1
|
Khezerlou E, Saenz J, Prakash SS, Pan PY. Protocol for live neuron imaging analysis of basal surface fraction and dynamic availability of the dopamine transporter using DAT-pHluorin. STAR Protoc 2024; 5:103358. [PMID: 39368094 PMCID: PMC11490699 DOI: 10.1016/j.xpro.2024.103358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 08/03/2024] [Accepted: 09/12/2024] [Indexed: 10/07/2024] Open
Abstract
Surface availability of the dopamine (DA) transporter (DAT) critically influences DA transmission. Here, we present a protocol that describes the preparation of mouse ventral midbrain neurons, the expression of a new optical sensor, DAT-pHluorin, and the utilization of this sensor to analyze the surface availability of DAT in live neurons via fluorescent microscopy. This approach allows quantitative measures of basal surface DAT fraction under genetic backgrounds of interest and live trafficking of DAT in response to psychoactive substances. For complete details on the use and execution of this protocol, please refer to Saenz et al.1.
Collapse
Affiliation(s)
- Elnaz Khezerlou
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ 08854, USA; Rutgers Addiction Research Center, Rutgers Brain Health Institute, 683 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Jacqueline Saenz
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ 08854, USA; Rutgers School of Graduate Studies, Molecular Biosciences Graduate Program, 25 Bishop Place, New Brunswick, NJ 08901-1178, USA; Rutgers Addiction Research Center, Rutgers Brain Health Institute, 683 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Sanjana Surya Prakash
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ 08854, USA; Rutgers School of Graduate Studies, Biomedical Science Master's Program, 25 Bishop Place, New Brunswick, NJ 08901-1178, USA
| | - Ping-Yue Pan
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ 08854, USA; Rutgers Addiction Research Center, Rutgers Brain Health Institute, 683 Hoes Lane West, Piscataway, NJ 08854, USA.
| |
Collapse
|
2
|
Cui H, Shu C, Peng Y, Wei Z, Ni X, Zheng L, Shang J, Liu F, Liu J. Long-life triclosan exposure induces ADHD-like behavior in rats via prefrontal cortex dopaminergic deficiency. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 282:116766. [PMID: 39047361 DOI: 10.1016/j.ecoenv.2024.116766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024]
Abstract
In recent years, exposure to triclosan (TCS) has been linked to an increase in psychiatric disorders. Nonetheless, the precise mechanisms of this occurrence remain elusive. Therefore, this study developed a long-life TCS-exposed rat model, an SH-SY5Y cell model, and an atomoxetine hydrochloride (ATX) treatment model to explore and validate the neurobehavioral mechanisms of TCS from multiple perspectives. In the long-life TCS-exposed model, pregnant rats received either 0 mg/kg (control) or 50 mg/kg TCS by oral gavage throughout pregnancy, lactation, and weaning of their offspring (up to 8 weeks old). In the ATX treatment model, weanling rats received daily injections of either 0 mg/kg (control) or 3 mg/kg ATX via intraperitoneal injection until they reached 8 weeks old. Unlike the TCS model, ATX exposure only occurred after the pups were weaned. The results indicated that long-life TCS exposure led to attention-deficit hyperactivity disorder (ADHD)-like behaviors in male offspring rats accompanied by dopamine-related mRNA and protein expression imbalances in the prefrontal cortex (PFC). Moreover, in vitro experiments also confirmed these findings. Mechanistically, TCS reduced dopamine (DA) synthesis, release, and transmission, and increased reuptake in PFC, thereby reducing synaptic gap DA levels and causing dopaminergic deficits. Additional experiments revealed that increased DA concentration in PFC by ATX effectively alleviated TCS-induced ADHD-like behavior in male offspring rats. These findings suggest that long-life TCS exposure causes ADHD-like behavior in male offspring rats through dopaminergic deficits. Furthermore, ATX treatment not only reduce symptoms in the rats, but also reveals valuable insights into the neurotoxic mechanisms induced by TCS.
Collapse
Affiliation(s)
- He Cui
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang 110122, PR China
| | - Chang Shu
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang 110122, PR China
| | - Yuxuan Peng
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang 110122, PR China
| | - Ziyun Wei
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang 110122, PR China
| | - Xiao Ni
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang 110122, PR China
| | - Linlin Zheng
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang 110122, PR China
| | - Jianing Shang
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang 110122, PR China
| | - Fu Liu
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang 110122, PR China
| | - Jieyu Liu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, PR China; Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang 110122, PR China.
| |
Collapse
|
3
|
Carboni E, Ibba M, Carboni E, Carta AR. Adolescent stress differentially modifies dopamine and norepinephrine release in the medial prefrontal cortex of adult rats. Prog Neuropsychopharmacol Biol Psychiatry 2024; 134:111055. [PMID: 38879069 DOI: 10.1016/j.pnpbp.2024.111055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 04/10/2024] [Accepted: 06/11/2024] [Indexed: 06/22/2024]
Abstract
Adolescent stress (AS) has been associated with higher vulnerability to psychiatric disorders such as schizophrenia, depression, or drug dependence. Moreover, the alteration of brain catecholamine (CAT) transmission in the medial prefrontal cortex (mPFC) has been found to play a major role in the etiology of psychiatric disturbances. We investigated the effect of adolescent stress on CAT transmission in the mPFC of freely moving adult rats because of the importance of this area in the etiology of psychiatric disorders, and because CAT transmission is the target of a relevant group of drugs used in the therapy of depression and psychosis. We assessed basal dopamine (DA) and norepinephrine (NE) extracellular concentrations (output) by brain microdialysis in in the mPFC of adult rats that were exposed to chronic mild stress in adolescence. To ascertain the role of an altered release or reuptake, we stimulated DA and NE output by administering either different doses of amphetamine (0.5 and 1.0 mg / kg s.c.), which by a complex mechanism determines a dose dependent increase in the CAT output, or reboxetine (10 mg/kg i.p.), a selective NE reuptake inhibitor. The results showed the following: (i) basal DA output in AS rats was lower than in controls, while no difference in basal NE output was observed; (ii) amphetamine, dose dependently, stimulated DA and NE output to a greater extent in AS rats than in controls; (iii) reboxetine stimulated NE output to a greater extent in AS rats than in controls, while no difference in stimulated DA output was observed between the two groups. These results show that AS determines enduring effects on DA and NE transmission in the mPFC and might lead to the occurrence of psychiatric disorders or increase the vulnerability to drug addiction.
Collapse
Affiliation(s)
- Ezio Carboni
- Department of Biomedical Sciences, University of Cagliari, Italy.
| | - Marcello Ibba
- Department of Biomedical Sciences, University of Cagliari, Italy
| | - Elena Carboni
- Unit of Paediatrics, ASST Cremona Maggiore Hospital, Cremona, Italy
| | - Anna R Carta
- Department of Biomedical Sciences, University of Cagliari, Italy
| |
Collapse
|
4
|
Kwon H, Hong E, Lee YS, Cheong JH, Kim HJ, Kim S, Yun J. Augmentation of intracranial self-stimulation induced by amphetamine-like drugs in Period circadian regulator 2 knockout mice is associated with intracellular Ca 2+ levels. Neurosci Res 2024:S0168-0102(24)00099-3. [PMID: 39094980 DOI: 10.1016/j.neures.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/18/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024]
Abstract
Over the past decade, new psychoactive substances (NPS) have emerged in the illegal drug market and have continued to attract attention from the international community. Among these, amphetamine-like NPS, classified as stimulants, constitute a significant proportion. However, the pharmacological characteristics and mechanisms underlying addiction to amphetamine-like NPS remain poorly understood. Given that circadian rhythms are linked to the brain stimulation effects of methamphetamine (METH) and amphetamine, we investigated the effects of METH, 1-(4-methoxyphenyl)-N-methylpropan-2-amine (PMMA), and 1-(benzofuran-5-yl)-N-ethylpropan-2-amine (5-EAPB) on intracranial self-stimulation (ICSS) in wild-type (WT) or Period circadian regulator 2 knockout mice. Amphetamine-like drugs increase intracellular Ca2+ levels to provoke dopamine release, so we examined the impact of Per2 knockdown on intracellular Ca2+ levels in PC12 cells to elucidate a potential mechanism underlying NPS-induced ICSS enhancement. Our ICSS results showed that METH and PMMA significantly increased brain stimulation in Per2 knockout mice compared to WT mice. Similarly, METH and PMMA induced higher Ca2+ fluorescence intensity in Per2 knockdown PC12 cells than in control cells. In contrast, 5-EAPB did not produce significant changes in either ICSS or Ca2+ signaling. These findings suggest that Per2 plays a crucial role in the brain stimulation effects of amphetamine-like drugs through the regulation of intracellular Ca2+.
Collapse
Affiliation(s)
- Hyeokjun Kwon
- College of Pharmacy, Chungbuk National University, 194-31 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, Republic of Korea
| | - Eunchong Hong
- College of Pharmacy, Chungbuk National University, 194-31 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, Republic of Korea
| | - Yong Sup Lee
- College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Jae Hoon Cheong
- College of Pharmacy, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do 54896, Republic of Korea
| | - Hee Jin Kim
- College of Pharmacy, Sahmyook University, 567 Hwarang-ro, Nowon-gu, Seoul 01795, Republic of Korea
| | - Soyoung Kim
- Medistation, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, Republic of Korea
| | - Jaesuk Yun
- College of Pharmacy, Chungbuk National University, 194-31 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, Republic of Korea.
| |
Collapse
|
5
|
Jiménez-Torres AC, Porter KD, Hastie JA, Adeniran C, Moukha-Chafiq O, Nguyen TH, Ananthan S, Augelli-Szafran CE, Zhan CG, Zhu J. Effects of SRI-32743, a Novel Quinazoline Structure-Based Compound, on HIV-1 Tat and Cocaine Interaction with Norepinephrine Transporter. Int J Mol Sci 2024; 25:7881. [PMID: 39063123 PMCID: PMC11277056 DOI: 10.3390/ijms25147881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Prolonged exposure to HIV-1 transactivator of transcription (Tat) protein dysregulates monoamine transmission, a physiological change implicated as a key factor in promoting neurocognitive disorders among people living with HIV. We have demonstrated that in vivo expression of Tat in Tat transgenic mice decreases dopamine uptake through both dopamine transporter (DAT) and norepinephrine transporter (NET) in the prefrontal cortex. Further, our novel allosteric inhibitor of monoamine transporters, SRI-32743, has been shown to attenuate Tat-inhibited dopamine transport through DAT and alleviates Tat-potentiated cognitive impairments. The current study reports the pharmacological profiles of SRI-32743 in basal and Tat-induced inhibition of human NET (hNET) function. SRI-32743 exhibited less affinity for hNET binding than desipramine, a classical NET inhibitor, but displayed similar potency for inhibiting hDAT and hNET activity. SRI-32743 concentration-dependently increased hNET affinity for [3H]DA uptake but preserved the Vmax of dopamine transport. SRI-32743 slowed the cocaine-mediated dissociation of [3H]Nisoxetine binding and reduced both [3H]DA and [3H]MPP+ efflux but did not affect d-amphetamine-mediated [3H]DA release through hNET. Finally, we determined that SRI-32743 attenuated a recombinant Tat1-86-induced decrease in [3H]DA uptake via hNET. Our findings demonstrated that SRI-32743 allosterically disrupts the recombinant Tat1-86-hNET interaction, suggesting a potential treatment for HIV-infected individuals with concurrent cocaine abuse.
Collapse
Affiliation(s)
- Ana Catya Jiménez-Torres
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter Street, Columbia, SC 29208, USA; (A.C.J.-T.); (K.D.P.); (J.A.H.)
| | - Katherine D. Porter
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter Street, Columbia, SC 29208, USA; (A.C.J.-T.); (K.D.P.); (J.A.H.)
| | - Jamison A. Hastie
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter Street, Columbia, SC 29208, USA; (A.C.J.-T.); (K.D.P.); (J.A.H.)
| | - Charles Adeniran
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA; (C.A.); (C.-G.Z.)
- Molecular Modeling and Biopharmaceutical Center, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - Omar Moukha-Chafiq
- Department of Chemistry, Scientific Platforms Division, Southern Research, Birmingham, AL 35205, USA; (O.M.-C.); (T.H.N.); (S.A.); (C.E.A.-S.)
| | - Theresa H. Nguyen
- Department of Chemistry, Scientific Platforms Division, Southern Research, Birmingham, AL 35205, USA; (O.M.-C.); (T.H.N.); (S.A.); (C.E.A.-S.)
| | - Subramaniam Ananthan
- Department of Chemistry, Scientific Platforms Division, Southern Research, Birmingham, AL 35205, USA; (O.M.-C.); (T.H.N.); (S.A.); (C.E.A.-S.)
| | - Corinne E. Augelli-Szafran
- Department of Chemistry, Scientific Platforms Division, Southern Research, Birmingham, AL 35205, USA; (O.M.-C.); (T.H.N.); (S.A.); (C.E.A.-S.)
| | - Chang-Guo Zhan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA; (C.A.); (C.-G.Z.)
- Molecular Modeling and Biopharmaceutical Center, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - Jun Zhu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter Street, Columbia, SC 29208, USA; (A.C.J.-T.); (K.D.P.); (J.A.H.)
| |
Collapse
|
6
|
Gobira PH, Joca SR, Moreira FA. Roles of cannabinoid CB1 and CB2 receptors in the modulation of psychostimulant responses. Acta Neuropsychiatr 2024; 36:67-77. [PMID: 35993329 DOI: 10.1017/neu.2022.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Addiction to psychostimulant drugs, such as cocaine, D-amphetamine, and methamphetamine, is a public health issue that substantially contributes to the global burden of disease. Psychostimulant drugs promote an increase in dopamine levels within the mesocorticolimbic system, which is central to the rewarding properties of such drugs. Cannabinoid receptors (CB1R and CB2R) are expressed in the main areas of this system and implicated in the neuronal mechanisms underlying the rewarding effect of psychostimulant drugs. Here, we reviewed studies focusing on pharmacological intervention targeting cannabinoid CB1R and CB2R and their interaction in the modulation of psychostimulant responses.
