1
|
Priyanka, Qamar SH, Visanji NP. Toward an animal model of Progressive Supranuclear Palsy. Front Neurosci 2024; 18:1433465. [PMID: 39420986 PMCID: PMC11484047 DOI: 10.3389/fnins.2024.1433465] [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: 05/15/2024] [Accepted: 08/19/2024] [Indexed: 10/19/2024] Open
Abstract
Progressive Supranuclear Palsy (PSP) is a rare and fatal neurodegenerative tauopathy which, with a rapid clinical progression coupled to a strong degree of clinico-pathologic correlation, has been suggested to be a "frontrunner" in translational development for neurodegenerative proteinopathies. Elegant studies in animals have contributed greatly to our understanding of disease pathogenesis in PSP. However, presently no animal model replicates the key anatomical and cytopathologic hallmarks, the spatiotemporal spread of pathology, progressive neurodegeneration, or locomotor and cognitive symptoms that characterize PSP. Current models therefore likely fail to recapitulate the key mechanisms that underly the pathological progression of PSP, impeding their translational value. Here we review what we have learned about PSP from work in animals to date, examine the gaps in modeling the disease and discuss strategies for the development of refined animal models that will improve our understanding of disease pathogenesis and provide a critical platform for the testing of novel therapeutics for this devastating disease.
Collapse
Affiliation(s)
- Priyanka
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Syeda Hania Qamar
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Naomi P. Visanji
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Rossy Progressive Supranuclear Palsy Centre, Toronto Western Hospital, Toronto, ON, Canada
| |
Collapse
|
2
|
Lu X, Wickens JR, Hyland BI. Multimodal convergence in the pedunculopontine tegmental nucleus: Motor, sensory and theta-frequency inputs influence activity of single neurons. Eur J Neurosci 2024; 60:3643-3658. [PMID: 38698531 DOI: 10.1111/ejn.16367] [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: 11/29/2022] [Accepted: 04/03/2024] [Indexed: 05/05/2024]
Abstract
The pedunculopontine tegmental nucleus of the brainstem (PPTg) has extensive interconnections and neuronal-behavioural correlates. It is implicated in movement control and sensorimotor integration. We investigated whether single neuron activity in freely moving rats is correlated with components of skilled forelimb movement, and whether individual neurons respond to both motor and sensory events. We found that individual PPTg neurons showed changes in firing rate at different times during the reach. This type of temporally specific modulation is like activity seen elsewhere in voluntary movement control circuits, such as the motor cortex, and suggests that PPTg neural activity is related to different specific events occurring during the reach. In particular, many neuronal modulations were time-locked to the end of the extension phase of the reach, when fine distal movements related to food grasping occur, indicating strong engagement of PPTg in this phase of skilled individual forelimb movements. In addition, some neurons showed brief periods of apparent oscillatory firing in the theta range at specific phases of the reach-to-grasp movement. When movement-related neurons were tested with tone stimuli, many also responded to this auditory input, allowing for sensorimotor integration at the cellular level. Together, these data extend the concept of the PPTg as an integrative structure in generation of complex movements, by showing that this function extends to the highly coordinated control of the forelimb during skilled reach to grasp movement, and that sensory and motor-related information converges on single neurons, allowing for direct integration at the cellular level.
Collapse
Affiliation(s)
- Xiaodong Lu
- Department of Physiology, School of Biomedical Sciences and Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- Brain Research New Zealand Centre of Research Excellence, Dunedin and Auckland, New Zealand
| | - Jeffery R Wickens
- Okinawa Institute for Science and Technology Graduate University, Okinawa, Japan
| | - Brian Ian Hyland
- Department of Physiology, School of Biomedical Sciences and Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- Brain Research New Zealand Centre of Research Excellence, Dunedin and Auckland, New Zealand
| |
Collapse
|
3
|
Ryczko D. The Mesencephalic Locomotor Region: Multiple Cell Types, Multiple Behavioral Roles, and Multiple Implications for Disease. Neuroscientist 2024; 30:347-366. [PMID: 36575956 PMCID: PMC11107129 DOI: 10.1177/10738584221139136] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The mesencephalic locomotor region (MLR) controls locomotion in vertebrates. In humans with Parkinson disease, locomotor deficits are increasingly associated with decreased activity in the MLR. This brainstem region, commonly considered to include the cuneiform and pedunculopontine nuclei, has been explored as a target for deep brain stimulation to improve locomotor function, but the results are variable, from modest to promising. However, the MLR is a heterogeneous structure, and identification of the best cell type to target is only beginning. Here, I review the studies that uncovered the role of genetically defined MLR cell types, and I highlight the cells whose activation improves locomotor function in animal models of Parkinson disease. The promising cell types to activate comprise some glutamatergic neurons in the cuneiform and caudal pedunculopontine nuclei, as well as some cholinergic neurons of the pedunculopontine nucleus. Activation of MLR GABAergic neurons should be avoided, since they stop locomotion or evoke bouts flanked with numerous stops. MLR is also considered a potential target in spinal cord injury, supranuclear palsy, primary progressive freezing of gait, or stroke. Better targeting of the MLR cell types should be achieved through optimized deep brain stimulation protocols, pharmacotherapy, or the development of optogenetics for human use.
