1
|
Shirazpour S, Taheri F, Sepehri G, Zangiabadizadeh M, Zangiabadi M, Hosseini NS, Sheikhi S, Raeini AS, Tezerji SS. Amelioration of Chronic Ethanol Administration-Induced Learning and Memory Impairments by High-Intensity Interval Training (HIIT) and Ritalin. Brain Behav 2025; 15:e70539. [PMID: 40341875 PMCID: PMC12060225 DOI: 10.1002/brb3.70539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 02/24/2025] [Accepted: 04/20/2025] [Indexed: 05/11/2025] Open
Abstract
OBJECTIVES The current study aimed to investigate the impacts of 8-week high-intensity interval training (HIIT) and Ritalin (RIT), alone and in combination, on cognitive functions and hippocampal oxidative parameters following chronic ethanol consumption in male rats. METHODS A total of 56 adult male rats were divided into 8 groups and received one of the following treatments: ethanol 20% (ET) (3 mL/kg/day, orally, 5 consecutive days/week in weeks 1-4, and 3 consecutive days/week in weeks 4-8), RIT (10 mg/kg, intraperitoneally, three consecutive times/week for 8 weeks), HIIT + SAL (five consecutive times/week for 8 weeks + saline injection), or saline (1 mL/day, intraperitoneally, three consecutive times/week for 8 weeks). Cognitive performance was assessed using the Morris water maze (MWM) and passive avoidance tasks. Oxidative stress markers, including malondialdehyde (MDA), glutathione peroxidase (GPx), and total antioxidant capacity (TAC), were measured in the hippocampus using thiobarbituric acid reactive substances (TBARS) and ferric reduction antioxidant power (FRAP). Nitric oxide (NO) level in the hippocampus was determined using an NO Assay Kit (Natrix, Arman Biotech, Iran). RESULTS Chronic ethanol administration impaired cognitive functions. However, RIT, HIIT, and their combination significantly improved these impairments. Furthermore, RIT increased ethanol-induced oxidative stress, whereas HIIT reduced it, even in the combination group. CONCLUSION Chronic ethanol consumption caused learning and memory deficits and disrupted oxidant/antioxidant balance in the hippocampus of rats. HIIT potentially improved memory impairments by restoring this balance, whereas RIT ameliorated cognitive dysfunction through a mechanism that requires further investigation.
Collapse
Affiliation(s)
- Sara Shirazpour
- Institute of NeuropharmacologyNeuroscience Research CenterKerman University of Medical SciencesKermanIran
| | - Farahnaz Taheri
- Neurology Research Center, Institute of NeuropharmacologyKerman University of Medical SciencesKermanIran
| | - Gholamreza Sepehri
- Institute of NeuropharmacologyNeuroscience Research CenterKerman University of Medical SciencesKermanIran
| | - Mahla Zangiabadizadeh
- Physiology Research CenterInstitute of Neuropharmacology, Kerman University of Medical Sciences, KermanIran
| | - Mostafa Zangiabadi
- Veterinary Medicine Student, School of Veterinary MedicineShahid Bahonar University of KermanKermanIran
| | - Najmeh Sadat Hosseini
- Bone Vascular and Microcirculation LaboratoryDepartment of Kinesiology, University of Texas at Arlington, Arlington, TX, 76019USA
| | - Sara Sheikhi
- Veterinary Medicine Student, School of Veterinary MedicineShahid Bahonar University of KermanKermanIran
| | - Azadeh Shahrokhi Raeini
- Institute of NeuropharmacologyNeuroscience Research CenterKerman University of Medical SciencesKermanIran
| | - Sara Sheibani Tezerji
- Department of Behavioral and Molecular NeurobiologyRegensburg Center for NeuroscienceUniversity of RegensburgRegensburgGermany
| |
Collapse
|
2
|
Mohammadgholi-Beiki A, Aghamiri H, Rashidian R, Jafari-Sabet M, Motevalian M, Rahimi-Moghaddam P, Sheibani M. Investigation of the Protective Effects of Aripiprazole on Methylphenidate-induced Neurotoxicity in Rats. Mol Neurobiol 2025:10.1007/s12035-025-04994-3. [PMID: 40299297 DOI: 10.1007/s12035-025-04994-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/22/2025] [Indexed: 04/30/2025]
Abstract
Methylphenidate (MPH) serves as a frequently utilized stimulant in the treatment of attention deficit hyperactivity disorder (ADHD). However, long-term administration of this medication has been linked to neurotoxic sequels such as cognitive decline and increased oxidative stress, especially in the hippocampus. In contrast, aripiprazole (ARP), an atypical antipsychotic, has shown promise in providing neuroprotection and reducing inflammation. This research seeks to investigate the protective role of ARP on MPH-induced neurotoxicity in rats. A total of 40 male Wistar rats participated in this study, which were organized into five groups. All the experimental groups received daily intraperitoneal injections of MPH at 10 mg/kg, combined with varying doses of ARP at 3, 10, and 30 mg/kg for 21 days. Behavioral assessments included the open field test and forced swimming test to evaluate motor activity and depressive-like behaviors, as well as the inhibitory avoidance test for cognitive function. Seizure susceptibility was assessed using pentylenetetrazol (PTZ). Following these evaluations, hippocampal tissues were collected for biochemical analysis (oxidative stress markers and neurotrophic factor) and histopathological studies. ARP at specific doses significantly mitigated locomotor activity impairment, depressive-like behaviors, and memory deficits induced by chronic MPH administration. Additionally, ARP reduced seizure susceptibility compared to the MPH-treated group. Biochemical analyses indicated that ARP decreased oxidative stress markers, including malondialdehyde (MDA) and myeloperoxidase (MPO), while enhancing levels of glutathione and the expression of brain-derived neurotrophic factor (BDNF), nuclear factor erythroid 2-related factor 2 (NRF2), and protein kinase B (Akt) in hippocampus. Aripiprazole reduces behavioral and biochemical deficits from chronic MPH use in rats, suggesting it may help mitigate adverse effects of long-term MPH treatment.
Collapse
Affiliation(s)
- Afrooz Mohammadgholi-Beiki
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, P.O. Box: 14155-6183, Tehran, Iran
| | - Helia Aghamiri
- Department of Pharmacology and Toxicology, School of Pharmacy, Iran University of Medical Sciences, Tehran, Iran
| | - Rojin Rashidian
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, P.O. Box: 14155-6183, Tehran, Iran
| | - Majid Jafari-Sabet
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, P.O. Box: 14155-6183, Tehran, Iran
- Razi Drug Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Manijeh Motevalian
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, P.O. Box: 14155-6183, Tehran, Iran
- Razi Drug Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Parvaneh Rahimi-Moghaddam
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, P.O. Box: 14155-6183, Tehran, Iran.
| | - Mohammad Sheibani
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, P.O. Box: 14155-6183, Tehran, Iran.
- Razi Drug Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Visternicu M, Rarinca V, Burlui V, Halitchi G, Ciobică A, Singeap AM, Dobrin R, Mavroudis I, Trifan A. Investigating the Impact of Nutrition and Oxidative Stress on Attention Deficit Hyperactivity Disorder. Nutrients 2024; 16:3113. [PMID: 39339712 PMCID: PMC11435085 DOI: 10.3390/nu16183113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/30/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Background/Objectives: Attention deficit hyperactivity disorder (ADHD) is the most common childhood-onset neurodevelopmental disorder, characterized by difficulty maintaining attention, impulsivity, and hyperactivity. While the cause of this disorder is still unclear, recent studies have stated that heredity is important in the development of ADHD. This is linked to a few comorbidities, including depression, criminal behavior, and anxiety. Although genetic factors influence ADHD symptoms, there are also non-genetic factors, one of which is oxidative stress (OS), which plays a role in the pathogenesis and symptoms of ADHD. This review aims to explore the role of OS in ADHD and its connection to antioxidant enzyme levels, as well as the gut-brain axis (GBA), focusing on diet and its influence on ADHD symptoms, particularly in adults with comorbid conditions. Methods: The literature search included the main available databases (e.g., Science Direct, PubMed, and Google Scholar). Articles in the English language were taken into consideration and our screening was conducted based on several words such as "ADHD", "oxidative stress", "diet", "gut-brain axis", and "gut microbiota." The review focused on studies examining the link between oxidative stress and ADHD, the role of the gut-brain axis, and the potential impact of dietary interventions. Results: Oxidative stress plays a critical role in the development and manifestation of ADHD symptoms. Studies have shown that individuals with ADHD exhibit reduced levels of key antioxidant enzymes, including glutathione peroxidase (GPx), catalase (CAT), and superoxide dismutase (SOD), as well as a diminished total antioxidant status (TOS) compared to healthy controls. Additionally, there is evidence of a close bidirectional interaction between the nervous system and gut microbiota, mediated by the gut-brain axis. This relationship suggests that dietary interventions targeting gut health may influence ADHD symptoms and related comorbidities. Conclusions: Oxidative stress and the gut-brain axis are key factors in the pathogenesis of ADHD, particularly in adults with comorbid conditions. A better understanding of these mechanisms could lead to more targeted treatments, including dietary interventions, to mitigate ADHD symptoms. Further research is required to explore the therapeutic potential of modulating oxidative stress and gut microbiota in the management of ADHD.
Collapse
Affiliation(s)
- Malina Visternicu
- Faculty of Biology, “Alexandru Ioan Cuza” University of Iași, Carol I Avenue, No. 20A, 700505 Iași, Romania;
- “Ioan Haulica” Institute, Apollonia University, Pacurari Street 11, 700511 Iași, Romania; (V.R.); (V.B.); (A.C.)
| | - Viorica Rarinca
- “Ioan Haulica” Institute, Apollonia University, Pacurari Street 11, 700511 Iași, Romania; (V.R.); (V.B.); (A.C.)
