1
|
Li Y, Li H, Wang H, Wang X. Utilizing Caenorhabditis Elegans as a Rapid and Precise Model for Assessing Amphetamine-Type Stimulants: A Novel Approach to Evaluating New Psychoactive Substances Activity and Mechanisms. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2500808. [PMID: 40068108 PMCID: PMC12061310 DOI: 10.1002/advs.202500808] [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] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/27/2025] [Indexed: 05/10/2025]
Abstract
The surge of new psychoactive substances (NPS) poses significant public health challenges due to their unregulated status and diverse effects. However, existing in vivo models for evaluating their activities are limited. To address this gap, this study utilizes the model organism Caenorhabditis elegans (C. elegans) to evaluate the activity of amphetamine-type stimulants (ATS) and their analogs. The swimming-induced paralysis (SWIP) assay is employed to measure the acute responses of C. elegans to various ATS, including amphetamine (AMPH), methamphetamine (METH), 3,4-methylenedioxymethamphetamine (MDMA) and their enantiomers. The findings reveal distinct responses in wild-type and mutant C. elegans, highlighting the roles of dopaminergic and serotonergic pathways, particularly DOP-3 and SER-4 receptors. The assay also revealed that C. elegans can distinguish between the chiral forms of ATS. Additionally, structural activity relationships (SAR) are observed, with meta-R amphetamines showing more pronounced effects than ortho-R and para-R analogs. This study demonstrates the utility of C. elegans in rapidly assessing ATS activity and toxicity, providing a cost-effective and precise method for high-throughput testing of NPS. These results contribute to a better understanding of ATS pharmacology and offer a valuable framework for future research and potential regulatory applications.
Collapse
Affiliation(s)
- Yuanpeng Li
- Laboratory of Chemical BiologyChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchunJilin130022China
- School of Applied Chemistry and EngineeringUniversity of Science and Technology of ChinaHefeiAnhui230026China
| | - Hongyuan Li
- Laboratory of Chemical BiologyChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchunJilin130022China
| | - Hongshuang Wang
- Laboratory of Chemical BiologyChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchunJilin130022China
| | - Xiaohui Wang
- Laboratory of Chemical BiologyChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchunJilin130022China
- School of Applied Chemistry and EngineeringUniversity of Science and Technology of ChinaHefeiAnhui230026China
- State Key Laboratory of Brain Machine IntelligenceZhejiang UniversityHangzhou310027China
| |
Collapse
|
2
|
Lange G, Gnazzo F, Faust RP, Beeler JA. Accumbal Dopamine and Acetylcholine Dynamics during Psychostimulant Sensitization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.29.646091. [PMID: 40236122 PMCID: PMC11996294 DOI: 10.1101/2025.03.29.646091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Behavioral sensitization to repeated psychostimulant exposure is believed to contribute to the development of addiction. Nucleus accumbens (NAcc) dopamine (DA) is known to be a key substrate in sensitization, though recent work suggests that striatal acetylcholine (ACh) may also play a critical role. However, underlying ACh changes and their relationship to DA signaling have not been characterized. Here, we used dual-color fiber photometry to simultaneously measure DA and ACh in the NAcc shell of mice across repeated injections of cocaine or amphetamine. Repeated exposure progressively elevated locomotor activity and increased slow extracellular DA while attenuating transient DA release. Psychostimulants reduced phasic ACh transient amplitude and frequency, an effect that sensitized with repeated injections. However, the temporal coupling of DA and ACh remained unchanged. To determine whether D2 receptors (D2Rs) on cholinergic interneurons (CINs) drive this effect, we generated CIN-selective D2R knockout (KO) mice. Surprisingly, KOs continued to show an acute decrease in ACh and intact DA-ACh correlations after psychostimulant administration. However, they failed to exhibit sensitization of either DA or ACh in response to repeated psychostimulant administration. Despite this lack of sensitization in underlying neuromodulator signaling, the KO mice nevertheless exhibited behavioral sensitization, though at a slower rate than wild-type. These findings suggest that neural sensitization to psychostimulants is dependent on D2R expressed on CINs, but that behavioral sensitization is not dependent on sensitization of these underlying signals.
Collapse
|
3
|
Fogaça MV, Daher F, Picciotto MR. Effects of ketamine on GABAergic and glutamatergic activity in the mPFC: biphasic recruitment of GABA function in antidepressant-like responses. Neuropsychopharmacology 2025; 50:673-684. [PMID: 39390105 PMCID: PMC11845475 DOI: 10.1038/s41386-024-02002-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 10/12/2024]
Abstract
Major depressive disorder (MDD) is associated with disruptions in glutamatergic and GABAergic activity in the medial prefrontal cortex (mPFC), leading to altered synaptic formation and function. Low doses of ketamine rapidly rescue these deficits, inducing fast and sustained antidepressant effects. While it is suggested that ketamine produces a rapid glutamatergic enhancement in the mPFC, the temporal dynamics and the involvement of GABA interneurons in its sustained effects remain unclear. Using simultaneous photometry recordings of calcium activity in mPFC pyramidal and GABA neurons, as well as chemogenetic approaches in Gad1-Cre mice, we explored the hypothesis that initial effects of ketamine on glutamate signaling trigger subsequent enhancement of GABAergic responses, contributing to its sustained antidepressant responses. Calcium recordings revealed a biphasic effect of ketamine on activity of mPFC GABA neurons, characterized by an initial transient decrease (phase 1, <30 min) followed by an increase (phase 2, >60 min), in parallel with a transient increase in excitation/inhibition levels (10 min) and lasting enhancement of glutamatergic activity (30-120 min). Previous administration of ketamine enhanced GABA neuron activity during the sucrose splash test (SUST) and novelty suppressed feeding test (NSFT), 24 h and 72 h post-treatment, respectively. Chemogenetic inhibition of GABA interneurons during the surge of GABAergic activity (phase 2), or immediately before the SUST or NSFT, occluded ketamine's behavioral actions. These results indicate that time-dependent modulation of GABAergic activity is required for the sustained antidepressant-like responses induced by ketamine, suggesting that approaches to enhance GABAergic plasticity and function are promising therapeutic targets for antidepressant development.
Collapse
Affiliation(s)
- Manoela V Fogaça
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA.
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT, 06519, USA.
| | - Fernanda Daher
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Marina R Picciotto
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT, 06519, USA
| |
Collapse
|
4
|
Koda M, Kawai H, Shirakawa H, Kaneko S, Nagayasu K. Effect of antidepressants and social defeat stress on the activity of dorsal raphe serotonin neurons in free-moving animals. J Pharmacol Sci 2025; 157:113-123. [PMID: 39828391 DOI: 10.1016/j.jphs.2025.01.001] [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: 12/02/2024] [Revised: 12/28/2024] [Accepted: 01/01/2025] [Indexed: 01/22/2025] Open
Abstract
Major depressive disorder (MDD) is among the most common mental disorders worldwide and is characterized by dysregulated reward processing associated with anhedonia. Selective serotonin reuptake inhibitors (SSRIs) are the first-line treatment for MDD; however, their onset of action is delayed. Recent reports have shown that serotonin neurons in the dorsal raphe nucleus (DRN) are activated by rewards and play a vital role in reward processing. However, whether antidepressant treatment affects the DRN serotonin neuronal response to rewards in awake animals remains unknown. In this study, we measured the activity of DRN serotonin neurons in awake mice and determined the effects of antidepressants and chronic stress on DRN serotonin neuronal activity. We found that acute treatment with citalopram, an SSRI, significantly decreased sucrose-induced activation of DRN serotonin neurons. The decrease in response to acute citalopram treatment was attenuated by chronic citalopram treatment. Acute treatment with (S)-WAY100135, a 5-HT1A receptor antagonist, dose-dependently inhibited the response to acute citalopram treatment. These results indicate that autoinhibition by activating 5-HT1A receptors via acute SSRI treatment may blunt the reward response, which can be recovered after chronic SSRI treatment.
Collapse
Affiliation(s)
- Masashi Koda
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Hiroyuki Kawai
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan; Department of Anatomy and Neuroscience, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3 Asahi-cho, Abeno-ku, Osaka, 545-8585, Japan
| | - Hisashi Shirakawa
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Shuji Kaneko
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Kazuki Nagayasu
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan; Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, 565-0871, Japan; Project for Neural Networks, Drug Innovation Center, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, 565-0871, Japan.
| |
Collapse
|
5
|
Galiza Soares JA, Sutley-Koury SN, Pomrenze MB, Tucciarone JM. Opioidergic tuning of social attachment: reciprocal relationship between social deprivation and opioid abuse. Front Neuroanat 2025; 18:1521016. [PMID: 39917739 PMCID: PMC11798945 DOI: 10.3389/fnana.2024.1521016] [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: 11/01/2024] [Accepted: 12/24/2024] [Indexed: 02/09/2025] Open
Abstract
Individuals misusing opioids often report heightened feelings of loneliness and decreased ability to maintain social connections. This disruption in social functioning further promotes addiction, creating a cycle in which increasing isolation drives drug use. Social factors also appear to impact susceptibility and progression of opioid dependence. In particular, increasing evidence suggests that poor early social bond formation and social environments may increase the risk of opioid abuse later in life. The brain opioid theory of social attachment suggests that endogenous opioids are key to forming and sustaining social bonds. Growing literature describes the opioid system as a powerful modulator of social separation distress and attachment formation in rodents and primates. In this framework, disruptions in opioidergic signaling due to opioid abuse may mediate social reward processing and behavior. While changes in endogenous opioid peptides and receptors have been reported in these early-life adversity models, the underlying mechanisms remain poorly understood. This review addresses the apparent bidirectional causal relationship between social deprivation and opioid addiction susceptibility, investigating the role of opioid transmission in attachment bond formation and prosocial behavior. We propose that early social deprivation disrupts the neurobiological substrates associated with opioid transmission, leading to deficits in social attachment and reinforcing addictive behaviors. By examining the literature, we discuss potential overlapping neural pathways between social isolation and opioid addiction, focusing on major reward-aversion substrates known to respond to opioids.
Collapse
Affiliation(s)
- Julia A. Galiza Soares
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States
| | - Samantha N. Sutley-Koury
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States
| | - Matthew B. Pomrenze
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States
| | - Jason M. Tucciarone
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States
| |
Collapse
|
6
|
Du C, Jeong H, Koretsky AP, Pan Y. Review of cocaine-induced brain vascular and cellular function changes measured in vivo with optical imaging. NEUROPHOTONICS 2025; 12:S14611. [PMID: 40438148 PMCID: PMC12118881 DOI: 10.1117/1.nph.12.s1.s14611] [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] [Received: 12/05/2024] [Revised: 03/01/2025] [Accepted: 04/02/2025] [Indexed: 06/01/2025]
Abstract
Significance Cocaine exerts effects on vascular and cellular functions in the brain. The interactions among cerebrovasculature, neurons, and astrocytes and their dynamic changes during exposure complicate the understanding of its effects. Therefore, there is a need for simultaneous, multiparameter in vivo measurements to accurately distinguish these effects. Aim A multimodal optical imaging approach that is tailored to investigate cocaine's effects on cerebrovasculature, neurons, and astrocytes in high-spatiotemporal resolution and large field of view is presented with comparisons to other modalities. Approach This approach integrates optical coherence tomography, fluorescence, and spectral absorption imaging to permit high-resolution imaging of 3D cerebrovessels, cerebral blood flow (CBF), changes in oxygenated/deoxygenated hemoglobin, and large-scale cellular activities via intracellular calcium fluorescence expressed through genetically encoded calcium indicators in the mouse cortex. Results Results show that cocaine induces vasoconstriction and reduces CBF, thus increasing the susceptibility of the brain to ischemia with chronic exposure. Moreover, cocaine alters neuronal activity and frontal responses to deep brain stimulation. Conclusions These findings on cocaine's effects on the neuro-astroglial-vascular network in the prefrontal cortex highlight the unique capacity of optical imaging to reveal the cellular and vascular mechanisms underlying cocaine's neurotoxic effects on brain function.
Collapse
Affiliation(s)
- Congwu Du
- Stony Brook University, Department of Biomedical Engineering, Stony Brook, New York, United States
| | - Hyomin Jeong
- Stony Brook University, Department of Biomedical Engineering, Stony Brook, New York, United States
| | - Alan P. Koretsky
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States
| | - Yingtian Pan
- Stony Brook University, Department of Biomedical Engineering, Stony Brook, New York, United States
| |
Collapse
|
7
|
Chen Y, Gu Y, Wang B, Wei A, Dong N, Jiang Y, Liu X, Zhu L, Zhu F, Tan T, Jing Z, Mao F, Zhang Y, Yao J, Yang Y, Wang H, Wu H, Li H, Zheng C, Duan X, Huo J, Wu X, Hu S, Zhao A, Li Z, Cheng X, Qin Y, Song Q, Zhan S, Qu Q, Guan F, Xu H, Kang X, Wang C. Synaptotagmin-11 deficiency mediates schizophrenia-like behaviors in mice via dopamine over-transmission. Nat Commun 2024; 15:10571. [PMID: 39632880 PMCID: PMC11618495 DOI: 10.1038/s41467-024-54604-4] [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: 03/27/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024] Open
Abstract
Schizophrenia is a severe neuropsychiatric disease, but the initiation mechanisms are unclear. Although antipsychotics are effective against positive symptoms, therapeutic interventions for negative symptoms are limited due to the lack of pathophysiological mechanisms. Here we identify synaptotagmin-11 (Syt11) as a potential genetic risk factor and dopamine over-transmission as a mechanism in the development of schizophrenia. Syt11 expression is reduced in individuals with schizophrenia but restored following the treatment with antipsychotics. Syt11 deficiency in dopamine neurons in early adolescence, but not in adults, leads to persistent social deficits and other schizophrenia-like behaviors by mediating dopamine over-transmission in mice. Accordingly, dopamine neuron over-excitation before late adolescence induces persistent schizophrenia-associated behavioral deficits, along with the structural and functional alternations in the mPFC. Notably, local intervention of D2R with clinical drugs presynaptically or postsynaptically exhibits both acute and long-lasting therapeutic effects on social deficits in schizophrenia mice models. These findings not only define Syt11 as a risk factor and DA over-transmission as a potential risk factor initiating schizophrenia, but also propose two D2R-targeting strategies for the comprehensive and long-term recovery of schizophrenia-associated social withdrawal.
