1
|
Newton K, De Biase L. Substance Use and Addiction. ADVANCES IN NEUROBIOLOGY 2024; 37:343-355. [PMID: 39207701 DOI: 10.1007/978-3-031-55529-9_19] [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: 09/04/2024]
Abstract
Efforts to reveal the molecular, cellular, and circuit mechanisms of addiction have largely focused on neurons. Yet accumulating data regarding the ability of glial cells to impact synaptic function, circuit activity, and behavior demands that we explore how these nonneuronal cells contribute to substance use disorders and addiction. Important work has shown that glial cells, including microglia, exhibit changes in phenotype following exposure to drugs of abuse and that modification of glial responses can impact behaviors related to drug seeking and drug taking. While these are critical first steps to understanding how microglia can impact addiction, there are still substantial gaps in knowledge that need to be addressed. This chapter reviews some of the key studies that have shown how microglia are affected by and can contribute to addiction. It also discusses areas where more knowledge is urgently needed to reveal new therapeutic and preventative approaches.
Collapse
Affiliation(s)
- Keionna Newton
- Neuroscience Interdepartmental Graduate Program, University of California, Los Angeles, CA, USA
| | - Lindsay De Biase
- Department of Physiology, University of California, Los Angeles, CA, USA.
| |
Collapse
|
2
|
Shi H, He Y, Zhou Y, Huang J, Maher K, Wang B, Tang Z, Luo S, Tan P, Wu M, Lin Z, Ren J, Thapa Y, Tang X, Chan KY, Deverman BE, Shen H, Liu A, Liu J, Wang X. Spatial atlas of the mouse central nervous system at molecular resolution. Nature 2023; 622:552-561. [PMID: 37758947 PMCID: PMC10709140 DOI: 10.1038/s41586-023-06569-5] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/22/2023] [Indexed: 09/29/2023]
Abstract
Spatially charting molecular cell types at single-cell resolution across the 3D volume is critical for illustrating the molecular basis of brain anatomy and functions. Single-cell RNA sequencing has profiled molecular cell types in the mouse brain1,2, but cannot capture their spatial organization. Here we used an in situ sequencing method, STARmap PLUS3,4, to profile 1,022 genes in 3D at a voxel size of 194 × 194 × 345 nm3, mapping 1.09 million high-quality cells across the adult mouse brain and spinal cord. We developed computational pipelines to segment, cluster and annotate 230 molecular cell types by single-cell gene expression and 106 molecular tissue regions by spatial niche gene expression. Joint analysis of molecular cell types and molecular tissue regions enabled a systematic molecular spatial cell-type nomenclature and identification of tissue architectures that were undefined in established brain anatomy. To create a transcriptome-wide spatial atlas, we integrated STARmap PLUS measurements with a published single-cell RNA-sequencing atlas1, imputing single-cell expression profiles of 11,844 genes. Finally, we delineated viral tropisms of a brain-wide transgene delivery tool, AAV-PHP.eB5,6. Together, this annotated dataset provides a single-cell resource that integrates the molecular spatial atlas, brain anatomy and the accessibility to genetic manipulation of the mammalian central nervous system.
Collapse
Affiliation(s)
- Hailing Shi
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yichun He
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
| | - Yiming Zhou
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jiahao Huang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kamal Maher
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Computational and Systems Biology PhD Program, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Brandon Wang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Zefang Tang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Shuchen Luo
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Peng Tan
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Morgan Wu
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Zuwan Lin
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Jingyi Ren
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yaman Thapa
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Xin Tang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
| | - Ken Y Chan
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Benjamin E Deverman
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hao Shen
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
| | - Albert Liu
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jia Liu
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA.
| | - Xiao Wang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
3
|
Hosseini MJ, Arabiyan A, Mobassem S, Ghavimi H. Metformin attenuates depressive-like behaviour of methamphetamine withdrawal in mice: A mechanistic approach. World J Biol Psychiatry 2023; 24:209-222. [PMID: 35673936 DOI: 10.1080/15622975.2022.2086294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVES Methamphetamine (METH) as a potent psychostimulant drug with a high potency of dependence rate that results in neurotoxicity has become a major drug of abuse in many parts of the world. Unfortunately, there is limited evidence regarding treatment of METH withdrawal syndrome. Therefore, we aimed to investigate whether metformin mitigate the methamphetamine (METH) withdrawal syndrome in male mice. Based on the literature, depression and anxiety are the major METH withdrawal symptoms. METHODS Here, METH (2 mg/kg) was administered to mice twice a day for 14 constitutive days to induce animal model of METH-induced withdrawal syndrome. To do this, mice in control group and those with METH withdrawal syndrome were divided into treatment (receiving metformin in 3 doses of 50, 100 and 200 mg/kg for 10 days) and non-treatment sub-groups. Following the behavioural test, the animals were sacrificed; their hippocampus was dissected to measure oxidative stress parameters and expression of cellular energy homeostasis and immune-inflammatory genes. RESULTS Our data revealed that metformin provoked antidepressant effects in behavioural tests through AMPK overexpression as an important mitochondrial energetic sensor and inhibition of Tlr4 overexpression in the immune system gene expression. In addition, metformin was able to improve oxidative stress biomarkers and neuronal damage in the hippocampus and restore cellular energy homeostasis and immune system gene expression. CONCLUSIONS The data suggested that metformin can influence the hippocampus through targeting mitochondria and their performance, and consequently, neuroinflammation responses and brain metabolic changes. It is supposed to be a new therapeutic option in clinical trials of depression and anxiety following METH withdrawal treatment.
