1
|
Prates‐Rodrigues M, Schweizer BLA, de Paula Gomes C, Ribeiro ÂM, Padovan‐Neto FE, Masini D, Lopes‐Aguiar C. Challenges and Opportunities in Exploring Non-Motor Symptoms in 6-Hydroxydopamine Models of Parkinson's Disease: A Systematic Review. J Neurochem 2025; 169:e70008. [PMID: 39901598 PMCID: PMC11791392 DOI: 10.1111/jnc.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/18/2024] [Accepted: 01/08/2025] [Indexed: 02/05/2025]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the progressive loss of midbrain dopaminergic neurons, leading to motor symptoms such as tremors, rigidity, and bradykinesia. Non-motor symptoms, including depression, hyposmia, and sleep disturbances, often emerge in the early stages of PD, but their mechanisms remain poorly understood. The 6-hydroxydopamine (6-OHDA) rodent model is a well-established tool for preclinical research, replicating key motor and non-motor symptoms of PD. In this review, we systematically analyzed 135 studies that used 6-OHDA rodent models of PD to investigate non-motor symptoms. The review process adhered to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Our analysis highlights the growing use of 6-OHDA PD models for experimental research of non-motor symptoms. It also reveals significant variability in methodologies, including choices of brain target, toxin dosage, lesion verification strategies, and behavioral assessment reporting. Factors that hinder reproducibility and comparability of findings across studies. We highlight the need for standardization in 6-OHDA-based models with particular emphasis on consistent evaluation of lesion extent and reporting of the co-occurrence of non-motor symptoms. By fostering methodological coherence, this framework aims to enhance the reproducibility, reliability, and translational value of 6-OHDA models in PD non-motor symptom research.
Collapse
Affiliation(s)
- Mateus Prates‐Rodrigues
- Departamento de Fisiologia e Biofísica, Laboratório de Neurociências Comportamental e Molecular (LANEC)Universidade Federal de Minas GeraisBelo HorizonteBrazil
| | - Beatriz Lage Araújo Schweizer
- Departamento de Fisiologia e Biofísica, Laboratório de Neurociências Comportamental e Molecular (LANEC)Universidade Federal de Minas GeraisBelo HorizonteBrazil
| | - Clara de Paula Gomes
- Departamento de Fisiologia e Biofísica, Laboratório de Neurociências Comportamental e Molecular (LANEC)Universidade Federal de Minas GeraisBelo HorizonteBrazil
| | - Ângela Maria Ribeiro
- Departamento de Fisiologia e Biofísica, Laboratório de Neurociências Comportamental e Molecular (LANEC)Universidade Federal de Minas GeraisBelo HorizonteBrazil
| | - Fernando E. Padovan‐Neto
- Department of Psychology, Faculty of Philosophy, Sciences and Letters of Ribeirão PretoUniversity of São PauloRibeirão PretoSPBrazil
| | - Debora Masini
- Department of Biochemistry and BiophysicsStockholm UniversityStockholmSweden
| | - Cleiton Lopes‐Aguiar
- Departamento de Fisiologia e Biofísica, Laboratório de Neurociências Comportamental e Molecular (LANEC)Universidade Federal de Minas GeraisBelo HorizonteBrazil
| |
Collapse
|
2
|
Statz M, Weber H, Weis F, Kober M, Bathel H, Plocksties F, van Rienen U, Timmermann D, Storch A, Fauser M. Subthalamic nucleus deep brain stimulation induces functional deficits in norepinephrinergic neurotransmission in a Parkinson's disease model. Brain Res 2024; 1841:149128. [PMID: 39053685 DOI: 10.1016/j.brainres.2024.149128] [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: 03/17/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Deep brain stimulation of the subthalamic nucleus (STN-DBS) is a successful treatment option in Parkinson's disease (PD) for different motor and non-motor symptoms, but has been linked to postoperative cognitive impairment. AIM Since both dopaminergic and norepinephrinergic neurotransmissions play important roles in symptom development, we analysed STN-DBS effects on dopamine and norepinephrine availability in different brain regions and morphological alterations of catecholaminergic neurons in the 6-hydroxydopamine PD rat model. METHODS We applied one week of continuous unilateral STN-DBS or sham stimulation, respectively, in groups of healthy and 6-hydroxydopamine-lesioned rats to quantify dopamine and norepinephrine contents in the striatum, olfactory bulb and dentate gyrus. In addition, we analysed dopaminergic cell counts in the substantia nigra pars compacta and area tegmentalis ventralis and norepinephrinergic neurons in the locus coeruleus after one and six weeks of STN-DBS. RESULTS In 6-hydroxydopamine-lesioned animals, one week of STN-DBS did not alter dopamine levels, while striatal norepinephrine levels were decreased. However, neither one nor six weeks of STN-DBS altered dopaminergic neuron numbers in the midbrain or norepinephrinergic neuron counts in the locus coeruleus. Dopaminergic fibre density in the dorsal and ventral striatum also remained unchanged after six weeks of STN-DBS. In healthy animals, one week of STN-DBS resulted in increased dopamine levels in the olfactory bulb and decreased contents in the dentate gyrus, but had no effects on norepinephrine availability. CONCLUSIONS STN-DBS modulates striatal norepinephrinergic neurotransmission in a PD rat model. Additional behavioural studies are required to investigate the functional impact of this finding.
Collapse
Affiliation(s)
- Meike Statz
- Department of Neurology, University of Rostock, Gehlsheimer Straße 20, 18147 Rostock, Germany
| | - Hanna Weber
- Department of Neurology, University of Rostock, Gehlsheimer Straße 20, 18147 Rostock, Germany
| | - Frederike Weis
- Department of Neurology, University of Rostock, Gehlsheimer Straße 20, 18147 Rostock, Germany
| | - Maria Kober
- Department of Neurology, University of Rostock, Gehlsheimer Straße 20, 18147 Rostock, Germany
| | - Henning Bathel
- Institute of General Electrical Engineering, University of Rostock, 18059 Rostock, Germany
| | - Franz Plocksties
- Institute of Applied Microelectronics and Computer Engineering, University of Rostock, 18059 Rostock, Germany
| | - Ursula van Rienen
- Institute of General Electrical Engineering, University of Rostock, 18059 Rostock, Germany; Department Life, Light and Matter, University of Rostock, 18059 Rostock, Germany; Department of Ageing of Individuals and Society, Interdisciplinary Faculty, University of Rostock, 18059 Rostock, Germany
| | - Dirk Timmermann
- Institute of Applied Microelectronics and Computer Engineering, University of Rostock, 18059 Rostock, Germany
| | - Alexander Storch
- Department of Neurology, University of Rostock, Gehlsheimer Straße 20, 18147 Rostock, Germany; German Centre for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, Gehlsheimer Straße 20, 18147 Rostock, Germany
| | - Mareike Fauser
- Department of Neurology, University of Rostock, Gehlsheimer Straße 20, 18147 Rostock, Germany.
| |
Collapse
|
3
|
Wang Z, Huang PE, Wang N, Zhang Q, Kang J, Fang Y, Ning B, Li L. β-asarone inhibits autophagy by activating the PI3K/Akt/mTOR pathway in a rat model of depression in Parkinson's disease. Behav Brain Res 2024; 465:114966. [PMID: 38518853 DOI: 10.1016/j.bbr.2024.114966] [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: 02/08/2024] [Revised: 03/19/2024] [Accepted: 03/19/2024] [Indexed: 03/24/2024]
Abstract
OBJECTIVE It is unclear whether β-asarone has a good antidepressant effect and what is the main mechanism in Depression in Parkinson's disease (DPD) model rats. METHODS In this study, DPD model rats were screened from 6-OHDA induced rats by sucrose preference test (SPT) and forced swimming test (FST). DPD model rats were divided into eight groups: model group, pramipexole group, β-asarone low-dose group (β-asarone 7.5 group), β-asarone medium-dose group (β-asarone 15 group), β-asarone high-dose group (β-asarone 30 group), 3-MA group, rapamycin group, and PI3K inhibitor group. 28 days after the end of treatment, open field test (OFT), SPT and FST were conducted in rats. The level of α-synuclein (α-syn) in the striatum was determined by enzyme-linked immunosorbent assay (ELISA). The expression of Beclin-1, p62 in the striatum was determined by western blot. The expression of PI3K, p-PI3K, Akt, p-Akt, mTOR, p-mTOR, Beclin-1, and p62 in the hippocampus was determined by western blot. The spine density of neurons in the hippocampus was detected by golgi staining. RESULTS The results showed that 4-week oral administration of β-asarone improve the motor and depressive symptoms of DPD model rats, and decrease the content of α-syn in the striatum. β-asarone inhibited the expression of autophagy in the striatum of DPD model rats. Furthermore, β-asarone decreased the levels of Beclin-1 protein, increased the expression of p62, p-PI3K, p-AKT, and p-mTOR, and improved the density of neuron dendritic spine in the hippocampus. CONCLUSIONS We concluded that β-asarone might improve the behavior of DPD model rats by activating the PI3K/Akt/mTOR pathway, inhibiting autophagy and protecting neuron.