Collapse
Affiliation(s)
- Yang Chen
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yuhao Gu
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Bianbian Wang
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Anqi Wei
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Nan Dong
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yong Jiang
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiaoying Liu
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, and the Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
- College of Life Sciences, Liaocheng University, Liaocheng, 252059, China
| | - Li Zhu
- Key Laboratory of National Health Commission for Forensic Sciences, College of Medicine & Forensics, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Feng Zhu
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Tao Tan
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Zexin Jing
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Fenghan Mao
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yichi Zhang
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Jingyu Yao
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yuxin Yang
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
- College of Life Sciences, Liaocheng University, Liaocheng, 252059, China
| | - Hongyan Wang
- College of Life Sciences, Liaocheng University, Liaocheng, 252059, China
| | - Hao Wu
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Hua Li
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Chaowen Zheng
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xueting Duan
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Jingxiao Huo
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xuanang Wu
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Shaoqin Hu
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Anran Zhao
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Ziyang Li
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xu Cheng
- Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, and the Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Yuhao Qin
- Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, and the Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Qian Song
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Shuqin Zhan
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Qiumin Qu
- Department of Neurology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Fanglin Guan
- Key Laboratory of National Health Commission for Forensic Sciences, College of Medicine & Forensics, Xi'an Jiaotong University, Xi'an, 710061, China.
| | - Huadong Xu
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| | - Xinjiang Kang
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
- Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, and the Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China.
- College of Life Sciences, Liaocheng University, Liaocheng, 252059, China.
| | - Changhe Wang
- Department of Neurology, the Second Affiliated Hospital, Neuroscience Research Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
- Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, and the Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China.
- Department of Psychology, Chengwu People's Hospital, Heze, 274200, China.
| |
Collapse
|
8
|
Fogaça MV, Daher F, Picciotto MR. Effects of ketamine on GABAergic and glutamatergic activity in the mPFC: biphasic recruitment of GABA function in antidepressant-like responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.29.605610. [PMID: 39131322 PMCID: PMC11312475 DOI: 10.1101/2024.07.29.605610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Major depressive disorder (MDD) is associated with disruptions in glutamatergic and GABAergic activity in the medial prefrontal cortex (mPFC), leading to altered synaptic formation and function. Low doses of ketamine rapidly rescue these deficits, inducing fast and sustained antidepressant effects. While it is suggested that ketamine produces a rapid glutamatergic enhancement in the mPFC, the temporal dynamics and the involvement of GABA interneurons in its sustained effects remain unclear. Using simultaneous photometry recordings of calcium activity in mPFC pyramidal and GABA neurons, as well as chemogenetic approaches in Gad1-Cre mice, we explored the hypothesis that initial effects of ketamine on glutamate signaling trigger subsequent enhancement of GABAergic responses, contributing to its sustained antidepressant responses. Calcium recordings revealed a biphasic effect of ketamine on activity of mPFC GABA neurons, characterized by an initial transient decrease (phase 1, <30 min) followed by an increase (phase 2, >60 min), in parallel with a transient increase in excitation/inhibition levels (10 min) and lasting enhancement of glutamatergic activity (30-120 min). Previous administration of ketamine enhanced GABA neuron activity during the sucrose splash test (SUST) and novelty suppressed feeding test (NSFT), 24 h and 72 h post-treatment, respectively. Chemogenetic inhibition of GABA interneurons during the surge of GABAergic activity (phase 2), or immediately before the SUST or NSFT, occluded ketamine's behavioral actions. These results indicate that time-dependent modulation of GABAergic activity is required for the sustained antidepressant-like responses induced by ketamine, suggesting that approaches to enhance GABAergic plasticity and function are promising therapeutic targets for antidepressant development.
Collapse
Affiliation(s)
- Manoela V. Fogaça
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT 06519, USA
| | - Fernanda Daher
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Marina R. Picciotto
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT 06519, USA
| |
Collapse
|
9
|
Dejoie JM, Senia N, Konova A, Smith D, Fareri D. Common and Distinct Drug Cue Reactivity Patterns Associated with Cocaine and Heroin: An fMRI Meta-Analysis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.10.19.23297268. [PMID: 37905133 PMCID: PMC10615011 DOI: 10.1101/2023.10.19.23297268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Substance use and substance use disorders represent ongoing major public health crises. Specifically, the use of substances such as cocaine and heroin are responsible for over 50,000 drug related deaths combined annually. We used a comparative meta-analysis procedure to contrast activation patterns associated with cocaine and heroin cue reactivity, which may reflect substance use risk for these substances. PubMed and Google Scholar were searched for studies with within-subject whole brain analyses comparing drug to neutral cues for users of cocaine and heroin published between 1995 and 2022. A total of 18 studies were included, 9 in each subgroup. Voxel-based meta-analyses were performed using seed-based d mapping with permuted subject images (SDM-PSI) for subgroup mean analyses and a contrast meta-regression comparing the two substances. Results from our mean analysis indicated that users of heroin showed more widespread activation in the nucleus accumbens, right inferior and left middle temporal gyrus, right thalamus, and right cerebellum. Cocaine use was associated with recruitment of dorsolateral prefrontal cortex during cue reactivity. Direct comparison of cue reactivity studies in heroin relative to cocaine users revealed greater activation in dopaminergic targets for users of heroin compared to users of cocaine. Differential activation patterns between substances may underlie differences in the clinical characteristics observed in users of cocaine and heroin, including seeking emotional blunting in users of heroin. More consistent research methodology is needed to provide adequate studies for stringent meta-analyses examining common and distinct neural activation patterns across substances and moderation by clinically relevant factors.
Collapse
|
10
|
Dwivedi I, Haddad GG. Investigating the neurobiology of maternal opioid use disorder and prenatal opioid exposure using brain organoid technology. Front Cell Neurosci 2024; 18:1403326. [PMID: 38812788 PMCID: PMC11133580 DOI: 10.3389/fncel.2024.1403326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/01/2024] [Indexed: 05/31/2024] Open
Abstract
Over the past two decades, Opioid Use Disorder (OUD) among pregnant women has become a major global public health concern. OUD has been characterized as a problematic pattern of opioid use despite adverse physical, psychological, behavioral, and or social consequences. Due to the relapsing-remitting nature of this disorder, pregnant mothers are chronically exposed to exogenous opioids, resulting in adverse neurological and neuropsychiatric outcomes. Collateral fetal exposure to opioids also precipitates severe neurodevelopmental and neurocognitive sequelae. At present, much of what is known regarding the neurobiological consequences of OUD and prenatal opioid exposure (POE) has been derived from preclinical studies in animal models and postnatal or postmortem investigations in humans. However, species-specific differences in brain development, variations in subject age/health/background, and disparities in sample collection or storage have complicated the interpretation of findings produced by these explorations. The ethical or logistical inaccessibility of human fetal brain tissue has also limited direct examinations of prenatal drug effects. To circumvent these confounding factors, recent groups have begun employing induced pluripotent stem cell (iPSC)-derived brain organoid technology, which provides access to key aspects of cellular and molecular brain development, structure, and function in vitro. In this review, we endeavor to encapsulate the advancements in brain organoid culture that have enabled scientists to model and dissect the neural underpinnings and effects of OUD and POE. We hope not only to emphasize the utility of brain organoids for investigating these conditions, but also to highlight opportunities for further technical and conceptual progress. Although the application of brain organoids to this critical field of research is still in its nascent stages, understanding the neurobiology of OUD and POE via this modality will provide critical insights for improving maternal and fetal outcomes.
Collapse
Affiliation(s)
- Ila Dwivedi
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Gabriel G. Haddad
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA, United States
- Department of Neurosciences, School of Medicine, University of California, San Diego, La Jolla, CA, United States
- Rady Children’s Hospital, San Diego, CA, United States
| |
Collapse
|
11
|
Wei C, Jiang W, Wang R, Zhong H, He H, Gao X, Zhong S, Yu F, Guo Q, Zhang L, Schiffelers LDJ, Zhou B, Trepel M, Schmidt FI, Luo M, Shao F. Brain endothelial GSDMD activation mediates inflammatory BBB breakdown. Nature 2024; 629:893-900. [PMID: 38632402 DOI: 10.1038/s41586-024-07314-2] [Citation(s) in RCA: 82] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/14/2024] [Indexed: 04/19/2024]
Abstract
The blood-brain barrier (BBB) protects the central nervous system from infections or harmful substances1; its impairment can lead to or exacerbate various diseases of the central nervous system2-4. However, the mechanisms of BBB disruption during infection and inflammatory conditions5,6 remain poorly defined. Here we find that activation of the pore-forming protein GSDMD by the cytosolic lipopolysaccharide (LPS) sensor caspase-11 (refs. 7-9), but not by TLR4-induced cytokines, mediates BBB breakdown in response to circulating LPS or during LPS-induced sepsis. Mice deficient in the LBP-CD14 LPS transfer and internalization pathway10-12 resist BBB disruption. Single-cell RNA-sequencing analysis reveals that brain endothelial cells (bECs), which express high levels of GSDMD, have a prominent response to circulating LPS. LPS acting on bECs primes Casp11 and Cd14 expression and induces GSDMD-mediated plasma membrane permeabilization and pyroptosis in vitro and in mice. Electron microscopy shows that this features ultrastructural changes in the disrupted BBB, including pyroptotic endothelia, abnormal appearance of tight junctions and vasculature detachment from the basement membrane. Comprehensive mouse genetic analyses, combined with a bEC-targeting adeno-associated virus system, establish that GSDMD activation in bECs underlies BBB disruption by LPS. Delivery of active GSDMD into bECs bypasses LPS stimulation and opens the BBB. In CASP4-humanized mice, Gram-negative Klebsiella pneumoniae infection disrupts the BBB; this is blocked by expression of a GSDMD-neutralizing nanobody in bECs. Our findings outline a mechanism for inflammatory BBB breakdown, and suggest potential therapies for diseases of the central nervous system associated with BBB impairment.
Collapse
Affiliation(s)
- Chao Wei
- Chinese Institute for Brain Research, Beijing, P. R. China
| | - Wei Jiang
- National Institute of Biological Sciences, Beijing, P. R. China
- Research Unit of Pyroptosis and Immunity, Chinese Academy of Medical Sciences and National Institute of Biological Sciences, Beijing, P. R. China
| | - Ruiyu Wang
- National Institute of Biological Sciences, Beijing, P. R. China
| | - Haoyu Zhong
- National Institute of Biological Sciences, Beijing, P. R. China
| | - Huabin He
- National Institute of Biological Sciences, Beijing, P. R. China
- Research Unit of Pyroptosis and Immunity, Chinese Academy of Medical Sciences and National Institute of Biological Sciences, Beijing, P. R. China
| | - Xinwei Gao
- Chinese Institute for Brain Research, Beijing, P. R. China
| | - Shilin Zhong
- National Institute of Biological Sciences, Beijing, P. R. China
| | - Fengting Yu
- Chinese Institute for Brain Research, Beijing, P. R. China
| | - Qingchun Guo
- Chinese Institute for Brain Research, Beijing, P. R. China
| | - Li Zhang
- Chinese Institute for Brain Research, Beijing, P. R. China
| | - Lisa D J Schiffelers
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Bin Zhou
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, P. R. China
| | - Martin Trepel
- Department of Hematology and Medical Oncology, University Medical Center Augsburg, Augsburg, Germany
| | - Florian I Schmidt
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Minmin Luo
- Chinese Institute for Brain Research, Beijing, P. R. China.
- National Institute of Biological Sciences, Beijing, P. R. China.
- Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing, P. R. China.
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, P. R. China.
- New Cornerstone Science Laboratory, Shenzhen, P. R. China.
| | - Feng Shao
- National Institute of Biological Sciences, Beijing, P. R. China.
- Research Unit of Pyroptosis and Immunity, Chinese Academy of Medical Sciences and National Institute of Biological Sciences, Beijing, P. R. China.
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, P. R. China.
- New Cornerstone Science Laboratory, Shenzhen, P. R. China.
- Changping Laboratory, Beijing, P. R. China.
| |
Collapse
|
12
|
Everett T, Ten Eyck TW, Wu CH, Shelowitz AL, Stansbury SM, Firek A, Setlow B, McIntyre JC. Cilia loss on distinct neuron populations differentially alters cocaine-induced locomotion and reward. J Psychopharmacol 2024; 38:200-212. [PMID: 38151883 PMCID: PMC11078551 DOI: 10.1177/02698811231219058] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
BACKGROUND Neuronal primary cilia are being recognized for their role in mediating signaling associated with a variety of neurobehaviors, including responses to drugs of abuse. They function as signaling hubs, enriched with a diverse array of G-protein coupled receptors (GPCRs), including several associated with motivation and drug-related behaviors. However, our understanding of how cilia regulate neuronal function and behavior is still limited. AIMS The objective of the current study was to investigate the contributions of primary cilia on specific neuronal populations to behavioral responses to cocaine. METHODS To test the consequences of cilia loss on cocaine-induced locomotion and reward-related behavior, we selectively ablated cilia from dopaminergic or GAD2-GABAergic neurons in mice. RESULTS Cilia ablation on either population of neurons failed to significantly alter acute locomotor responses to cocaine at a range of doses. With repeated administration, mice lacking cilia on GAD2-GABAergic neurons showed no difference in locomotor sensitization to cocaine compared to wild-type (WT) littermates, whereas mice lacking cilia on dopaminergic neurons exhibited reduced locomotor sensitization to cocaine at 10 and 30 mg/kg. Mice lacking cilia on GAD2-GABAergic neurons showed no difference in cocaine conditioned place preference (CPP), whereas mice lacking cilia on dopaminergic neurons exhibited reduced CPP compared to WT littermates. CONCLUSIONS Combined with previous findings using amphetamine, our results show that behavioral effects of cilia ablation are cell- and drug type-specific, and that neuronal cilia contribute to modulation of both the locomotor-inducing and rewarding properties of cocaine.
