1
|
Roustaee S, Sani M, Mehranpour M, Raee P, Moghaddam MH, Bahar R, Nourirad SN, Golzarian MJ, Beirami A, Jafary H, Aalipour MA, Taghizadeh M, Abdollahifar MA, Vakili K, Fathi M, Heidari MH, Abbaszadeh HA, Aliaghaei A, Nazarian H. Chronic Administration of Lisdexamfetamine Induces Apoptosis and Inflammation and Reduces Sperm Quality in Adult Male Rats. Reprod Sci 2024; 31:1278-1289. [PMID: 38228974 DOI: 10.1007/s43032-023-01449-9] [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: 09/11/2023] [Accepted: 12/28/2023] [Indexed: 01/18/2024]
Abstract
Concerns have been raised about potentially irreversible brain damage and damage to the neuroendocrine system during development when treating attention-deficit/hyperactivity disorder with lisdexamfetamine (LDX), a norepinephrine dopamine reuptake inhibitor. This study aims to elucidate the potential adverse effects of LDX on the male reproductive system due to its widespread use and potential for abuse. In this study, adult male rats were randomized into control and LDX groups. Thirty milligrams per kilogram LDX was administered orally for 3 weeks. After isolation of epididymal spermatozoa, the rats were euthanized and testicular tissues were collected for stereological and molecular analyses. The LDX group showed a decrease in sperm motility and an increase in DNA fragmentation compared to the control group. There was also a dramatic decrease in testosterone in the LDX group. Testicular expression of caspase-3 and TNF-α was significantly increased in the LDX group. According to our findings, prolonged use of LDX leads to reduced sperm quality. It also induces apoptosis, inflammatory response, and pathological changes in the testicular tissue. What we have observed in this study is noteworthy but requires further investigation, particularly in people who use LDX over a longer period of time.
Collapse
Affiliation(s)
- Susan Roustaee
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mojtaba Sani
- Department of Educational Neuroscience, Aras International Campus, University of Tabriz, Tabriz, Iran
| | - Maryam Mehranpour
- Department of Genetics, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Pourya Raee
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Meysam Hassani Moghaddam
- Department of Anatomical Sciences, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Reza Bahar
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyedeh Naghmeh Nourirad
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad-Jasim Golzarian
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirreza Beirami
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hosein Jafary
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Aalipour
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taghizadeh
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad-Amin Abdollahifar
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kimia Vakili
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mobina Fathi
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossain Heidari
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hojjat-Allah Abbaszadeh
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Aliaghaei
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hamid Nazarian
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Schifano N, Chiappini S, Mosca A, Miuli A, Santovito MC, Pettorruso M, Capogrosso P, Dehò F, Martinotti G, Schifano F. Recreational Drug Misuse and Its Potential Contribution to Male Fertility Levels' Decline: A Narrative Review. Brain Sci 2022; 12:1582. [PMID: 36421906 PMCID: PMC9688450 DOI: 10.3390/brainsci12111582] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 10/14/2023] Open
Abstract
Recreational drug intake may be associated with a range of medical untoward consequences, including male infertility. However, as the related evidence is still limited, the main outcome of this review is to provide a better understanding of the existence of any association between recreational drug misuse and male fertility levels' decline. Whilst searching the MEDLINE/PubMed, a comprehensive overview of the literature regarding male infertility and substances of abuse (e.g., phytocannabinoids, opiates/opioids, stimulants, 'herbal highs', psychedelics, and 'novel psychoactive substances) was here undertaken. Due to the paucity of robust, high-quality, empirical, human studies, a narrative strategy was here preferred over a systematic approach. Relevant data are qualitatively analyzed and presented in a table. Although most available evidence is in support of a detrimental role of cannabis on human spermatogenesis, a few remaining studies failed to document any effect of this drug on seminal quality, and it is not clear to which extent this drug impacts fertility rates/time to pregnancy. The current understanding of the impact of opiate-, cocaine- and amphetamine/stimulant-misuse on human reproduction is widely unknown, and most studies dealing with this matter represent only an extrapolation of data derived from specific clinical circumstances. Although the message of 'no smoking, no alcohol and no street drugs' should always be offered as good health advice to all patients seeking medical help for fertility issues, robust scientific clinical evidence in support of a direct detrimental impact of recreational drugs on spermatogenesis is scant to date.