Collapse
Affiliation(s)
- P H Gobira
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - S R Joca
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - F A Moreira
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
7
|
Gölöncsér F, Baranyi M, Tod P, Maácz F, Sperlágh B. P2X7 receptor inhibition alleviates mania-like behavior independently of interleukin-1β. iScience 2024; 27:109284. [PMID: 38444608 PMCID: PMC10914489 DOI: 10.1016/j.isci.2024.109284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 12/15/2023] [Accepted: 02/16/2024] [Indexed: 03/07/2024] Open
Abstract
Purinergic dysfunctions are associated with mania and depression pathogenesis. P2X7 receptor (P2X7R) mediates the IL-1β maturation via NLRP3 inflammasome activation. We tested in a mouse model of the subchronic amphetamine (AMPH)-induced hyperactivity whether P2X7R inhibition alleviated mania-like behavior through IL-1β. Treatment with JNJ-47965567, a P2X7R antagonist, abolished AMPH-induced hyperlocomotion in wild-type and IL-1α/β-knockout male mice. The NLRP3 inhibitor MCC950 failed to reduce AMPH-induced locomotion in WT mice, whereas the IL-1 receptor antagonist anakinra slightly increased it. AMPH increased IL-10, TNF-α, and TBARS levels, but did not influence BDNF levels, serotonin, dopamine, and noradrenaline content in brain tissues in either genotypes. JNJ-47965567 and P2rx7-gene deficiency, but not IL-1α/β-gene deficiency, attenuated AMPH-induced [3H]dopamine release from striatal slices. In wild-type and IL-1α/β-knockout female mice, JNJ-47965567 was also effective in attenuating AMPH-induced hyperlocomotion. This study suggests that AMPH-induced hyperactivity is modulated by P2X7Rs, but not through IL-1β.
Collapse
Affiliation(s)
- Flóra Gölöncsér
- Laboratory of Molecular Pharmacology, HUN-REN Institute of Experimental Medicine, 1083 Budapest, Hungary
| | - Mária Baranyi
- Laboratory of Molecular Pharmacology, HUN-REN Institute of Experimental Medicine, 1083 Budapest, Hungary
| | - Pál Tod
- Laboratory of Molecular Pharmacology, HUN-REN Institute of Experimental Medicine, 1083 Budapest, Hungary
| | - Fruzsina Maácz
- Laboratory of Molecular Pharmacology, HUN-REN Institute of Experimental Medicine, 1083 Budapest, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University School of Ph.D Studies, 1083 Budapest, Hungary
| | - Beáta Sperlágh
- Laboratory of Molecular Pharmacology, HUN-REN Institute of Experimental Medicine, 1083 Budapest, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University School of Ph.D Studies, 1083 Budapest, Hungary
| |
Collapse
|
8
|
Vaughan RA, Henry LK, Foster JD, Brown CR. Post-translational mechanisms in psychostimulant-induced neurotransmitter efflux. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 99:1-33. [PMID: 38467478 DOI: 10.1016/bs.apha.2023.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
The availability of monoamine neurotransmitters in the brain is under the control of dopamine, norepinephrine, and serotonin transporters expressed on the plasma membrane of monoaminergic neurons. By regulating transmitter levels these proteins mediate crucial functions including cognition, attention, and reward, and dysregulation of their activity is linked to mood and psychiatric disorders of these systems. Amphetamine-based transporter substrates stimulate non-exocytotic transmitter efflux that induces psychomotor stimulation, addiction, altered mood, hallucinations, and psychosis, thus constituting a major component of drug neurochemical and behavioral outcomes. Efflux is under the control of transporter post-translational modifications that synergize with other regulatory events, and this review will summarize our knowledge of these processes and their role in drug mechanisms.
Collapse
Affiliation(s)
- Roxanne A Vaughan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States.
| | - L Keith Henry
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| | - James D Foster
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| | - Christopher R Brown
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| |
Collapse
|
9
|
Bagalkot T, Sorkin A. Amphetamine Induces Sex-Dependent Loss of the Striatal Dopamine Transporter in Sensitized Mice. eNeuro 2024; 11:ENEURO.0491-23.2023. [PMID: 38164591 PMCID: PMC10849026 DOI: 10.1523/eneuro.0491-23.2023] [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: 11/23/2023] [Revised: 12/10/2023] [Accepted: 12/12/2023] [Indexed: 01/03/2024] Open
Abstract
Dopamine transporter (DAT) controls dopamine signaling in the brain through the reuptake of synaptically released dopamine. DAT is a target of abused psychostimulants such as amphetamine (Amph). Acute Amph administration induces transient DAT endocytosis, which, among other Amph effects on dopaminergic neurons, elevates extracellular dopamine. However, the effects of repeated Amph abuse, leading to behavioral sensitization and drug addiction, on DAT are unknown. Hence, we developed a 14 d Amph-sensitization protocol in knock-in mice expressing HA-epitope-tagged DAT (HA-DAT) and investigated the effects of Amph challenge on sensitized HA-DAT animals. The Amph challenge resulted in the highest locomotor activity on Day 14 in both sexes, which was sustained for 1 h in male but not female mice. Strikingly, significant (by 30-60%) loss of the HA-DAT protein in the striatum was caused by the Amph challenge of sensitized males but not females. Amph also reduced V max of dopamine transport in the striatal synaptosomes of males without changing K m values. Consistently, immunofluorescence microscopy revealed a significant increase of HA-DAT colocalization with the endosomal protein VPS35 only in Amph-challenged males. Amph-induced loss of striatal HA-DAT in sensitized mice was blocked by chloroquine, vacuolin-1, and inhibitor of Rho-associated kinases ROCK1/2, indicative of the involvement of endocytic trafficking in the DAT protein loss. Interestingly, an apparent degradation of HA-DAT protein was observed in the nucleus accumbens and not in the dorsal striatum. We propose that Amph challenge in sensitized mice triggers Rho-mediated endocytosis and post-endocytic trafficking of DAT in a brain-region-specific and sex-dependent manner.
Collapse
Affiliation(s)
- Tarique Bagalkot
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh 15261, Pennsylvania
| | - Alexander Sorkin
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh 15261, Pennsylvania
| |
Collapse
|
10
|
Mayer FP, Niello M, Bulling S, Zhang YW, Li Y, Kudlacek O, Holy M, Kooti F, Sandtner W, Rudnick G, Schmid D, Sitte HH. Mephedrone induces partial release at human dopamine transporters but full release at human serotonin transporters. Neuropharmacology 2023; 240:109704. [PMID: 37703919 DOI: 10.1016/j.neuropharm.2023.109704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 07/07/2023] [Accepted: 08/31/2023] [Indexed: 09/15/2023]
Abstract
Mephedrone (4-methylmethcathinone) is a cathinone derivative that is recreationally consumed for its energizing and empathogenic effects. The stimulating properties are believed to arise from the ability of mephedrone to interact with the high-affinity transporters for dopamine (DA) (DAT) and norepinephrine (NET), whereas the entactogenic effect presumably relies on its activity at the serotonin (5-HT) transporter (SERT). Early studies found that mephedrone acts as a releaser at NET, DAT and SERT, and thus promotes efflux of the respective monoamines. Evidence linked drug-induced reverse transport of 5-HT via SERT to prosocial effects, whereas activity at DAT is strongly correlated with abuse liability. Consequently, we sought to evaluate the pharmacology of mephedrone at human (h) DAT and SERT, heterologously expressed in human embryonic kidney 293 cells, in further detail. In line with previous studies, we report that mephedrone evokes carrier-mediated release via hDAT and hSERT. We found this effect to be sensitive to the protein kinase C inhibitor GF109203X. Electrophysiological recordings revealed that mephedrone is actively transported by hDAT and hSERT. However, mephedrone acts as a full substrate of hSERT but as a partial substrate of hDAT. Furthermore, when compared to fully efficacious releasing agents at hDAT and hSERT (i.e. S(+)-amphetamine and para-chloroamphetamine, respectively) mephedrone displays greater efficacy as a releaser at hSERT than at hDAT. In summary, this study provides additional insights into the molecular mechanism of action of mephedrone at hDAT and hSERT.
Collapse
Affiliation(s)
- Felix P Mayer
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria
| | - Marco Niello
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria
| | - Simon Bulling
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria
| | - Yuan-Wei Zhang
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8066, USA
| | - Yang Li
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria
| | - Oliver Kudlacek
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria
| | - Marion Holy
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria
| | - Fatemeh Kooti
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria
| | - Walter Sandtner
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria
| | - Gary Rudnick
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8066, USA
| | - Diethart Schmid
- Institute for Physiology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria
| | - Harald H Sitte
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria; Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman, Jordan; Center for Addiction Research and Science - AddRess, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria.
| |
Collapse
|
11
|
Zhang J, Nguyen AH, Jilani D, Trigo Torres RS, Schmiess-Heine L, Le T, Xia X, Cao H. Consecutive treatments of methamphetamine promote the development of cardiac pathological symptoms in zebrafish. PLoS One 2023; 18:e0294322. [PMID: 37976248 PMCID: PMC10655962 DOI: 10.1371/journal.pone.0294322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 10/30/2023] [Indexed: 11/19/2023] Open
Abstract
Chronic methamphetamine use, a widespread drug epidemic, has been associated with cardiac morphological and electrical remodeling, leading to the development of numerous cardiovascular diseases. While methamphetamine has been documented to induce arrhythmia, most results originate from clinical trials from users who experienced different durations of methamphetamine abuse, providing no documentation on the use of methamphetamine in standardized settings. Additionally, the underlying molecular mechanism on how methamphetamine affects the cardiovascular system remains elusive. A relationship was sought between cardiotoxicity and arrhythmia with associated methamphetamine abuse in zebrafish to identify and to understand the adverse cardiac symptoms associated with methamphetamine. Zebrafish were first treated with methamphetamine 3 times a week over a 2-week duration. Immediately after treatment, zebrafish underwent electrocardiogram (ECG) measurement using an in-house developed acquisition system for electrophysiological analysis. Subsequent analyses of cAMP expression and Ca2+ regulation in zebrafish cardiomyocytes were conducted. cAMP is vital to development of myocardial fibrosis and arrhythmia, prominent symptoms in the development of cardiovascular diseases. Ca2+ dysregulation is also a factor in inducing arrhythmias. During the first week of treatment, zebrafish that were administered with methamphetamine displayed a decrease in heart rate, which persisted throughout the second week and remained significantly lower than the heart rate of untreated fish. Results also indicate an increased heart rate variability during the early stage of treatment followed by a decrease in the late stage for methamphetamine-treated fish over the duration of the experiment, suggesting a biphasic response to methamphetamine exposure. Methamphetamine-treated fish also exhibited reduced QTc intervals throughout the experiment. Results from the cAMP and Ca2+ assays demonstrate that cAMP was upregulated and Ca2+ was dysregulated in response to methamphetamine treatment. Collagenic assays indicated significant fibrotic response to methamphetamine treatment. These results provide potential insight into the role of methamphetamine in the development of fibrosis and arrhythmia due to downstream effectors of cAMP.
Collapse
Affiliation(s)
- Jimmy Zhang
- Department of Biomedical Engineering, University of California-Irvine, Irvine, CA, United States of America
| | - Anh H. Nguyen
- Department of Electrical Engineering and Computer Science, University of California-Irvine, Irvine, CA, United States of America
- Sensoriis, Inc., Edmonds, WA, United States of America
| | - Daniel Jilani
- Department of Electrical Engineering and Computer Science, University of California-Irvine, Irvine, CA, United States of America
| | | | - Lauren Schmiess-Heine
- Department of Electrical Engineering and Computer Science, University of California-Irvine, Irvine, CA, United States of America
| | - Tai Le
- Department of Biomedical Engineering, University of California-Irvine, Irvine, CA, United States of America
| | - Xing Xia
- Department of Electrical Engineering and Computer Science, University of California-Irvine, Irvine, CA, United States of America
| | - Hung Cao
- Department of Biomedical Engineering, University of California-Irvine, Irvine, CA, United States of America
- Department of Electrical Engineering and Computer Science, University of California-Irvine, Irvine, CA, United States of America
- Sensoriis, Inc., Edmonds, WA, United States of America
| |
Collapse
|
12
|
Chang SH, Pai PY, Hsu CH, Marthandam Asokan S, Tsai BCK, Weng WT, Kuo WW, Shih TC, Kao HC, Chen WST, Huang CY. Estimating the impact of drug addiction causes heart damage. Drug Chem Toxicol 2023; 46:1044-1050. [PMID: 36216784 DOI: 10.1080/01480545.2022.2122984] [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: 04/06/2022] [Revised: 07/26/2022] [Accepted: 09/01/2022] [Indexed: 11/03/2022]
Abstract
To date, few studies have investigated the toxicological effects of the combined use of amphetamine and heroin in the heart. Hence, the aim of this study was to identify indicators for clinical evaluation and prevention of cardiac injury induced by the combined use of amphetamine and heroin. Four different groups were analyzed: (1) normal group (n=25;average age=35 ± 6.8); (2) heart disease group (n=25;average age=58 ± 17.2); (3) drug abusers (n = 27; average age = 37 ± 7.7); (4) drug abstainers (previous amphetamine-heroin users who had been drug-free for more than two weeks; n = 22; average age = 35 ± 5.6). The activity of MMPs, and levels of TNF-α, IL-6, GH, IGF-I, and several serum biomarkers were examined to evaluate the impact of drug abuse on the heart. The selected plasma biomarkers and classic cardiac biomarkers were significantly increased compared to the normal group. The zymography data showed the changes in cardiac-remodeling enzymes MMP-9 and MMP-2 among combined users of amphetamine and heroin. The levels of TNF-α and IL-6 only increased in the heart disease group. Growth hormone was increased; however, IGF-I level decreased with drug abuse and the level was not restored by abstinence. We speculated that the amphetamine-heroin users might pose risk to initiate heart disease even though the users abstained for more than two weeks. The activity change of MMP-9 and MMP-2 can be a direct reason affecting heart function. The indirect reason may be related to liver damage by drug abuse reduce IGF-1 production to protect heart function.
Collapse
Affiliation(s)
- Sheng-Huang Chang
- Department of Health, Executive Yuan, Tsao-Tun Psychiatric Center, Nantou, Taiwan
| | - Pei-Ying Pai
- Division of Cardiology, China Medical University Hospital, Taichung, Taiwan
| | - Chiung-Hung Hsu
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Shibu Marthandam Asokan
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Bruce Chi-Kang Tsai
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Wen-Tsan Weng
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
- Ph.D. Program for Biotechnology Industry, China Medical University, Taichung, Taiwan
| | - Tzu-Ching Shih
- Department of Biomedical Imaging and Radiological Science College of Medicine, China Medical University, Taichung, Taiwan
| | - Hui-Chuan Kao
- Department of Public Health, Tzu Chi University, Hualien, Taiwan
| | - William Shao-Tsu Chen
- Department of Psychiatry, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Department of Human Development and Psychology, Tzu Chi University, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan
- Center of General Education, Tzu Chi University of Science and Technology, Hualien, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
13
|
Miller EJ, Khoshbouei H. Immunity on ice: The impact of methamphetamine on peripheral immunity. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2023; 99:217-250. [PMID: 38467482 DOI: 10.1016/bs.apha.2023.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Methamphetamine (METH) regulation of the dopamine transporter (DAT) and central nervous system (CNS) dopamine transmission have been extensively studied. However, our understanding of how METH influences neuroimmune communication and innate and adaptive immunity is still developing. Recent studies have shed light on the bidirectional communication between the CNS and the peripheral immune system. They have established a link between CNS dopamine levels, dopamine neuronal activity, and peripheral immunity. Akin to dopamine neurons in the CNS, a majority of peripheral immune cells also express DAT, implying that in addition to their effect in the CNS, DAT ligands such as methamphetamine may have a role in modulating peripheral immunity. For example, by directly influencing DAT-expressing peripheral immune cells and thus peripheral immunity, METH can trigger a feed-forward cascade that impacts the bidirectional communication between the CNS and peripheral immune system. In this review, we aim to discuss the current understanding of how METH modulates both innate and adaptive immunity and identify areas where knowledge gaps exist. These gaps will then be considered in guiding future research directions.