Collapse
Affiliation(s)
- Mir-Jamal Hosseini
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.,Departments of Pharmacology and Toxicology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Aisan Arabiyan
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.,Departments of Pharmacology and Toxicology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Sina Mobassem
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.,Departments of Pharmacology and Toxicology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hamed Ghavimi
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.,Departments of Pharmacology and Toxicology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
4
|
Barone A, De Simone G, Ciccarelli M, Buonaguro EF, Tomasetti C, Eramo A, Vellucci L, de Bartolomeis A. A Postsynaptic Density Immediate Early Gene-Based Connectome Analysis of Acute NMDAR Blockade and Reversal Effect of Antipsychotic Administration. Int J Mol Sci 2023; 24:ijms24054372. [PMID: 36901803 PMCID: PMC10002165 DOI: 10.3390/ijms24054372] [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: 01/25/2023] [Revised: 02/20/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Although antipsychotics' mechanisms of action have been thoroughly investigated, they have not been fully elucidated at the network level. We tested the hypothesis that acute pre-treatment with ketamine (KET) and administration of asenapine (ASE) would modulate the functional connectivity of brain areas relevant to the pathophysiology of schizophrenia, based on transcript levels of Homer1a, an immediate early gene encoding a key molecule of the dendritic spine. Sprague-Dawley rats (n = 20) were assigned to KET (30 mg/kg) or vehicle (VEH). Each pre-treatment group (n = 10) was randomly split into two arms, receiving ASE (0.3 mg/kg), or VEH. Homer1a mRNA levels were evaluated by in situ hybridization in 33 regions of interest (ROIs). We computed all possible pairwise Pearson correlations and generated a network for each treatment group. Acute KET challenge was associated with negative correlations between the medial portion of cingulate cortex/indusium griseum and other ROIs, not detectable in other treatment groups. KET/ASE group showed significantly higher inter-correlations between medial cingulate cortex/indusium griseum and lateral putamen, the upper lip of the primary somatosensory cortex, septal area nuclei, and claustrum, in comparison to the KET/VEH network. ASE exposure was associated with changes in subcortical-cortical connectivity and an increase in centrality measures of the cingulate cortex and lateral septal nuclei. In conclusion, ASE was found to finely regulate brain connectivity by modelling the synaptic architecture and restoring a functional pattern of interregional co-activation.
Collapse
Affiliation(s)
- Annarita Barone
- Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University Medical School of Naples “Federico II”, 80131 Naples, Italy
| | - Giuseppe De Simone
- Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University Medical School of Naples “Federico II”, 80131 Naples, Italy
| | - Mariateresa Ciccarelli
- Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University Medical School of Naples “Federico II”, 80131 Naples, Italy
| | - Elisabetta Filomena Buonaguro
- Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University Medical School of Naples “Federico II”, 80131 Naples, Italy
| | - Carmine Tomasetti
- Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University Medical School of Naples “Federico II”, 80131 Naples, Italy
| | | | - Licia Vellucci
- Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University Medical School of Naples “Federico II”, 80131 Naples, Italy
| | - Andrea de Bartolomeis
- Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University Medical School of Naples “Federico II”, 80131 Naples, Italy
- Correspondence: ; Tel.: +39-081-7463673; Fax: +39-081-7462644
| |
Collapse
|
5
|
Machado da Silva MC, Iglesias LP, Candelario-Jalil E, Khoshbouei H, Moreira FA, de Oliveira ACP. Role of Microglia in Psychostimulant Addiction. Curr Neuropharmacol 2023; 21:235-259. [PMID: 36503452 PMCID: PMC10190137 DOI: 10.2174/1570159x21666221208142151] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 12/14/2022] Open
Abstract
The use of psychostimulant drugs can modify brain function by inducing changes in the reward system, mainly due to alterations in dopaminergic and glutamatergic transmissions in the mesocorticolimbic pathway. However, the etiopathogenesis of addiction is a much more complex process. Previous data have suggested that microglia and other immune cells are involved in events associated with neuroplasticity and memory, which are phenomena that also occur in addiction. Nevertheless, how dependent is the development of addiction on the activity of these cells? Although the mechanisms are not known, some pathways may be involved. Recent data have shown psychoactive substances may act directly on immune cells, alter their functions and induce various inflammatory mediators that modulate synaptic activity. These could, in turn, be involved in the pathological alterations that occur in substance use disorder. Here, we extensively review the studies demonstrating how cocaine and amphetamines modulate microglial number, morphology, and function. We also describe the effect of these substances in the production of inflammatory mediators and a possible involvement of some molecular signaling pathways, such as the toll-like receptor 4. Although the literature in this field is scarce, this review compiles the knowledge on the neuroimmune axis that is involved in the pathogenesis of addiction, and suggests some pharmacological targets for the development of pharmacotherapy.