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iași, Carol I Avenue, No. 20A, 700505 Iași, Romania
- Doctoral School of Geosciences, Faculty of Geography and Geology, “Alexandru Ioan Cuza” University of Iași, Carol I Avenue, No. 20A, 700505 Iași, Romania
| | - Vasile Burlui
- “Ioan Haulica” Institute, Apollonia University, Pacurari Street 11, 700511 Iași, Romania; (V.R.); (V.B.); (A.C.)
| | - Gabriela Halitchi
- “Ioan Haulica” Institute, Apollonia University, Pacurari Street 11, 700511 Iași, Romania; (V.R.); (V.B.); (A.C.)
| | - Alin Ciobică
- “Ioan Haulica” Institute, Apollonia University, Pacurari Street 11, 700511 Iași, Romania; (V.R.); (V.B.); (A.C.)
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iași, Carol I Avenue, No. 20A, 700505 Iași, Romania
- CENEMED Platform for Interdisciplinary Research, “Grigore T. Popa” University of Medicine and Pharmacy of Iasi, University Street No. 16, 700115 Iași, Romania
- Academy of Romanian Scientists, No. 54, Independence Street, Sector 5, 050094 Bucharest, Romania
| | - Ana-Maria Singeap
- Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy of Iasi, University Street No. 16, 700115 Iași, Romania;
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700115 Iași, Romania
| | - Romeo Dobrin
- Institute of Psychiatry “Socola”, 36 Bucium Street, 700282 Iași, Romania;
- Department of Psychiatry, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy of Iasi, University Street No. 16, 700115 Iași, Romania
| | - Ioannis Mavroudis
- Department of Neurology, Leeds Teaching Hospitals, NHS Trust, Leeds LS2 9JT, UK;
- Faculty of Medicine, Leeds University, Leeds LS2 9JT, UK
| | - Anca Trifan
- Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy of Iasi, University Street No. 16, 700115 Iași, Romania;
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700115 Iași, Romania
| |
Collapse
|
4
|
Novo JP, Muga M, Lourenço T, Sanches ES, Leitão RA, Silva AP. Dichotomous effect of methylphenidate on microglia and astrocytes: Insights from in vitro and animal studies. Toxicol Lett 2023; 389:1-10. [PMID: 37844808 DOI: 10.1016/j.toxlet.2023.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/27/2023] [Accepted: 10/09/2023] [Indexed: 10/18/2023]
Abstract
Methylphenidate (MPH) has been used for decades to treat attention-deficit/hyperactivity disorder (ADHD) and narcolepsy. Moreover, several studies have shown that it is subject to misuse, particularly among college students and adolescents, for cognitive enhancement or as a recreational drug. This phenomenon causes concern, and it is critical to clarify better how MPH impacts brain cells. In fact, data has suggested that MPH could result in neuroinflammation and neurodegeneration across several brain regions; however, little is known about the effect of MPH on glial cells. To address this, we used microglia N9 cell line and primary cultures of cortical astrocytes that were exposed to MPH (0.01 - 2 mM), as well as Wistar Kyoto rats (WKY) chronically administered with MPH (1.5 mg/kg/day). Several parameters were analyzed, and we concluded that MPH has no significant direct effect on microglial cells, apart from cell migration impairment. On the contrary, MPH promotes astrogliosis, oxidative/nitrosative stress, and increases proinflammatory cytokine TNF levels by astrocytes, which was concordant with the results obtained in the hippocampus of WKY rats. Overall, the present results suggest that brain cells respond differently to MPH, with a more prominent direct effect on astrocytes when compared to microglia.
Collapse
Affiliation(s)
- João P Novo
- Univ Coimbra, Faculty of Medicine, Institute of Pharmacology and Experimental Therapeutics, and Coimbra Institute for Clinical and Biomedical Research (iCBR), Coimbra, Portugal; Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Portugal
| | - Mariana Muga
- Univ Coimbra, Faculty of Medicine, Institute of Pharmacology and Experimental Therapeutics, and Coimbra Institute for Clinical and Biomedical Research (iCBR), Coimbra, Portugal
| | - Teresa Lourenço
- Univ Coimbra, Faculty of Medicine, Institute of Pharmacology and Experimental Therapeutics, and Coimbra Institute for Clinical and Biomedical Research (iCBR), Coimbra, Portugal
| | - Eliane S Sanches
- Univ Coimbra, Faculty of Medicine, Institute of Pharmacology and Experimental Therapeutics, and Coimbra Institute for Clinical and Biomedical Research (iCBR), Coimbra, Portugal; Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Portugal
| | - Ricardo A Leitão
- Univ Coimbra, Faculty of Medicine, Institute of Pharmacology and Experimental Therapeutics, and Coimbra Institute for Clinical and Biomedical Research (iCBR), Coimbra, Portugal; Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Ana Paula Silva
- Univ Coimbra, Faculty of Medicine, Institute of Pharmacology and Experimental Therapeutics, and Coimbra Institute for Clinical and Biomedical Research (iCBR), Coimbra, Portugal; Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal.
| |
Collapse
|
5
|
Vázquez-González D, Corona JC. Pioglitazone enhances brain mitochondrial biogenesis and phase II detoxification capacity in neonatal rats with 6-OHDA-induced unilateral striatal lesions. Front Neurosci 2023; 17:1186520. [PMID: 37575308 PMCID: PMC10416244 DOI: 10.3389/fnins.2023.1186520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 07/17/2023] [Indexed: 08/15/2023] Open
Abstract
The psychostimulant methylphenidate (MPH) is the first-line pharmacological treatment for attention-deficit/hyperactivity disorder (ADHD), but has numerous adverse side effects. The PPARγ receptor agonist pioglitazone (PIO) is known to improve mitochondrial bioenergetics and antioxidant capacity, both of which may be deficient in ADHD, suggesting utility as an adjunct therapy. Here, we assessed the effects of PIO on ADHD-like symptoms, mitochondrial biogenesis and antioxidant pathways in multiple brain regions of neonate rats with unilateral striatal lesions induced by 6-hydroxydopamine (6-OHDA) as an experimental ADHD model. Unilateral striatal injection of 6-OHDA reduced ipsilateral dopaminergic innervation by 33% and increased locomotor activity. This locomotor hyperactivity was not altered by PIO treatment for 14 days. However, PIO increased the expression of proteins contributing to mitochondrial biogenesis in the striatum, hippocampus, cerebellum and prefrontal cortex of 6-OHDA-lesioned rats. In addition, PIO treatment enhanced the expression of the phase II transcription factor Nrf2 in the striatum, prefrontal cortex and cerebellum. In contrast, no change in the antioxidant enzyme catalase was observed in any of the brain regions analyzed. Thus, PIO may improve mitochondrial biogenesis and phase 2 detoxification in the ADHD brain. Further studies are required to determine if different dose regimens can exert more comprehensive therapeutic effects against ADHD neuropathology and behavior.
Collapse
Affiliation(s)
| | - Juan Carlos Corona
- Laboratory of Neurosciences, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| |
Collapse
|
6
|
Gutiérrez-Casares JR, Quintero J, Segú-Vergés C, Rodríguez Monterde P, Pozo-Rubio T, Coma M, Montoto C. In silico clinical trial evaluating lisdexamfetamine's and methylphenidate's mechanism of action computational models in an attention-deficit/hyperactivity disorder virtual patients' population. Front Psychiatry 2023; 14:939650. [PMID: 37333910 PMCID: PMC10273406 DOI: 10.3389/fpsyt.2023.939650] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 04/21/2023] [Indexed: 06/20/2023] Open
Abstract
Introduction Attention-deficit/hyperactivity disorder (ADHD) is an impairing psychiatric condition with the stimulants, lisdexamfetamine (LDX), and methylphenidate (MPH), as the first lines pharmacological treatment. Methods Herein, we applied a novel in silico method to evaluate virtual LDX (vLDX) and vMPH as treatments for ADHD applying quantitative systems pharmacology (QSP) models. The objectives were to evaluate the model's output, considering the model characteristics and the information used to build them, to compare both virtual drugs' efficacy mechanisms, and to assess how demographic (age, body mass index, and sex) and clinical characteristics may affect vLDX's and vMPH's relative efficacies. Results and Discussion We molecularly characterized the drugs and pathologies based on a bibliographic search, and generated virtual populations of adults and children-adolescents totaling 2,600 individuals. For each virtual patient and virtual drug, we created physiologically based pharmacokinetic and QSP models applying the systems biology-based Therapeutic Performance Mapping System technology. The resulting models' predicted protein activity indicated that both virtual drugs modulated ADHD through similar mechanisms, albeit with some differences. vMPH induced several general synaptic, neurotransmitter, and nerve impulse-related processes, whereas vLDX seemed to modulate neural processes more specific to ADHD, such as GABAergic inhibitory synapses and regulation of the reward system. While both drugs' models were linked to an effect over neuroinflammation and altered neural viability, vLDX had a significant impact on neurotransmitter imbalance and vMPH on circadian system deregulation. Among demographic characteristics, age and body mass index affected the efficacy of both virtual treatments, although the effect was more marked for vLDX. Regarding comorbidities, only depression negatively impacted both virtual drugs' efficacy mechanisms and, while that of vLDX were more affected by the co-treatment of tic disorders, the efficacy mechanisms of vMPH were disturbed by wide-spectrum psychiatric drugs. Our in silico results suggested that both drugs could have similar efficacy mechanisms as ADHD treatment in adult and pediatric populations and allowed raising hypotheses for their differential impact in specific patient groups, although these results require prospective validation for clinical translatability.
