1
|
Witzig V, Pjontek R, Tan SKH, Schulz JB, Holtbernd F. Modulating the cholinergic system-Novel targets for deep brain stimulation in Parkinson's disease. J Neurochem 2025; 169:e16264. [PMID: 39556446 PMCID: PMC11808463 DOI: 10.1111/jnc.16264] [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: 06/11/2024] [Revised: 10/25/2024] [Accepted: 10/25/2024] [Indexed: 11/20/2024]
Abstract
Parkinson's disease (PD) is the second-fastest growing neurodegenerative disease in the world. The major clinical symptoms rigor, tremor, and bradykinesia derive from the degeneration of the nigrostriatal pathway. However, PD is a multi-system disease, and neurodegeneration extends beyond the degradation of the dopaminergic pathway. Symptoms such as postural instability, freezing of gait, falls, and cognitive decline are predominantly caused by alterations of transmitter systems outside the classical dopaminergic axis. While levodopa and deep brain stimulation (DBS) of the subthalamic nucleus or globus pallidus internus effectively address PD primary motor symptoms, they often fall short in mitigating axial symptoms and cognitive impairment. Along these lines, the cholinergic system is increasingly recognized to play a crucial role in governing locomotion, postural stability, and cognitive function. Thus, there is a growing interest in bolstering the cholinergic tone by DBS of cholinergic targets such as the pedunculopontine nucleus (PPN) and nucleus basalis of Meynert (NBM), aiming to alleviate these debilitating symptoms resistant to traditional treatment strategies targeting the dopaminergic network. This review offers a comprehensive overview of the role of cholinergic dysfunction in PD. We discuss the impact of PPN and NBM DBS on the management of symptoms not readily accessible to established DBS targets and pharmacotherapy in PD and seek to provide guidance on patient selection, surgical approach, and stimulation paradigms.
Collapse
Affiliation(s)
- V. Witzig
- Department of NeurologyRWTH Aachen UniversityAachenGermany
| | - R. Pjontek
- Department of NeurosurgeryRWTH Aachen UniversityAachenGermany
- Department of Stereotactic and Functional NeurosurgeryUniversity Hospital CologneCologneGermany
| | - S. K. H. Tan
- Department of NeurosurgeryAntwerp University HospitalEdegemBelgium
- Translational Neurosciences, Faculty of Medicine and Health SciencesUniversity of AntwerpAntwerpBelgium
| | - J. B. Schulz
- Department of NeurologyRWTH Aachen UniversityAachenGermany
- JARA‐BRAIN Institute Molecular Neuroscience and NeuroimagingJülich Research Center GmbH and RWTH Aachen UniversityAachenGermany
| | - F. Holtbernd
- Department of NeurologyRWTH Aachen UniversityAachenGermany
- JARA‐BRAIN Institute Molecular Neuroscience and NeuroimagingJülich Research Center GmbH and RWTH Aachen UniversityAachenGermany
- Jülich Research Center, Institutes of Neuroscience and Medicine (INM‐4, INM‐11)JülichGermany
| |
Collapse
|
2
|
Shan AD, Zhang H, Gao MX, Wang LN, Cao XY, Gan CT, Sun HM, Lu QL, Zhang L, Yuan YS, Zhang KZ. Altered effective connectivity in Parkinson's disease patients with rapid eye movement sleep behavior disorder: a resting-state functional magnetic resonance imaging study and support vector machine analysis. Quant Imaging Med Surg 2025; 15:352-369. [PMID: 39839023 PMCID: PMC11744126 DOI: 10.21037/qims-24-1196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 11/06/2024] [Indexed: 01/23/2025]
Abstract
Background Rapid eye movement sleep behavior disorder (RBD) is associated with pathological α-synuclein deposition and may have different damage directions due to α-synuclein spreading orientations. Recent functional imaging studies of Parkinson's disease (PD) with RBD have identified abnormalities in connectivity, but effective connectivity (EC) for this altered orientation is understudied. Here, we aimed to explore altered intrinsic functional connectivity (FC) and EC in PD patients with probable RBD (pRBD). Methods This was a cross-sectional study. A total of 31 PD patients with pRBD (PD-pRBD), 35 PD without pRBD (PD-npRBD), and 32 healthy controls (HCs) underwent resting-state functional magnetic resonance imaging (RS-fMRI) scans. The voxel-wise degree centrality (DC) calculation was first performed to investigate the inherent connectivity of the PD-pRBD patients. Subsequently, we applied Granger causality analysis (GCA) to probe the causal effects of anomalous brain regions. Finally, the support vector machine (SVM) method was executed to evaluate the DC values in identifying PD-pRBD. Results PD-pRBD patients exhibited reduced z-DC values in the right precentral gyrus relative to PD-npRBD (voxel-level P<0.001, cluster-level P<0.05), as well as decreased z-DC values in the right postcentral gyrus and the superior parietal lobule compared to HCs. Then, our GCA revealed that decreased EC was located predominantly from the right precentral gyrus to the right caudate nucleus in the PD-pRBD group. Additionally, the SVM results revealed that the z-DC values of the right precentral gyrus could discriminate PD-pRBD from the PD-npRBD group [area under the curve (AUC) =0.905]. Conclusions The altered z-DC in the right precentral gyrus and the anomaly causal effects from the precentral motor cortex to the ipsilateral striatum represented by the caudate nucleus might play vital roles in the pathogenesis of PD-pRBD. It was speculated that the attenuation of FC from the precentral motor cortex to the subcortical striatum might be associated with nocturnal muscle dyskinesia and behavioral abnormalities in PD-pRBD patients. This disruption pattern may be a prospective imaging marker in the characterization of PD with pRBD.
Collapse
Affiliation(s)
- Ai-Di Shan
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Heng Zhang
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Meng-Xi Gao
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Li-Na Wang
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xing-Yue Cao
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Cai-Ting Gan
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hui-Min Sun
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qian-Ling Lu
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Li Zhang
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yong-Sheng Yuan
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ke-Zhong Zhang
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
3
|
Beretta VS, Orcioli-Silva D, Zampier VC, Moraca GAG, Pereira MP, Gobbi LTB, Vitório R. Eight sessions of transcranial electrical stimulation for postural response in people with Parkinson's disease: A randomized trial. Gait Posture 2024; 114:1-7. [PMID: 39197335 DOI: 10.1016/j.gaitpost.2024.08.076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 09/01/2024]
Abstract
BACKGROUND Impairments in postural responses to perturbation are common in people with Parkinson's disease (PwPD) and lack effective treatment. We recently showed that a single session of transcranial direct current stimulation (tDCS) promotes acute improvement of postural response to perturbation in PwPD. However, the effects of multiple tDCS sessions remain unclear. RESEARCH QUESTION What is the efficacy of eight sessions of anodal tDCS on postural responses to external perturbation in PwPD? METHODS Twenty-two PwPD participated in this randomized, double-blind, parallel-arm, and sham-controlled study. Participants were randomly distributed into active (a-tDCS; n=11) or sham stimulation (s-tDCS; n=11). Eight tDCS sessions were applied over the primary motor cortex (M1), with the a-tDCS group receiving 2 mA for 20 minutes. Postural responses to external perturbations were assessed before, 48 hours after, and one month after (follow-up) the completion of tDCS sessions. Primary outcome measures included the onset latency of medial gastrocnemius (MG) muscle and range of center of pressure. Secondary outcomes included electromyography and CoP parameters, and prefrontal cortex (PFC) activity. RESULTS ANOVA revealed a trend for Group*Moment interaction for MG onset latency (p=0.058). a-tDCS tended to have shorter MG onset latency at post-test (p=0.040; SRM = -0.63) compared to pre-test. For the secondary outcomes, only a-tDCS decreased the time taken to recover balance after the perturbation at post-test and follow-up compared to pre-test (both p<0.001; SRM=-1.42 and -1.53, respectively). Also, only a-tDCS demonstrated lower PFC activity at post-test compared to pre-test (p=0.017; SRM = -0.82) and follow-up (p=0.001). SIGNIFICANCE Eight sessions of tDCS over M1 improved postural response to perturbation in PwPD. Some benefits lasted for at least a month. Neuromuscular and behavioral changes observed after the intervention were accompanied by decreased PFC activity (executive-attentional control), suggesting that tDCS applied over M1 can improve movement automaticity.
