1
|
Li H, Mei L, Nie X, Wu L, Lv L, Ren X, Yang J, Cao H, Wu J, Zhang Y, Hu Y, Wang W, Turck CW, Shi B, Li J, Xu L, Hu X. The Tree Shrew Model of Parkinson Disease: A Cost-Effective Alternative to Nonhuman Primate Models. J Transl Med 2024; 104:102145. [PMID: 39343009 DOI: 10.1016/j.labinv.2024.102145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 09/13/2024] [Accepted: 09/20/2024] [Indexed: 10/01/2024] Open
Abstract
The surge in demand for experimental monkeys has led to a rapid increase in their costs. Consequently, there is a growing need for a cost-effective model of Parkinson disease (PD) that exhibits all core clinical and pathologic phenotypes. Evolutionarily, tree shrews (Tupaia belangeri) are closer to primates in comparison with rodents and could be an ideal species for modeling PD. To develop a tree shrew PD model, we used the 1-methyl-4-phenylpyridinium (MPP+), a metabolite derived from 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, to induce lesions in dopaminergic neurons of the unilateral substantia nigra. The induced tree shrew model consistently exhibited and maintained all classic clinical manifestations of PD for a 5-month period. The symptoms included bradykinesia, rest tremor, and postural instability, and ∼50% individuals showed apomorphine-induced rotations, a classic phenotype of unilateral PD models. All these are closely resembled the ones observed in PD monkeys. Meanwhile, this model was also sensitive to L-dopa treatment in a dose-dependent manner, which suggested that the motor deficits are dopamine dependent. Immunostaining showed a significant loss of dopaminergic neurons (∼95%) in the lesioned substantia nigra, which is a crucial PD pathological marker. Moreover, a control group of nigral saline injection did not show any motor deficits and pathological changes. Cytomorphologic analysis revealed that the size of nigral dopaminergic neurons in tree shrews is much bigger than that of rodents and is close to that of macaques. The morphologic similarity may be an important structural basis for the manifestation of the highly similar phenotypes between monkey and tree shrew PD models. Collectively, in this study, we have successfully developed a PD model in a small animal species that faithfully recapitulated the classic clinical symptoms and key pathological indicators of PD monkeys, providing a novel and low-cost avenue for evaluation of PD treatments and underlying mechanisms.
Collapse
Affiliation(s)
- Hao Li
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, and National Resource Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Leyi Mei
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, and National Resource Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Institutes of Physical Science and Information Technology, Anhui University, Hefei, China
| | - Xiupeng Nie
- Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Liping Wu
- Department of Endocrinology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Longbao Lv
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, and National Resource Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, China
| | - Xiaofeng Ren
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, and National Resource Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, China
| | - Jitong Yang
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, and National Resource Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, China
| | - Haonan Cao
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, and National Resource Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Jing Wu
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, and National Resource Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yuhua Zhang
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, and National Resource Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yingzhou Hu
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, and National Resource Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Wenchao Wang
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, and National Resource Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Christoph W Turck
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, and National Resource Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Max Planck Institute of Psychiatry, Munich, Germany.
| | - Bingyin Shi
- Department of Endocrinology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Jiali Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Department of Neurology, Hackensack Meridian School of Medicine, Nutley, New Jersey.
| | - Lin Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.
| | - Xintian Hu
- National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, and National Resource Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.
| |
Collapse
|
2
|
Togawa S, Usui N, Doi M, Kobayashi Y, Koyama Y, Nakamura Y, Shinoda K, Kobayashi H, Shimada S. Neuroprotective effects of Si-based hydrogen-producing agent on 6-hydroxydopamine-induced neurotoxicity in juvenile mouse model. Behav Brain Res 2024; 468:115040. [PMID: 38723675 DOI: 10.1016/j.bbr.2024.115040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024]
Abstract
Neurotoxins have been extensively investigated, particularly in the field of neuroscience. They induce toxic damage, oxidative stress, and inflammation on neurons, triggering neuronal dysfunction and neurodegenerative diseases. Here we demonstrate the neuroprotective effect of a silicon (Si)-based hydrogen-producing agent (Si-based agent) in a juvenile neurotoxic mouse model induced by 6-hydroxydopamine (6-OHDA). The Si-based agent produces hydrogen in bowels and functions as an antioxidant and anti-inflammatory agent. However, the effects of the Si-based agent on neural degeneration in areas other than the lesion and behavioral alterations caused by it are largely unknown. Moreover, the neuroprotective effects of Si-based agent in the context of lactation and use during infancy have not been explored in prior studies. In this study, we show the neuroprotective effect of the Si-based agent on 6-OHDA during lactation period and infancy using the mouse model. The Si-based agent safeguards against the degradation and neuronal cell death of dopaminergic neurons and loss of dopaminergic fibers in the striatum (STR) and ventral tegmental area (VTA) caused by 6-OHDA. Furthermore, the Si-based agent exhibits a neuroprotective effect on the length of axon initial segment (AIS) in the layer 2/3 (L2/3) neurons of the medial prefrontal cortex (mPFC). As a result, the Si-based agent mitigates hyperactive behavior in a juvenile neurotoxic mouse model induced by 6-OHDA. These results suggest that the Si-based agent serves as an effective neuroprotectant and antioxidant against neurotoxic effects in the brain, offering the possibility of the Si-based agent as a neuroprotectant for nervous system diseases.
Collapse
Affiliation(s)
- Shogo Togawa
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Yamaguchi 755-8505, Japan
| | - Noriyoshi Usui
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; Omics Center, Center of Medical Innovation and Translational Research, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; United Graduate School of Child Development, Osaka University, Suita 565-0871, Japan; Global Center for Medical Engineering and Informatics, Osaka University, Suita 565-0871, Japan; Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka 541-8567, Japan.
| | - Miyuki Doi
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka 541-8567, Japan
| | - Yuki Kobayashi
- SANKEN (Institute of Scientific and Industrial Research), Osaka University, Ibaraki, 567-0047, Japan
| | - Yoshihisa Koyama
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka 541-8567, Japan
| | - Yukiko Nakamura
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka 541-8567, Japan
| | - Koh Shinoda
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Yamaguchi 755-8505, Japan
| | - Hikaru Kobayashi
- SANKEN (Institute of Scientific and Industrial Research), Osaka University, Ibaraki, 567-0047, Japan
| | - Shoichi Shimada
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; United Graduate School of Child Development, Osaka University, Suita 565-0871, Japan; Global Center for Medical Engineering and Informatics, Osaka University, Suita 565-0871, Japan; Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka 541-8567, Japan
| |
Collapse
|
3
|
Sif-Eddine W, Ba-M'hamed S, Lefranc B, Leprince J, Boukhzar L, Anouar Y, Bennis M. Selenoprotein T, a potential treatment of attention-deficit/hyperactivity disorder and comorbid pain in neonatal 6-OHDA lesioned mice. Exp Mol Pathol 2024; 137:104905. [PMID: 38797131 DOI: 10.1016/j.yexmp.2024.104905] [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: 12/17/2023] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 05/29/2024]
Abstract
pathological pain and Attention-deficit/hyperactivity disorder (ADHD) are two complex multifactorial syndromes. The comorbidity of ADHD and altered pain perception is well documented in children, adolescents, and adults. According to pathophysiological investigations, the dopaminergic system's dysfunction provides a common basis for ADHD and comorbid pain. Growing evidence suggests that oxidative stress may be crucial in both pathologies. Recent studies revealed that a small peptide encompassing the redox-active site of selenoprotein T (PSELT), protects dopaminergic neurons and fibers as well as lesioned nerves in animal models. The current study aims to examine the effects of PSELT treatment on ADHD-like symptoms and pain sensitivity, as well as the role of catecholaminergic systems in these effects. Our results demonstrated that intranasal administration of PSELT reduced the hyperactivity in the open field, decreased the impulsivity displayed by 6-OHDA-lesioned male mice in the 5-choice serial reaction time task test and improved attentional performance. In addition, PSELT treatment significantly increased the nociception threshold in both normal and inflammatory conditions. Furthermore, anti-hyperalgesic activity was antagonized with sulpiride pre-treatment, but not by phentolamine, or propranolol pre-treatments. The present study suggests that PSELT reduces the severity of ADHD symptoms in mice and possesses potent antinociceptive effects which could be related to the involvement of D2/D3 dopaminergic receptors.
Collapse
Affiliation(s)
- Wahiba Sif-Eddine
- Laboratory of Pharmacology, Neurobiology, Anthropobiology, and Environment, Faculty of Sciences, Cadi Ayyad University, Marrakesh, Morocco
| | - Saadia Ba-M'hamed
- Laboratory of Pharmacology, Neurobiology, Anthropobiology, and Environment, Faculty of Sciences, Cadi Ayyad University, Marrakesh, Morocco
| | - Benjamin Lefranc
- Univ Rouen Normandie, INSERM, NorDiC, UMR 1239, Rouen, France; Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Jérôme Leprince
- Univ Rouen Normandie, INSERM, NorDiC, UMR 1239, Rouen, France; Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Loubna Boukhzar
- Laboratory of Pharmacology, Neurobiology, Anthropobiology, and Environment, Faculty of Sciences, Cadi Ayyad University, Marrakesh, Morocco; Univ Rouen Normandie, INSERM, NorDiC, UMR 1239, Rouen, France; Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Youssef Anouar
- Univ Rouen Normandie, INSERM, NorDiC, UMR 1239, Rouen, France; Institute for Research and Innovation in Biomedicine, Rouen, France.
| | - Mohamed Bennis
- Laboratory of Pharmacology, Neurobiology, Anthropobiology, and Environment, Faculty of Sciences, Cadi Ayyad University, Marrakesh, Morocco
| |
Collapse
|
4
|
Choi SH, Bae HJ, Kim SY, Mony TJ, Kim HJ, Cho YE, Choi YY, An JY, Cho SY, Kim DH, Park SJ. Particulate matter (PM 10) exacerbates on MK-801-induced schizophrenia-like behaviors through the inhibition of ERK-CREB-BDNF signaling pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 276:116294. [PMID: 38574646 DOI: 10.1016/j.ecoenv.2024.116294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 03/02/2024] [Accepted: 04/01/2024] [Indexed: 04/06/2024]
Abstract
Particulate matter (PM), released into the air by a variety of natural and human activities, is a key indicator of air pollution. Although PM is known as the extensive health hazard to affect a variety of illness, few studies have specifically investigated the effects of PM10 exposure on schizophrenic development. In the present study, we aimed to investigate the impact of PM10 on MK-801, N-methyl-D-aspartate (NMDA) receptor antagonist, induced schizophrenia-like behaviors in C57BL/6 mouse. Preadolescent mice were exposed PM10 to 3.2 mg/m3 concentration for 4 h/day for 2 weeks through a compartmentalized whole-body inhalation chamber. After PM10 exposure, we conducted behavioral tests during adolescence and adulthood to investigate longitudinal development of schizophrenia. We found that PM10 exacerbated schizophrenia-like behavior, such as psychomotor agitation, social interaction deficits and cognitive deficits at adulthood in MK-801-induced schizophrenia animal model. Furthermore, the reduced expression levels of brain-derived neurotrophic factor (BDNF) and the phosphorylation of BDNF related signaling molecules, extracellular signal-regulated kinase (ERK) and cAMP response element-binding protein (CREB), were exacerbated by PM10 exposure in the adult hippocampus of MK-801-treated mice. Thus, our present study demonstrates that exposure to PM10 in preadolescence exacerbates the cognitive impairment in animal model of schizophrenia, which are considered to be facilitated by the decreased level of BDNF through reduced ERK-CREB expression.