Collapse
Affiliation(s)
- Thomas Everett
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | - Tyler W. Ten Eyck
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | - Chang-Hung Wu
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | | | - Sofia M. Stansbury
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | - Alexandra Firek
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | - Barry Setlow
- Department of Psychiatry, University of Florida, Gainesville, FL 32610
- Center for Addiction Research and Education, University of Florida, Gainesville, FL 32610
| | - Jeremy C. McIntyre
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
- Center for Addiction Research and Education, University of Florida, Gainesville, FL 32610
| |
Collapse
|
13
|
Welsch L, Colantonio E, Frison M, Johnson DA, McClain SP, Mathis V, Banghart MR, Ben Hamida S, Darcq E, Kieffer BL. Mu Opioid Receptor-Expressing Neurons in the Dorsal Raphe Nucleus Are Involved in Reward Processing and Affective Behaviors. Biol Psychiatry 2023; 94:842-851. [PMID: 37285896 PMCID: PMC10850692 DOI: 10.1016/j.biopsych.2023.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 05/12/2023] [Accepted: 05/24/2023] [Indexed: 06/09/2023]
Abstract
BACKGROUND Mu opioid receptors (MORs) are key for reward processing, mostly studied in dopaminergic pathways. MORs are also expressed in the dorsal raphe nucleus (DRN), which is central for the modulation of reward and mood, but MOR function in the DRN remains underexplored. Here, we investigated whether MOR-expressing neurons of the DRN (DRN-MOR neurons) participate in reward and emotional responses. METHODS We characterized DRN-MOR neurons anatomically using immunohistochemistry and functionally using fiber photometry in responses to morphine and rewarding/aversive stimuli. We tested the effect of opioid uncaging on the DRN on place conditioning. We examined the effect of DRN-MOR neuron optostimulation on positive reinforcement and mood-related behaviors. We mapped their projections and selected DRN-MOR neurons projecting to the lateral hypothalamus for a similar optogenetic experimentation. RESULTS DRN-MOR neurons form a heterogeneous neuronal population essentially composed of GABAergic (gamma-aminobutyric acidergic) and glutamatergic neurons. Calcium activity of DRN-MOR neurons was inhibited by rewarding stimuli and morphine. Local photo-uncaging of oxymorphone in the DRN produced conditioned place preference. DRN-MOR neuron optostimulation triggered real-time place preference and was self-administered, promoted social preference, and reduced anxiety and passive coping. Finally, specific optostimulation of DRN-MOR neurons projecting to the lateral hypothalamus recapitulated the reinforcing effects of total DRN-MOR neuron stimulation. CONCLUSIONS Our data show that DRN-MOR neurons respond to rewarding stimuli and that their optoactivation has reinforcing effects and promotes positive emotional responses, an activity which is partially mediated by their projections to the lateral hypothalamus. Our study also suggests a complex regulation of DRN activity by MOR opioids, involving mixed inhibition/activation mechanisms that fine-tune DRN function.
Collapse
Affiliation(s)
- Lola Welsch
- Douglas Research Center, Department of Psychiatry, McGill University, Montréal, Quebec, Canada; INSERM U1114, Department of Psychiatry, University of Strasbourg, Strasbourg, France
| | - Esther Colantonio
- INSERM U1114, Department of Psychiatry, University of Strasbourg, Strasbourg, France
| | - Mathilde Frison
- Douglas Research Center, Department of Psychiatry, McGill University, Montréal, Quebec, Canada
| | - Desiree A Johnson
- Neurobiology Department, School of the Biological Sciences, University of California San Diego, La Jolla, California
| | - Shannan P McClain
- Neurobiology Department, School of the Biological Sciences, University of California San Diego, La Jolla, California
| | - Victor Mathis
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, UPR 3212, Strasbourg, France
| | - Matthew R Banghart
- Neurobiology Department, School of the Biological Sciences, University of California San Diego, La Jolla, California
| | - Sami Ben Hamida
- Douglas Research Center, Department of Psychiatry, McGill University, Montréal, Quebec, Canada; INSERM UMR 1247, Université de Picardie Jules Verne, Amiens, France
| | - Emmanuel Darcq
- Douglas Research Center, Department of Psychiatry, McGill University, Montréal, Quebec, Canada; INSERM U1114, Department of Psychiatry, University of Strasbourg, Strasbourg, France
| | - Brigitte L Kieffer
- Douglas Research Center, Department of Psychiatry, McGill University, Montréal, Quebec, Canada; INSERM U1114, Department of Psychiatry, University of Strasbourg, Strasbourg, France.
| |
Collapse
|
14
|
Welsch L, Colantonio E, Falconnier C, Champagnol-DiLiberti C, Allain F, Ben Hamida S, Darcq E, Lutz PE, Kieffer BL. Mu Opioid Receptor-Positive Neurons in the Dorsal Raphe Nucleus Are Impaired by Morphine Abstinence. Biol Psychiatry 2023; 94:852-862. [PMID: 37393045 PMCID: PMC10851617 DOI: 10.1016/j.biopsych.2023.06.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 06/21/2023] [Accepted: 06/21/2023] [Indexed: 07/03/2023]
Abstract
BACKGROUND Chronic opioid exposure leads to hedonic deficits and enhanced vulnerability to addiction, which are observed and even strengthen after a period of abstinence, but the underlying circuit mechanisms are poorly understood. In this study, using both molecular and behavioral approaches, we tested the hypothesis that neurons expressing mu opioid receptors (MORs) in the dorsal raphe nucleus (DRN) are involved in addiction vulnerability associated with morphine abstinence. METHODS MOR-Cre mice were exposed to chronic morphine and then went through spontaneous withdrawal for 4 weeks, a well-established mouse model of morphine abstinence. We studied DRN-MOR neurons of abstinent mice using 1) viral translating ribosome affinity for transcriptome profiling, 2) fiber photometry to measure neuronal activity, and 3) an opto-intracranial self-stimulation paradigm applied to DRN-MOR neurons to assess responses related to addiction vulnerability including persistence to respond, motivation to obtain the stimulation, self-stimulation despite punishment, and cue-induced reinstatement. RESULTS DRN-MOR neurons of abstinent animals showed a downregulation of genes involved in ion conductance and MOR-mediated signaling, as well as altered responding to acute morphine. Opto-intracranial self-stimulation data showed that abstinent animals executed more impulsive-like and persistent responses during acquisition and scored higher on addiction-like criteria. CONCLUSIONS Our data suggest that protracted abstinence to chronic morphine leads to reduced MOR function in DRN-MOR neurons and abnormal self-stimulation of these neurons. We propose that DRN-MOR neurons have partially lost their reward-facilitating properties, which in turn may lead to increased propensity to perform addiction-related behaviors.
Collapse
Affiliation(s)
- Lola Welsch
- Douglas Research Center, Department of Psychiatry, McGill University, Montréal, Quebec, Canada; INSERM U1114, University of Strasbourg, Strasbourg, France
| | | | - Camille Falconnier
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives UPR3212, Strasbourg, France
| | | | - Florence Allain
- Douglas Research Center, Department of Psychiatry, McGill University, Montréal, Quebec, Canada; INSERM U1114, University of Strasbourg, Strasbourg, France
| | - Sami Ben Hamida
- Douglas Research Center, Department of Psychiatry, McGill University, Montréal, Quebec, Canada; INSERM UMR 1247, Research Group on Alcohol & Pharmacodependences, Université de Picardie Jules Verne, Amiens, France
| | - Emmanuel Darcq
- Douglas Research Center, Department of Psychiatry, McGill University, Montréal, Quebec, Canada; INSERM U1114, University of Strasbourg, Strasbourg, France
| | - Pierre-Eric Lutz
- Douglas Research Center, Department of Psychiatry, McGill University, Montréal, Quebec, Canada; Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives UPR3212, Strasbourg, France
| | - Brigitte L Kieffer
- Douglas Research Center, Department of Psychiatry, McGill University, Montréal, Quebec, Canada; INSERM U1114, University of Strasbourg, Strasbourg, France.
| |
Collapse
|
15
|
Ganaway A, Tatsuta K, Castillo VCG, Okada R, Sunaga Y, Ohta Y, Ohta J, Ohsawa M, Akay M, Akay YM. Investigating the Influence of Morphine and Cocaine on the Mesolimbic Pathway Using a Novel Microimaging Platform. Int J Mol Sci 2023; 24:16303. [PMID: 38003493 PMCID: PMC10671016 DOI: 10.3390/ijms242216303] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Dopamine (DA)'s relationship with addiction is complex, and the related pathways in the mesocorticolimbic system are used to deliver DA, regulating both behavioral and perceptual actions. Specifically, the mesolimbic pathway connecting the ventral tegmental area (VTA) and the nucleus accumbens (NAc) is crucial in regulating memory, emotion, motivation, and behavior due to its responsibility to modulate dopamine. To better investigate the relationship between DA and addiction, more advanced mapping methods are necessary to monitor its production and propagation accurately and efficiently. In this study, we incorporate dLight1.2 adeno-associated virus (AAV) into our latest CMOS (complementary metal-oxide semiconductor) imaging platform to investigate the effects of two pharmacological substances, morphine and cocaine, in the NAc using adult mice. By implanting our self-fabricated CMOS imaging device into the deep brain, fluorescence imaging of the NAc using the dLight1.2 AAV allows for the visualization of DA molecules delivered from the VTA in real time. Our results suggest that changes in extracellular DA can be observed with this adapted system, showing potential for new applications and methods for approaching addiction studies. Additionally, we can identify the unique characteristic trend of DA release for both morphine and cocaine, further validating the underlying biochemical mechanisms used to modulate dopaminergic activation.
Collapse
Affiliation(s)
- Austin Ganaway
- Biomedical Engineering Department, University of Houston, 3517 Cullen Blvd, Houston, TX 77204, USA; (A.G.); (M.A.)
| | - Kousuke Tatsuta
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8601, Japan; (K.T.); (M.O.)
| | - Virgil Christian Garcia Castillo
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma 630-0101, Japan; (V.C.G.C.); (R.O.); (Y.S.); (Y.O.); (J.O.)
| | - Ryoma Okada
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma 630-0101, Japan; (V.C.G.C.); (R.O.); (Y.S.); (Y.O.); (J.O.)
| | - Yoshinori Sunaga
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma 630-0101, Japan; (V.C.G.C.); (R.O.); (Y.S.); (Y.O.); (J.O.)
| | - Yasumi Ohta
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma 630-0101, Japan; (V.C.G.C.); (R.O.); (Y.S.); (Y.O.); (J.O.)
| | - Jun Ohta
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma 630-0101, Japan; (V.C.G.C.); (R.O.); (Y.S.); (Y.O.); (J.O.)
| | - Masahiro Ohsawa
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8601, Japan; (K.T.); (M.O.)
| | - Metin Akay
- Biomedical Engineering Department, University of Houston, 3517 Cullen Blvd, Houston, TX 77204, USA; (A.G.); (M.A.)
| | - Yasemin M. Akay
- Biomedical Engineering Department, University of Houston, 3517 Cullen Blvd, Houston, TX 77204, USA; (A.G.); (M.A.)
| |
Collapse
|
16
|
Rezayof A, Ghasemzadeh Z, Sahafi OH. Addictive drugs modify neurogenesis, synaptogenesis and synaptic plasticity to impair memory formation through neurotransmitter imbalances and signaling dysfunction. Neurochem Int 2023; 169:105572. [PMID: 37423274 DOI: 10.1016/j.neuint.2023.105572] [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: 04/19/2023] [Revised: 07/01/2023] [Accepted: 07/05/2023] [Indexed: 07/11/2023]
Abstract
Drug abuse changes neurophysiological functions at multiple cellular and molecular levels in the addicted brain. Well-supported scientific evidence suggests that drugs negatively affect memory formation, decision-making and inhibition, and emotional and cognitive behaviors. The mesocorticolimbic brain regions are involved in reward-related learning and habitual drug-seeking/taking behaviors to develop physiological and psychological dependence on the drugs. This review highlights the importance of specific drug-induced chemical imbalances resulting in memory impairment through various neurotransmitter receptor-mediated signaling pathways. The mesocorticolimbic modifications in the expression levels of brain-derived neurotrophic factor (BDNF) and the cAMP-response element binding protein (CREB) impair reward-related memory formation following drug abuse. The contributions of protein kinases and microRNAs (miRNAs), along with the transcriptional and epigenetic regulation have also been considered in memory impairment underlying drug addiction. Overall, we integrate the research on various types of drug-induced memory impairment in distinguished brain regions and provide a comprehensive review with clinical implications addressing the upcoming studies.