Collapse
Affiliation(s)
- Victor Spiandor Beretta
- São Paulo State University (Unesp), Institute of Biosciences, Graduate Program in Movement Sciences, Posture and Gait Studies Laboratory (LEPLO), Rio Claro, Brazil; São Paulo State University (Unesp), School of Technology and Sciences, Physical Education Department, Presidente Prudente, Brazil.
| | - Diego Orcioli-Silva
- São Paulo State University (Unesp), Institute of Biosciences, Graduate Program in Movement Sciences, Posture and Gait Studies Laboratory (LEPLO), Rio Claro, Brazil; University of Campinas (UNICAMP), School of Applied Sciences (FCA), Laboratory of Applied Sport Physiology (LAFAE), Limeira, Brazil
| | - Vinicius Cavassano Zampier
- São Paulo State University (Unesp), Institute of Biosciences, Graduate Program in Movement Sciences, Posture and Gait Studies Laboratory (LEPLO), Rio Claro, Brazil
| | - Gabriel Antonio Gazziero Moraca
- São Paulo State University (Unesp), Institute of Biosciences, Graduate Program in Movement Sciences, Posture and Gait Studies Laboratory (LEPLO), Rio Claro, Brazil
| | - Marcelo Pinto Pereira
- São Paulo State University (Unesp), Institute of Biosciences, Graduate Program in Movement Sciences, Posture and Gait Studies Laboratory (LEPLO), Rio Claro, Brazil
| | - Lilian Teresa Bucken Gobbi
- São Paulo State University (Unesp), Institute of Biosciences, Graduate Program in Movement Sciences, Posture and Gait Studies Laboratory (LEPLO), Rio Claro, Brazil
| | - Rodrigo Vitório
- Department of Sport, Exercise and Rehabilitation, Northumbria University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
4
|
Bastos-Gonçalves R, Coimbra B, Rodrigues AJ. The mesopontine tegmentum in reward and aversion: From cellular heterogeneity to behaviour. Neurosci Biobehav Rev 2024; 162:105702. [PMID: 38718986 DOI: 10.1016/j.neubiorev.2024.105702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/06/2024] [Accepted: 05/01/2024] [Indexed: 05/18/2024]
Abstract
The mesopontine tegmentum, comprising the pedunculopontine tegmentum (PPN) and the laterodorsal tegmentum (LDT), is intricately connected to various regions of the basal ganglia, motor systems, and limbic systems. The PPN and LDT can regulate the activity of different brain regions of these target systems, and in this way are in a privileged position to modulate motivated behaviours. Despite recent findings, the PPN and LDT have been largely overlooked in discussions about the neural circuits associated with reward and aversion. This review aims to provide a timely and comprehensive resource on past and current research, highlighting the PPN and LDT's connectivity and influence on basal ganglia and limbic, and motor systems. Seminal studies, including lesion, pharmacological, and optogenetic/chemogenetic approaches, demonstrate their critical roles in modulating reward/aversive behaviours. The review emphasizes the need for further investigation into the associated cellular mechanisms, in order to clarify their role in behaviour and contribution for different neuropsychiatric disorders.
Collapse
Affiliation(s)
- Ricardo Bastos-Gonçalves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Bárbara Coimbra
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Ana João Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
5
|
Watanabe H, Dijkstra JM, Nagatsu T. Parkinson's Disease: Cells Succumbing to Lifelong Dopamine-Related Oxidative Stress and Other Bioenergetic Challenges. Int J Mol Sci 2024; 25:2009. [PMID: 38396687 PMCID: PMC10888576 DOI: 10.3390/ijms25042009] [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: 12/29/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
The core pathological event in Parkinson's disease (PD) is the specific dying of dopamine (DA) neurons of the substantia nigra pars compacta (SNc). The reasons why SNc DA neurons are especially vulnerable and why idiopathic PD has only been found in humans are still puzzling. The two main underlying factors of SNc DA neuron vulnerability appear related to high DA production, namely (i) the toxic effects of cytoplasmic DA metabolism and (ii) continuous cytosolic Ca2+ oscillations in the absence of the Ca2+-buffer protein calbindin. Both factors cause oxidative stress by producing highly reactive quinones and increasing intra-mitochondrial Ca2+ concentrations, respectively. High DA expression in human SNc DA neuron cell bodies is suggested by the abundant presence of the DA-derived pigment neuromelanin, which is not found in such abundance in other species and has been associated with toxicity at higher levels. The oxidative stress created by their DA production system, despite the fact that the SN does not use unusually high amounts of energy, explains why SNc DA neurons are sensitive to various genetic and environmental factors that create mitochondrial damage and thereby promote PD. Aging increases multiple risk factors for PD, and, to a large extent, PD is accelerated aging. To prevent PD neurodegeneration, possible approaches that are discussed here are (1) reducing cytoplasmic DA accumulation, (2) blocking cytoplasmic Ca2+ oscillations, and (3) providing bioenergetic support.
Collapse
Affiliation(s)
- Hirohisa Watanabe
- Department of Neurology, School of Medicine, Fujita Health University, Toyoake 470-1192, Aichi, Japan
| | - Johannes M. Dijkstra
- Center for Medical Science, Fujita Health University, Toyoake 470-1192, Aichi, Japan
| | - Toshiharu Nagatsu
- Center for Research Promotion and Support, Fujita Health University, Toyoake 470-1192, Aichi, Japan;
| |
Collapse
|
6
|
Nordengen K, Morland C. From Synaptic Physiology to Synaptic Pathology: The Enigma of α-Synuclein. Int J Mol Sci 2024; 25:986. [PMID: 38256059 PMCID: PMC10815905 DOI: 10.3390/ijms25020986] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/06/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Alpha-synuclein (α-syn) has gained significant attention due to its involvement in neurodegenerative diseases, particularly Parkinson's disease. However, its normal function in the human brain is equally fascinating. The α-syn protein is highly dynamic and can adapt to various conformational stages, which differ in their interaction with synaptic elements, their propensity to drive pathological aggregation, and their toxicity. This review will delve into the multifaceted role of α-syn in different types of synapses, shedding light on contributions to neurotransmission and overall brain function. We describe the physiological role of α-syn at central synapses, including the bidirectional interaction between α-syn and neurotransmitter systems.
Collapse
Affiliation(s)
- Kaja Nordengen
- Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
| | - Cecilie Morland
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, The Faculty of Mathematics and Natural Sciences, University of Oslo, 1068 Oslo, Norway
| |
Collapse
|
7
|
Tubert C, Zampese E, Pancani T, Tkatch T, Surmeier DJ. Feed-forward metabotropic signaling by Cav1 Ca 2+ channels supports pacemaking in pedunculopontine cholinergic neurons. Neurobiol Dis 2023; 188:106328. [PMID: 37852390 PMCID: PMC10792542 DOI: 10.1016/j.nbd.2023.106328] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/14/2023] [Accepted: 10/15/2023] [Indexed: 10/20/2023] Open
Abstract
Like a handful of other neuronal types in the brain, cholinergic neurons (CNs) in the pedunculopontine nucleus (PPN) are lost during Parkinson's disease (PD). Why this is the case is unknown. One neuronal trait implicated in PD selective neuronal vulnerability is the engagement of feed-forward stimulation of mitochondrial oxidative phosphorylation (OXPHOS) to meet high bioenergetic demand, leading to sustained oxidant stress and ultimately degeneration. The extent to which this trait is shared by PPN CNs is unresolved. To address this question, a combination of molecular and physiological approaches were used. These studies revealed that PPN CNs are autonomous pacemakers with modest spike-associated cytosolic Ca2+ transients. These Ca2+ transients were partly attributable to the opening of high-threshold Cav1.2 Ca2+ channels, but not Cav1.3 channels. Cav1.2 channel signaling through endoplasmic reticulum ryanodine receptors stimulated mitochondrial OXPHOS to help maintain cytosolic adenosine triphosphate (ATP) levels necessary for pacemaking. Inhibition of Cav1.2 channels led to the recruitment of ATP-sensitive K+ channels and the slowing of pacemaking. A 'side-effect' of Cav1.2 channel-mediated stimulation of mitochondria was increased oxidant stress. Thus, PPN CNs have a distinctive physiological phenotype that shares some, but not all, of the features of other neurons that are selectively vulnerable in PD.