Collapse
Affiliation(s)
- Zhifang Wang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ping-E Huang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Nanbu Wang
- Guangzhou University of Chinese Medicine, Guangzhou, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | | | - Jian Kang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongqi Fang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Baile Ning
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Ling Li
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
4
|
Liu F, Huang S, Guo D, Li X, Han Y. Deep brain stimulation of ventromedial prefrontal cortex reverses depressive-like behaviors via BDNF/TrkB signaling pathway in rats. Life Sci 2023; 334:122222. [PMID: 38084673 DOI: 10.1016/j.lfs.2023.122222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/18/2023] [Accepted: 10/26/2023] [Indexed: 12/18/2023]
Abstract
AIM Deep brain stimulation (DBS) is currently under investigation as a potential therapeutic approach for managing major depressive disorder (MDD) and ventromedial prefrontal cortex (vmPFC) is recognized as a promising target region. Therefore, the present study aimed to investigate a preclinical paradigm of bilateral vmPFC DBS and examine the molecular mechanisms underlying its antidepressant-like effects using chronic unpredictable stress (CUS) model in rats. MAIN METHODS Male rats were subjected to stereotaxic surgery and deep brain stimulation paradigm in non-stressed and CUS rats respectively, and the therapeutic effect of DBS were assessed by a series of behavioral tests including sucrose preference test, open field test, elevated plus maze test, and forced swim test. The potential involvement of the BDNF/TrkB signaling pathway and its downstream effects in this process were also investigated using western blot. KEY FINDINGS We identified that a stimulation protocol consisting of 130 Hz, 200 μA, 90 μs pulses administered for 5 h per day over a period of 7 days effectively mitigated CUS-induced depressive-like and anxiety-like behaviors in rats. These therapeutic effects were associated with the enhancement of the BDNF/TrkB signaling pathway and its downstream ERK1/2 activity. SIGNIFICANCE These findings provide valuable insights into the potential clinical utility of vmPFC DBS as an approach of improving the symptoms experienced by individuals with MDD. This evidence contributes to our understanding of the neurobiological basis of depression and offers promise for the development of more effective treatments.
Collapse
Affiliation(s)
- Fanglin Liu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China; Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Shihao Huang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China; Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Dan Guo
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China; Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Xin Li
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China
| | - Ying Han
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China.
| |
Collapse
|
5
|
Grembecka B, Majkutewicz I, Harackiewicz O, Wrona D. Deep-Brain Subthalamic Nucleus Stimulation Enhances Food-Related Motivation by Influencing Neuroinflammation and Anxiety Levels in a Rat Model of Early-Stage Parkinson's Disease. Int J Mol Sci 2023; 24:16916. [PMID: 38069238 PMCID: PMC10706602 DOI: 10.3390/ijms242316916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
Deep-brain subthalamic nucleus stimulation (DBS-STN) has become a well-established therapeutic option for advanced Parkinson's disease (PD). While the motor benefits of DBS-STN are widely acknowledged, the neuropsychiatric effects are still being investigated. Beyond its immediate effects on neuronal circuits, emerging research suggests that DBS-STN might also modulate the peripheral inflammation and neuroinflammation. In this work, we assessed the effects of DBS-STN on food-related motivation, food intake pattern, and the level of anxiety and compared them with markers of cellular and immune activation in nigrostriatal and mesolimbic areas in rats with the 6-OHDA model of early PD. To evaluate the potential mechanism of observed effects, we also measured corticosterone concentration in plasma and leukocyte distribution in peripheral blood. We found that DBS-STN applied during neurodegeneration has beneficial effects on food intake pattern and motivation and reduces anxiety. These behavioral effects occur with reduced percentages of IL-6-labeled cells in the ventral tegmental area and substantia nigra pars compacta in the stimulated brain hemisphere. At the same brain structures, the cFos cell activations were confirmed. Simultaneously, the corticosterone plasma concentration was elevated, and the peripheral blood lymphocytes were reduced after DBS-STN. We believe that comprehending the relationship between the effects of DBS-STN on inflammation and its therapeutic results is essential for optimizing DBS therapy in PD.
Collapse
Affiliation(s)
- Beata Grembecka
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland; (I.M.); (O.H.); (D.W.)
| | | | | | | |
Collapse
|
6
|
Diao Y, Hu T, Xie H, Fan H, Meng F, Yang A, Bai Y, Zhang J. Premature drug reduction after subthalamic nucleus deep brain stimulation leading to worse depression in patients with Parkinson's disease. Front Neurol 2023; 14:1270746. [PMID: 37928164 PMCID: PMC10620523 DOI: 10.3389/fneur.2023.1270746] [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: 08/07/2023] [Accepted: 09/26/2023] [Indexed: 11/07/2023] Open
Abstract
Background Reduction of medication in Parkinson's disease (PD) following subthalamic nucleus deep brain stimulation (STN-DBS) has been recognized, but the optimal timing for medication adjustments remains unclear, posing challenges in postoperative patient management. Objective This study aimed to provide evidence for the timing of medication reduction post-DBS using propensity score matching (PSM). Methods In this study, initial programming and observation sessions were conducted over 1 week for patients 4-6 weeks postoperatively. Patients were subsequently categorized into medication reduction or non-reduction groups based on their dyskinesia evaluation using the 4.2-item score from the MDS-UPDRS-IV. PSM was employed to maintain baseline comparability. Short-term motor and neuropsychiatric symptom assessments for both groups were conducted 3-6 months postoperatively. Results A total of 123 PD patients were included. Baseline balance in motor and non-motor scores was achieved between the two groups based on PSM. Short-term efficacy revealed a significant reduction in depression scores within the non-reduction group compared to baseline (P < 0.001) and a significant reduction compared to the reduction group (P = 0.037). No significant differences were observed in UPDRS-III and HAMA scores between the two groups. Within-group analysis showed improvements in motor symptoms, depression, anxiety, and subdomains in the non-reduction group, while the reduction group exhibited improvements only in motor symptoms. Conclusion This study provides evidence for the timing of medication reduction following DBS. Our findings suggest that early maintenance of medication stability is more favorable for improving neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Yu Diao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tianqi Hu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hutao Xie
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Houyou Fan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fangang Meng
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Anchao Yang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yutong Bai
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jianguo Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
Campos ACP, Pople C, Silk E, Surendrakumar S, Rabelo TK, Meng Y, Gouveia FV, Lipsman N, Giacobbe P, Hamani C. Neurochemical mechanisms of deep brain stimulation for depression in animal models. Eur Neuropsychopharmacol 2023; 68:11-26. [PMID: 36640729 DOI: 10.1016/j.euroneuro.2022.12.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 01/13/2023]
Abstract
Deep brain stimulation (DBS) has emerged as a neuromodulation therapy for treatment-resistant depression, but its actual efficacy and mechanisms of action are still unclear. Changes in neurochemical transmission are important mechanisms of antidepressant therapies. Here, we review the preclinical DBS literature reporting behavioural and neurochemical data associated with its antidepressant-like effects. The most commonly studied target in preclinical models was the ventromedial prefrontal cortex (vmPFC). In rodents, DBS delivered to this target induced serotonin (5-HT) release and increased 5-HT1B receptor expression. The antidepressant-like effects of vmPFC DBS seemed to be independent of the serotonin transporter and potentially mediated by the direct modulation of prefrontal projections to the raphe. Adenosinergic and glutamatergic transmission might have also play a role. Medial forebrain bundle (MFB) DBS increased dopamine levels and reduced D2 receptor expression, whereas nucleus accumbens (NAcc), and lateral habenula (LHb) stimulation increased catecholamine levels in different brain regions. In rodents, subthalamic nucleus (STN) DBS induced robust depression-like responses associated with a reduction in serotonergic transmission, as revealed by a decrease in serotonin release. Some of these effects seemed to be mediated by 5HT1A receptors. In conclusion, the antidepressant-like effects of DBS in preclinical models have been well documented in multiple targets. Though variable mechanisms have been proposed, DBS-induced acute and long-term changes in neurochemical substrates seem to play an important role in the antidepressant-like effects of this therapy.
Collapse
Affiliation(s)
- Ana Carolina P Campos
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - Christopher Pople
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - Esther Silk
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - Shanan Surendrakumar
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - Thallita K Rabelo
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - Ying Meng
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - Flavia Venetucci Gouveia
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - Nir Lipsman
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; Hurvitz Brain Sciences Centre, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; Division of Neurosurgery, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Peter Giacobbe
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; Hurvitz Brain Sciences Centre, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; Neuropsychiatry Program, Department of Psychiatry, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Clement Hamani
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; Hurvitz Brain Sciences Centre, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; Division of Neurosurgery, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada.
| |
Collapse
|
8
|
Alosaimi F, Boonstra JT, Tan S, Temel Y, Jahanshahi A. The role of neurotransmitter systems in mediating deep brain stimulation effects in Parkinson’s disease. Front Neurosci 2022; 16:998932. [PMID: 36278000 PMCID: PMC9579467 DOI: 10.3389/fnins.2022.998932] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 09/14/2022] [Indexed: 11/24/2022] Open
Abstract
Deep brain stimulation (DBS) is among the most successful paradigms in both translational and reverse translational neuroscience. DBS has developed into a standard treatment for movement disorders such as Parkinson’s disease (PD) in recent decades, however, specific mechanisms behind DBS’s efficacy and side effects remain unrevealed. Several hypotheses have been proposed, including neuronal firing rate and pattern theories that emphasize the impact of DBS on local circuitry but detail distant electrophysiological readouts to a lesser extent. Furthermore, ample preclinical and clinical evidence indicates that DBS influences neurotransmitter dynamics in PD, particularly the effects of subthalamic nucleus (STN) DBS on striatal dopaminergic and glutamatergic systems; pallidum DBS on striatal dopaminergic and GABAergic systems; pedunculopontine nucleus DBS on cholinergic systems; and STN-DBS on locus coeruleus (LC) noradrenergic system. DBS has additionally been associated with mood-related side effects within brainstem serotoninergic systems in response to STN-DBS. Still, addressing the mechanisms of DBS on neurotransmitters’ dynamics is commonly overlooked due to its practical difficulties in monitoring real-time changes in remote areas. Given that electrical stimulation alters neurotransmitter release in local and remote regions, it eventually exhibits changes in specific neuronal functions. Consequently, such changes lead to further modulation, synthesis, and release of neurotransmitters. This narrative review discusses the main neurotransmitter dynamics in PD and their role in mediating DBS effects from preclinical and clinical data.