Collapse
Affiliation(s)
- José Ramón Gutiérrez-Casares
- Unidad Ambulatoria de Psiquiatría y Salud Mental de la Infancia, Niñez y Adolescencia, Hospital Perpetuo Socorro, Badajoz, Spain
| | - Javier Quintero
- Servicio de Psiquiatría, Hospital Universitario Infanta Leonor, Universidad Complutense, Madrid, Spain
| | - Cristina Segú-Vergés
- Anaxomics Biotech, Barcelona, Spain
- Structural Bioinformatics Group, Research Programme on Biomedical Informatics, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | | | | | | | - Carmen Montoto
- Medical Department, Takeda Farmacéutica España, Madrid, Spain
| |
Collapse
|
7
|
Carreón-Trujillo S, Vázquez-González D, Corona JC. Atomoxetine Decreases Mitochondrial Biogenesis, Fission and Fusion In Human Neuron-like Cells But Does Not Alter Antioxidant Defences. Cell Biochem Biophys 2023; 81:105-115. [PMID: 36346546 DOI: 10.1007/s12013-022-01116-x] [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: 11/29/2021] [Accepted: 10/30/2022] [Indexed: 11/10/2022]
Abstract
Atomoxetine (ATX) is a presynaptic norepinephrine transporter (NET) inhibitor widely prescribed for attention-deficit/hyperactivity disorder (ADHD) due to its low abuse potential and absence of psychostimulant effects. While NET inhibition is implicated in the clinical response, several additional pharmacoactivities may contribute to clinical efficacy or unwanted side effects. We recently reported that ATX can dose-dependently alter mitochondrial function and cellular redox status. Here, we assessed potential alterations in mitochondrial biogenesis, mitochondrial dynamics and cellular antioxidant capacity following high- and low-dose ATX treatment of differentiated human neuroblastoma cells. Human SH-SY5Y neuroblastoma cells were treated with ATX (1, 5, 10, 20 and 50 μM) for 7 days under differentiation culture conditions. Changes in the expression levels of protein markers for mitochondrial biogenesis, fusion and fission as well as of antioxidant proteins were analysed by Western blot. High-dose ATX (50 μM) reduced while low-dose ATX (10 μM) increased mitochondrial biogenesis as evidenced by parallel changes in SDHA, COX-I, PGC1α and TFAM expression. High-dose ATX also reduced mitochondrial fusion as evidenced by OPA1 and MFN2 downregulation, and mitochondrial fission as indicated by DRP1 and Fis1 downregulation. In contrast, ATX did not alter expression of the antioxidant enzymes SOD1 and catalase, the phase II transcription factor Nfr2, or the Nfr2-regulated antioxidant enzyme NQO1. Clinical responses and side effects of ATX may be mediated by dose-dependent modulation of mitochondrial biogenesis and dynamics as well as NET inhibition.
Collapse
Affiliation(s)
- Sonia Carreón-Trujillo
- Laboratory of Neurosciences, Hospital Infantil de México Federico Gómez, 06720, Mexico City, Mexico
| | - Daniela Vázquez-González
- Laboratory of Neurosciences, Hospital Infantil de México Federico Gómez, 06720, Mexico City, Mexico
| | - Juan Carlos Corona
- Laboratory of Neurosciences, Hospital Infantil de México Federico Gómez, 06720, Mexico City, Mexico.
| |
Collapse
|
8
|
Vázquez-González D, Carreón-Trujillo S, Alvarez-Arellano L, Abarca-Merlin DM, Domínguez-López P, Salazar-García M, Corona JC. A Potential Role for Neuroinflammation in ADHD. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1411:327-356. [PMID: 36949317 DOI: 10.1007/978-981-19-7376-5_15] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Attention deficit hyperactivity disorder (ADHD) is a neurobehavioural disorder in children and adolescents. Although increases in oxidative stress and disturbances of neurotransmitter system such as the dopaminergic and abnormalities in several brain regions have been demonstrated, the pathophysiology of ADHD is not fully understood. Nevertheless, ADHD involves several factors that have been associated with an increase in neuroinflammation. This chapter presents an overview of factors that may increase neuroinflammation and play a potential role in the development and pathophysiology of ADHD. The altered immune response, polymorphisms in inflammatory-related genes, ADHD comorbidity with autoimmune and inflammatory disorders and prenatal exposure to inflammation are associated with alterations in offspring brain development and are a risk factor; genetic and environmental risk factors that may increase the risk for ADHD and medications can increase neuroinflammation. Evidence of an association between these factors has been an invaluable tool for research on inflammation in ADHD. Therefore, evidence studies have made it possible to generate alternative therapeutic interventions using natural products as anti-inflammatories that could have great potential against neuroinflammation in ADHD.
Collapse
Affiliation(s)
| | - Sonia Carreón-Trujillo
- Laboratory of Neurosciences, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | | | | | - Pablo Domínguez-López
- Unidad de Investigación Médica en Medicina Reproductiva, Hospital Gineco-Obstetricia, IMSS, Mexico City, Mexico
| | - Marcela Salazar-García
- Laboratorio de Investigación en Biología del Desarrollo y Teratogénesis Experimental, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Juan Carlos Corona
- Laboratory of Neurosciences, Hospital Infantil de México Federico Gómez, Mexico City, Mexico.
| |
Collapse
|
9
|
Evidence of methylphenidate effect on mitochondria, redox homeostasis, and inflammatory aspects: Insights from animal studies. Prog Neuropsychopharmacol Biol Psychiatry 2022; 116:110518. [PMID: 35092763 DOI: 10.1016/j.pnpbp.2022.110518] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 01/19/2022] [Accepted: 01/21/2022] [Indexed: 11/22/2022]
Abstract
Methylphenidate (MPH) is a central nervous system (CNS) stimulant known for its effectiveness in the treatment of Attention Deficit Hyperactivity Disorder (ADHD), a neuropsychiatric condition that has a high incidence in childhood and affects behavior and cognition. However, the increase in its use among individuals who do not present all the diagnostic criteria for ADHD has become a serious public health problem since the neurological and psychiatric consequences of this unrestricted use are not widely known. In addition, since childhood is a critical period for the maturation of the CNS, the high prescription of MPH for preschool children also raises several concerns. This review brings new perspectives on how MPH (in different doses, routes of administration and ages) affects the CNS, focusing on animal studies that evaluated changes in mitochondrial (bioenergetics), redox balance and apoptosis, as well as inflammatory parameters. MPH alters brain energy homeostasis, increasing glucose consumption and impairing the activity of enzymes in the Krebs cycle and electron transport chain, as well as ATP levels and Na+,K+-ATPase activity. MPH induces oxidative stress, increasing the levels of reactive oxygen and nitrogen species and altering enzymatic and non-enzymatic antioxidant defenses, which, consequently, is related to damage to proteins, lipids, and DNA. Among the harmful effects of MPH, studies also demonstrate its ability to induce inflammation as well as alter the apoptosis pathway. It is important to highlight that age, treatment time, administration route, and dose are factors that can influence MPH effects. However, young animals seem to be more susceptible to damage caused by MPH. It is possible that changes in mitochondrial function and markers of status oxidative, apoptosis and inflammation may be exerting important mechanisms associated with MPH toxicity and, therefore, the unrestricted use of this drug can cause brain damage.
Collapse
|
10
|
Ceyhun HA, Gürbüzer N. New Hematological Parameters as Inflammatory Biomarkers: Systemic Immune Inflammation Index, Platerethritis, and Platelet Distribution Width in Patients with Adult Attention Deficit Hyperactivity Disorder. ADVANCES IN NEURODEVELOPMENTAL DISORDERS 2022; 6:211-223. [PMID: 35573104 PMCID: PMC9091147 DOI: 10.1007/s41252-022-00258-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 04/26/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVES The potential role of neuroinflammation in adult attention deficit hyperactivity disorder (ADHD) patients has been investigated with peripheral hemogram-related inflammatory markers. Systemic immune inflammation index (SII) is defined as a new index that has been developed for the balance of inflammatory and immune status. METHODS Our study was based on a prospective routine complete blood count(CBC) analysis of 74 Adult ADHD patients and 70 healthy participants. The DSM-5-Clinician version and Barratt impulsivity scale-11 were used to evaluate the participants. RESULTS There was no statistical difference in the comparison of the SII ratios, platelet distribution width (PDW), and plateretritis (PCT) (p>0.05 for each) in a group of a young adults with ADHD and in a comparison control group. These inflammatory indicators were found to be similar between patients newly diagnosed with ADHD (n=40) and patients using methylphenidate (n=34) (p>0.05 for each). The relationship between ADHD clinical symptoms and severity and inflammation was evaluated. A significant negative correlation was observed between attention deficit scores and PCT (r=-0.301, p=0.009). A positive significant correlation was found between hyperactivity scores and SII (r=0.247, p=0.034). A significant positive correlation was found between Barrat motor scores and PDW(r=241, p=0.038). In the regression analysis, the PCT variable changed the attention deficit variable (β=.33, t(70)= -2.703, p=.009, pr 2= .094) predicted negatively and significantly. CONCLUSIONS We demonstrated the association of SII, which is independently associated with adverse outcomes in many diseases, and the severity of hyperactivity symptoms in adult ADHD. The fact that PCT predicts attention deficit negatively and decisively shows the importance of inflammatory assessments specific to clinical presentations. The critical importance of platelets in inflammatory processes in ADHD has been demonstrated once again with inflammatory markers such as SII, PLT, and PDW, which can be accessed by an easily applicable complete blood count method.
