1
|
Serra M, Simola N, Pollack AE, Costa G. Brain dysfunctions and neurotoxicity induced by psychostimulants in experimental models and humans: an overview of recent findings. Neural Regen Res 2024; 19:1908-1918. [PMID: 38227515 DOI: 10.4103/1673-5374.390971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 11/10/2023] [Indexed: 01/17/2024] Open
Abstract
Preclinical and clinical studies indicate that psychostimulants, in addition to having abuse potential, may elicit brain dysfunctions and/or neurotoxic effects. Central toxicity induced by psychostimulants may pose serious health risks since the recreational use of these substances is on the rise among young people and adults. The present review provides an overview of recent research, conducted between 2018 and 2023, focusing on brain dysfunctions and neurotoxic effects elicited in experimental models and humans by amphetamine, cocaine, methamphetamine, 3,4-methylenedioxymethamphetamine, methylphenidate, caffeine, and nicotine. Detailed elucidation of factors and mechanisms that underlie psychostimulant-induced brain dysfunction and neurotoxicity is crucial for understanding the acute and enduring noxious brain effects that may occur in individuals who use psychostimulants for recreational and/or therapeutic purposes.
Collapse
Affiliation(s)
- Marcello Serra
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - Nicola Simola
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - Alexia E Pollack
- Department of Biology, University of Massachusetts-Boston, Boston, MA, USA
| | - Giulia Costa
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| |
Collapse
|
2
|
Vo TTL, Shin D, Ha E, Seo JH. Dysfunction of the Neurovascular Unit by Psychostimulant Drugs. Int J Mol Sci 2023; 24:15154. [PMID: 37894832 PMCID: PMC10606839 DOI: 10.3390/ijms242015154] [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: 08/27/2023] [Revised: 10/08/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
'Drug abuse' has been recognized as one of the most pressing epidemics in contemporary society. Traditional research has primarily focused on understanding how drugs induce neurotoxicity or degeneration within the central nervous system (CNS) and influence systems related to reward, motivation, and cravings. However, recent investigations have increasingly shifted their attention toward the detrimental consequences of drug abuse on the blood-brain barrier (BBB). The BBB is a structural component situated in brain vessels, responsible for separating brain tissue from external substances to maintain brain homeostasis. The BBB's function is governed by cellular interactions involving various elements of the 'neurovascular unit (NVU),' such as neurons, endothelial cells, astrocytes, pericytes, and microglia. Disruption of the NVU is closely linked to serious neurodegeneration. This review provides a comprehensive overview of the harmful effects of psychostimulant drugs on the BBB, highlighting the mechanisms through which drugs can damage the NVU. Additionally, the review proposes novel therapeutic targets aimed at protecting the BBB. By understanding the intricate relationships between drug abuse, BBB integrity, and NVU function, researchers and clinicians may uncover new strategies to mitigate the damaging impact of drug abuse on brain health.
Collapse
Affiliation(s)
- Tam Thuy Lu Vo
- Department of Biochemistry, Keimyung University School of Medicine, Daegu 42601, Republic of Korea; (T.T.L.V.); (E.H.)
| | - Dain Shin
- Keimyung University School of Medicine, Daegu 42601, Republic of Korea;
| | - Eunyoung Ha
- Department of Biochemistry, Keimyung University School of Medicine, Daegu 42601, Republic of Korea; (T.T.L.V.); (E.H.)
| | - Ji Hae Seo
- Department of Biochemistry, Keimyung University School of Medicine, Daegu 42601, Republic of Korea; (T.T.L.V.); (E.H.)
| |
Collapse
|
3
|
Bravo J, Magalhães C, Andrade EB, Magalhães A, Summavielle T. The impact of psychostimulants on central and peripheral neuro-immune regulation: a scoping review of cytokine profiles and their implications for addiction. Front Cell Neurosci 2023; 17:1109611. [PMID: 37305435 PMCID: PMC10251407 DOI: 10.3389/fncel.2023.1109611] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 04/06/2023] [Indexed: 06/13/2023] Open
Abstract
It is now well-accepted that psychostimulants act on glial cells causing neuroinflammation and adding to the neurotoxic effects of such substances. Neuroinflammation can be described as an inflammatory response, within the CNS, mediated through several cytokines, reactive oxygen species, chemokines and other inflammatory markers. These inflammatory players, in particular cytokines, play important roles. Several studies have demonstrated that psychostimulants impact on cytokine production and release, both centrally and at the peripheral level. Nevertheless, the available data is often contradictory. Because understanding how cytokines are modulated by psychoactive substances seems crucial to perspective successful therapeutic interventions, here, we conducted a scoping review of the available literature. We have focused on how different psychostimulants impact on the cytokine profile. Publications were grouped according to the substance addressed (methamphetamine, cocaine, methylphenidate, MDMA or other amphetamines), the type of exposure and period of evaluation (acute, short- or long-term exposure, withdrawal, and reinstatement). Studies were further divided in those addressing central cytokines, circulating (peripheral) levels, or both. Our analysis showed that the classical pro-inflammatory cytokines TNF-α, IL-6, and IL-1β were those more investigated. The majority of studies have reported increased levels of these cytokines in the central nervous system after acute or repeated drug. However, studies investigating cytokine levels during withdrawal or reinstatement have shown higher variability in their findings. Although we have identified fewer studies addressing circulating cytokines in humans, the available data suggest that the results may be more robust in animal models than in patients with problematic drug use. As a major conclusion, an extensive use of arrays for relevant cytokines should be considered to better determine which cytokines, upon the classical ones, may be involved in the progression from episodic use to the development of addiction. A concerted effort is still necessary to address the link between peripheral and central immune players, including from a longitudinal perspective. Until there, the identification of new biomarkers and therapeutic targets to envision personalized immune-based therapeutics will continue to be unlikely.