Collapse
Affiliation(s)
- Seung-Hyuk Choi
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Ho Jung Bae
- Agriculture and Life Science Research Institute, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - So-Yeon Kim
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Tamanna Jahan Mony
- Agriculture and Life Science Research Institute, Kangwon National University, Chuncheon 24341, Republic of Korea; Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO, USA
| | - Hyun-Jeong Kim
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Ye Eun Cho
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Yu-Yeong Choi
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Ju-Yeon An
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - So-Young Cho
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Dong Hyun Kim
- Department of Pharmacology and Department of Advanced Translational Medicine, School of Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Se Jin Park
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon 24341, Republic of Korea; School of Natural Resources and Environmental Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea; Agriculture and Life Science Research Institute, Kangwon National University, Chuncheon 24341, Republic of Korea.
| |
Collapse
|
5
|
Bou Sader Nehme S, Sanchez-Sarasua S, Adel R, Tuifua M, Ali A, Essawy AE, Abdel Salam S, Hleihel W, Boué-Grabot E, Landry M. P2X4 signalling contributes to hyperactivity but not pain sensitization comorbidity in a mouse model of attention deficit/hyperactivity disorder. Front Pharmacol 2024; 14:1288994. [PMID: 38239187 PMCID: PMC10794506 DOI: 10.3389/fphar.2023.1288994] [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: 09/05/2023] [Accepted: 12/12/2023] [Indexed: 01/22/2024] Open
Abstract
Introduction: Attention deficit/hyperactivity disorder (ADHD) is a common neurodevelopmental disorder characterized by hyperactivity, inattention, and impulsivity that often persist until adulthood. Frequent comorbid disorders accompany ADHD and two thirds of children diagnosed with ADHD also suffer from behavioural disorders and from alteration of sensory processing. We recently characterized the comorbidity between ADHD-like symptoms and pain sensitisation in a pharmacological mouse model of ADHD, and we demonstrated the implication of the anterior cingulate cortex and posterior insula. However, few studies have explored the causal mechanisms underlying the interactions between ADHD and pain. The implication of inflammatory mechanisms has been suggested but the signalling pathways involved have not been explored. Methods: We investigated the roles of purinergic signalling, at the crossroad of pain and neuroinflammatory pathways, by using a transgenic mouse line that carries a total deletion of the P2X4 receptor. Results: We demonstrated that P2X4 deletion prevents hyperactivity in the mouse model of ADHD. In contrast, the absence of P2X4 lowered thermal pain thresholds in sham conditions and did not affect pain sensitization in ADHD-like conditions. We further analysed microglia reactivity and the expression of inflammatory markers in wild type and P2X4KO mice. Our results revealed that P2X4 deletion limits microglia reactivity but at the same time exerts proinflammatory effects in the anterior cingulate cortex and posterior insula. Conclusion: This dual role of P2X4 could be responsible for the differential effects noted on ADHD-like symptoms and pain sensitization and calls for further studies to investigate the therapeutic benefit of targeting the P2X4 receptor in ADHD patients.
Collapse
Affiliation(s)
- Sarah Bou Sader Nehme
- University of Bordeaux, CNRS, Institute of Neurodegenerative Diseases, IMN, UMR 5293, Bordeaux, France
- Department of Biology, Faculty of Arts and Sciences, Holy Spirit University of Kaslik, Jounieh, Lebanon
| | - Sandra Sanchez-Sarasua
- University of Bordeaux, CNRS, Institute of Neurodegenerative Diseases, IMN, UMR 5293, Bordeaux, France
- Faculty of Health Sciences, University of Jaume I, Castellon, Spain
| | - Ramy Adel
- Zoology Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Marie Tuifua
- University of Bordeaux, CNRS, Institute of Neurodegenerative Diseases, IMN, UMR 5293, Bordeaux, France
| | - Awatef Ali
- Zoology Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Amina E. Essawy
- Zoology Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Sherine Abdel Salam
- Zoology Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Walid Hleihel
- Department of Biology, Faculty of Arts and Sciences, Holy Spirit University of Kaslik, Jounieh, Lebanon
| | - Eric Boué-Grabot
- University of Bordeaux, CNRS, Institute of Neurodegenerative Diseases, IMN, UMR 5293, Bordeaux, France
| | - Marc Landry
- University of Bordeaux, CNRS, Institute of Neurodegenerative Diseases, IMN, UMR 5293, Bordeaux, France
| |
Collapse
|
6
|
Runkel MT, Tarabishi A, Shay-Winkler K, Emmert ME, Goh Q, Cornwall R. The role of sympathetic innervation in neonatal muscle growth and neuromuscular contractures. FEBS J 2023; 290:4877-4898. [PMID: 37462535 PMCID: PMC10592371 DOI: 10.1111/febs.16908] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 06/05/2023] [Accepted: 07/17/2023] [Indexed: 07/26/2023]
Abstract
Neonatal brachial plexus injury (NBPI), a leading cause of pediatric upper limb paralysis, results in disabling and incurable muscle contractures that are driven by impaired longitudinal growth of denervated muscles. A rare form of NBPI, which maintains both afferent and sympathetic muscle innervation despite motor denervation, protects against contractures. We have previously ruled out a role for NRG/ErbB signaling, the predominant pathway governing antegrade afferent neuromuscular transmission, in modulating the formation of contractures. Our current study therefore investigated the contributions of sympathetic innervation of skeletal muscle in modulating NBPI-induced contractures. Through chemical sympathectomy and pharmacologic modification with a β2 -adrenergic agonist, we discovered that sympathetic innervation alone is neither required nor sufficient to modulate contracture formation in neonatal mice. Despite this, sympathetic innervation plays an intriguing sex-specific role in mediating neonatal muscle growth, as the cross-sectional area (CSA) and volume of normally innervated male muscles were diminished by ablation of sympathetic neurons and increased by β-adrenergic stimulation. Intriguingly, the robust alterations in CSA occurred with minimal changes to normal longitudinal muscle growth as determined by sarcomere length. Instead, β-adrenergic stimulation exacerbated sarcomere overstretch in denervated male muscles, indicating potentially discrete regulation of muscle width and length. Future investigations into the mechanistic underpinnings of these distinct aspects of muscle growth are thus essential for improving clinical outcomes in patients affected by muscle disorders in which both length and width are affected.
Collapse
Affiliation(s)
- Mason T. Runkel
- Department of Health Sciences, Butler University, Indianapolis, IN, USA
| | - Albaraa Tarabishi
- Department of Biochemistry, University of Cincinnati, Cincinnati, OH, USA
| | - Kritton Shay-Winkler
- Division of Orthopaedic Surgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Marianne E. Emmert
- Division of Orthopaedic Surgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Qingnian Goh
- Division of Orthopaedic Surgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Orthopaedic Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Roger Cornwall
- Division of Orthopaedic Surgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Orthopaedic Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
7
|
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
|
8
|
Senior D, Ahmed R, Arnavut E, Carvalho A, Lee WX, Blum K, Komatsu DE, Hadjiargyrou M, Badgaiyan RD, Thanos PK. Behavioral, Neurochemical and Developmental Effects of Chronic Oral Methylphenidate: A Review. J Pers Med 2023; 13:jpm13040574. [PMID: 37108960 PMCID: PMC10144804 DOI: 10.3390/jpm13040574] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/07/2023] [Accepted: 03/13/2023] [Indexed: 04/29/2023] Open
Abstract
The majority of animal studies on methylphenidate (MP) use intraperitoneal (IP) injections, subcutaneous (SC) injections, or the oral gavage route of administration. While all these methods allow for delivery of MP, it is the oral route that is clinically relevant. IP injections commonly deliver an immediate and maximum dose of MP due to their quick absorption. This quick-localized effect can give timely results but will only display a small window of the psychostimulant's effects on the animal model. On the opposite side of the spectrum, a SC injection does not accurately represent the pathophysiology of an oral exposure because the metabolic rate of the drug would be much slower. The oral-gavage method, while providing an oral route, possesses some adverse effects such as potential animal injury and can be stressful to the animal compared to voluntary drinking. It is thus important to allow the animal to have free consumption of MP, and drinking it to more accurately mirror human treatment. The use of a two-bottle drinking method allows for this. Rodents typically have a faster metabolism than humans, which means this needs to be considered when administering MP orally while reaching target pharmacokinetic levels in plasma. With this oral two-bottle approach, the pathophysiological effects of MP on development, behavior, neurochemistry and brain function can be studied. The present review summarizes these effects of oral MP which have important implications in medicine.