Collapse
Affiliation(s)
- Faisal Alosaimi
- Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, Netherlands
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Rabigh, Saudi Arabia
- *Correspondence: Faisal Alosaimi,
| | - Jackson Tyler Boonstra
- Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Sonny Tan
- Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Yasin Temel
- Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Ali Jahanshahi
- Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, Netherlands
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, Netherlands
- Ali Jahanshahi,
| |
Collapse
|
9
|
Canonica T, Zalachoras I. Motivational disturbances in rodent models of neuropsychiatric disorders. Front Behav Neurosci 2022; 16:940672. [PMID: 36051635 PMCID: PMC9426724 DOI: 10.3389/fnbeh.2022.940672] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
Motivated behavior is integral to the survival of individuals, continuously directing actions toward rewards or away from punishments. The orchestration of motivated behavior depends on interactions among different brain circuits, primarily within the dopaminergic system, that subserve the analysis of factors such as the effort necessary for obtaining the reward and the desirability of the reward. Impairments in motivated behavior accompany a wide range of neuropsychiatric disorders, decreasing the patients’ quality of life. Despite its importance, motivation is often overlooked as a parameter in neuropsychiatric disorders. Here, we review motivational impairments in rodent models of schizophrenia, depression, and Parkinson’s disease, focusing on studies investigating effort-related behavior in operant conditioning tasks and on pharmacological interventions targeting the dopaminergic system. Similar motivational disturbances accompany these conditions, suggesting that treatments aimed at ameliorating motivation levels may be beneficial for various neuropsychiatric disorders.
Collapse
|
10
|
Santana-Gómez CE, Pérez-Pérez D, Fonseca-Barriendos D, Arias-Carrión O, Besio W, Rocha L. Transcranial Focal Electrical Stimulation Modifies Biogenic Amines' Alterations Induced by 6-Hydroxydopamine in Rat Brain. Pharmaceuticals (Basel) 2021; 14:ph14080706. [PMID: 34451804 PMCID: PMC8401891 DOI: 10.3390/ph14080706] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 07/02/2021] [Accepted: 07/19/2021] [Indexed: 01/12/2023] Open
Abstract
Transcranial focal stimulation (TFS) is a non-invasive neuromodulation strategy with neuroprotective effects. On the other hand, 6-hidroxidopamine (6-OHDA) induces neurodegeneration of the nigrostriatal system producing modifications in the dopaminergic, serotoninergic, and histaminergic systems. The present study was conducted to test whether repetitive application of TFS avoids the biogenic amines' changes induced by the intrastriatal injection of 6-OHDA. Experiments were designed to determine the tissue content of dopamine, serotonin, and histamine in the brain of animals injected with 6-OHDA and then receiving daily TFS for 21 days. Tissue content of biogenic amines was evaluated in the cerebral cortex, hippocampus, amygdala, and striatum, ipsi- and contralateral to the side of 6-OHDA injection. Results obtained were compared to animals with 6-OHDA, TFS alone, and a Sham group. The present study revealed that TFS did not avoid the changes in the tissue content of dopamine in striatum. However, TFS was able to avoid several of the changes induced by 6-OHDA in the tissue content of dopamine, serotonin, and histamine in the different brain areas evaluated. Interestingly, TFS alone did not induce significant changes in the different brain areas evaluated. The present study showed that repetitive TFS avoids the biogenic amines' changes induced by 6-OHDA. TFS can represent a new therapeutic strategy to avoid the neurotoxicity induced by 6-OHDA.
Collapse
Affiliation(s)
| | - Daniel Pérez-Pérez
- Plan of Combined Studies in Medicine (PECEM), Faculty of Medicine, UNAM, México City 04510, Mexico;
| | | | - Oscar Arias-Carrión
- Unidad de Trastornos del Movimiento y Sueño, Hospital General Dr. Manuel Gea González, Secretaria de Salud, Mexico City 14080, Mexico;
| | - Walter Besio
- Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, RI 02881, USA
- Correspondence: (W.B.); (L.R.)
| | - Luisa Rocha
- Pharmacobiology Department, Center for Research and Advanced Studies, México City 14330, Mexico;
- Correspondence: (W.B.); (L.R.)
| |
Collapse
|
11
|
Angelopoulou E, Paudel YN, Bougea A, Piperi C. Impact of the apelin/APJ axis in the pathogenesis of Parkinson's disease with therapeutic potential. J Neurosci Res 2021; 99:2117-2133. [PMID: 34115895 DOI: 10.1002/jnr.24895] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/07/2021] [Accepted: 05/12/2021] [Indexed: 12/18/2022]
Abstract
The pathogenesis of Parkinson's disease (PD) remains elusive. There is still no available disease-modifying strategy against PD, whose management is mainly symptomatic. A growing amount of preclinical evidence shows that a complex interplay between autophagy dysregulation, mitochondrial impairment, endoplasmic reticulum stress, oxidative stress, and excessive neuroinflammation underlies PD pathogenesis. Identifying key molecules linking these pathological cellular processes may substantially aid in our deeper understanding of PD pathophysiology and the development of novel effective therapeutic approaches. Emerging preclinical evidence indicates that apelin, an endogenous neuropeptide acting as a ligand of the orphan G protein-coupled receptor APJ, may play a key neuroprotective role in PD pathogenesis, via inhibition of apoptosis and dopaminergic neuronal loss, autophagy enhancement, antioxidant effects, endoplasmic reticulum stress suppression, as well as prevention of synaptic dysregulation in the striatum, excessive neuroinflammation, and glutamate-induced excitotoxicity. Underlying signaling pathways involve phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin, extracellular signal-regulated kinase 1/2, and inositol requiring kinase 1α/XBP1/C/EBP homologous protein. Herein, we discuss the role of apelin/APJ axis and associated molecular mechanisms on the pathogenesis of PD in vitro and in vivo and provide evidence for its challenging therapeutic potential.
Collapse
Affiliation(s)
- Efthalia Angelopoulou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece.,Department of Neurology, Eginition University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Yam Nath Paudel
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Anastasia Bougea
- Department of Neurology, Eginition University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
12
|
Bouali-Benazzouz R, Landry M, Benazzouz A, Fossat P. Neuropathic pain modeling: Focus on synaptic and ion channel mechanisms. Prog Neurobiol 2021; 201:102030. [PMID: 33711402 DOI: 10.1016/j.pneurobio.2021.102030] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 02/22/2021] [Indexed: 12/28/2022]
Abstract
Animal models of pain consist of modeling a pain-like state and measuring the consequent behavior. The first animal models of neuropathic pain (NP) were developed in rodents with a total lesion of the sciatic nerve. Later, other models targeting central or peripheral branches of nerves were developed to identify novel mechanisms that contribute to persistent pain conditions in NP. Objective assessment of pain in these different animal models represents a significant challenge for pre-clinical research. Multiple behavioral approaches are used to investigate and to validate pain phenotypes including withdrawal reflex to evoked stimuli, vocalizations, spontaneous pain, but also emotional and affective behaviors. Furthermore, animal models were very useful in investigating the mechanisms of NP. This review will focus on a detailed description of rodent models of NP and provide an overview of the assessment of the sensory and emotional components of pain. A detailed inventory will be made to examine spinal mechanisms involved in NP-induced hyperexcitability and underlying the current pharmacological approaches used in clinics with the possibility to present new avenues for future treatment. The success of pre-clinical studies in this area of research depends on the choice of the relevant model and the appropriate test based on the objectives of the study.
Collapse
Affiliation(s)
- Rabia Bouali-Benazzouz
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.
| | - Marc Landry
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Abdelhamid Benazzouz
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Pascal Fossat
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| |
Collapse
|
13
|
Stefani A, Cerroni R, Pierantozzi M, D’Angelo V, Grandi L, Spanetta M, Galati S. Deep brain stimulation in Parkinson’s disease patients and routine 6‐OHDA rodent models: Synergies and pitfalls. Eur J Neurosci 2020; 53:2322-2343. [DOI: 10.1111/ejn.14950] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 08/09/2020] [Accepted: 08/12/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Alessandro Stefani
- Department of System Medicine Faculty of Medicine and Surgery University of Rome “Tor Vergata” Rome Italy
| | - Rocco Cerroni
- Department of System Medicine Faculty of Medicine and Surgery University of Rome “Tor Vergata” Rome Italy
| | - Mariangela Pierantozzi
- Department of System Medicine Faculty of Medicine and Surgery University of Rome “Tor Vergata” Rome Italy
| | - Vincenza D’Angelo
- Department of System Medicine Faculty of Medicine and Surgery University of Rome “Tor Vergata” Rome Italy
| | - Laura Grandi
- Center for Movement Disorders Neurocenter of Southern Switzerland Lugano Switzerland
| | - Matteo Spanetta
- Department of System Medicine Faculty of Medicine and Surgery University of Rome “Tor Vergata” Rome Italy
| | - Salvatore Galati
- Center for Movement Disorders Neurocenter of Southern Switzerland Lugano Switzerland
- Faculty of Biomedical Sciences Università della Svizzera Italiana Lugano Switzerland
| |
Collapse
|
14
|
Effects of Subthalamic Nucleus Deep Brain Stimulation on Facial Emotion Recognition in Parkinson's Disease: A Critical Literature Review. Behav Neurol 2020; 2020:4329297. [PMID: 32724481 PMCID: PMC7382738 DOI: 10.1155/2020/4329297] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 06/12/2020] [Indexed: 01/04/2023] Open
Abstract
Deep brain stimulation (DBS) of the subthalamic nucleus (STN) is an effective therapy for Parkinson's disease (PD). Nevertheless, DBS has been associated with certain nonmotor, neuropsychiatric effects such as worsening of emotion recognition from facial expressions. In order to investigate facial emotion recognition (FER) after STN DBS, we conducted a literature search of the electronic databases MEDLINE and Web of science. In this review, we analyze studies assessing FER after STN DBS in PD patients and summarize the current knowledge of the effects of STN DBS on FER. The majority of studies, which had clinical and methodological heterogeneity, showed that FER is worsening after STN DBS in PD patients, particularly for negative emotions (sadness, fear, anger, and tendency for disgust). FER worsening after STN DBS can be attributed to the functional role of the STN in limbic circuits and the interference of STN stimulation with neural networks involved in FER, including the connections of the STN with the limbic part of the basal ganglia and pre- and frontal areas. These outcomes improve our understanding of the role of the STN in the integration of motor, cognitive, and emotional aspects of behaviour in the growing field of affective neuroscience. Further studies using standardized neuropsychological measures of FER assessment and including larger cohorts are needed, in order to draw definite conclusions about the effect of STN DBS on emotional recognition and its impact on patients' quality of life.