Collapse
Affiliation(s)
- Joana Bravo
- Addiction Biology, i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
- Escola Superior de Saúde, Polytechnic of Porto, Porto, Portugal
| | - Catarina Magalhães
- Addiction Biology, i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Centro Hospitalar Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
| | - Elva B. Andrade
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
- Escola Superior de Saúde, Polytechnic of Porto, Porto, Portugal
- Immunobiology, i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Ana Magalhães
- Addiction Biology, i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
- Instituto Universitário de Ciências da Saúde, Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Gandra, Portugal
| | - Teresa Summavielle
- Addiction Biology, i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Escola Superior de Saúde, Polytechnic of Porto, Porto, Portugal
| |
Collapse
|
4
|
Costa G, Serra M, Maccioni R, Casu MA, Kasture SB, Acquas E, Morelli M. Withania somnifera influences MDMA-induced hyperthermic, cognitive, neurotoxic and neuroinflammatory effects in mice. Biomed Pharmacother 2023; 161:114475. [PMID: 36905810 DOI: 10.1016/j.biopha.2023.114475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/11/2023] Open
Abstract
Withania somnifera (WS) is utilized in Ayurvedic medicine owing to its central and peripheral beneficial properties. Several studies have accrued indicating that the recreational amphetamine-related drug (+/-)- 3,4-methylenedioxymethamphetamine (MDMA; Ecstasy) targets the nigrostriatal dopaminergic system in mice, inducing neurodegeneration and gliosis, causing acute hyperthermia and cognitive impairment. This study aimed to investigate the effect of a standardized extract of W. somnifera (WSE) on MDMA-induced neurotoxicity, neuroinflammation, memory impairment and hyperthermia. Mice received a 3-day pretreatment with vehicle or WSE. Thereafter, vehicle- and WSE-pretreated mice were randomly divided into four groups: saline, WSE, MDMA alone, WSE plus MDMA. Body temperature was recorded throughout treatment, and memory performance was assessed by a novel object recognition (NOR) task at the end of treatment. Thereafter, immunohistochemistry was performed to evaluate in the substantia nigra pars compacta (SNc) and striatum the levels of tyrosine hydroxylase (TH), as marker of dopaminergic degeneration, and of glial fibrillary acidic protein (GFAP) and TMEM119, as markers of astrogliosis or microgliosis, respectively. MDMA-treated mice showed a decrease in TH-positive neurons and fibers in the SNc and striatum respectively, an increase in gliosis and body temperature, and a decrease in NOR performance, irrespective of vehicle or WSE pretreatment. Acute WSE plus MDMA counteracted the modifications in TH-positive cells in SNc, GFAP-positive cells in striatum, TMEM in both areas and NOR performance, as compared to MDMA alone, while no differences were observed as compared to saline. Results indicate that WSE acutely administered in combination with MDMA, but not as pretreatment, protects mice against the noxious central effects of MDMA.
Collapse
Affiliation(s)
- Giulia Costa
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy.
| | - Marcello Serra
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - Riccardo Maccioni
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy; Department of Immunology and Microbiology, Scripps Research, La Jolla, CA, USA
| | - Maria Antonietta Casu
- National Research Council of Italy, Institute of Translational Pharmacology, UOS of Cagliari, Scientific and Technological Park of Sardinia POLARIS, Pula, Italy
| | - Sanjay B Kasture
- Rajarshi Shahu College of Pharmacy, Buldhana, Maharashtra, India
| | - Elio Acquas
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy; National Research Council of Italy, Neuroscience Institute, Cagliari, Italy
| |
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: 0] [Impact Index Per Article: 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
|
Ruggiero M, Calvello R, Porro C, Messina G, Cianciulli A, Panaro MA. Neurodegenerative Diseases: Can Caffeine Be a Powerful Ally to Weaken Neuroinflammation? Int J Mol Sci 2022; 23:ijms232112958. [PMID: 36361750 PMCID: PMC9658704 DOI: 10.3390/ijms232112958] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/18/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022] Open
Abstract
In recent years, there has been considerable research showing that coffee consumption seems to be beneficial to human health, as it contains a mixture of different bioactive compounds such as chlorogenic acids, caffeic acid, alkaloids, diterpenes and polyphenols. Neurodegenerative diseases (NDs) are debilitating, and non-curable diseases associated with impaired central, peripheral and muscle nervous systems. Several studies demonstrate that neuroinflammation mediated by glial cells—such as microglia and astrocytes—is a critical factor contributing to neurodegeneration that causes the dysfunction of brain homeostasis, resulting in a progressive loss of structure, function, and number of neuronal cells. This happens over time and leads to brain damage and physical impairment. The most known chronic NDs are represented by Alzheimer’s disease (AD), Parkinson’s disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS) and Huntington’s disease (HD). According to epidemiological studies, regular coffee consumption is associated with a lower risk of neurodegenerative diseases. In this review, we summarize the latest research about the potential effects of caffeine in neurodegenerative disorders prevention and discuss the role of controlled caffeine delivery systems in maintaining high plasma caffeine concentrations for an extended time.
Collapse
Affiliation(s)
- Melania Ruggiero
- Department of Biosciences, Biotechnologies and Environment, University of Bari, 70125 Bari, Italy
| | - Rosa Calvello
- Department of Biosciences, Biotechnologies and Environment, University of Bari, 70125 Bari, Italy
| | - Chiara Porro
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy
| | - Giovanni Messina
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy
| | - Antonia Cianciulli
- Department of Biosciences, Biotechnologies and Environment, University of Bari, 70125 Bari, Italy
| | - Maria Antonietta Panaro
- Department of Biosciences, Biotechnologies and Environment, University of Bari, 70125 Bari, Italy
- Correspondence:
| |
Collapse
|
7
|
Magenis ML, Damiani AP, Franca IB, de Marcos PS, Effting PS, Muller AP, de Bem Silveira G, Borges Correa MEA, Medeiros EB, Silveira PCL, Budni J, Boeck CR, de Andrade VM. Behavioral, genetic and biochemical changes in the brain of the offspring of female mice treated with caffeine during pregnancy and lactation. Reprod Toxicol 2022; 112:119-135. [PMID: 35868513 DOI: 10.1016/j.reprotox.2022.07.005] [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: 01/17/2022] [Revised: 07/07/2022] [Accepted: 07/16/2022] [Indexed: 11/17/2022]
Abstract
The intrauterine environment is a critical location for exposure to exogenous and endogenous factors that trigger metabolic changes through fetal programming. Among the external factors, chemical compounds stand out, which include caffeine, since its consumption is common among women, including during pregnancy. Thereby, the aim of the present study was to evaluate the behavioral, genetic, and biochemical parameters in the offspring of female mice treated with caffeine during pregnancy and lactation. Swiss female mice (60 days old) received tap water or caffeine at 0.3 or 1.0 mg/mL during copulation (7 days), pregnancy (21 days) and lactation (21 days). After the end of the lactation period, the offspring were divided into groups (water, caffeine 0.3 or 1.0 mg/mL) with 20 animals (10 animals aged 30 days and 10 animals aged 60 days per group per sex). Initially, the offspring were submitted to behavioral tasks and then euthanized for genetic and biochemical analysis in the brain (cortex, striatum, and hippocampus). Behavioral changes in memory, depression, and anxiety were observed in the offspring: 30-day-old female offspring at 1.0 mg /mL dose presented anxiogenic behavior and male offspring the 0.3 mg/mL dose at 30 days of age did not alter long-term memory. Furthermore, an increase in DNA damage and oxidative stress in the brain were observed in the offspring of both sexes. Furthermore, there were changes in Ape-1, BAX, and Bcl-2 in the female offspring hippocampus at 30 days of life. Thus, with this study, we can suggest genotoxicity, oxidative stress, and behavioral changes caused by caffeine during pregnancy and lactation in the offspring that were not treated directly, but received through their mothers; thus, it is important to raise awareness regarding caffeine consumption among pregnant and lactating females.