Collapse
Affiliation(s)
- Daniela Senior
- Behavioral Neuropharmacology & Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Rania Ahmed
- Behavioral Neuropharmacology & Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Eliz Arnavut
- Behavioral Neuropharmacology & Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Alexandra Carvalho
- Behavioral Neuropharmacology & Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Wen Xuan Lee
- Behavioral Neuropharmacology & Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Kenneth Blum
- Division of Addiction Research & Education, Center for Psychiatry, Medicine & Primary Care (Office of the Provost), Western University Health Sciences, Pomona, CA 91766, USA
| | - David E Komatsu
- Department of Orthopaedics and Rehabilitation, Stony Brook University, New York, NY 11794, USA
| | - Michael Hadjiargyrou
- Department of Biological and Chemical Sciences, New York Institute of Technology, Old Westbury, NY 11568, USA
| | | | - Panayotis K Thanos
- Behavioral Neuropharmacology & Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| |
Collapse
|
9
|
Sifeddine W, Ba-M'hamed S, Landry M, Bennis M. Effect of atomoxetine on ADHD-pain hypersensitization comorbidity in 6-OHDA lesioned mice. Pharmacol Rep 2023; 75:342-357. [PMID: 36787018 DOI: 10.1007/s43440-023-00459-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/26/2023] [Accepted: 01/31/2023] [Indexed: 02/15/2023]
Abstract
BACKGROUND Methylphenidate and atomoxetine are used for the treatment of attention-deficit/hyperactivity disorder (ADHD). Our previous studies established the validity of the 6-hydroxydopamine (6-OHDA) mouse model of ADHD and demonstrated hypersensitivity to pain, in line with clinical reports in ADHD patients. Acute methylphenidate treatment reduces hyperactivity and increases attention, but does not affect pain behaviors in this mouse model. Whereas atomoxetine has been shown to be effective against some symptoms of ADHD, nothing is known about its possible action on comorbid pain hypersensitivity. The objectives of the present research are (1) to investigate the effects of acute and chronic treatment with atomoxetine on ADHD-like symptoms and nociceptive thresholds, and (2) to explore the catecholaminergic systems underlying these effects. METHODS Sham and 6-OHDA cohorts of male mice were tested for hyperactivity (open field), attention and impulsivity (5-choice serial reaction time task test), and thermal (hot plate test) and mechanical (von Frey test) thresholds after acute or repeated treatment with vehicle or atomoxetine (1, 3 or 10 mg/kg). RESULTS Acute administration of atomoxetine (10 mg/kg) reduced the hyperactivity and impulsivity displayed by 6-OHDA mice, without affecting attention or nociception. However, atomoxetine administered at 3 mg/kg/day for 7 days alleviated the ADHD-like core symptoms and attenuated the hyperalgesic responses. Furthermore, hyperlocomotion and anti-hyperalgesic activity were antagonized with phentolamine, propranolol, and sulpiride pre-treatments. CONCLUSION These findings demonstrated that when administered chronically, atomoxetine has a significant effect on ADHD-associated pain hypersensitization, likely mediated by both α- and β-adrenergic and D2/D3 dopaminergic receptors, and suggest new indications for atomoxetine that will need to be confirmed by well-designed clinical trials.
Collapse
Affiliation(s)
- Wahiba Sifeddine
- Laboratory of Pharmacology, Neurobiology, Anthropobiology, and Environment, Faculty of Sciences, Cadi Ayyad University, Avenue Prince My Abdellah, B.P. 2390, 40000, Marrakesh, Morocco
| | - Saadia Ba-M'hamed
- Laboratory of Pharmacology, Neurobiology, Anthropobiology, and Environment, Faculty of Sciences, Cadi Ayyad University, Avenue Prince My Abdellah, B.P. 2390, 40000, Marrakesh, Morocco
| | - Marc Landry
- University of Bordeaux, CNRS, Institute of Neurodegenerative Diseases, UMR 5293, Bordeaux, France.,University of Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, UMS 3420, US 4, Bordeaux, France
| | - Mohamed Bennis
- Laboratory of Pharmacology, Neurobiology, Anthropobiology, and Environment, Faculty of Sciences, Cadi Ayyad University, Avenue Prince My Abdellah, B.P. 2390, 40000, Marrakesh, Morocco.
| |
Collapse
|
10
|
Targeting Neuroinflammation with Abscisic Acid Reduces Pain Sensitivity in Females and Hyperactivity in Males of an ADHD Mice Model. Cells 2023; 12:cells12030465. [PMID: 36766806 PMCID: PMC9914171 DOI: 10.3390/cells12030465] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 01/28/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Attention deficit/hyperactivity disorder (ADHD) is a neurodevelopmental syndrome characterized by dopaminergic dysfunction. In this study, we aimed to demonstrate that there is a link between dopaminergic deficit and neuroinflammation that underlies ADHD symptoms. We used a validated ADHD mice model involving perinatal 6-OHDA lesions. The animals received abscisic acid (ABA), an anti-inflammatory phytohormone, at a concentration of 20 mg/L (drinking water) for one month. We tested a battery of behavior tests, learning and memory, anxiety, social interactions, and pain thresholds in female and male mice (control and lesioned, with or without ABA treatment). Postmortem, we analyzed microglia morphology and Ape1 expression in specific brain areas related to the descending pain inhibitory pathway. In females, the dopaminergic deficit increased pain sensitivity but not hyperactivity. In contrast, males displayed hyperactivity but showed no increased pain sensitivity. In females, pain sensitivity was associated with inflammatory microglia and lower Ape1 levels in the anterior cingulate cortex (ACC) and posterior insula cortex (IC). In addition, ABA treatment alleviated pain sensitivity concomitant with reduced inflammation and normalized APE1. In males, ABA reduced hyperactivity but had no significant effect on inflammation in these areas. This is the first study proving a sex-dependent association between dopamine dysfunction and inflammation in specific brain areas, hence leading to different behavioral outcomes in a mouse model of ADHD. These findings provide new clues for potential treatments for ADHD.
Collapse
|
11
|
Cabana-Domínguez J, Antón-Galindo E, Fernàndez-Castillo N, Singgih EL, O'Leary A, Norton WH, Strekalova T, Schenck A, Reif A, Lesch KP, Slattery D, Cormand B. The translational genetics of ADHD and related phenotypes in model organisms. Neurosci Biobehav Rev 2023; 144:104949. [PMID: 36368527 DOI: 10.1016/j.neubiorev.2022.104949] [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: 07/01/2022] [Revised: 11/02/2022] [Accepted: 11/05/2022] [Indexed: 11/10/2022]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a highly prevalent neurodevelopmental disorder resulting from the interaction between genetic and environmental risk factors. It is well known that ADHD co-occurs frequently with other psychiatric disorders due, in part, to shared genetics factors. Although many studies have contributed to delineate the genetic landscape of psychiatric disorders, their specific molecular underpinnings are still not fully understood. The use of animal models can help us to understand the role of specific genes and environmental stimuli-induced epigenetic modifications in the pathogenesis of ADHD and its comorbidities. The aim of this review is to provide an overview on the functional work performed in rodents, zebrafish and fruit fly and highlight the generated insights into the biology of ADHD, with a special focus on genetics and epigenetics. We also describe the behavioral tests that are available to study ADHD-relevant phenotypes and comorbid traits in these models. Furthermore, we have searched for new models to study ADHD and its comorbidities, which can be useful to test potential pharmacological treatments.
Collapse
Affiliation(s)
- Judit Cabana-Domínguez
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain; Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Catalonia, Spain.
| | - Ester Antón-Galindo
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain; Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Catalonia, Spain
| | - Noèlia Fernàndez-Castillo
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain; Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Catalonia, Spain
| | - Euginia L Singgih
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Aet O'Leary
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany; Division of Neuropsychopharmacology, Department of Psychology, University of Tartu, Tartu, Estonia
| | - William Hg Norton
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Tatyana Strekalova
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany, and Department of Neuropsychology and Psychiatry, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, the Netherlands
| | - Annette Schenck
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany, and Department of Neuropsychology and Psychiatry, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, the Netherlands
| | - David Slattery
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Bru Cormand
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain; Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Catalonia, Spain.
| |
Collapse
|
12
|
Kerekes N, Lundqvist S, Schubert Hjalmarsson E, Torinsson Naluai Å, Kantzer AK, Knez R. The associations between ADHD, pain, inflammation, and quality of life in children and adolescents-a clinical study protocol. PLoS One 2022; 17:e0273653. [PMID: 36083951 PMCID: PMC9462574 DOI: 10.1371/journal.pone.0273653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 08/10/2022] [Indexed: 11/19/2022] Open
Abstract
New research shows that the prevalence of neurodevelopmental disorders, such as attention-deficit/hyperactivity disorder (ADHD), is increased in children and adolescents as well as in adults with chronic pain, compared to those without chronic pain. Children and adolescents with ADHD also have an increased incidence of various physical conditions associated with pain, and they more frequently suffer from inflammatory diseases. Moreover, parents of children with ADHD can often suffer from pain conditions. These epidemiological and clinical observations form the scientific basis of our study, which aims to map the relationships between ADHD, altered pain experiences/central sensitization, and inflammation in children and adolescents. We will investigate the presence of central sensitization in children and adolescents with newly diagnosed ADHD and compare it with those who have not been diagnosed with ADHD. Participants (and their biological parents) will complete surveys about their somatic health, pain experience, and quality of life. Biological samples (saliva and stool) will be collected, aiming to utilize proteome and metabolome data to discover disease mechanisms and to predict, prevent and treat them. The results from our investigation should enable an expanded understanding of the pathophysiology behind both ADHD and pain/central sensitization. Presently, there are no established protocols for addressing psychiatric symptoms when examining patients with pain conditions in a somatic care setting, nor is there any knowledge of offering patients with ADHD or other neurodevelopmental disorders adapted treatments for pain conditions. Our results, therefore, can contribute to the development of new treatment strategies for pathological pain conditions in children and adolescents with ADHD. They may also increase awareness about and provide opportunities for the treatment of attention and impulse control problems in children and adolescents with pain syndromes.
Collapse
Affiliation(s)
- Nóra Kerekes
- Department of Health Sciences, University West, Trollhättan, Sweden
| | - Sara Lundqvist
- Child and Adolescent Psychiatry, Queen Silvia Children’s Hospital, Gothenburg, Sweden
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Elke Schubert Hjalmarsson
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Physiotherapy, Queen Silvia Children’s Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Åsa Torinsson Naluai
- Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | | | - Rajna Knez
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Pediatrics, Skaraborg Hospital, Skövde, Sweden
| |
Collapse
|
13
|
Fox SN, McMeekin LJ, Savage CH, Joyce KL, Boas SM, Simmons MS, Farmer CB, Ryan J, Pereboeva L, Becker K, Auwerx J, Sudarshan S, Ma J, Lee A, Roberts RC, Crossman DK, Kralli A, Cowell RM. Estrogen-related receptor gamma regulates mitochondrial and synaptic genes and modulates vulnerability to synucleinopathy. NPJ Parkinsons Dis 2022; 8:106. [PMID: 35982091 PMCID: PMC9388660 DOI: 10.1038/s41531-022-00369-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 07/12/2022] [Indexed: 11/17/2022] Open
Abstract
Many studies implicate mitochondrial dysfunction as a key contributor to cell loss in Parkinson disease (PD). Previous analyses of dopaminergic (DAergic) neurons from patients with Lewy-body pathology revealed a deficiency in nuclear-encoded genes for mitochondrial respiration, many of which are targets for the transcription factor estrogen-related receptor gamma (Esrrg/ERRγ). We demonstrate that deletion of ERRγ from DAergic neurons in adult mice was sufficient to cause a levodopa-responsive PD-like phenotype with reductions in mitochondrial gene expression and number, that partial deficiency of ERRγ hastens synuclein-mediated toxicity, and that ERRγ overexpression reduces inclusion load and delays synuclein-mediated cell loss. While ERRγ deletion did not fully recapitulate the transcriptional alterations observed in postmortem tissue, it caused reductions in genes involved in synaptic and mitochondrial function and autophagy. Altogether, these experiments suggest that ERRγ-deficient mice could provide a model for understanding the regulation of transcription in DAergic neurons and that amplifying ERRγ-mediated transcriptional programs should be considered as a strategy to promote DAergic maintenance in PD.