Collapse
Affiliation(s)
- Hacer Akgül Ceyhun
- Department of Psychiatry, Ataturk University School of Medicine, Atatürk University, 25 240 Erzurum, Turkey
| | - Nilifer Gürbüzer
- Department of Psychiatry, Regional Training and Research Hospital, University of Health Sciences, Erzurum, Turkey
| |
Collapse
|
11
|
Mohammadzadeh Honarvar N, Samadi M, Seyedi Chimeh M, Gholami F, Bahrampour N, Jalali M, Effatpanah M, Yekaninejad MS, Abdolahi M, Chamari M. Effect of Vitamin D on Paraxonase-1, Total Antioxidant Capacity, and 8-Isoprostan in Children with Attention Deficit Hyperactivity Disorder. Int J Clin Pract 2022; 2022:4836731. [PMID: 35685610 PMCID: PMC9159115 DOI: 10.1155/2022/4836731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/19/2021] [Accepted: 12/25/2021] [Indexed: 11/17/2022] Open
Abstract
METHOD In this double-blind, randomized, placebo-controlled trial, 75 children (aged 6-12) diagnosed with ADHD were randomly assigned into two groups. The supplementation group received vitamin D3 (2000 IU), and the control group received a placebo for 3 months. Blood samples were collected at baseline and after intervention to analyze the 25(OH)D, paraxonase-1 activity (PON-1), Total Antioxidant Capacity (TAC), and 8-isoprostan levels. RESULTS A significant rise in circulating 25(OH)D was observed in the vitamin D group versus the placebo group at the end of the study. There was no reduction in 8-isoprostan levels in the vitamin D group compared to the placebo group. Serum paraxonase-1 and TAC concentration decreased in both groups, but these alterations were not statistically significant in the treatment group versus the placebo group at the end of the intervention. CONCLUSION Vitamin D supplementation for 3 months did not have beneficial effects on biomarkers of oxidative stress status. To confirm these findings, further studies on children are suggested.
Collapse
Affiliation(s)
- Niyaz Mohammadzadeh Honarvar
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Samadi
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Marzieh Seyedi Chimeh
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Gholami
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Niki Bahrampour
- Department of Nutrition, Science and Research Branch Islamic Azad University (SRBIAU), Tehran, Iran
| | - Mahmoud Jalali
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Effatpanah
- School of Medicine, Ziaeian Hospital, International Campus, Tehran University of Medical Sciences, Tehran, Iran
| | - Mir Saeid Yekaninejad
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Science, Tehran, Iran
| | - Mina Abdolahi
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Chamari
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Samadi M, Gholami F, Seyedi M, Jalali M, Effatpanah M, Yekaninejad MS, Abdolahi M, Chamari M, Mohammadzadeh Honarvar N. Effect of Vitamin D Supplementation on Inflammatory Biomarkers in School-Aged Children with Attention Deficit Hyperactivity Disorder. Int J Clin Pract 2022; 2022:1256408. [PMID: 36052304 PMCID: PMC9423974 DOI: 10.1155/2022/1256408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 03/31/2022] [Indexed: 11/29/2022] Open
Abstract
METHOD This randomized double-blind, placebo-controlled trial was conducted on 75 school-aged children with a diagnosis of ADHD based on DSM-V criteria. Children were randomly allocated to receive either vitamin D3 (2000 IU/day) or a placebo for 3 months. Serum IL-6, TNF-α, and 25(OH) D were assessed before and after the intervention to determine the effects of vitamin D on the highlighted parameters. RESULTS Serum levels of 25(OH) D increased significantly in the vitamin D group (P=0.01). However, no significant differences in serum IL-6 and TNF-α were found between both groups at the baseline and at the end of the intervention. CONCLUSION The findings revealed that vitamin D supplementation for 3 months is not efficacious in reducing inflammatory cytokines in children with ADHD. Further studies are required to confirm these results.
Collapse
Affiliation(s)
- Mahsa Samadi
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Gholami
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Marzieh Seyedi
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Jalali
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Effatpanah
- School of Medicine, Ziaeian Hospital, International Campus, Tehran University of Medical Sciences, Tehran, Iran
| | - Mir Saeid Yekaninejad
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Science, Tehran, Iran
| | - Mina Abdolahi
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Chamari
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Niyaz Mohammadzadeh Honarvar
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Evidence That Methylphenidate Treatment Evokes Anxiety-Like Behavior Through Glucose Hypometabolism and Disruption of the Orbitofrontal Cortex Metabolic Networks. Neurotox Res 2021; 39:1830-1845. [PMID: 34797528 DOI: 10.1007/s12640-021-00444-9] [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: 07/21/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 10/19/2022]
Abstract
Methylphenidate (MPH) has been widely misused by children and adolescents who do not meet all diagnostic criteria for attention-deficit/hyperactivity disorder without a consensus about the consequences. Here, we evaluate the effect of MPH treatment on glucose metabolism and metabolic network in the rat brain, as well as on performance in behavioral tests. Wistar male rats received intraperitoneal injections of MPH (2.0 mg/kg) or an equivalent volume of 0.9% saline solution (controls), once a day, from the 15th to the 44th postnatal day. Fluorodeoxyglucose-18 was used to investigate cerebral metabolism, and a cross-correlation matrix was used to examine the brain metabolic network in MPH-treated rats using micro-positron emission tomography imaging. Performance in the light-dark transition box, eating-related depression, and sucrose preference tests was also evaluated. While MPH provoked glucose hypermetabolism in the auditory, parietal, retrosplenial, somatosensory, and visual cortices, hypometabolism was identified in the left orbitofrontal cortex. MPH-treated rats show a brain metabolic network more efficient and connected, but careful analyses reveal that the MPH interrupts the communication of the orbitofrontal cortex with other brain areas. Anxiety-like behavior was also observed in MPH-treated rats. This study shows that glucose metabolism evaluated by micro-positron emission tomography in the brain can be affected by MPH in different ways according to the region of the brain studied. It may be related, at least in part, to a rewiring in the brain the metabolic network and behavioral changes observed, representing an important step in exploring the mechanisms and consequences of MPH treatment.
Collapse
|
14
|
Attention-deficit/hyperactivity disorder has a state-dependent association with asthma: The role of systemic inflammation in a population-based birth cohort followed from childhood to adulthood. Brain Behav Immun 2021; 97:239-249. [PMID: 34371132 DOI: 10.1016/j.bbi.2021.08.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 07/28/2021] [Accepted: 08/03/2021] [Indexed: 12/29/2022] Open
Abstract
There is a high comorbidity between attention-deficit/hyperactivity disorder (ADHD) and asthma, and inflammation has been proposed as a potential pathophysiological mechanism behind this association. Most studies conducted so far have used a cross-sectional design, and none has evaluated the prevalence of asthma symptoms in patients with ADHD followed from childhood to adulthood. We relied on data from the 1993 Pelotas birth cohort to evaluate the association between ADHD and asthma in patients with distinct patterns of incidence, persistence and remission, and to explore the potential role of inflammatory markers in the comorbidity. We analyzed data from 3281 individuals from the 1993 Pelotas birth cohort collected at birth (1993), 11 years (2004), 18 years (2011), and 22 years (2015). Subjects were first classified according to their ADHD and asthma status as early-onset (EO) persistent (positive screening for ADHD at 11 years and diagnosis of ADHD according to DSM-5, except criterion E, at either 18 or 22 years), EO-remittent (positive screening for ADHD at 11 years only), late-onset (diagnosis of ADHD according to DSM-5, except criterion E, at 18 or 22 years only), or healthy subjects (negative for both conditions in all evaluation). After controlling for confounders, significant associations were observed between EO-remittent ADHD and EO-remittent asthma (OR 1.68, 95% CI 1.11-2.55), EO-persistent ADHD and EO-persistent asthma (OR 4.33, 95% CI 1.65-11.34), and between late-onset ADHD and late-onset asthma (OR 1.86, 95% CI 1.28-2.70), suggesting a state-dependent association. Serum interleukin-6 (IL-6) and C-reactive protein (CRP) were measured at the 18- and 22-year evaluations and compared between subjects positive for ADHD, asthma, and subjects with both or none conditions, regardless of the previously defined trajectories. Subjects with comorbid ADHD and asthma presented higher levels of IL-6 at the 18- and 22-year evaluations when compared to subjects negative for both conditions. Our results demonstrate a state-dependent association between ADHD and asthma despite underlying trajectories. Higher levels of serum IL-6 in patients with both conditions suggest that a pro-inflammatory environment might have a role in the pathophysiological mechanisms underlying the comorbidity.