Collapse
Affiliation(s)
- Marina Lummertz Magenis
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Adriani Paganini Damiani
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Ive Bahia Franca
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Pamela Souza de Marcos
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Pauline Souza Effting
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Alexandre Pastoris Muller
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Gustavo de Bem Silveira
- Laboratory of Experimental Pathophysiology, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Maria Eduarda Anastácio Borges Correa
- Laboratory of Experimental Pathophysiology, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Eduarda Behenck Medeiros
- Laboratory of Experimental Neurology, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Paulo Cesar Lock Silveira
- Laboratory of Experimental Pathophysiology, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Josiane Budni
- Laboratory of Experimental Neurology, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil
| | - Carina Rodrigues Boeck
- Graduate Program in Nanosciences, Franciscan University Center - UNIFRA, Santa Maria, RS, Brazil
| | - Vanessa Moraes de Andrade
- Laboratory of Translational Biomedicine, Graduate Program of Health Sciences, University of Southern Santa Catarina - UNESC, Criciúma, SC, Brazil.
| |
Collapse
|
8
|
Epigenetic Studies for Evaluation of NPS Toxicity: Focus on Synthetic Cannabinoids and Cathinones. Biomedicines 2022; 10:biomedicines10061398. [PMID: 35740419 PMCID: PMC9219842 DOI: 10.3390/biomedicines10061398] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/02/2022] [Accepted: 06/06/2022] [Indexed: 11/26/2022] Open
Abstract
In the recent decade, numerous new psychoactive substances (NPSs) have been added to the illicit drug market. These are synthetized to mimic the effects of classic drugs of abuse (i.e., cannabis, cocaine, etc.), with the purpose of bypassing substance legislations and increasing the pharmacotoxicological effects. To date, research into the acute pharmacological effects of new NPSs is ongoing and necessary in order to provide an appropriate contribution to public health. In fact, multiple examples of NPS-related acute intoxication and mortality have been recorded in the literature. Accordingly, several in vitro and in vivo studies have investigated the pharmacotoxicological profiles of these compounds, revealing that they can cause adverse effects involving various organ systems (i.e., cardiovascular, respiratory effects) and highlighting their potential increased consumption risks. In this sense, NPSs should be regarded as a complex issue that requires continuous monitoring. Moreover, knowledge of long-term NPS effects is lacking. Because genetic and environmental variables may impact NPS responses, epigenetics may aid in understanding the processes behind the harmful events induced by long-term NPS usage. Taken together, “pharmacoepigenomics” may provide a new field of combined study on genetic differences and epigenetic changes in drug reactions that might be predictive in forensic implications.
Collapse
|
9
|
Costa G, Gołembiowska K. Neurotoxicity of MDMA: Main effects and mechanisms. Exp Neurol 2021; 347:113894. [PMID: 34655576 DOI: 10.1016/j.expneurol.2021.113894] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 10/01/2021] [Accepted: 10/08/2021] [Indexed: 01/19/2023]
Abstract
Preclinical and clinical studies indicate that 3,4-methylenedioxymethamphetamine (MDMA; 'ecstasy'), in addition to having abuse potential, may elicit acute and persistent abnormalities of varying severity at the central level. Importantly, neurotoxic effects of MDMA have been demonstrated in experimental animals. Accordingly, central toxicity induced by MDMA may pose a serious harm for health, since MDMA is among the substances that are used for recreational purposes by young and adult people. This review provides a concise overview of recent findings from preclinical and clinical studies that evaluated the central effects of MDMA, and the mechanisms involved in the neurotoxicity induced by this amphetamine-related drug.
Collapse
Affiliation(s)
- Giulia Costa
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy.
| | - Krystyna Gołembiowska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, 12 Smętna, 31-343 Kraków, Poland
| |
Collapse
|
10
|
Costa G, Caputi FF, Serra M, Simola N, Rullo L, Stamatakos S, Sanna F, Germain M, Martinoli MG, Candeletti S, Morelli M, Romualdi P. Activation of Antioxidant and Proteolytic Pathways in the Nigrostriatal Dopaminergic System After 3,4-Methylenedioxymethamphetamine Administration: Sex-Related Differences. Front Pharmacol 2021; 12:713486. [PMID: 34512343 PMCID: PMC8430399 DOI: 10.3389/fphar.2021.713486] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 07/12/2021] [Indexed: 12/29/2022] Open
Abstract
3,4-Methylenedioxymethamphetamine (MDMA, “ecstasy”) is an amphetamine-related drug that may damage the dopaminergic nigrostriatal system. To investigate the mechanisms that sustain this toxic effect and ascertain their sex-dependence, we evaluated in the nigrostriatal system of MDMA-treated (4 × 20 mg/kg, 2 h apart) male and female mice the activity of superoxide dismutase (SOD), the gene expression of SOD type 1 and 2, together with SOD1/2 co-localization with tyrosine hydroxylase (TH)-positive neurons. In the same mice and brain areas, activity of glutathione peroxidase (GPx) and of β2/β5 subunits of the ubiquitin-proteasome system (UPS) were also evaluated. After MDMA, SOD1 increased in striatal TH-positive terminals, but not nigral neurons, of males and females, while SOD2 increased in striatal TH-positive terminals and nigral neurons of males only. Moreover, after MDMA, SOD1 gene expression increased in the midbrain of males and females, whereas SOD2 increased only in males. Finally, MDMA increased the SOD activity in the midbrain of females, without affecting GPx activity, decreased the β2/β5 activities in the striatum of males and the β2 activity in the midbrain of females. These results suggest that the mechanisms of MDMA-induced neurotoxic effects are sex-dependent and dopaminergic neurons of males could be more sensitive to SOD2- and UPS-mediated toxic effects.