Collapse
Affiliation(s)
- Nicolò Schifano
- Department of Urology, ASST Sette Laghi–Circolo e Fondazione Macchi Hospital, 21100 Varese, Italy
| | - Stefania Chiappini
- Psychopharmacology, Drug Misuse and Novel Psychoactive Substances Research Unit, School of Life and Medical Sciences, University of Hertfordshire, Hertfordshire AL10 9EU, UK
- Department of Neuroscience, Imaging and Clinical Sciences, “G. D’Annunzio” University, 66100 Chieti, Italy
| | - Alessio Mosca
- Department of Neuroscience, Imaging and Clinical Sciences, “G. D’Annunzio” University, 66100 Chieti, Italy
| | - Andrea Miuli
- Department of Neuroscience, Imaging and Clinical Sciences, “G. D’Annunzio” University, 66100 Chieti, Italy
| | - Maria Chiara Santovito
- Department of Neuroscience, Imaging and Clinical Sciences, “G. D’Annunzio” University, 66100 Chieti, Italy
| | - Mauro Pettorruso
- Department of Neuroscience, Imaging and Clinical Sciences, “G. D’Annunzio” University, 66100 Chieti, Italy
| | - Paolo Capogrosso
- Department of Urology, ASST Sette Laghi–Circolo e Fondazione Macchi Hospital, 21100 Varese, Italy
| | - Federico Dehò
- Department of Urology, ASST Sette Laghi–Circolo e Fondazione Macchi Hospital, 21100 Varese, Italy
| | - Giovanni Martinotti
- Department of Neuroscience, Imaging and Clinical Sciences, “G. D’Annunzio” University, 66100 Chieti, Italy
| | - Fabrizio Schifano
- Psychopharmacology, Drug Misuse and Novel Psychoactive Substances Research Unit, School of Life and Medical Sciences, University of Hertfordshire, Hertfordshire AL10 9EU, UK
| |
Collapse
|
3
|
Does methylphenidate use affect sperm parameters in patients undergoing infertility investigation? A retrospective analysis of 9769 semen samples. Arch Gynecol Obstet 2021; 304:539-546. [PMID: 33433701 DOI: 10.1007/s00404-020-05938-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 12/11/2020] [Indexed: 10/22/2022]
Abstract
OBJECTIVES Methylphenidate (MPH) is the most widely prescribed therapy for attention deficit hyperactivity disorder. Animal studies have shown a potential adverse effect of MPH exposure on male fertility. We examined the impact of MPH on human male sperm parameters. DESIGN Sperm parameters of 9769 samples from patients 18 years of age or older, collected as part of the basic evaluation of couples referred to the Infertility Clinic were analyzed retrospectively. We divided the study population into three groups according to MPH purchasing information: MPH purchased ≤ 90 days prior to sperm analysis-current users (n = 83), MPH purchased > 90 days prior to sperm analysis-past users (n = 293), and MPH-naïve patients (n = 9393). METHODS All sperm samples were analyzed by the same laboratory technician team for the following routine parameters: semen volume, sperm concentration, percentage of motile sperm, and percentage of normal morphology according to World Health Organization. The analysis of the samples was completed by evaluation of total sperm count, total sperm motility, and percentage of fast and slow motile cells. Sperm morphology was evaluated by a laboratory technician using methodological examination according to the strict Kruger-Tygerberg criteria. RESULTS Methylphenidate exposure did not affect sperm morphology but was associated with increased sperm concentration as well as increased total sperm count and total sperm motility among current and past users compared with MPH-naïve patients. In particular, progressive motility and total motile sperm count were significantly increased following MPH use. A multivariate analysis adjusting for age and current smoking was conducted, further supporting a positive correlation between current MPH use and increased values of total sperm count and total sperm motility. LIMITATIONS Our study has several inherent weaknesses, foremost of which is its retrospective nature. Another notable weakness is that medication purchasing data may not accurately reflect MPH exposure in the study population. Patients may be purchasing MPH and not taking it as prescribed. CONCLUSIONS In the present study, we could not demonstrate a negative impact of methylphenidate treatment on sperm parameters in adults with ADHD. Hence, we may assume that methylphenidate does not negatively affect male fertility.
Collapse
|
4
|
Raoofi A, Delbari A, Mahdian D, Mojadadi MS, Akhlaghi M, Dadashizadeh G, Ebrahimi V, Amini A, Golmohammadi R, Javadinia SS, Khaneghah AM. Effects of curcumin nanoparticle on the histological changes and apoptotic factors expression in testis tissue after methylphenidate administration in rats. Acta Histochem 2021; 123:151656. [PMID: 33249311 DOI: 10.1016/j.acthis.2020.151656] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 11/08/2020] [Accepted: 11/13/2020] [Indexed: 12/27/2022]
Abstract
The present article sought to evaluate the impact of curcumin-loaded superparamagnetic iron oxide (Fe3O4) nanoparticles (NPs) on the histological variables and apoptotic agents in adult male rats after 3-weeks of methylphenidate (MPH) oral administration (20 mg/kg) versus vehicle therapy on the testis. Twenty-four male rats have been categorized randomly into four groups, in which Group 1 has been chosen as the controls, and Group 2 has been a vehicle and taken the sesame oil as curcumin carrier. Moreover, Group 3 has been taken MPH (20 mg/kg by gavage for 21 consecutive days). Group 4 received MPH plus Curcumin nanoparticles (5.4 mg/100 g) for twenty-one consecutive days. Then, testis histology, apoptosis as well as stereology have been examined. According to the examinations, curcumin nanoparticles are significantly capable of improving the sperms and stereological variables; for example, round spermatid and Leydig cells by enhancing the level of the serum testosterone in comparison with the MPH and vehicle groups. Besides, it was found that the gene expression in inflammation pathways and apoptosis genes largely diminished in the treatment group by curcumin nanoparticles in comparison with the MPH and vehicle groups, also we observed considerable differences for the weight of testes between the examined groups. Therefore, Curcumin effectively inhibited the testis damages and MPH-induced apoptosis, indicating possible protecting features of the Curcumin nanoparticles in opposition to MPH.