Collapse
|
15
|
Gamma oryzanol impairs alcohol-induced anxiety-like behavior in mice via upregulation of central monoamines associated with Bdnf and Il-1β signaling. Sci Rep 2020; 10:10677. [PMID: 32606350 PMCID: PMC7326911 DOI: 10.1038/s41598-020-67689-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 06/10/2020] [Indexed: 02/06/2023] Open
Abstract
Adolescent alcohol exposure may increase anxiety-like behaviors by altering central monoaminergic functions and other important neuronal pathways. The present study was designed to investigate the anxiolytic effect of 0.5% γ-oryzanol (GORZ) and its neurochemical and molecular mechanisms under chronic 10% ethanol consumption. Five-week-old ICR male mice received either control (14% casein, AIN 93 M) or GORZ (14% casein, AIN 93 M + 0.5% GORZ) diets in this study. We showed that GORZ could potentially attenuate alcohol-induced anxiety-like behaviors by significantly improving the main behavioral parameters measured by the elevated plus maze test. Moreover, GORZ treatment significantly restored the alcohol-induced downregulation of 5-hydroxytryptophan and 5-hydroxyindole acetic acid in the hippocampus and improved homovanillic acid levels in the cerebral cortex. Furthermore, a recovery increase in the level of 3-methoxy-4-hydroxyphenylglycol both in the hippocampus and cerebral cortex supported the anxiolytic effect of GORZ. The significant elevation and reduction in the hippocampus of relative mRNA levels of brain-derived neurotrophic factor and interleukin 1β, respectively, also showed the neuroprotective role of GORZ in ethanol-induced anxiety. Altogether, these results suggest that 0.5% GORZ is a promising neuroprotective drug candidate with potential anxiolytic, neurogenic, and anti-neuroinflammatory properties for treating adolescent alcohol exposure.
Collapse
|
16
|
In vivo validation of a new portable stimulator for chronic deep brain stimulation in freely moving rats. J Neurosci Methods 2020; 333:108577. [PMID: 31899208 DOI: 10.1016/j.jneumeth.2019.108577] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 12/18/2019] [Accepted: 12/30/2019] [Indexed: 11/23/2022]
Abstract
BACKGROUND Deep brain stimulation (DBS) of the subthalamic nucleus (STN) is considered as a gold standard therapy for the alleviation of motor symptoms in Parkinson's disease (PD). This success paved the way to its application for other neurological and psychiatric disorders. In this context, we aimed to develop a rodent-specific stimulator with characteristics similar to those used in patients. NEW METHOD We designed a stimulator that can be connected to an electrode container with options for bilateral or unilateral stimulation selection and offers a wide range of frequencies, pulse widths and intensities, constant current, biphasic current-control and charge balancing. Dedicated software was developed to program these parameters and the device was tested on a bilateral 6-hydroxydopamine (6-OHDA) rat model of PD. RESULTS The equipment was well tolerated by the animals with a good general welfare. STN stimulation (130 Hz frequency, 0.06 ms pulse width, 150 μA average intensity) improved the motor deficits induced by 6-OHDA as it significantly increased the number of movements compared to the values obtained in the same animals without STN stimulation. Furthermore, it restored motor coordination by significantly increasing the time spent on the rotarod bar. CONCLUSION We successfully developed and validated a new portable and programmable stimulator for freely moving rats that delivers a large range of stimulation parameters using bilateral biphasic current-control and charge balancing to maximize tissue safety. This device can be used to test deep brain stimulation in different animal models of human brain diseases.
Collapse
|
17
|
Dujardin K, Sgambato V. Neuropsychiatric Disorders in Parkinson's Disease: What Do We Know About the Role of Dopaminergic and Non-dopaminergic Systems? Front Neurosci 2020; 14:25. [PMID: 32063833 PMCID: PMC7000525 DOI: 10.3389/fnins.2020.00025] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 01/10/2020] [Indexed: 12/13/2022] Open
Abstract
Besides the hallmark motor symptoms (rest tremor, hypokinesia, rigidity, and postural instability), patients with Parkinson’s disease (PD) have non-motor symptoms, namely neuropsychiatric disorders. They are frequent and may influence the other symptoms of the disease. They have also a negative impact on the quality of life of patients and their caregivers. In this article, we will describe the clinical manifestations of the main PD-related behavioral disorders (depression, anxiety disorders, apathy, psychosis, and impulse control disorders). We will also provide an overview of the clinical and preclinical literature regarding the underlying mechanisms with a focus on the role of the dopaminergic and non-dopaminergic systems.
Collapse
Affiliation(s)
- Kathy Dujardin
- Inserm U1171 Degenerative and Vascular Cognitive Disorders, Lille University Medical Center, Lille, France
| | - Véronique Sgambato
- CNRS, Institut des Sciences Cognitives Marc Jeannerod, UMR 5229, Lyon University, Bron, France
| |
Collapse
|
18
|
Spay C, Albares M, Lio G, Thobois S, Broussolle E, Lau B, Ballanger B, Boulinguez P. Clonidine modulates the activity of the subthalamic-supplementary motor loop: evidence from a pharmacological study combining deep brain stimulation and electroencephalography recordings in Parkinsonian patients. J Neurochem 2019; 146:333-347. [PMID: 29675956 DOI: 10.1111/jnc.14447] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 03/15/2018] [Accepted: 04/04/2018] [Indexed: 12/24/2022]
Abstract
Clonidine is an anti-hypertensive medication which acts as an alpha-adrenergic receptor agonist. As the noradrenergic system is likely to support cognitive functions including attention and executive control, other clinical uses of clonidine have recently gained popularity for the treatment of neuropsychiatric disorders like attention-deficit hyperactivity disorder or Tourette syndrome, but the mechanism of action is still unclear. Here, we test the hypothesis that the noradrenergic system regulates the activity of subthalamo-motor cortical loops, and that this influence can be modulated by clonidine. We used pharmacological manipulation of clonidine in a placebo-controlled study in combination with subthalamic nucleus-deep brain stimulation (STN-DBS) in 16 Parkinson's disease patients performing a reaction time task requiring to refrain from reacting (proactive inhibition). We recorded electroencephalographical activity of the whole cortex, and applied spectral analyses directly at the source level after advanced blind source separation. We found only one cortical source localized to the supplementary motor area (SMA) that supported an interaction of pharmacological and subthalamic stimulation. Under placebo, STN-DBS reduced proactive alpha power in the SMA, a marker of local inhibitory activity. This effect was associated with the speeding-up of movement initiation. Clonidine substantially increased proactive alpha power from the SMA source, and canceled out the benefits of STN-DBS on movement initiation. These results provide the first direct neural evidence in humans that the tonic inhibitory activity of the subthalamocortical loops underlying the control of movement initiation is coupled to the noradrenergic system, and that this activity can be targeted by pharmacological agents acting on alpha-adrenergic receptors.
Collapse
Affiliation(s)
- Charlotte Spay
- Université de Lyon, Lyon, France.,Université Lyon 1, Villeurbanne, France.,INSERM, U 1028, Lyon Neuroscience Research Center, Lyon, France.,CNRS, UMR 5292, Lyon Neuroscience Research Center, Lyon, France
| | - Marion Albares
- Université de Lyon, Lyon, France.,Université Lyon 1, Villeurbanne, France.,CNRS, UMR 5229, Institut des Sciences Cognitives Marc Jeannerod, Bron, France.,Sorbonne Universités, UPMC Université Pierre et Marie Curie Paris 06, UMR 7225, Paris, France.,INSERM UMR 1127, Institut du cerveau et de la moelle épinière, ICM, Paris, France.,CNRS, UMR 7225, Institut du cerveau et de la moelle épinière, ICM, Paris, France
| | - Guillaume Lio
- Université de Lyon, Lyon, France.,Université Lyon 1, Villeurbanne, France.,CNRS, UMR 5229, Institut des Sciences Cognitives Marc Jeannerod, Bron, France
| | - Stephane Thobois
- Université de Lyon, Lyon, France.,Université Lyon 1, Villeurbanne, France.,CNRS, UMR 5229, Institut des Sciences Cognitives Marc Jeannerod, Bron, France.,Hospices civils de Lyon, hôpital neurologique Pierre Wertheimer, Bron, France
| | - Emmanuel Broussolle
- Université de Lyon, Lyon, France.,Université Lyon 1, Villeurbanne, France.,CNRS, UMR 5229, Institut des Sciences Cognitives Marc Jeannerod, Bron, France.,Hospices civils de Lyon, hôpital neurologique Pierre Wertheimer, Bron, France
| | - Brian Lau
- Sorbonne Universités, UPMC Université Pierre et Marie Curie Paris 06, UMR 7225, Paris, France.,INSERM UMR 1127, Institut du cerveau et de la moelle épinière, ICM, Paris, France.,CNRS, UMR 7225, Institut du cerveau et de la moelle épinière, ICM, Paris, France
| | - Benedicte Ballanger
- Université de Lyon, Lyon, France.,Université Lyon 1, Villeurbanne, France.,INSERM, U 1028, Lyon Neuroscience Research Center, Lyon, France.,CNRS, UMR 5292, Lyon Neuroscience Research Center, Lyon, France
| | - Philippe Boulinguez
- Université de Lyon, Lyon, France.,Université Lyon 1, Villeurbanne, France.,INSERM, U 1028, Lyon Neuroscience Research Center, Lyon, France.,CNRS, UMR 5292, Lyon Neuroscience Research Center, Lyon, France
| |
Collapse
|
19
|
Accolla EA, Pollo C. Mood Effects After Deep Brain Stimulation for Parkinson's Disease: An Update. Front Neurol 2019; 10:617. [PMID: 31258509 PMCID: PMC6587122 DOI: 10.3389/fneur.2019.00617] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 05/28/2019] [Indexed: 11/23/2022] Open
Abstract
Depression in Parkinson's Disease (PD) is a prevalent and invalidating symptom. Deep brain stimulation (DBS) allows for an improvement of PD motor features, but its effects on mood are difficult to predict. Here, we review the evidence regarding mood effects after DBS of either subthalamic nucleus (STN) or globus pallidus pars interna (GPi). Different influences of multiple factors contribute to impact the neuropsychiatric outcome after surgery. Psychosocial presurgical situation, postsurgical coping mechanisms, dopaminergic treatment modifications, and direct effects of the stimulation of either target are all playing a distinct role on the psychological well-being of patients undergoing DBS. No clear advantage of either target (STN vs. GPi) has been consistently found, both being effective and with a favorable profile on depression symptoms. However, specific patients' characteristics or anatomical considerations can guide the neurosurgeon in the target choice. Further research together with technological advances are expected to confine the stimulation area within dysfunctional circuits causing motor symptoms of PD.