Collapse
Affiliation(s)
- Giulia Costa
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - Francesca Felicia Caputi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Marcello Serra
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - Nicola Simola
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - Laura Rullo
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Serena Stamatakos
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Fabrizio Sanna
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - Marc Germain
- Department of Medical Biology, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada.,CERMO-FC UQAM, Québec, QC, Canada
| | - Maria-Grazia Martinoli
- Department of Medical Biology, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada.,Department of Psychiatry and Neuroscience, Université Laval and CHU Research Center, Québec, QC, Canada
| | - Sanzio Candeletti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy.,National Research Council of Italy, Neuroscience Institute, Cagliari, Italy
| | - Patrizia Romualdi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| |
Collapse
|
11
|
Costa G, Spulber S, Paci E, Casu MA, Ceccatelli S, Simola N, Morelli M. In utero exposure to dexamethasone causes a persistent and age-dependent exacerbation of the neurotoxic effects and glia activation induced by MDMA in dopaminergic brain regions of C57BL/6J mice. Neurotoxicology 2021; 83:1-13. [PMID: 33338551 DOI: 10.1016/j.neuro.2020.12.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 11/03/2020] [Accepted: 12/04/2020] [Indexed: 12/22/2022]
Abstract
Clinical and preclinical evidence indicates that prenatal exposure to glucocorticoids may induce detrimental effects in the offspring, including reduction in fetal growth and alterations in the CNS. On this basis, the present study investigated whether in utero exposure to high levels of glucocorticoids is a risk factor that may lead to an exacerbation of the central noxious effects induced by psychoactive drugs consumed later in life. To this end, pregnant C57BL6/J dams were treated with dexamethasone (DEX, 0.05 mg/kg per day) from gestational day 14 until delivery. Thereafter, the male offspring were evaluated to ascertain the magnitude of dopaminergic damage, astrogliosis and microgliosis elicited in the nigrostriatal tract by the amphetamine-related drug 3,4--methylenedioxymethamphetamine (MDMA, 4 × 20 mg/kg, 2 h apart, sacrificed 48 h later) administered at either adolescence or adulthood. Immunohistochemistry was performed in the substantia nigra pars compacta (SNc) and striatum, to evaluate dopaminergic degeneration by measuring tyrosine hydroxylase (TH), as well as astrogliosis and microgliosis by measuring glial fibrillary acidic protein (GFAP) and ionized calcium-binding adapter molecule 1 (IBA-1), respectively. Moreover, immunohistochemistry was used to ascertain the co-localization of IBA-1 with either the pro-inflammatory interleukin (IL) IL-1β or the anti-inflammatory IL IL-10, in order to determine the microglial phenotype. In utero administration of DEX induced dopaminergic damage by decreasing the density of TH-positive fibers in the striatum, although only in adult mice. MDMA administration induced dopaminergic damage and glia activation in the nigrostriatal tract of adolescent and adult mice. Mice exposed to DEX in utero and treated with MDMA later in life showed a more pronounced loss of dopaminergic neurons (adolescent mice) and astrogliosis (adolescent and adult mice) in the SNc, compared with control mice. These results suggest that prenatal exposure to glucocorticoids may induce an age-dependent and persistent increase in the susceptibility to central toxicity of amphetamine-related drugs used later in life.
Collapse
Affiliation(s)
- Giulia Costa
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy.
| | - Stefan Spulber
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Elena Paci
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Maria Antonietta Casu
- National Research Council of Italy, Institute of Translational Pharmacology, UOS of Cagliari, Scientific and Technological Park of Sardinia POLARIS, Pula, Italy
| | - Sandra Ceccatelli
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Nicola Simola
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy; National Research Council of Italy, Neuroscience Institute, Cagliari, Italy
| |
Collapse
|
12
|
Do caffeine and more selective adenosine A 2A receptor antagonists protect against dopaminergic neurodegeneration in Parkinson's disease? Parkinsonism Relat Disord 2020; 80 Suppl 1:S45-S53. [PMID: 33349580 PMCID: PMC8102090 DOI: 10.1016/j.parkreldis.2020.10.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 09/26/2020] [Accepted: 10/11/2020] [Indexed: 12/15/2022]
Abstract
The adenosine A2A receptor is a major target of caffeine, the most widely used psychoactive substance worldwide. Large epidemiological studies have long shown caffeine consumption is a strong inverse predictor of Parkinson’s disease (PD). In this review, we first examine the epidemiology of caffeine use vis-à-vis PD and follow this by looking at the evidence for adenosine A2A receptor antagonists as potential neuroprotective agents. There is a wealth of accumulating biological, epidemiological and clinical evidence to support the further investigation of selective adenosine A2A antagonists, as well as caffeine, as promising candidate therapeutics to fill the unmet need for disease modification of PD.
Collapse
|
13
|
Ren X, Chen JF. Caffeine and Parkinson's Disease: Multiple Benefits and Emerging Mechanisms. Front Neurosci 2020; 14:602697. [PMID: 33390888 PMCID: PMC7773776 DOI: 10.3389/fnins.2020.602697] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disorder, characterized by dopaminergic neurodegeneration, motor impairment and non-motor symptoms. Epidemiological and experimental investigations into potential risk factors have firmly established that dietary factor caffeine, the most-widely consumed psychoactive substance, may exerts not only neuroprotective but a motor and non-motor (cognitive) benefits in PD. These multi-benefits of caffeine in PD are supported by convergence of epidemiological and animal evidence. At least six large prospective epidemiological studies have firmly established a relationship between increased caffeine consumption and decreased risk of developing PD. In addition, animal studies have also demonstrated that caffeine confers neuroprotection against dopaminergic neurodegeneration using PD models of mitochondrial toxins (MPTP, 6-OHDA, and rotenone) and expression of α-synuclein (α-Syn). While caffeine has complex pharmacological profiles, studies with genetic knockout mice have clearly revealed that caffeine’s action is largely mediated by the brain adenosine A2A receptor (A2AR) and confer neuroprotection by modulating neuroinflammation and excitotoxicity and mitochondrial function. Interestingly, recent studies have highlighted emerging new mechanisms including caffeine modulation of α-Syn degradation with enhanced autophagy and caffeine modulation of gut microbiota and gut-brain axis in PD models. Importantly, since the first clinical trial in 2003, United States FDA has finally approved clinical use of the A2AR antagonist istradefylline for the treatment of PD with OFF-time in Sept. 2019. To realize therapeutic potential of caffeine in PD, genetic study of caffeine and risk genes in human population may identify useful pharmacogenetic markers for predicting individual responses to caffeine in PD clinical trials and thus offer a unique opportunity for “personalized medicine” in PD.