Collapse
Affiliation(s)
- Maria Carolina Machado da Silva
- Department of Pharmacology, Neuropharmacology Laboratory, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil;
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Lia Parada Iglesias
- Department of Pharmacology, Neuropsychopharmacology Laboratory, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Habibeh Khoshbouei
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Fabrício Araujo Moreira
- Department of Pharmacology, Neuropsychopharmacology Laboratory, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | |
Collapse
|
6
|
Lafuente JV, Sharma A, Feng L, Muresanu DF, Nozari A, Tian ZR, Buzoianu AD, Sjöquist PO, Wiklund L, Sharma HS. Nanowired Delivery of Mesenchymal Stem Cells with Antioxidant Compound H-290/51 Reduces Exacerbation of Methamphetamine Neurotoxicity in Hot Environment. ADVANCES IN NEUROBIOLOGY 2023; 32:317-352. [PMID: 37480465 DOI: 10.1007/978-3-031-32997-5_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
Military personnel are often exposed to hot environments either for combat operations or peacekeeping missions. Hot environment is a severe stressful situation leading to profound hyperthermia, fatigue and neurological impairments. To avoid stressful environment, some people frequently use methamphetamine (METH) or other psychostimulants to feel comfortable under adverse situations. Our studies show that heat stress alone induces breakdown of the blood-brain barrier (BBB) and edema formation associated with reduced cerebral blood flow (CBF). On the other hand, METH alone induces hyperthermia and neurotoxicity. These effects of METH are exacerbated at high ambient temperatures as seen with greater breakdown of the BBB and brain pathology. Thus, a combination of METH use at hot environment may further enhance the brain damage-associated behavioral dysfunctions. METH is well known to induce severe oxidative stress leading to brain pathology. In this investigation, METH intoxication at hot environment was examined on brain pathology and to explore suitable strategies to induce neuroprotection. Accordingly, TiO2-nanowired delivery of H-290/51 (150 mg/kg, i.p.), a potent chain-breaking antioxidant in combination with mesenchymal stem cells (MSCs), is investigated in attenuating METH-induced brain damage at hot environment in model experiments. Our results show that nanodelivery of H-290/51 with MSCs significantly enhanced CBF and reduced BBB breakdown, edema formation and brain pathology following METH exposure at hot environment. These observations are the first to point out that METH exacerbated brain pathology at hot environment probably due to enhanced oxidative stress, and MSCs attenuate these adverse effects, not reported earlier.
Collapse
Affiliation(s)
- José Vicente Lafuente
- LaNCE, Department Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Zhongshan, Hebei Province, China
| | - Dafin F Muresanu
- Department Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania
- "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Ala Nozari
- Anesthesiology & Intensive Care, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Z Ryan Tian
- Department Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, USA
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Per-Ove Sjöquist
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
7
|
Kermanian F, Seghatoleslam M, Mahakizadeh S. MDMA related neuro-inflammation and adenosine receptors. Neurochem Int 2022; 153:105275. [PMID: 34990730 DOI: 10.1016/j.neuint.2021.105275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 12/25/2021] [Accepted: 12/30/2021] [Indexed: 12/20/2022]
Abstract
3,4-methylenedioxymethamphetamine (MDMA) is a world-wide abused psychostimulant, which has the neurotoxic effects on dopaminergic and serotonergic neurons in both rodents and non-human primates. Adenosine acts as a neurotransmitter in the brain through the activation of four specific G-protein-coupled receptors and it acts as a neuromodulator of dopamine neurotransmission. Recent studies suggest that stimulation of adenosine receptors oppose many behavioral effects of methamphetamines. This review summarizes the specific cellular mechanisms involved in MDMA neuroinflammatory effects, along with the protective effects of adenosine receptors.
Collapse
Affiliation(s)
- Fatemeh Kermanian
- Department of Anatomy, Faculty of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Masoumeh Seghatoleslam
- Evaluative Clinical Sciences, Sunnybrook Research Institute, University of Toronto, ON, Canada
| | - Simin Mahakizadeh
- Department of Anatomy, Faculty of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
8
|
Apóstol Del Rosal GD, Limón ID, Martínez I, Patricio-Martínez A. The Chronic Oral Administration of Clobenzorex or Amphetamine Decreases Motor Behavior and Induces Glial Activation in the Striatum Without Dopaminergic Degeneration. Neurotox Res 2021; 39:1405-1417. [PMID: 34279823 DOI: 10.1007/s12640-021-00395-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/22/2021] [Accepted: 07/12/2021] [Indexed: 01/12/2023]
Abstract
Described as amphetamine-like due to their structural and stimulant similarities, clobenzorex is one of the five most-commonly used drugs in Mexico for the treatment of obesity. Various studies have shown that amphetamines induce dopaminergic neurotoxicity and neuroinflammation in the striatum, symptoms which are associated with motor damage. For this reason, the present study aimed to evaluate the effect of chronic clobenzorex administration on motor behaviors, TH immunoreactivity, gliosis, and the neurodegenerative process in the striatum and substantia nigra pars compacta (SNpc). The present research was conducted on three experimental groups of male Wistar rats: the vehicle group, the amphetamine group (2 mg/kg), and the clobenzorex group (30 mg/kg). All groups were subject to oral administration every 24 h for 31 days. Motor activity and motor coordination were evaluated in the open field test and the beam walking test, respectively. The animals were euthanized after the last day of treatment to enable the extraction of their brains for the evaluation of tyrosine hydroxylase (TH) levels, the immunoreactivity of the glial cells, and the neurodegeneration of both the striatum and SNpc via amino-cupric-silver stain. The results obtained show that amphetamine and clobenzorex administration decrease motor activity and motor coordination in the beam walking test and cause increased gliosis in the striatum, while no significant changes were observed in terms of immunoreactivity to TH and neurodegeneration in both the striatum and SNpc. These results suggest that the chronic administration of clobenzorex may decrease motor function in a manner similar to amphetamine, via the neuroadaptive and non-neurotoxic changes caused to the striatum under this administration scheme.