Collapse
Affiliation(s)
- C Tubert
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Universidad de Buenos Aires - CONICET. Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Facultad de Medicina, Departamento de Ciencias Fisiológicas, Grupo de Neurociencia de Sistemas, Buenos Aires, Argentina
| | - E Zampese
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - T Pancani
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Max Planck Florida Institute for Neuroscience, 1 Max Planck Way, Jupiter, FL 33458, USA
| | - T Tkatch
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - D J Surmeier
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA.
| |
Collapse
|
8
|
Onder H, Korkmaz B, Comoglu S. Temporal Investigations of the Changes in Presynaptic Inhibition Associated With Subthalamic Nucleus-Deep-Brain Stimulation. J Clin Neurol 2023; 19:539-546. [PMID: 37488958 PMCID: PMC10622716 DOI: 10.3988/jcn.2022.0439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/27/2023] [Accepted: 02/17/2023] [Indexed: 07/26/2023] Open
Abstract
BACKGROUND AND PURPOSE There are controversies regarding the role of presynaptic inhibition (PSI) in the mechanisms underlying the efficacy of deep-brain stimulation (DBS) in Parkinson's disease (PD). We sought to determine the involvement of PSI in DBS-related mechanisms and clinical correlates. METHODS We enrolled PD subjects who had received subthalamic nucleus DBS (STN-DBS) therapy and had been admitted to our clinic between January 2022 and March 2022. The tibial H-reflex was studied bilaterally during the medication-off state, and all tests were repeated 10 and 20 minutes after the simulation was turned off. Simultaneous evaluations based on the Movement-Disorder-Society-sponsored revision of the Unified Parkinson's Disease Rating Scale part III (MDS-UPDRS-III) were performed in all of the patients. RESULTS Ultimately we enrolled 18 patients aged 58.7±9.3 years (mean±standard deviation, 10 females). Fifty percent of the patients showed a decrease in the MDS-UPDRS-III score of more than 60% during the stimulation-on period. Comparative analyses of the repeated measurements made according to the stimulation status revealed significant differences only in the left H-reflex/M-response amplitude ratio (H/M ratio). However, no difference in the left H/M ratio was found in the subgroup of patients with a prominent clinical response to stimulation (n=9). Analyses of the less-affected side revealed differences in the H-reflex amplitude and H/M ratio. CONCLUSIONS We found evidence of PSI recovery on the less-affected side of our PD subjects associated with STN-DBS. We hypothesize that the involvement of this spinal pathway and its contribution to the mechanisms of DBS differ between individuals based on the severity of the disease and which brainstem regions and descending tracts are involved.
Collapse
Affiliation(s)
- Halil Onder
- Neurology Clinic, Diskapi Yildirim Beyazit Training and Research Hospital, Ankara, Turkey.
| | - Bektas Korkmaz
- Neurology Clinic, Diskapi Yildirim Beyazit Training and Research Hospital, Ankara, Turkey
| | - Selcuk Comoglu
- Neurology Clinic, Diskapi Yildirim Beyazit Training and Research Hospital, Ankara, Turkey
| |
Collapse
|
9
|
Deli A, Zamora M, Fleming JE, Divanbeighi Zand A, Benjaber M, Green AL, Denison T. Bioelectronic Zeitgebers: targeted neuromodulation to re-establish circadian rhythms. CONFERENCE PROCEEDINGS. IEEE INTERNATIONAL CONFERENCE ON SYSTEMS, MAN, AND CYBERNETICS 2023; 2023:2301-2308. [PMID: 38343562 PMCID: PMC7615625 DOI: 10.1109/smc53992.2023.10394632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/16/2024]
Abstract
Existing neurostimulation systems implanted for the treatment of neurodegenerative disorders generally deliver invariable therapy parameters, regardless of phase of the sleep/wake cycle. However, there is considerable evidence that brain activity in these conditions varies according to this cycle, with discrete patterns of dysfunction linked to loss of circadian rhythmicity, worse clinical outcomes and impaired patient quality of life. We present a targeted concept of circadian neuromodulation using a novel device platform. This system utilises stimulation of circuits important in sleep and wake regulation, delivering bioelectronic cues (Zeitgebers) aimed at entraining rhythms to more physiological patterns in a personalised and fully configurable manner. Preliminary evidence from its first use in a clinical trial setting, with brainstem arousal circuits as a surgical target, further supports its promising impact on sleep/wake pathology. Data included in this paper highlight its versatility and effectiveness on two different patient phenotypes. In addition to exploring acute and long-term electrophysiological and behavioural effects, we also discuss current caveats and future feature improvements of our proposed system, as well as its potential applicability in modifying disease progression in future therapies.
Collapse
Affiliation(s)
- Alceste Deli
- Functional Neurosurgery Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, UK
| | - Mayela Zamora
- Institute of Biomedical Engineering, Department of Engineering Sciences, University of Oxford, Oxford OX3 7DQ, UK
| | - John E. Fleming
- Institute of Biomedical Engineering, Department of Engineering Sciences, University of Oxford, Oxford OX3 7DQ, UK
- MRC Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Amir Divanbeighi Zand
- Functional Neurosurgery Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, UK
| | - Moaad Benjaber
- Institute of Biomedical Engineering, Department of Engineering Sciences, University of Oxford, Oxford OX3 7DQ, UK
- MRC Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Alexander L. Green
- Functional Neurosurgery Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, UK
| | - Timothy Denison
- Institute of Biomedical Engineering, Department of Engineering Sciences, University of Oxford, Oxford OX3 7DQ, UK
- MRC Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| |
Collapse
|
10
|
Hu Y, Ma TC, Alberico SL, Ding Y, Jin L, Kang UJ. Substantia Nigra Pars Reticulata Projections to the Pedunculopontine Nucleus Modulate Dyskinesia. Mov Disord 2023; 38:1850-1860. [PMID: 37461292 PMCID: PMC10932617 DOI: 10.1002/mds.29558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/02/2023] [Accepted: 06/29/2023] [Indexed: 07/21/2023] Open
Abstract
BACKGROUND Long-term use of levodopa for Parkinson's disease (PD) treatment is often hindered by development of motor complications, including levodopa-induced dyskinesia (LID). The substantia nigra pars reticulata (SNr) and globus pallidus internal segment (GPi) are the output nuclei of the basal ganglia. Dysregulation of SNr and GPi activity contributes to PD pathophysiology and LID. OBJECTIVE The objective of this study was to determine whether direct modulation of SNr GABAergic neurons and SNr projections to the pedunculopontine nucleus (PPN) regulates PD symptoms and LID in a mouse model. METHODS We expressed Cre-recombinase activated channelrhodopsin-2 (ChR2) or halorhodopsin adeno-associated virus-2 (AAV2) vectors selectively in SNr GABAergic neurons of Vgat-IRES-Cre mice in a 6-hydroxydopamine model of PD to investigate whether direct optogenetic modulation of SNr neurons or their projections to the PPN regulates PD symptoms and LID expression. The forepaw stepping task, mouse LID rating scale, and open-field locomotion were used to assess akinesia and LID to test the effect of SNr modulation. RESULTS Akinesia was improved by suppressing SNr neuron activity with halorhodopsin. LID was significantly reduced by increasing SNr neuronal activity with ChR2, which did not interfere with the antiakinetic effect of levodopa. Optical stimulation of ChR2 in SNr projections to the PPN recapitulated direct SNr stimulation. CONCLUSIONS Modulation of SNr GABAergic neurons alters akinesia and LID expression in a manner consistent with the rate model of basal ganglia circuitry. Moreover, the projections from SNr to PPN likely mediate the antidyskinetic effect of increasing SNr neuronal activity, identifying a potential novel role for the PPN in LID. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Yong Hu
- Department of Neurology, NYU Langone Health, New York, NY 10016, USA
| | - Thong C. Ma
- Department of Neurology, NYU Langone Health, New York, NY 10016, USA
| | | | - Yunmin Ding
- Department of Neurology, NYU Langone Health, New York, NY 10016, USA
| | - Lingjing Jin
- Department of Neurology and Neurological Rehabilitation, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai 200092, China
| | - Un Jung Kang
- Department of Neurology, NYU Langone Health, New York, NY 10016, USA
| |
Collapse
|
11
|
Figorilli M, Meloni M, Lanza G, Casaglia E, Lecca R, Saibene FL, Congiu P, Puligheddu M. Considering REM Sleep Behavior Disorder in the Management of Parkinson's Disease. Nat Sci Sleep 2023; 15:333-352. [PMID: 37180094 PMCID: PMC10167974 DOI: 10.2147/nss.s266071] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Rapid eye movement (REM) sleep behavior disorder (RBD) is the result of the loss of physiological inhibition of muscle tone during REM sleep, characterized by dream-enacting behavior and widely recognized as a prodromal manifestation of alpha-synucleinopathies. Indeed, patients with isolated RBD (iRBD) have an extremely high estimated risk to develop a neurodegenerative disease after a long follow up. Nevertheless, in comparison with PD patients without RBD (PDnoRBD), the occurrence of RBD in the context of PD (PDRBD) seems to identify a unique, more malignant phenotype, characterized by a more severe burden of disease in terms of both motor and non-motor symptoms and increased risk for cognitive decline. However, while some medications (eg, melatonin, clonazepam, etc.) and non-pharmacological options have been found to have some therapeutic benefits on RBD there is no available treatment able to modify the disease course or, at least, slow down the neurodegenerative process underlying phenoconversion. In this scenario, the long prodromal phase may allow an early therapeutic window and, therefore, the identification of multimodal biomarkers of disease onset and progression is becoming increasingly crucial. To date, several clinical (motor, cognitive, olfactory, visual, and autonomic features) neurophysiological, neuroimaging, biological (biofluids or tissue biopsy), and genetic biomarkers have been identified and proposed, also in combination, as possible diagnostic or prognostic markers, along with a potential role for some of them as outcome measures and index of treatment response. In this review, we provide an insight into the present knowledge on both existing and future biomarkers of iRBD and highlight the difference with PDRBD and PDnoRBD, including currently available treatment options.