Collapse
Affiliation(s)
- Emily J Miller
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States.
| | - Habibeh Khoshbouei
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States.
| |
Collapse
|
14
|
Zanfino G, Puzzo C, de Laurenzi V, Adriani W. Characterization of Behavioral Phenotypes in Heterozygous DAT Rat Based on Pedigree. Biomedicines 2023; 11:2565. [PMID: 37761006 PMCID: PMC10526166 DOI: 10.3390/biomedicines11092565] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Dopamine is an essential neurotransmitter whose key roles include movement control, pleasure and reward, attentional and cognitive skills, and regulation of the sleep/wake cycle. Reuptake is carried out by the dopamine transporter (DAT; DAT1 SLC6A3 gene). In order to study the effects of hyper-dopaminergia syndrome, the gene was silenced in rats. DAT-KO rats show stereotypical behavior, hyperactivity, a deficit in working memory, and an altered circadian cycle. In addition to KO rats, heterozygous (DAT-HET) rats show relative hypofunction of DAT; exact phenotypic effects are still unknown and may depend on whether the sire or the dam was KO. Our goal was to elucidate the potential importance of the parental origin of the healthy or silenced allele and its impact across generations, along with the potential variations in maternal care. We thus generated specular lines to study the effects of (grand) parental roles in inheriting the wild or mutated allele. MAT-HETs are the progeny of a KO sire and a WT dam; by breeding MAT-HET males and KO females, we obtained subjects with a DAT -/- epigenotype, named QULL, to reflect additional epigenetic DAT modulation when embryos develop within a hyper-dopaminergic KO uterus. We aimed to verify if any behavioral anomaly was introduced by a QULL (instead of KO) rat acting as a direct father or indirect maternal grandfather (or both). We thus followed epigenotypes obtained after three generations and observed actual effects on impaired maternal care of the offspring (based on pedigree). In particular, offspring of MAT-HET-dam × QULL-sire breeding showed a compulsive and obsessive phenotype. Although the experimental groups were all heterozygous, the impact of having a sire of epigenotype QULL (who developed in the uterus of a KO grand-dam) has emerged clearly. Along the generations, the effects of the DAT epigenotype on the obsessive/compulsive phenotype do vary as a function of the uterine impact on either allele in one's genealogical line.
Collapse
Affiliation(s)
- Gioia Zanfino
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.Z.); (C.P.)
| | - Concetto Puzzo
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.Z.); (C.P.)
- Faculty of Psychology, International Telematic University Uninettuno, 00186 Rome, Italy
| | - Vincenzo de Laurenzi
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy;
- Department of Innovative Technologies in Medicine and Dentistry, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Walter Adriani
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.Z.); (C.P.)
- Faculty of Psychology, International Telematic University Uninettuno, 00186 Rome, Italy
| |
Collapse
|
15
|
Belcher AM, Rorabaugh BR. Maternal use of methamphetamine alters cardiovascular function in the adult offspring. Biochem Cell Biol 2023; 101:198-203. [PMID: 36763967 PMCID: PMC10563036 DOI: 10.1139/bcb-2022-0349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023] Open
Abstract
Methamphetamine is one of the most commonly used illicit drugs during pregnancy. Most studies investigating the impact of maternal use of methamphetamine on children have focused on neurological outcomes. In contrast, cardiovascular outcomes in these children have not been characterized. Recent studies in rodents provide evidence that prenatal exposure to methamphetamine induces changes in cardiac gene expression, changes in the heart's susceptibility to ischemic injury, and changes in vascular function that may increase the risk of developing cardiovascular disorders later in life. Importantly, these changes are sex-dependent. This review summarizes our current understanding of how methamphetamine use during pregnancy impacts the cardiovascular function of adult offspring and highlights gaps in our knowledge of the potential cardiovascular risks associated with prenatal exposure to methamphetamine.
Collapse
Affiliation(s)
- Adam M. Belcher
- Department of Pharmaceutical Sciences, Marshall University School of Pharmacy, Huntington, WV, USA
| | - Boyd R. Rorabaugh
- Department of Pharmaceutical Sciences, Marshall University School of Pharmacy, Huntington, WV, USA
- Department of Biomedical Sciences, Marshall University School of Medicine, Huntingotn, WV, USA
| |
Collapse
|
16
|
Bagalkot T, Sorkin A. Endocytic down-regulation of the striatal dopamine transporter by amphetamine in sensitized mice in sex-dependent manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.17.541165. [PMID: 37293021 PMCID: PMC10245703 DOI: 10.1101/2023.05.17.541165] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Dopamine transporter (DAT) controls dopamine signaling in the brain through the reuptake of synaptically released dopamine. DAT is a target of abused psychostimulants such as amphetamine (Amph). Acute Amph is proposed to cause transient DAT endocytosis which among other Amph effects on dopaminergic neurons elevates extracellular dopamine. However, the effects of repeated Amph abuse, leading to behavioral sensitization and drug addiction, on DAT traffic are unknown. Hence, we developed a 14-day Amph-sensitization protocol in knock-in mice expressing HA-epitope tagged DAT (HA-DAT) and investigated effects of Amph challenge on HA-DAT in sensitized animals. Amph challenge resulted in the highest locomotor activity on day 14 in both sexes, which was however sustained for 1 hour in male but not female mice. Strikingly, significant (by 30-60%) reduction in the amount of the HA-DAT protein in striatum was observed in response to Amph challenge of sensitized males but not females. Amph reduced Vmax of dopamine transport in striatal synaptosomes of males without changing Km values. Consistently, immunofluorescence microscopy revealed a significant increase of HA-DAT co-localization with the endosomal protein VPS35 only in males. Amph-induced HA-DAT down-regulation in the striatum of sensitized mice was blocked by chloroquine, vacuolin-1 (inhibitor of PIKfive kinase), and inhibitor of Rho-associated kinases (ROCK1/2), indicative of the involvement of endocytic trafficking in DAT down-regulation. Interestingly, HA-DAT protein down-regulation was observed in nucleus accumbens and not in dorsal striatum. We propose that Amph challenge in sensitized mice leads to ROCK-dependent endocytosis and post-endocytic traffic of DAT in a brain-region-specific and sex-dependent manner.
Collapse
|
17
|
Conn KA, Zou S, Das J, Alexander S, Burne TH, Kesby JP. Activating the dorsomedial and ventral midbrain projections to the striatum differentially impairs goal-directed action in male mice. Neuropharmacology 2023; 234:109550. [PMID: 37085011 DOI: 10.1016/j.neuropharm.2023.109550] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 03/29/2023] [Accepted: 04/17/2023] [Indexed: 04/23/2023]
Abstract
The cognitive symptoms of schizophrenia are wide ranging and include impaired goal-directed action. This could be driven by an increase in dopamine transmission in the dorsomedial striatum, a pathophysiological hallmark of schizophrenia. Although commonly associated with psychotic symptoms, dopamine signalling in this region also modulates associative learning that aids in the execution of actions. To gain a better understanding of the role of subcortical dopamine in learning and decision-making, we assessed goal-directed action in male mice using the cross-species outcome-specific devaluation task (ODT). First, we administered systemic amphetamine during training to determine the impact of altered dopaminergic signaling on associative learning. Second, we used pathway-specific chemogenetic approaches to activate the dorsomedial and ventral striatal pathways (that originate in the midbrain) to separately assess learning and performance. Amphetamine treatment during learning led to a dose-dependent impairment in goal-directed action. Activation of both striatal pathways during learning also impaired performance. However, when these pathways were activated during choice, only activation of the ventral pathway impaired goal-directed action. This suggests that elevated transmission in the dorsomedial striatal pathway impairs associative learning processes that guide the goal-directed execution of actions. By contrast, elevated transmission of the ventral striatal pathway disrupts the encoding of outcome values that are important for both associative learning and choice performance. These findings highlight the differential roles of the dorsomedial and ventral inputs into the striatum in goal-directed action and provides insight into how striatal dopamine signaling may contribute to the cognitive problems in those with schizophrenia.
Collapse
Affiliation(s)
- Kyna-Anne Conn
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Simin Zou
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Joyosmita Das
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Suzy Alexander
- Queensland Centre for Mental Health Research, Wacol, QLD, 4076, Australia
| | - Thomas Hj Burne
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia; Queensland Centre for Mental Health Research, Wacol, QLD, 4076, Australia
| | - James P Kesby
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia; Queensland Centre for Mental Health Research, Wacol, QLD, 4076, Australia; QIMR Berghofer Medical Research Institute, Herston, QLD, 4029, Australia.
| |
Collapse
|
18
|
Milanesi LH, Rossato DR, Rosa JLO, D'avila LF, Metz VG, Rampelotto CR, Pereira VG, Schaffazick SR, de Bona da Silva C, Burger ME. Ferulic acid-loaded nanostructure prevents morphine reinstatement: the involvement of dopamine system, NRF2, and ΔFosB in the striatum brain area of rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023:10.1007/s00210-023-02420-w. [PMID: 36790483 DOI: 10.1007/s00210-023-02420-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/06/2023] [Indexed: 02/16/2023]
Abstract
Morphine is among the most powerful analgesics and pain-relieving agents. However, its addictive properties limit their medical use because patients may be susceptible to abuse and reinstatement. Morphine addiction occurs because of dopamine release in the mesolimbic brain area, implying in an increase in oxidative stress. Ferulic acid (FA), a phenolic phytochemical found in a variety of foods, has been reported to exert antioxidant and neuroprotective effects; however, its low bioavailability makes its nano-encapsulated form a promising alternative. This study aimed to evaluate the protective effects of a novel nanosystem with FA on morphine reinstatement and the consequent molecular neuroadaptations and oxidative status in the mesolimbic region. Rats previously exposed to morphine in conditioned place preference (CPP) paradigm were treated with ferulic acid-loaded nanocapsules (FA-Nc) or nonencapsulated FA during morphine-preference extinction. Following the treatments, animals were re-exposed to morphine to induce the reinstatement. While morphine-preference extinction was comparable among all experimental groups, FA-Nc treatment prevented morphine reinstatement. In the dorsal striatum, while morphine exposure increased lipid peroxidation (LP) and reactive species (RS), FA-Nc decreased LP and FA decreased RS levels. Morphine exposure increased the dopaminergic markers (D1R, D3R, DAT) and ΔFosB immunoreactivity in the ventral striatum; however, FA-Nc treatment decreased D1R, D3R, and ΔFosB and increased D2R, DAT, and NRF2. In conclusion, FA-Nc treatment prevented the morphine reinstatement, promoted antioxidant activity, and modified the dopaminergic neurotransmission, NRF2, and ΔFosB, what may indicate a neuroprotective and antioxidant role of this nanoformulation.
Collapse
Affiliation(s)
- Laura Hautrive Milanesi
- Programa de Pós-Graduação Em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima 1000, Santa Maria, RS, Brazil
| | | | - Jéssica Leandra Oliveira Rosa
- Programa de Pós-Graduação Em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima 1000, Santa Maria, RS, Brazil
| | - Lívia Ferraz D'avila
- Programa de Pós-Graduação Em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima 1000, Santa Maria, RS, Brazil
| | - Vinícia Garzella Metz
- Programa de Pós-Graduação Em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima 1000, Santa Maria, RS, Brazil
| | - Camila Reck Rampelotto
- Programa de Pós-Graduação Em Ciências Farmacêuticas, Universidade Federal de Santa Maria (UFSM), Av, Roraima 1000, Santa Maria, RS, Brazil
| | | | | | - Cristiane de Bona da Silva
- Programa de Pós-Graduação Em Ciências Farmacêuticas, Universidade Federal de Santa Maria (UFSM), Av, Roraima 1000, Santa Maria, RS, Brazil
| | - Marilise E Burger
- Programa de Pós-Graduação Em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima 1000, Santa Maria, RS, Brazil. .,Departamento de Fisiologia E Farmacologia, UFSM, Santa Maria, RS, 97105-900, Brazil. .,Graduation Program of Pharmacology, Physiology and Pharmacology Department, Universidade Federal de Santa Maria (UFSM), Av. Roraima 1000, 97105-900, Santa Maria, RS, Brazil.
| |
Collapse
|
19
|
Elder HJ, Varshneya NB, Walentiny DM, Beardsley PM. Amphetamines modulate fentanyl-depressed respiration in a bidirectional manner. Drug Alcohol Depend 2023; 243:109740. [PMID: 36608481 PMCID: PMC9881117 DOI: 10.1016/j.drugalcdep.2022.109740] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 12/12/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND The opioid epidemic remains one of the most pressing public health crises facing the United States. Fentanyl and related synthetic opioid agonists have largely driven the rising rates of associated overdose deaths, in part, because of their surreptitious use as substitutes for other opioids and as adulterants in psychostimulants. Deaths involving opioids typically result from lethal respiratory depression, and it is currently unknown how co-use of psychostimulants with opioids affects respiratory toxicity. Considering psychostimulant overdoses have increased over 3-fold since 2013, and half of those co-involved opioids, this is a cardinal question. METHODS Naloxone, d-amphetamine (AMPH), and (±)-methamphetamine (METH) were evaluated for their effects on basal and fentanyl-depressed respiration. Minute volume (MVb) was measured in awake, freely moving mice via whole-body plethysmography to quantify fentanyl-induced respiratory depression and its modulation by dose ranges of each test drug. RESULTS Naloxone immediately reversed respiratory depression induced by fentanyl only at the highest dose tested (10 mg/kg). Both AMPH and METH exhibited bidirectional effects on MVb under basal conditions, producing significant (p ≤ 0.05) depressions then elevations of respiration as dose increased. Under depressed conditions the bidirectional effects of AMPH and METH on respiration were exaggerated, exacerbating and then reversing fentanyl-induced depression as dose increased. CONCLUSIONS These results indicate that co-use of amphetamines with fentanyl may worsen respiratory depression, but conversely, monoaminergic components of the amphetamines may possibly be exploited to mitigate fentanyl overdose.