Collapse
Affiliation(s)
- Ameneh Rezayof
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| | - Zahra Ghasemzadeh
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Oveis Hosseinzadeh Sahafi
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| |
Collapse
|
17
|
Ou TS, Huber L, Macy JT, Bray BC, Lin HC. Stressful Life Events and Patterns of Polysubstance Use Among U.S. Late Middle-Aged and Older Adults: A Latent Class Analysis. J Appl Gerontol 2023; 42:1867-1876. [PMID: 36988206 DOI: 10.1177/07334648231165256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023] Open
Abstract
The goals of this study were to identify patterns of polysubstance use and their associations with stressful life events among U.S. late middle-aged and older adults and examine whether gender moderates these associations. Adults aged 50 and older (N = 14,738) from the National Epidemiological Survey on Alcohol and Related Conditions-III were included. Latent class analysis was conducted to identify patterns of polysubstance use. Weighted multinomial logistic regression was estimated with a generalized structural equation model. Three different polysubstance use patterns (non-users/low substance users; cannabis and excessive alcohol users; painkiller and sedative/tranquilizer misusers) were identified. Higher levels of stressful life events were associated with patterns of polysubstance use. Gender moderated the association between stressful life events and co-misusing painkillers and sedatives/tranquilizers (p < 0.05). Substance use prevention efforts should consider aging adults' patterns of polysubstance use and associated stressful life events when designing and implementing gender-specific polysubstance use prevention interventions.
Collapse
Affiliation(s)
- Tzung-Shiang Ou
- Department of Applied Health Science, School of Public Health, Indiana University, Bloomington, IN, USA
| | - Lesa Huber
- Department of Applied Health Science, School of Public Health, Indiana University, Bloomington, IN, USA
| | - Jonathan T Macy
- Department of Applied Health Science, School of Public Health, Indiana University, Bloomington, IN, USA
| | - Bethany C Bray
- Institute for Health Research and Policy, The University of Illinois at Chicago, Chicago, IL, USA
| | - Hsien-Chang Lin
- Department of Applied Health Science, School of Public Health, Indiana University, Bloomington, IN, USA
| |
Collapse
|
18
|
Lalani E, Menon R, Mufti MA, Kumfa C, Raji M. Mirtazapine: A One-Stop Strategy for Treatment of Opioid Withdrawal Symptoms. Cureus 2023; 15:e43821. [PMID: 37736438 PMCID: PMC10509332 DOI: 10.7759/cureus.43821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2023] [Indexed: 09/23/2023] Open
Abstract
Public health efforts to reduce the opioid overdose epidemic and treat opioid use disorder (OUD) have met with challenges associated with current non-standardized approaches to managing opioid withdrawal symptoms, such as itching, jitteriness, anxiety, depression, craving, vomiting, diarrhea, insomnia, and anorexia. These symptoms pose substantial obstacles to the safe initiation of medications for OUD, maintenance of long-term sobriety, and prevention of relapse. In clinical practice, multiple medications (polypharmacy) are prescribed to manage these withdrawal symptoms, including ondansetron and promethazine for vomiting and nausea, loperamide and Lomotil for diarrhea, hydroxyzine and doxepin for pruritus, benzodiazepines, the Z-drugs, and melatonin for insomnia, and benzos, tricyclic antidepressants (TCAs), and various serotonergic agents for anxiety. This polypharmacy is associated with an increased risk of adverse drug-drug interactions and adverse drug events, increased medical costs, and increased odds of medication non-adherence and relapse. We propose an alternative single medication, mirtazapine, a noradrenergic and specific serotonergic receptor antagonist, that can be used for myriad symptoms of opioid withdrawal. Case series, clinical studies, and clinical trials have shown mirtazapine to be effective for treating nausea and vomiting resulting from multiple etiologies, including hyperemesis gravidarum and chemotherapy-induced emesis. Other evidence supports the salutary effects of mirtazapine on itching and craving. Research findings support mirtazapine's beneficial effects on diarrhea and anxiety, a consequence of its modulating effects on serotonergic receptors mediating mood and gastrointestinal symptoms. There is also evidence supporting its efficacy as a potent and non-addictive sleep aid, which presents itself as a solution for insomnia associated with opioid withdrawal. The current review presents evidence from extant literature supporting mirtazapine as a one-drug strategy to treat the variety of symptoms of opioid withdrawal. This one-drug strategy has much potential to decrease polypharmacy, adverse drug events, relapse, and healthcare cost and increase the likelihood of prolonged sobriety and better quality of life for people living with OUD.
Collapse
Affiliation(s)
- Elisha Lalani
- Department of Internal Medicine, Division of General Medicine, University of Texas Medical Branch, Galveston, USA
| | - Raakhi Menon
- Department of Internal Medicine, Division of General Medicine, University of Texas Medical Branch, Galveston, USA
| | - Mariam A Mufti
- Department of Internal Medicine, Division of Geriatrics & Palliative Medicine, University of Texas Medical Branch, Galveston, USA
| | - Cecil Kumfa
- Department of Internal Medicine, Division of Geriatrics & Palliative Medicine, University of Texas Medical Branch, Galveston, USA
| | - Mukaila Raji
- Department of Internal Medicine, Division of Geriatrics & Palliative Medicine, University of Texas Medical Branch, Galveston, USA
| |
Collapse
|
19
|
Barrell A. Current Evidence on Tobacco Harm Reduction in Pneumology: Interviews with Two Key Opinion Leaders. EUROPEAN MEDICAL JOURNAL 2022. [DOI: 10.33590/emj/10044289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The negative impact of smoking on the lung is well documented. Cigarette smoke is the cause of 90% of cases of chronic obstructive pulmonary disease (COPD), which includes chronic bronchitis and emphysema. COPD is a progressive and debilitating condition with morbidity and mortality rates similar to myocardial infarction (MI) and stroke. Despite the widely recognised risks, millions of people continue to smoke, in some cases even after receiving a diagnosis of COPD and despite knowing that the habit will accelerate disease progression. While there is no doubt that smoking cessation is the most important health intervention for all cigarette smokers, it can be challenging. Nicotine addiction, social norms and culture, and the length of time it can take smokers to feel the adverse consequences of the habit all contribute to the high cessation failure rate. However, increasing clinical and epidemiological evidence indicates that for those who are unwilling or cannot stop smoking, harm reduction strategies can help reduce exposure to the harmful chemicals and carcinogens released during tobacco combustion. Electronic devices heat processed tobacco without combusting it to deliver an aerosol containing fewer toxic products or harmful and potentially harmful constituents (HPHC) than cigarette smoke. Clinical evidence to support their use is growing. In Japan, for example, a decrease in hospitalisations due to COPD exacerbation after the introduction of such heat-not-burn (HNB), or tobacco heating systems (THS), has been reported.
In this article, Wolfgang Popp, Döbling Doctor’s Center, Privatklinik Döbling, Vienna, Austria, and Klara Szondy, Semmelweis University, Budapest, Hungary, discuss the impact of cigarette smoke on the lung, and how to support smoking cessation in those willing and able to quit smoking. They also explain when strategies that reduce the harmful effects of smoking may be useful, and outline the latest evidence supporting the use of HNB systems.
Collapse
|
20
|
Glantz S, Jeffers A, Winickoff JP. Nicotine Addiction and Intensity of e-Cigarette Use by Adolescents in the US, 2014 to 2021. JAMA Netw Open 2022; 5:e2240671. [PMID: 36342713 PMCID: PMC9641541 DOI: 10.1001/jamanetworkopen.2022.40671] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
IMPORTANCE As e-cigarettes have become more effective at delivering the addictive drug nicotine, they have become the dominant form of tobacco use by US adolescents. OBJECTIVE To measure intensity of use of e-cigarettes, cigarettes, and other tobacco products among US adolescents and their dependence level over time. DESIGN, SETTING, AND PARTICIPANTS This survey study analyzed the cross-sectional National Youth Tobacco Surveys from 2014 to 2021. Confirmatory analysis was conducted using Youth Behavioral Risk Factor Surveillance System from 2015 to 2019. The surveys were administered to national probability samples of US students in grades 6 to 12. EXPOSURES Use of e-cigarettes and other tobacco products before and after the introduction of e-cigarettes delivering high levels of nicotine. MAIN OUTCOMES AND MEASURES First tobacco product used, age at initiation of use, intensity of use (days per month), and nicotine addiction (measured as time after waking to first use of any tobacco product). RESULTS A total of 151 573 respondents were included in the analysis (51.1% male and 48.9% female; mean [SEM] age, 14.57 [0.03] years). Prevalence of e-cigarette use peaked in 2019 and then declined. Between 2014 and 2021, the age at initiation of e-cigarette use decreased, and intensity of use and addiction increased. By 2017, e-cigarettes became the most common first product used (77.0%). Age at initiation of use did not change for cigarettes or other tobacco products, and changes in intensity of use were minimal. By 2019, more e-cigarette users were using their first tobacco product within 5 minutes of waking than for cigarettes and all other products combined. Median e-cigarette use also increased from 3 to 5 d/mo in 2014 to 2018 to 6 to 9 d/mo in 2019 to 2020 and 10 to 19 d/mo in 2021. CONCLUSIONS AND RELEVANCE The changes detected in this survey study may reflect the higher levels of nicotine delivery and addiction liability of modern e-cigarettes that use protonated nicotine to make nicotine easier to inhale. The increasing intensity of use of modern e-cigarettes highlights the clinical need to address youth addiction to these new high-nicotine products over the course of many clinical encounters. In addition, stronger regulation, including comprehensive bans on the sale of flavored tobacco products, should be implemented.
Collapse
Affiliation(s)
| | - Abra Jeffers
- Division of General Academic Pediatrics, Massachusetts General Hospital for Children, Boston
- Tobacco Research and Treatment Center, Massachusetts General Hospital, Boston
| | - Jonathan P. Winickoff
- Division of General Academic Pediatrics, Massachusetts General Hospital for Children, Boston
- Tobacco Research and Treatment Center, Massachusetts General Hospital, Boston
- Julius B. Richmond Center, American Academy of Pediatrics, Itasca, Illinois
| |
Collapse
|
21
|
Lambert TP, Gazi AH, Harrison AB, Gharehbaghi S, Chan M, Obideen M, Alavi P, Murrah N, Shallenberger L, Driggers EG, Alvarado Ortega R, Washington B, Walton KM, Tang YL, Gupta R, Nye JA, Welsh JW, Vaccarino V, Shah AJ, Bremner JD, Inan OT. Leveraging Accelerometry as a Prognostic Indicator for Increase in Opioid Withdrawal Symptoms. BIOSENSORS 2022; 12:924. [PMID: 36354433 PMCID: PMC9688173 DOI: 10.3390/bios12110924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/14/2022] [Accepted: 10/21/2022] [Indexed: 06/16/2023]
Abstract
Treating opioid use disorder (OUD) is a significant healthcare challenge in the United States. Remaining abstinent from opioids is challenging for individuals with OUD due to withdrawal symptoms that include restlessness. However, to our knowledge, studies of acute withdrawal have not quantified restlessness using involuntary movements. We hypothesized that wearable accelerometry placed mid-sternum could be used to detect withdrawal-related restlessness in patients with OUD. To study this, 23 patients with OUD undergoing active withdrawal participated in a protocol involving wearable accelerometry, opioid cues to elicit craving, and non-invasive Vagal Nerve Stimulation (nVNS) to dampen withdrawal symptoms. Using accelerometry signals, we analyzed how movements correlated with changes in acute withdrawal severity, measured by the Clinical Opioid Withdrawal Scale (COWS). Our results revealed that patients demonstrating sinusoidal-i.e., predominantly single-frequency oscillation patterns in their motion almost exclusively demonstrated an increase in the COWS, and a strong relationship between the maximum power spectral density and increased withdrawal over time, measured by the COWS (R = 0.92, p = 0.029). Accelerometry may be used in an ambulatory setting to indicate the increased intensity of a patient's withdrawal symptoms, providing an objective, readily-measurable marker that may be captured ubiquitously.
Collapse
Affiliation(s)
- Tamara P. Lambert
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Asim H. Gazi
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Anna B. Harrison
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Sevda Gharehbaghi
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Michael Chan
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Malik Obideen
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Parvaneh Alavi
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Nancy Murrah
- Department of Epidemiology, Rollins School of Public Health, Atlanta, GA 30322, USA
| | - Lucy Shallenberger
- Department of Epidemiology, Rollins School of Public Health, Atlanta, GA 30322, USA
| | - Emily G. Driggers
- Department of Epidemiology, Rollins School of Public Health, Atlanta, GA 30322, USA
| | - Rebeca Alvarado Ortega
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Brianna Washington
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Kevin M. Walton
- Clinical Research Grants Branch, Division of Therapeutics and Medical Consequences, National Institute on Drug Abuse, Bethesda, MD 20877, USA
| | - Yi-Lang Tang
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- Atlanta VA Medical Center, Decatur, GA 30033, USA
| | - Rahul Gupta
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- Atlanta VA Medical Center, Decatur, GA 30033, USA
| | - Jonathon A. Nye
- Department of Radiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Justine W. Welsh
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Viola Vaccarino
- Department of Epidemiology, Rollins School of Public Health, Atlanta, GA 30322, USA
- Division of Cardiology, Department of Internal Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Amit J. Shah
- Department of Epidemiology, Rollins School of Public Health, Atlanta, GA 30322, USA
- Atlanta VA Medical Center, Decatur, GA 30033, USA
- Division of Cardiology, Department of Internal Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - J. Douglas Bremner
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- Atlanta VA Medical Center, Decatur, GA 30033, USA
- Department of Radiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Omer T. Inan
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
22
|
Fulton SL, Mitra S, Lepack AE, Martin JA, Stewart AF, Converse J, Hochstetler M, Dietz DM, Maze I. Histone H3 dopaminylation in ventral tegmental area underlies heroin-induced transcriptional and behavioral plasticity in male rats. Neuropsychopharmacology 2022; 47:1776-1783. [PMID: 35094023 PMCID: PMC9372029 DOI: 10.1038/s41386-022-01279-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 01/10/2022] [Accepted: 01/14/2022] [Indexed: 12/14/2022]
Abstract
Persistent transcriptional events in ventral tegmental area (VTA) and other reward relevant brain regions contribute to enduring behavioral adaptations that characterize substance use disorder. Recent data from our laboratory indicate that aberrant accumulation of the newly discovered histone post-translational modification (PTM), H3 dopaminylation at glutamine 5 (H3Q5dop), contributes significantly to cocaine-seeking behavior following prolonged periods of abstinence. It remained unclear, however, whether this modification is important for relapse vulnerability in the context of other drugs of abuse, such as opioids. Here, we showed that H3Q5dop plays a critical role in heroin-mediated transcriptional plasticity in midbrain regions, particularly the VTA. In rats undergoing abstinence from heroin self-administration (SA), we found acute and persistent accumulation of H3Q5dop in VTA. Attenuation of H3Q5dop during abstinence induced persistent changes in gene expression programs associated with neuronal signaling and dopaminergic function in heroin abstinence and led to reduced heroin-seeking behavior. Interestingly, the observed changes in molecular pathways after heroin SA showed significant yet reversed overlap with the same genes altered in cocaine SA. These findings establish an essential role for H3Q5dop, and its downstream transcriptional consequences, in heroin-induced functional plasticity in VTA.