Collapse
Affiliation(s)
- Amir Raoofi
- Leishmaniasis Research Center, Department of Anatomy, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Ali Delbari
- Leishmaniasis Research Center, Department of Anatomy, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Davood Mahdian
- Cellular and Molecular Research Center, Department of Pharmacology, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Mohammad-Shafi Mojadadi
- Leishmaniasis Research Center, Department of Immunology, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Maedeh Akhlaghi
- Leishmaniasis Research Center, Department of Anatomy, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Ghazaleh Dadashizadeh
- Cellular and Molecular Research Center, Department of Anatomy, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Vahid Ebrahimi
- Department of Anatomy, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abdollah Amini
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rahim Golmohammadi
- Cellular and Molecular Research Center, Department of Anatomy, Sabzevar University of Medical Sciences, Sabzevar, Iran.
| | - Sara Sadat Javadinia
- Cellular and Molecular Research Center, Department of Anatomy, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Amin Mousavi Khaneghah
- Department of Food Science, Faculty of Food Engineering, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
| |
Collapse
|
5
|
Effects of Chronic Use of Methylphenidate on Spermatogenesis and Sexual Hormones in Adult Male Rats. Nephrourol Mon 2020. [DOI: 10.5812/numonthly.101792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: The growing prevalence of Attention Deficit Hyperactivity Disorder (ADHD) and the non-medical use of Methylphenidate (MPH) among the youth have lead male infertility to be a major health problem. Objectives: The present study was conducted to investigate the impacts of MPH administration on different aspects of productivity, including total body weight, testis weight, spermatogenesis, sperm motility, histopathology changes, and sex hormone serum levels in male rats. Methods: This study was performed with 54 eight-week-old male rats divided into one control and two experimental groups. The experimental groups were gavaged with 2 and 10 mg/kg methylphenidate daily while the control group was gavaged with normal saline (at the same dosage). After 60 days, rats were subjected to blood sampling and bilateral orchidoepididymectomy under anesthesia. Spermogram, histological, and hormonal evaluations were performed on the samples. Testes weight and total body weight were also recorded. Results: The results revealed significant differences between the MPH and experimental groups in terms of hormonal, spermographic, and histopathologic features, as well as weight. Luteinizing hormone and testosterone levels, sperm count and motility, Leydig cell hyperplasia, spermatogenesis, congestion and necrosis levels, total body weight, and testis weight were significantly different between the experimental and control groups. However, no difference was observed between the experimental and control groups concerning follicle-stimulating hormone, maturation arrest, and edema levels. Conclusions: Based on the findings, MPH exposure exerts a significant effect on the testis and total body weight, as well as hormonal, spermatographic, and histopathologic characteristics. Accordingly, the present study provided an insight into the negative impression of MPH on sexual parameters.
Collapse
|
6
|
Caballero-Puntiverio M, Lerdrup LS, Arvastson L, Aznar S, Andreasen JT. ADHD medication and the inverted U-shaped curve: A pharmacological study in female mice performing the rodent Continuous Performance Test (rCPT). Prog Neuropsychopharmacol Biol Psychiatry 2020; 99:109823. [PMID: 31765714 DOI: 10.1016/j.pnpbp.2019.109823] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 11/13/2019] [Accepted: 11/14/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND The rodent Continuous Performance Test (rCPT) is an analogue of human CPTs where mice have to discriminate between target and non-target stimuli. The rCPT offers a readout of attentional performance and impulsive behaviour. This study aimed to determine if female C57BL/6 J mice could be trained in the rCPT since previously published rCPT studies have only used male mice and to study whether the effects of methylphenidate (MPH), atomoxetine (ATX), and dexamphetamine (AMPH) on attention and impulsivity depend on baseline (reference) levels of performance. METHODS 48 female mice underwent rCPT training. Effects of MPH (1, 2, and 3 mg/kg), ATX (1, 3, and 5 mg/kg) and AMPH (0.3, 0.6, and 1 mg/kg) were assessed in a variable stimulus duration probe. Drugs were administered intraperitoneally and sequentially tested following a Latin-square design. Data were analysed using a repeated measurements mixed effect model and reference-dependent effects were studied. RESULTS ATX and AMPH improved performance as seen by increases in discriminability. These improvements were a result of a decreased false-alarm rate. AMPH showed a reference-dependent effect, improving the task performance of low-performing mice and decreasing the performance of high-performing mice. MPH also showed this reference-dependent effects, albeit to a lesser extent. ATX and AMPH decreased premature responses and increased response criterion, but no reference-dependent effects were observed for these parameters. CONCLUSION This study presents a novel method to analyse baseline-dependent effects. It shows that the rCPT can be successfully used in pharmacological studies in female mice and demonstrates that the effect of ADHD medication is in line with the inverted U-shape theory of performance-arousal relationship.