Collapse
Affiliation(s)
- Harrison J Elder
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Neil B Varshneya
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA; Center for Drug Evaluation and Research, Food and Drug Administration, United States Department of Health and Human Services, Silver Spring, MD, USA
| | - D Matthew Walentiny
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Patrick M Beardsley
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA; Center for Biomarker Research & Precision Medicine, Virginia Commonwealth University School of Pharmacy, Richmond, VA, USA.
| |
Collapse
|
20
|
Coliță D, Coliță CI, Hermann DM, Coliță E, Doeppner TR, Udristoiu I, Popa-Wagner A. Therapeutic Use and Chronic Abuse of CNS Stimulants and Anabolic Drugs. Curr Issues Mol Biol 2022; 44:4902-4920. [PMID: 36286048 PMCID: PMC9600088 DOI: 10.3390/cimb44100333] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 11/29/2022] Open
Abstract
The available evidence suggests that affective disorders, such as depression and anxiety, increase risk for accelerated cognitive decline and late-life dementia in aging individuals. Behavioral neuropsychology studies also showed that cognitive decline is a central feature of aging impacting the quality of life. Motor deficits are common after traumatic brain injuries and stroke, affect subjective well-being, and are linked with reduced quality of life. Currently, restorative therapies that target the brain directly to restore cognitive and motor tasks in aging and disease are available. However, the very same drugs used for therapeutic purposes are employed by athletes as stimulants either to increase performance for fame and financial rewards or as recreational drugs. Unfortunately, most of these drugs have severe side effects and pose a serious threat to the health of athletes. The use of performance-enhancing drugs by children and teenagers has increased tremendously due to the decrease in the age of players in competitive sports and the availability of various stimulants in many forms and shapes. Thus, doping may cause serious health-threatening conditions including, infertility, subdural hematomas, liver and kidney dysfunction, peripheral edema, cardiac hypertrophy, myocardial ischemia, thrombosis, and cardiovascular disease. In this review, we focus on the impact of doping on psychopathological disorders, cognition, and depression. Occasionally, we also refer to chronic use of therapeutic drugs to increase physical performance and highlight the underlying mechanisms. We conclude that raising awareness on the health risks of doping in sport for all shall promote an increased awareness for healthy lifestyles across all generations.
Collapse
Affiliation(s)
- Daniela Coliță
- Doctoral School, University of Medicine and Pharmacy “Carol Davila”, 020276 Bucharest, Romania
| | - Cezar-Ivan Coliță
- Doctoral School, University of Medicine and Pharmacy “Carol Davila”, 020276 Bucharest, Romania
- Correspondence: (C.-I.C.); (I.U.); (A.P.-W.)
| | - Dirk M. Hermann
- Chair of Vascular Neurology, Dementia and Ageing, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Eugen Coliță
- Doctoral School, University of Medicine and Pharmacy “Carol Davila”, 020276 Bucharest, Romania
| | - Thorsten R. Doeppner
- Department of Neurology, University Medical Center Göttingen, 37075 Gottingen, Germany
- Department of Neurology, University Hospital Giessen, 35394 Giessen, Germany
| | - Ion Udristoiu
- Department of Psychiatry, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Correspondence: (C.-I.C.); (I.U.); (A.P.-W.)
| | - Aurel Popa-Wagner
- Chair of Vascular Neurology, Dementia and Ageing, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
- Department of Psychiatry, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Correspondence: (C.-I.C.); (I.U.); (A.P.-W.)
| |
Collapse
|
21
|
Lycas MD, Ejdrup AL, Sørensen AT, Haahr NO, Jørgensen SH, Guthrie DA, Støier JF, Werner C, Newman AH, Sauer M, Herborg F, Gether U. Nanoscopic dopamine transporter distribution and conformation are inversely regulated by excitatory drive and D2 autoreceptor activity. Cell Rep 2022; 40:111431. [PMID: 36170827 PMCID: PMC9617621 DOI: 10.1016/j.celrep.2022.111431] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 07/22/2022] [Accepted: 09/08/2022] [Indexed: 11/30/2022] Open
Abstract
The nanoscopic organization and regulation of individual molecular components in presynaptic varicosities of neurons releasing modulatory volume neurotransmitters like dopamine (DA) remain largely elusive. Here we show, by application of several super-resolution microscopy techniques to cultured neurons and mouse striatal slices, that the DA transporter (DAT), a key protein in varicosities of dopaminergic neurons, exists in the membrane in dynamic equilibrium between an inward-facing nanodomain-localized and outward-facing unclustered configuration. The balance between these configurations is inversely regulated by excitatory drive and DA D2 autoreceptor activation in a manner dependent on Ca2+ influx via N-type voltage-gated Ca2+ channels. The DAT nanodomains contain tens of transporters molecules and overlap with nanodomains of PIP2 (phosphatidylinositol-4,5-bisphosphate) but show little overlap with D2 autoreceptor, syntaxin-1, and clathrin nanodomains. The data reveal a mechanism for rapid alterations of nanoscopic DAT distribution and show a striking link of this to the conformational state of the transporter.
Collapse
Affiliation(s)
- Matthew D Lycas
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Maersk Tower 7.5, 2200 Copenhagen, Denmark
| | - Aske L Ejdrup
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Maersk Tower 7.5, 2200 Copenhagen, Denmark
| | - Andreas T Sørensen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Maersk Tower 7.5, 2200 Copenhagen, Denmark
| | - Nicolai O Haahr
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Maersk Tower 7.5, 2200 Copenhagen, Denmark
| | - Søren H Jørgensen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Maersk Tower 7.5, 2200 Copenhagen, Denmark
| | - Daryl A Guthrie
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Jonatan F Støier
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Maersk Tower 7.5, 2200 Copenhagen, Denmark
| | - Christian Werner
- Department of Biotechnology and Biophysics, Biocenter, Julius-Maximilians-Universität Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Amy Hauck Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Markus Sauer
- Department of Biotechnology and Biophysics, Biocenter, Julius-Maximilians-Universität Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Freja Herborg
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Maersk Tower 7.5, 2200 Copenhagen, Denmark
| | - Ulrik Gether
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Maersk Tower 7.5, 2200 Copenhagen, Denmark.
| |
Collapse
|
22
|
Markowitz JS, Melchert PW. The Pharmacokinetics and Pharmacogenomics of Psychostimulants. Child Adolesc Psychiatr Clin N Am 2022; 31:393-416. [PMID: 35697392 DOI: 10.1016/j.chc.2022.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The psychostimulants-amphetamine and methylphenidate-have been in clinical use for well more than 60 years. In general, both stimulants are rapidly absorbed with relatively poor bioavailability and short half-lives. The pharmacokinetics of both stimulants are generally linear and dose proportional although substantial interindividual variability in pharmacokinetics is in evidence. Amphetamine (AMP) is highly metabolized by several oxidative enzymes forming multiple metabolites while methylphenidate (MPH) is primarily metabolized by hydrolysis to the inactive metabolite ritalinic acid. At present, pharmacogenomic testing as an aid to guide dosing and personalized treatment cannot be recommended for either agent. Few pharmacokinetically based drug-drug interactions (DDIs) have been documented for either stimulant.
Collapse
Affiliation(s)
- John S Markowitz
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL 32610-0486, USA; Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL 32610-0486, USA.
| | - Philip W Melchert
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL 32610-0486, USA
| |
Collapse
|
23
|
Rossato DR, Rosa HZ, Rosa JLO, Milanesi LH, Metz VG, D'Àvila LF, Burger ME. Tactile Stimulation in Adult Rats Modulates Dopaminergic Molecular Parameters in the Nucleus accumbens Preventing Amphetamine Relapse. Mol Neurobiol 2022; 59:5564-5573. [PMID: 35732868 PMCID: PMC9217176 DOI: 10.1007/s12035-022-02927-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 06/14/2022] [Indexed: 11/28/2022]
Abstract
Amphetamine (AMPH) is a psychostimulant drug frequently related to addiction, which is characterized by functional and molecular changes in the brain reward system, favoring relapse development, and pharmacotherapies have shown low effectiveness. Considering the beneficial influences of tactile stimulation (TS) in different diseases that affect the central nervous system (CNS), here we evaluated if TS applied in adult rats could prevent or minimize the AMPH-relapse behavior also accessing molecular neuroadaptations in the nucleus accumbens (NAc). Following AMPH conditioning in the conditioned place preference (CPP) paradigm, male rats were submitted to TS (15-min session, 3 times a day, for 8 days) during the drug abstinence period, which were re-exposed to the drug in the CPP paradigm for additional 3 days for relapse observation and molecular assessment. Our findings showed that besides AMPH relapse, TS prevented the dopamine transporter (DAT), dopamine 1 receptor (D1R), tyrosine hydroxylase (TH), mu opioid receptor (MOR) increase, and AMPH-induced delta FosB (ΔFosB). Based on these outcomes, we propose TS as a useful tool to treat psychostimulant addiction, which is subsequent to clinical studies; it could be included in detoxification programs together with pharmacotherapies and psychological treatments already conventionally established.
Collapse
Affiliation(s)
- D R Rossato
- Graduation Program in Pharmacology, Universidade Federal de Santa Maria (UFSM), Santa Maria, Rio Grande do Sul, Brazil
| | - H Z Rosa
- Graduation Program in Pharmacology, Universidade Federal de Santa Maria (UFSM), Santa Maria, Rio Grande do Sul, Brazil
| | - J L O Rosa
- Graduation Program in Pharmacology, Universidade Federal de Santa Maria (UFSM), Santa Maria, Rio Grande do Sul, Brazil
| | - L H Milanesi
- Department of Physiology and Pharmacology (UFSM), Santa Maria, Rio Grande do Sul, Brazil
| | - V G Metz
- Graduation Program in Pharmacology, Universidade Federal de Santa Maria (UFSM), Santa Maria, Rio Grande do Sul, Brazil
| | - L F D'Àvila
- Graduation Program in Pharmacology, Universidade Federal de Santa Maria (UFSM), Santa Maria, Rio Grande do Sul, Brazil
| | - M E Burger
- Graduation Program in Pharmacology, Universidade Federal de Santa Maria (UFSM), Santa Maria, Rio Grande do Sul, Brazil. .,Department of Physiology and Pharmacology (UFSM), Santa Maria, Rio Grande do Sul, Brazil.
| |
Collapse
|
24
|
The dopamine transporter antiports potassium to increase the uptake of dopamine. Nat Commun 2022; 13:2446. [PMID: 35508541 PMCID: PMC9068915 DOI: 10.1038/s41467-022-30154-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 04/18/2022] [Indexed: 12/11/2022] Open
Abstract
The dopamine transporter facilitates dopamine reuptake from the extracellular space to terminate neurotransmission. The transporter belongs to the neurotransmitter:sodium symporter family, which includes transporters for serotonin, norepinephrine, and GABA that utilize the Na+ gradient to drive the uptake of substrate. Decades ago, it was shown that the serotonin transporter also antiports K+, but investigations of K+-coupled transport in other neurotransmitter:sodium symporters have been inconclusive. Here, we show that ligand binding to the Drosophila- and human dopamine transporters are inhibited by K+, and the conformational dynamics of the Drosophila dopamine transporter in K+ are divergent from the apo- and Na+-states. Furthermore, we find that K+ increases dopamine uptake by the Drosophila dopamine transporter in liposomes, and visualize Na+ and K+ fluxes in single proteoliposomes using fluorescent ion indicators. Our results expand on the fundamentals of dopamine transport and prompt a reevaluation of the impact of K+ on other transporters in this pharmacologically important family. The dopamine transporter, DAT, controls dopamine signaling by facilitating its reuptake using the Na+ gradient as driving force. Here, the authors uncover that an antiport of K+ ions also contributes to setting the rate of DAT-mediated dopamine clearance.
Collapse
|
25
|
Sustkova-Fiserova M, Charalambous C, Khryakova A, Certilina A, Lapka M, Šlamberová R. The Role of Ghrelin/GHS-R1A Signaling in Nonalcohol Drug Addictions. Int J Mol Sci 2022; 23:761. [PMID: 35054944 PMCID: PMC8776007 DOI: 10.3390/ijms23020761] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 01/27/2023] Open
Abstract
Drug addiction causes constant serious health, social, and economic burden within the human society. The current drug dependence pharmacotherapies, particularly relapse prevention, remain limited, unsatisfactory, unreliable for opioids and tobacco, and even symptomatic for stimulants and cannabinoids, thus, new more effective treatment strategies are researched. The antagonism of the growth hormone secretagogue receptor type A (GHS-R1A) has been recently proposed as a novel alcohol addiction treatment strategy, and it has been intensively studied in experimental models of other addictive drugs, such as nicotine, stimulants, opioids and cannabinoids. The role of ghrelin signaling in these drugs effects has also been investigated. The present review aims to provide a comprehensive overview of preclinical and clinical studies focused on ghrelin's/GHS-R1A possible involvement in these nonalcohol addictive drugs reinforcing effects and addiction. Although the investigation is still in its early stage, majority of the existing reviewed experimental results from rodents with the addition of few human studies, that searched correlations between the genetic variations of the ghrelin signaling or the ghrelin blood content with the addictive drugs effects, have indicated the importance of the ghrelin's/GHS-R1As involvement in the nonalcohol abused drugs pro-addictive effects. Further research is necessary to elucidate the exact involved mechanisms and to verify the future potential utilization and safety of the GHS-R1A antagonism use for these drug addiction therapies, particularly for reducing the risk of relapse.
Collapse
Affiliation(s)
- Magdalena Sustkova-Fiserova
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 100 00 Prague, Czech Republic; (C.C.); (A.K.); (A.C.); (M.L.)
| | - Chrysostomos Charalambous
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 100 00 Prague, Czech Republic; (C.C.); (A.K.); (A.C.); (M.L.)
| | - Anna Khryakova
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 100 00 Prague, Czech Republic; (C.C.); (A.K.); (A.C.); (M.L.)
| | - Alina Certilina
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 100 00 Prague, Czech Republic; (C.C.); (A.K.); (A.C.); (M.L.)
| | - Marek Lapka
- Department of Pharmacology, Third Faculty of Medicine, Charles University, Ruska 87, 100 00 Prague, Czech Republic; (C.C.); (A.K.); (A.C.); (M.L.)
| | - Romana Šlamberová
- Department of Physiology, Third Faculty of Medicine, Charles University, Ke Karlovu 4, 120 00 Prague, Czech Republic;
| |
Collapse
|
26
|
Xu L, Chen LY. Effects of the N-terminal dynamics on the conformational states of human dopamine transporter. Biophys Chem 2022; 283:106765. [PMID: 35101818 PMCID: PMC8898274 DOI: 10.1016/j.bpc.2022.106765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 11/02/2022]
Abstract
Dopamine transporter mediates the neurotransmitter dopamine homeostasis in a sodium-dependent manner. The transport process involves an alternating access of a substrate to the extracellular and intracellular spaces, which is associated with different conformational states of the transporter. However, the underlying mechanism of modulation of the state transition remains elusive. Here we present a computational simulation study of human dopamine transporter to explore its two end states (outward-facing open and inward-facing open) that have not been determined experimentally. We show that the full-length transporter may tend to adopt the inward-facing open state in its free state. The binding of an amphetamine may not trap the transporter in the outward-facing open state with increasing length of the N-terminal. Furthermore, we identify distinct patterns in the interaction networks between the N-terminal and the intracellular region that could stabilize the state of the transporter, independent of substrate binding and phosphorylation. Our results reveal the essential role of the N-terminal dynamics in modulating the functional states of the dopamine transporter, providing molecular insights into the coupling of conformational transition and substrate passage in neurotransmitter transporters.