Collapse
Affiliation(s)
- Grego David Apóstol Del Rosal
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Ilhuicamina Daniel Limón
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Isabel Martínez
- Laboratorio de Neuroquímica, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Aleidy Patricio-Martínez
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico.
- Facultad de Ciencias Biológicas, Benemérita Universidad Autónoma de Puebla, Blvd. Valsequillo y Av. San Claudio Edificio BIO-1 C.U. Col. Jardines de San Manuel A.P, 72570, Puebla, Mexico.
| |
Collapse
|
9
|
Gholami M, Hozuri F, Abdolkarimi S, Mahmoudi M, Motaghinejad M, Safari S, Sadr S. Pharmacological and Molecular Evidence of Neuroprotective Curcumin Effects Against Biochemical and Behavioral Sequels Caused by Methamphetamine: Possible Function of CREB-BDNF Signaling Pathway. Basic Clin Neurosci 2021; 12:325-338. [PMID: 34917292 PMCID: PMC8666919 DOI: 10.32598/bcn.2021.1176.3] [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: 12/15/2018] [Revised: 02/20/2019] [Accepted: 04/20/2020] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION The neuroprotective impact of curcumin and the role of CREB (Cyclic AMP Response Element Binding protein)-BDNF (Brain-Derived Neurotrophic Factor) signaling pathway was evaluated in Methamphetamine (METH)-induced neurodegeneration in rats. METHODS Sixty adult male rats were randomly divided into 6 groups. While normal saline and 10 mg/kg METH were administered intraperitoneally in groups 1 and 2, groups 3, 4, 5, and 6 received METH (10 mg/kg) and curcumin (10, 20, 40, and 80 mg/kg, respectively) simultaneously. Morris water maze test was administered, and oxidative hippocampal, antioxidant, inflammatory, apoptotic, and CREB and BDNF were assessed. RESULTS We found that METH disturbs learning and memory. Concurrent curcumin therapy (40 and 80 mg/kg) decreased cognitive disturbance caused by METH. Multiple parameters, such as lipid peroxidation, the oxidized form of glutathione, interleukin 1 beta, tumor necrosis factor-alpha, and Bax were increased by METH therapy, while the reduced type of glutathione, Bcl-2, P-CREB, and BDNF concentrations in the hippocampus were decreased. CONCLUSION Different doses of curcumin adversely attenuated METH-induced apoptosis, oxidative stress, and inflammation but enhanced the concentrations of P-CREB and BDNF. The neuroprotection caused by curcumin against METH-induced neurodegeneration is mediated through P-CREB-BDNF signaling pathway activation.
Collapse
Affiliation(s)
- Mina Gholami
- Department of Medicinal Chemistry, School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzad Hozuri
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Chemistry, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Setayesh Abdolkarimi
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Chemistry, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Mahsa Mahmoudi
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Chemistry, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Majid Motaghinejad
- Research Center for Addiction and Risky Behaviors (ReCARB), Iran Psychiatric Center, Iran University of Medical Sciences, Tehran, Iran
| | - Sepideh Safari
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Samira Sadr
- Department of Research and Development, Parsian-Exir-Aria Pharmaceutical Company, Tehran, Iran
| |
Collapse
|
10
|
Brain-wide genetic mapping identifies the indusium griseum as a prenatal target of pharmacologically unrelated psychostimulants. Proc Natl Acad Sci U S A 2019; 116:25958-25967. [PMID: 31796600 DOI: 10.1073/pnas.1904006116] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Psychostimulant use is an ever-increasing socioeconomic burden, including a dramatic rise during pregnancy. Nevertheless, brain-wide effects of psychostimulant exposure are incompletely understood. Here, we performed Fos-CreERT2-based activity mapping, correlated for pregnant mouse dams and their fetuses with amphetamine, nicotine, and caffeine applied acutely during midgestation. While light-sheet microscopy-assisted intact tissue imaging revealed drug- and age-specific neuronal activation, the indusium griseum (IG) appeared indiscriminately affected. By using GAD67gfp/+ mice we subdivided the IG into a dorsolateral domain populated by γ-aminobutyric acidergic interneurons and a ventromedial segment containing glutamatergic neurons, many showing drug-induced activation and sequentially expressing Pou3f3/Brn1 and secretagogin (Scgn) during differentiation. We then combined Patch-seq and circuit mapping to show that the ventromedial IG is a quasi-continuum of glutamatergic neurons (IG-Vglut1 +) reminiscent of dentate granule cells in both rodents and humans, whose dendrites emanate perpendicularly toward while their axons course parallel with the superior longitudinal fissure. IG-Vglut1 + neurons receive VGLUT1+ and VGLUT2+ excitatory afferents that topologically segregate along their somatodendritic axis. In turn, their efferents terminate in the olfactory bulb, thus being integral to a multisynaptic circuit that could feed information antiparallel to the olfactory-cortical pathway. In IG-Vglut1 + neurons, prenatal psychostimulant exposure delayed the onset of Scgn expression. Genetic ablation of Scgn was then found to sensitize adult mice toward methamphetamine-induced epilepsy. Overall, our study identifies brain-wide targets of the most common psychostimulants, among which Scgn +/Vglut1 + neurons of the IG link limbic and olfactory circuits.