Collapse
Affiliation(s)
- S N Fox
- Neuroscience Department, Drug Discovery Division, Southern Research, Birmingham, AL, 35205, USA
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - L J McMeekin
- Neuroscience Department, Drug Discovery Division, Southern Research, Birmingham, AL, 35205, USA
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - C H Savage
- Neuroscience Department, Drug Discovery Division, Southern Research, Birmingham, AL, 35205, USA
| | - K L Joyce
- Neuroscience Department, Drug Discovery Division, Southern Research, Birmingham, AL, 35205, USA
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - S M Boas
- Neuroscience Department, Drug Discovery Division, Southern Research, Birmingham, AL, 35205, USA
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - M S Simmons
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - C B Farmer
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - J Ryan
- NeuroInitiative, LLC, Jacksonville, FL, 32207, USA
| | - L Pereboeva
- Department of Pediatrics, Infectious Disease, Neuroscience Vector and Virus Core, University of Alabama at Birmingham, Birmingham, AL, 35223, USA
| | - K Becker
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - J Auwerx
- Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - S Sudarshan
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - J Ma
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - A Lee
- NeuroInitiative, LLC, Jacksonville, FL, 32207, USA
| | - R C Roberts
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - D K Crossman
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - A Kralli
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - R M Cowell
- Neuroscience Department, Drug Discovery Division, Southern Research, Birmingham, AL, 35205, USA.
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
14
|
Pain hypersensitivity in a pharmacological mouse model of attention-deficit/hyperactivity disorder. Proc Natl Acad Sci U S A 2022; 119:e2114094119. [PMID: 35858441 PMCID: PMC9335339 DOI: 10.1073/pnas.2114094119] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Clinical evidence suggests that pain hypersensitivity develops in patients with attention-deficit/hyperactivity disorder (ADHD). However, the mechanisms and neural circuits involved in these interactions remain unknown because of the paucity of studies in animal models. We previously validated a mouse model of ADHD obtained by neonatal 6-hydroxydopamine (6-OHDA) injection. Here, we have demonstrated that 6-OHDA mice exhibit a marked sensitization to thermal and mechanical stimuli, suggesting that phenotypes associated with ADHD include increased nociception. Moreover, sensitization to pathological inflammatory stimulus is amplified in 6-OHDA mice as compared to shams. In this ADHD model, spinal dorsal horn neuron hyperexcitability was observed. Furthermore, ADHD-related hyperactivity and anxiety, but not inattention and impulsivity, are worsened in persistent inflammatory conditions. By combining in vivo electrophysiology, optogenetics, and behavioral analyses, we demonstrated that anterior cingulate cortex (ACC) hyperactivity alters the ACC-posterior insula circuit and triggers changes in spinal networks that underlie nociceptive sensitization. Altogether, our results point to shared mechanisms underlying the comorbidity between ADHD and nociceptive sensitization. This interaction reinforces nociceptive sensitization and hyperactivity, suggesting that overlapping ACC circuits may be targeted to develop better treatments.
Collapse
|
15
|
Schmitz F, Ferreira FS, Silveira JS, V. R. Júnior O, T. S. Wyse A. Effects of methylphenidate after a long period of discontinuation include changes in exploratory behavior and increases brain activities of Na+,K+-ATPase and acetylcholinesterase. Neurobiol Learn Mem 2022; 192:107637. [DOI: 10.1016/j.nlm.2022.107637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/04/2022] [Accepted: 05/13/2022] [Indexed: 11/28/2022]
|
16
|
Contreras D, Piña R, Carvallo C, Godoy F, Ugarte G, Zeise M, Rozas C, Morales B. Methylphenidate Restores Behavioral and Neuroplasticity Impairments in the Prenatal Nicotine Exposure Mouse Model of ADHD: Evidence for Involvement of AMPA Receptor Subunit Composition and Synaptic Spine Morphology in the Hippocampus. Int J Mol Sci 2022; 23:ijms23137099. [PMID: 35806103 PMCID: PMC9266648 DOI: 10.3390/ijms23137099] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/22/2022] [Accepted: 06/22/2022] [Indexed: 02/04/2023] Open
Abstract
In ADHD treatment, methylphenidate (MPH) is the most frequently used medication. The present work provides evidence that MPH restored behavioral impairments and neuroplasticity due to changes in AMPAR subunit composition and distribution, as well as maturation of dendritic spines, in a prenatal nicotine exposure (PNE) ADHD mouse model. PNE animals and controls were given a single oral dose of MPH (1 mg/kg), and their behavior was tested for attention, hyperactivity, and working memory. Long-term potentiation (LTP) was induced and analyzed at the CA3/CA1 synapse in hippocampal slices taken from the same animals tested behaviorally, measuring fEPSPs and whole-cell patch-clamp EPSCs. By applying crosslinking and Western blots, we estimated the LTP effects on AMPAR subunit composition and distribution. The density and types of dendritic spines were quantified by using the Golgi staining method. MPH completely restored the behavioral impairments of PNE mice. Reduced LTP and AMPA-receptor-mediated EPSCs were also restored. EPSC amplitudes were tightly correlated with numbers of GluA1/GluA1 AMPA receptors at the cell surface. Finally, we found a lower density of dendritic spines in hippocampal pyramidal neurons in PNE mice, with a higher fraction of thin-type immature spines and a lower fraction of mushroom mature spines; the latter effect was also reversed by MPH.
Collapse
Affiliation(s)
- Darwin Contreras
- Laboratory of Neuroscience, Faculty of Chemistry and Biology, University of Santiago de Chile, Alameda 3363, Santiago 9170022, Chile; (D.C.); (F.G.); (G.U.)
| | - Ricardo Piña
- Departamento de Biología, Facultad de Ciencias Básicas, Universidad Metropolitana de Ciencias de la Educación, Santiago 7760197, Chile;
- Departamento de Ciencias Pedagógicas, Facultad de Educación, Universidad Bernardo O’Higgins, Santiago 8370993, Chile
| | - Claudia Carvallo
- Centro de investigación e innovación en Gerontología Aplicada (CIGAP), Facultad de Salud, Universidad Santo Tomás, Santiago 8370003, Chile;
| | - Felipe Godoy
- Laboratory of Neuroscience, Faculty of Chemistry and Biology, University of Santiago de Chile, Alameda 3363, Santiago 9170022, Chile; (D.C.); (F.G.); (G.U.)
| | - Gonzalo Ugarte
- Laboratory of Neuroscience, Faculty of Chemistry and Biology, University of Santiago de Chile, Alameda 3363, Santiago 9170022, Chile; (D.C.); (F.G.); (G.U.)
| | - Marc Zeise
- School of Psychology, Faculty of Humanities, University of Santiago de Chile, Santiago 9170022, Chile;
| | - Carlos Rozas
- Laboratory of Neuroscience, Faculty of Chemistry and Biology, University of Santiago de Chile, Alameda 3363, Santiago 9170022, Chile; (D.C.); (F.G.); (G.U.)
- Correspondence: (C.R.); (B.M.)
| | - Bernardo Morales
- Laboratory of Neuroscience, Faculty of Chemistry and Biology, University of Santiago de Chile, Alameda 3363, Santiago 9170022, Chile; (D.C.); (F.G.); (G.U.)
- Correspondence: (C.R.); (B.M.)
| |
Collapse
|
17
|
Herrera-Morales WV, Ramírez-Lugo L, Cauich-Kumul R, Murillo-Rodríguez E, Núñez-Jaramillo L. Personalization of pharmacological treatments for ADHD: Why it is advisable and possible options to achieve it. Curr Top Med Chem 2022; 22:1236-1249. [DOI: 10.2174/1568026622666220509155413] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 02/08/2022] [Accepted: 02/18/2022] [Indexed: 11/22/2022]
Abstract
Abstract:
Attention-deficit hyperactivity disorder is a neurodevelopmental disorder diagnosed primarily in children, although it is also present in adults. Patients present inattention, impulsivity, and hyperactivity symptoms that create difficulties in their daily lives. Pharmacological treatment with stimulants or non-stimulants is used most commonly to reduce ADHD symptoms. Although generally effective and safe, pharmacological treatments have different effects among patients, including lack of response and adverse reactions. The reasons for these differences are not fully understood, but they may derive from the highly diverse etiology of ADHD. Strategies to guide optimal pharmacological treatment selection on the basis of individual patients’ physiological markers are being developed. In this review, we describe the main pharmacological ADHD treatments used and their main drawbacks. We present alternatives under study that would allow the customization of pharmacological treatments to overcome these drawbacks and achieve more reliable improvement of ADHD symptoms.