Collapse
|
27
|
Krief S, Berrebi‐Bertrand I, Nagmar I, Giret M, Belliard S, Perrin D, Uguen M, Robert P, Lecomte J, Schwartz J, Finance O, Ligneau X. Pitolisant, a wake-promoting agent devoid of psychostimulant properties: Preclinical comparison with amphetamine, modafinil, and solriamfetol. Pharmacol Res Perspect 2021; 9:e00855. [PMID: 34423920 PMCID: PMC8381683 DOI: 10.1002/prp2.855] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 01/04/2023] Open
Abstract
Several therapeutic options are currently available to treat excessive daytime sleepiness (EDS) in patients suffering from narcolepsy or obstructive sleep apnea. However, there are no comparisons between the various wake-promoting agents in terms of mechanism of action, efficacy, or safety. The goal of this study was to compare amphetamine, modafinil, solriamfetol, and pitolisant at their known primary pharmacological targets, histamine H3 receptors (H3R), dopamine, norepinephrine, and serotonin transporters, and in various in vivo preclinical models in relation to neurochemistry, locomotion, behavioral sensitization, and food intake. Results confirmed that the primary pharmacological effect of amphetamine, modafinil, and solriamfetol was to increase central dopamine neurotransmission, in part by inhibiting its transporter. Furthermore, solriamfetol increased levels of extracellular dopamine in the nucleus accumbens, and decreased the 3,4-dihydroxyphenyl acetic acid (DOPAC)/DA ratio in the striatum, as reported for modafinil and amphetamine. All these compounds produced hyperlocomotion, behavioral sensitization, and hypophagia, which are common features of psychostimulants and of compounds with abuse potential. In contrast, pitolisant, a selective and potent H3R antagonist/inverse agonist that promotes wakefulness, had no effect on striatal dopamine, locomotion, or food intake. In addition, pitolisant, devoid of behavioral sensitization by itself, attenuated the hyperlocomotion induced by either modafinil or solriamfetol. Therefore, pitolisant presents biochemical, neurochemical, and behavioral profiles different from those of amphetamine and other psychostimulants such as modafinil or solriamfetol. In conclusion, pitolisant is a differentiated therapeutic option, when compared with psychostimulants, for the treatment of EDS, as this agent does not show any amphetamine-like properties within in vivo preclinical models.
Collapse
|
28
|
Aguilar JI, Cheng MH, Font J, Schwartz AC, Ledwitch K, Duran A, Mabry SJ, Belovich AN, Zhu Y, Carter AM, Shi L, Kurian MA, Fenollar-Ferrer C, Meiler J, Ryan RM, Mchaourab HS, Bahar I, Matthies HJ, Galli A. Psychomotor impairments and therapeutic implications revealed by a mutation associated with infantile Parkinsonism-Dystonia. eLife 2021; 10:68039. [PMID: 34002696 PMCID: PMC8131106 DOI: 10.7554/elife.68039] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/02/2021] [Indexed: 12/30/2022] Open
Abstract
Parkinson disease (PD) is a progressive, neurodegenerative disorder affecting over 6.1 million people worldwide. Although the cause of PD remains unclear, studies of highly penetrant mutations identified in early-onset familial parkinsonism have contributed to our understanding of the molecular mechanisms underlying disease pathology. Dopamine (DA) transporter (DAT) deficiency syndrome (DTDS) is a distinct type of infantile parkinsonism-dystonia that shares key clinical features with PD, including motor deficits (progressive bradykinesia, tremor, hypomimia) and altered DA neurotransmission. Here, we define structural, functional, and behavioral consequences of a Cys substitution at R445 in human DAT (hDAT R445C), identified in a patient with DTDS. We found that this R445 substitution disrupts a phylogenetically conserved intracellular (IC) network of interactions that compromise the hDAT IC gate. This is demonstrated by both Rosetta molecular modeling and fine-grained simulations using hDAT R445C, as well as EPR analysis and X-ray crystallography of the bacterial homolog leucine transporter. Notably, the disruption of this IC network of interactions supported a channel-like intermediate of hDAT and compromised hDAT function. We demonstrate that Drosophila melanogaster expressing hDAT R445C show impaired hDAT activity, which is associated with DA dysfunction in isolated brains and with abnormal behaviors monitored at high-speed time resolution. We show that hDAT R445C Drosophila exhibit motor deficits, lack of motor coordination (i.e. flight coordination) and phenotypic heterogeneity in these behaviors that is typically associated with DTDS and PD. These behaviors are linked with altered dopaminergic signaling stemming from loss of DA neurons and decreased DA availability. We rescued flight coordination with chloroquine, a lysosomal inhibitor that enhanced DAT expression in a heterologous expression system. Together, these studies shed some light on how a DTDS-linked DAT mutation underlies DA dysfunction and, possibly, clinical phenotypes shared by DTDS and PD.
Collapse
Affiliation(s)
- Jenny I Aguilar
- Department of Pharmacology, Vanderbilt University, Nashville, United States.,Department of Surgery, University of Alabama at Birmingham, Birmingham, United States
| | - Mary Hongying Cheng
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, United States
| | - Josep Font
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Alexandra C Schwartz
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, United States
| | - Kaitlyn Ledwitch
- Center for Structural Biology, Vanderbilt University, Nashville, United States.,Department of Chemistry, Vanderbilt University, Nashville, United States
| | - Amanda Duran
- Center for Structural Biology, Vanderbilt University, Nashville, United States.,Department of Chemistry, Vanderbilt University, Nashville, United States
| | - Samuel J Mabry
- Department of Surgery, University of Alabama at Birmingham, Birmingham, United States
| | - Andrea N Belovich
- Department of Biomedical Sciences, Idaho College of Osteopathic Medicine, Meridian, United States
| | - Yanqi Zhu
- Department of Surgery, University of Alabama at Birmingham, Birmingham, United States
| | - Angela M Carter
- Department of Surgery, University of Alabama at Birmingham, Birmingham, United States
| | - Lei Shi
- Computational Chemistry and Molecular Biophysics Section, NIDA, NIH, Baltimore, United States
| | - Manju A Kurian
- Molecular Neurosciences, Developmental Neurosciences, University College London (UCL), London, United Kingdom
| | | | - Jens Meiler
- Center for Structural Biology, Vanderbilt University, Nashville, United States.,Department of Chemistry, Vanderbilt University, Nashville, United States.,Institute for Drug Discovery, Leipzig University Medical School, Leipzig, Germany
| | - Renae Monique Ryan
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Hassane S Mchaourab
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, United States
| | - Ivet Bahar
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, United States
| | - Heinrich Jg Matthies
- Department of Surgery, University of Alabama at Birmingham, Birmingham, United States
| | - Aurelio Galli
- Department of Surgery, University of Alabama at Birmingham, Birmingham, United States.,Center for Inter-systemic Networks and Enteric Medical Advances, University of Alabama at Birmingham, Birmingham, United States
| |
Collapse
|
29
|
Docherty JR, Alsufyani HA. Cardiovascular and temperature adverse actions of stimulants. Br J Pharmacol 2021; 178:2551-2568. [PMID: 33786822 DOI: 10.1111/bph.15465] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/22/2021] [Accepted: 03/10/2021] [Indexed: 12/11/2022] Open
Abstract
The vast majority of illicit stimulants act at monoaminergic systems, causing both psychostimulant and adverse effects. Stimulants can interact as substrates or antagonists at the nerve terminal monoamine transporter that mediates the reuptake of monoamines across the nerve synaptic membrane and at the vesicular monoamine transporter (VMAT-2) that mediates storage of monoamines in vesicles. Stimulants can act directly at presynaptic or postsynaptic receptors for monoamines or have indirect monoamine-mimetic actions due to the release of monoamines. Cocaine and other stimulants can acutely increase the risk of sudden cardiac death. Stimulants, particularly MDMA, in hot conditions, such as that occurring at a "rave," have caused fatalities from the consequences of hyperthermia, often compounding cardiac adverse actions. This review examines the pharmacology of the cardiovascular and temperature adverse actions of stimulants.
Collapse
Affiliation(s)
- James R Docherty
- Department of Physiology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Hadeel A Alsufyani
- Department of Physiology, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
30
|
Jîtcă G, Ősz BE, Tero-Vescan A, Vari CE. Psychoactive Drugs-From Chemical Structure to Oxidative Stress Related to Dopaminergic Neurotransmission. A Review. Antioxidants (Basel) 2021; 10:381. [PMID: 33806320 PMCID: PMC8000782 DOI: 10.3390/antiox10030381] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/24/2021] [Accepted: 03/01/2021] [Indexed: 12/26/2022] Open
Abstract
Nowadays, more and more young people want to experience illegal, psychoactive substances, without knowing the risks of exposure. Besides affecting social life, psychoactive substances also have an important effect on consumer health. We summarized and analyzed the published literature data with reference to the mechanism of free radical generation and the link between chemical structure and oxidative stress related to dopaminergic neurotransmission. This review presents data on the physicochemical properties, on the ability to cross the blood brain barrier, the chemical structure activity relationship (SAR), and possible mechanisms by which neuronal injuries occur due to oxidative stress as a result of drug abuse such as "bath salts", amphetamines, or cocaine. The mechanisms of action of ingested compounds or their metabolites involve intermediate steps in which free radicals are generated. The brain is strongly affected by the consumption of such substances, facilitating the induction of neurodegenerative diseases. It can be concluded that neurotoxicity is associated with drug abuse. Dependence and oxidative stress are linked to inhibition of neurogenesis and the onset of neuronal death. Understanding the pathological mechanisms following oxidative attack can be a starting point in the development of new therapeutic targets.
Collapse
Affiliation(s)
- George Jîtcă
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania; (G.J.); (C.E.V.)
| | - Bianca E. Ősz
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania; (G.J.); (C.E.V.)
| | - Amelia Tero-Vescan
- Department of Biochemistry, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania;
| | - Camil E. Vari
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania; (G.J.); (C.E.V.)
| |
Collapse
|
31
|
Colucci P, Santori A, Romanelli L, Zwergel C, Mai A, Scaccianoce S, Campolongo P. Amphetamine Modulation of Long-Term Object Recognition Memory in Rats: Influence of Stress. Front Pharmacol 2021; 12:644521. [PMID: 33716754 PMCID: PMC7943736 DOI: 10.3389/fphar.2021.644521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 01/25/2021] [Indexed: 11/13/2022] Open
Abstract
Amphetamine is a potent psychostimulant that increases brain monoamine levels. Extensive evidence demonstrated that norepinephrine is crucially involved in the regulation of memory consolidation for stressful experiences. Here, we investigated amphetamine effects on the consolidation of long-term recognition memory in rats exposed to different intensities of forced swim stress immediately after training. Furthermore, we evaluated whether such effects are dependent on the activation of the peripheral adrenergic system. To this aim, male adult Sprague Dawley rats were subjected to an object recognition task and intraperitoneally administered soon after training with amphetamine (0.5 or 1 mg/kg), or its corresponding vehicle. Rats were thereafter exposed to a mild (1 min, 25 ± 1°C) or strong (5 min, 19 ± 1°C) forced swim stress procedure. Recognition memory retention was assessed 24-h after training. Our findings showed that amphetamine enhances the consolidation of memory in rats subjected to mild stress condition, while it impairs long-term memory performance in rats exposed to strong stress. These dichotomic effects is dependent on stress-induced activation of the peripheral adrenergic response.
Collapse
Affiliation(s)
- Paola Colucci
- Dept. of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,Neurobiology of Behavior Laboratory, Section of Neuropsychopharmacology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Alessia Santori
- Dept. of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,Neurobiology of Behavior Laboratory, Section of Neuropsychopharmacology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Luca Romanelli
- Dept. of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Clemens Zwergel
- Dept. of Drug Chemistry & Technologies, Sapienza University of Rome, Rome, Italy
| | - Antonello Mai
- Dept. of Drug Chemistry & Technologies, Sapienza University of Rome, Rome, Italy
| | - Sergio Scaccianoce
- Dept. of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Patrizia Campolongo
- Dept. of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,Neurobiology of Behavior Laboratory, Section of Neuropsychopharmacology, IRCCS Santa Lucia Foundation, Rome, Italy
| |
Collapse
|
32
|
Adjunctive Approaches to Aphasia Rehabilitation: A Review on Efficacy and Safety. Brain Sci 2021; 11:brainsci11010041. [PMID: 33401678 PMCID: PMC7823462 DOI: 10.3390/brainsci11010041] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/24/2020] [Accepted: 12/31/2020] [Indexed: 12/14/2022] Open
Abstract
Aphasia is one of the most socially disabling post-stroke deficits. Although traditional therapies have been shown to induce adequate clinical improvement, aphasic symptoms often persist. Therefore, unconventional rehabilitation techniques which act as a substitute or as an adjunct to traditional approaches are urgently needed. The present review provides an overview of the efficacy and safety of the principal approaches which have been proposed over the last twenty years. First, we examined the effectiveness of the pharmacological approach, principally used as an adjunct to language therapy, reporting the mechanism of action of each single drug for the recovery of aphasia. Results are conflicting but promising. Secondly, we discussed the application of Virtual Reality (VR) which has been proven to be useful since it potentiates the ecological validity of the language therapy by using virtual contexts which simulate real-life everyday contexts. Finally, we focused on the use of Transcranial Direct Current Stimulation (tDCS), both discussing its applications at the cortical level and highlighting a new perspective, which considers the possibility to extend the use of tDCS over the motor regions. Although the review reveals an extraordinary variability among the different studies, substantial agreement has been reached on some general principles, such as the necessity to consider tDCS only as an adjunct to traditional language therapy.
Collapse
|
33
|
Sonneborn A, Greene RW. Norepinephrine transporter antagonism prevents dopamine-dependent synaptic plasticity in the mouse dorsal hippocampus. Neurosci Lett 2021; 740:135450. [PMID: 33127445 PMCID: PMC7725138 DOI: 10.1016/j.neulet.2020.135450] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/14/2020] [Accepted: 10/08/2020] [Indexed: 12/13/2022]
Abstract
The rodent dorsal hippocampus is essential for episodic memory consolidation, a process heavily modulated by dopamine D1-like receptor (D1/5R) activation. It was previously thought that the ventral tegmental area provided the only supply of dopamine release to dorsal hippocampus, but several recent studies have established the locus coeruleus (LC) as the major source for CA1. Here we show that selective blockade of the norepinephrine transporter (NET) prevents dopamine-dependent, late long-term synaptic potentiation (LTP) in dorsal CA1, a neural correlate of memory formation that relies on LC-mediated activation of D1/5Rs. Since dopamine activation of D1/5Rs by vesicular release is expected to be enhanced by NET antagonism, our data identify NET reversal as a plausible mechanism for LC-mediated DA release. We also show that genetic deletion of LC NMDA receptors (NMDARs) blocks D1R-mediated LTP, suggesting the requirement of both a functional NET and presynaptic NMDARs for this release. As LC activity is highly correlated with attentional processes and memory, these experiments provide insight into how selective attention influences memory formation at the synaptic and circuit levels.