Collapse
|
11
|
Catale C, Bussone S, Lo Iacono L, Carola V. Microglial alterations induced by psychoactive drugs: A possible mechanism in substance use disorder? Semin Cell Dev Biol 2019; 94:164-175. [PMID: 31004753 DOI: 10.1016/j.semcdb.2019.03.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/19/2019] [Accepted: 03/29/2019] [Indexed: 12/11/2022]
Abstract
Recently, the xenobiotic hypothesis has implicated the immune system in targeting substances of abuse as foreign molecules and stimulating inflammatory responses. Microglial cells are the resident immune cells of the central nervous system and function in homeostatic surveillance. Microglial changes that are induced by exposure to substances of abuse appear to mediate in part the establishment of addiction and the persistence of drug-mediated biological and behavioral changes. In this context, interest in the study of drug-microglia interactions has increased recently. This review summarizes the most recent preclinical rodent and clinical studies on the interaction between microglia and various classes of drugs of abuse, such as ethanol, psychostimulants, and opioids. The principal biological mechanisms of the communication between substances of abuse and microglia will be described to consider putative mechanisms of the establishment of drug addiction and future potential targets for treating substance use disorder.
Collapse
Affiliation(s)
- Clarissa Catale
- Department of Psychology, University of Rome "La Sapienza", Via dei Marsi, 78, 00185 Rome, Italy
| | - Silvia Bussone
- Department of Dynamic and Clinical Psychology, University of Rome "La Sapienza", Via degli Apuli 1, 00185 Rome, Italy
| | - Luisa Lo Iacono
- Department of Psychology, University of Rome "La Sapienza", Via dei Marsi, 78, 00185 Rome, Italy; IRCCS Santa Lucia Foundation, Via Fosso di Fiorano 64, 00143 Rome, Italy
| | - Valeria Carola
- Department of Dynamic and Clinical Psychology, University of Rome "La Sapienza", Via degli Apuli 1, 00185 Rome, Italy; IRCCS Santa Lucia Foundation, Via Fosso di Fiorano 64, 00143 Rome, Italy.
| |
Collapse
|
12
|
Du SH, Zhang W, Yue X, Luo XQ, Tan XH, Liu C, Qiao DF, Wang H. Role of CXCR1 and Interleukin-8 in Methamphetamine-Induced Neuronal Apoptosis. Front Cell Neurosci 2018; 12:230. [PMID: 30123110 PMCID: PMC6085841 DOI: 10.3389/fncel.2018.00230] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 07/16/2018] [Indexed: 12/12/2022] Open
Abstract
Methamphetamine (METH), an extremely and widely abused illicit drug, can cause serious nervous system damage and social problems. Previous research has shown that METH use causes dopaminergic neuron apoptosis and astrocyte-related neuroinflammation. However, the relationship of astrocytes and neurons in METH-induced neurotoxicity remains unclear. We hypothesized that chemokine interleukin (IL) eight released by astrocytes and C-X-C motif chemokine receptor 1 (CXCR1) in neurons are involved in METH-induced neuronal apoptosis. We tested our hypothesis by examining the changes of CXCR1 in SH-SY5Y cells and in the brain of C57BL/6 mice exposed to METH by western blotting and immunolabeling. We also determined the effects of knocking down CXCR1 expression with small interfering ribonucleic acid (siRNA) on METH-exposed SH-SY5Y cells. Furthermore, we detected the expression levels of IL-8 and the nuclear factor-kappa B (NF-κB) pathway in U87MG cells and then co-cultured the two cell types to determine the role of CXCR1 and IL-8 in neuronal apoptosis. Our results indicated that METH exposure increased CXCR1 expression both in vitro and in vivo, with the effects obtained in vitro being dose-dependent. Silencing of CXCR1 expression with siRNAs reduced the expression of cleaved caspase-3, cleaved poly (ADP-ribose) polymerase (PARP), and other related proteins. In addition, IL-8 expression and release were increased in METH-exposed U87MG cells, which is regulated by NF-κB pathway. Neuronal apoptosis was attenuated by siCXCR1 after METH treatment in the co-cultured cells, which can be reversed after exposure to recombinant IL-8. These results demonstrate that CXCR1 plays an important role in neuronal apoptosis induced by METH and may be a potential target for METH-induced neurotoxicity therapy. Highlights -Methamphetamine exposure upregulated the expression of CXCR1.-Methamphetamine exposure increased the expression of interleukin-8 through nuclear factor-kappa B pathway.-Activation of CXCR1 by interleukin-8 induces an increase in methamphetamine-related neuronal apoptosis.