Collapse
Affiliation(s)
- Xiangpeng Ren
- Molecular Neuropharmacology Lab, School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, China.,Department of Biochemistry, Medical College, Jiaxing University, Jiaxing, China
| | - Jiang-Fan Chen
- Molecular Neuropharmacology Lab, School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, China
| |
Collapse
|
14
|
Costa G, Sisalli MJ, Simola N, Della Notte S, Casu MA, Serra M, Pinna A, Feliciello A, Annunziato L, Scorziello A, Morelli M. Gender Differences in Neurodegeneration, Neuroinflammation and Na +-Ca 2+ Exchangers in the Female A53T Transgenic Mouse Model of Parkinson's Disease. Front Aging Neurosci 2020; 12:118. [PMID: 32477098 PMCID: PMC7232579 DOI: 10.3389/fnagi.2020.00118] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 04/08/2020] [Indexed: 12/20/2022] Open
Abstract
Twelve-month-old male mice expressing the human A53T variant of α-synuclein (A53T) develop dopamine neuron degeneration, neuroinflammation, and motor deficits, along with dysfunctions of the mitochondrial Na+-Ca2+ exchanger (NCX) isoforms 1 (NCX1) and 3 (NCX3) in the nigrostriatal system. Since gender is thought to play a role in the etiology of Parkinson's disease (PD), we characterized neurochemical and behavioral alterations in 12-month-old female A53T transgenic mice. We investigated the presence of dopaminergic degeneration, astrogliosis and microgliosis using immunohistochemistry for tyrosine hydroxylase (TH), glial fibrillary acidic protein (GFAP) and ionized calcium-binding adaptor molecule-1 (IBA-1) in both the substantia nigra pars compacta (SNc) and striatum. In the same regions, we also evaluated the co-localization of NCX1 in cells positive for IBA-1 and the co-localization of NCX3 in TH-positive neurons and fibers. Furthermore, in both male and female mice, we performed motor (beam walking and pole tests) and memory [novel object recognition (NOR) and spontaneous alternation] tasks, together with tests to evaluate peripheral deficits (olfactory and stool collection tests). Female A53T transgenic mice displayed degeneration of nigral dopaminergic neurons, but neither microgliosis nor astrogliosis in the SNc and striatum. Moreover, female A53T transgenic mice displayed co-localization between NCX1 and IBA-1 positive cells in the striatum but not SNc, whereas NCX3 did not co-localize with either TH-positive terminals or neuronal bodies in the nigrostriatal system. Furthermore, female A53T transgenic mice showed increased crossing time in the beam walking test, but no impairments in the pole or memory tests, and in tests that evaluated peripheral deficits, whereas male A53T transgenic mice displayed motor, memory and peripheral deficits. Immunohistochemical and behavioral results obtained here in the female mice differ from those previously observed in males, and suggest a dissimilar influence of NCX1 and NCX3 on dopaminergic function in female and male A53T transgenic mice, strengthening the validity of these mice as a model for studying the etiological factors of PD.
Collapse
Affiliation(s)
- Giulia Costa
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - Maria Jose Sisalli
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, University of Naples Federico II, Naples, Italy
| | - Nicola Simola
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy.,National Institute of Neuroscience (INN), University of Cagliari, Cagliari, Italy
| | - Salvatore Della Notte
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, University of Naples Federico II, Naples, Italy
| | - Maria Antonietta Casu
- National Research Council of Italy, Institute of Translational Pharmacology, UOS of Cagliari, Scientific and Technological Park of Sardinia POLARIS, Pula, Italy
| | - Marcello Serra
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy
| | - Annalisa Pinna
- National Research Council of Italy, Neuroscience Institute, Cagliari, Italy
| | - Antonio Feliciello
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Lucio Annunziato
- SDN Research Institute Diagnostics and Nuclear (IRCCS SDN), Naples, Italy
| | - Antonella Scorziello
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, University of Naples Federico II, Naples, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy.,National Institute of Neuroscience (INN), University of Cagliari, Cagliari, Italy.,National Research Council of Italy, Neuroscience Institute, Cagliari, Italy
| |
Collapse
|
15
|
Aguilar MA, García-Pardo MP, Parrott AC. Of mice and men on MDMA: A translational comparison of the neuropsychobiological effects of 3,4-methylenedioxymethamphetamine ('Ecstasy'). Brain Res 2020; 1727:146556. [PMID: 31734398 DOI: 10.1016/j.brainres.2019.146556] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 11/09/2019] [Accepted: 11/12/2019] [Indexed: 11/19/2022]
Abstract
MDMA (3,4-methylendioxymethamphetamine), also known as Ecstasy, is a stimulant drug recreationally used by young adults usually in dance clubs and raves. Acute MDMA administration increases serotonin, dopamine and noradrenaline by reversing the action of the monoamine transporters. In this work, we review the studies carried out over the last 30 years on the neuropsychobiological effects of MDMA in humans and mice and summarise the current knowledge. The two species differ with respect to the neurochemical consequences of chronic MDMA, since it preferentially induces serotonergic dysfunction in humans and dopaminergic neurotoxicity in mice. However, MDMA alters brain structure and function and induces hormonal, psychomotor, neurocognitive, psychosocial and psychiatric outcomes in both species, as well as physically damaging and teratogen effects. Pharmacological and genetic studies in mice have increased our knowledge of the neurochemical substrate of the multiple effects of MDMA. Future work in this area may contribute to developing pharmacological treatments for MDMA-related disorders.