Collapse
Affiliation(s)
- Wendy Verónica Herrera-Morales
- Departamento de Ciencias Médicas. División de Ciencias de la Salud. Universidad de Quintana Roo. Chetumal, Quintana Roo. México
| | - Leticia Ramírez-Lugo
- Instituto de Fisiología Celular. Universidad Nacional Autónoma de México. Ciudad de México. México
| | - Roger Cauich-Kumul
- Departamento de Ciencias Farmaceúticas. División de Ciencias de la Salud. Universidad de Quintana Roo. Chetumal, Quintana Roo. México
| | - Eric Murillo-Rodríguez
- Laboratorio de Neurociencias Moleculares e Integrativas. Escuela de Medicina, División Ciencias de la Salud, Universidad Anáhuac Mayab Mérida, México
- Intercontinental Neuroscience Research Group, Mérida, Yucatán, México
| | - Luis Núñez-Jaramillo
- Departamento de Ciencias Médicas. División de Ciencias de la Salud. Universidad de Quintana Roo. Chetumal, Quintana Roo. México
| |
Collapse
|
18
|
Mu S, Lin Y, Xu Y, Wei X, Zeng Z, Lin K, Zhu L, Liu Q, Qi X, Wei L, Liang S, Wang S. A novel rat model for cerebral venous sinus thrombosis: verification of similarity to human disease via clinical analysis and experimental validation. J Transl Med 2022; 20:174. [PMID: 35410343 PMCID: PMC8996223 DOI: 10.1186/s12967-022-03374-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 04/01/2022] [Indexed: 11/10/2022] Open
Abstract
Background Cerebral venous sinus thrombosis (CVST) is a rare neurovascular disorder with highly variable manifestations and clinical courses. Animal models properly matched to the clinical form of CVST are necessary for elucidating the pathophysiology of the disease. In this study, we aimed to establish a rat model that accurately recapitulates the clinical features of CVST in human patients. Methods This study consisted of a clinical analysis and animal experiments. Clinical data for two centres obtained between January 2016 and May 2021 were collected and analysed retrospectively. In addition, a Sprague–Dawley rat model of CVST was established by inserting a water-swellable rubber device into the superior sagittal sinus, following which imaging, histological, haematological, and behavioural tests were used to investigate pathophysiological changes. Principal component analysis and hierarchical clustering heatmaps were used to evaluate the similarity between the animal models and human patients. Results The imaging results revealed the possibility of vasogenic oedema in animal models. Haematological analysis indicated an inflammatory and hypercoagulable state. These findings were mostly matched with the retrospective clinical data. Pathological and serological tests further revealed brain parenchymal damage related to CVST in animal models. Conclusions We successfully established a stable and reproducible rat model of CVST. The high similarity between clinical patients and animal models was verified via cluster analysis. This model may be useful for the study of CVST pathophysiology and potential therapies. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03374-y.
Collapse
|
19
|
Kantak KM. Rodent models of attention-deficit hyperactivity disorder: An updated framework for model validation and therapeutic drug discovery. Pharmacol Biochem Behav 2022; 216:173378. [DOI: 10.1016/j.pbb.2022.173378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/22/2022] [Accepted: 03/28/2022] [Indexed: 01/21/2023]
|
20
|
Kenton JA, Young JW. Preclinical Evaluation of Attention and Impulsivity Relevant to Determining ADHD Mechanisms and Treatments. Curr Top Behav Neurosci 2022; 57:291-320. [PMID: 35606639 DOI: 10.1007/7854_2022_340] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
People with Attention-Deficit Hyperactivity Disorder (ADHD) exhibit inattention, hyperactivity, and/or impulsivity. Symptoms of ADHD emerge in childhood and can continue throughout adulthood. Clinical assessments to diagnose ADHD can include administration of continuous performance tests (CPTs). CPTs provide an objective measure of inattention, requiring individuals to respond to targets (attention), and inhibit response to non-targets (impulsivity). When investigating the mechanisms of, and novel treatments for, ADHD it is important to measure such behavioral domains (attention and impulsivity). Some well-established preclinical tasks purport to assess attention in rodents but, unlike CPTs, do not require non-target inhibition, limiting their ADHD-relevance.Recently developed tasks recreate CPTs for rodents. The 5-Choice CPT (5C-CPT) contains non-target stimuli, enabling use of signal detection theory to evaluate performance, consistent with CPTs. The 5C-CPT has been adapted for use in humans, enabling direct cross-species comparisons of performance. A newer task, the rodent CPT (rCPT), is a touchscreen-based analog of CPTs, utilizing symbols instead of a simple stimulus array. Currently, the rCPT may be more akin to a go/no-go task, equally presenting targets/non-targets, although numerous variants exist - a strength. The 5C-CPT and rCPT emulate human CPTs and provide the most up-to-date information on ADHD-relevant studies for understanding attention/impulsivity.
Collapse
Affiliation(s)
- Johnny A Kenton
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - 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.
| |
Collapse
|
21
|
Stanford SC. Animal Models of ADHD? Curr Top Behav Neurosci 2022; 57:363-393. [PMID: 35604570 DOI: 10.1007/7854_2022_342] [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] [Indexed: 06/15/2023]
Abstract
To describe animals that express abnormal behaviors as a model of Attention-Deficit Hyperactivity Disorder (ADHD) implies that the abnormalities are analogous to those expressed by ADHD patients. The diagnostic features of ADHD comprise inattentiveness, impulsivity, and hyperactivity and so these behaviors are fundamental for validation of any animal model of this disorder. Several experimental interventions such as neurotoxic lesion of neonatal rats with 6-hydroxydopamine (6-OHDA), genetic alterations, or selective inbreeding of rodents have produced animals that express each of these impairments to some extent. This article appraises the validity of claims that these procedures have produced a model of ADHD, which is essential if they are to be used to investigate the underlying cause(s) of ADHD and its abnormal neurobiology.
Collapse
Affiliation(s)
- S Clare Stanford
- Department of Neuroscience Physiology and Pharmacology, University College London, London, UK.
| |
Collapse
|
22
|
Usui N, Tian X, Harigai W, Togawa S, Utsunomiya R, Doi T, Miyoshi K, Shinoda K, Tanaka J, Shimada S, Katayama T, Yoshimura T. Length impairments of the axon initial segment in rodent models of attention-deficit hyperactivity disorder and autism spectrum disorder. Neurochem Int 2021; 153:105273. [PMID: 34971749 DOI: 10.1016/j.neuint.2021.105273] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 11/25/2022]
Abstract
The axon initial segment (AIS) is a structural neuronal compartment of the proximal axon that plays key roles in sodium channel clustering, action potential initiation, and signal propagation of neuronal outputs. Mutations in constitutive genes of the AIS, such as ANK3, have been identified in patients with neurodevelopmental disorders. Nevertheless, morphological changes in the AIS in neurodevelopmental disorders have not been characterized. In this study, we investigated the length of the AIS in rodent models of attention-deficit hyperactivity disorder (ADHD) and autism spectrum disorder (ASD). We observed abnormalities in AIS length in both animal models. In ADHD model rodents, we observed shorter AIS length in layer 2/3 (L2/3) neurons of the medial prefrontal cortex (mPFC) and primary somatosensory barrel field (S1BF). Further, we observed shorter AIS length in S1BF L5 neurons. In ASD model mice, we observed shorter AIS length in L2/3 and L5 neurons of the S1BF. These results suggest that impairments in AIS length are common phenomena in neurodevelopmental disorders such as ADHD and ASD and may be conserved across species. Our findings provide novel insight into the potential contribution of the AIS to the pathophysiology and pathogenesis of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Noriyoshi Usui
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan; United Graduate School of Child Development, Osaka University, Suita, 565-0871, Japan; Global Center for Medical Engineering and Informatics, Osaka University, Suita, 565-0871, Japan; Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Hirakata, 573-0022, Japan.
| | - Xiaoye Tian
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, Suita, 565-0871, Japan
| | - Wakana Harigai
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, Suita, 565-0871, Japan
| | - Shogo Togawa
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan; Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Yamaguchi, 755-8505, Japan
| | - Ryo Utsunomiya
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Ehime, 791-0295, Japan
| | - Tomomi Doi
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Ehime, 791-0295, Japan
| | - Ko Miyoshi
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, Suita, 565-0871, Japan
| | - Koh Shinoda
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Yamaguchi, 755-8505, Japan
| | - Junya Tanaka
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Ehime, 791-0295, Japan
| | - Shoichi Shimada
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan; United Graduate School of Child Development, Osaka University, Suita, 565-0871, Japan; Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Hirakata, 573-0022, Japan
| | - Taiichi Katayama
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, Suita, 565-0871, Japan
| | - Takeshi Yoshimura
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, Suita, 565-0871, Japan.
| |
Collapse
|
23
|
Effects of neonatal dopaminergic lesion on oral cocaine self-administration in rats: Higher female vulnerability to cocaine consumption. Pharmacol Biochem Behav 2021; 212:173315. [PMID: 34942237 DOI: 10.1016/j.pbb.2021.173315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 12/12/2021] [Accepted: 12/13/2021] [Indexed: 11/22/2022]
Abstract
The dopaminergic system is associated with cocaine-seeking behaviors, being influenced by other neurotransmitters such as GABA and deregulated by chronic cocaine self-administration. Administration of 6-hydroxydopamine (6-OHDA) to neonatal rats produces a depletion of brain dopamine, mainly, that results in behavioral alterations in adulthood. This model can be applied to better understanding of the role of the dopaminergic system in cocaine use and how its behavioral effects can modulate drug intake. Though there are well-established sex differences in the pattern of drug use, there are no published studies investigating sex-dependent effects of neonatal lesions with 6-OHDA on cocaine self-administration nor regarding GABAA receptor (GABAAR) subunits expression. Herein, neurotoxic lesion was induced in male and female neonatal rats by intracisternal injection of 6-OHDA at PND 4, and locomotion was evaluated before and after cocaine self-administration. Cocaine was diluted in a sweet solution (sucrose 1.5%) and offered for 27 consecutive 3-h daily sessions via a dispenser for oral intake, in an operant chamber under a fixed-ratio 1 (FR1) schedule. The 6-OHDA lesion reduced oral cocaine self-administration in male and female rats. Female rats, independent of dopaminergic condition, consumed more cocaine-containing solution than sucrose-only solution. Furthermore, as expected, 6-OHDA-lesioned animals presented a higher basal locomotor activity when compared to sham rats. We evaluated GABAAR subunit expression and found no statistically significant differences between rats that self-administered a sucrose-only solution and those that self-administered a cocaine-containing solution. Even when the reward system is depleted, some behavioral differences remain in females, providing more data that highlight the female vulnerability to cocaine consumption.
Collapse
|
24
|
Kerekes N, Sanchéz-Pérez AM, Landry M. Neuroinflammation as a possible link between attention-deficit/hyperactivity disorder (ADHD) and pain. Med Hypotheses 2021; 157:110717. [PMID: 34717072 DOI: 10.1016/j.mehy.2021.110717] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 09/01/2021] [Accepted: 10/17/2021] [Indexed: 12/17/2022]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) and pathological pain are two complex syndromes of multifactorial origin. Despite their prevalence and broad impacts, these conditions are seldom recognized and managed simultaneously. The co-existence of neuropsychiatric conditions (such as ADHD) and altered pain perception and chronic pain has been noted in children, and the comorbidity of ADHD and chronic pain is well documented in adults. Pathophysiological studies have suggested dysfunction of the dopaminergic system as a common neurochemical basis for comorbid ADHD and pain. Considerable evidence supports the role of neuroinflammation in the pathophysiology of both. We suggest that central neuroinflammation underlies altered pain perception and pain sensitization in persons with ADHD. Based on our hypothesis, targeting neuroinflammation may serve as a potential new therapeutic intervention to treat ADHD and comorbid pain in children and adolescents and a preventive strategy for the development of chronic pain in adults with ADHD.