Collapse
Affiliation(s)
- Sasha L. Fulton
- grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Swarup Mitra
- grid.273335.30000 0004 1936 9887Department of Pharmacology and Toxicology, Program in Neuroscience, University at Buffalo, Buffalo, NY 14214 USA
| | - Ashley E. Lepack
- grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Jennifer A. Martin
- grid.273335.30000 0004 1936 9887Department of Pharmacology and Toxicology, Program in Neuroscience, University at Buffalo, Buffalo, NY 14214 USA
| | - Andrew F. Stewart
- grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Jacob Converse
- grid.273335.30000 0004 1936 9887Department of Pharmacology and Toxicology, Program in Neuroscience, University at Buffalo, Buffalo, NY 14214 USA
| | - Mason Hochstetler
- grid.273335.30000 0004 1936 9887Department of Pharmacology and Toxicology, Program in Neuroscience, University at Buffalo, Buffalo, NY 14214 USA
| | - David M. Dietz
- grid.273335.30000 0004 1936 9887Department of Pharmacology and Toxicology, Program in Neuroscience, University at Buffalo, Buffalo, NY 14214 USA
| | - Ian Maze
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA. .,Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA. .,Howard Hughes Medical Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
23
|
Paquelet GE, Carrion K, Lacefield CO, Zhou P, Hen R, Miller BR. Single-cell activity and network properties of dorsal raphe nucleus serotonin neurons during emotionally salient behaviors. Neuron 2022; 110:2664-2679.e8. [PMID: 35700737 PMCID: PMC9575686 DOI: 10.1016/j.neuron.2022.05.015] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 11/23/2021] [Accepted: 05/13/2022] [Indexed: 12/20/2022]
Abstract
The serotonin system modulates a wide variety of emotional behaviors and states, including reward processing, anxiety, and social interaction. To reveal the underlying patterns of neural activity, we visualized serotonergic neurons in the dorsal raphe nucleus (DRN5-HT) of mice using miniaturized microscopy during diverse emotional behaviors. We discovered ensembles of cells with highly correlated activity and found that DRN5-HT neurons are preferentially recruited by emotionally salient stimuli as opposed to neutral stimuli. Individual DRN5-HT neurons responded to diverse combinations of salient stimuli, with some preference for valence and sensory modality. Anatomically defined subpopulations projecting to either a reward-related structure (the ventral tegmental area) or an anxiety-related structure (the bed nucleus of the stria terminalis) contained all response types but were enriched in reward- and anxiety-responsive cells, respectively. Our results suggest that the DRN serotonin system responds to emotional salience using ensembles with mixed selectivity and biases in downstream connectivity.
Collapse
Affiliation(s)
- Grace E Paquelet
- Department of Neuroscience, Columbia University, New York, NY 10032, USA
| | - Kassandra Carrion
- Division of Systems Neuroscience, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Clay O Lacefield
- Division of Systems Neuroscience, New York State Psychiatric Institute, New York, NY 10032, USA; Department of Psychiatry, Columbia University Medical Center, New York, NY 10032, USA
| | - Pengcheng Zhou
- Department of Neuroscience, Columbia University, New York, NY 10032, USA; Department of Statistics, Columbia University, New York, NY 10027, USA; Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027
| | - René Hen
- Department of Neuroscience, Columbia University, New York, NY 10032, USA; Division of Systems Neuroscience, New York State Psychiatric Institute, New York, NY 10032, USA; Department of Psychiatry, Columbia University Medical Center, New York, NY 10032, USA
| | - Bradley R Miller
- Division of Systems Neuroscience, New York State Psychiatric Institute, New York, NY 10032, USA; Department of Psychiatry, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
24
|
Altered Accumbal Dopamine Terminal Dynamics Following Chronic Heroin Self-Administration. Int J Mol Sci 2022; 23:ijms23158106. [PMID: 35897682 PMCID: PMC9332320 DOI: 10.3390/ijms23158106] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/19/2022] [Accepted: 07/19/2022] [Indexed: 11/30/2022] Open
Abstract
Administration of heroin results in the engagement of multiple brain regions and the rewarding and addictive effects are mediated, at least partially, through activation of the mesolimbic dopamine system. However, less is known about dopamine system function following chronic exposure to heroin. Withdrawal from chronic heroin exposure is likely to drive a state of low dopamine in the nucleus accumbens (NAc), as previously observed during withdrawal from other drug classes. Thus, we aimed to investigate alterations in NAc dopamine terminal function following chronic heroin self-administration to identify a mechanism for dopaminergic adaptations. Adult male Long Evans rats were trained to self-administer heroin (0.05 mg/kg/inf, IV) and then placed on a long access (FR1, 6-h, unlimited inf, 0.05 mg/kg/inf) protocol to induce escalation of intake. Following heroin self-administration, rats had decreased basal extracellular levels of dopamine and blunted dopamine response following a heroin challenge (0.1 mg/kg/inf, IV) in the NAc compared to saline controls. FSCV revealed that heroin-exposed rats exhibited reduced stimulated dopamine release during tonic-like, single-pulse stimulations, but increased phasic-like dopamine release during multi-pulse stimulation trains (5 pulses, 5–100 Hz) in addition to an altered dynamic range of release stimulation intensities when compared to controls. Further, we found that presynaptic D3 autoreceptor and kappa-opioid receptor agonist responsivity were increased following heroin self-administration. These results reveal a marked low dopamine state following heroin exposure and suggest the combination of altered dopamine release dynamics may contribute to increased heroin seeking.
Collapse
|
25
|
Hao Y, Liu W, Xu Z, Jin X, Ye Y, Yu C, Hu C, Zuo H, Li Y. High-Power Electromagnetic Pulse Exposure of Healthy Mice: Assessment of Effects on Mice Cognitions, Neuronal Activities, and Hippocampal Structures. Front Cell Neurosci 2022; 16:898164. [PMID: 35966202 PMCID: PMC9374008 DOI: 10.3389/fncel.2022.898164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/30/2022] [Indexed: 11/17/2022] Open
Abstract
Electromagnetic pulse (EMP) is a high-energy pulse with an extremely rapid rise time and a broad bandwidth. The brain is a target organ sensitive to electromagnetic radiation (EMR), the biological effects and related mechanisms of EMPs on the brain remain unclear. The objectives of the study were to assess the effects of EMP exposure on mouse cognitions, and the neuronal calcium activities in vivo under different cases of real-time exposure and post exposure. EMP-treated animal model was established by exposing male adult C57BL/6N mice to 300 kV/m EMPs. First, the effects of EMPs on the cognitions, including the spatial learning and memory, avoidance learning and memory, novelty-seeking behavior, and anxiety, were assessed by multiple behavioral experiments. Then, the changes in the neuronal activities of the hippocampal CA1 area in vivo were detected by fiber photometry in both cases of during real-time EMP radiation and post-exposure. Finally, the structures of neurons in hippocampi were observed by optical microscope and transmission electron microscope. We found that EMPs under this condition caused a decline in the spatial learning and memory ability in mice, but no effects on the avoidance learning and memory, novelty-seeking behavior, and anxiety. The neuron activities of hippocampal CA1 were disturbed by EMP exposure, which were inhibited during EMP exposure, but activated immediately after exposure end. Additionally, the CA1 neuron activities, when mice entered the central area in an Open field (OF) test or explored the novelty in a Novel object exploration (NOE) test, were inhibited on day 1 and day 7 after radiation. Besides, damaged structures in hippocampal neurons were observed after EMP radiation. In conclusion, EMP radiation impaired the spatial learning and memory ability and disturbed the neuronal activities in hippocampal CA1 in mice.
Collapse
Affiliation(s)
- Yanhui Hao
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Weiqi Liu
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, China
- Life Science Department, Foshan University, Foshan, China
| | - Zhengtao Xu
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, China
- Life Science Department, Foshan University, Foshan, China
| | - Xing Jin
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yumeng Ye
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Chao Yu
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Cuicui Hu
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, China
- Academy of Life Sciences, Anhui Medical University, Hefei, China
| | - Hongyan Zuo
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, China
- *Correspondence: Yang Li ; Hongyan Zuo
| | - Yang Li
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, China
- Academy of Life Sciences, Anhui Medical University, Hefei, China
- *Correspondence: Yang Li ; Hongyan Zuo
| |
Collapse
|
26
|
Sharp JL, Smith MA. The Effects of Drugs on Behavior Maintained by Social Contact: Role of Monoamines in Social Reinforcement. Front Behav Neurosci 2022; 15:805139. [PMID: 35264935 PMCID: PMC8899311 DOI: 10.3389/fnbeh.2021.805139] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/27/2021] [Indexed: 11/13/2022] Open
Abstract
Drug use is highly concordant among members of adolescent and young adult peer groups. One potential explanation for this observation is that drugs may increase the reinforcing effects of social contact, leading to greater motivation to establish and maintain contact with other members of the peer group. Several classes of drugs, particularly drugs that increase synaptic dopamine, increase the reinforcing effects of contextual stimuli, but the extent to which these drugs enhance the reinforcing effects of social contact is not known. The purpose of this study was to determine the extent to which drugs that increase synaptic dopamine, norepinephrine, and serotonin enhance the positive reinforcing effects of social contact. To this end, male and female Long-Evans rats were pretreated with acute doses of the selective dopamine reuptake inhibitor, WIN-35,428, the selective norepinephrine reuptake inhibitor, atomoxetine, the selective serotonin reuptake inhibitor, fluoxetine, the non-selective monoamine reuptake inhibitor, cocaine, and the non-selective monoamine releasers d-amphetamine and (±)-MDMA. Ten minutes later, the positive reinforcing effects of 30-s access to a same-sex social partner was examined on a progressive ratio schedule of reinforcement. To determine whether the reinforcement-altering effects of these drugs were specific to the social stimulus, the reinforcing effects of a non-social stimulus (30-s access to an athletic sock of similar size and coloring as another rat) was determined in control subjects. WIN-35,428, d-amphetamine, and cocaine, but not atomoxetine, fluoxetine, or MDMA, dose-dependently increased breakpoints maintained by a social partner under conditions in which responding maintained by a non-social stimulus was not affected. These data indicate that increases in extracellular dopamine, but not extracellular norepinephrine or serotonin, increases the positive reinforcing effects of social contact in both male and female rats. These data also provide support for the hypothesis that some drugs with high abuse liability increase the motivation to establish and maintain contact with social peers.
Collapse
|
27
|
Kermanian F, Seghatoleslam M, Mahakizadeh S. MDMA related neuro-inflammation and adenosine receptors. Neurochem Int 2022; 153:105275. [PMID: 34990730 DOI: 10.1016/j.neuint.2021.105275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 12/25/2021] [Accepted: 12/30/2021] [Indexed: 12/20/2022]
Abstract
3,4-methylenedioxymethamphetamine (MDMA) is a world-wide abused psychostimulant, which has the neurotoxic effects on dopaminergic and serotonergic neurons in both rodents and non-human primates. Adenosine acts as a neurotransmitter in the brain through the activation of four specific G-protein-coupled receptors and it acts as a neuromodulator of dopamine neurotransmission. Recent studies suggest that stimulation of adenosine receptors oppose many behavioral effects of methamphetamines. This review summarizes the specific cellular mechanisms involved in MDMA neuroinflammatory effects, along with the protective effects of adenosine receptors.
Collapse
Affiliation(s)
- Fatemeh Kermanian
- Department of Anatomy, Faculty of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Masoumeh Seghatoleslam
- Evaluative Clinical Sciences, Sunnybrook Research Institute, University of Toronto, ON, Canada
| | - Simin Mahakizadeh
- Department of Anatomy, Faculty of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
28
|
Dorsal Raphe Nucleus Serotoninergic Neurons Mediate Morphine Rewarding Effect and Conditioned Place Preference. Neuroscience 2022; 480:108-116. [PMID: 34762982 DOI: 10.1016/j.neuroscience.2021.10.031] [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: 06/24/2021] [Revised: 10/25/2021] [Accepted: 10/30/2021] [Indexed: 11/21/2022]
Abstract
Morphine rewarding properties are the main reasons for drug-craving in behaviors occurring during morphine addiction. It has been suggested that morphine addiction relies on signals to the mesolimbic dopamine system, although the mechanisms outside the dopaminergic system are still unclear. Notably, the role of the dorsal raphe nucleus (DRN) serotoninergic (5-hydroxytryptamine, 5-HT) system remains unexplored. Using in vivo electrophysiological and optogenetic approaches, we found that morphine treatment increased DRN 5-TH neurons firing rate and optogenetic activation of DRN 5-HT neurons induced a rewarding effect, indicating that morphine reward is related to DRN 5-HT neurons. Accordingly, optogenetic inhibition of DRN 5-HT neurons following morphine injection reversed conditioned place preference (CPP) during chronic morphine treatment. These findings aid our understanding of the new functions of the DRN 5-HT neurons for morphine rewarding effect and provide a potential approach for the treatment of morphine addiction.