Collapse
Affiliation(s)
- M Caballero-Puntiverio
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, Copenhagen 2100, Denmark; Symptoms Biology, H. Lundbeck A/S, Ottiliavej 9, Valby 2500, Denmark
| | - L S Lerdrup
- Symptoms Biology, H. Lundbeck A/S, Ottiliavej 9, Valby 2500, Denmark
| | - L Arvastson
- Bioinformatics, H. Lundbeck A/S, Ottiliavej 9, Valby 2500, Denmark
| | - S Aznar
- Molecular Biology, Bispebjerg Hospital, Copenhagen University Hospital, Bispebjerg Bakke 23, Copenhagen 2400, Denmark
| | - J T Andreasen
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, Copenhagen 2100, Denmark.
| |
Collapse
|
7
|
Pillidge K, Porter AJ, Young JW, Stanford SC. Perseveration by NK1R-/- ('knockout') mice is blunted by doses of methylphenidate that affect neither other aspects of their cognitive performance nor the behaviour of wild-type mice in the 5-Choice Continuous Performance Test. J Psychopharmacol 2016; 30:837-47. [PMID: 27097734 PMCID: PMC4994704 DOI: 10.1177/0269881116642541] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The underlying cause(s) of abnormalities expressed by patients with attention deficit hyperactivity disorder (ADHD) have yet to be delineated. One factor that has been associated with increased vulnerability to ADHD is polymorphism(s) of TACR1, which is the human equivalent of the rodent NK1 (substance P-preferring) receptor gene (Nk1r). We have reported previously that genetically altered mice, lacking functional NK1R (NK1R-/-), express locomotor hyperactivity, which was blunted by the first-line treatment for ADHD, methylphenidate. Here, we compared the effects of this psychostimulant (3, 10 and 30 mg/kg, intraperitoneally) on the behaviour of NK1R-/- mice and their wild types in the 5-Choice Continuous Performance Test, which emulates procedures used to study attention and response control in ADHD patients. Methylphenidate increased total trials (a measure of 'productivity') completed by wild types, but not by NK1R-/- mice. Conversely, this drug reduced perseveration by NK1R-/- mice, but not by wild types. Other drug-induced changes in key behaviours were not genotype dependent, especially at the highest dose: for example, % omissions (an index of inattentiveness) was increased, whereas % false alarms and % premature responses (measures of impulsivity) declined in both genotypes, indicating reduced overall response. These findings are discussed in the context of the efficacy of methylphenidate in the treatment of ADHD. Moreover, they lead to several testable proposals. First, methylphenidate does not improve attention in a subgroup of ADHD patients with a functional deficit of TACR1. Second, these patients do not express excessive false alarms when compared with other groups of subjects, but they do express excessive perseveration, which would be ameliorated by methylphenidate.
Collapse
Affiliation(s)
- Katharine Pillidge
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Ashley J Porter
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Jared W Young
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA,Research Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - S Clare Stanford
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| |
Collapse
|
8
|
Yang X, Morris SM, Gearhart JM, Ruark CD, Paule MG, Slikker W, Mattison DR, Vitiello B, Twaddle NC, Doerge DR, Young JF, Fisher JW. Development of a physiologically based model to describe the pharmacokinetics of methylphenidate in juvenile and adult humans and nonhuman primates. PLoS One 2014; 9:e106101. [PMID: 25184666 PMCID: PMC4153582 DOI: 10.1371/journal.pone.0106101] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 07/28/2014] [Indexed: 11/18/2022] Open
Abstract
The widespread usage of methylphenidate (MPH) in the pediatric population has received considerable attention due to its potential effect on child development. For the first time a physiologically based pharmacokinetic (PBPK) model has been developed in juvenile and adult humans and nonhuman primates to quantitatively evaluate species- and age-dependent enantiomer specific pharmacokinetics of MPH and its primary metabolite ritalinic acid. The PBPK model was first calibrated in adult humans using in vitro enzyme kinetic data of MPH enantiomers, together with plasma and urine pharmacokinetic data with MPH in adult humans. Metabolism of MPH in the small intestine was assumed to account for the low oral bioavailability of MPH. Due to lack of information, model development for children and juvenile and adult nonhuman primates primarily relied on intra- and interspecies extrapolation using allometric scaling. The juvenile monkeys appear to metabolize MPH more rapidly than adult monkeys and humans, both adults and children. Model prediction performance is comparable between juvenile monkeys and children, with average root mean squared error values of 4.1 and 2.1, providing scientific basis for interspecies extrapolation of toxicity findings. Model estimated human equivalent doses in children that achieve similar internal dose metrics to those associated with pubertal delays in juvenile monkeys were found to be close to the therapeutic doses of MPH used in pediatric patients. This computational analysis suggests that continued pharmacovigilance assessment is prudent for the safe use of MPH.