Collapse
Affiliation(s)
- Michela Figorilli
- Sleep Disorder Research Center, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Mario Meloni
- IRCCS, Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| | - Giuseppe Lanza
- Department of Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
- Clinical Neurophysiology Research Unit, Oasi Research Institute-IRCCS, Troina, Italy
| | - Elisa Casaglia
- Sleep Disorder Research Center, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Rosamaria Lecca
- Sleep Disorder Research Center, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | | | - Patrizia Congiu
- Sleep Disorder Research Center, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Monica Puligheddu
- Sleep Disorder Research Center, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| |
Collapse
|
12
|
Witzig VS, Alosaimi F, Temel Y, Schulz JB, Jahanshahi A, Tan SKH. Gait improvement by high-frequency stimulation of the subthalamic nucleus in Parkinsonian mice is not associated with changes of the cholinergic system in the pedunculopontine nucleus. Neurosci Lett 2023; 802:137134. [PMID: 36801348 DOI: 10.1016/j.neulet.2023.137134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/30/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023]
Abstract
Deep brain stimulation (DBS) of the subthalamic nucleus (STN) is standard care for severe motor symptoms of Parkinson's disease (PD). However, a challenge of DBS remains improving gait. Gait has been associated with the cholinergic system in the pedunculopontine nucleus (PPN). In this study, we investigated the effects of long-term intermittent bilateral STN-DBS on PPN cholinergic neurons in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) Parkinsonian mouse model. Motor behavior, previously assessed by the automated Catwalk gait analysis, demonstrated a parkinsonian-like motor phenotype with static and dynamic gait impairments, which were reversed by STN-DBS. In this study, a subset of brains was further immunohistochemically processed for choline acetyltransferase (ChAT) and the neuronal activation marker c-Fos. MPTP treatment resulted in a significant reduction of PPN ChAT expressing neurons compared to saline treatment. STN-DBS did not alter the number of ChAT expressing neurons, nor the number of double-labelled PPN neurons for ChAT and c-Fos. Although STN-DBS improved gait in our model this was not associated with an altered expression or activation of PPN acetylcholine neurons. Motor and gait effects of STN-DBS are therefore less likely to be mediated by the STN-PPN connection and PPN cholinergic system.
Collapse
Affiliation(s)
- V S Witzig
- Department of Neurology, RWTH Aachen University, Aachen, Germany.
| | - F Alosaimi
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - Y Temel
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - J B Schulz
- Department of Neurology, RWTH Aachen University, Aachen, Germany; JARA Institute Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich and RWTH Aachen University, Aachen, Germany
| | - A Jahanshahi
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - S K H Tan
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, Netherlands; Department of Neurosurgery, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
13
|
Influence of RBD onset on the clinical characteristics of Parkinson's disease patients: a retrospective study. J Neurol 2023; 270:3171-3178. [PMID: 36897388 DOI: 10.1007/s00415-023-11659-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/27/2023] [Accepted: 03/02/2023] [Indexed: 03/11/2023]
Abstract
INTRODUCTION In Parkinson's disease (PD), rapid eye movement (REM) sleep behavior disorder (RBD) might either precede the appearance of motor symptoms, or develop during the disease course. PD patients with RBD are characterized by a higher burden of cognitive impairment and hallucinations. However, few studies have analyzed the clinical characteristics of PD patients according to the timeline of RBD onset. METHODS PD patients have been retrospectively enrolled. Presence and onset of probable RBD (pRBD) has been evaluated using RBD Screening Questionnaire (score ≥ 6). Presence of Mild Cognitive Impairment (MCI) at baseline has been evaluated using the MDS criteria level II. Presence of motor complications and hallucinations has been evaluated at a 5-year follow-up. RESULTS A total of 115 PD patients (65 men, 56.5%; mean age 62.5 ± 9.7 years; mean disease duration 3.7 ± 3.9 years) have been enrolled. Out of these, 63 fulfilled the diagnosis of pRBD (54.8%) with 21 (33.3%) reporting the RBD onset before the onset of the motor symptoms (PD-RBDpre), and 42 (66.7%) after the motor symptoms (PD-RBDpost). At enrolment presence of MCI was associated with PD-RBDpre patients (OR 5.04; 95% CI 1.33-19.05; p value 0.02). At follow-up, a higher risk of developing hallucinations was also associated with PD-RBDpre (OR 4.68; 95% CI 1.24-17.63; p = 0.022). CONCLUSIONS PD patients with RBD occurring before the onset of motor symptoms represent a subgroup of patients with a more severe cognitive phenotype and with a higher risk of developing hallucinations along the disease course, with significant implications in terms of prognostic stratification and therapeutic approach.
Collapse
|
14
|
The Pedunculopontine Tegmental Nucleus is not Important for Breathing Impairments Observed in a Parkinson's Disease Model. Neuroscience 2023; 512:32-46. [PMID: 36690033 DOI: 10.1016/j.neuroscience.2022.12.022] [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: 09/01/2022] [Revised: 12/15/2022] [Accepted: 12/20/2022] [Indexed: 01/21/2023]
Abstract
Parkinson's disease (PD) is a motor disorder resulting from degeneration of dopaminergic neurons of substantia nigra pars compacta (SNpc), with classical and non-classical symptoms such as respiratory instability. An important region for breathing control, the Pedunculopontine Tegmental Nucleus (PPTg), is composed of cholinergic, glutamatergic, and GABAergic neurons. We hypothesize that degenerated PPTg neurons in a PD model contribute to the blunted respiratory activity. Adult mice (40 males and 29 females) that express the fluorescent green protein in cholinergic, glutamatergic or GABAergic cells were used (Chat-cre Ai6, Vglut2-cre Ai6 and Vgat-cre Ai6) and received bilateral intrastriatal injections of vehicle or 6-hydroxydopamine (6-OHDA). Ten days later, the animals were exposed to hypercapnia or hypoxia to activate PPTg neurons. Vglut2-cre Ai6 animals also received retrograde tracer injections (cholera toxin b) into the retrotrapezoid nucleus (RTN) or preBötzinger Complex (preBötC) and anterograde tracer injections (AAV-mCherry) into the SNpc. In 6-OHDA-injected mice, there is a 77% reduction in the number of dopaminergic neurons in SNpc without changing the number of neurons in the PPTg. Hypercapnia activated fewer Vglut2 neurons in PD, and hypoxia did not activate PPTg neurons. PPTg neurons do not input RTN or preBötC regions but receive projections from SNpc. Although our results did not show a reduction in the number of glutamatergic neurons in PPTg, we observed a reduction in the number of neurons activated by hypercapnia in the PD animal model, suggesting that PPTg may participate in the hypercapnia ventilatory response.
Collapse
|
15
|
Gu W, Xu L, Wang J, Ou Y. Control mechanisms of pathological low-frequency oscillations under different targets in Parkinson's disease. Biomed Signal Process Control 2023. [DOI: 10.1016/j.bspc.2022.104257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
16
|
Lin C, Ridder MC, Sah P. The PPN and motor control: Preclinical studies to deep brain stimulation for Parkinson's disease. Front Neural Circuits 2023; 17:1095441. [PMID: 36925563 PMCID: PMC10011138 DOI: 10.3389/fncir.2023.1095441] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/31/2023] [Indexed: 03/04/2023] Open
Abstract
The pedunculopontine nucleus (PPN) is the major part of the mesencephalic locomotor region, involved in the control of gait and locomotion. The PPN contains glutamatergic, cholinergic, and GABAergic neurons that all make local connections, but also have long-range ascending and descending connections. While initially thought of as a region only involved in gait and locomotion, recent evidence is showing that this structure also participates in decision-making to initiate movement. Clinically, the PPN has been used as a target for deep brain stimulation to manage freezing of gait in late Parkinson's disease. In this review, we will discuss current thinking on the role of the PPN in locomotor control. We will focus on the cytoarchitecture and functional connectivity of the PPN in relationship to motor control.