Collapse
Affiliation(s)
- Alex Sonneborn
- Department of Psychiatry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75205, USA; Department of Neuroscience, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75205, USA.
| | - Robert W Greene
- Department of Psychiatry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75205, USA; Department of Neuroscience, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75205, USA.
| |
Collapse
|
34
|
Loser D, Schaefer J, Danker T, Möller C, Brüll M, Suciu I, Ückert AK, Klima S, Leist M, Kraushaar U. Human neuronal signaling and communication assays to assess functional neurotoxicity. Arch Toxicol 2021; 95:229-252. [PMID: 33269408 PMCID: PMC7811517 DOI: 10.1007/s00204-020-02956-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/16/2020] [Indexed: 01/08/2023]
Abstract
Prediction of drug toxicity on the human nervous system still relies mainly on animal experiments. Here, we developed an alternative system allowing assessment of complex signaling in both individual human neurons and on the network level. The LUHMES cultures used for our approach can be cultured in 384-well plates with high reproducibility. We established here high-throughput quantification of free intracellular Ca2+ concentrations [Ca2+]i as broadly applicable surrogate of neuronal activity and verified the main processes by patch clamp recordings. Initially, we characterized the expression pattern of many neuronal signaling components and selected the purinergic receptors to demonstrate the applicability of the [Ca2+]i signals for quantitative characterization of agonist and antagonist responses on classical ionotropic neurotransmitter receptors. This included receptor sub-typing and the characterization of the anti-parasitic drug suramin as modulator of the cellular response to ATP. To exemplify potential studies on ion channels, we characterized voltage-gated sodium channels and their inhibition by tetrodotoxin, saxitoxin and lidocaine, as well as their opening by the plant alkaloid veratridine and the food-relevant marine biotoxin ciguatoxin. Even broader applicability of [Ca2+]i quantification as an end point was demonstrated by measurements of dopamine transporter activity based on the membrane potential-changing activity of this neurotransmitter carrier. The substrates dopamine or amphetamine triggered [Ca2+]i oscillations that were synchronized over the entire culture dish. We identified compounds that modified these oscillations by interfering with various ion channels. Thus, this new test system allows multiple types of neuronal signaling, within and between cells, to be assessed, quantified and characterized for their potential disturbance.
Collapse
Affiliation(s)
- Dominik Loser
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, 72770, Reutlingen, Germany
- NMI TT GmbH, 72770, Reutlingen, Germany
- Life Sciences Faculty, Albstadt-Sigmaringen University, 72488, Sigmaringen, Germany
| | - Jasmin Schaefer
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, 72770, Reutlingen, Germany
- NMI TT GmbH, 72770, Reutlingen, Germany
| | | | - Clemens Möller
- Life Sciences Faculty, Albstadt-Sigmaringen University, 72488, Sigmaringen, Germany
| | - Markus Brüll
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany
| | - Ilinca Suciu
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany
| | - Anna-Katharina Ückert
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany
| | - Stefanie Klima
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany.
| | - Udo Kraushaar
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, 72770, Reutlingen, Germany
| |
Collapse
|
35
|
Mice with dopaminergic neuron-specific deletion of DTNBP-1 gene show blunted nucleus accumbens dopamine release and associated behaviors. Neuropharmacology 2020; 184:108440. [PMID: 33340529 DOI: 10.1016/j.neuropharm.2020.108440] [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: 03/31/2020] [Revised: 12/05/2020] [Accepted: 12/12/2020] [Indexed: 11/21/2022]
Abstract
Reduced expression of a schizophrenia-associated gene Dystrobrevin Binding Protein 1 (DTNBP1) and its protein product dysbindin-1, has been reported in the brains of schizophrenia patients. DTNBP1-null mutant Sdy (Sandy) mice exhibit several behavioral features relevant to schizophrenia. Changes in dopaminergic as well as glutamatergic and GABAergic neurotransmission in cortico-limbic regions have been reported in Sdy mice. Since dysbindin-1 is expressed in multiple brain regions, it is not known whether dopamine (DA) changes observed in Sdy null mutants are due to dysbindin-1 deficiency in DAergic neurons specifically. Here, using a mouse line with conditional knockout (cKO) of DTNBP1 in DA neurons, we studied the effects of dysbindin-1 deficiency on DA release and DA-dependent behaviors. Spontaneous locomotor activity of cKO mice in novel environment was significantly reduced initially but was comparable at later time points with littermate controls. However, the locomotion-enhancing effect of a low dose of d-amphetamine (d-AMPH; 2.5 mg/kg, ip) was significantly attenuated in the cKO mice suggesting a dampened mesolimbic DA transmission. Similarly, the prepulse inhibition disrupting effect of d-AMPH was found to be significantly reduced in the mutant mice. No significant differences between the cKO and control mice were observed in tests of anxiety, spatial learning and memory and social interaction. In- vivo microdialysis in the nucleus accumbens (NAc) showed a decrease in d-AMPH-induced extracellular DA release in the cKO mice. No significant alterations in protein levels of DA transporter, phosphorylated CaM kinase-II or Akt308 in the NAc were observed in the cKO mice. Taken together, our data suggest an important role of dysbindin-1 in maintaining mesolimbic DA tone and call for further investigations identifying mechanisms linking dysbindin-1, DA and schizophrenia.
Collapse
|
36
|
Sun J, Chen F, Chen C, Zhang Z, Zhang Z, Tian W, Yu J, Wang K. Intestinal mRNA expression profile and bioinformatics analysis in a methamphetamine-induced mouse model of inflammatory bowel disease. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1669. [PMID: 33490181 PMCID: PMC7812166 DOI: 10.21037/atm-20-7741] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Background Methamphetamine use has become a serious global public health problem and puts increasing burdens on healthcare services. Abdominal complications caused by methamphetamine use are uncommon and often go ignored by clinicians. The exact intestinal pathological alterations and transcriptomic responses associated with methamphetamine use are not well understood. This study sought to investigate the transcriptome in a methamphetamine-induced mouse model of inflammatory bowel disease (IBD) using next-generation RNA sequencing. Methods Tissues from the ileum of methamphetamine-treated mice (n=5) and control mice (n=5) were dissected, processed and applied to RNA-sequencing. Bioinformatics and histopathological analysis were then performed. The expression profiles of intestinal tissue samples were analyzed and their expression profiles were integrated to obtain the differentially expressed genes and analyzed using bioinformatics. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses of the differentially expressed genes were performed using Metascape. Results A total of 326 differentially expressed genes were identified; of these genes, 120 were upregulated and 206 were downregulated. The Gene Ontology analysis showed that the biological processes of the differentially expressed genes were focused primarily on the regulation of cellular catabolic processes, endocytosis, and autophagy. The main cellular components included the endoplasmic and endocytic vesicles, cytoskeleton, adherens junctions, focal adhesions, cell body, and lysosomes. Molecular functions included protein transferase, GTPase and proteinase activities, actin-binding, and protein-lipid complex binding. The Kyoto Encyclopedia of Genes and Genomes pathway analysis showed that the differentially expressed genes were mainly involved in bacterial invasion of epithelial cells, protein processing in the endoplasmic reticulum, regulation of the actin cytoskeleton, and T-cell receptor signaling pathways. A set of overlapping genes between IBD and methamphetamine-treated intestinal tissues was discovered. Conclusions The present study is the first to analyze intestinal samples from methamphetamine-treated mice using high-throughput RNA sequencing. This study revealed key molecules that might be involved in the pathogenesis of a special type of methamphetamine-induced IBD. These results offer new insights into the relationship between methamphetamine abuse and IBD.
Collapse
Affiliation(s)
- Jiaxue Sun
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China.,Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, China.,The Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Fengrong Chen
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China.,The Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Cheng Chen
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China.,Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, China.,The Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zherui Zhang
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China.,Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, China.,The Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zunyue Zhang
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China.,The Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Weiwei Tian
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China.,The Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Juehua Yu
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China.,The Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Kunhua Wang
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
37
|
Amphetamine-induced alteration to gaze parameters: A novel conceptual pathway and implications for naturalistic behavior. Prog Neurobiol 2020; 199:101929. [PMID: 33091542 DOI: 10.1016/j.pneurobio.2020.101929] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 09/03/2020] [Accepted: 10/08/2020] [Indexed: 12/25/2022]
Abstract
Amphetamine produces a multiplicity of well-documented end-order biochemical, pharmacological and biobehavioural effects. Mechanistically, amphetamine downregulates presynaptic and postsynaptic striatal monoamine (primarily dopaminergic) systems, producing alterations to key brain regions which manifest as stereotyped ridged behaviour which occurs under both acute and chronic dosing schedules and persists beyond detoxification. Despite evidence of amphetamine-induced visual attentional dysfunction, no conceptual synthesis has yet captured how characteristic pharmaco-behavioural processes are critically implicated via these pathways, nor described the potential implications for safety-sensitive behaviours. Drawing on known pathomechanisms, we propose a cross-disciplinary, novel conceptual functional system framework for delineating the biobehavioural consequences of amphetamine use on visual attentional capacity and discuss the implications for functional and behavioural outcomes. Specifically, we highlight the manifest implications for behaviours that are conceptually driven and highly dependent on visual information processing for timely execution of visually-guided movements. Following this, we highlight the potential impact on safety-sensitive, but common behaviours, such as driving a motor vehicle. The close pathophysiological relationship between oculomotor control and higher-order cognitive processes further suggests that dynamic measurement of movement related to the motion of the eye (gaze behaviour) may be a simple, effective and direct measure of behavioural performance capabilities in naturalistic settings. Consequently, we discuss the potential efficacy of ocular monitoring for the detection and monitoring of driver states for this drug user group, and potential wider application. Significance statement: We propose a novel biochemical-physiological-behavioural pathway which delineates how amphetamine use critically alters oculomotor function, visual-attentional performance and information processing capabilities. Given the manifest implications for behaviours that are conceptually driven and highly dependent on these processes, we recommend oculography as a novel means of detecting and monitoring gaze behaviours during naturalistic tasks such as driving. Real-word examination of gaze behaviour therefore present as an effective means to detect driver impairment and prevent performance degradation due to these drugs.
Collapse
|
38
|
Liu P, Lei G, Chu Z, Deng LS, Yang L, He JL, Dang YH. The role of HINT1 in methamphetamine-induced behavioral sensitization. Psychopharmacology (Berl) 2020; 237:2345-2351. [PMID: 32430517 DOI: 10.1007/s00213-020-05538-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 04/27/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Drug addiction is a chronically relapsing disorder in humans yet the underlying mechanism remained unclear. Recent studies suggested that histidine triad nucleotide binding protein1 (HINT1) may play significant roles in diverse neuropsychiatric diseases including drug addiction. METHODS In the current study, we used different batches of mice to establish different stages of methamphetamine (METH)-induced behavioral sensitization (BS) to explore the dynamic changes throughout the process of addiction in different brain regions, including the prefrontal cortex (PFC), nucleus accumbens (NAc), caudate putamen (CPu), and hippocampus (Hip). In addition, we used HINT1 knockout (KO) mice to investigate the effect of HINT1 protein deletion on METH-induced BS. RESULTS We found that in PFC of the METH group mice, the HINT1 expression level initially increased after development phase, and then dropped to the normal level during expression phase. However, there was no statistical difference in the HINT1 expression level in the other three encephalic regions (NAc, CPu, and Hip). The absence of HINT1 could promote METH-mediated addictive behavior to a certain extent, while the significant difference between genotypes only occurred in the development phase. CONCLUSIONS Using the new technique, hip fractures were correctly predicted in 78% of cases compared with 36% when using the T-score. The accuracy of the prediction was not greatly reduced when using SSM and SAM (78% and 74% correct, respectively). Various geometric and BMD distribution traits were identified in the fractured and non-fractured groups.
Collapse
Affiliation(s)
- Peng Liu
- Department of Pharmacology and Toxicology, Institute of Basic Medicine Science, Xi'an Medical University, Xi'an, 710021, China
| | - Gang Lei
- College of Medicine & Forensics, Key Laboratory of the Health Ministry for Forensic Medicine, Key Laboratory of Environment and Genes Related to Diseases of the Education Ministry, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Zheng Chu
- College of Medicine & Forensics, Key Laboratory of the Health Ministry for Forensic Medicine, Key Laboratory of Environment and Genes Related to Diseases of the Education Ministry, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Li-Sha Deng
- College of Medicine & Forensics, Key Laboratory of the Health Ministry for Forensic Medicine, Key Laboratory of Environment and Genes Related to Diseases of the Education Ministry, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Liu Yang
- College of Medicine & Forensics, Key Laboratory of the Health Ministry for Forensic Medicine, Key Laboratory of Environment and Genes Related to Diseases of the Education Ministry, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Jun-Liang He
- College of Medicine & Forensics, Key Laboratory of the Health Ministry for Forensic Medicine, Key Laboratory of Environment and Genes Related to Diseases of the Education Ministry, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Yong-Hui Dang
- College of Medicine & Forensics, Key Laboratory of the Health Ministry for Forensic Medicine, Key Laboratory of Environment and Genes Related to Diseases of the Education Ministry, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.
| |
Collapse
|
39
|
Koch J, Shi WX, Dashtipour K. VMAT2 inhibitors for the treatment of hyperkinetic movement disorders. Pharmacol Ther 2020; 212:107580. [PMID: 32454050 DOI: 10.1016/j.pharmthera.2020.107580] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 05/14/2020] [Indexed: 11/24/2022]
Abstract
Hyperkinetic movement disorders comprise a variety of conditions characterized by involuntary movements, which include but are not limited to tardive dyskinesia, chorea associated with Huntington's Disease, and tic disorders. The class of medications that have been used to treat these conditions includes Vesicular Monoamine Transporter-2 (VMAT2) inhibitors. In 2008, the FDA approved tetrabenazine as a treatment for chorea associated with Huntington's Disease. Optimization of the pharmacology of tetrabenazine has since led to the approval of two new VMAT2 inhibitors, deutetrabenazine and valbenazine. The objective of this review is to provide background on the role of VMAT in monoamine neurotransmission, the mechanism of VMAT2 inhibition on the treatment of hyperkinetic disorders (specifically tardive dyskinesia and chorea associated with Huntington's Disease), the pharmacology and pharmacokinetics of the commercially available VMAT2 inhibitors, and a summary of the clinical data to support application of these medications.