Collapse
Affiliation(s)
- Xiaoxia Yang
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
- * E-mail:
| | - Suzanne M. Morris
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - Jeffery M. Gearhart
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Wright-Patterson Air Force Base, Ohio, United States of America
| | - Christopher D. Ruark
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Wright-Patterson Air Force Base, Ohio, United States of America
| | - Merle G. Paule
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - William Slikker
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - Donald R. Mattison
- Risk Sciences International, Ottawa, Ontario, Canada
- University of Ottawa, Ottawa, Ontario, Canada
| | - Benedetto Vitiello
- National Institute of Mental Health, Bethesda, Maryland, United States of America
| | - Nathan C. Twaddle
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - Daniel R. Doerge
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - John F. Young
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - Jeffrey W. Fisher
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| |
Collapse
|
9
|
Combs CC, Hankins EL, Copeland CL, Brown SD, Pond BB. Quantitative determination of d- and l-threo enantiomers of methylphenidate in brain tissue by liquid chromatography-mass spectrometry. Biomed Chromatogr 2013; 27:1587-9. [PMID: 23813595 DOI: 10.1002/bmc.2975] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 05/21/2013] [Accepted: 05/23/2013] [Indexed: 11/09/2022]
Abstract
Methylphenidate, a psychostimulant used for the treatment of attention deficit hyperactivity disorder and narcolepsy, is administered as a 50:50 racemic mixture, despite the fact that d-methylphenidate has been shown to have greater pharmacologic activity. This paper presents a validated LC-MS/MS approach to separation and quantification of methylphenidate enantiomers using a vancomycin column and triethylammonium acetate to enhance the chiral separation. The method is applicable to the monitoring of these enantiomers in mouse brain, with a limit of detection of 0.5 ng/mL and a lower limit of quantification of 7.5 ng/mL.
Collapse
Affiliation(s)
- Carolyn C Combs
- Bill Gatton College of Pharmacy, East Tennessee State University, Pharmaceutical Sciences, Johnson City, Tennessee, USA
| | | | | | | | | |
Collapse
|
10
|
Griffin WC, McGovern RW, Bell GH, Randall PK, Middaugh LD, Patrick KS. Interactive effects of methylphenidate and alcohol on discrimination, conditioned place preference and motor coordination in C57BL/6J mice. Psychopharmacology (Berl) 2013; 225:613-25. [PMID: 22955568 PMCID: PMC3547134 DOI: 10.1007/s00213-012-2849-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 08/10/2012] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Prior research indicates methylphenidate (MPH) and alcohol (ethanol, EtOH) interact to significantly affect responses humans and mice. The present studies tested the hypothesis that MPH and EtOH interact to potentiate ethanol-related behaviors in mice. METHODS We used several behavioral tasks including: drug discrimination in MPH-trained and EtOH-trained mice, conditioned place preference (CPP), rota-rod and the parallel rod apparatus. We also used gas chromatographic methods to measure brain tissue levels of EtOH and the D- and L-isomers of MPH and the metabolite, ethylphenidate (EPH). RESULTS In discrimination, EtOH (1 g/kg) produced a significant leftward shift in the MPH generalization curve (1-2 mg/kg) for MPH-trained mice, but no effects of MPH (0.625-1.25 mg/kg) on EtOH discrimination in EtOH-trained mice (0-2.5 g/kg) were observed. In CPP, the MPH (1.25 mg/kg) and EtOH (1.75 g/kg) combination significantly increased time on the drug paired side compared to vehicle (30.7 %), but this was similar to MPH (28.8 %) and EtOH (33.6 %). Footslip errors measured in a parallel rod apparatus indicated that the drug combination was very ataxic, with footslips increasing 29.5 % compared to EtOH. Finally, brain EtOH concentrations were not altered by 1.75 g/kg EtOH combined with 1.25 mg/kg MPH. However, EtOH significantly increased D-MPH and L-EPH without changing L-MPH brain concentrations. CONCLUSIONS The enhanced behavioral effects when EtOH is combined with MPH are likely due to the selective increase in brain D-MPH concentrations. These studies are consistent with observations in humans of increased interoceptive awareness of the drug combination and provide new clinical perspectives regarding enhanced ataxic effects of this drug combination.