Collapse
Affiliation(s)
- Maria A Aguilar
- Department of Psychobiology, Faculty of Psychology, Valencia University, Valencia, Spain.
| | | | - Andrew C Parrott
- Department of Psychology, Swansea University, Swansea, United Kingdom; Centre for Human Psychopharmacology, Swinburne University, Melbourne, Australia
| |
Collapse
|
16
|
Costa G, De Luca MA, Piras G, Marongiu J, Fattore L, Simola N. Neuronal and peripheral damages induced by synthetic psychoactive substances: an update of recent findings from human and animal studies. Neural Regen Res 2020; 15:802-816. [PMID: 31719240 PMCID: PMC6990793 DOI: 10.4103/1673-5374.268895] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Preclinical and clinical studies indicate that synthetic psychoactive substances, in addition to having abuse potential, may elicit toxic effects of varying severity at the peripheral and central levels. Nowadays, toxicity induced by synthetic psychoactive substances poses a serious harm for health, since recreational use of these substances is on the rise among young and adult people. The present review summarizes recent findings on the peripheral and central toxicity elicited by “old” and “new” synthetic psychoactive substances in humans and experimental animals, focusing on amphetamine derivatives, hallucinogen and dissociative drugs and synthetic cannabinoids.
Collapse
Affiliation(s)
- Giulia Costa
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Maria Antonietta De Luca
- Department of Biomedical Sciences; National Institute of Neuroscience (INN), University of Cagliari, Cagliari, Italy
| | - Gessica Piras
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Jacopo Marongiu
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Liana Fattore
- National Research Council of Italy, Institute of Neuroscience, Cagliari, Italy
| | - Nicola Simola
- Department of Biomedical Sciences; National Institute of Neuroscience (INN), University of Cagliari, Cagliari, Italy
| |
Collapse
|
17
|
Lack of Rhes Increases MDMA-Induced Neuroinflammation and Dopamine Neuron Degeneration: Role of Gender and Age. Int J Mol Sci 2019; 20:ijms20071556. [PMID: 30925704 PMCID: PMC6480667 DOI: 10.3390/ijms20071556] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/21/2019] [Accepted: 03/24/2019] [Indexed: 02/06/2023] Open
Abstract
Ras homolog enriched in striatum (Rhes) is a protein that exerts important physiological functions and modulates psychostimulant drug effects. On this basis, the object of this study was to assess 3,4-methylenedioxymethamphetamine (MDMA) effects on microglial (CD11b) and astroglial (GFAP) activation and on dopamine neuron degeneration (TH) in wild-type (WT) and Rhes knockout (KO) male and female mice of different ages. Motor activity was also evaluated. Adult (3 months) MDMA-treated mice displayed an increase in GFAP-positive cells in striatum (STR), whereas the substantia nigra pars compacta (SNc) was affected only in male mice. In these mice, the increase of CD11b was more extensive including STR, SNc, motor cortex (CTX), ventral tegmental area (VTA), and nucleus accumbens (NAc). MDMA administration also affected TH immunoreactivity in both STR and SNc of male but not female WT and Rhes KO mice. In middle-aged mice (12 months), MDMA administration further increased GFAP and CD11b and decreased TH immunoreactivity in STR and SNc of all mice. Finally, MDMA induced a higher increase of motor activity in adult Rhes KO male, but not female mice. The results show that Rhes protein plays an important role on MDMA-mediated neuroinflammation and neurodegeneration dependent on gender and age, and confirm the important role of Rhes protein in neuroinflammatory and neurodegenerative processes.
Collapse
|
18
|
Schiavone S, Neri M, Maffione AB, Frisoni P, Morgese MG, Trabace L, Turillazzi E. Increased iNOS and Nitrosative Stress in Dopaminergic Neurons of MDMA-Exposed Rats. Int J Mol Sci 2019; 20:E1242. [PMID: 30871034 PMCID: PMC6429174 DOI: 10.3390/ijms20051242] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/28/2019] [Accepted: 03/05/2019] [Indexed: 12/29/2022] Open
Abstract
Several mechanisms underlying 3,4-Methylenedioxy-N-methylamphetamine (MDMA) neurotoxicity have been proposed, including neurochemical alterations and excitotoxicity mediated by reactive oxygen species (ROS), nitric oxide (NO), and reactive nitrogen species (RNS). However, ROS, NO, and RNS sources in the brain are not fully known. We aimed to investigate possible alterations in the expression of the ROS producer NOX enzymes (NOX2, NOX1, and NOX4), NO generators (iNOS, eNOS, and nNOS), markers of oxidative (8-hydroxy-2'-deoxyguanosine, 8OHdG), and nitrosative (3-nitrotyrosine, NT) stress, as well as the colocalization between cells positive for the dopamine transporter (DT1) and cells expressing the neuronal nuclei (NeuN) marker, in the frontal cortex of rats receiving saline or MDMA, sacrificed 6 h, 16 h, or 24 h after its administration. MDMA did not affect NOX2, NOX1, and NOX4 immunoreactivity, whereas iNOS expression was enhanced. The number of NT-positive cells was increased in MDMA-exposed animals, whereas no differences were detected in 8OHdG expression among experimental groups. MDMA and NT markers colocalized with DT1 positive cells. DT1 immunostaining was found in NeuN-positive stained cells. Virtually no colocalization was observed with microglia and astrocytes. Moreover, MDMA immunostaining was not found in NOX2-positive cells. Our results suggest that iNOS-derived nitrosative stress, but not NOX enzymes, may have a crucial role in the pathogenesis of MDMA-induced neurotoxicity, highlighting the specificity of different enzymatic systems in the development of neuropathological alterations induced by the abuse of this psychoactive compound.
Collapse
Affiliation(s)
- Stefania Schiavone
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli, 20, 71122 Foggia, Italy.
| | - Margherita Neri
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Fossato di Mortara, 70, 44100 Ferrara, Italy.
| | - Angela Bruna Maffione
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli, 20, 71122 Foggia, Italy.
| | - Paolo Frisoni
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Fossato di Mortara, 70, 44100 Ferrara, Italy.
| | - Maria Grazia Morgese
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli, 20, 71122 Foggia, Italy.