Collapse
Affiliation(s)
- Ettore A. Accolla
- Neurology Unit, Department of Medicine, HFR – Hôpital Cantonal Fribourg and Fribourg University, Fribourg, Switzerland
| | - Claudio Pollo
- Department of Neurosurgery, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| |
Collapse
|
20
|
The effect of docosahexaenoic acid on apelin distribution of nervous system in the experimental mouse model of Parkinson’s disease. Tissue Cell 2019; 56:41-51. [DOI: 10.1016/j.tice.2018.12.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 11/19/2018] [Accepted: 12/05/2018] [Indexed: 12/21/2022]
|
21
|
Sancandi M, Schul EV, Economides G, Constanti A, Mercer A. Structural Changes Observed in the Piriform Cortex in a Rat Model of Pre-motor Parkinson's Disease. Front Cell Neurosci 2018; 12:479. [PMID: 30618629 PMCID: PMC6296349 DOI: 10.3389/fncel.2018.00479] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 11/22/2018] [Indexed: 12/11/2022] Open
Abstract
Early diagnosis of Parkinson’s disease (PD) offers perhaps, the most promising route to a successful clinical intervention, and the use of an animal model exhibiting symptoms comparable to those observed in PD patients in the early stage of the disease, may facilitate screening of novel therapies for delaying the onset of more debilitating motor and behavioral abnormalities. In this study, a rat model of pre-motor PD was used to study the etiology of hyposmia, a non-motor symptom linked to the early stage of the disease when the motor symptoms have yet to be experienced. The study focussed on determining the effect of a partial reduction of both dopamine and noradrenaline levels on the olfactory cortex. Neuroinflammation and striking structural changes were observed in the model. These changes were prevented by treatment with a neuroprotective drug, a glucagon-like peptide-1 (GLP1) receptor agonist, exendin-4 (EX-4).
Collapse
|
22
|
Loiodice S, Wing Young H, Rion B, Méot B, Montagne P, Denibaud AS, Viel R, Drieu La Rochelle C. Implication of nigral dopaminergic lesion and repeated L-dopa exposure in neuropsychiatric symptoms of Parkinson's disease. Behav Brain Res 2018; 360:120-127. [PMID: 30521934 DOI: 10.1016/j.bbr.2018.12.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 10/30/2018] [Accepted: 12/02/2018] [Indexed: 12/11/2022]
Abstract
This study aims to investigate the contribution of nigral dopaminergic (DA) cell loss, repeated exposure to DA medication and the combination of both to the development of neuropsychiatric symptoms observed in Parkinson's disease (PD). A bilateral 6-OHDA lesion of the substantia nigra pars compacta (SNc) was performed in rats. A set of animals was repeatedly administered with L-dopa (20 mg/kg/day) and benserazide (5 mg/kg/day) over 10 days starting from day 11 post-lesion. Behavioural testing was performed in week 3 post-lesion: novel object recognition (NOR), elevated plus maze (EPM) social interaction (SI) tests, and amphetamine-induced hyperlocomotion (AIH). Immunohistochemical analysis revealed a significant partial lesion (48%) in 6-OHDA versus sham rats. This lesion was not associated with motor impairment. However, lesioned rats displayed a significant deficit in the NOR, which was reversed by acute treatment with l-dopa/benserazide (12.5 mg/kg and 15 mg/kg respectively). Lesioned rats also displayed a deficit in the EPM which was not reversed by acute treatment with l-dopa. No difference was observed in the SI test or in the AIH assay. In all assays, no effect of chronic l-dopa exposure was observed. This study provides new insights into the neuropathophysiology associated with neuropsychiatric symptoms of PD. Our data strongly emphasises a not previously clearly identified critical role in cognition for the SNc. The results suggest that DA pathways were less directly involved in lesion-induced anxiety-like behaviour. We did not report any effect of chronic l-dopa exposure in the context of partial nigral cell loss.
Collapse
Affiliation(s)
- Simon Loiodice
- Biotrial Pharmacology, 7-9 rue Jean-Louis Bertrand, 35042, Rennes, France.
| | - Harry Wing Young
- Biotrial Pharmacology, 7-9 rue Jean-Louis Bertrand, 35042, Rennes, France
| | - Bertrand Rion
- Biotrial Pharmacology, 7-9 rue Jean-Louis Bertrand, 35042, Rennes, France
| | - Benoît Méot
- Biotrial Pharmacology, 7-9 rue Jean-Louis Bertrand, 35042, Rennes, France
| | - Pierre Montagne
- Biotrial Pharmacology, 7-9 rue Jean-Louis Bertrand, 35042, Rennes, France
| | | | - Roselyne Viel
- Plate-Forme H2P2, Université de Rennes 1, Biosit, 2 Av. du Prof. Léon Bernard, 35043, Rennes, France
| | | |
Collapse
|
23
|
Li Y, Jiao Q, Du X, Bi M, Han S, Jiao L, Jiang H. Investigation of Behavioral Dysfunctions Induced by Monoamine Depletions in a Mouse Model of Parkinson's Disease. Front Cell Neurosci 2018; 12:241. [PMID: 30135645 PMCID: PMC6092512 DOI: 10.3389/fncel.2018.00241] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 07/17/2018] [Indexed: 01/10/2023] Open
Abstract
Parkinson's disease (PD) is characterized not only by typical motor symptoms, but also by nonmotor symptoms in the early stages. In addition to the loss of dopaminergic (DAergic) neurons, progressive degenerations of noradrenergic (NA) and serotonergic (5-HT) neurons were also observed. However, the respective effects and interactions of these monoamine depletions on certain nonmotor symptoms are still largely unknown. In the present study, we performed selective depletions of NA, 5-HT and DA in mice by intraperitioneal injection of N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine hydrochloride (DSP-4), 4-chloro-L-phenylalanine (pCPA) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), respectively. DSP-4 led to a 34% decrease in the number of NAergic neurons in the locus coeruleus, and MPTP led to a 30% decrease in the number of DAergic neurons in the substantia nigra. Although there was no obvious change in the number of 5-HTergic neurons in the dorsal raphe nucleus after pCPA treatment, the levels of 5-HT and its metabolite in the frontal cortex and hippocampus were reduced, respectively. Locomotor activity deficit was induced by DA depletion and a decrease in traveled distance was potentiated by additional NA depletion. Despair-associated depressive-like behavior could be observed in every group. Anxiety states emerged only from the combined depletion of two or three monoamines. However, combined depletion of the three monoamines dramatically induced anhedonia, and it could also aggravate the depressive-like and anxiety behavior. Furthermore, NA depletion significantly reduced spatial learning and memory ability, which was not enhanced by additional 5-HT or DA depletion. Our data highlighted the interactive role of NA, 5-HT and DA in the motor, emotional and cognitive deficits, providing new insight into the complex orchestration of impaired monoaminergic systems that related to the pathology of PD.