Collapse
Affiliation(s)
- William C. Griffin
- Charleston Alcohol Research Center, Center for Drug and Alcohol Programs, Department of Psychiatry and Behavioral Science, Medical University of South Carolina, Charleston, SC 29425-0742
| | | | - Guinevere H. Bell
- Department of Pharmaceutical and Biomedical Sciences Medical University of South Carolina, Charleston, SC 29425-0742
| | - Patrick K. Randall
- Charleston Alcohol Research Center, Center for Drug and Alcohol Programs, Department of Psychiatry and Behavioral Science, Medical University of South Carolina, Charleston, SC 29425-0742
| | - Lawrence D. Middaugh
- Charleston Alcohol Research Center, Center for Drug and Alcohol Programs, Department of Psychiatry and Behavioral Science, Medical University of South Carolina, Charleston, SC 29425-0742
,Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425-0742
| | - Kennerly S. Patrick
- Department of Pharmaceutical and Biomedical Sciences Medical University of South Carolina, Charleston, SC 29425-0742
| |
Collapse
|
11
|
Abstract
People with attention-deficit/hyperactivity disorder (ADHD) often experience sleep problems, and these are frequently exacerbated by the methylphenidate they take to manage their ADHD symptoms. Many of the changes to sleep are consistent with a change in the underlying circadian clock. The present study was designed to determine if methylphenidate alone could alter properties of the circadian clock. Young male mice were examined in light-dark cycles and in constant darkness and recordings were performed on behavioral activity, sleep, and electrical activity in the suprachiasmatic nucleus (SCN) of freely moving mice. Methylphenidate in the drinking water (0.08%) significantly increased activity in the mid-to-late night, and led to a delay in the onset of activity and sleep relative to the light-dark cycle. While locomotor levels returned to baseline after treatment ended, the phase angle of entrainment required at least a week to return to baseline levels. In constant darkness, the free-running period of both wheel-running and general locomotor rhythms was lengthened by methylphenidate. When the treatment ended, the free-running period either remained stable or only partially reverted to baseline levels. Methylphenidate also altered the electrical firing rate rhythms in the SCN. It induced a delay in the trough of the rhythm, an increment in rhythm amplitude, and a reduction in rhythm variability. These observations suggest that methylphenidate alters the underlying circadian clock. The observed changes are consistent with clock alterations that would promote sleep-onset insomnia.
Collapse
|
12
|
Levi MS, Divine B, Hanig JP, Doerge DR, Vanlandingham MM, George NI, Twaddle NC, Bowyer JF. A comparison of methylphenidate-, amphetamine-, and methamphetamine-induced hyperthermia and neurotoxicity in male Sprague–Dawley rats during the waking (lights off) cycle. Neurotoxicol Teratol 2012; 34:253-62. [DOI: 10.1016/j.ntt.2012.01.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Revised: 01/12/2012] [Accepted: 01/14/2012] [Indexed: 10/14/2022]
|
13
|
Morris SM, Petibone DM, Lin WJ, Chen JJ, Vitiello B, Witt KL, Mattison DR. The genetic toxicity of methylphenidate: a review of the current literature. J Appl Toxicol 2012; 32:756-64. [PMID: 22337063 DOI: 10.1002/jat.2721] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Accepted: 12/23/2011] [Indexed: 12/15/2022]
Abstract
Attention deficit/hyperactivity disorder (ADHD), a common children's behavioral disorder, is characterized by inattention, hyperactivity and impulsivity. The disorder is thought to stem from abnormalities in the catecholamine pathway and the symptoms of the disorder have been successfully treated with methylphenidate (MPH) since the FDA approved the drug in the 1950s. MPH underwent the appropriate safety testing as part of the FDA approval process; however, a publication in 2005 that reported significant increases in cytogenetic damage in the lymphocytes of MPH-treated pediatric patients caused concern for patients and their families, the pharmaceutical industry and regulatory agencies. This communication will review the many studies that were subsequently initiated worldwide to address the genetic safety of MPH in both animal models and human subjects. Animal experiments broadened the study protocols used in the 2005 investigation to include a wider dose-range, a longer treatment period and automated scoring of biological endpoints, where possible, to reduce observer bias. The human subject studies replicated the experimental design used in the 2005 study, but increased the treatment periods and the sizes of the study populations. Neither the laboratory animal nor human subject studies found an increase in any of the measures of genetic damage that were evaluated. Taken together, these new studies are consistent with the original safety evaluation of the FDA and do not support the hypothesis that MPH treatment increases the risk of genetic damage in ADHD patients. Published 2012. This article is a US Government work and is in the public domain in the USA.
Collapse
Affiliation(s)
- Suzanne M Morris
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US FDA, Jefferson, AR 72079, USA.
| | | | | | | | | | | | | |
Collapse
|
14
|
Zamora-Perez AL, Lazalde-Ramos BP, Sosa-Macías MG, Gómez-Meda BC, Torres-Bugarín O, Zúñiga-González GM. Methylphenidate lacks genotoxic effects in mouse peripheral blood erythrocytes. Drug Chem Toxicol 2011; 34:294-9. [PMID: 21649484 DOI: 10.3109/01480545.2010.536770] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Methylphenidate (MPH; Ritalin®; Novartis Pharmaceuticals, Inc., Basel, Switzerland) has been prescribed to treat attention deficit/hyperactivity disorder (ADHD) since its approval by the U.S. Food and Drug Administration over 50 years ago. Due to concerns that MPH might induce cytogenetic alterations in children, treatment with this drug has been a controversial issue. In the present study, we assessed the frequency of micronucleated erythrocytes (MNEs), micronucleated polychromatic erythrocytes (MNPCEs), and polychromatic erythrocytes (PCEs) in peripheral blood samples from mice treated with three different doses of MPH (30, 60, or 125 mg/kg). We found no evidence of increased MNEs or MNPCEs, nor did PCEs decline. These results add to the accumulating evidence that MPH does not induce genotoxic or cytotoxic damage.