Collapse
Affiliation(s)
- Nóra Kerekes
- Department of Health Sciences, University West, Trollhättan 461 86, Sweden.
| | - Ana Maria Sanchéz-Pérez
- Neurobiotechnology Laboratory, Faculty of Health Sciences, Institute of Advanced Materials (INAM), University Jaume I, Castellon 120 71, Spain
| | - Marc Landry
- University of Bordeaux, CNRS, Institute for Neurodegenrative Diseases, IMN, UMR 5293, F-33000 Bordeaux, France
| |
Collapse
|
25
|
McLaurin KA, Harris M, Madormo V, Harrod SB, Mactutus CF, Booze RM. HIV-Associated Apathy/Depression and Neurocognitive Impairments Reflect Persistent Dopamine Deficits. Cells 2021; 10:2158. [PMID: 34440928 PMCID: PMC8392364 DOI: 10.3390/cells10082158] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/10/2021] [Accepted: 08/18/2021] [Indexed: 12/12/2022] Open
Abstract
Individuals living with human immunodeficiency virus type 1 (HIV-1) are often plagued by debilitating neurocognitive impairments and affective alterations;the pathophysiology underlying these deficits likely includes dopaminergic system dysfunction. The present review utilized four interrelated aims to critically examine the evidence for dopaminergic alterations following HIV-1 viral protein exposure. First, basal dopamine (DA) values are dependent upon both brain region andexperimental approach (i.e., high-performance liquid chromatography, microdialysis or fast-scan cyclic voltammetry). Second, neurochemical measurements overwhelmingly support decreased DA concentrations following chronic HIV-1 viral protein exposure. Neurocognitive impairments, including alterations in pre-attentive processes and attention, as well as apathetic behaviors, provide an additional line of evidence for dopaminergic deficits in HIV-1. Third, to date, there is no compelling evidence that combination antiretroviral therapy (cART), the primary treatment regimen for HIV-1 seropositive individuals, has any direct pharmacological action on the dopaminergic system. Fourth, the infection of microglia by HIV-1 viral proteins may mechanistically underlie the dopamine deficit observed following chronic HIV-1 viral protein exposure. An inclusive and critical evaluation of the literature, therefore, supports the fundamental conclusion that long-term HIV-1 viral protein exposure leads to a decreased dopaminergic state, which continues to persist despite the advent of cART. Thus, effective treatment of HIV-1-associated apathy/depression and neurocognitive impairments must focus on strategies for rectifying decreases in dopamine function.
Collapse
Affiliation(s)
| | | | | | | | | | - Rosemarie M. Booze
- Department of Psychology, University of South Carolina, Columbia, SC 29208, USA; (K.A.M.); (M.H.); (V.M.); (S.B.H.); (C.F.M.)
| |
Collapse
|
26
|
Fan HC, Chang YK, Tsai JD, Chiang KL, Shih JH, Yeh KY, Ma KH, Li IH. The Association Between Parkinson's Disease and Attention-Deficit Hyperactivity Disorder. Cell Transplant 2021; 29:963689720947416. [PMID: 33028106 PMCID: PMC7784516 DOI: 10.1177/0963689720947416] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
While Parkinson’s disease (PD) and attention-deficit hyperactivity disorder (ADHD) are two distinct conditions, it has been hypothesized that they share several overlapping anatomical and neurochemical changes. In order to investigate that hypothesis, this study used claims data from Taiwan’s Longitudinal Health Insurance Database 2000 to provide the significant nationwide population-based evidence of an increased risk of PD among ADHD patients, and the connection between the two conditions was not the result of other comorbidities. Moreover, this study showed that the patients with PD were 2.8 times more likely to have a prior ADHD diagnosis compared with those without a prior history of ADHD. Furthermore, an animal model of ADHD was generated by neonatally injecting rats with 6-hydroxydopamine (6-OHDA). These rats were subjected to behavior tests and the 99mTc-TRODAT-1 brain imaging at the juvenile stage. Compared to control group rats, the 6-OHDA rats showed a significantly reduced specific uptake ratio in the striatum, indicating an underlying PD-linked pathology in the brains of these ADHD phenotype-expressing rats. Overall, these results support that ADHD shares a number of anatomical and neurochemical changes with PD. As such, improved knowledge of the neurochemical mechanisms underlying ADHD could result in improved treatments for various debilitating neurological disorders, including PD.
Collapse
Affiliation(s)
- Hueng-Chuen Fan
- Department of Pediatrics, 59084Tungs' Taichung Metroharbor Hospital, Wuchi, Taichung.,Department of Medical research, 68866Tungs' Taichung Metroharbor Hospital, Wuchi, Taichung.,Department of Life Sciences, 59084National Chung Hsing University, Taichung.,Department of Rehabilitation, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli
| | - Yu-Kang Chang
- Department of Medical research, 68866Tungs' Taichung Metroharbor Hospital, Wuchi, Taichung.,Department of Life Sciences, 59084National Chung Hsing University, Taichung.,Department of Rehabilitation, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli
| | - Jeng-Dau Tsai
- School of Medicine, 34899Chung Shan Medical University, Taichung.,Department of Pediatrics, 34899Chung Shan Medical University Hospital, Taichung
| | - Kuo-Liang Chiang
- Department of Pediatric Neurology, 38009Kuang-Tien General Hospital, Taichung.,Department of Nutrition, Hungkuang University, Taichung
| | - Jui-Hu Shih
- Department of Pharmacy Practice, 63452Tri-Service General Hospital, Taipei.,School of Pharmacy, 71548National Defense Medical Center, Taipei
| | - Kuan-Yi Yeh
- Department of Biology and Anatomy, 71548National Defense Medical Center, Taipei
| | - Kuo-Hsing Ma
- Department of Biology and Anatomy, 71548National Defense Medical Center, Taipei
| | - I-Hsun Li
- Department of Pharmacy Practice, 63452Tri-Service General Hospital, Taipei.,School of Pharmacy, 71548National Defense Medical Center, Taipei
| |
Collapse
|
27
|
Rahi V, Kumar P. Animal models of attention-deficit hyperactivity disorder (ADHD). Int J Dev Neurosci 2021; 81:107-124. [PMID: 33428802 DOI: 10.1002/jdn.10089] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 12/14/2020] [Accepted: 01/06/2021] [Indexed: 12/15/2022] Open
Abstract
Attention-deficit hyperactivity disorder (ADHD) is a heterogeneous neuropsychiatric disorder characterized by three primary symptoms hyperactivity, attention deficit, and impulsiveness, observed in both children and adults. In childhood, this disorder is more common in boys than in girls, and at least 75% will continue to suffer from the disorder until adulthood. Individuals with ADHD generally have poor academic, occupational, and social functioning resulting from developmentally inappropriate levels of hyperactivity and impulsivity, as well as impaired ability to maintain attention on motivationally relevant tasks. Very few drugs available in clinical practice altogether abolish the symptoms of ADHD, therefore, to find new drugs and target it is essential to understand the neuropathological, neurochemical, and genetic alterations that lead to the progression of ADHD. With this contrast, an animal study is the best approach because animal models provide relatively fast invasive manipulation, rigorous hypothesis testing, as well as it provides a better angle to understand the pathological mechanisms involved in disease progression. Moreover, animal models, especially for ADHD, serve with good predictive validity would allow the assessment and development of new therapeutic interventions, with this aim, the present review collect the various animal models on a single platform so that the research can select an appropriate model to pursue his study.
Collapse
Affiliation(s)
- Vikrant Rahi
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, India
| | - Puneet Kumar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, India
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| |
Collapse
|
28
|
Rinehart L, Spencer S. Which came first: Cannabis use or deficits in impulse control? Prog Neuropsychopharmacol Biol Psychiatry 2021; 106:110066. [PMID: 32795592 PMCID: PMC7750254 DOI: 10.1016/j.pnpbp.2020.110066] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/12/2020] [Accepted: 08/05/2020] [Indexed: 12/16/2022]
Abstract
Impulse control deficits are often found to co-occur with substance use disorders (SUDs). On the one hand, it is well known that chronic intake of drugs of abuse remodels the brain with significant consequences for a range of cognitive behaviors. On the other hand, individual variation in impulse control may contribute to differences in susceptibility to SUDs. Both of these relationships have been described, thus leading to a "chicken or the egg" debate which remains to be fully resolved. Does impulsivity precede drug use or does it manifest as a function of problematic drug usage? The link between impulsivity and SUDs has been most strongly established for cocaine and alcohol use disorders using both preclinical models and clinical data. Much less is known about the potential link between impulsivity and cannabis use disorder (CUD) or the directionality of this relationship. The initiation of cannabis use occurs most often during adolescence prior to the brain's maturation, which is recognized as a critical period of development. The long-term effects of chronic cannabis use on the brain and behavior have started to be explored. In this review we will summarize these observations, especially as they pertain to the relationship between impulsivity and CUD, from both a psychological and biological perspective. We will discuss impulsivity as a multi-dimensional construct and attempt to reconcile the results obtained across modalities. Finally, we will discuss possible avenues for future research with emerging longitudinal data.
Collapse
Affiliation(s)
- Linda Rinehart
- University of Minnesota, Department of Psychiatry and Behavioral Sciences
| | - Sade Spencer
- University of Minnesota, Department of Pharmacology, Minneapolis, MN, USA.
| |
Collapse
|
29
|
Pantic I, Cumic J, Skodric SR, Dugalic S, Brodski C. Oxidopamine and oxidative stress: Recent advances in experimental physiology and pharmacology. Chem Biol Interact 2021; 336:109380. [PMID: 33450287 DOI: 10.1016/j.cbi.2021.109380] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 01/01/2021] [Accepted: 01/10/2021] [Indexed: 12/20/2022]
Abstract
Oxidopamine (6-hydroxydopamine, 6-OHDA) is a toxin commonly used for the creation of experimental animal models of Parkinson's disease, attention-deficit hyperactivity disorder, and Lesch-Nyhan syndrome. Its exact mechanism of action is not completely understood, although there are many indications that it is related to the generation of reactive oxygen species (ROS), primarily in dopaminergic neurons. In certain experimental conditions, oxidopamine may also cause programmed cell death via various signaling pathways. Oxidopamine may also have a significant impact on chromatin structure and nuclear structural organization in some cells. Today, many researchers use oxidopamine-associated oxidative damage to evaluate different antioxidant-based pharmacologically active compounds as drug candidates for various neurological and non-neurological diseases. Additional research is needed to clarify the exact biochemical pathways associated with oxidopamine toxicity, related ROS generation and apoptosis. In this short review, we focus on the recent research in experimental physiology and pharmacology, related to the cellular and animal experimental models of oxidopamine - mediated toxicity.