Collapse
Affiliation(s)
- Jessa Koch
- Pharmacy Practice, Loma Linda University School of Pharmacy, Shryock Hall, 24745 Stewart St, Loma Linda, CA 92350, United States of America; Department of Neurology, Loma Linda University School of Medicine, Faculty Medical Offices, 11370 Anderson, Suite B-100, Loma Linda, CA 92350, United States of America.
| | - Wei-Xing Shi
- Pharmaceutical, Administrative and Basic Sciences, Loma Linda University Schools of Pharmacy and Medicine, 11175 Campus St, CSP21020, Loma Linda, CA 92350, United States of America.
| | - Khashayar Dashtipour
- Pharmacy Practice, Loma Linda University School of Pharmacy, Shryock Hall, 24745 Stewart St, Loma Linda, CA 92350, United States of America; Department of Neurology, Loma Linda University School of Medicine, Faculty Medical Offices, 11370 Anderson, Suite B-100, Loma Linda, CA 92350, United States of America.
| |
Collapse
|
40
|
Heterogeneity of Striatal Dopamine Function in Schizophrenia: Meta-analysis of Variance. Biol Psychiatry 2020; 87:215-224. [PMID: 31561858 DOI: 10.1016/j.biopsych.2019.07.008] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 06/24/2019] [Accepted: 07/10/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND It has been hypothesized that dopamine function in schizophrenia exhibits heterogeneity in excess of that seen in the general population. However, no previous study has systematically tested this hypothesis. METHODS We employed meta-analysis of variance to investigate interindividual variability of striatal dopaminergic function in patients with schizophrenia and in healthy control subjects. We included 65 studies that reported molecular imaging measures of dopamine synthesis or release capacities, dopamine D2/3 receptor (D2/3R) or dopamine transporter (DAT) availabilities, or synaptic dopamine levels in 983 patients and 968 control subjects. Variability differences were quantified using variability ratio (VR) and coefficient of variation ratio. RESULTS Interindividual variability of striatal D2/3R (VR = 1.26, p < .0001) and DAT (VR = 1.31, p = .01) availabilities and synaptic dopamine levels (VR = 1.38, p = .045) but not dopamine synthesis (VR = 1.12, p = .13) or release (VR = 1.08, p = .70) capacities were significantly greater in patients than in control subjects. Findings were robust to variability measure. Mean dopamine synthesis (g = 0.65, p = .004) and release (g = 0.66, p = .03) capacities, as well as synaptic levels (g = 0.78, p = .0006), were greater in patients overall, but mean synthesis capacity did not differ from that of control subjects in treatment-resistant patients (p > .3). Mean D2/3R (g = 0.17, p = .14) and DAT (g = -0.20, p = .28) availabilities did not differ between groups. CONCLUSIONS Our findings demonstrate significant heterogeneity of striatal dopamine function in schizophrenia. They suggest that while elevated dopamine synthesis and release capacities may be core features of the disorder, altered D2/3R and DAT availabilities and synaptic dopamine levels may occur only in a subgroup of patients. This heterogeneity may contribute to variation in treatment response and side effects.
Collapse
|
41
|
Korchynska S, Krassnitzer M, Malenczyk K, Prasad RB, Tretiakov EO, Rehman S, Cinquina V, Gernedl V, Farlik M, Petersen J, Hannes S, Schachenhofer J, Reisinger SN, Zambon A, Asplund O, Artner I, Keimpema E, Lubec G, Mulder J, Bock C, Pollak DD, Romanov RA, Pifl C, Groop L, Hökfelt TGM, Harkany T. Life-long impairment of glucose homeostasis upon prenatal exposure to psychostimulants. EMBO J 2020; 39:e100882. [PMID: 31750562 PMCID: PMC6939201 DOI: 10.15252/embj.2018100882] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 10/10/2019] [Accepted: 10/21/2019] [Indexed: 12/21/2022] Open
Abstract
Maternal drug abuse during pregnancy is a rapidly escalating societal problem. Psychostimulants, including amphetamine, cocaine, and methamphetamine, are amongst the illicit drugs most commonly consumed by pregnant women. Neuropharmacology concepts posit that psychostimulants affect monoamine signaling in the nervous system by their affinities to neurotransmitter reuptake and vesicular transporters to heighten neurotransmitter availability extracellularly. Exacerbated dopamine signaling is particularly considered as a key determinant of psychostimulant action. Much less is known about possible adverse effects of these drugs on peripheral organs, and if in utero exposure induces lifelong pathologies. Here, we addressed this question by combining human RNA-seq data with cellular and mouse models of neuroendocrine development. We show that episodic maternal exposure to psychostimulants during pregnancy coincident with the intrauterine specification of pancreatic β cells permanently impairs their ability of insulin production, leading to glucose intolerance in adult female but not male offspring. We link psychostimulant action specifically to serotonin signaling and implicate the sex-specific epigenetic reprogramming of serotonin-related gene regulatory networks upstream from the transcription factor Pet1/Fev as determinants of reduced insulin production.
Collapse
Affiliation(s)
- Solomiia Korchynska
- Department of Molecular NeurosciencesCenter for Brain ResearchMedical University of ViennaViennaAustria
| | - Maria Krassnitzer
- Department of Molecular NeurosciencesCenter for Brain ResearchMedical University of ViennaViennaAustria
| | - Katarzyna Malenczyk
- Department of Molecular NeurosciencesCenter for Brain ResearchMedical University of ViennaViennaAustria
| | - Rashmi B Prasad
- Department of Clinical Sciences, Diabetes and Endocrinology CRCSkåne University Hospital MalmöMalmöSweden
| | - Evgenii O Tretiakov
- Department of Molecular NeurosciencesCenter for Brain ResearchMedical University of ViennaViennaAustria
| | - Sabah Rehman
- Department of Molecular NeurosciencesCenter for Brain ResearchMedical University of ViennaViennaAustria
| | - Valentina Cinquina
- Department of Molecular NeurosciencesCenter for Brain ResearchMedical University of ViennaViennaAustria
| | - Victoria Gernedl
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Matthias Farlik
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Julian Petersen
- Department of Molecular NeurosciencesCenter for Brain ResearchMedical University of ViennaViennaAustria
| | - Sophia Hannes
- Department of Molecular NeurosciencesCenter for Brain ResearchMedical University of ViennaViennaAustria
| | - Julia Schachenhofer
- Department of Molecular NeurosciencesCenter for Brain ResearchMedical University of ViennaViennaAustria
| | - Sonali N Reisinger
- Department of Neurophysiology and NeuropharmacologyCenter for Physiology and PharmacologyMedical University of ViennaViennaAustria
| | - Alice Zambon
- Department of Neurophysiology and NeuropharmacologyCenter for Physiology and PharmacologyMedical University of ViennaViennaAustria
| | - Olof Asplund
- Department of Clinical Sciences, Diabetes and Endocrinology CRCSkåne University Hospital MalmöMalmöSweden
| | - Isabella Artner
- Stem Cell CenterLund UniversityLundSweden
- Endocrine Cell Differentiation and FunctionLund University Diabetes CenterLund UniversityMalmöSweden
| | - Erik Keimpema
- Department of Molecular NeurosciencesCenter for Brain ResearchMedical University of ViennaViennaAustria
| | - Gert Lubec
- Paracelsus Medical UniversitySalzburgAustria
| | - Jan Mulder
- Science for Life LaboratoryKarolinska InstitutetSolnaSweden
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Daniela D Pollak
- Department of Neurophysiology and NeuropharmacologyCenter for Physiology and PharmacologyMedical University of ViennaViennaAustria
| | - Roman A Romanov
- Department of Molecular NeurosciencesCenter for Brain ResearchMedical University of ViennaViennaAustria
| | - Christian Pifl
- Department of Molecular NeurosciencesCenter for Brain ResearchMedical University of ViennaViennaAustria
| | - Leif Groop
- Department of Clinical Sciences, Diabetes and Endocrinology CRCSkåne University Hospital MalmöMalmöSweden
- Institute for Molecular Medicine Finland (FIMM)Helsinki UniversityHelsinkiFinland
| | | | - Tibor Harkany
- Department of Molecular NeurosciencesCenter for Brain ResearchMedical University of ViennaViennaAustria
- Department of NeuroscienceKarolinska InstitutetSolnaSweden
| |
Collapse
|
42
|
Colucci P, Mancini GF, Santori A, Zwergel C, Mai A, Trezza V, Roozendaal B, Campolongo P. Amphetamine and the Smart Drug 3,4-Methylenedioxypyrovalerone (MDPV) Induce Generalization of Fear Memory in Rats. Front Mol Neurosci 2019; 12:292. [PMID: 31849606 PMCID: PMC6895769 DOI: 10.3389/fnmol.2019.00292] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 11/14/2019] [Indexed: 11/13/2022] Open
Abstract
Human studies have consistently shown that drugs of abuse affect memory function. The psychostimulants amphetamine and the "bath salt" 3,4-methylenedioxypyrovalerone (MDPV) increase brain monoamine levels through a similar, yet not identical, mechanism of action. Findings indicate that amphetamine enhances the consolidation of memory for emotional experiences, but still MDPV effects on memory function are underinvestigated. Here, we tested the effects induced by these two drugs on generalization of fear memory and their relative neurobiological underpinnings. To this aim, we used a modified version of the classical inhibitory avoidance task, termed inhibitory avoidance discrimination task. According to such procedure, adult male Sprague-Dawley rats were first exposed to one inhibitory avoidance apparatus and, with a 1-min delay, to a second apparatus where they received an inescapable footshock. Forty-eight hours later, retention latencies were tested, in a randomized order, in the two training apparatuses as well as in a novel contextually modified apparatus to assess both strength and generalization of memory. Our results indicated that both amphetamine and MDPV induced generalization of fear memory, whereas only amphetamine enhanced memory strength. Co-administration of the β-adrenoceptor antagonist propranolol prevented the effects of both amphetamine and MDPV on the strength and generalization of memory. The dopaminergic receptor blocker cis-flupenthixol selectively reversed the amphetamine effect on memory generalization. These findings indicate that amphetamine and MDPV induce generalization of fear memory through different modulations of noradrenergic and dopaminergic neurotransmission.
Collapse
Affiliation(s)
- Paola Colucci
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,Neurobiology of Behavior Laboratory, Santa Lucia Foundation, Rome, Italy
| | - Giulia Federica Mancini
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,Neurobiology of Behavior Laboratory, Santa Lucia Foundation, Rome, Italy
| | - Alessia Santori
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,Neurobiology of Behavior Laboratory, Santa Lucia Foundation, Rome, Italy
| | - Clemens Zwergel
- Department of Drug Chemistry & Technologies, Sapienza University of Rome, Rome, Italy.,Department of Medicine of Precision, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Antonello Mai
- Department of Drug Chemistry & Technologies, Sapienza University of Rome, Rome, Italy
| | - Viviana Trezza
- Department of Science, Section of Biomedical Sciences and Technologies, University Roma Tre, Rome, Italy
| | - Benno Roozendaal
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, Netherlands.,Donders Institute for Brain, Cognition and Behavior, Radboud University, Nijmegen, Netherlands
| | - Patrizia Campolongo
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,Neurobiology of Behavior Laboratory, Santa Lucia Foundation, Rome, Italy
| |
Collapse
|
43
|
Kevil CG, Goeders NE, Woolard MD, Bhuiyan MS, Dominic P, Kolluru GK, Arnold CL, Traylor JG, Orr AW. Methamphetamine Use and Cardiovascular Disease. Arterioscler Thromb Vasc Biol 2019; 39:1739-1746. [PMID: 31433698 DOI: 10.1161/atvbaha.119.312461] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
While the opioid epidemic has garnered significant attention, the use of methamphetamines is growing worldwide independent of wealth or region. Following overdose and accidents, the leading cause of death in methamphetamine users is cardiovascular disease, because of significant effects of methamphetamine on vasoconstriction, pulmonary hypertension, atherosclerotic plaque formation, cardiac arrhythmias, and cardiomyopathy. In this review, we examine the current literature on methamphetamine-induced changes in cardiovascular health, discuss the potential mechanisms regulating these varied effects, and highlight our deficiencies in understanding how to treat methamphetamine-associated cardiovascular dysfunction.
Collapse
Affiliation(s)
- Christopher G Kevil
- From the Departments of Pathology and Translational Pathobiology (C.G.K., M.S.B., G.K.K., J.G.T., A.W.O.), LSU Health Sciences Center, Shreveport, LA.,Cellular and Molecular Physiology (C.G.K., M.S.B., A.W.O.), LSU Health Sciences Center, Shreveport, LA.,Cell Biology and Anatomy (C.G.K., A.W.O.), LSU Health Sciences Center, Shreveport, LA
| | - Nicholas E Goeders
- Pharmacology, Toxicology, and Neuroscience (N.E.G.), LSU Health Sciences Center, Shreveport, LA
| | - Matthew D Woolard
- Microbiology and Immunology (M.D.W.), LSU Health Sciences Center, Shreveport, LA
| | - Md Shenuarin Bhuiyan
- From the Departments of Pathology and Translational Pathobiology (C.G.K., M.S.B., G.K.K., J.G.T., A.W.O.), LSU Health Sciences Center, Shreveport, LA.,Cellular and Molecular Physiology (C.G.K., M.S.B., A.W.O.), LSU Health Sciences Center, Shreveport, LA
| | - Paari Dominic
- Medicine (P.D., C.L.A.), LSU Health Sciences Center, Shreveport, LA
| | - Gopi K Kolluru
- From the Departments of Pathology and Translational Pathobiology (C.G.K., M.S.B., G.K.K., J.G.T., A.W.O.), LSU Health Sciences Center, Shreveport, LA
| | - Connie L Arnold
- Medicine (P.D., C.L.A.), LSU Health Sciences Center, Shreveport, LA
| | - James G Traylor
- From the Departments of Pathology and Translational Pathobiology (C.G.K., M.S.B., G.K.K., J.G.T., A.W.O.), LSU Health Sciences Center, Shreveport, LA
| | - A Wayne Orr
- From the Departments of Pathology and Translational Pathobiology (C.G.K., M.S.B., G.K.K., J.G.T., A.W.O.), LSU Health Sciences Center, Shreveport, LA.,Cellular and Molecular Physiology (C.G.K., M.S.B., A.W.O.), LSU Health Sciences Center, Shreveport, LA.,Cell Biology and Anatomy (C.G.K., A.W.O.), LSU Health Sciences Center, Shreveport, LA
| |
Collapse
|
44
|
Monoamine transporters: structure, intrinsic dynamics and allosteric regulation. Nat Struct Mol Biol 2019; 26:545-556. [PMID: 31270469 DOI: 10.1038/s41594-019-0253-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/14/2019] [Indexed: 12/31/2022]
Abstract
Monoamine transporters (MATs) regulate neurotransmission via the reuptake of dopamine, serotonin and norepinephrine from extra-neuronal regions and thus maintain neurotransmitter homeostasis. As targets of a wide range of compounds, including antidepressants, substances of abuse and drugs for neuropsychiatric and neurodegenerative disorders, their mechanism of action and their modulation by small molecules have long been of broad interest. Recent advances in the structural characterization of dopamine and serotonin transporters have opened the way for structure-based modeling and simulations, which, together with experimental data, now provide mechanistic understanding of their transport function and interactions. Here we review recent progress in the elucidation of the structural dynamics of MATs and their conformational landscape and transitions, as well as allosteric regulation mechanisms.