Collapse
|
15
|
Li P, Huang Y, Yang Y, Huang X. Methylphenidate exerts neuroprotective effects through the AMPK signaling pathway. Hum Exp Toxicol 2021; 40:1422-1433. [PMID: 33660552 DOI: 10.1177/0960327121996021] [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] [Indexed: 12/12/2022]
Abstract
PURPOSE Cerebral ischemia is the main cause of permanent adult disabilities worldwide. This study investigated the reparative effects and potential mechanisms of methylphenidate (MPH), a medication for the treatment of attention-deficit/hyperactivity disorder. METHODS In vitro oxygen-glucose deprivation/reperfusion (OGD/R) and in vivo cerebral ischemia-reperfusion models were established. Sprague-Dawley (SD) rats were randomly divided into four groups (n = 20): Sham, Model, and MPH (0.5 and 1 mg/kg). Rats in MPH groups were treated with 0.5 or 1 mg/kg MPH via intraperitoneal injection for 7 days. Rats in the Sham and Model groups were treated with PBS during the same period. Cell viability was measured using MTT assay. Apoptosis was detected by Annexin V/PI staining. Protein expression was detected by Western blot. The volume of cerebral infarction was detected by triphenyltetrazolium chloride (TTC) staining. The DNA damage in ischemic brain tissues was detected by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. RESULTS MPH treatment significantly reduced OGD/R-induced cell damage, shown by the increased cell viability and decreased apoptotic rate. p-AMPK and p-ACC protein expression increased in the OGD/R model after MPH treatment. The addition of AMPK inhibitor largely abolished the neuroprotective effects of MPH, evidenced by the reduced cell viability, increased apoptotic rate, and decreased protein expression of p-AMPK as well as p-ACC. Moreover, MPH treatment significantly alleviated the cerebral ischemia-reperfusion injury and decreased apoptosis in brain tissues, which may be associated with the AMPK/ACC pathway. CONCLUSIONS MPH exerted protective activities against oxidative stress in the OGD/R model and ameliorated brain damage of rats in the middle cerebral artery occlusion model, at least in part, through activating the AMPK pathway. These data demonstrated neuroprotective properties of MPH and highlighted it as a potential therapeutic agent against cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- P Li
- Department of Neurology, Hunan Provincial Brain Hospital, Changsha, China
| | - Y Huang
- Department of Neurology, Hunan Provincial Brain Hospital, Changsha, China
| | - Y Yang
- Department of Neurology, Hunan Provincial Brain Hospital, Changsha, China
| | - X Huang
- Department of Neurology, Hunan Provincial Brain Hospital, Changsha, China
| |
Collapse
|
16
|
Ho HY, Wong CK, Wu SY, Hsiao RC, Chen YL, Yen CF. Increased Alopecia Areata Risk in Children with Attention-Deficit/Hyperactivity Disorder and the Impact of Methylphenidate Use: A Nationwide Population-Based Cohort Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18031286. [PMID: 33535410 PMCID: PMC7908272 DOI: 10.3390/ijerph18031286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 01/27/2021] [Accepted: 01/29/2021] [Indexed: 11/16/2022]
Abstract
Alopecia areata (AA) is an autoimmune disease that causes sudden hair loss. Although few studies have reported the association between AA and attention-deficit/hyperactivity disorder (ADHD), the impact of methylphenidate (MPH) on AA has not been examined. This study examined whether AA risk is higher in children with ADHD than in those without ADHD as well as the impact of MPH use on AA risk in children with ADHD. From the Taiwan Maternal and Child Health Database, we enrolled all 1,750,456 newborns from 2004 to 2017 in Taiwan. Of them, 90,016 children received a diagnosis of ADHD whereas the remaining 1,660,440 did not. To compare AA risk in ADHD and the impact of MPH treatment on it, multiple Cox regression with adjustments for covariates (i.e., age, sex, and psychiatric comorbidities) was performed. The results indicated that 88 (0.098%) children with ADHD and 1191 (0.072%) children without ADHD had AA. Nevertheless, after adjustment for the covariates, AA risk was higher in children with ADHD than in those without ADHD (adjusted hazard ratio [aHR]: 1.30, 95% confidence interval [CI]: 1.04–1.64). Our data indicated a considerable reduction in AA risk (aHR: 0.64) among children with ADHD who received MPH than among those who did not receive MPH; however, this difference was nonsignificant, indicated by a wide 95% CI (0.32–1.25). In conclusion, ADHD and AA may share some underlying mechanisms.
Collapse
Affiliation(s)
- Hsing-Ying Ho
- Department of Healthcare Administration, College of Medical and Health Science, Asia University, Taichung 41354, Taiwan; (H.-Y.H.); (S.-Y.W.)
- Department of Psychology, College of Medical and Health Science, Asia University, Taichung 41354, Taiwan
| | - Chih-Kai Wong
- Department of Dermatology, MacKay Memorial Hospital, Taipei 10449, Taiwan;
| | - Szu-Yuan Wu
- Department of Healthcare Administration, College of Medical and Health Science, Asia University, Taichung 41354, Taiwan; (H.-Y.H.); (S.-Y.W.)
- Department of Food Nutrition and Health Biotechnology, College of Medical and Health Science, Asia University, Taichung 41354, Taiwan
- Big Data Center, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan 26546, Taiwan
- Division of Radiation Oncology, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan 26546, Taiwan
- Graduate Institute of Business Administration, Fu Jen Catholic University, Taipei 24205, Taiwan
| | - Ray C. Hsiao
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine and Children’s Hospital, Seattle, WA 98105, USA;
| | - Yi-Lung Chen
- Department of Healthcare Administration, College of Medical and Health Science, Asia University, Taichung 41354, Taiwan; (H.-Y.H.); (S.-Y.W.)
- Department of Psychology, College of Medical and Health Science, Asia University, Taichung 41354, Taiwan
- Correspondence: (Y.-L.C.); (C.-F.Y.); Tel.: +886-4-23323456 (ext. 20106) (Y.-L.C.); +886-7-3121101 (ext. 6822) (C.-F.Y.)
| | - Cheng-Fang Yen
- Department of Psychiatry, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Department of Psychiatry, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Correspondence: (Y.-L.C.); (C.-F.Y.); Tel.: +886-4-23323456 (ext. 20106) (Y.-L.C.); +886-7-3121101 (ext. 6822) (C.-F.Y.)
| |
Collapse
|
17
|
Alvarez-Arellano L, Salazar-García M, Corona JC. Neuroprotective Effects of Quercetin in Pediatric Neurological Diseases. Molecules 2020; 25:E5597. [PMID: 33260783 PMCID: PMC7731313 DOI: 10.3390/molecules25235597] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/20/2020] [Accepted: 11/26/2020] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress is a crucial event underlying several pediatric neurological diseases, such as the central nervous system (CNS) tumors, autism spectrum disorder (ASD) and attention-deficit/hyperactivity disorder (ADHD). Neuroprotective therapy with natural compounds used as antioxidants has the potential to delay, ameliorate or prevent several pediatric neurological diseases. The present review provides an overview of the most recent research outcomes following quercetin treatment for CNS tumors, ASD and ADHD as well as describes the potential in vitro and in vivo ameliorative effect on oxidative stress of bioactive natural compounds, which seems like a promising future therapy for these diseases. The neuroprotective effects of quercetin against oxidative stress can also be applied in the management of several neurodegenerative disorders with effects such as anti-cancer, anti-inflammatory, anti-viral, anti-obesity and anti-microbial. Therefore, quercetin appears to be a suitable adjuvant for therapy against pediatric neurological diseases.
Collapse
Affiliation(s)
| | - Marcela Salazar-García
- Laboratorio de Investigación en Biología del Desarrollo y Teratogénesis Experimental, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico;
| | - Juan Carlos Corona
- Laboratory of Neurosciences, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico
| |
Collapse
|
18
|
Corona JC. Role of Oxidative Stress and Neuroinflammation in Attention-Deficit/Hyperactivity Disorder. Antioxidants (Basel) 2020; 9:antiox9111039. [PMID: 33114154 PMCID: PMC7690797 DOI: 10.3390/antiox9111039] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/03/2020] [Accepted: 09/08/2020] [Indexed: 02/06/2023] Open
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a neurodevelopmental disorder of childhood. Although abnormalities in several brain regions and disturbances of the catecholaminergic pathway have been demonstrated, the pathophysiology of ADHD is not completely understood, but as a multifactorial disorder, has been associated with an increase in oxidative stress and neuroinflammation. This review presents an overview of factors that increase oxidative stress and neuroinflammation. The imbalance between oxidants and antioxidants and also the treatment with medications are two factors that can increase oxidative damage, whereas the comorbidity between ADHD and inflammatory disorders, altered immune response, genetic and environmental associations, and polymorphisms in inflammatory-related genes can increase neuroinflammation. Evidence of an association with these factors has become valuable for research on ADHD. Such evidence opens up new intervention routes for the use of natural products as antioxidants that could have potential as a treatment against oxidative stress and neuroinflammation in ADHD.
Collapse
Affiliation(s)
- Juan Carlos Corona
- Laboratory of Neurosciences, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico
| |
Collapse
|
19
|
Khalid A, Abbasi UA, Amber S, Sumera, Mirza FJ, Asif M, Javed A, Zahid S. Methylphenidate and Rosmarinus officinalis improves cognition and regulates inflammation and synaptic gene expression in AlCl 3-induced neurotoxicity mouse model. Mol Biol Rep 2020; 47:7861-7870. [PMID: 33011892 DOI: 10.1007/s11033-020-05864-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 09/04/2020] [Accepted: 09/25/2020] [Indexed: 12/27/2022]
Abstract
Methylphenidate (MPH), a psychotropic medication is commonly used for children with attention deficit hyperactivity disorder (ADHD). In this study we elucidated the neuroprotective and anti-inflammatory effects of MPH and Rosmarinus officinalis (rosemary) extract, an ancient aromatic herb with several applications in traditional medicine. Briefly, six groups of mice (n = 8 each group), were specified for the study and behavioral analysis was performed to analyze spatial memory followed by histological assessment and gene expression analysis of synaptic (Syn I, II and III) and inflammatory markers (IL-6, TNFα and GFAP) via qRT-PCR, in an AlCl3-induced mouse model for neurotoxicity. The behavioral analysis demonstrated significant cognitive decline, memory defects and altered gene expression in AlCl3-treated group. Rosemary extract significantly decreased the expression of inflammatory and synaptic markers to the similar levels as that of MPH. The present findings suggested the neuroprotective potential of Rosmarinus officinalis extract. However, further characterization of its anti-inflammatory and neuroprotective properties and MPH is required to strategize future treatments for several neurological and neurodegenerative disorders, including Alzheimer's disease.