Collapse
Affiliation(s)
- Igor Pantic
- University of Belgrade, Faculty of Medicine, Dr Subotica 8, RS-11129, Belgrade, Serbia; University of Haifa, 199 Abba Hushi Blvd, Mount Carmel, Haifa, IL-3498838, Israel; Institute of medical physiology, Visegradska 26/II, RS-11129, Belgrade, Serbia.
| | - Jelena Cumic
- University of Belgrade, Faculty of Medicine, Dr Subotica 8, RS-11129, Belgrade, Serbia; Clinical Center of Serbia, Dr. KosteTodorovića 8, RS-11129, Belgrade, Serbia
| | | | - Stefan Dugalic
- Clinical Center of Serbia, Dr. KosteTodorovića 8, RS-11129, Belgrade, Serbia
| | - Claude Brodski
- Ben-Gurion University of the Negev, Zlotowski Center for Neuroscience, Faculty of Health Sciences, Department of Physiology and Cell Biology, P.O.B. 653, Beersheba, Israel
| |
Collapse
|
30
|
Cai Y, Xing L, Yang T, Chai R, Wang J, Bao J, Shen W, Ding S, Chen G. The neurodevelopmental role of dopaminergic signaling in neurological disorders. Neurosci Lett 2020; 741:135540. [PMID: 33278505 DOI: 10.1016/j.neulet.2020.135540] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/10/2020] [Accepted: 11/27/2020] [Indexed: 12/11/2022]
Abstract
Dopamine (DA), a critical neurotransmitter of both the central and peripheral nerve system, plays important roles in a series of biological processes. Dysfunction of dopaminergic signalling may lead to a series of developmental disorders, including attention deficit/hyperactivity disorder, autism and schizophrenia. However, the exact roles of dopaminergic signalling in these diseases are far from fully understood. We analyse the roles of dopaminergic signalling in multiple physiological and pathological processes, focusing on brain development and related disorders. By summarizing current research in this area, we provide guidance for future studies. This review seeks to deepen our understanding of dopaminergic signalling in developmental disorders, which may offer clues for developing more effective therapeutic drugs.
Collapse
Affiliation(s)
- Yunyun Cai
- Department of Physiology, School of Medicine, Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Lingyan Xing
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Tuo Yang
- Department of Hand Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, China
| | - Rui Chai
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Jiaqi Wang
- Department of Physiology, School of Medicine, Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Jingyin Bao
- Basic Medical Research Centre, Medical College of Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Weixing Shen
- Department of Physiology, School of Medicine, Nantong University, Nantong, Jiangsu Province, 226001, China.
| | - Sujun Ding
- Department of Ultrasound, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, 226001, China.
| | - Gang Chen
- Department of Tissue and Embryology, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, 226001, China; Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, 226001, China.
| |
Collapse
|
31
|
Exogenous Flupirtine as Potential Treatment for CLN3 Disease. Cells 2020; 9:cells9081872. [PMID: 32796515 PMCID: PMC7464162 DOI: 10.3390/cells9081872] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 02/07/2023] Open
Abstract
CLN3 disease is a fatal neurodegenerative disorder affecting children. Hallmarks include brain atrophy, accelerated neuronal apoptosis, and ceramide elevation. Treatment regimens are supportive, highlighting the importance of novel, disease-modifying drugs. Flupirtine and its new allyl carbamate derivative (compound 6) confer neuroprotective effects in CLN3-deficient cells. This study lays the groundwork for investigating beneficial effects in Cln3Δex7/8 mice. WT/Cln3Δex7/8 mice received flupirtine/compound 6/vehicle for 14 weeks. Short-term effect of flupirtine or compound 6 was tested using a battery of behavioral testing. For flupirtine, gene expression profiles, astrogliosis, and neuronal cell counts were determined. Flupirtine improved neurobehavioral parameters in open field, pole climbing, and Morris water maze tests in Cln3Δex7/8 mice. Several anti-apoptotic markers and ceramide synthesis/degradation enzymes expression was dysregulated in Cln3Δex7/8 mice. Flupirtine reduced astrogliosis in hippocampus and motor cortex of male and female Cln3Δex7/8 mice. Flupirtine increased neuronal cell counts in male mice. The newly synthesized compound 6 showed promising results in open field and pole climbing. In conclusion, flupirtine improved behavioral, neuropathological and biochemical parameters in Cln3Δex7/8 mice, paving the way for potential therapies for CLN3 disease.
Collapse
|
32
|
Areal LB, Blakely RD. Neurobehavioral changes arising from early life dopamine signaling perturbations. Neurochem Int 2020; 137:104747. [PMID: 32325191 PMCID: PMC7261509 DOI: 10.1016/j.neuint.2020.104747] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/11/2020] [Accepted: 04/14/2020] [Indexed: 12/11/2022]
Abstract
Dopamine (DA) signaling is critical to the modulation of multiple brain functions including locomotion, reinforcement, attention and cognition. The literature provides strong evidence that altered DA availability and actions can impact normal neurodevelopment, with both early and enduring consequences on anatomy, physiology and behavior. An appreciation for the developmental contributions of DA signaling to brain development is needed to guide efforts to preclude and remedy neurobehavioral disorders, such as attention-deficit/hyperactivity disorder, addiction, bipolar disorder, schizophrenia and autism spectrum disorder, each of which exhibits links to DA via genetic, cellular and/or pharmacological findings. In this review, we highlight research pursued in preclinical models that use genetic and pharmacological approaches to manipulate DA signaling at sensitive developmental stages, leading to changes at molecular, circuit and/or behavioral levels. We discuss how these alterations can be aligned with traits displayed by neuropsychiatric diseases. Lastly, we review human studies that evaluate contributions of developmental perturbations of DA systems to increased risk for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Lorena B Areal
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Randy D Blakely
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, 33458, USA; Brain Institute, Florida Atlantic University, Jupiter, FL, 33458, USA.
| |
Collapse
|
33
|
Miguel PM, Deniz BF, Confortim HD, de Almeida W, Bronauth LP, Vieira MC, Bertoldi K, Siqueira IR, Silveira PP, Pereira LO. Methylphenidate treatment increases hippocampal BDNF levels but does not improve memory deficits in hypoxic-ischemic rats. J Psychopharmacol 2020; 34:750-758. [PMID: 32255391 DOI: 10.1177/0269881120913153] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Methylphenidate (MPH) is a stimulant drug mainly prescribed to treat cognitive impairments in attention-deficit/hyperactivity disorder (ADHD). We demonstrated that neonatal hypoxia-ischemia (HI) induced attentional deficits in rats and MPH administration reversed these deficits. However, MPH effects on memory deficits after the HI procedure have not been evaluated yet. AIMS We aimed to analyze learning and memory performance of young hypoxic-ischemic rats after MPH administration and associate their performance with brain-derived neurotrophic factor (BDNF) levels in the prefrontal cortex and hippocampus. METHODS Male Wistar rats were divided into four groups (n=11-13/group): control saline (CTS), control MPH (CTMPH), HI saline (HIS) and HIMPH. The HI procedure was conducted at post-natal day (PND) 7 and memory tasks between PND 30 and 45. MPH administration (2.5 mg/kg, i.p.) occurred 30 min prior to each behavioral session and daily, for 15 days, for the BDNF assay (n=5-7/group). RESULTS As expected, hypoxic-ischemic animals demonstrated learning and memory deficits in the novel-object recognition (NOR) and Morris water maze (MWM) tasks. However, MPH treatment did not improve learning and memory deficits of these animals in the MWM-and even disrupted the animals' performance in the NOR task. Increased BDNF levels were found in the hippocampus of HIMPH animals, which seem to have been insufficient to improve memory deficits observed in this group. CONCLUSIONS The MPH treatment was not able to improve memory deficits resulting from the HI procedure considering a dose of 2.5 mg/kg. Further studies investigating different MPH doses would be necessary to determine a dose-response relationship in this model.
Collapse
Affiliation(s)
- Patrícia Maidana Miguel
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Departamento de Ciências Morfológicas, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Bruna Ferrary Deniz
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Departamento de Ciências Morfológicas, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Heloísa Deola Confortim
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Departamento de Ciências Morfológicas, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Wellington de Almeida
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Departamento de Ciências Morfológicas, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Loise Peres Bronauth
- Departamento de Ciências Morfológicas, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Milene Cardoso Vieira
- Departamento de Ciências Morfológicas, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Karine Bertoldi
- Departamento de Farmacologia, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Ionara Rodrigues Siqueira
- Departamento de Farmacologia, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Programa de Pós-Graduação em Ciências Biológicas, Fisiologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Patrícia Pelufo Silveira
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, Canada.,Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Canada.,Sackler Program for Epigenetics & Psychobiology at McGill University, Montreal, Canada
| | - Lenir Orlandi Pereira
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Departamento de Ciências Morfológicas, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
34
|
Bouchatta O, Manouze H, Ba-M'Hamed S, Landry M, Bennis M. Neonatal 6-OHDA Lesion Model in Mouse Induces Cognitive Dysfunctions of Attention-Deficit/Hyperactivity Disorder (ADHD) During Young Age. Front Behav Neurosci 2020; 14:27. [PMID: 32174817 PMCID: PMC7054716 DOI: 10.3389/fnbeh.2020.00027] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 02/05/2020] [Indexed: 11/13/2022] Open
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a syndrome characterized by impaired attention, impulsivity and hyperactivity in children. These symptoms are often maintained in adults. During adolescence, prefrontal cortex develops connectivity with other brain regions to engage executive functions such as, latent inhibition, attention and inhibitory control. In our previous work, we demonstrated the validity of the neonatal 6-Hydroxydopamine (6-OHDA) mouse model, a classical neurodevelopmental model mimicking major symptoms of the human ADHD pathology. In order to evaluate pathological forms of executive functions and impulsive behavior in 6-OHDA mice during young age, we first tested latent inhibition (LI) after weaning, and then we evaluated the impulsive behavior using a cliff avoidance reaction test. Our results demonstrated that 6-OHDA mice showed disruption in latent inhibition, suggesting a deficit in selective attention, and displayed repetitive peering-down behavior, indicating a maladaptive impulsive behavior. Subsequently, to assess impulsivity and attention in young mice, we performed a modified 5-choice serial reaction time task test (5-CSRTT), optimizing the degree of food restriction for young animals and shortening the training duration. This test allowed us to demonstrate a deficit in inhibitory control and a loss of accuracy of 6-OHDA mice in the 5-CSRTT. In conclusion, we demonstrated that the 6-OHDA mouse model reproduces human symptoms of ADHD in childhood and early adulthood periods, as seen in human. Taken together, the 6-OHDA mouse model will be useful alongside other animal models to understand the neurobiological mechanisms underlying complex, heterogeneous neurological disorders.