Collapse
Affiliation(s)
- Caixia Lin
- Queensland Brain Institute, University of Queensland, St Lucia, QLD, Australia.,Joint Centre for Neuroscience and Neural Engineering, and Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Margreet C Ridder
- Queensland Brain Institute, University of Queensland, St Lucia, QLD, Australia
| | - Pankaj Sah
- Queensland Brain Institute, University of Queensland, St Lucia, QLD, Australia.,Joint Centre for Neuroscience and Neural Engineering, and Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| |
Collapse
|
17
|
Carvajal-Oliveros A, Dominguez-Baleón C, Sánchez-Díaz I, Zambrano-Tipan D, Hernández-Vargas R, Campusano JM, Narváez-Padilla V, Reynaud E. Parkinsonian phenotypes induced by Synphilin-1 expression are differentially contributed by serotonergic and dopaminergic circuits and suppressed by nicotine treatment. PLoS One 2023; 18:e0282348. [PMID: 36857384 PMCID: PMC9977059 DOI: 10.1371/journal.pone.0282348] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 02/13/2023] [Indexed: 03/02/2023] Open
Abstract
Synphilin-1 is a protein encoded by the human SNCAIP gene whose function has yet to be fully understood. However, it has been linked to familial Parkinson's disease (PD). Synphilin-1 is a major component of the Lewy bodies found in neurons in the substantia nigra pars compacta of PD patients. Synphilin-1 expression in serotonergic and/or dopaminergic neurons of Drosophila melanogaster induces neurodegeneration, as well as motor and non-motor PD like symptoms. In this work, we examined the contribution of the serotonergic and dopaminergic circuits in the development of PD-like phenotypes. We found that olfactory and visual symptoms are majorly contributed by the serotonergic system, and that motor symptoms and reduction in survival are mainly contributed by the dopaminergic system. Chronic nicotine treatment was able to suppress several of these symptoms. These results indicate that both the serotonergic and dopaminergic systems contribute to different aspects of PD symptomatology and that nicotine has beneficial effects on specific symptoms.
Collapse
Affiliation(s)
- Angel Carvajal-Oliveros
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, México
| | - Carmen Dominguez-Baleón
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, México
| | - Iván Sánchez-Díaz
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, México
| | - Diego Zambrano-Tipan
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, México
| | - René Hernández-Vargas
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, México
| | - Jorge M. Campusano
- Facultad de Ciencias Biológicas, Departamento de Biología Celular y Molecular, Laboratorio Neurogenética de la Conducta, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Verónica Narváez-Padilla
- Centro de Investigación en Dinámica Celular, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, México
| | - Enrique Reynaud
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, México
- * E-mail:
| |
Collapse
|
18
|
Su JH, Hu YW, Yang Y, Li RY, Teng F, Li LX, Jin LJ. Dystonia and the pedunculopontine nucleus: Current evidences and potential mechanisms. Front Neurol 2022; 13:1065163. [PMID: 36504662 PMCID: PMC9727297 DOI: 10.3389/fneur.2022.1065163] [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: 10/09/2022] [Accepted: 11/08/2022] [Indexed: 11/24/2022] Open
Abstract
Being a major component of the midbrain locomotion region, the pedunculopontine nucleus (PPN) is known to have various connections with the basal ganglia, the cerebral cortex, thalamus, and motor regions of the brainstem and spinal cord. Functionally, the PPN is associated with muscle tone control and locomotion modulation, including motor initiation, rhythm and speed. In addition to its motor functions, the PPN also contribute to level of arousal, attention, memory and learning. Recent studies have revealed neuropathologic deficits in the PPN in both patients and animal models of dystonia, and deep brain stimulation of the PPN also showed alleviation of axial dystonia in patients of Parkinson's disease. These findings indicate that the PPN might play an important role in the development of dystonia. Moreover, with increasing preclinical evidences showed presence of dystonia-like behaviors, muscle tone changes, impaired cognitive functions and sleep following lesion or neuromodulation of the PPN, it is assumed that the pathological changes of the PPN might contribute to both motor and non-motor manifestations of dystonia. In this review, we aim to summarize the involvement of the PPN in dystonia based on the current preclinical and clinical evidences. Moreover, potential mechanisms for its contributions to the manifestation of dystonia is also discussed base on the dystonia-related basal ganglia-cerebello-thalamo-cortical circuit, providing fundamental insight into the targeting of the PPN for the treatment of dystonia in the future.
Collapse
Affiliation(s)
- Jun-hui Su
- Department of Neurology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China,Department of Neurology and Neurological Rehabilitation, Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Yao-wen Hu
- Department of Neurology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yi Yang
- Department of Neurology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ruo-yu Li
- Department of Neurology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fei Teng
- Department of Neurology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Li-xi Li
- Department of Neurology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ling-jing Jin
- Department of Neurology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China,Department of Neurology and Neurological Rehabilitation, Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China,*Correspondence: Ling-jing Jin
| |
Collapse
|
19
|
Increased Expression of Alpha-, Beta-, and Gamma-Synucleins in Brainstem Regions of a Non-Human Primate Model of Parkinson’s Disease. Int J Mol Sci 2022; 23:ijms23158586. [PMID: 35955716 PMCID: PMC9369189 DOI: 10.3390/ijms23158586] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 12/02/2022] Open
Abstract
Parkinson’s disease (PD) is characterized by cell loss in the substantia nigra and the presence of alpha-synuclein (α-syn)-containing neuronal Lewy bodies. While α-syn has received major interest in the pathogenesis of PD, the function of beta- and gamma-synucleins (β-syn and γ-syn, respectively) is not really known. Yet, these proteins are members of the same family and also concentrated in neuronal terminals. The current preclinical study investigated the expression levels of α-, β-, and γ-synucleins in brainstem regions involved in PD physiopathology. We analyzed synuclein expression in the substantia nigra, raphe nuclei, pedunculopontine nucleus, and locus coeruleus from control and parkinsonian (by MPTP) macaques. MPTP-intoxicated monkeys developed a more or less severe parkinsonian score and were sacrificed after a variable post-MPTP period ranging from 1 to 20 months. The expression of the three synucleins was increased in the substantia nigra after MPTP, and this increase correlates positively, although not very strongly, with cell loss and motor score and not with the time elapsed after intoxication. In the dorsal raphe nucleus, the expression of the three synucleins was also increased, but only α- and γ-Syn are linked to the motor score and associated cell loss. Finally, although no change in synuclein expression was demonstrated in the locus coeruleus after MPTP, we found increased expression levels of γ-Syn, which are only correlated with cell loss in the pedunculopontine nucleus. Altogether, our data suggest that these proteins may play a key role in brainstem regions and mesencephalic tegmentum. Given the involvement of these brain regions in non-motor symptoms of PD, these data also strengthen the relevance of the MPTP macaque model of PD, which exhibits pathological changes beyond nigral DA cell loss and α-synucleinopathy.
Collapse
|
20
|
Radulovic J, Ivkovic S, Adzic M. From chronic stress and anxiety to neurodegeneration: Focus on neuromodulation of the axon initial segment. HANDBOOK OF CLINICAL NEUROLOGY 2022; 184:481-495. [PMID: 35034756 DOI: 10.1016/b978-0-12-819410-2.00025-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
To adapt to the sustained demands of chronic stress, discrete brain circuits undergo structural and functional changes often resulting in anxiety disorders. In some individuals, anxiety disorders precede the development of motor symptoms of Parkinson's disease (PD) caused by degeneration of neurons in the substantia nigra (SN). Here, we present a circuit framework for probing a causal link between chronic stress, anxiety, and PD, which postulates a central role of abnormal neuromodulation of the SN's axon initial segment by brainstem inputs. It is grounded in findings demonstrating that the earliest PD pathologies occur in the stress-responsive, emotion regulation network of the brainstem, which provides the SN with dense aminergic and cholinergic innervation. SN's axon initial segment (AIS) has unique features that support the sustained and bidirectional propagation of activity in response to synaptic inputs. It is therefore, especially sensitive to circuit-mediated stress-induced imbalance of neuromodulation, and thus a plausible initiating site of neurodegeneration. This could explain why, although secondary to pathophysiologies in other brainstem nuclei, SN degeneration is the most extensive. Consequently, the cardinal symptom of PD, severe motor deficits, arise from degeneration of the nigrostriatal pathway rather than other brainstem nuclei. Understanding when and how circuit dysfunctions underlying anxiety can progress to neurodegeneration, raises the prospect of timed interventions for reversing, or at least impeding, the early pathophysiologies that lead to PD and possibly other neurodegenerative disorders.