Collapse
Affiliation(s)
- Anibah Khalid
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Umme Aimen Abbasi
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Sanila Amber
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Sumera
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Fatima Javed Mirza
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Muhammad Asif
- Department of Histopathology, Armed Forces Institute of Pathology, Rawalpindi, Pakistan
| | - Aneela Javed
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Saadia Zahid
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan.
| |
Collapse
|
20
|
Feizipour S, Sobhani S, Mehrafza S, Gholami M, Motaghinejad M, Motevalian M, Safari S, Davoudizadeh R. Selegiline acts as neuroprotective agent against methamphetamine-prompted mood and cognitive related behavior and neurotoxicity in rats: Involvement of CREB/BDNF and Akt/GSK3 signal pathways. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 23:606-615. [PMID: 32742598 PMCID: PMC7374985 DOI: 10.22038/ijbms.2020.38827.9221] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Objective(s): Present study investigated the neuroprotective effects of selegiline and the molecular mechanisms involved in methamphetamine-induced neurotoxicity. Materials and Methods: Male wistar rats were randomly divided into six groups (10 rats in each group). Group 1 and group 2 received normal saline and methamphetamine (10 mg/kg), respectively. Groups 3, 4, 5 and 6 were treated simultaneously with methamphetamine and selegiline. From day 22 to day 28, forced swim test, elevated plus maze, and open field test were conducted to assess mood (anxiety and depression) levels, and from day 17 to day 21, Morris Water Maze was conducted for cognition assessment. On day 29, hippocampus of the animals were isolated and evaluated by ELISA method for oxidative, antioxidant, and inflammatory factors and expression levels of active (total) and inactive (phosphorylated) forms of cyclic AMP response element binding protein (CREB), brain-derived neurotrophic factor (BDNF), Akt (Protein Kinase B) and glycogen synthase kinase 3 (GSK3) proteins. Results: Selegiline reduced behavioral impacts caused by methamphetamine in all doses. Methamphetamine administration may improve malondialdehyde, tumor necrosis factor-alpha, interleukin-1 beta and GSK3 (both forms). Moreover, methamphetamine reduced the activity of superoxide dismutase, glutathione peroxidase, glutathione reductase, amount of BDNF, CREB and Akt (both forms). Conclusion: Current research showed that selegiline can protect the brain from methamphetamine-prompted neurodegeneration, and this could be intervened by CREB -BDNF or Akt-GSK3 signaling pathways.
Collapse
Affiliation(s)
- Saba Feizipour
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Chemistry, Pharmaceutical Sciences Branch, Islamic Azad University (IUAPS), Tehran, Iran
| | - Sarvenaz Sobhani
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Shafagh Mehrafza
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Chemistry, Pharmaceutical Sciences Branch, Islamic Azad University (IUAPS), Tehran, Iran
| | - Mina Gholami
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Motaghinejad
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Medicine, Qom branch, Islamic Azad University, Iran
| | | | - Sepideh Safari
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
21
|
Antioxidants as a Potential Target against Inflammation and Oxidative Stress in Attention-Deficit/Hyperactivity Disorder. Antioxidants (Basel) 2020; 9:antiox9020176. [PMID: 32098021 PMCID: PMC7070894 DOI: 10.3390/antiox9020176] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/29/2020] [Accepted: 01/30/2020] [Indexed: 12/22/2022] Open
Abstract
Psychostimulants and non-psychostimulants are the medications prescribed for the treatment of attention-deficit/hyperactivity disorder (ADHD). However, several adverse results have been linked with an increased risk of substance use and side effects. The pathophysiology of ADHD is not completely known, although it has been associated with an increase in inflammation and oxidative stress. This review presents an overview of findings following antioxidant treatment for ADHD and describes the potential amelioration of inflammation and oxidative stress using antioxidants that might have a future as multi-target adjuvant therapy in ADHD. The use of antioxidants against inflammation and oxidative conditions is an emerging field in the management of several neurodegenerative and neuropsychiatric disorders. Thus, antioxidants could be promising as an adjuvant ADHD therapy.
Collapse
|
22
|
Jansen K, Hanusch B, Pross S, Hanff E, Drabert K, Bollenbach A, Dugave I, Carmann C, Siefen RG, Emons B, Juckel G, Legenbauer T, Tsikas D, Lücke T. Enhanced Nitric Oxide (NO) and Decreased ADMA Synthesis in Pediatric ADHD and Selective Potentiation of NO Synthesis by Methylphenidate. J Clin Med 2020; 9:jcm9010175. [PMID: 31936392 PMCID: PMC7019361 DOI: 10.3390/jcm9010175] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 12/27/2019] [Accepted: 01/04/2020] [Indexed: 12/12/2022] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) is a common pediatric psychiatric disorder, frequently treated with methylphenidate (MPH). Recently, MPH’s cardiovascular safety has been questioned by observational studies describing an increased cardiovascular risk in adults and blood pressure alterations in children. We considered members of the L-arginine (Arg)/nitric oxide (NO) pathway as possible early cardiovascular risk factors in pediatric ADHD children. They include the NO metabolites, nitrite and nitrate, the NO precursor Arg, and asymmetric dimethylarginine (ADMA), an endogenous NO synthase (NOS) inhibitor and a cardiovascular risk factor in adults. We conducted a prospective clinical trial with 42 ADHD children (aged 6–16 years) with (n = 19) and without (n = 23) MPH treatment. Age-matched children without ADHD (n = 43) served as controls. All plasma and urine metabolites were determined by gas chromatography-mass spectrometry. We observed higher plasma nitrite and lower plasma ADMA concentrations in the ADHD children. MPH-treated ADHD children had higher plasma nitrite concentrations than MPH-untreated ADHD children. As NOS activity is basally inhibited by ADMA, MPH treatment seems to have decreased the inhibitory potency of ADMA. Percentiles of systolic blood pressure were higher in MPH-treated ADHD children. The underlying mechanisms and their implications in the MPH therapy of pediatric ADHD with MPH remain to be elucidated in larger cohorts.
Collapse
Affiliation(s)
- Kathrin Jansen
- University Children’s Hospital, Ruhr University Bochum, 44791 Bochum, Germany
- Correspondence: ; Tel.: +49-234-5092615
| | - Beatrice Hanusch
- University Children’s Hospital, Ruhr University Bochum, 44791 Bochum, Germany
| | - Saskia Pross
- University Children’s Hospital, Ruhr University Bochum, 44791 Bochum, Germany
- Children’s Hospital, St., Clemens-Hospital Geldern, 47608 Geldern, Germany
| | - Erik Hanff
- Institute of Toxicology, Core Unit Proteomics, Hannover Medical School, 30625 Hannover, Germany
| | - Kathrin Drabert
- Institute of Toxicology, Core Unit Proteomics, Hannover Medical School, 30625 Hannover, Germany
| | - Alexander Bollenbach
- Institute of Toxicology, Core Unit Proteomics, Hannover Medical School, 30625 Hannover, Germany
| | - Irina Dugave
- University Children’s Hospital, Ruhr University Bochum, 44791 Bochum, Germany
- Department of Psychiatry, Alexius/Josef Hospital, 41464 Neuss, Germany
| | - Christina Carmann
- University Children’s Hospital, Ruhr University Bochum, 44791 Bochum, Germany
| | - Rainer Georg Siefen
- University Children’s Hospital, Ruhr University Bochum, 44791 Bochum, Germany
| | - Barbara Emons
- Department of Psychiatry, LWL Institute of Mental Health, LWL University Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
- Department of Psychiatry, LWL University Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Georg Juckel
- Department of Psychiatry, LWL Institute of Mental Health, LWL University Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
- Department of Psychiatry, LWL University Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Tanja Legenbauer
- LWL University Hospital Hamm for Child and Adolescent Psychiatry, Psychotherapy and Psychosomatic, Ruhr University Bochum, 59071 Hamm, Germany
| | - Dimitrios Tsikas
- Institute of Toxicology, Core Unit Proteomics, Hannover Medical School, 30625 Hannover, Germany
| | - Thomas Lücke
- University Children’s Hospital, Ruhr University Bochum, 44791 Bochum, Germany
| |
Collapse
|
23
|
Mohammadi N, Taheri P, Shahmoradi E, Motaghinejad M, Gholami M, Motevalian M. Preventive Effects of Duloxetine Against Methamphetamine Induced Neurodegeneration and Motor Activity Disorder in Rat: Possible Role of CREB/BDNF Signaling Pathway. Int J Prev Med 2019; 10:195. [PMID: 31772727 PMCID: PMC6868645 DOI: 10.4103/ijpvm.ijpvm_53_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 01/31/2019] [Indexed: 11/23/2022] Open
Abstract
Background: The neuroprotective effects of duloxetine and neurodegenerative effects of methamphetamine have been shown in previous studies, but their exact mechanism remain unclear. In the current study it involved molecular mechanisms of neuroprotective effects of duloxetine against methamphetamine induced neurodegeneration were clarified. Methods: About 40 adult male rats randomly were divided to 5 groups. Group 1 and 2, as control and methamphetamine treated, received normal saline and methamphetamine (10 mg/kg) respectively. Groups 3, 4 and 5 concurrently treated with methamphetamine and duloxetine at doses of 10, 20 and 30 mg/kg respectively. All treatments were undertaken for 21 days. On day 22 Open Field Test (OFT) were used to examine the level of motor activity disturbance and anxiety in animals. After that hippocampus was isolated from each rat and oxidative, antioxidant, inflammatory factors and also level or expression of total and phosphorylated forms of CREB and P-CREB and BDNF proteins were measured. Results: Duloxetine in all mentioned doses could inhibit the effects of methamphetamine induced motor activity disturbance in MWM. Chronic abuse of methamphetamine could increase malondialdehyde (MDA), tumor necrosis factor-Alpha (TNF-α) and interleukine-1beta (IL-1β) while caused decreases in superoxide dismutase (SOD), glutathione peroxidase (GPx) and glutathione reductase (GR) activities and decreased CREB (both forms) and BDNF proteins, while duloxetine could prevent these malicious effects of methamphetamine. Conclusions: We conclude that P-CREB/BDNF signaling pathways might have critical role in duloxetine neuroprotective effects against methamphetamine induced neurodegeneration.