Collapse
Affiliation(s)
- Yong Li
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines, Physiology, Qingdao University Medical College, Qingdao University, Qingdao, China
| | - Qian Jiao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines, Physiology, Qingdao University Medical College, Qingdao University, Qingdao, China
| | - Xixun Du
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines, Physiology, Qingdao University Medical College, Qingdao University, Qingdao, China
| | - Mingxia Bi
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines, Physiology, Qingdao University Medical College, Qingdao University, Qingdao, China
| | - Shuaishuai Han
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines, Physiology, Qingdao University Medical College, Qingdao University, Qingdao, China
| | - Lingling Jiao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines, Physiology, Qingdao University Medical College, Qingdao University, Qingdao, China
| | - Hong Jiang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines, Physiology, Qingdao University Medical College, Qingdao University, Qingdao, China
| |
Collapse
|
24
|
Charles KA, Naudet F, Bouali-Benazzouz R, Landry M, De Deurwaerdère P, Fossat P, Benazzouz A. Alteration of nociceptive integration in the spinal cord of a rat model of Parkinson's disease. Mov Disord 2018; 33:1010-1015. [PMID: 29774960 DOI: 10.1002/mds.27377] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 02/06/2018] [Accepted: 02/11/2018] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Pain is a major non motor symptom that contributes to impaired quality of life in PD. However, its mechanism is unknown. OBJECTIVES AND METHODS We sought to identify the pain phenotypes and parallel changes in spinal integration of peripheral stimuli in a rat model of PD induced by lesions of SN dopamine neurons, using behavioral plantar and von Frey tests as well as electrophysiology of the dorsal horn. RESULTS We show that dopamine depletion by 6-OHDA induced hypersensitivity to mechanical and thermal stimuli. These abnormal behaviors were paralleled by increased neuronal responses and hyperexcitability of wide dynamic range neurons of lamina V of the dorsal horn of the spinal cord in response to electrical stimulation of the sciatic nerve in the 6-OHDA model as compared to sham rats. CONCLUSIONS These results provide evidence for alteration of nociceptive integration in the spinal dorsal horn neurons in 6-OHDA rats that can reflect changes in pain behavior. © 2018 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Keri-Ann Charles
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Frédéric Naudet
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | | | - Marc Landry
- CNRS, Intitut Interdisciplinaire de Neurosciences, UMR5297, Bordeaux, France
| | - Philippe De Deurwaerdère
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Pascal Fossat
- CNRS, Intitut Interdisciplinaire de Neurosciences, UMR5297, Bordeaux, France
| | - Abdelhamid Benazzouz
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| |
Collapse
|
25
|
Dandekar MP, Fenoy AJ, Carvalho AF, Soares JC, Quevedo J. Deep brain stimulation for treatment-resistant depression: an integrative review of preclinical and clinical findings and translational implications. Mol Psychiatry 2018; 23:1094-1112. [PMID: 29483673 DOI: 10.1038/mp.2018.2] [Citation(s) in RCA: 183] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 12/05/2017] [Accepted: 12/15/2017] [Indexed: 02/07/2023]
Abstract
Although deep brain stimulation (DBS) is an established treatment choice for Parkinson's disease (PD), essential tremor and movement disorders, its effectiveness for the management of treatment-resistant depression (TRD) remains unclear. Herein, we conducted an integrative review on major neuroanatomical targets of DBS pursued for the treatment of intractable TRD. The aim of this review article is to provide a critical discussion of possible underlying mechanisms for DBS-generated antidepressant effects identified in preclinical studies and clinical trials, and to determine which brain target(s) elicited the most promising outcomes considering acute and maintenance treatment of TRD. Major electronic databases were searched to identify preclinical and clinical studies that have investigated the effects of DBS on depression-related outcomes. Overall, 92 references met inclusion criteria, and have evaluated six unique DBS targets namely the subcallosal cingulate gyrus (SCG), nucleus accumbens (NAc), ventral capsule/ventral striatum or anterior limb of internal capsule (ALIC), medial forebrain bundle (MFB), lateral habenula (LHb) and inferior thalamic peduncle for the treatment of unrelenting TRD. Electrical stimulation of these pertinent brain regions displayed differential effects on mood transition in patients with TRD. In addition, 47 unique references provided preclinical evidence for putative neurobiological mechanisms underlying antidepressant effects of DBS applied to the ventromedial prefrontal cortex, NAc, MFB, LHb and subthalamic nucleus. Preclinical studies suggest that stimulation parameters and neuroanatomical locations could influence DBS-related antidepressant effects, and also pointed that modulatory effects on monoamine neurotransmitters in target regions or interconnected brain networks following DBS could have a role in the antidepressant effects of DBS. Among several neuromodulatory targets that have been investigated, DBS in the neuroanatomical framework of the SCG, ALIC and MFB yielded more consistent antidepressant response rates in samples with TRD. Nevertheless, more well-designed randomized double-blind, controlled trials are warranted to further assess the efficacy, safety and tolerability of these more promising DBS targets for the management of TRD as therapeutic effects have been inconsistent across some controlled studies.
Collapse
Affiliation(s)
- M P Dandekar
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - A J Fenoy
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - A F Carvalho
- Department of Clinical Medicine and Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - J C Soares
- Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - J Quevedo
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.,Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.,Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA.,Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, Brazil
| |
Collapse
|
26
|
Premature aging in behavior and immune functions in tyrosine hydroxylase haploinsufficient female mice. A longitudinal study. Brain Behav Immun 2018; 69:440-455. [PMID: 29341892 DOI: 10.1016/j.bbi.2018.01.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 12/21/2017] [Accepted: 01/11/2018] [Indexed: 01/03/2023] Open
Abstract
Aging is accompanied by impairment in the nervous, immune, and endocrine systems as well as in neuroimmunoendocrine communication. In this context, there is an age-related alteration of the physiological response to acute stress, which is modulated by catecholamine (CA), final products of the sympathetic-adreno-medullary axis. The involvement of CA in essential functions of the nervous system is consistent with the neuropsychological deficits found in mice with haploinsufficiency (hemizygous; HZ) of tyrosine hydroxylase (TH) enzyme (TH-HZ). However, other possible alterations in regulatory systems have not been studied in these animals. The aim of the present work was to analyze whether adult TH-HZ female mice presented the impairment of behavioral traits and immunological responses that occurs with aging and whether they had affected their mean lifespan. ICR-CD1 female TH-HZ and wild type (WT) mice were used in a longitudinal study. Behavioral tests were performed on adult and old mice in order to evaluate their sensorimotor abilities and exploratory capacity, as well as anxiety-like behaviors. At the ages of 2 ± 1, 4 ± 1, 9 ± 1, 13 ± 1 and 20 ± 1 months, peritoneal leukocytes were extracted and several immune functions were assessed (phagocytic capacity, Natural Killer (NK) cytotoxicity, and lymphoproliferative response to lipopolysaccharide (LPS) and concanavalin A (ConA)). In addition, several oxidative stress parameters (catalase, glutathione reductase and glutathione peroxidase activities, and reduced glutathione (GSH) concentrations as antioxidant compounds as well as xanthine oxidase activity, oxidized glutathione (GSSG) concentrations, and GSSG/GSH ratio as oxidants) were analyzed. As inflammatory stress parameters TNF-alpha and IL-10 concentrations, and TNF-alpha/IL-10 ratios as inflammatory/anti-inflammatory markers, were measured. Animals were maintained in standard conditions until their natural death. The results indicate that adult TH-HZ mice presented worse sensorimotor abilities and exploratory capacity than their WT littermates as well as greater anxiety-like behaviors. With regards to the immune system, adult TH-HZ animals exhibited lower values of phagocytic capacity, NK cytotoxicity, and lymphoproliferative response to LPS and ConA than WT mice. Moreover, immune cells of TH-HZ mice showed higher oxidative and inflammatory stress than those of WT animals. Although these differences between TH-HZ and WT, in general, decreased with aging, this premature immunosenescence and impairment of behavior of TH-HZ mice was accompanied by a shorter mean lifespan in comparison to WT counterparts. In conclusion, haploinsufficiency of th gene in female mice appears to provoke premature aging of the regulatory systems affecting mean lifespan.
Collapse
|
27
|
Bonato JM, Bassani TB, Milani H, Vital MABF, de Oliveira RMW. Pioglitazone reduces mortality, prevents depressive-like behavior, and impacts hippocampal neurogenesis in the 6-OHDA model of Parkinson's disease in rats. Exp Neurol 2018; 300:188-200. [DOI: 10.1016/j.expneurol.2017.11.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 10/23/2017] [Accepted: 11/15/2017] [Indexed: 12/20/2022]
|
28
|
Faggiani E, Naudet F, Janssen ML, Temel Y, Benazzouz A. Serotonergic neurons mediate the anxiolytic effect of l -DOPA: Neuronal correlates in the amygdala. Neurobiol Dis 2018; 110:20-28. [DOI: 10.1016/j.nbd.2017.11.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 10/11/2017] [Accepted: 11/02/2017] [Indexed: 10/18/2022] Open
|
29
|
Depression in Parkinson's Disease: The Contribution from Animal Studies. PARKINSONS DISEASE 2017; 2017:9124160. [PMID: 29158943 PMCID: PMC5660814 DOI: 10.1155/2017/9124160] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 09/07/2017] [Indexed: 02/06/2023]
Abstract
Besides being better known for causing motor impairments, Parkinson's disease (PD) can also cause many nonmotor symptoms, like depression and anxiety, which can cause significant loss of life quality and may not respond to regular drugs treatment. In this review, we discuss the depression in PD, based on data from studies in humans and rodents. Depression frequency seems higher in PD patients than in general population, despite high variation in data due to diagnosis disparities. Development of depression in PD seems more likely to be caused by the nigrostriatal pathway degeneration than as a consequence of the awareness of disease prognostic, and it seems to be related to dopaminergic, noradrenergic, and serotoninergic synapses deficits. The dopaminergic role could be more significant, since it can modulate the release of the others, and its depletion is progressive, due to the degenerative feature of PD. Highly regarded in major depression, serotonin can be depleted in rats after nigrostriatal damage, but data from human patients are more conflicting. Animal studies can help in understanding the neurobiological mechanisms of depression in PD and the pursuit for more effective drugs for its treatment, but they lack the complexity of the disease progression, especially the nondopaminergic degeneration.