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli, 20, 71122 Foggia, Italy.
| | - Emanuela Turillazzi
- Section of Legal Medicine, Department of Surgical, Medical, Molecular and Critical Pathology, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| |
Collapse
|
19
|
Costa G, Serra M, Pintori N, Casu MA, Zanda MT, Murtas D, De Luca MA, Simola N, Fattore L. The novel psychoactive substance methoxetamine induces persistent behavioral abnormalities and neurotoxicity in rats. Neuropharmacology 2019; 144:219-232. [DOI: 10.1016/j.neuropharm.2018.10.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 10/01/2018] [Accepted: 10/21/2018] [Indexed: 10/28/2022]
|
20
|
Bessada SM, Alves RC, Oliveira MBP. Caffeine-based food supplements and beverages: Trends of consumption for performance purposes and safety concerns. Food Res Int 2018; 109:310-319. [DOI: 10.1016/j.foodres.2018.04.050] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 04/20/2018] [Accepted: 04/22/2018] [Indexed: 01/12/2023]
|
21
|
Costa G, Pinna A, Porceddu PF, Casu MA, Di Maio A, Napolitano F, Usiello A, Morelli M. Rhes Counteracts Dopamine Neuron Degeneration and Neuroinflammation Depending on Gender and Age. Front Aging Neurosci 2018; 10:163. [PMID: 29904346 PMCID: PMC5990628 DOI: 10.3389/fnagi.2018.00163] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 05/15/2018] [Indexed: 01/11/2023] Open
Abstract
We have recently shown that male Rhes knockout (KO) mice develop a mild form of spontaneous Parkinson’s disease (PD)-like phenotype, characterized by motor impairment and a decrease in nigrostriatal dopamine (DA) neurons. Experimental evidence has implicated neuroinflammation in PD progression, and the presence of activated glial cells has been correlated with DA neuron degeneration. Despite this, several factors, such as gender, have been found to affect DAergic neuron degeneration and influence neuroinflammation, explaining the differences between men and women in the etiology of PD. On these basis, we studied age and gender differences in DA neuron degeneration and gliosis in the nigrostriatal system of adult (3-month-old) and middle aged (12-month-old) male and female Rhes wild-type (WT) and KO mice. Through immunohistochemistry, tyrosine hydroxylase (TH), microglial (complement type 3 receptor [CD11b]) and astroglial (glial fibrillary acid protein [GFAP]) increase, were evaluated. Adult male Rhes KO mice showed a decrease in TH and an increase in CD11b, both in the caudate putamen (CPu) and substantia nigra pars compacta (SNc), and an increase in GFAP in the CPu. In contrast, adult female Rhes KO mice showed only a decrease in TH in the SNc, whereas no modifications to the levels of GFAP and CD11b were observed in the CPu or SNc. Middle aged male Rhes KO mice showed a decrease in TH in the CPu and SNc, and an increase in GFAP and CD11b in the SNc. Middle aged female Rhes KO mice showed a decrease in TH in the CPu and SNc and an increase in CD11b only in the CPu, but no modifications to GFAP levels. The more marked DA neuron degeneration and neuroinflammation in male compared with female Rhes KO mice, while confirming the role of Rhes as an important protein for DA neuron survival, gives support to Rhes KO mice as a valuable preclinical model for studying the vulnerability factors of DA neuron degeneration as in PD.
Collapse
Affiliation(s)
- Giulia Costa
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Cagliari, Italy
| | - Annalisa Pinna
- National Research Council of Italy, Neuroscience Institute, Cagliari, Italy
| | - Pier Francesca Porceddu
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Cagliari, Italy
| | - Maria Antonietta Casu
- National Research Council of Italy, Institute of Translational Pharmacology, UOS of Cagliari, Scientific and Technological Park of Sardinia POLARIS, Pula, Italy
| | - Anna Di Maio
- IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Francesco Napolitano
- Laboratory of Behavioral Neuroscience, Ceinge Biotecnologie Avanzate, Naples, Italy.,Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Alessandro Usiello
- Laboratory of Behavioral Neuroscience, Ceinge Biotecnologie Avanzate, Naples, Italy.,Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania, Caserta, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Cagliari, Italy.,National Research Council of Italy, Neuroscience Institute, Cagliari, Italy.,National Institute of Neuroscience (INN), University of Cagliari, Cagliari, Italy
| |
Collapse
|
22
|
Costa G, Morelli M, Simola N. Repeated Administration of 3,4-Methylenedioxymethamphetamine (MDMA) Elevates the Levels of Neuronal Nitric Oxide Synthase in the Nigrostriatal System: Possible Relevance to Neurotoxicity. Neurotox Res 2018; 34:763-768. [PMID: 29629511 DOI: 10.1007/s12640-018-9892-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 03/01/2018] [Accepted: 03/20/2018] [Indexed: 12/12/2022]
Abstract
Previous studies have consistently demonstrated that the amphetamine-related drug 3,4-methylenedioxymethamphetamine (MDMA) induces dopaminergic damage in the mouse brain, and that this effect is most marked in the nigrostriatal system. Moreover, it has been suggested that the overproduction of nitric oxide (NO) may participate in the dopaminergic damage induced by MDMA. To further elucidate this issue, we evaluated the levels of the enzyme nitric oxide synthase (nNOS), which catalyzes the production of NO, in mice treated with regimens of MDMA that induce progressive and persistent neurotoxicity in the dopaminergic nigrostriatal system. Mice received 14, 28, or 36 administrations of MDMA (10 mg/kg i.p.), twice a day/twice a week, and were sacrificed at different time-points after treatment discontinuation. Thereafter, the number of nNOS-positive neurons was quantified by immunohistochemistry in the caudate-putamen (CPu) and substantia nigra pars compacta (SNc). MDMA elevated the numbers of nNOS-positive neurons in the CPu of mice that received 28 or 36 drug administrations. This effect was still detectable at 3 months after treatment discontinuation. Moreover, MDMA elevated the numbers of nNOS-positive neurons in the SNc. However, this effect occurred only in mice that received 28 drug administrations and were sacrificed 3 days after treatment discontinuation. These results are in line with the hypothesis that activation of the NO cascade participates in the toxic effects induced by MDMA in the dopaminergic nigrostriatal system. Moreover, they suggest that activation of the NO cascade induces toxic effects that are more marked in striatal terminals, compared with nigral neurons.