Collapse
Affiliation(s)
- Ana L Zamora-Perez
- Instituto de Investigación en Odontología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México.
| | | | | | | | | | | |
Collapse
|
15
|
Carrey N, Wilkinson M. A review of psychostimulant-induced neuroadaptation in developing animals. Neurosci Bull 2011; 27:197-214. [PMID: 21614102 DOI: 10.1007/s12264-011-1004-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The effects of clinically relevant doses of commonly prescribed stimulants methylphenidate (MPH), d-amphetamine (d-AMPH), and dl-AMPH or mixed amphetamine salts (MAS) such as Adderall, on short- and long-term gene neuroadaptations in developing animals have not been widely investigated. In the present review, the effects of oral stimulant administration were compared with those of the subcutaneous or intra-peritoneal route. A selective set of studies between 1979 and 2010, which incorporated in their design developmental period, clinically relevant doses of stimulants, and repeated daily doses were reviewed. These studies indicate that neuroadaptation to chronic stimulants includes blunting of stimulated immediate early gene expression, sensitivity of younger (prepubertal) brain to smaller dosages of stimulants, and the persistence of some effects, especially behavioral neuroadaptations, into adulthood. In addition, oral amphetamines (MAS) have more profound effects than does oral MPH. Further animal developmental studies are required to understand potential long-term neuroadaptations to low, daily oral doses of stimulants. Implications for clinical practice were also discussed.
Collapse
Affiliation(s)
- Normand Carrey
- Department of Psychiatry, IWK Health Centre, Halifax, Nova Scotia, Canada.
| | | |
Collapse
|
16
|
Bell GH, Novak AJ, Griffin WC, Patrick KS. Transdermal and oral dl-methylphenidate-ethanol interactions in C57BL/6J mice: transesterification to ethylphenidate and elevation of d-methylphenidate concentrations. J Pharm Sci 2011; 100:2966-78. [PMID: 21240977 DOI: 10.1002/jps.22476] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2010] [Revised: 10/12/2010] [Accepted: 12/13/2010] [Indexed: 01/17/2023]
Abstract
We tested the hypothesis that C57BL/6J mice will model human metabolic interactions between dl-methylphenidate (MPH) and ethanol, placing an emphasis on the MPH transdermal system (MTS). Specifically, we asked: (1) will ethanol increase d-MPH biological concentrations, (2) will MTS facilitate the systemic bioavailability of l-MPH, and (3) will l-MPH enantioselectively interact with ethanol to yield l-ethylphenidate (l-EPH)? Mice were dosed with MTS (¼ of a 12.5 cm(2) patch on shaved skin) or a comparable oral dl-MPH dose (7.5 mg/kg), with or without ethanol (3.0 g/kg), and then placed in metabolic cages for 3 h. MPH and EPH isomer concentrations in blood, brain, and urine were analyzed by gas chromatographic-mass spectrometry monitoring of N-(S)-prolylpiperidyl fragments. As in humans, MTS greatly facilitated the absorption of l-MPH in this mouse strain. Similarly, ethanol led to the enantioselective formation of l-EPH and to an elevation in d-MPH concentrations with both MTS and oral MPH. Although only guarded comparisons between MTS and oral MPH can be made due to route-dependent drug absorption rate differences, MTS was associated with significant MPH-ethanol interactions. Ethanol-mediated increases in circulating concentrations of d-MPH carry toxicological and abuse liability implications should this animal model hold for ethanol-consuming attention-deficit hyperactivity disorder patients or coabusers.
Collapse
Affiliation(s)
- Guinevere H Bell
- Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina 29425, USA.