Collapse
|
30
|
Sabbar M, Dkhissi-Benyahya O, Benazzouz A, Lakhdar-Ghazal N. Circadian Clock Protein Content and Daily Rhythm of Locomotor Activity Are Altered after Chronic Exposure to Lead in Rat. Front Behav Neurosci 2017; 11:178. [PMID: 28970786 PMCID: PMC5609114 DOI: 10.3389/fnbeh.2017.00178] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 09/08/2017] [Indexed: 12/13/2022] Open
Abstract
Lead exposure has been reported to produce many clinical features, including parkinsonism. However, its consequences on the circadian rhythms are still unknown. Here we aimed to examine the circadian rhythms of locomotor activity following lead intoxication and investigate the mechanisms by which lead may induce alterations of circadian rhythms in rats. Male Wistar rats were injected with lead or sodium acetate (10 mg/kg/day, i.p.) during 4 weeks. Both groups were tested in the “open field” to quantify the exploratory activity and in the rotarod to evaluate motor coordination. Then, animals were submitted to continuous 24 h recordings of locomotor activity under 14/10 Light/dark (14/10 LD) cycle and in complete darkness (DD). At the end of experiments, the clock proteins BMAL1, PER1-2, and CRY1-2 were assayed in the suprachiasmatic nucleus (SCN) using immunohistochemistry. We showed that lead significantly reduced the number of crossing in the open field, impaired motor coordination and altered the daily locomotor activity rhythm. When the LD cycle was advanced by 6 h, both groups adjusted their daily locomotor activity to the new LD cycle with high onset variability in lead-intoxicated rats compared to controls. Lead also led to a decrease in the number of immunoreactive cells (ir-) of BMAL1, PER1, and PER2 without affecting the number of ir-CRY1 and ir-CRY2 cells in the SCN. Our data provide strong evidence that lead intoxication disturbs the rhythm of locomotor activity and alters clock proteins expression in the SCN. They contribute to the understanding of the mechanism by which lead induce circadian rhythms disturbances.
Collapse
Affiliation(s)
- Mariam Sabbar
- Équipe de Recherche sur les Rythmes Biologiques, Neurosciences et Environnement, Faculté des Sciences, Université Mohammed VRabat, Morocco
| | - Ouria Dkhissi-Benyahya
- INSERM, Stem Cell and Brain Research Institute U1208, University of Lyon, Université Claude Bernard Lyon 1Lyon, France
| | - Abdelhamid Benazzouz
- Institut des Maladies Neurodégénératives, Univ. de Bordeaux, UMR5293Bordeaux, France.,Centre National de la Recherche Scientifique, Institut des Maladies Neurodégénératives, UMR5293Bordeaux, France
| | - Nouria Lakhdar-Ghazal
- Équipe de Recherche sur les Rythmes Biologiques, Neurosciences et Environnement, Faculté des Sciences, Université Mohammed VRabat, Morocco
| |
Collapse
|
31
|
Faggiani E, Benazzouz A. Deep brain stimulation of the subthalamic nucleus in Parkinson’s disease: From history to the interaction with the monoaminergic systems. Prog Neurobiol 2017; 151:139-156. [DOI: 10.1016/j.pneurobio.2016.07.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 07/08/2016] [Indexed: 11/16/2022]
|
32
|
Stefani A, Trendafilov V, Liguori C, Fedele E, Galati S. Subthalamic nucleus deep brain stimulation on motor-symptoms of Parkinson's disease: Focus on neurochemistry. Prog Neurobiol 2017; 151:157-174. [PMID: 28159574 DOI: 10.1016/j.pneurobio.2017.01.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 01/20/2017] [Accepted: 01/22/2017] [Indexed: 12/19/2022]
Abstract
Deep brain stimulation (DBS) has become a standard therapy for Parkinson's disease (PD) and it is also currently under investigation for other neurological and psychiatric disorders. Although many scientific, clinical and ethical issues are still unresolved, DBS delivered into the subthalamic nucleus (STN) has improved the quality of life of several thousands of patients. The mechanisms underlying STN-DBS have been debated extensively in several reviews; less investigated are the biochemical consequences, which are still under scrutiny. Crucial and only partially understood, for instance, are the complex interplays occurring between STN-DBS and levodopa (LD)-centred therapy in the post-surgery follow-up. The main goal of this review is to address the question of whether an improved motor control, based on STN-DBS therapy, is also achieved through the additional modulation of other neurotransmitters, such as noradrenaline (NA) and serotonin (5-HT). A critical issue is to understand not only acute DBS-mediated effects, but also chronic changes, such as those involving cyclic nucleotides, capable of modulating circuit plasticity. The present article will discuss the neurochemical changes promoted by STN-DBS and will document the main results obtained in microdialysis studies. Furthermore, we will also examine the preliminary achievements of voltammetry applied to humans, and discuss new hypothetical investigational routes, taking into account novel players such as glia, or subcortical regions such as the pedunculopontine (PPN) area. Our further understanding of specific changes in brain chemistry promoted by STN-DBS would further disseminate its utilisation, at any stage of disease, avoiding an irreversible lesioning approach.
Collapse
Affiliation(s)
- A Stefani
- Department of System Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - V Trendafilov
- Laboratory for Biomedical Neurosciences (LBN), Neurocenter of Southern Switzerland (NSI), Lugano, Switzerland
| | - C Liguori
- Department of System Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - E Fedele
- Department of Pharmacy, Pharmacology and Toxicology Unit and Center of Excellence for Biomedical Research, University of Genoa, 16148 Genoa, Italy
| | - S Galati
- Laboratory for Biomedical Neurosciences (LBN), Neurocenter of Southern Switzerland (NSI), Lugano, Switzerland.
| |
Collapse
|
33
|
Benazzouz A. Twenty-four years of deep brain stimulation of the subthalamic nucleus. Rev Neurol (Paris) 2016; 173:103-105. [PMID: 28041608 DOI: 10.1016/j.neurol.2016.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 12/08/2016] [Indexed: 11/15/2022]
Affiliation(s)
- A Benazzouz
- Institut des maladies neurodégénératives, université de Bordeaux, CNRS UMR 5293, 146, rue Léo-Saignat, 33076 Bordeaux, France.
| |
Collapse
|
34
|
De Deurwaerdère P, Di Giovanni G, Millan MJ. Expanding the repertoire of L-DOPA's actions: A comprehensive review of its functional neurochemistry. Prog Neurobiol 2016; 151:57-100. [PMID: 27389773 DOI: 10.1016/j.pneurobio.2016.07.002] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 06/18/2016] [Accepted: 07/03/2016] [Indexed: 01/11/2023]
Abstract
Though a multi-facetted disorder, Parkinson's disease is prototypically characterized by neurodegeneration of nigrostriatal dopaminergic neurons of the substantia nigra pars compacta, leading to a severe disruption of motor function. Accordingly, L-DOPA, the metabolic precursor of dopamine (DA), is well-established as a treatment for the motor deficits of Parkinson's disease despite long-term complications such as dyskinesia and psychiatric side-effects. Paradoxically, however, despite the traditional assumption that L-DOPA is transformed in residual striatal dopaminergic neurons into DA, the mechanism of action of L-DOPA is neither simple nor entirely clear. Herein, focussing on its influence upon extracellular DA and other neuromodulators in intact animals and experimental models of Parkinson's disease, we highlight effects other than striatal generation of DA in the functional profile of L-DOPA. While not excluding a minor role for glial cells, L-DOPA is principally transformed into DA in neurons yet, interestingly, with a more important role for serotonergic than dopaminergic projections. Moreover, in addition to the striatum, L-DOPA evokes marked increases in extracellular DA in frontal cortex, nucleus accumbens, the subthalamic nucleus and additional extra-striatal regions. In considering its functional profile, it is also important to bear in mind the marked (probably indirect) influence of L-DOPA upon cholinergic, GABAergic and glutamatergic neurons in the basal ganglia and/or cortex, while anomalous serotonergic transmission is incriminated in the emergence of L-DOPA elicited dyskinesia and psychosis. Finally, L-DOPA may exert intrinsic receptor-mediated actions independently of DA neurotransmission and can be processed into bioactive metabolites. In conclusion, L-DOPA exerts a surprisingly complex pattern of neurochemical effects of much greater scope that mere striatal transformation into DA in spared dopaminergic neurons. Their further experimental and clinical clarification should help improve both L-DOPA-based and novel strategies for controlling the motor and other symptoms of Parkinson's disease.
Collapse
Affiliation(s)
- Philippe De Deurwaerdère
- CNRS (Centre National de la Recherche Scientifique), Institut des Maladies Neurodégénératives, UMR CNRS 5293, F-33000 Bordeaux, France.
| | - Giuseppe Di Giovanni
- Neuroscience Division, School of Biosciences, Cardiff University, Cardiff, UK; Department of Physiology & Biochemistry, Faculty of Medicine and Surgery, University of Malta, Malta
| | - Mark J Millan
- Institut de Recherche Servier, Pole for Therapeutic Innovation in Neuropsychiatry, 78290 Croissy/Seine,Paris, France
| |
Collapse
|
35
|
Wan OW, Shin E, Mattsson B, Caudal D, Svenningsson P, Björklund A. α-Synuclein induced toxicity in brain stem serotonin neurons mediated by an AAV vector driven by the tryptophan hydroxylase promoter. Sci Rep 2016; 6:26285. [PMID: 27211987 PMCID: PMC4876322 DOI: 10.1038/srep26285] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 04/27/2016] [Indexed: 02/01/2023] Open
Abstract
We studied the impact of α-synuclein overexpression in brainstem serotonin neurons using a novel vector construct where the expression of human wildtype α-synuclein is driven by the tryptophan hydroxylase promoter, allowing expression of α-synuclein at elevated levels, and with high selectivity, in serotonergic neurons. α-Synuclein induced degenerative changes in axons and dendrites, displaying a distorted appearance, suggesting accumulation and aggregation of α-synuclein as a result of impaired axonal transport, accompanied by a 40% loss of terminals, as assessed in the hippocampus. Tissue levels of serotonin and its major metabolite 5-HIAA remained largely unaltered, and the performance of the α-synuclein overexpressing rats in tests of spatial learning (water maze), anxiety related behavior (elevated plus maze) and depressive-like behavior (forced swim test) was not different from control, suggesting that the impact of the developing axonal pathology on serotonin neurotransmission was relatively mild. Overexpression of α-synuclein in the raphe nuclei, combined with overexpression in basal forebrain cholinergic neurons, resulted in more pronounced axonal pathology and significant impairment in the elevated plus maze. We conclude that α-synuclein pathology in serotonergic or cholinergic neurons alone is not sufficient to impair non-motor behaviors, but that it is their simultaneous involvement that determines severity of such symptoms.