Collapse
Affiliation(s)
- Jelena Radulovic
- Department of Neuroscience, Albert Einstein Medical College, Bronx, NY, United States; Department of Psychiatry and Behavioral Sciences, Albert Einstein Medical College, Bronx, NY, United States.
| | - Sanja Ivkovic
- Department of Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Miroslav Adzic
- Department of Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
21
|
Insola A, Mazzone P, Della Marca G, Capozzo A, Vitale F, Scarnati E. Pedunculopontine tegmental Nucleus-evoked prepulse inhibition of the blink reflex in Parkinson's disease. Clin Neurophysiol 2021; 132:2729-2738. [PMID: 34417108 DOI: 10.1016/j.clinph.2021.06.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To investigate the effects on the blink reflex (BR) of single stimuli applied to the pedunculopontine tegmental nucleus (PPTg). METHODS The BR was evoked by stimulating the supraorbital nerve (SON) in fifteen patients suffering from idiopathic Parkinson's disease (PD) who had electrodes monolaterally or bilaterally implanted in the PPTg for deep brain stimulation (DBS). Single stimuli were delivered to the PPTg through externalized electrode connection wires 3-4 days following PPTg implantation. RESULTS PPTg stimuli increased the latency and reduced duration, amplitude and area of the R2 component of the BR in comparison to the response recorded in the absence of PPTg stimulation. These effects were independent of the side of SON stimulation and were stable for interstimulus interval (ISI) between PPTg prepulse and SON stimulus from 0 to 110 ms. The PPTg-induced prepulse inhibition of the BR was bilaterally present in the brainstem. The R1 component was unaffected. CONCLUSIONS The prepulse inhibition of the R2 component may be modulated by the PPTg. SIGNIFICANCE These findings suggest that abnormalities of BR occurring in PD may be ascribed to a reduction of basal ganglia-mediated inhibition of brainstem excitability.
Collapse
Affiliation(s)
- Angelo Insola
- Clinical Neurophysiopathology, CTO Andrea Alesini Hospital, ASL Roma 2, Via San Nemesio 21, 00145 Rome, Italy.
| | - Paolo Mazzone
- Functional and Stereotactic Neurosurgery, CTO Andrea Alesini Hospital, ASL Roma 2, Via San Nemesio 21, 00145 Rome, Italy
| | - Giacomo Della Marca
- Institute of Neurology, Catholic University, Largo A.Gemelli 8, 00168 Rome, Italy
| | - Annamaria Capozzo
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L'Aquila, Via Vetoio Coppito 2, 67100 L'Aquila, Italy
| | - Flora Vitale
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L'Aquila, Via Vetoio Coppito 2, 67100 L'Aquila, Italy
| | - Eugenio Scarnati
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L'Aquila, Via Vetoio Coppito 2, 67100 L'Aquila, Italy
| |
Collapse
|
22
|
Bayasgalan T, Stupniki S, Kovács A, Csemer A, Szentesi P, Pocsai K, Dionisio L, Spitzmaul G, Pál B. Alteration of Mesopontine Cholinergic Function by the Lack of KCNQ4 Subunit. Front Cell Neurosci 2021; 15:707789. [PMID: 34381336 PMCID: PMC8352570 DOI: 10.3389/fncel.2021.707789] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 06/28/2021] [Indexed: 11/16/2022] Open
Abstract
The pedunculopontine nucleus (PPN), a structure known as a cholinergic member of the reticular activating system (RAS), is source and target of cholinergic neuromodulation and contributes to the regulation of the sleep–wakefulness cycle. The M-current is a voltage-gated potassium current modulated mainly by cholinergic signaling. KCNQ subunits ensemble into ion channels responsible for the M-current. In the central nervous system, KCNQ4 expression is restricted to certain brainstem structures such as the RAS nuclei. Here, we investigated the presence and functional significance of KCNQ4 in the PPN by behavioral studies and the gene and protein expressions and slice electrophysiology using a mouse model lacking KCNQ4 expression. We found that this mouse has alterations in the adaptation to changes in light–darkness cycles, representing the potential role of KCNQ4 in the regulation of the sleep–wakefulness cycle. As cholinergic neurons from the PPN participate in the regulation of this cycle, we investigated whether the cholinergic PPN might also possess functional KCNQ4 subunits. Although the M-current is an electrophysiological hallmark of cholinergic neurons, only a subpopulation of them had KCNQ4-dependent M-current. Interestingly, the absence of the KCNQ4 subunit altered the expression patterns of the other KCNQ subunits in the PPN. We also determined that, in wild-type animals, the cholinergic inputs of the PPN modulated the M-current, and these in turn can modulate the level of synchronization between neighboring PPN neurons. Taken together, the KCNQ4 subunit is present in a subpopulation of PPN cholinergic neurons, and it may contribute to the regulation of the sleep–wakefulness cycle.
Collapse
Affiliation(s)
- T Bayasgalan
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - S Stupniki
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional Del Sur (UNS), Bahía Blanca, Argentina.,Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, Bahía Blanca, Argentina
| | - A Kovács
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - A Csemer
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - P Szentesi
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - K Pocsai
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - L Dionisio
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional Del Sur (UNS), Bahía Blanca, Argentina.,Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, Bahía Blanca, Argentina
| | - G Spitzmaul
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional Del Sur (UNS), Bahía Blanca, Argentina.,Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, Bahía Blanca, Argentina
| | - B Pál
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
23
|
Rodríguez MÁ, Crespo I, Del Valle M, Olmedillas H. Home-Based Vigorous Tele-Exercise in People with Parkinson's Disease: Feasibility Beyond Complexity. JOURNAL OF PARKINSONS DISEASE 2021; 11:843-845. [PMID: 33554926 DOI: 10.3233/jpd-212541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
| | - Irene Crespo
- Department of Functional Biology, Universidad de Oviedo, Oviedo, Spain.,Institute of Biomedicine, Universidad de León, León, Spain
| | - Miguel Del Valle
- Department of Cellular Morphology and Biology, Universidad de Oviedo, Oviedo, Spain
| | - Hugo Olmedillas
- Department of Functional Biology, Universidad de Oviedo, Oviedo, Spain.,Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Spain
| |
Collapse
|
24
|
The correlation of non-motor symptoms and sleep on balance in Parkinson's disease patients with normal cognition and mild cognitive impairment. Ir J Med Sci 2021; 190:1577-1584. [PMID: 33449322 DOI: 10.1007/s11845-020-02462-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/03/2020] [Indexed: 10/22/2022]
Abstract
BACKGROUND Parkinson's disease (PD) is characterized by non-motor symptoms (NMS) as well as by motor symptoms. Together with the impairment of cognitive functions, NMS and sleep also affect motor symptoms negatively. The aim of our study is to examine the correlation of NMS and sleep on balance in PD patients with normal cognition (PD-NC) and with mild cognitive impairment (PD-MCI). METHODS A total of 69 patients were included in our study. Using the Standardized Mini-Mental State Examination, participants were divided into 2 groups, PD-NC and PD-MCI. Patients were assessed with the Unified Parkinson's Disease Rating Scale (UPDRS), the Berg Balance Scale (BBS), the Tinetti Balance Assessment Tool (TBAT), the Non-Motor Symptoms Questionnaire (NMSQ), and the Parkinson's Disease Questionnaire (PDQ-39). RESULTS PD-MCI patients had statistically significant worse motor symptoms and more balance disorder compared to PD-NC (UPDRS: p = 0.009; BBS: p = 0.010; TBAT: p = 0.004). PD-MCI patients had greater severity of non-motor symptoms and worse sleep quality than the PD-NC group (NMSQ-total: p = 0.02; NMSQ-sleep total: p = 0.01). The evaluation has shown that with a diagnosis of MCI, NMS, and sleeping problems were correlated, and the correlation was associated with impairment of the balance function. While being more pronounced in the PD-MCI group, quality of life was affected in both groups (p < 0.05). CONCLUSION Our data demonstrate a negative effect on the balance function in patients with cognitive impairment suffering increased NMS and sleeping disorders. Treatment of these patients needs to concentrate on NMS and cognitive functions as much as on motor symptoms.
Collapse
|
25
|
Chambers NE, Coyle M, Sergio J, Lanza K, Saito C, Topping B, Clark SD, Bishop C. Effects of pedunculopontine nucleus cholinergic lesion on gait and dyskinesia in hemiparkinsonian rats. Eur J Neurosci 2021; 53:2835-2847. [PMID: 33426708 DOI: 10.1111/ejn.15106] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/02/2021] [Accepted: 01/04/2021] [Indexed: 11/30/2022]
Abstract
Pedunculopontine nucleus (PPN) cholinergic neurons are implicated in freezing of gait in Parkinson's disease (PD) and motor stereotypy in normal animals, but the causal role of these neurons on specific gait parameters and treatment-induced dyskinesia remains speculative. Therefore, we examined whether selective cholinergic lesion of the rostral PPN affects PD motor and gait deficits, L-DOPA-induced dyskinesia and motor improvement, and DA-agonist-induced dyskinesia. Sprague-Dawley rats were assigned to one unilaterally lesioned group: Sham lesion, PPN cholinergic lesion with diphtheria urotensin II fusion toxin, medial forebrain bundle dopamine lesion with 6-hydroxydopamine, or dual acetylcholine and dopamine lesion. We used gait analysis and forepaw adjusting steps to examine PD gait and motor deficits. Forepaw adjusting steps were also used to assess motor improvement with L-DOPA treatment. The abnormal involuntary movements scale measured L-DOPA and dopamine D1- and D2-receptor agonist-induced dyskinesia. Lesions, verified via tyrosine hydroxylase and choline acetyltransferase immunohistochemistry reduced an average of 95% of nigral dopamine neurons and 80% of PPN cholinergic neurons, respectively. Rats receiving acetylcholine and dual lesion demonstrated enhanced freezing, and acetylcholine lesioned rats exhibited increased print area and stand index. Dopamine and dual lesion produced similar forepaw adjusting steps task on and off L-DOPA. Relative to DA lesioned rats, dual lesioned rats displayed reduced L-DOPA and DA agonist-induced dyskinesia at specific time points. Our results indicate that PPN cholinergic neurons affect gait parameters related to postural stability. Therefore, therapeutically targeting PPN cholinergic neurons could reduce intractable postural instability in PD without affecting motor benefits or side effects of L-DOPA treatment.