| | | | | | | |
Collapse
|
17
|
Mattison DR. Research on cytogenetic risk of ADHD treatments in children. J Atten Disord 2010; 14:203-4. [PMID: 20978275 DOI: 10.1177/1087054710381477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Donald R. Mattison
- Eunice Kennedy Shriver National Institute of Child Health and Human Development
| |
Collapse
|
18
|
Walitza S, Kämpf K, Gnana Oli R, Warnke A, Gerlach M, Stopper H. Prospective follow-up studies found no chromosomal mutagenicity of methylphenidate therapy in ADHD affected children. Toxicol Lett 2010; 193:4-8. [DOI: 10.1016/j.toxlet.2009.12.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Revised: 12/14/2009] [Accepted: 12/14/2009] [Indexed: 10/20/2022]
|
19
|
Dobrovolsky VN, Boctor SY, Twaddle NC, Doerge DR, Bishop ME, Manjanatha MG, Kimoto T, Miura D, Heflich RH, Ferguson SA. Flow cytometric detection of Pig-A mutant red blood cells using an erythroid-specific antibody: application of the method for evaluating the in vivo genotoxicity of methylphenidate in adolescent rats. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2010; 51:138-145. [PMID: 19658152 DOI: 10.1002/em.20519] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
A modified flow cytometry assay for Pig-A mutant rat red blood cells (RBCs) was developed using an antibody that positively identifies rat RBCs (monoclonal antibody HIS49). The assay was used in conjunction with a flow cytometric micronucleus (MN) assay to evaluate gene mutation and clastogenicity/aneugenicity in adolescent male and female rats treated with methylphenidate hydrochloride (MPH). Sprague-Dawley rats were treated orally with 3 mg/kg MPH (70/sex) or water (40/sex) 3 x /day on postnatal days (PNDs) 29-50. Eight additional rats (4/sex) were injected i.p. with N-ethyl-N-nitrosourea (ENU) on PND 28. Blood was collected on PNDs 29, 50, and 90, and used for determining serum MPH levels and/or conducting genotoxicity assays. On the first and last days of MPH treatment (PNDs 29 and 50), serum MPH levels averaged 21 pg/microl, well within the clinical treatment range. Relative to our previously published method (Miura et al. [2008]; Environ Mol Mutagen 49: 614-629), the HIS49 Pig-A mutation assay significantly reduced the background RBC mutant frequency; in the experiments with ENU-treated rats, the modification increased the overall sensitivity of the assay 2-3 fold. Even with the increased assay sensitivity, the 21 consecutive days of MPH treatment produced no evidence of Pig-A mutation induction (measured at PND 90); in addition, MPH treatment did not increase MN frequency (measured at PND 50). These results support the consensus view that the genotoxicity of MPH in pediatric patients reported earlier (El-Zein et al. [2005]: Cancer Lett 230: 284-291) cannot be reproduced in animal models, suggesting that MPH at clinically relevant levels may be nongenotoxic in humans.
Collapse
Affiliation(s)
- Vasily N Dobrovolsky
- Division of Genetic and Reproductive Toxicology; National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Witt KL, Malarkey DE, Hobbs CA, Davis JP, Kissling GE, Caspary W, Travlos G, Recio L. No increases in biomarkers of genetic damage or pathological changes in heart and brain tissues in male rats administered methylphenidate hydrochloride (Ritalin) for 28 days. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2010; 51:80-88. [PMID: 19634155 PMCID: PMC2807377 DOI: 10.1002/em.20515] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Following a 2005 report of chromosomal damage in children with attention deficit/hyperactivity disorder (ADHD) who were treated with the commonly prescribed medication methylphenidate (MPH), numerous studies have been conducted to clarify the risk for MPH-induced genetic damage. Although most of these studies reported no changes in genetic damage endpoints associated with exposure to MPH, one recent study (Andreazza et al. [2007]: Prog Neuropsychopharmacol Biol Psychiatry 31:1282-1288) reported an increase in DNA damage detected by the Comet assay in blood and brain cells of Wistar rats treated by intraperitoneal injection with 1, 2, or 10 mg/kg MPH; no increases in micronucleated lymphocyte frequencies were observed in these rats. To clarify these findings, we treated adult male Wistar Han rats with 0, 2, 10, or 25 mg/kg MPH by gavage once daily for 28 consecutive days and measured micronucleated reticulocyte (MN-RET) frequencies in blood, and DNA damage in blood, brain, and liver cells 4 hr after final dosing. Flow cytometric evaluation of blood revealed no significant increases in MN-RET. Comet assay evaluations of blood leukocytes and cells of the liver, as well as of the striatum, hippocampus, and frontal cortex of the brain showed no increases in DNA damage in MPH-treated rats in any of the three treatment groups. Thus, the previously reported observations of DNA damage in blood and brain tissue of rats exposed to MPH for 28 days were not confirmed in this study. Additionally, no histopathological changes in brain or heart, or elevated serum biomarkers of cardiac injury were observed in these MPH-exposed rats.
Collapse
Affiliation(s)
- Kristine L Witt
- Biomolecular Screening Branch, NIEHS/NTP, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Manjanatha MG, Shelton SD, Dobrovolsky VN, Shaddock JG, McGarrity LG, Twaddle NW, Moore MM, Mattison DR, Slikker W, Morris SM. Evaluation of mutagenic mode of action in Big Blue mice fed methylphenidate for 24 weeks. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2009; 680:43-8. [DOI: 10.1016/j.mrgentox.2009.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2009] [Revised: 09/05/2009] [Accepted: 09/11/2009] [Indexed: 11/16/2022]
|
22
|
Tucker JD, Suter W, Petibone DM, Thomas RA, Bailey NL, Zhou Y, Zhao Y, Muniz R, Kumar V. Cytogenetic assessment of methylphenidate treatment in pediatric patients treated for attention deficit hyperactivity disorder. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2009; 677:53-8. [DOI: 10.1016/j.mrgentox.2009.05.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2009] [Revised: 05/12/2009] [Accepted: 05/13/2009] [Indexed: 12/15/2022]
|
23
|
The genetic toxicology of methylphenidate hydrochloride in non-human primates. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2009; 673:59-66. [DOI: 10.1016/j.mrgentox.2008.12.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2008] [Revised: 12/09/2008] [Accepted: 12/10/2008] [Indexed: 02/06/2023]
|