Collapse
Affiliation(s)
- Niloofar Mohammadi
- School of Behavioral Sciences and Mental Health (Tehran Institute of Psychiatry), Tehran, Iran
| | - Parastoo Taheri
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Elaheh Shahmoradi
- Department of Chemistry, Faculty of Sciences, University of Zanjan 45371-38791, Zanjan, Iran
| | - Majid Motaghinejad
- Research Center for Addiction and Risky Behaviors (ReCARB), Iran Psychiatric Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mina Gholami
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Manijeh Motevalian
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Salehi P, Shahmirzadi ZY, Mirrezaei FS, Shirvani Boushehri F, Mayahi F, Songhori M, Abofazeli M, Motaghinejad M, Safari S. A hypothetic role of minocycline as a neuroprotective agent against methylphenidate-induced neuronal mitochondrial dysfunction and tau protein hyper-phosphorylation: Possible role of PI3/Akt/GSK3β signaling pathway. Med Hypotheses 2019; 128:6-10. [PMID: 31203911 DOI: 10.1016/j.mehy.2019.04.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 04/14/2019] [Accepted: 04/23/2019] [Indexed: 02/08/2023]
Abstract
The underlining mechanism in neural mitochondrial dysfunction and consequences neurotoxicity, and cognitive behavior after methylphenidate (MPH) prolonged uses is unclear and proposing of therapeutic approaches for treatment of these types of neurotoxicity is one of the main goals of scientist in this manner. MPH-induced mitochondrial dysfunction in neural cells caused induction of oxidative stress, apoptosis, inflammation and cognition impairment, which leads to neurotoxicity, was reported previously but role of key neural cells proteins and involved signaling pathway in this manner remained indeterminate. Tau protein aggregation is a biomarker for mitochondrial dysfunction, neurodegenerative event and cognition impairment. Tau aggregation occur by stimulation effects of Glycogen synthase kinase-3(GSK3β) and phosphatidylinositol 3-kinase (PI3K) which activates protein kinase B(Akt) and causes inhibition of phosphorylation(activation) of GSK3β, thus Akt activation can cause inhibition of tau aggregation (hyper-phosphorylation). Management of mentioned MPH-induced mitochondrial dysfunction and consequences of neurotoxicity, and cognitive behavior through a new generation neuroprotective combination, based on modulation of disturbed in Akt function and inhibition of GSK3β and tau hyper-phosphorylation can be a prefect therapeutic interventions. Therefore, finding, introduction and development of new neuroprotective properties and explanation of their effects with potential capacity for modulation of tau hyper-phosphorylation via PI3/Akt/GSK signaling pathway is necessitated. During recent years, using new neuroprotective compounds with therapeutic probability for treatment of psychostimulant-induced mitochondrial dysfunction, neurotoxicity and cognitive malicious effects have been amazingly increased. Many previous studies have reported the neuroprotective roles of minocycline (a broad-spectrum and long-acting antibiotic) in multiple neurodegenerative events and diseases in animal model. But the role of neuroprotective effects of this agent against MPH induced mitochondrial dysfunction, neurotoxicity and cognitive malicious and also role of tau hyper-phosphorylation by modulation of PI3/Akt/GSK signaling pathway in this manner remain unknown. Thus we suggested and theorized that by using minocycline in MPH addicted subject, it would provide neuroprotection against MPH-induced mitochondrial dysfunction, neurotoxicity and cognitive malicious. Also we hypothesized that minocycline, via modulation of PI3/Akt/GSK and inhibition of tau hyper-phosphorylation, can inhibit MPH-induced mitochondrial dysfunction, neurotoxicity and cognitive malicious. In this article, we tried to discuss our hypothesis regarding the possible role of minocycline, as a powerful neuroprotective agent, and also role of tau hyper-phosphorylation related to PI3/Akt/GSK signaling pathway in treatment of MPH-induced mitochondrial dysfunction, neurotoxicity and cognitive disturbance.
Collapse
Affiliation(s)
- Pegah Salehi
- Research Center for Addiction and Risky Behaviors (ReCARB), Iran Psychiatric Center, Iran University of Medical Sciences, Tehran, Iran
| | - Zhara Yaraei Shahmirzadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Chemistry, Pharmaceutical Sciences Branch, Islamic Azad University (IUAPS), Tehran, Iran
| | - Fatemeh Sadat Mirrezaei
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Chemistry, Pharmaceutical Sciences Branch, Islamic Azad University (IUAPS), Tehran, Iran
| | - Farima Shirvani Boushehri
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Chemistry, Pharmaceutical Sciences Branch, Islamic Azad University (IUAPS), Tehran, Iran
| | - Fatemeh Mayahi
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Chemistry, Pharmaceutical Sciences Branch, Islamic Azad University (IUAPS), Tehran, Iran
| | - Mojtaba Songhori
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Chemistry, Pharmaceutical Sciences Branch, Islamic Azad University (IUAPS), Tehran, Iran
| | - Maryam Abofazeli
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Chemistry, Pharmaceutical Sciences Branch, Islamic Azad University (IUAPS), 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, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Rahimi Borumand M, Motaghinejad M, Motevalian M, Gholami M. Duloxetine by Modulating the Akt/GSK3 Signaling Pathways Has Neuroprotective Effects against Methamphetamine-Induced Neurodegeneration and Cognition Impairment in Rats. IRANIAN JOURNAL OF MEDICAL SCIENCES 2019; 44:146-154. [PMID: 30936601 PMCID: PMC6423432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND The neuroprotective effects of duloxetine, as an antidepressant agent, and the neurodegenerative effects of methamphetamine have been shown in previous studies. Nonetheless, their exact neurochemical and behavioral effects are still unclear. In the current study, we sought to clarify the molecular mechanisms involved in the protective effects of duloxetine against methamphetamine-induced neurodegeneration. METHODS Forty adult male rats were divided randomly into 5 groups. Group 1 was the negative control and received normal saline, Group 2 was the positive control and received methamphetamine, and Groups 3, 4, and 5 were concurrently treated with methamphetamine (10 mg/kg) and duloxetine (5, 10, and 15 mg/kg, respectively). All the treatments were continued for 21 days. Between days 17 and 21, the Morris Water Maze (MWM) was used to assess learning and memory in the treated groups. On day 22, the hippocampus was isolated from each rat and oxidative, antioxidant, and inflammatory factors were measured. Additionally, the expression levels of the total and phosphorylated forms of the Akt and GSK3 proteins were evaluated via the ELISA method. RESULTS Duloxetine in all the administered doses ameliorated the effects of the methamphetamine-induced cognition impairment in the MWM. The chronic abuse of methamphetamine increased malondialdehyde, tumor necrosis factor-α, and interleukin-1β, while it decreased superoxide dismutase, glutathione peroxidase, and glutathione reductase activities. Duloxetine not only prevented these malicious effects of methamphetamine but also activated the expression of Akt (both forms) and inhibited the expression of GSK3 (both forms) in the methamphetamine-treated rats. CONCLUSION We conclude that the Akt/GSK3 signaling pathways might have a critical role in the protective effects of duloxetine against methamphetamine-induced neurodegeneration and cognition impairment.
Collapse
Affiliation(s)
- Mehrasa Rahimi Borumand
- Department of Pharmaceutical Biomaterials and Medical Biomaterial Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran;
| | - Majid Motaghinejad
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran;
| | - Manijeh Motevalian
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran;
| | - Mina Gholami
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
26
|
Carias E, Hamilton J, Robison LS, Delis F, Eiden R, Quattrin T, Hadjiargyrou M, Komatsu D, Thanos PK. Chronic oral methylphenidate treatment increases microglial activation in rats. J Neural Transm (Vienna) 2018; 125:1867-1875. [PMID: 30238340 DOI: 10.1007/s00702-018-1931-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 09/17/2018] [Indexed: 12/18/2022]
Abstract
Methylphenidate (MP) is a widely prescribed psychostimulant used to treat attention deficit hyperactivity disorder. Previously, we established a drinking paradigm to deliver MP to rats at doses that result in pharmacokinetic profiles similar to treated patients. In the present study, adolescent male rats were assigned to one of three groups: control (water), low-dose MP (LD; 4/10 mg/kg), and high dose MP (HD; 30/60 mg/kg). Following 3 months of treatment, half of the rats in each group were euthanized, and the remaining rats received only water throughout a 1-month-long abstinence phase. In vitro autoradiography using [3H] PK 11195 was performed to measure microglial activation. HD MP rats showed increased [3H] PK 11195 binding compared to control rats in several cerebral cortical areas: primary somatosensory cortex including jaw (68.6%), upper lip (80.1%), barrel field (88.9%), and trunk (78%) regions, forelimb sensorimotor area (87.3%), secondary somatosensory cortex (72.5%), motor cortices 1 (73.2%) and 2 (69.3%), insular cortex (59.9%); as well as subcortical regions including the thalamus (62.9%), globus pallidus (79.4%) and substantia nigra (22.7%). Additionally, HD MP rats showed greater binding compared to LD MP rats in the hippocampus (60.6%), thalamus (59.6%), substantia nigra (38.5%), and motor 2 cortex (55.3%). Following abstinence, HD MP rats showed no significant differences compared to water controls; however, LD MP rats showed increased binding in pre-limbic cortex (78.1%) and ventromedial caudate putamen (113.8%). These findings indicate that chronic MP results in widespread microglial activation immediately after treatment and following the cessation of treatment in some brain regions.