Collapse
Affiliation(s)
- Si-Hao Du
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Wei Zhang
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Xia Yue
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Xiao-Qing Luo
- School of Forensic Medicine, Southern Medical University, Guangzhou, China.,Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiao-Hui Tan
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Chao Liu
- Guangzhou Forensic Science Institute, Guangzhou Public Security Bureau, Guangzhou, China
| | - Dong-Fang Qiao
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Huijun Wang
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
13
|
Granado N, Ares-Santos S, Tizabi Y, Moratalla R. Striatal Reinnervation Process after Acute Methamphetamine-Induced Dopaminergic Degeneration in Mice. Neurotox Res 2018; 34:627-639. [PMID: 29934756 DOI: 10.1007/s12640-018-9925-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/28/2018] [Accepted: 06/07/2018] [Indexed: 01/03/2023]
Abstract
Methamphetamine (METH), an amphetamine derivate, may increase the risk of developing Parkinson's disease (PD). Human and animal studies have shown that METH produces persistent dopaminergic neurotoxicity in the nigrostriatal pathway, despite initial partial recovery. To determine the processes leading to early compensation, we studied the detailed morphology and distribution of tyrosine hydroxylase immunoreactive fibers (TH-ir) classified by their thickness (types I-IV) before and after METH. Applying three established neurotoxic regimens of METH: single high dose (1 × 30 mg/kg), multiple lower doses (3 × 5 mg/kg) or (3 × 10 mg/kg), we show that METH primarily damages type I fibers (the thinner ones), and to a much lesser extend types II-IV fibers including sterile axons. The striatal TH terminal partial recovery process, consisting of a progressive regrowth increases in types II, III, and IV fibers, demonstrated by co-localization of GAP-43, a sprouting marker, was observed 3 days post-METH treatment. In addition, we demonstrate the presence of growth-cone-like TH-ir structures, indicative of new terminal generation as well as improvement in motor functions after 3 days. A temporal relationship was observed between decreases in TH-expression and increases in silver staining, a marker of degeneration. Striatal regeneration was associated with an increase in astroglia and decrease in microglia expression, suggesting a possible role for the neuroimmune system in regenerative processes. Identification of regenerative compensatory mechanisms in response to neurotoxic agents could point to novel mechanisms in countering the neurotoxicity and/or enhancing the regenerative processes.
Collapse
Affiliation(s)
- Noelia Granado
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Avda Dr Arce 37, 28002, Madrid, Spain.,CIBERNED, ISCIII, Madrid, Spain
| | - Sara Ares-Santos
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Avda Dr Arce 37, 28002, Madrid, Spain.,CIBERNED, ISCIII, Madrid, Spain
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington DC, USA
| | - Rosario Moratalla
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Avda Dr Arce 37, 28002, Madrid, Spain. .,CIBERNED, ISCIII, Madrid, Spain.
| |
Collapse
|
14
|
Du SH, Qiao DF, Chen CX, Chen S, Liu C, Lin Z, Wang H, Xie WB. Toll-Like Receptor 4 Mediates Methamphetamine-Induced Neuroinflammation through Caspase-11 Signaling Pathway in Astrocytes. Front Mol Neurosci 2017; 10:409. [PMID: 29311802 PMCID: PMC5733023 DOI: 10.3389/fnmol.2017.00409] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 11/27/2017] [Indexed: 02/02/2023] Open
Abstract
Methamphetamine (METH) is an amphetamine-typed stimulant drug that is increasingly being abused worldwide. Previous studies have shown that METH toxicity is systemic, especially targeting dopaminergic neurons in the central nervous system (CNS). However, the role of neuroinflammation in METH neurotoxicity remains unclear. We hypothesized that Toll-like receptor 4 (TLR4) and Caspase-11 are involved in METH-induced astrocyte-related neuroinflammation. We tested our hypothesis by examining the changes of TLR4 and Caspase-11 protein expression in primary cultured C57BL/6 mouse astrocytes and in the midbrain and striatum of mice exposed to METH with western blot and double immunofluorescence labeling. We also determined the effects of blocking Caspase-11 expression with wedelolactone (a specific inhibitor of Caspase-11) or siRNA on METH-induced neuroinflammation in astrocytes. Furthermore, we determined the effects of blocking TLR4 expression with TAK-242 (a specific inhibitor of TLR4) or siRNA on METH-induced neuroinflammation in astrocytes. METH exposure increased Caspase-11 and TLR4 expression both in vitro and in vivo, with the effects in vitro being dose-dependent. Inhibition of Caspase-11 expression with either wedelolactone or siRNAs reduced the expression of inflammasome NLRP3 and pro-inflammatory cytokines. In addition, blocking TLR4 expression inhibited METH-induced activation of NF-κB and Caspase-11 in vitro and in vivo, suggesting that TLR4-Caspase-11 pathway is involved in METH-induced neuroinflammation. These results indicate that Caspase-11 and TLR4 play an important role in METH-induced neuroinflammation and may be potential gene targets for therapeutics in METH-caused neurotoxicity.