Collapse
Affiliation(s)
- Nicole E Chambers
- Department of Psychology, Behavioral Neuroscience Program, Binghamton University, Binghamton, NY, USA
| | - Michael Coyle
- Department of Psychology, Behavioral Neuroscience Program, Binghamton University, Binghamton, NY, USA
| | - Jordan Sergio
- Department of Psychology, Behavioral Neuroscience Program, Binghamton University, Binghamton, NY, USA
| | - Kathryn Lanza
- Department of Psychology, Behavioral Neuroscience Program, Binghamton University, Binghamton, NY, USA
| | - Carolyn Saito
- Department of Psychology, Behavioral Neuroscience Program, Binghamton University, Binghamton, NY, USA
| | - Brent Topping
- Department of Psychology, Behavioral Neuroscience Program, Binghamton University, Binghamton, NY, USA
| | - Stewart D Clark
- Department of Pharmacology and Toxicology, Jacobs School of Medicine & Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Christopher Bishop
- Department of Psychology, Behavioral Neuroscience Program, Binghamton University, Binghamton, NY, USA
| |
Collapse
|
26
|
Taximaimaiti R, Luo X, Wang XP. Pharmacological and Non-pharmacological Treatments of Sleep Disorders in Parkinson's Disease. Curr Neuropharmacol 2021; 19:2233-2249. [PMID: 33998990 PMCID: PMC9185775 DOI: 10.2174/1570159x19666210517115706] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 11/22/2022] Open
Abstract
Sleep disorders are one of the most common non-motor symptoms in Parkinson's disease (PD). It can cause a notable decrease in quality of life and functioning in PD patients, as well as place a huge burden on both patients and caregivers. The most cited sleep disorders in PD included insomnia, restless legs syndrome (RLS), rapid eye movement (REM), sleep behavior disorders (RBD), excessive daytime sleepiness (EDS) and sleep disordered breathing (SDB), which can appear alone or several at the same time. In this review, we listed the recommended pharmacological treatments for common sleep disorders in PD, and discussed the recommended dosages, benefits and side effects of relative drugs. We also discussed non-pharmacological treatments to improve sleep quality, including sleep hygiene education, exercise, deep brain stimulation, cognitive behavior therapy and complementary therapies. We tried to find proper interventions for different types of sleep disorders in PD, while minimizing relative side effects.
Collapse
Affiliation(s)
| | | | - Xiao-Ping Wang
- Address correspondence to this author at the Department of Neurology, Shanghai TongRen Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China; Tel: +86-021-52039999-72223; Fax: +86-021-52039999-72223; E-mail:
| |
Collapse
|
27
|
Calderón-Garcidueñas L, Torres-Solorio AK, Kulesza RJ, Torres-Jardón R, González-González LO, García-Arreola B, Chávez-Franco DA, Luévano-Castro SC, Hernández-Castillo A, Carlos-Hernández E, Solorio-López E, Crespo-Cortés CN, García-Rojas E, Mukherjee PS. Gait and balance disturbances are common in young urbanites and associated with cognitive impairment. Air pollution and the historical development of Alzheimer's disease in the young. ENVIRONMENTAL RESEARCH 2020; 191:110087. [PMID: 32890478 PMCID: PMC7467072 DOI: 10.1016/j.envres.2020.110087] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 05/03/2023]
Abstract
To determine whether gait and balance dysfunction are present in young urbanites exposed to fine particular matter PM2.5 ≥ annual USEPA standard, we tested gait and balance with Tinetti and Berg tests in 575 clinically healthy subjects, age 21.0 ± 5.7 y who were residents in Metropolitan Mexico City, Villahermosa and Reynosa. The Montreal Cognitive Assessment was also applied to an independent cohort n:76, age 23.3 ± 9.1 y. In the 575 cohort, 75.4% and 34.4% had abnormal total Tinetti and Berg scores and high risk of falls in 17.2% and 5.7% respectively. BMI impacted negatively Tinetti and Berg performance. Gait dysfunction worsen with age and males performed worse than females. Gait and balance dysfunction were associated with mild cognitive impairment MCI (19.73%) and dementia (55.26%) in 57/76 and 19 cognitively intact subjects had gait and balance dysfunction. Seventy-five percent of urbanites exposed to PM2.5 had gait and balance dysfunction. For MMC residents-with historical documented Alzheimer disease (AD) and CSF abnormalities, these findings suggest Alzheimer Continuum is in progress. Early development of a Motoric Cognitive Risk Syndrome ought to be considered in city dwellers with normal cognition and gait dysfunction. The AD research frame in PM2.5 exposed young urbanites should include gait and balance measurements. Multicity teens and young adult cohorts are warranted for quantitative gait and balance measurements and neuropsychological and brain imaging studies in high vs low PM2.5 exposures. Early identification of gait and balance impairment in young air pollution-exposed urbanites would facilitate multidisciplinary prevention efforts for modifying the course of AD.
Collapse
Affiliation(s)
| | | | - Randy J Kulesza
- Auditory Research Center, Lake Erie College of Osteopathic Medicine, Erie, PA, 16509, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Rodríguez MÁ, Albillos-Almaraz L, López-Aguado I, Crespo I, Del Valle M, Olmedillas H. Vigorous Aerobic Exercise in the Management of Parkinson Disease: A Systematic Review. PM R 2020; 13:890-900. [PMID: 32978867 DOI: 10.1002/pmrj.12500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 09/02/2020] [Accepted: 09/09/2020] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To summarize the findings from studies examining the effects of vigorous-intensity aerobic exercise in the management of Parkinson disease. TYPE: Systematic review. LITERATURE SURVEY PubMed/MEDLINE, EMBASE, Scopus, Web of Science, Cochrane Library, SPORTDiscus, and ScienceDirect databases were searched up to May 2020. Reference lists of the included articles were also searched for additional studies. Searches were restricted to English language. METHODOLOGY Seven papers, including six studies, five randomized controlled trials and one controlled trial, were identified. The studies examined the effects of vigorous-intensity aerobic exercise in participants with Parkinson disease. Studies in which the minimal intensity required was ≥77% of maximum heart rate, 60% of heart rate reserve or 64% of maximal oxygen uptake met the inclusion criteria. Method appraisal showed a mean score of 5.3 in the Physiotherapy Evidence Database (PEDro) scale. SYNTHESIS No statistically significant differences were found between vigorous-intensity aerobic exercise and moderate/low-intensity aerobic exercise for the main outcomes (disease severity and motor function). Only one study concluded a significant higher aerobic fitness in favor of the group that exercised at vigorous intensity compared to the moderate intensity group. CONCLUSIONS Vigorous-intensity aerobic exercise has not shown statistically significant improvements in motor and nonmotor impairments in individuals with Parkinson disease as compared to moderate/low-intensity aerobic exercise. Hence, the current evidence is too limited to allow recommendations for clinical practice.