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
|
S S, Nair AJS, Sandhya KY. Highly Stable Copper Nano Cluster on Nitrogen-Doped Graphene Quantum Dots for the Simultaneous Electrochemical Sensing of Dopamine, Serotonin, and Nicotine; a Possible Addiction Scrutinizing Strategy. J Mater Chem B 2022; 10:3974-3988. [DOI: 10.1039/d1tb02368c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A highly stable copper nanocluster CuNC@N-GQD which exhibited stability for more than one year was synthesized using nitrogen doped graphene quantum dots (N-GQDs) as reducing and capping agents and smaller...
Collapse
|
31
|
|
32
|
van Zessen R, Li Y, Marion-Poll L, Hulo N, Flakowski J, Lüscher C. Dynamic dichotomy of accumbal population activity underlies cocaine sensitization. eLife 2021; 10:e66048. [PMID: 34608866 PMCID: PMC8523149 DOI: 10.7554/elife.66048] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 09/28/2021] [Indexed: 11/28/2022] Open
Abstract
Locomotor sensitization (LS) is an early behavioral adaptation to addictive drugs, driven by the increase of dopamine in the Nucleus Accumbens (NAc). However, the effect on accumbal population activity remains elusive. Here, we used single-cell calcium imaging in mice to record the activity of dopamine-1-receptor (D1R) and dopamine-2-receptor (D2R) expressing spiny projection neurons (SPNs) during cocaine LS. Acute exposure to cocaine elevated D1R SPN activity and reduced D2R SPN activity, albeit with high variability between neurons. During LS, the number of D1R and D2R neurons responding in opposite directions increased. Moreover, preventing LS by inhibition of the ERK signaling pathway decreased the number of cocaine responsive D1R SPNs, but had little effect on D2R SPNs. These results indicate that accumbal population dichotomy is dynamic and contains a subgroup of D1R SPNs that eventually drives LS. Insights into the drug-related activity dynamics provides a foundation for understanding the circuit-level addiction pathogenesis.
Collapse
Affiliation(s)
- Ruud van Zessen
- Department of Basic Neurosciences, Faculty of Medicine, University of GenevaGenevaSwitzerland
| | - Yue Li
- Department of Basic Neurosciences, Faculty of Medicine, University of GenevaGenevaSwitzerland
| | - Lucile Marion-Poll
- Department of Basic Neurosciences, Faculty of Medicine, University of GenevaGenevaSwitzerland
| | - Nicolas Hulo
- Institute of Genetics and Genomics of Geneva (IGE3), University of GenevaGenevaSwitzerland
| | - Jérôme Flakowski
- Department of Basic Neurosciences, Faculty of Medicine, University of GenevaGenevaSwitzerland
| | - Christian Lüscher
- Department of Basic Neurosciences, Faculty of Medicine, University of GenevaGenevaSwitzerland
- Clinic of Neurology, Dept. of Clinical Neurosciences, Geneva University HospitalGenevaSwitzerland
| |
Collapse
|
33
|
George BE, Barth SH, Kuiper LB, Holleran KM, Lacy RT, Raab-Graham KF, Jones SR. Enhanced heroin self-administration and distinct dopamine adaptations in female rats. Neuropsychopharmacology 2021; 46:1724-1733. [PMID: 34040157 PMCID: PMC8358024 DOI: 10.1038/s41386-021-01035-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 05/03/2021] [Accepted: 05/07/2021] [Indexed: 02/04/2023]
Abstract
Increasing evidence suggests that females are more vulnerable to the harmful effects of drugs of abuse, including opioids. Additionally, rates of heroin-related deaths substantially increased in females from 1999 to 2017 [1], underscoring the need to evaluate sex differences in heroin vulnerability. Moreover, the neurobiological substrates underlying sexually dimorphic responding to heroin are not fully defined. Thus, we evaluated male and female Long Evans rats on acquisition, dose-responsiveness, and seeking for heroin self-administration (SA) as well as using a long access model to assess escalation of intake at low and high doses of heroin, 0.025 and 0.1 mg/kg/inf, respectively. We paired this with ex vivo fast-scan cyclic voltammetry (FSCV) in the medial nucleus accumbens (NAc) shell and quantification of mu-opioid receptor (MOR) protein in the ventral tegmental area (VTA) and NAc. While males and females had similar heroin SA acquisition rates, females displayed increased responding and intake across doses, seeking for heroin, and escalation on long access. However, we found that males and females had similar expression levels of MORs in the VTA and NAc, regardless of heroin exposure. FSCV results revealed that heroin exposure did not change single-pulse elicited dopamine release, but caused an increase in dopamine transporter activity in both males and females compared to their naïve counterparts. Phasic-like stimulations elicited robust increases in dopamine release in heroin-exposed females compared to heroin-naïve females, with no differences seen in males. Together, our results suggest that differential adaptations of dopamine terminals may underlie the increased heroin SA behaviors seen in females.
Collapse
Affiliation(s)
- Brianna E. George
- grid.241167.70000 0001 2185 3318Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC USA
| | - Samuel H. Barth
- grid.241167.70000 0001 2185 3318Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC USA
| | - Lindsey B. Kuiper
- grid.241167.70000 0001 2185 3318Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC USA
| | - Katherine M. Holleran
- grid.241167.70000 0001 2185 3318Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC USA
| | - Ryan T. Lacy
- grid.256069.eDepartment of Psychology, Franklin and Marshall College, Lancaster, PA USA
| | - Kimberly F. Raab-Graham
- grid.241167.70000 0001 2185 3318Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC USA
| | - Sara R. Jones
- grid.241167.70000 0001 2185 3318Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC USA
| |
Collapse
|
34
|
Dagra A, Miller DR, Lin M, Gopinath A, Shaerzadeh F, Harris S, Sorrentino ZA, Støier JF, Velasco S, Azar J, Alonge AR, Lebowitz JJ, Ulm B, Bu M, Hansen CA, Urs N, Giasson BI, Khoshbouei H. α-Synuclein-induced dysregulation of neuronal activity contributes to murine dopamine neuron vulnerability. NPJ Parkinsons Dis 2021; 7:76. [PMID: 34408150 PMCID: PMC8373893 DOI: 10.1038/s41531-021-00210-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 07/09/2021] [Indexed: 02/07/2023] Open
Abstract
Pathophysiological damages and loss of function of dopamine neurons precede their demise and contribute to the early phases of Parkinson's disease. The presence of aberrant intracellular pathological inclusions of the protein α-synuclein within ventral midbrain dopaminergic neurons is one of the cardinal features of Parkinson's disease. We employed molecular biology, electrophysiology, and live-cell imaging to investigate how excessive α-synuclein expression alters multiple characteristics of dopaminergic neuronal dynamics and dopamine transmission in cultured dopamine neurons conditionally expressing GCaMP6f. We found that overexpression of α-synuclein in mouse (male and female) dopaminergic neurons altered neuronal firing properties, calcium dynamics, dopamine release, protein expression, and morphology. Moreover, prolonged exposure to the D2 receptor agonist, quinpirole, rescues many of the alterations induced by α-synuclein overexpression. These studies demonstrate that α-synuclein dysregulation of neuronal activity contributes to the vulnerability of dopaminergic neurons and that modulation of D2 receptor activity can ameliorate the pathophysiology. These findings provide mechanistic insights into the insidious changes in dopaminergic neuronal activity and neuronal loss that characterize Parkinson's disease progression with significant therapeutic implications.
Collapse
Affiliation(s)
- Abeer Dagra
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Douglas R. Miller
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Min Lin
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Adithya Gopinath
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Fatemeh Shaerzadeh
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Sharonda Harris
- grid.15276.370000 0004 1936 8091Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL USA
| | - Zachary A. Sorrentino
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Jonatan Fullerton Støier
- grid.5254.60000 0001 0674 042XMolecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sophia Velasco
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Janelle Azar
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Adetola R. Alonge
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Joseph J. Lebowitz
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Brittany Ulm
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Mengfei Bu
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Carissa A. Hansen
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Nikhil Urs
- grid.15276.370000 0004 1936 8091Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL USA
| | - Benoit I. Giasson
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| | - Habibeh Khoshbouei
- grid.15276.370000 0004 1936 8091Department of Neuroscience, University of Florida, Gainesville, FL USA
| |
Collapse
|
35
|
Abstract
This paper is the forty-second consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2019 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides and receptors as well as effects of opioid/opiate agonists and antagonists. The review is subdivided into the following specific topics: molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors (1), the roles of these opioid peptides and receptors in pain and analgesia in animals (2) and humans (3), opioid-sensitive and opioid-insensitive effects of nonopioid analgesics (4), opioid peptide and receptor involvement in tolerance and dependence (5), stress and social status (6), learning and memory (7), eating and drinking (8), drug abuse and alcohol (9), sexual activity and hormones, pregnancy, development and endocrinology (10), mental illness and mood (11), seizures and neurologic disorders (12), electrical-related activity and neurophysiology (13), general activity and locomotion (14), gastrointestinal, renal and hepatic functions (15), cardiovascular responses (16), respiration and thermoregulation (17), and immunological responses (18).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, 65-30 Kissena Blvd., Flushing, NY, 11367, United States.
| |
Collapse
|
36
|
Lin R, Liang J, Luo M. The Raphe Dopamine System: Roles in Salience Encoding, Memory Expression, and Addiction. Trends Neurosci 2021; 44:366-377. [PMID: 33568331 DOI: 10.1016/j.tins.2021.01.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/21/2020] [Accepted: 01/13/2021] [Indexed: 02/06/2023]
Abstract
Dopamine (DA) neurons of the dorsal raphe nucleus (DRN) were traditionally viewed as an extension of the ventral tegmental area (VTA) DA population. While the VTA DA population is known to play important roles in reward processing, emerging evidence now supports the view that DRN DA neurons are a specialized midbrain DA subsystem that performs distinct functions in parallel to the VTA DA population. Recent studies have shed new light on the roles of DRN DA neurons in encoding incentive salience and in regulating memory expression and arousal. Here, we review recent findings using mouse models about the physiology and behavioral functions of DRN DA neurons, highlight the engagement of DRN DA neurons and their upstream circuits in opioid addiction, and discuss emerging lines of investigation that reveal multifaceted heterogeneity among DRN DA neurons.
Collapse
Affiliation(s)
- Rui Lin
- National Institute of Biological Sciences (NIBS), Beijing 102206, China.
| | - Jingwen Liang
- National Institute of Biological Sciences (NIBS), Beijing 102206, China
| | - Minmin Luo
- National Institute of Biological Sciences (NIBS), Beijing 102206, China; Chinese Institute for Brain Research, Beijing 102206, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
37
|
Choi MR, Jin YB, Kim HN, Chai YG, Im CN, Lee SR, Kim DJ. Gene expression in the striatum of cynomolgus monkeys after chronic administration of cocaine and heroin. Basic Clin Pharmacol Toxicol 2021; 128:686-698. [PMID: 33404192 DOI: 10.1111/bcpt.13554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 12/26/2020] [Accepted: 01/01/2021] [Indexed: 01/16/2023]
Abstract
Cocaine and heroin cause impairment of neural plasticity in the brain including striatum. This study aimed to identify genes differentially expressed in the striatum of cynomolgus monkeys in response to cocaine and heroin. After chronic administration of cocaine and heroin in the monkeys, we performed large-scale transcriptome profiling in the striatum using RNA-Seq technology and analysed functional annotation. We found that 547 and 1238 transcripts were more than 1.5-fold up- or down-regulated in cocaine- and heroin-treated groups, respectively, compared to the control group, and 3432 transcripts exhibited differential expression between cocaine- and heroin-treated groups. Functional annotation analysis indicated that genes associated with nervous system development (NAGLU, MOBP and TTL7) and stress granule disassembly (KIF5B and KLC1) were differentially expressed in the cocaine-treated group compared to the control group, whereas gene associated with neuron apoptotic process (ERBB3) was differentially expressed in the heroin-treated group. In addition, IPA network analysis indicated that genes (TRAF6 and TRAF3IP2) associated with inflammation were increased by the chronic administration of cocaine and heroin. These results provide insight into the correlated molecular mechanisms as well as the upregulation and down-regulation of genes in the striatum after chronic exposure to cocaine and heroin.
Collapse
Affiliation(s)
- Mi Ran Choi
- Department of Psychiatry, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Republic of Korea
| | - Yeung-Bae Jin
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Han-Na Kim
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Republic of Korea
| | - Young Gyu Chai
- Department of Molecular and Life Sciences, Hanyang University, Ansan, Republic of Korea
| | - Chang-Nim Im
- Department of Psychiatry, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Republic of Korea
| | - Sang-Rae Lee
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Republic of Korea.,Department of Functional Genomics, University of Science and Technology, Daejeon, Republic of Korea
| | - Dai-Jin Kim
- Department of Psychiatry, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
38
|
Kim CK, Sanchez MI, Hoerbelt P, Fenno LE, Malenka RC, Deisseroth K, Ting AY. A Molecular Calcium Integrator Reveals a Striatal Cell Type Driving Aversion. Cell 2020; 183:2003-2019.e16. [PMID: 33308478 PMCID: PMC9839359 DOI: 10.1016/j.cell.2020.11.015] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 09/18/2020] [Accepted: 11/06/2020] [Indexed: 01/17/2023]
Abstract
The ability to record transient cellular events in the DNA or RNA of cells would enable precise, large-scale analysis, selection, and reprogramming of heterogeneous cell populations. Here, we report a molecular technology for stable genetic tagging of cells that exhibit activity-related increases in intracellular calcium concentration (FLiCRE). We used FLiCRE to transcriptionally label activated neural ensembles in the nucleus accumbens of the mouse brain during brief stimulation of aversive inputs. Using single-cell RNA sequencing, we detected FLiCRE transcripts among the endogenous transcriptome, providing simultaneous readout of both cell-type and calcium activation history. We identified a cell type in the nucleus accumbens activated downstream of long-range excitatory projections. Taking advantage of FLiCRE's modular design, we expressed an optogenetic channel selectively in this cell type and showed that direct recruitment of this otherwise genetically inaccessible population elicits behavioral aversion. The specificity and minute resolution of FLiCRE enables molecularly informed characterization, manipulation, and reprogramming of activated cellular ensembles.