Collapse
Affiliation(s)
- Si-Hao Du
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Dong-Fang Qiao
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Chuan-Xiang Chen
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Si Chen
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Chao Liu
- Guangzhou Forensic Science Institute, Guangzhou, China
| | - Zhoumeng Lin
- Department of Anatomy and Physiology, Institute of Computational Comparative Medicine (ICCM), College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States
| | - Huijun Wang
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Wei-Bing Xie
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
15
|
Intranasal insulin treatment alleviates methamphetamine induced anxiety-like behavior and neuroinflammation. Neurosci Lett 2017; 660:122-129. [DOI: 10.1016/j.neulet.2017.09.026] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 09/11/2017] [Accepted: 09/12/2017] [Indexed: 01/03/2023]
|
16
|
Occhieppo VB, Marchese NA, Rodríguez ID, Basmadjian OM, Baiardi G, Bregonzio C. Neurovascular unit alteration in somatosensory cortex and enhancement of thermal nociception induced by amphetamine involves central AT1receptor activation. Eur J Neurosci 2017; 45:1586-1593. [DOI: 10.1111/ejn.13594] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/20/2017] [Accepted: 04/21/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Victoria Belén Occhieppo
- Instituto de Farmacología Experimental Córdoba (IFEC-CONICET) Departamento de Farmacología; Facultad de Ciencias Químicas Universidad Nacional de Córdoba; Edificio Nuevo de Ciencias I Ciudad Universitaria Córdoba; Haya de la Torre S/N, esquina Medina Allende Córdoba Argentina
| | - Natalia Andrea Marchese
- Instituto de Farmacología Experimental Córdoba (IFEC-CONICET) Departamento de Farmacología; Facultad de Ciencias Químicas Universidad Nacional de Córdoba; Edificio Nuevo de Ciencias I Ciudad Universitaria Córdoba; Haya de la Torre S/N, esquina Medina Allende Córdoba Argentina
| | - Iara Diamela Rodríguez
- Instituto de Farmacología Experimental Córdoba (IFEC-CONICET) Departamento de Farmacología; Facultad de Ciencias Químicas Universidad Nacional de Córdoba; Edificio Nuevo de Ciencias I Ciudad Universitaria Córdoba; Haya de la Torre S/N, esquina Medina Allende Córdoba Argentina
| | - Osvaldo Martin Basmadjian
- Instituto de Farmacología Experimental Córdoba (IFEC-CONICET) Departamento de Farmacología; Facultad de Ciencias Químicas Universidad Nacional de Córdoba; Edificio Nuevo de Ciencias I Ciudad Universitaria Córdoba; Haya de la Torre S/N, esquina Medina Allende Córdoba Argentina
| | - Gustavo Baiardi
- Laboratorio de Neurofarmacología (IIBYT-CONICET); Universidad Nacional de Córdoba Facultad de Ciencias Químicas; Universidad Católica de Córdoba; Córdoba Argentina
| | - Claudia Bregonzio
- Instituto de Farmacología Experimental Córdoba (IFEC-CONICET) Departamento de Farmacología; Facultad de Ciencias Químicas Universidad Nacional de Córdoba; Edificio Nuevo de Ciencias I Ciudad Universitaria Córdoba; Haya de la Torre S/N, esquina Medina Allende Córdoba Argentina
| |
Collapse
|
17
|
Frau L, Simola N, Porceddu PF, Morelli M. Effect of crowding, temperature and age on glia activation and dopaminergic neurotoxicity induced by MDMA in the mouse brain. Neurotoxicology 2016; 56:127-138. [PMID: 27451954 DOI: 10.1016/j.neuro.2016.07.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 07/15/2016] [Accepted: 07/20/2016] [Indexed: 12/13/2022]
Abstract
3,4-methylenedyoxymethamphetamine (MDMA or "ecstasy"), a recreational drug of abuse, can induce glia activation and dopaminergic neurotoxicity. Since MDMA is often consumed in crowded environments featuring high temperatures, we studied how these factors influenced glia activation and dopaminergic neurotoxicity induced by MDMA. C57BL/6J adolescent (4 weeks old) and adult (12 weeks old) mice received MDMA (4×20mg/kg) in different conditions: 1) while kept 1, 5, or 10×cage at room temperature (21°C); 2) while kept 5×cage at either room (21°C) or high (27°C) temperature. After the last MDMA administration, immunohistochemistry was performed in the caudate-putamen for CD11b and GFAP, to mark microglia and astroglia, and in the substantia nigra pars compacta for tyrosine hydroxylase, to mark dopaminergic neurons. MDMA induced glia activation and dopaminergic neurotoxicity, compared with vehicle administration. Crowding (5 or 10 mice×cage) amplified MDMA-induced glia activation (in adult and adolescent mice) and dopaminergic neurotoxicity (in adolescent mice). Conversely, exposure to a high environmental temperature (27°C) potentiated MDMA-induced glia activation in adult and adolescent mice kept 5×cage, but not dopaminergic neurotoxicity. Crowding and exposure to a high environmental temperature amplified MDMA-induced hyperthermia, and a positive correlation between body temperature and activation of either microglia or astroglia was found in adult and adolescent mice. These results provide further evidence that the administration setting influences the noxious effects of MDMA in the mouse brain. However, while crowding amplifies both glia activation and dopaminergic neurotoxicity, a high environmental temperature exacerbates glia activation only.