Collapse
Affiliation(s)
- Emily Carias
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 1021 Main St., Buffalo, NY, USA
| | - John Hamilton
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 1021 Main St., Buffalo, NY, USA
| | - Lisa S Robison
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Ave., Albany, NY, USA
| | - Foteini Delis
- Department of Pharmacology, Medical School, University of Ioannina, 45110, Ioannina, Greece
| | - Rina Eiden
- Department of Psychology, University at Buffalo, 1021 Main St., Buffalo, NY, USA
| | - Teresa Quattrin
- Women and Children's Hospital of Buffalo, Department of Pediatrics, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Michael Hadjiargyrou
- Department of Life Sciences, New York Institute of Technology, Northern Blvd., Old Westbury, NY, USA
| | - David Komatsu
- Department of Orthopedics, Stony Brook University, 100 Nicolls Rd., Stony Brook, NY, USA
| | - Panayotis K Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 1021 Main St., Buffalo, NY, USA.
| |
Collapse
|
27
|
Faraone SV. The pharmacology of amphetamine and methylphenidate: Relevance to the neurobiology of attention-deficit/hyperactivity disorder and other psychiatric comorbidities. Neurosci Biobehav Rev 2018; 87:255-270. [PMID: 29428394 DOI: 10.1016/j.neubiorev.2018.02.001] [Citation(s) in RCA: 366] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/25/2018] [Accepted: 02/05/2018] [Indexed: 12/20/2022]
Abstract
Psychostimulants, including amphetamines and methylphenidate, are first-line pharmacotherapies for individuals with attention-deficit/hyperactivity disorder (ADHD). This review aims to educate physicians regarding differences in pharmacology and mechanisms of action between amphetamine and methylphenidate, thus enhancing physician understanding of psychostimulants and their use in managing individuals with ADHD who may have comorbid psychiatric conditions. A systematic literature review of PubMed was conducted in April 2017, focusing on cellular- and brain system-level effects of amphetamine and methylphenidate. The primary pharmacologic effect of both amphetamine and methylphenidate is to increase central dopamine and norepinephrine activity, which impacts executive and attentional function. Amphetamine actions include dopamine and norepinephrine transporter inhibition, vesicular monoamine transporter 2 (VMAT-2) inhibition, and monoamine oxidase activity inhibition. Methylphenidate actions include dopamine and norepinephrine transporter inhibition, agonist activity at the serotonin type 1A receptor, and redistribution of the VMAT-2. There is also evidence for interactions with glutamate and opioid systems. Clinical implications of these actions in individuals with ADHD with comorbid depression, anxiety, substance use disorder, and sleep disturbances are discussed.
Collapse
Affiliation(s)
- Stephen V Faraone
- Departments of Psychiatry and of Neuroscience and Physiology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States; K.G. Jebsen Centre for Research on Neuropsychiatric Disorders, University of Bergen, Bergen, Norway.
| |
Collapse
|
28
|
Topiramate via NMDA, AMPA/kainate, GABA A and Alpha2 receptors and by modulation of CREB/BDNF and Akt/GSK3 signaling pathway exerts neuroprotective effects against methylphenidate-induced neurotoxicity in rats. J Neural Transm (Vienna) 2017; 124:1369-1387. [PMID: 28795276 DOI: 10.1007/s00702-017-1771-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 07/23/2017] [Indexed: 12/18/2022]
Abstract
Chronic abuse of methylphenidate (MPH) often causes neuronal cell death. Topiramate (TPM) carries neuroprotective effects, but its exact mechanism of action remains unclear. In the present study, the role of various doses of TPM and its possible mechanisms, receptors and signaling pathways involved against MPH-induced hippocampal neurodegeneration were evaluated in vivo. Thus, domoic acid (DOM) was used as AMPA/kainate receptor agonist, bicuculline (BIC) as GABAA receptor antagonist, ketamine (KET) as NMDA receptor antagonist, yohimbine (YOH) as α2 adrenergic receptor antagonist and haloperidol (HAL) was used as dopamine D2 receptor antagonist. Open field test (OFT) was used to investigate the disturbances in motor activity. Hippocampal neurodegenerative parameters were evaluated. Protein expressions of CREB/BDNF and Akt/GSK3 signaling pathways were also evaluated. Cresyl violet staining was performed to show and confirm the changes in the shape of the cells. TPM (70 and 100 mg/kg) reduced MPH-induced rise in lipid peroxidation, oxidized form of glutathione (GSSG), IL-1β and TNF-α levels, Bax expression and motor activity disturbances. In addition, TPM treatment increased Bcl-2 expression, the level of reduced form of glutathione (GSH) and the levels and activities of superoxide dismutase, glutathione peroxidase and glutathione reductase enzymes. TPM also inhibited MPH-induced hippocampal degeneration. Pretreatment of animals with DOM, BIC, KET and YOH inhibited TPM-induced neuroprotection and increased oxidative stress, neuroinflammation, neuroapoptosis and neurodegeneration while reducing CREB, BDNF and Akt protein expressions. Also pretreatment with DOM, BIC, KET and YOH inhibited TPM-induced decreases in GSK3. It can be concluded that the mentioned receptors by modulation of CREB/BDNF and Akt/GSK3 pathways, are involved in neuroprotection of TPM against MPH-induced neurodegeneration.
Collapse
|
29
|
Motaghinejad M, Motevalian M, Babalouei F, Abdollahi M, Heidari M, Madjd Z. Possible involvement of CREB/BDNF signaling pathway in neuroprotective effects of topiramate against methylphenidate induced apoptosis, oxidative stress and inflammation in isolated hippocampus of rats: Molecular, biochemical and histological evidences. Brain Res Bull 2017; 132:82-98. [PMID: 28552672 DOI: 10.1016/j.brainresbull.2017.05.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 05/13/2017] [Accepted: 05/19/2017] [Indexed: 12/29/2022]
Abstract
Chronic abuse of methylphenidate (MPH) can cause serious neurotoxicity. The neuroprotective effects of topiramate (TPM) were approved, but its putative mechanism remains unclear. In current study the role of CREB/BDNF signaling pathway in TPM protection against methylphenidate-induced neurotoxicity in rat hippocampus was evaluated. 60 adult male rats were divided randomly into six groups. Groups received MPH (10mg/kg) only and concurrently with TPM (50mg/kg and 100mg/kg) and TPM (50 and 100mg/kg) only for 14 days. Open field test (OFT) was used to investigate motor activity. Some biomarkers of apoptotic, anti-apoptotic, oxidative, antioxidant and inflammatory factors were also measured in hippocampus. Expression of total (inactive) and phosphorylated (active) CREB and BDNF were also measured in gene and protein levels in dentate gyrus (DG) and CA1 areas of hippocampus. MPH caused significant decreases in motor activity in OFT while TPM (50 and 100mg/kg) inhibited MPH-induced decreases in motor activity. On the other hand, MPH caused remarkable increases in Bax protein level, lipid peroxidation, catalase activity, IL-1β and TNF-α levels in hippocampal tissue. MPH also caused significant decreases of superoxide dismutase, activity and also decreased CREB, in both forms, BDNF and Bcl-2 protein levels. TPM, by the mentioned doses, attenuated these effects and increased superoxide dismutase, glutathione peroxidase and glutathione reductase activities and also increased CREB, in both forms, BDNF and Bcl-2 protein levels and inhibited MPH induced increase in Bax protein level, lipid peroxidation, catalase activity, IL-1β and TNF-α levels. TPM also inhibited MPH induced decreases in cell number and changes in cell shapes in DG and CA1 areas. TPM can probably act as a neuroprotective agent against MPH induced neurotoxicity and this might have been mediated by CREB/BDNF signaling pathway.
Collapse
Affiliation(s)
- Majid Motaghinejad
- Razi Drug Research Center & Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Manijeh Motevalian
- Razi Drug Research Center & Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Babalouei
- Deparemten of Chemistry, Faculty of Science, Islamic Azad University, Share-Qods Brach, Tehran, Iran
| | - Mohammad Abdollahi
- Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mansour Heidari
- Department of Medical Genetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center and Department of Pathology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
30
|
Motaghinejad M, Motevalian M, Abdollahi M, Heidari M, Madjd Z. Topiramate Confers Neuroprotection Against Methylphenidate-Induced Neurodegeneration in Dentate Gyrus and CA1 Regions of Hippocampus via CREB/BDNF Pathway in Rats. Neurotox Res 2017; 31:373-399. [PMID: 28078543 DOI: 10.1007/s12640-016-9695-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 12/21/2016] [Accepted: 12/26/2016] [Indexed: 12/18/2022]
Abstract
Methylphenidate (MPH) abuse can cause serious neurological damages. The neuroprotective effects of topiramate (TPM) have been reported already, but its mechanism of action still remains unclear. The current study evaluates in vivo role of CREB/BDNF in TPM protection of the rat hippocampal cells from methylphenidate-induced apoptosis, oxidative stress, and inflammation. A total of 60 adult male rats were divided into six groups. Groups 1 and 2 received normal saline (0.7 ml/rat) and MPH (10 mg/kg) respectively for 14 days. Groups 3 and 4 were concurrently treated with MPH (10 mg/kg) and TPM 50 and 100 mg/kg respectively for 14 days. Groups 5 and 6 were treated with 50 and 100 mg/kg TPM only respectively. After drug administration, open field test (OFT) was used to investigate motor activity. The hippocampus was then isolated and the apoptotic, antiapoptotic, oxidative, antioxidant, and inflammatory factors were measured. Expression of the total and phosphorylated CREB and BDNF in gene and protein levels, and gene expression of Ak1, CaMK4, MAPK3, PKA, and c-Fos levels were also measured. MPH significantly decreased motor activity in OFT. TPM (50 and 100 mg/kg) decreased MPH-induced motor activity disturbance. Additionally, MPH significantly increased Bax protein level, CaMK4 gene expression, lipid peroxidation, catalase activity, mitochondrial GSH, IL-1β, and TNF-α levels in isolated hippocampal cells. Also CREB, in total and phosphorylated forms, BDNF and Bcl-2 protein levels, Ak1, MAPK3, PKA and c-Fos gene expression, superoxide dismutase, glutathione peroxidase, and glutathione reductase activities decreased significantly by MPH. TPM (50 and 100 mg/kg), both in the presence and absence of MPH, attenuated the effects of MPH. Immunohistochemistry data showed that TPM increased localization of the total and phosphorylated forms of CREB in dentate gyrus (DG) and CA1 areas of the hippocampus. It seems that TPM can be used as a neuroprotective agent against apoptosis, oxidative stress, and neuroinflammation induced by frequent use of MPH. This might be probably mediated by the CREB/BDNF and their upstream signaling pathways.
Collapse
Affiliation(s)
- Majid Motaghinejad
- Razi Drug Research Center & Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Hemmat high way, Beside Milad Tower, Tehran, 14496-14525, Iran
| | - Manijeh Motevalian
- Razi Drug Research Center & Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Hemmat high way, Beside Milad Tower, Tehran, 14496-14525, Iran.
| | - Mohammad Abdollahi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mansour Heidari
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center and Department of pathology, Faculty of medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|