Collapse
Affiliation(s)
| | | | | | - Irene Crespo
- Department of Functional Biology, Universidad de Oviedo, Oviedo, Spain.,Institute of Biomedicine, Universidad de León, León, Spain
| | - Miguel Del Valle
- Department of Cellular Morphology and Biology, Universidad de Oviedo, Oviedo, Spain
| | - Hugo Olmedillas
- Department of Functional Biology, Universidad de Oviedo, Oviedo, Spain.,Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Spain
| |
Collapse
|
29
|
Petrovic J, Radovanovic L, Saponjic J. Prodromal local sleep disorders in a rat model of Parkinson's disease cholinopathy, hemiparkinsonism and hemiparkinsonism with cholinopathy. Behav Brain Res 2020; 397:112957. [PMID: 33038348 DOI: 10.1016/j.bbr.2020.112957] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 10/02/2020] [Accepted: 10/03/2020] [Indexed: 01/14/2023]
Abstract
We investigated the prodromal alterations of local sleep, particularly the motor cortical and hippocampal sleep, along with spontaneous locomotor activity in the rat models of Parkinson's disease (PD). We performed our experiments in adult, male Wistar rats, chronically implanted for sleep recording and divided into four experimental groups: the control (implanted controls), the bilateral pedunculopontine tegmental nucleus (PPT) lesions (PD cholinopathy), the unilateral substantia nigra pars compacta (SNpc) lesions (hemiparkinsonism) and the unilateral SNpc/bilateral PPT lesions (hemiparkinsonism with PD cholinopathy). We followed their sleep, basal locomotor activity and spatial habituation for 14 days following the surgical procedures. Severe prodromal local sleep disturbances in the hemiparkinsonian rats were expressed as sleep fragmentation and distinct local NREM/REM EEG microstructure alterations in both the motor cortex and the hippocampus. Alongside the state-unrelated role of the dopaminergic control of theta oscillations and NREM/REM related sigma and beta oscillations, we demonstrated that the REM neurochemical regulatory substrate is particularly important in the dopaminergic control of beta oscillations. In addition, hippocampal prodromal sleep disorders in the hemiparkinsonian rats were expressed as NREM/REM fragmentation and the opposite impact of dopaminergic versus cholinergic control of the NREM delta and beta oscillation amplitudes in the hippocampus, likewise in the motor cortex versus the hippocampus. All these distinct prodromal local sleep disorders and the dopaminergic vs. cholinergic impact on NREM/REM EEG microstructure alterations are of fundamental importance for the further development and follow-up of PD-modifying therapies, and for the identification of patients who are at risk of developing PD.
Collapse
Affiliation(s)
- Jelena Petrovic
- Institute for Biological Research, Sinisa Stankovic - National Institute of Republic of Serbia, Department of Neurobiology, University of Belgrade, Despot Stefan Blvd., 142, 11060, Belgrade, Serbia.
| | - Ljiljana Radovanovic
- Institute for Biological Research, Sinisa Stankovic - National Institute of Republic of Serbia, Department of Neurobiology, University of Belgrade, Despot Stefan Blvd., 142, 11060, Belgrade, Serbia
| | - Jasna Saponjic
- Institute for Biological Research, Sinisa Stankovic - National Institute of Republic of Serbia, Department of Neurobiology, University of Belgrade, Despot Stefan Blvd., 142, 11060, Belgrade, Serbia
| |
Collapse
|
30
|
Troncoso-Escudero P, Sepulveda D, Pérez-Arancibia R, Parra AV, Arcos J, Grunenwald F, Vidal RL. On the Right Track to Treat Movement Disorders: Promising Therapeutic Approaches for Parkinson's and Huntington's Disease. Front Aging Neurosci 2020; 12:571185. [PMID: 33101007 PMCID: PMC7497570 DOI: 10.3389/fnagi.2020.571185] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/17/2020] [Indexed: 12/17/2022] Open
Abstract
Movement disorders are neurological conditions in which patients manifest a diverse range of movement impairments. Distinct structures within the basal ganglia of the brain, an area involved in movement regulation, are differentially affected for every disease. Among the most studied movement disorder conditions are Parkinson's (PD) and Huntington's disease (HD), in which the deregulation of the movement circuitry due to the loss of specific neuronal populations in basal ganglia is the underlying cause of motor symptoms. These symptoms are due to the loss principally of dopaminergic neurons of the substantia nigra (SN) par compacta and the GABAergic neurons of the striatum in PD and HD, respectively. Although these diseases were described in the 19th century, no effective treatment can slow down, reverse, or stop disease progression. Available pharmacological therapies have been focused on preventing or alleviating motor symptoms to improve the quality of life of patients, but these drugs are not able to mitigate the progressive neurodegeneration. Currently, considerable therapeutic advances have been achieved seeking a more efficacious and durable therapeutic effect. Here, we will focus on the new advances of several therapeutic approaches for PD and HD, starting with the available pharmacological treatments to alleviate the motor symptoms in both diseases. Then, we describe therapeutic strategies that aim to restore specific neuronal populations or their activity. Among the discussed strategies, the use of Neurotrophic factors (NTFs) and genetic approaches to prevent the neuronal loss in these diseases will be described. We will highlight strategies that have been evaluated in both Parkinson's and Huntington's patients, and also the ones with strong preclinical evidence. These current therapeutic techniques represent the most promising tools for the safe treatment of both diseases, specifically those aimed to avoid neuronal loss during disease progression.
Collapse
Affiliation(s)
- Paulina Troncoso-Escudero
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health, and Metabolism, University of Chile, Santiago, Chile
| | - Denisse Sepulveda
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health, and Metabolism, University of Chile, Santiago, Chile
| | - Rodrigo Pérez-Arancibia
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health, and Metabolism, University of Chile, Santiago, Chile
| | - Alejandra V. Parra
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health, and Metabolism, University of Chile, Santiago, Chile
| | - Javiera Arcos
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health, and Metabolism, University of Chile, Santiago, Chile
| | - Felipe Grunenwald
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health, and Metabolism, University of Chile, Santiago, Chile
| | - Rene L. Vidal
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health, and Metabolism, University of Chile, Santiago, Chile
| |
Collapse
|
31
|
Antonyová V, Kejík Z, Brogyányi T, Kaplánek R, Pajková M, Talianová V, Hromádka R, Masařík M, Sýkora D, Mikšátková L, Martásek P, Jakubek M. Role of mtDNA disturbances in the pathogenesis of Alzheimer's and Parkinson's disease. DNA Repair (Amst) 2020; 91-92:102871. [PMID: 32502755 DOI: 10.1016/j.dnarep.2020.102871] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/08/2020] [Accepted: 05/09/2020] [Indexed: 12/11/2022]
Abstract
Neurodegenerative diseases (e.g. Alzheimer's and Parkinson's disease) are becoming increasingly problematic to healthcare systems. Therefore, their underlying mechanisms are trending topics of study in medicinal research. Numerous studies have evidenced a strong association between mitochondrial DNA disturbances (e.g. oxidative damage, mutations, and methylation shifts) and the initiation and progression of neurodegenerative diseases. Therefore, this review discusses the risk and development of neurodegenerative diseases in terms of disturbances in mitochondrial DNA and as a part of a complex ecosystem that includes other important mechanisms (e.g. neuroinflammation and the misfolding and aggregation of amyloid-β peptides, α-synuclein, and tau proteins). In addition, the influence of individual mitochondrial DNA haplogroups on the risk and development of neurodegenerative diseases is also described and discussed.
Collapse
Affiliation(s)
- Veronika Antonyová
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Kateřinská 32, 121 08 Prague 2, Czech Republic
| | - Zdeněk Kejík
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Kateřinská 32, 121 08 Prague 2, Czech Republic; Department of Analytical Chemistry, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Dejvice, Czech Republic
| | - Tereza Brogyányi
- Depertment of Pathological Physiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 2, 121 00 Prague 2, Czech Republic
| | - Robert Kaplánek
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Kateřinská 32, 121 08 Prague 2, Czech Republic; BIOCEV, First Faculty of Medicine, Charles University, Kateřinská 32, 121 08 Prague 2, Czech Republic; Department of Analytical Chemistry, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Dejvice, Czech Republic
| | - Martina Pajková
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Kateřinská 32, 121 08 Prague 2, Czech Republic; BIOCEV, First Faculty of Medicine, Charles University, Kateřinská 32, 121 08 Prague 2, Czech Republic
| | - Veronika Talianová
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Kateřinská 32, 121 08 Prague 2, Czech Republic; BIOCEV, First Faculty of Medicine, Charles University, Kateřinská 32, 121 08 Prague 2, Czech Republic
| | - Róbert Hromádka
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Kateřinská 32, 121 08 Prague 2, Czech Republic
| | - Michal Masařík
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Kateřinská 32, 121 08 Prague 2, Czech Republic; BIOCEV, First Faculty of Medicine, Charles University, Kateřinská 32, 121 08 Prague 2, Czech Republic
| | - David Sýkora
- BIOCEV, First Faculty of Medicine, Charles University, Kateřinská 32, 121 08 Prague 2, Czech Republic; Department of Analytical Chemistry, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Dejvice, Czech Republic
| | - Lucie Mikšátková
- BIOCEV, First Faculty of Medicine, Charles University, Kateřinská 32, 121 08 Prague 2, Czech Republic; Department of Analytical Chemistry, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Dejvice, Czech Republic
| | - Pavel Martásek
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Kateřinská 32, 121 08 Prague 2, Czech Republic.
| | - Milan Jakubek
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Kateřinská 32, 121 08 Prague 2, Czech Republic; BIOCEV, First Faculty of Medicine, Charles University, Kateřinská 32, 121 08 Prague 2, Czech Republic; Department of Analytical Chemistry, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Dejvice, Czech Republic.
| |
Collapse
|