Collapse
Affiliation(s)
- Giulia Costa
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Building A, Monserrato University Campus, SP 8, Km 0.700, 09042, Monserrato, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Building A, Monserrato University Campus, SP 8, Km 0.700, 09042, Monserrato, Italy. .,National Research Council of Italy (CNR), Neuroscience Institute, Cagliari, Italy. .,National Institute of Neuroscience (INN), University of Cagliari, Cagliari, Italy.
| | - Nicola Simola
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Building A, Monserrato University Campus, SP 8, Km 0.700, 09042, Monserrato, Italy.,National Institute of Neuroscience (INN), University of Cagliari, Cagliari, Italy
| |
Collapse
|
23
|
Górska AM, Kamińska K, Wawrzczak-Bargieła A, Costa G, Morelli M, Przewłocki R, Kreiner G, Gołembiowska K. Neurochemical and Neurotoxic Effects of MDMA (Ecstasy) and Caffeine After Chronic Combined Administration in Mice. Neurotox Res 2018; 33:532-548. [PMID: 29134560 PMCID: PMC5871650 DOI: 10.1007/s12640-017-9831-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 10/04/2017] [Accepted: 10/18/2017] [Indexed: 02/04/2023]
Abstract
MDMA (3,4-methylenedioxymethamphetamine) is a psychostimulant popular as a recreational drug because of its effect on mood and social interactions. MDMA acts at dopamine (DA) transporter (DAT) and serotonin (5-HT) transporter (SERT) and is known to induce damage of dopamine and serotonin neurons. MDMA is often ingested with caffeine. Caffeine as a non-selective adenosine A1/A2A receptor antagonist affects dopaminergic and serotonergic transmissions. The aim of the present study was to determine the changes in DA and 5-HT release in the mouse striatum induced by MDMA and caffeine after their chronic administration. To find out whether caffeine aggravates MDMA neurotoxicity, the content of DA and 5-HT, density of brain DAT and SERT, and oxidative damage of nuclear DNA were determined. Furthermore, the effect of caffeine on MDMA-induced changes in striatal dynorphin and enkephalin and on behavior was assessed. The DA and 5-HT release was determined with in vivo microdialysis, and the monoamine contents were measured by HPLC with electrochemical detection. DNA damage was assayed with the alkaline comet assay. DAT and SERT densities were determined by immunohistochemistry, while prodynorphin (PDYN) and proenkephalin were determined by quantitative PCR reactions. The behavioral changes were measured by the open-field (OF) test and novel object recognition (NOR) test. Caffeine potentiated MDMA-induced DA release while inhibiting 5-HT release in the mouse striatum. Caffeine also exacerbated the oxidative damage of nuclear DNA induced by MDMA but diminished DAT decrease in the striatum and worsened a decrease in SERT density produced by MDMA in the frontal cortex. Neither the striatal PDYN expression, increased by MDMA, nor exploratory and locomotor activities of mice, decreased by MDMA, were affected by caffeine. The exploration of novel object in the NOR test was diminished by MDMA and caffeine. Our data provide evidence that long-term caffeine administration has a powerful influence on functions of dopaminergic and serotonergic neurons in the mouse brain and on neurotoxic effects evoked by MDMA.
Collapse
Affiliation(s)
- Anna Maria Górska
- Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12 Street, 31-343, Kraków, Poland
| | - Katarzyna Kamińska
- Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12 Street, 31-343, Kraków, Poland
| | - Agnieszka Wawrzczak-Bargieła
- Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Giulia Costa
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Cagliari, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Cagliari, Italy
| | - Ryszard Przewłocki
- Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Grzegorz Kreiner
- Department of Brain Biochemistry, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Krystyna Gołembiowska
- Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12 Street, 31-343, Kraków, Poland.
| |
Collapse
|
24
|
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: 1.6] [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
|
25
|
Selected Literature Watch. JOURNAL OF CAFFEINE RESEARCH 2016. [DOI: 10.1089/jcr.2016.29000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
26
|
Muñiz JA, Gomez G, González B, Rivero-Echeto MC, Cadet JL, García-Rill E, Urbano FJ, Bisagno V. Combined Effects of Simultaneous Exposure to Caffeine and Cocaine in the Mouse Striatum. Neurotox Res 2016; 29:525-38. [PMID: 26858178 DOI: 10.1007/s12640-016-9601-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 01/08/2016] [Accepted: 01/25/2016] [Indexed: 12/31/2022]
Abstract
Caffeine is the world's most popular psychoactive drug and is also an active adulterant found in many drugs of abuse, including seized cocaine samples. Despite several studies which examine the effects of caffeine or cocaine administered as single agents, little data are available for these agents when given in combination. The purpose of the present study was to determine if combined intake of both psychostimulants can lead to maladaptive changes in striatal function. Mice were injected with a binge regimen (intermittent treatment for 13 days) of caffeine (3 × 5 mg/kg), cocaine (3 × 10 mg/kg), or combined administration. We found that chronic caffeine potentiated locomotion induced by cocaine and that both caffeine-treated groups showed sensitization. Striatal tissue was obtained 24 h and 7 days after last injection (withdrawal) for immunohistochemistry and mRNA expression. Our results show that combined intake of both psychostimulants can increase GFAP immunoreactivity in the striatum at both times post treatment. Gene expression analysis, targeted at dopamine, adenosine, and glutamate receptor subunit genes, revealed significant transcript down-regulation in the dorsal striatum of AMPA, NMDA, D1 and D2 receptor subunit mRNA expression in the group that received combined treatment, but not after individual administration. At withdrawal, we found increased D1 receptor mRNA expression along with increased A1, AMPA, NMDA, and metabotropic subunit expression. A2A mRNA showed decreased expression after both times in all experimental groups. Our study provides evidence that there are striatal alterations mediated by combined caffeine and cocaine administration, and highlights negative outcomes of chronic intake of both psychostimulants.
Collapse
Affiliation(s)
- Javier A Muñiz
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Gimena Gomez
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Betina González
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - María Celeste Rivero-Echeto
- Laboratorio de Fisiología y Biología Molecular, Instituto de Fisiología, Biología Molecular y Neurociencias, Departamento de Fisiología, Biología Molecular y Celular "Dr. Hector Maldonado" (DFBMC), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas, Junín 956, piso 5, Ciudad Autónoma de Buenos Aires, C1113, Buenos Aires, Argentina
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, MD, USA
| | - Edgar García-Rill
- Department of Neurobiology and Developmental Sciences, Center for Translational Neuroscience, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Francisco J Urbano
- Laboratorio de Fisiología y Biología Molecular, Instituto de Fisiología, Biología Molecular y Neurociencias, Departamento de Fisiología, Biología Molecular y Celular "Dr. Hector Maldonado" (DFBMC), Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas, Junín 956, piso 5, Ciudad Autónoma de Buenos Aires, C1113, Buenos Aires, Argentina
| | - Veronica Bisagno
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|