Collapse
|
45
|
Yang G, Wu H, Pembere AMS, Luo Z. Deep Ultraviolet Laser Radiation Causes Brittle Fracture of C
α
‐C
β
Bonds in Neurotransmitters. ChemistrySelect 2019. [DOI: 10.1002/slct.201900936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Guanhua Yang
- University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Haiming Wu
- Beijing National Laboratory for Molecular Sciences (BNLMS), State Key Laboratory for Structural Chemistry of Unstable and Stable SpeciesInstitute of ChemistryChinese Academy of Sciences. Beijing 100190 P. R. China
- University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Anthony M. S. Pembere
- Beijing National Laboratory for Molecular Sciences (BNLMS), State Key Laboratory for Structural Chemistry of Unstable and Stable SpeciesInstitute of ChemistryChinese Academy of Sciences. Beijing 100190 P. R. China
- University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Zhixun Luo
- Beijing National Laboratory for Molecular Sciences (BNLMS), State Key Laboratory for Structural Chemistry of Unstable and Stable SpeciesInstitute of ChemistryChinese Academy of Sciences. Beijing 100190 P. R. China
- University of Chinese Academy of Sciences Beijing 100049 P. R. China
| |
Collapse
|
46
|
Ferrucci M, Limanaqi F, Ryskalin L, Biagioni F, Busceti CL, Fornai F. The Effects of Amphetamine and Methamphetamine on the Release of Norepinephrine, Dopamine and Acetylcholine From the Brainstem Reticular Formation. Front Neuroanat 2019; 13:48. [PMID: 31133823 PMCID: PMC6524618 DOI: 10.3389/fnana.2019.00048] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 04/26/2019] [Indexed: 12/22/2022] Open
Abstract
Amphetamine (AMPH) and methamphetamine (METH) are widely abused psychostimulants, which produce a variety of psychomotor, autonomic and neurotoxic effects. The behavioral and neurotoxic effects of both compounds (from now on defined as AMPHs) stem from a fair molecular and anatomical specificity for catecholamine-containing neurons, which are placed in the brainstem reticular formation (RF). In fact, the structural cross-affinity joined with the presence of shared molecular targets between AMPHs and catecholamine provides the basis for a quite selective recruitment of brainstem catecholamine neurons following AMPHs administration. A great amount of investigations, commentary manuscripts and books reported a pivotal role of mesencephalic dopamine (DA)-containing neurons in producing behavioral and neurotoxic effects of AMPHs. Instead, the present review article focuses on catecholamine reticular neurons of the low brainstem. In fact, these nuclei add on DA mesencephalic cells to mediate the effects of AMPHs. Among these, we also include two pontine cholinergic nuclei. Finally, we discuss the conundrum of a mixed neuronal population, which extends from the pons to the periaqueductal gray (PAG). In this way, a number of reticular nuclei beyond classic DA mesencephalic cells are considered to extend the scenario underlying the neurobiology of AMPHs abuse. The mechanistic approach followed here to describe the action of AMPHs within the RF is rooted on the fine anatomy of this region of the brainstem. This is exemplified by a few medullary catecholamine neurons, which play a pivotal role compared with the bulk of peripheral sympathetic neurons in sustaining most of the cardiovascular effects induced by AMPHs.
Collapse
Affiliation(s)
- Michela Ferrucci
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Fiona Limanaqi
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Larisa Ryskalin
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | | | | | - Francesco Fornai
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
- IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
47
|
Metz VG, Segat HJ, Dias VT, Barcelos RCS, Maurer LH, Stiebe J, Emanuelli T, Burger ME, Pase CS. Omega-3 decreases D1 and D2 receptors expression in the prefrontal cortex and prevents amphetamine-induced conditioned place preference in rats. J Nutr Biochem 2019; 67:182-189. [PMID: 30951972 DOI: 10.1016/j.jnutbio.2019.02.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 02/13/2019] [Accepted: 02/28/2019] [Indexed: 01/09/2023]
Abstract
Amphetamine (AMPH) abuse is a serious public health problem due to the high addictive potential of this drug, whose use is related to severe brain neurotoxicity and memory impairments. So far, therapies for psychostimulant addiction have had limited efficacy. Omega-3 polyunsaturated fatty acids (n-3 PUFA) have shown beneficial influences on the prevention and treatment of several diseases that affect the central nervous system. Here, we assessed the influence of fish oil (FO), which is rich in n-3 PUFA, on withdrawal and relapse symptoms following re-exposure to AMPH. Male Wistar rats received d,l-AMPH or vehicle in the conditioned place preference (CPP) paradigm for 14 days. Then, half of each experimental group was treated with FO (3 g/kg, p.o.) for 14 days. Subsequently, animals were re-exposed to AMPH-CPP for three additional days, in order to assess relapse behavior. Our findings have evidenced that FO prevented relapse induced by AMPH reconditioning. While FO prevented AMPH-induced oxidative damages in the prefrontal cortex, molecular assays allowed us to observe that it was also able to modulate dopaminergic cascade markers (DAT, TH, VMAT-2, D1R and D2R) in the same brain area, thus preventing AMPH-induced molecular changes. To the most of our knowledge, this is the first study to show a natural alternative tool which is able to prevent psychostimulant relapse following drug withdrawal. This non-invasive and healthy nutraceutical may be considered as an adjuvant treatment in detoxification clinics.
Collapse
Affiliation(s)
- Vinícia Garzella Metz
- Programa de Pós-Graduação em Farmacologia-Universidade Federal de Santa Maria, RS, Brazil
| | - Hecson Jesser Segat
- Programa de Pós-Graduação em Bioquímica Toxicológica - Universidade Federal de Santa Maria, RS, Brazil
| | - Verônica Tironi Dias
- Programa de Pós-Graduação em Farmacologia-Universidade Federal de Santa Maria, RS, Brazil
| | | | - Luana Haselein Maurer
- Programa de Pós-Graduação em Ciência e Tecnologia dos Alimentos-Universidade Federal de Santa Maria, RS, Brazil
| | - Jéssica Stiebe
- Departamento de Tecnologia e Ciências dos Alimentos - Universidade Federal de Santa Maria, RS, Brazil
| | - Tatiana Emanuelli
- Programa de Pós-Graduação em Ciência e Tecnologia dos Alimentos-Universidade Federal de Santa Maria, RS, Brazil
| | | | - Camila Simonetti Pase
- Programa de Pós-Graduação em Farmacologia-Universidade Federal de Santa Maria, RS, Brazil; Universidade Federal do Pampa, Campus Uruguaiana, RS, Brazil.
| |
Collapse
|
48
|
Mutational effects of human dopamine transporter at tyrosine88, lysine92, and histidine547 on basal and HIV-1 Tat-inhibited dopamine transport. Sci Rep 2019; 9:3843. [PMID: 30846720 PMCID: PMC6405875 DOI: 10.1038/s41598-019-39872-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 12/12/2018] [Indexed: 01/06/2023] Open
Abstract
Dysregulation of dopaminergic system induced by HIV-1 Tat protein-mediated direct inhibition of the dopamine transporter (DAT) has been implicated as a mediating factor of HIV-1 associated neurocognitive disorders. We have reported that single point mutations on human DAT (hDAT) at tyrosine88 (Y88F), lysine92 (K92M), and histidine547 (H547A) differentially regulate basal dopamine uptake but diminish Tat-induced inhibition of dopamine uptake by changing dopamine transport process. This study evaluated the effects of double (Y88F/H547A) and triple (Y88F/K92M/H547A) mutations on basal dopamine uptake, Tat-induced inhibition of DAT function, and dynamic transport process. Compared to wild-type hDAT, the Vmax values of [3H]Dopamine uptake were increased by 96% in Y88F/H547A but decreased by 97% in Y88F/K92M/H547A. [3H]WIN35,428 binding sites were not altered in Y88F/H547A but decreased in Y88F/K92M/H547A. Y88F/H547A mutant attenuated Tat-induced inhibition of dopamine uptake observed in wild-type hDAT. Y88F/H547A displayed an attenuation of zinc-augmented [3H]WIN35,428 binding, increased basal dopamine efflux, and reduced amphetamine-induced dopamine efflux, indicating this mutant alters transporter conformational transitions. These findings further demonstrate that both tyrosine88 and histidine547 on hDAT play a key role in stabilizing basal dopamine transport and Tat-DAT integration. This study provides mechanistic insights into developing small molecules to block multiple sites in DAT for Tat binding.
Collapse
|
49
|
Mauna JC, Harris SS, Pino JA, Edwards CM, DeChellis-Marks MR, Bassi CD, Garcia-Olivares J, Amara SG, Guajardo FG, Sotomayor-Zarate R, Terminel M, Castañeda E, Vergara M, Baust T, Thiels E, Torres GE. G protein βγ subunits play a critical role in the actions of amphetamine. Transl Psychiatry 2019; 9:81. [PMID: 30745563 PMCID: PMC6370791 DOI: 10.1038/s41398-019-0387-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 10/30/2018] [Accepted: 01/01/2019] [Indexed: 11/18/2022] Open
Abstract
Abnormal levels of dopamine (DA) are thought to contribute to several neurological and psychiatric disorders including drug addiction. Extracellular DA levels are regulated primarily via reuptake by the DA transporter (DAT). Amphetamine, a potent psychostimulant, increases extracellular DA by inducing efflux through DAT. Recently, we discovered that G protein βγ subunits (Gβγ) interact with DAT, and that in vitro activation of Gβγ promotes DAT-mediated efflux. Here, we investigated the role of Gβγ in the actions of amphetamine in DA neurons in culture, ex vivo nucleus accumbens (NAc), and freely moving rats. Activation of Gβγ with the peptide myr-Ser-Ile-Arg-Lys-Ala-Leu-Asn-Ile-Leu-Gly-Tyr-Pro-Asp-Tyr-Asp (mSIRK) in the NAc potentiated amphetamine-induced hyperlocomotion, but not cocaine-induced hyperlocomotion, and systemic or intra-accumbal administration of the Gβγ inhibitor gallein attenuated amphetamine-induced, but not cocaine-induced hyperlocomotion. Infusion into the NAc of a TAT-fused peptide that targets the Gβγ-binding site on DAT (TAT-DATct1) also attenuated amphetamine-induced but not cocaine-induced hyperlocomotion. In DA neurons in culture, inhibition of Gβγ with gallein or blockade of the Gβγ-DAT interaction with the TAT-DATct1 peptide decreased amphetamine-induced DA efflux. Furthermore, activation of Gβγ with mSIRK potentiated and inhibition of Gβγ with gallein reduced amphetamine-induced increases of extracellular DA in the NAc in vitro and in freely moving rats. Finally, systemic or intra-accumbal inhibition of Gβγ with gallein blocked the development of amphetamine-induced, but not cocaine-induced place preference. Collectively, these results suggest that interaction between Gβγ and DAT plays a critical role in the actions of amphetamine and presents a novel target for modulating the actions of amphetamine in vivo.
Collapse
Affiliation(s)
- J. C. Mauna
- 0000 0004 1936 9000grid.21925.3dDepartment of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - S. S. Harris
- 0000 0004 1936 8091grid.15276.37Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL USA
| | - J. A. Pino
- 0000 0004 1936 8091grid.15276.37Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL USA
| | - C. M. Edwards
- 0000 0004 1936 9000grid.21925.3dDepartment of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - M. R. DeChellis-Marks
- 0000 0004 1936 9000grid.21925.3dDepartment of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - C. D. Bassi
- 0000 0004 1936 9000grid.21925.3dDepartment of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - J. Garcia-Olivares
- 0000 0001 2297 5165grid.94365.3dLaboratory of Cellular and Molecular Neurobiology, National Institute of Mental Health, NIH, Bethesda, MD USA
| | - S. G. Amara
- 0000 0001 2297 5165grid.94365.3dLaboratory of Cellular and Molecular Neurobiology, National Institute of Mental Health, NIH, Bethesda, MD USA
| | - F. G. Guajardo
- 0000 0004 1936 8091grid.15276.37Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL USA ,0000 0000 8912 4050grid.412185.bLaboratory of Neurochemistry and Neuropharmacology, Center for Neurobiology and Brain Plasticity, Universidad de Valparaíso, Valparaíso, Chile
| | - R. Sotomayor-Zarate
- 0000 0000 8912 4050grid.412185.bLaboratory of Neurochemistry and Neuropharmacology, Center for Neurobiology and Brain Plasticity, Universidad de Valparaíso, Valparaíso, Chile
| | - M. Terminel
- 0000 0001 0668 0420grid.267324.6Department of Psychology, University of Texas at El Paso, El Paso, TX USA
| | - E. Castañeda
- 0000 0001 0668 0420grid.267324.6Department of Psychology, University of Texas at El Paso, El Paso, TX USA
| | - M. Vergara
- 0000 0004 1936 8091grid.15276.37Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL USA
| | - T. Baust
- 0000 0004 1936 9000grid.21925.3dDepartment of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - E. Thiels
- 0000 0004 1936 9000grid.21925.3dDepartment of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - G. E. Torres
- 0000 0004 1936 8091grid.15276.37Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL USA ,0000 0004 1936 8091grid.15276.37Center for Addiction Research and Education, University of Florida College of Medicine, Gainesville, FL USA
| |
Collapse
|
50
|
Wu H, Guo M, Yang M, Luo Z, Hansen K. Selective C–C and C–N bond activation in dopamine and norepinephrine under deep ultraviolet laser irradiation. Chem Commun (Camb) 2019; 55:4015-4018. [DOI: 10.1039/c9cc00444k] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Selective C–C bond and C–N bond activation in dopamine and norepinephrine is analyzed utilizing deep-ultraviolet laser ionization mass spectrometry (DUV-LIMS).
Collapse
Affiliation(s)
- Haiming Wu
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species
- Institute of Chemistry
- Chinese Academy of Sciences
- Beijing 100190
- P. R. China
| | - Mengdi Guo
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species
- Institute of Chemistry
- Chinese Academy of Sciences
- Beijing 100190
- P. R. China
| | - Mengzhou Yang
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species
- Institute of Chemistry
- Chinese Academy of Sciences
- Beijing 100190
- P. R. China
| | - Zhixun Luo
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species
- Institute of Chemistry
- Chinese Academy of Sciences
- Beijing 100190
- P. R. China
| | - Klavs Hansen
- Joint Center for Quantum Studies
- School of Science
- Tianjin University
- Tianjin
- P. R. China
| |
Collapse
|