Collapse
Affiliation(s)
- Lucia Frau
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Via Ospedale 72, 09124, Cagliari, Italy.
| | - Nicola Simola
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Via Ospedale 72, 09124, Cagliari, Italy.
| | - Pier Francesca Porceddu
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Via Ospedale 72, 09124, Cagliari, Italy.
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Via Ospedale 72, 09124, Cagliari, Italy; CNR, Institute of Neuroscience, Cagliari, Italy.
| |
Collapse
|
18
|
Frau L, Costa G, Porceddu PF, Khairnar A, Castelli MP, Ennas MG, Madeddu C, Wardas J, Morelli M. Influence of caffeine on 3,4-methylenedioxymethamphetamine-induced dopaminergic neuron degeneration and neuroinflammation is age-dependent. J Neurochem 2015; 136:148-62. [DOI: 10.1111/jnc.13377] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 09/02/2015] [Accepted: 09/03/2015] [Indexed: 02/03/2023]
Affiliation(s)
- Lucia Frau
- Department of Biomedical Sciences; Section of Neuropsychopharmacology; University of Cagliari; Cagliari Italy
| | - Giulia Costa
- Department of Biomedical Sciences; Section of Neuropsychopharmacology; University of Cagliari; Cagliari Italy
| | - Pier Francesca Porceddu
- Department of Biomedical Sciences; Section of Neuropsychopharmacology; University of Cagliari; Cagliari Italy
| | - Amit Khairnar
- Applied Neuroscience Research Group; CEITEC - Central European Institute of Technology; Masaryk University; Brno Czech Republic
| | - Maria Paola Castelli
- Department of Biomedical Sciences; Section of Neuroscience and Clinical Pharmacology; University of Cagliari; Monserrato (CA) Italy
| | - Maria Grazia Ennas
- Department of Biomedical Sciences; Section of Neuroscience and Clinical Pharmacology; University of Cagliari; Monserrato (CA) Italy
| | - Camilla Madeddu
- Department of Biomedical Sciences; Section of Neuroscience and Clinical Pharmacology; University of Cagliari; Monserrato (CA) Italy
| | - Jadwiga Wardas
- Department of Neuropsychopharmacology; Institute of Pharmacology; Polish Academy of Sciences; Krakow Poland
| | - Micaela Morelli
- Department of Biomedical Sciences; Section of Neuropsychopharmacology; University of Cagliari; Cagliari Italy
- CNR; Institute of Neuroscience; Cagliari Italy
| |
Collapse
|
19
|
Moratalla R, Khairnar A, Simola N, Granado N, García-Montes JR, Porceddu PF, Tizabi Y, Costa G, Morelli M. Amphetamine-related drugs neurotoxicity in humans and in experimental animals: Main mechanisms. Prog Neurobiol 2015; 155:149-170. [PMID: 26455459 DOI: 10.1016/j.pneurobio.2015.09.011] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 09/04/2015] [Accepted: 09/15/2015] [Indexed: 12/13/2022]
Abstract
Amphetamine-related drugs, such as 3,4-methylenedioxymethamphetamine (MDMA) and methamphetamine (METH), are popular recreational psychostimulants. Several preclinical studies have demonstrated that, besides having the potential for abuse, amphetamine-related drugs may also elicit neurotoxic and neuroinflammatory effects. The neurotoxic potentials of MDMA and METH to dopaminergic and serotonergic neurons have been clearly demonstrated in both rodents and non-human primates. This review summarizes the species-specific cellular and molecular mechanisms involved in MDMA and METH-mediated neurotoxic and neuroinflammatory effects, along with the most important behavioral changes elicited by these substances in experimental animals and humans. Emphasis is placed on the neuropsychological and neurological consequences associated with the neuronal damage. Moreover, we point out the gap in our knowledge and the need for developing appropriate therapeutic strategies to manage the neurological problems associated with amphetamine-related drug abuse.
Collapse
Affiliation(s)
- Rosario Moratalla
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Madrid, Spain; CIBERNED, ISCIII, Madrid, Spain.
| | - Amit Khairnar
- Applied Neuroscience Research Group, CEITEC - Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Nicola Simola
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
| | - Noelia Granado
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Madrid, Spain; CIBERNED, ISCIII, Madrid, Spain
| | - Jose Ruben García-Montes
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Madrid, Spain; CIBERNED, ISCIII, Madrid, Spain
| | - Pier Francesca Porceddu
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA
| | - Giulia Costa
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy; Centre of Excellence for Neurobiology of Dependence, University of Cagliari, Cagliari, Italy; National Research Council (CNR), Institute of Neuroscience, Cagliari, Italy
| |
Collapse
|