Collapse
Affiliation(s)
- Oi Wan Wan
- Wallenberg Neuroscience Center, Department of Experimental Medical Sciences, Lund University, BMC A11, Lund 22184, Sweden
| | - Eunju Shin
- Wallenberg Neuroscience Center, Department of Experimental Medical Sciences, Lund University, BMC A11, Lund 22184, Sweden
| | - Bengt Mattsson
- Wallenberg Neuroscience Center, Department of Experimental Medical Sciences, Lund University, BMC A11, Lund 22184, Sweden
| | - Dorian Caudal
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institute, Stockholm 17176, Sweden
| | - Per Svenningsson
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institute, Stockholm 17176, Sweden
| | - Anders Björklund
- Wallenberg Neuroscience Center, Department of Experimental Medical Sciences, Lund University, BMC A11, Lund 22184, Sweden
| |
Collapse
|
36
|
De Deurwaerdère P, Di Giovanni G. Serotonergic modulation of the activity of mesencephalic dopaminergic systems: Therapeutic implications. Prog Neurobiol 2016; 151:175-236. [PMID: 27013075 DOI: 10.1016/j.pneurobio.2016.03.004] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 03/13/2016] [Accepted: 03/14/2016] [Indexed: 12/14/2022]
Abstract
Since their discovery in the mammalian brain, it has been apparent that serotonin (5-HT) and dopamine (DA) interactions play a key role in normal and abnormal behavior. Therefore, disclosure of this interaction could reveal important insights into the pathogenesis of various neuropsychiatric diseases including schizophrenia, depression and drug addiction or neurological conditions such as Parkinson's disease and Tourette's syndrome. Unfortunately, this interaction remains difficult to study for many reasons, including the rich and widespread innervations of 5-HT and DA in the brain, the plethora of 5-HT receptors and the release of co-transmitters by 5-HT and DA neurons. The purpose of this review is to present electrophysiological and biochemical data showing that endogenous 5-HT and pharmacological 5-HT ligands modify the mesencephalic DA systems' activity. 5-HT receptors may control DA neuron activity in a state-dependent and region-dependent manner. 5-HT controls the activity of DA neurons in a phasic and excitatory manner, except for the control exerted by 5-HT2C receptors which appears to also be tonically and/or constitutively inhibitory. The functional interaction between the two monoamines will also be discussed in view of the mechanism of action of antidepressants, antipsychotics, anti-Parkinsonians and drugs of abuse.
Collapse
Affiliation(s)
- Philippe De Deurwaerdère
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5293, 33076 Bordeaux Cedex, France.
| | - Giuseppe Di Giovanni
- Department of Physiology & Biochemistry, Faculty of Medicine and Surgery, University of Malta, Malta; Neuroscience Division, School of Biosciences, Cardiff University, Cardiff, UK.
| |
Collapse
|
37
|
Magnard R, Vachez Y, Carcenac C, Krack P, David O, Savasta M, Boulet S, Carnicella S. What can rodent models tell us about apathy and associated neuropsychiatric symptoms in Parkinson's disease? Transl Psychiatry 2016; 6:e753. [PMID: 26954980 PMCID: PMC4872443 DOI: 10.1038/tp.2016.17] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 01/11/2016] [Accepted: 01/19/2016] [Indexed: 12/19/2022] Open
Abstract
In addition to classical motor symptoms, Parkinson's disease (PD) patients display incapacitating neuropsychiatric manifestations, such as apathy, anhedonia, depression and anxiety. These hitherto generally neglected non-motor symptoms, have gained increasing interest in medical and scientific communities over the last decade because of the extent of their negative impact on PD patients' quality of life. Although recent clinical and functional imaging studies have provided useful information, the pathophysiology of apathy and associated affective impairments remains elusive. Our aim in this review is to summarize and discuss recent advances in the development of rodent models of PD-related neuropsychiatric symptoms using neurotoxin lesion-based approaches. The data collected suggest that bilateral and partial lesions of the nigrostriatal system aimed at inducing reliable neuropsychiatric-like deficits while avoiding severe motor impairments that may interfere with behavioral evaluation, is a more selective and efficient strategy than medial forebrain bundle lesions. Moreover, of all the different classes of pharmacological agents, D2/D3 receptor agonists such as pramipexole appear to be the most efficient treatment for the wide range of behavioral deficits induced by dopaminergic lesions. Lesion-based rodent models, therefore, appear to be relevant tools for studying the pathophysiology of the non-motor symptoms of PD. Data accumulated so far confirm the causative role of dopaminergic depletion, especially in the nigrostriatal system, in the development of behavioral impairments related to apathy, depression and anxiety. They also put forward D2/D3 receptors as potential targets for the treatment of such neuropsychiatric symptoms in PD.
Collapse
Affiliation(s)
- R Magnard
- Inserm U1216, Grenoble, France,Université Grenoble Alpes, Grenoble Institut des Neurosciences, Grenoble, France
| | - Y Vachez
- Inserm U1216, Grenoble, France,Université Grenoble Alpes, Grenoble Institut des Neurosciences, Grenoble, France
| | - C Carcenac
- Inserm U1216, Grenoble, France,Université Grenoble Alpes, Grenoble Institut des Neurosciences, Grenoble, France
| | - P Krack
- Inserm U1216, Grenoble, France,Université Grenoble Alpes, Grenoble Institut des Neurosciences, Grenoble, France,Movement Disorder Unit, Department of Psychiatry and Neurology, CHU de Grenoble, Grenoble, France
| | - O David
- Inserm U1216, Grenoble, France,Université Grenoble Alpes, Grenoble Institut des Neurosciences, Grenoble, France
| | - M Savasta
- Inserm U1216, Grenoble, France,Université Grenoble Alpes, Grenoble Institut des Neurosciences, Grenoble, France
| | - S Boulet
- Inserm U1216, Grenoble, France,Université Grenoble Alpes, Grenoble Institut des Neurosciences, Grenoble, France
| | - S Carnicella
- Inserm U1216, Grenoble, France,Université Grenoble Alpes, Grenoble Institut des Neurosciences, Grenoble, France,Inserm U1216, Grenoble Institute of Neuroscience, Site Santé La Tronche - BP 170, 38042 Grenoble, France. E-mail:
| |
Collapse
|
38
|
Tinakoua A, Bouabid S, Faggiani E, De Deurwaerdère P, Lakhdar-Ghazal N, Benazzouz A. The impact of combined administration of paraquat and maneb on motor and non-motor functions in the rat. Neuroscience 2015; 311:118-29. [PMID: 26477982 DOI: 10.1016/j.neuroscience.2015.10.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 10/09/2015] [Accepted: 10/12/2015] [Indexed: 11/29/2022]
Abstract
Paraquat (PQ) and maneb (MB) are potential risk factors for Parkinson's disease. However, their impact on non-motor disorders, monoamine neurotransmission and basal ganglia function is not clearly determined. Here we investigated the effects of combined treatment with PQ/MB on motor behavior, anxiety and "depressive-like" disorders, tissue content of monoamines, and subthalamic nucleus (STN) neuronal activity. Male Sprague-Dawley rats were intoxicated by PQ (10 mg/kg) and MB (30 mg/kg) twice a week. Two weeks later, the majority of animals (group 1, 16/26) showed a severe loss of body weight with tremor and respiratory distress and others (group 2, 6/26) showed only tremor. Animals of group 2 received PQ/MB during four weeks before developing weight loss. A last group (group 3, 4/26) was insensitive to PQ/MB after 6 weeks of injections. Groups 1 and 2 displayed a failure of motor activity and motor coordination. Group 3 showed slight motor deficits only after the last injection of PQ/MB. Moreover, PQ/MB induced anxiety and "depressive-like" behaviors in animals of groups 2 and 3. Biochemical analysis showed that PQ/MB reduced striatal dopamine (DA) tissue content paralleled by changes in the activity of STN neurons without changing the content of norepinephrine and serotonin in the cortex. Our data provide evidence that individuals are not equally sensitive to PQ/MB and show that the motor deficits in vulnerable animals, are not only a result of DA neuron degeneration, but may also be a consequence of peripheral disabilities. Nevertheless, the parkinsonian-like non-motor impairments may be a direct consequence of the bilateral DA depletion.
Collapse
Affiliation(s)
- A Tinakoua
- Univ. de Bordeaux, Institut des maladies neurodégénératives, UMR 5293, Bordeaux, France; CNRS, Institut des maladies neurodégénératives, UMR 5293, Bordeaux, France; Université Mohammed V, Faculté des Sciences, Equipe Rythmes Biologiques, Neurosciences et Environnement, Rabat, Morocco
| | - S Bouabid
- Univ. de Bordeaux, Institut des maladies neurodégénératives, UMR 5293, Bordeaux, France; CNRS, Institut des maladies neurodégénératives, UMR 5293, Bordeaux, France; Université Mohammed V, Faculté des Sciences, Equipe Rythmes Biologiques, Neurosciences et Environnement, Rabat, Morocco
| | - E Faggiani
- Univ. de Bordeaux, Institut des maladies neurodégénératives, UMR 5293, Bordeaux, France; CNRS, Institut des maladies neurodégénératives, UMR 5293, Bordeaux, France
| | - P De Deurwaerdère
- Univ. de Bordeaux, Institut des maladies neurodégénératives, UMR 5293, Bordeaux, France; CNRS, Institut des maladies neurodégénératives, UMR 5293, Bordeaux, France
| | - N Lakhdar-Ghazal
- Université Mohammed V, Faculté des Sciences, Equipe Rythmes Biologiques, Neurosciences et Environnement, Rabat, Morocco
| | - A Benazzouz
- Univ. de Bordeaux, Institut des maladies neurodégénératives, UMR 5293, Bordeaux, France; CNRS, Institut des maladies neurodégénératives, UMR 5293, Bordeaux, France.
| |
Collapse
|