Collapse
Affiliation(s)
- Otmane Bouchatta
- Laboratory of Pharmacology, Neurobiology and Behavior, Faculty of Sciences, Cadi Ayyad University, Marrakesh, Morocco.,University of Bordeaux, Bordeaux, France.,CNRS UMR 5297, Centre Paul Broca-Nouvelle Aquitaine, Interdisciplinary Institute of Neuroscience, Bordeaux, France
| | - Houria Manouze
- Laboratory of Pharmacology, Neurobiology and Behavior, Faculty of Sciences, Cadi Ayyad University, Marrakesh, Morocco
| | - Saadia Ba-M'Hamed
- Laboratory of Pharmacology, Neurobiology and Behavior, Faculty of Sciences, Cadi Ayyad University, Marrakesh, Morocco
| | - Marc Landry
- University of Bordeaux, Bordeaux, France.,CNRS UMR 5297, Centre Paul Broca-Nouvelle Aquitaine, Interdisciplinary Institute of Neuroscience, Bordeaux, France
| | - Mohamed Bennis
- Laboratory of Pharmacology, Neurobiology and Behavior, Faculty of Sciences, Cadi Ayyad University, Marrakesh, Morocco
| |
Collapse
|
35
|
Intracellular mechanisms and behavioral changes in mouse model of attention deficit hyperactivity disorder: Importance of age-specific NMDA receptor blockade. Pharmacol Biochem Behav 2019; 188:172830. [PMID: 31756355 DOI: 10.1016/j.pbb.2019.172830] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 11/18/2019] [Accepted: 11/18/2019] [Indexed: 11/21/2022]
Abstract
Exposure of NMDA receptor antagonists during developmental stages leads to behavioral consequences like attention deficit hyperactivity disorder (ADHD). However, the underlying molecular mechanisms have remained poorly understood. Herein, we studied the phosphorylated Akt (pAkt) and caspase-3, the key regulators of neuronal cell survival/death, as the probable downstream targets of MK-801 often used to engender ADHD-like condition. Swiss albino mice at postnatal days (PND) 7, 14 or 21 were injected with a single dose of MK-801 and evaluated for hyperactivity (open field test) and memory deficit at adolescence (PND 30) and adult stages (PND 60). PND 7 or 14 treatment groups (but not PND 21) consistently showed hyperactivity at the adolescence stage. A significant increase in working and reference memory errors in radial arm maze was noted at the adolescence age. PND 7 group continued to display the symptoms even in adulthood. All the treatment groups showed a significant decrease in the percent alterations (Y-maze) and discrimination index (novel object recognition test) at adolescence age. A significant increase in caspase-3 expression was noted in the prefrontal cortex (PFC) and hippocampus, whereas increased pAkt was noticed only in the hippocampus, following a single injection of MK-801 at PND 7. Concurrently, PND 7 treatment group showed significantly decreased neuronal nuclei (NeuN) expression (a marker for mature neurons) in the dentate gyrus, cornu ammonis-3 and PFC, but not in cornu ammonis-1, at adolescence age. We suggest that the observed symptoms of ADHD at adolescence and adulthood stages may be linked to alteration in pAkt and caspase-3 followed MK-801 treatment at PND 7.
Collapse
|
36
|
NURR1 deficiency is associated to ADHD-like phenotypes in mice. Transl Psychiatry 2019; 9:207. [PMID: 31455763 PMCID: PMC6712038 DOI: 10.1038/s41398-019-0544-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 06/09/2019] [Accepted: 07/17/2019] [Indexed: 01/10/2023] Open
Abstract
The transcription factor NURR1 regulates the dopamine (DA) signaling pathway and exerts a critical role in the development of midbrain dopaminergic neurons (mDA). NURR1 alterations have been linked to DA-associated brain disorders, such as Parkinson's disease and schizophrenia. However, the association between NURR1 defects and the attention-deficit hyperactivity disorder (ADHD), a DA-associated brain disease characterized by hyperactivity, impulsivity and inattention, has never been demonstrated. To date, a comprehensive murine model of ADHD truly reflecting the whole complex human psychiatric disorder still does not exist. NURR1-knockout (NURR1-KO) mice have been reported to exhibit increased spontaneous locomotor activity, but their complete characterization is still lacking. In the present study a wide-ranging test battery was used to perform a comprehensive analysis of the behavioral phenotype of the male NURR1-KO mice. As a result, their hyperactive phenotype was confirmed, while their impulsive behavior was reported for the first time. On the other hand, no anxiety and alterations in motor coordination, sociability and memory were observed. Also, the number of mDA expressing tyrosine hydroxylase, a rate-limiting enzyme of catecholamines biosynthesis, and DA level in brain were not impaired in NURR1-KO mice. Finally, hyperactivity has been shown to be recovered by treatment with methylphenidate, the first line psychostimulant drug used for ADHD. Overall, our study suggests that the NURR1 deficient male mouse may be a satisfactory model to study some ADHD behavioral phenotypes and to test the clinical efficacy of potential therapeutic agents.
Collapse
|
37
|
Schmitz J, Kumsta R, Moser D, Güntürkün O, Ocklenburg S. DNA methylation of dopamine-related gene promoters is associated with line bisection deviation in healthy adults. Sci Rep 2019; 9:5902. [PMID: 30976054 PMCID: PMC6459813 DOI: 10.1038/s41598-019-42553-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 04/03/2019] [Indexed: 11/09/2022] Open
Abstract
Handedness and language lateralization are the most investigated phenotypes among functional hemispheric asymmetries, i.e. differences in function between the left and the right half of the human brain. Both phenotypes are left hemisphere-dominant, while investigations of the molecular factors underlying right hemisphere-dominant phenotypes are less prominent. In the classical line bisection task, healthy subjects typically show a leftward attentional bias due to a relative dominance of the right hemisphere for visuospatial attention. Based on findings of variations in dopamine-related genes affecting performance in the line bisection task, we first tested whether DNA methylation in non-neuronal tissue in the promoter regions of DBH, SLC6A3, and DRD2 are associated with line bisection deviation. We replicated the typical behavioral pattern and found an effect of DNA methylation in the DBH promoter region on line bisection deviation in right-aligned trials. A second exploratory analysis indicated that an overall DNA methylation profile of genes involved in dopamine function predicts line bisection performance in right-aligned trials. Genetic variation in dopamine-related genes has been linked to attention deficit hyperactivity disorder (ADHD), a neurodevelopmental trait associated with rightward attentional bias. Overall, our findings point towards epigenetic markers for functional hemispheric asymmetries in non-neuronal tissue not only for left hemisphere-dominant, but also for right hemisphere-dominant phenotypes.
Collapse
Affiliation(s)
- Judith Schmitz
- Biopsychology, Institute of Cognitive Neuroscience, Department of Psychology, Ruhr University, Bochum, Germany.
| | - Robert Kumsta
- Genetic Psychology, Department of Psychology, Ruhr University, Bochum, Germany
| | - Dirk Moser
- Genetic Psychology, Department of Psychology, Ruhr University, Bochum, Germany
| | - Onur Güntürkün
- Biopsychology, Institute of Cognitive Neuroscience, Department of Psychology, Ruhr University, Bochum, Germany
| | - Sebastian Ocklenburg
- Biopsychology, Institute of Cognitive Neuroscience, Department of Psychology, Ruhr University, Bochum, Germany
| |
Collapse
|
38
|
Bailey RA, Gutierrez A, Kyser TL, Hemmerle AM, Hufgard JR, Seroogy KB, Vorhees CV, Williams MT. Effects of Preweaning Manganese in Combination with Adult Striatal Dopamine Lesions on Monoamines, BDNF, TrkB, and Cognitive Function in Sprague-Dawley Rats. Neurotox Res 2019; 35:606-620. [PMID: 30612279 DOI: 10.1007/s12640-018-9992-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 11/19/2018] [Accepted: 12/18/2018] [Indexed: 01/25/2023]
Abstract
Manganese (Mn) is an essential nutrient especially during development, but Mn overexposure (MnOE) produces long-term cognitive deficits. Evidence of long-term changes in dopamine in the neostriatum was found in rats from developmental MnOE previously. To examine the relationship between MnOE and dopamine, we tested whether the effects of developmental MnOE would be exaggerated by dopamine reductions induced by 6-hydroxydopamine (6-OHDA) neostriatal infusion when the rats were adults. The experiment consisted of four groups of females and males: Vehicle/Sham, MnOE/Sham, Vehicle/6-OHDA, and MnOE/6-OHDA. Both MnOE/Sham and Vehicle/6-OHDA groups displayed egocentric and allocentric memory deficits, whereas MnOE+6-OHDA had additive effects on spatial memory in the Morris water maze and egocentric learning in the Cincinnati water maze. 6-OHDA reduced dopamine in the neostriatum and nucleus accumbens, reduced norepinephrine in the hippocampus, reduced TH+ cells and TrkB and TH expression in the substantia nigra pars compacta (SNpc), but increased TrkB in the neostriatum. MnOE alone had no effect on monoamines or TrkB in the neostriatum or hippocampus but reduced BDNF in the hippocampus. A number of sex differences were noted; however, only a few significant interactions were found for MnOE and/or 6-OHDA exposure. These data further implicate dopamine and BDNF in the cognitive deficits arising from developmental MnOE.
Collapse
Affiliation(s)
- Rebecca A Bailey
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
- Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Arnold Gutierrez
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
- Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Tara L Kyser
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
- Department of Neurology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Ann M Hemmerle
- Department of Neurology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Jillian R Hufgard
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
- Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA
| | - Kim B Seroogy
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
- Department of Neurology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Charles V Vorhees
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
- Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Michael T Williams
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA.
- Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA.
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| |
Collapse
|