Collapse
Affiliation(s)
- Christina K Kim
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Mateo I Sanchez
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Paul Hoerbelt
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Lief E Fenno
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94035, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Robert C Malenka
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Karl Deisseroth
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94035, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA.
| | - Alice Y Ting
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA; Department of Biology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
39
|
Li H, Liu Y, Xing L, Yang X, Xu J, Ren Q, Su KP, Lu Y, Wang F. Association of Cigarette Smoking with Sleep Disturbance and Neurotransmitters in Cerebrospinal Fluid. Nat Sci Sleep 2020; 12:801-808. [PMID: 33122957 PMCID: PMC7591043 DOI: 10.2147/nss.s272883] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/14/2020] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Cigarette smoking has shown to be associated with sleep disturbance, especially prolonged sleep onset latency (SOL). Cigarette smoking stimulates the release of dopamine (DA) and serotonin (5-HT), which might promote awakening and inhibit rapid eye movement sleep. Dopamine transporter (DAT) and serotonin transporter play a key role in the reuptake of DA and 5-HT from the synaptic cleft into presynaptic neurons. However, the relationship among cigarette smoking, sleep disturbance and neurotransmitters has never been investigated in human cerebrospinal fluid (CSF). METHODS A total of 159 Chinese male subjects (81 active smokers and 78 non-smokers) who would undergo lumbar puncture before the surgery of anterior cruciate ligament reconstruction were recruited and 5mL-CSF samples were collected incidentally. CSF levels of DA, DAT, 5-HT, and serotonin transporter were measured using radioimmunoassay and ELISA. Sociodemographic data and the Pittsburgh Sleep Quality Index (PSQI) scale were collected before surgery. RESULTS PSQI global scores, SOL, and CSF DA levels were significantly higher in active smokers compared to non-smokers (2.00 [1.00-4.75] scores vs 4.00 [3.00-6.00] scores, p = 0.001; 10.00 [5.00-15.00] minutes vs 15.00 [10.00-30.00] minutes, p = 0.002; 87.20 [82.31-96.06]ng/mL vs 107.45 [92.78-114.38] ng/mL, p < 0.001), while CSF DAT levels were significantly lower in active smokers (0.35 [0.31-0.39] ng/mL vs 0.29 [0.26-0.34] ng/mL, p < 0.001). CONCLUSION Cigarette smoking was indeed associated with sleep disturbance, shown by prolonged SOL, higher DA levels and lower DAT levels in CSF of active smokers.
Collapse
Affiliation(s)
- Hui Li
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing100871, People’s Republic of China
- Xinjiang Key Laboratory of Neurological Disorder Research, The Second Affiliated Hospital, Xinjiang Medical University, Urumqi830063, People’s Republic of China
- Psychosomatic Medicine Research Division, Inner Mongolia Medical University, Huhhot010110, People’s Republic of China
| | - Yanlong Liu
- Zhuji Institute of Biomedicine, School of Pharmaceutical Sciences, Wenzhou Medical University, Shaoxing311800, People’s Republic of China
- School of Mental Health, Wenzhou Medical University, Wenzhou, 325035, People’s Republic of China
- The Affiliated Kangning Hospital, Wenzhou Medical University, Wenzhou325035, People’s Republic of China
| | - Lifei Xing
- Department of Neurology, Inner Mongolia North Heavy Industries Group Corp. Ltd Hospital, Baotou014030, People’s Republic of China
| | - Xiaoyu Yang
- Beijing Jishuitan Hospital, Beijing100035, People’s Republic of China
| | - Jinzhong Xu
- The Affiliated Wenling Hospital of Wenzhou Medical University, Wenling317500, People’s Republic of China
| | - Qiushi Ren
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing100871, People’s Republic of China
| | - Kuan-Pin Su
- Zhuji Institute of Biomedicine, School of Pharmaceutical Sciences, Wenzhou Medical University, Shaoxing311800, People’s Republic of China
- An-Nan Hospital, China Medical University, Tainan, Taiwan
- College of Medicine, China Medical University, Taichung, Taiwan
| | - Yanye Lu
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing100871, People’s Republic of China
| | - Fan Wang
- Xinjiang Key Laboratory of Neurological Disorder Research, The Second Affiliated Hospital, Xinjiang Medical University, Urumqi830063, People’s Republic of China
- Psychosomatic Medicine Research Division, Inner Mongolia Medical University, Huhhot010110, People’s Republic of China
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing100096, People’s Republic of China
| |
Collapse
|
40
|
Abstract
The brain serotonin systems participate in numerous aspects of reward processing, although it remains elusive how exactly serotonin signals regulate neural computation and reward-related behavior. The application of optogenetics and imaging techniques during the last decade has provided many insights. Here, we review recent progress on the organization and physiology of the dorsal raphe serotonin neurons and the relationships between their activity and behavioral functions in the context of reward processing. We also discuss several interesting theories on serotonin's function and how these theories may be reconciled by the possibility that serotonin, acting in synergy with coreleased glutamate, tracks and calculates the so-called beneficialness of the current state to guide an animal's behavior in dynamic environments.
Collapse
Affiliation(s)
- Zhixiang Liu
- National Institute of Biological Sciences, Beijing 102206, China
| | - Rui Lin
- National Institute of Biological Sciences, Beijing 102206, China
| | - Minmin Luo
- National Institute of Biological Sciences, Beijing 102206, China
- School of Life Sciences, Tsinghua University, Beijing 100081, China
- Chinese Institute for Brain Research, Beijing 102206, China
| |
Collapse
|
41
|
Lin R, Liang J, Wang R, Yan T, Zhou Y, Liu Y, Feng Q, Sun F, Li Y, Li A, Gong H, Luo M. The Raphe Dopamine System Controls the Expression of Incentive Memory. Neuron 2020; 106:498-514.e8. [PMID: 32145184 DOI: 10.1016/j.neuron.2020.02.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/03/2020] [Accepted: 02/10/2020] [Indexed: 02/07/2023]
Abstract
The brain dopamine (DA) system participates in forming and expressing memory. Despite a well-established role of DA neurons in the ventral tegmental area in memory formation, the exact DA circuits that control memory expression remain unclear. Here, we show that DA neurons in the dorsal raphe nucleus (DRN) and their medulla input control the expression of incentive memory. DRN DA neurons are activated by both rewarding and aversive stimuli in a learning-dependent manner and exhibit elevated activity during memory recall. Disrupting their physiological activity or DA synthesis blocks the expression of natural appetitive and aversive memories as well as drug memories associated with opioids. Moreover, a glutamatergic pathway from the lateral parabrachial nucleus to the DRN selectively regulates the expression of reward memories associated with opioids or foods. Our study reveals a specialized DA subsystem important for memory expression and suggests new targets for interventions against opioid addiction.
Collapse
Affiliation(s)
- Rui Lin
- National Institute of Biological Sciences (NIBS), Beijing 102206, China.
| | - Jingwen Liang
- National Institute of Biological Sciences (NIBS), Beijing 102206, China; Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Ruiyu Wang
- National Institute of Biological Sciences (NIBS), Beijing 102206, China; School of Life Sciences, Peking University, Beijing 100871, China
| | - Ting Yan
- National Institute of Biological Sciences (NIBS), Beijing 102206, China; School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Youtong Zhou
- National Institute of Biological Sciences (NIBS), Beijing 102206, China; Chinese Institute for Brain Research, Beijing 102206, China
| | - Yang Liu
- National Institute of Biological Sciences (NIBS), Beijing 102206, China; School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Qiru Feng
- National Institute of Biological Sciences (NIBS), Beijing 102206, China; School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Fangmiao Sun
- School of Life Sciences, Peking University, Beijing 100871, China
| | - Yulong Li
- School of Life Sciences, Peking University, Beijing 100871, China
| | - Anan Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China; MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China; HUST-Suzhou Institute for Brainsmatics, JITRI Institute for Brainsmatics, Suzhou 215100, China
| | - Hui Gong
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China; MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan 430074, China; HUST-Suzhou Institute for Brainsmatics, JITRI Institute for Brainsmatics, Suzhou 215100, China
| | - Minmin Luo
- National Institute of Biological Sciences (NIBS), Beijing 102206, China; Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China; School of Life Sciences, Tsinghua University, Beijing 100084, China; Chinese Institute for Brain Research, Beijing 102206, China.
| |
Collapse
|
42
|
McGrath-Morrow SA, Gorzkowski J, Groner JA, Rule AM, Wilson K, Tanski SE, Collaco JM, Klein JD. The Effects of Nicotine on Development. Pediatrics 2020; 145:peds.2019-1346. [PMID: 32047098 PMCID: PMC7049940 DOI: 10.1542/peds.2019-1346] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/05/2019] [Indexed: 01/08/2023] Open
Abstract
Recently, there has been a significant increase in the use of noncombustible nicotine-containing products, including electronic cigarettes (e-cigarettes). Of increasing popularity are e-cigarettes that can deliver high doses of nicotine over short periods of time. These devices have led to a rise in nicotine addiction in adolescent users who were nonsmokers. Use of noncombustible nicotine products by pregnant mothers is also increasing and can expose the developing fetus to nicotine, a known teratogen. In addition, young children are frequently exposed to secondhand and thirdhand nicotine aerosols generated by e-cigarettes, with little understanding of the effects these exposures can have on health. With the advent of these new nicotine-delivery systems, many concerns have arisen regarding the short- and long-term health effects of nicotine on childhood health during all stages of development. Although health studies on nicotine exposure alone are limited, educating policy makers and health care providers on the potential health effects of noncombustible nicotine is needed because public acceptance of these products has become so widespread. Most studies evaluating the effects of nicotine on health have been undertaken in the context of smoke exposure. Nevertheless, in vitro and in vivo preclinical studies strongly indicate that nicotine exposure alone can adversely affect the nervous, respiratory, immune, and cardiovascular systems, particularly when exposure occurs during critical developmental periods. In this review, we have included both preclinical and clinical studies to identify age-related health effects of nicotine exposure alone, examining the mechanisms underlying these effects.
Collapse
Affiliation(s)
- Sharon A. McGrath-Morrow
- Julius B. Richmond Center of Excellence, American Academy of Pediatrics, Itasca, Illinois;,Eudowood Division of Pediatric Respiratory Sciences, Department of Pediatrics, School of Medicine and
| | - Julie Gorzkowski
- Julius B. Richmond Center of Excellence, American Academy of Pediatrics, Itasca, Illinois
| | - Judith A. Groner
- Julius B. Richmond Center of Excellence, American Academy of Pediatrics, Itasca, Illinois;,Department of Pediatrics, Nationwide Children’s Hospital, Columbus, Ohio
| | - Ana M. Rule
- Julius B. Richmond Center of Excellence, American Academy of Pediatrics, Itasca, Illinois;,Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Karen Wilson
- Julius B. Richmond Center of Excellence, American Academy of Pediatrics, Itasca, Illinois;,Department of Pediatrics, Icahn School of Medicine at Mount Sinai and Kravis Children’s Hospital, New York, New York
| | - Susanne E. Tanski
- Julius B. Richmond Center of Excellence, American Academy of Pediatrics, Itasca, Illinois;,Department of Pediatrics, Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire; and
| | - Joseph M. Collaco
- Julius B. Richmond Center of Excellence, American Academy of Pediatrics, Itasca, Illinois;,Eudowood Division of Pediatric Respiratory Sciences, Department of Pediatrics, School of Medicine and
| | - Jonathan D. Klein
- Julius B. Richmond Center of Excellence, American Academy of Pediatrics, Itasca, Illinois;,Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
43
|
Vassilev P, Avvisati R, Koya E, Badiani A. Distinct Populations of Neurons Activated by Heroin and Cocaine in the Striatum as Assessed by catFISH. eNeuro 2020; 7:ENEURO.0394-19.2019. [PMID: 31937522 PMCID: PMC7005257 DOI: 10.1523/eneuro.0394-19.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/30/2019] [Accepted: 12/19/2019] [Indexed: 12/13/2022] Open
Abstract
Despite the still prevailing notion of a shared substrate of action for all addictive drugs, there is evidence suggesting that opioid and psychostimulant drugs differ substantially in terms of their neurobiological and behavioral effects. These differences may reflect separate neural circuits engaged by the two drugs. Here we used the catFISH (cellular compartment analysis of temporal activity by fluorescence in situ hybridization) technique to investigate the degree of overlap between neurons engaged by heroin versus cocaine in adult male Sprague Dawley rats. The catFISH technique is a within-subject procedure that takes advantage of the different transcriptional time course of the immediate-early genes homer 1a and arc to determine to what extent two stimuli separated by an interval of 25 min engage the same neuronal population. We found that throughout the striatal complex the neuronal populations activated by noncontingent intravenous injections of cocaine (800 μg/kg) and heroin (100 and 200 μg/kg), administered at an interval of 25 min from each other, overlapped to a much lesser extent than in the case of two injections of cocaine (800 μg/kg), also 25 min apart. The greatest reduction in overlap between populations activated by cocaine and heroin was in the dorsomedial and dorsolateral striatum (∼30% and ∼22%, respectively, of the overlap observed for the sequence cocaine-cocaine). Our results point toward a significant separation between neuronal populations activated by heroin and cocaine in the striatal complex. We propose that our findings are a proof of concept that these two drugs are encoded differently in a brain area believed to be a common neurobiological substrate to drug abuse.
Collapse
Affiliation(s)
- Philip Vassilev
- Sussex Addiction Research and Intervention Centre (SARIC), School of Psychology, University of Sussex, Brighton BN1 9RH, United Kingdom
| | - Riccardo Avvisati
- Sussex Addiction Research and Intervention Centre (SARIC), School of Psychology, University of Sussex, Brighton BN1 9RH, United Kingdom
| | - Eisuke Koya
- Sussex Addiction Research and Intervention Centre (SARIC), School of Psychology, University of Sussex, Brighton BN1 9RH, United Kingdom
| | - Aldo Badiani
- Sussex Addiction Research and Intervention Centre (SARIC), School of Psychology, University of Sussex, Brighton BN1 9RH, United Kingdom
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
44
|
Scida K, Plaxco KW, Jamieson BG. High frequency, real-time neurochemical and neuropharmacological measurements in situ in the living body. Transl Res 2019; 213:50-66. [PMID: 31361988 DOI: 10.1016/j.trsl.2019.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/20/2019] [Accepted: 07/11/2019] [Indexed: 12/18/2022]
Abstract
The beautiful and complex brain machinery is perfectly synchronized, and our bodies have evolved to protect it against a myriad of potential threats. Shielded physically by the skull and chemically by the blood brain barrier, the brain processes internal and external information so that we can efficiently relate to the world that surrounds us while simultaneously and unconsciously controlling our vital functions. When coupled with the brittle nature of its internal chemical and electric signals, the brain's "armor" render accessing it a challenging and delicate endeavor that has historically limited our understanding of its structural and neurochemical intricacies. In this review, we briefly summarize the advancements made over the past 10 years to decode the brain's neurochemistry and neuropharmacology in situ, at the site of interest in the brain, with special focus on what we consider game-changing emerging technologies (eg, genetically encoded indicators and electrochemical aptamer-based sensors) and the challenges these must overcome before chronic, in situ chemosensing measurements become routine.
Collapse
Affiliation(s)
- Karen Scida
- Diagnostic Biochips, Inc., Glen Burnie, Maryland
| | - Kevin W Plaxco
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California
| | | |
Collapse
|
45
|
Nickoloff E, Mackie P, Runner K, Matt S, Khoshbouei H, Gaskill P. Dopamine increases HIV entry into macrophages by increasing calcium release via an alternative signaling pathway. Brain Behav Immun 2019; 82:239-252. [PMID: 31470080 PMCID: PMC6941734 DOI: 10.1016/j.bbi.2019.08.191] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 08/22/2019] [Accepted: 08/26/2019] [Indexed: 12/19/2022] Open
Abstract
Dopaminergic dysfunction has long been connected to the development of HIV infection in the CNS. Our previous data showed that dopamine increases HIV infection in human macrophages by increasing the susceptibility of primary human macrophages to HIV entry through stimulation of both D1-like and D2-like receptors. These data suggest that, in macrophages, both dopamine receptor subtypes may act through a common signaling mechanism. To define better the mechanism(s) underlying this effect, this study examines the specific signaling processes activated by dopamine in primary human monocyte-derived macrophages (hMDM). In addition to confirming that the increase in entry is unique to dopamine, these studies show that dopamine increases HIV entry through a PKA insensitive, Ca2+ dependent pathway. Further examination demonstrated that dopamine can signal through a previously defined, non-canonical pathway in human macrophages. This pathway involves both Ca2+ release and PKC phosphorylation, and these data show that dopamine mediates both of these effects and that both were partially inhibited by the Gq/11 specific inhibitor YM-254890. Studies have shown that Gq/11 preferentially couples to the D1-like receptor D5, indicating an important role of the D1-like receptors in mediating these effects. These data indicate a role for Ca2+ flux in the HIV entry process, and suggest a distinct signaling mechanism mediating some of the effects of dopamine in macrophages. Together, the data indicate that targeting this alternative dopamine signaling pathway might provide new therapeutic options for individuals with elevated CNS dopamine suffering from NeuroHIV.
Collapse
Affiliation(s)
- E.A. Nickoloff
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102
| | - P. Mackie
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611
| | - K. Runner
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102
| | - S.M. Matt
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102
| | - H. Khoshbouei
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611,Department of Psychiatry, University of Florida, Gainesville, FL, 32611
| | - P.J. Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102
| |
Collapse
|
46
|
Task-Demand-Dependent Neural Representation of Odor Information in the Olfactory Bulb and Posterior Piriform Cortex. J Neurosci 2019; 39:10002-10018. [PMID: 31672791 DOI: 10.1523/jneurosci.1234-19.2019] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 10/16/2019] [Accepted: 10/19/2019] [Indexed: 02/03/2023] Open
Abstract
In awake rodents, the neural representation of olfactory information in the olfactory bulb is largely dependent on brain state and behavioral context. Learning-modified neural plasticity has been observed in mitral/tufted cells, the main output neurons of the olfactory bulb. Here, we propose that the odor information encoded by mitral/tufted cell responses in awake mice is highly dependent on the behavioral task demands. We used fiber photometry to record calcium signals from the mitral/tufted cell population in awake, head-fixed male mice under different task demands. We found that the mitral/tufted cell population showed similar responses to two distinct odors when the odors were presented in the context of a go/go task, in which the mice received a water reward regardless of the identity of the odor presented. However, when the same odors were presented in a go/no-go task, in which one odor was rewarded and the other was not, then the mitral cell population responded very differently to the two odors, characterized by a robust reduction in the response to the nonrewarded odor. Thus, the representation of odors in the mitral/tufted cell population depends on whether the task requires discrimination of the odors. Strikingly, downstream of the olfactory bulb, pyramidal neurons in the posterior piriform cortex also displayed a task-demand-dependent neural representation of odors, but the anterior piriform cortex did not, indicating that these two important higher olfactory centers use different strategies for neural representation.SIGNIFICANCE STATEMENT The most important task of the olfactory system is to generate a precise representation of odor information under different brain states. Whether the representation of odors by neurons in olfactory centers such as the olfactory bulb and the piriform cortex depends on task demands remains elusive. We find that odor representation in the mitral/tufted cells of the olfactory bulb depends on whether the task requires odor discrimination. A similar neural representation is found in the posterior piriform cortex but not the anterior piriform cortex, indicating that these higher olfactory centers use different representational strategies. The task-demand-dependent representational strategy is likely important for facilitating information processing in higher brain centers responsible for decision making and encoding of salience.
Collapse
|
47
|
Frankowska M, Miszkiel J, Pomierny-Chamioło L, Pomierny B, Borelli AC, Suder A, Filip M. Extinction training following cocaine or MDMA self-administration produces discrete changes in D 2-like and mGlu 5 receptor density in the rat brain. Pharmacol Rep 2019; 71:870-878. [PMID: 31408786 DOI: 10.1016/j.pharep.2019.05.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 04/11/2019] [Accepted: 05/06/2019] [Indexed: 11/17/2022]
Abstract
BACKGROUND Several studies strongly support the role of the dopamine D2-like and glutamate mGlu5 receptors in psychostimulant reward and relapse. METHODS The present study employed cocaine or MDMA self-administration with yoked-triad procedure in rats to explore whether extinction training affects the drug-seeking behavior and the D2-like and mGlu5 receptor Bmax and Kd values in several regions of the animal brain. RESULTS Both cocaine and MDMA rats developed maintenance of self-administration, but MDMA evoked lower response rates and speed of self-administration acquisition. During reinstatement tests, cocaine or MDMA seeking behavior was produced by either exposure to the drug-associated cues or drug-priming injections. The extinction training after cocaine self-administration did not alter significantly D2-like receptor expression the in the limbic and subcortical brain areas, while MDMA yoked rats showed a decrease of the D2-like binding density in the nucleus accumbens and increase in the hippocampus and a rise of affinity in the striatum and hippocampus. Interestingly, in the prefrontal cortex a reduction in the mGlu5 receptor density in cocaine- or MDMA-abstinent rats was demonstrated, with significant effects being observed after previous MDMA exposure. Moreover, rats self-administered cocaine showed a rise in the density of mGlu5 receptor for the nucleus accumbens. CONCLUSION This study first time shows that abstinence followed extinction training after cocaine or MDMA self- or passive-injections changes the D2-like and mGlu5 density and affinity. The observed changes in the expression of both receptors are brain-region specific and related to either pharmacological and/or motivational features of cocaine or MDMA.
Collapse
Affiliation(s)
- Małgorzata Frankowska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Kraków, Poland.
| | - Joanna Miszkiel
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Kraków, Poland
| | - Lucyna Pomierny-Chamioło
- Department of Toxicology, Faculty of Pharmacy, Jagiellonian University Collegium Medicum, Kraków, Poland
| | - Bartosz Pomierny
- Department of Toxicology, Faculty of Pharmacy, Jagiellonian University Collegium Medicum, Kraków, Poland
| | | | - Agata Suder
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Kraków, Poland
| | - Małgorzata Filip
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Kraków, Poland
| |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW Recent epidemiology, biological and clinical findings correlate high cigarette consumption in patients with schizophrenia, impeding both treatment strategies and the effectiveness of antipsychotics. RECENT FINDINGS New data suggests that despite world-wide efforts to curb cigarette consumption, smoking in patients with schizophrenia was still high. Recent reports could not confirm earlier findings regarding smoking's beneficial effects on cognitive dysfunction, however, the association between smoking, positive symptoms and suicidal behavior was revealed. As some patients smoked in an attempt to alleviate extrapyramidal symptoms (EPS) and negative symptoms, the molecular studies shared genetic roots correlating smoking and schizophrenia, revealing that smoking may increase the risk of developing schizophrenia. Preclinical and clinical studies clarified the complex relationship between schizophrenia's pathology and nicotine's effects on the human brain. SUMMARY Cigarette smoking continues to adversely affect the health of individuals with schizophrenia. Both smoking and heavy nicotine dependence, given the complex biological findings, might influence symptom severity in patients with schizophrenia. Regardless, ceasing smoking activities is strongly advocated to replace 'self-medication by nicotine' with safer and more effective medications.
Collapse
|
49
|
Nestler EJ, Lüscher C. The Molecular Basis of Drug Addiction: Linking Epigenetic to Synaptic and Circuit Mechanisms. Neuron 2019; 102:48-59. [PMID: 30946825 PMCID: PMC6587180 DOI: 10.1016/j.neuron.2019.01.016] [Citation(s) in RCA: 194] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/01/2019] [Accepted: 01/10/2019] [Indexed: 12/22/2022]
Abstract
Addiction is a disease in which, after a period of recreational use, a subset of individuals develops compulsive use that does not stop even in light of major negative consequences. Here, we review the evidence for underlying epigenetic remodeling in brain in two settings. First, excessive dopamine signaling during drug use may modulate gene expression, altering synaptic function and circuit activity and leading over time to maladaptive behaviors in vulnerable individuals. Second, on a longer timescale, life experience can shape the epigenetic landscape in brain and thereby may contribute to an individual's vulnerability by amplifying drug-induced changes in gene expression that drive the transition to addiction. We conclude by exploring how epigenetic mechanisms might serve as therapeutic targets for addiction treatments.
Collapse
Affiliation(s)
- Eric J Nestler
- Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christian Lüscher
- Department of Basic Neurosciences, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Clinic of Neurology, Departement of Clinical Neurosiences, Geneva University Hospital, Geneva, Switzerland.
| |
Collapse
|
50
|
Schiavone S, Neri M, Maffione AB, Frisoni P, Morgese MG, Trabace L, Turillazzi E. Increased iNOS and Nitrosative Stress in Dopaminergic Neurons of MDMA-Exposed Rats. Int J Mol Sci 2019; 20:E1242. [PMID: 30871034 PMCID: PMC6429174 DOI: 10.3390/ijms20051242] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/28/2019] [Accepted: 03/05/2019] [Indexed: 12/29/2022] Open
Abstract
Several mechanisms underlying 3,4-Methylenedioxy-N-methylamphetamine (MDMA) neurotoxicity have been proposed, including neurochemical alterations and excitotoxicity mediated by reactive oxygen species (ROS), nitric oxide (NO), and reactive nitrogen species (RNS). However, ROS, NO, and RNS sources in the brain are not fully known. We aimed to investigate possible alterations in the expression of the ROS producer NOX enzymes (NOX2, NOX1, and NOX4), NO generators (iNOS, eNOS, and nNOS), markers of oxidative (8-hydroxy-2'-deoxyguanosine, 8OHdG), and nitrosative (3-nitrotyrosine, NT) stress, as well as the colocalization between cells positive for the dopamine transporter (DT1) and cells expressing the neuronal nuclei (NeuN) marker, in the frontal cortex of rats receiving saline or MDMA, sacrificed 6 h, 16 h, or 24 h after its administration. MDMA did not affect NOX2, NOX1, and NOX4 immunoreactivity, whereas iNOS expression was enhanced. The number of NT-positive cells was increased in MDMA-exposed animals, whereas no differences were detected in 8OHdG expression among experimental groups. MDMA and NT markers colocalized with DT1 positive cells. DT1 immunostaining was found in NeuN-positive stained cells. Virtually no colocalization was observed with microglia and astrocytes. Moreover, MDMA immunostaining was not found in NOX2-positive cells. Our results suggest that iNOS-derived nitrosative stress, but not NOX enzymes, may have a crucial role in the pathogenesis of MDMA-induced neurotoxicity, highlighting the specificity of different enzymatic systems in the development of neuropathological alterations induced by the abuse of this psychoactive compound.
Collapse
Affiliation(s)
- Stefania Schiavone
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli, 20, 71122 Foggia, Italy.
| | - Margherita Neri
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Fossato di Mortara, 70, 44100 Ferrara, Italy.
| | - Angela Bruna Maffione
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli, 20, 71122 Foggia, Italy.
| | - Paolo Frisoni
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Fossato di Mortara, 70, 44100 Ferrara, Italy.
| | - Maria Grazia Morgese
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli, 20, 71122 Foggia, Italy.
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli, 20, 71122 Foggia, Italy.
| | - Emanuela Turillazzi
- Section of Legal Medicine, Department of Surgical, Medical, Molecular and Critical Pathology, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| |
Collapse
|