Collapse
Affiliation(s)
- Dimitri Ryczko
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Canada
- Centre de recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Canada
- Neurosciences Sherbrooke, Sherbrooke, Canada
- Institut de Pharmacologie de Sherbrooke, Sherbrooke, Canada
| |
Collapse
|
4
|
Miller LR, Bickel MA, Tarantini S, Runion ME, Matacchiera Z, Vance ML, Hibbs C, Vaden H, Nagykaldi D, Martin T, Bullen EC, Pinckard J, Kiss T, Howard EW, Yabluchanskiy A, Conley SM. IGF1R deficiency in vascular smooth muscle cells impairs myogenic autoregulation and cognition in mice. Front Aging Neurosci 2024; 16:1320808. [PMID: 38425784 PMCID: PMC10902040 DOI: 10.3389/fnagi.2024.1320808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/22/2024] [Indexed: 03/02/2024] Open
Abstract
Introduction Cerebrovascular pathologies contribute to cognitive decline during aging, leading to vascular cognitive impairment and dementia (VCID). Levels of circulating insulin-like growth factor 1 (IGF-1), a vasoprotective hormone, decrease during aging. Decreased circulating IGF-1 in animal models leads to the development of VCID-like symptoms, but the cellular mechanisms underlying IGF-1-deficiency associated pathologies in the aged cerebrovasculature remain poorly understood. Here, we test the hypothesis that vascular smooth muscle cells (VSMCs) play an integral part in mediating the vasoprotective effects of IGF-1. Methods We used a hypertension-based model of cerebrovascular dysfunction in mice with VSMC-specific IGF-1 receptor (Igf1r) deficiency and evaluated the development of cerebrovascular pathologies and cognitive dysfunction. Results VSMC-specific Igf1r deficiency led to impaired cerebral myogenic autoregulation, independent of blood pressure changes, which was also associated with impaired spatial learning and memory function as measured by radial arm water maze and impaired motor learning measured by rotarod. In contrast, VSMC-specific IGF-1 receptor knockdown did not lead to cerebral microvascular rarefaction. Discussion These studies suggest that VSMCs are key targets for IGF-1 in the context of cerebrovascular health, playing a role in vessel stability alongside other cells in the neurovascular unit, and that VSMC dysfunction in aging likely contributes to VCID.
Collapse
Affiliation(s)
- Lauren R. Miller
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Marisa A. Bickel
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Megan E. Runion
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Zoe Matacchiera
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Michaela L. Vance
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Clara Hibbs
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Hannah Vaden
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Domonkos Nagykaldi
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Teryn Martin
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Elizabeth C. Bullen
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Jessica Pinckard
- Division of Comparative Medicine, Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Tamas Kiss
- Pediatric Center, Semmelweis University, Budapest, Hungary
- Eötvös Loránd Research Network and Semmelweis University Cerebrovascular and Neurocognitive Disorders Research Group, Budapest, Hungary
| | - Eric W. Howard
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Shannon M. Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
5
|
Su JH, Hu YW, Song YP, Yang Y, Li RY, Zhou KG, Hu L, Wan XH, Teng F, Jin LJ. Dystonia-like behaviors and impaired sensory-motor integration following neurotoxic lesion of the pedunculopontine tegmental nucleus in mice. Front Neurol 2023; 14:1102837. [PMID: 37064180 PMCID: PMC10101329 DOI: 10.3389/fneur.2023.1102837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 03/02/2023] [Indexed: 04/18/2023] Open
Abstract
Introduction The pedunculopontine nucleus (PPTg) is a vital interface between the basal ganglia and cerebellum, participating in modulation of the locomotion and muscle tone. Pathological changes of the PPTg have been reported in patients and animal models of dystonia, while its effect and mechanism on the phenotyping of dystonia is still unknown. Methods In this study, a series of behavioral tests focusing on the specific deficits of dystonia were conducted for mice with bilateral and unilateral PPTg excitotoxic lesion, including the dystonia-like movements evaluation, different types of sensory-motor integrations, explorative behaviors and gait. In addition, neural dysfunctions including apoptosis, neuroinflammation, neurodegeneration and neural activation of PPTg-related motor areas in the basal ganglia, reticular formations and cerebellum were also explored. Results Both bilateral and unilateral lesion of the PPTg elicited dystonia-like behaviors featured by the hyperactivity of the hindlimb flexors. Moreover, proprioceptive and auditory sensory-motor integrations were impaired in bilaterally lesioned mice, while no overt alterations were found for the tactile sensory-motor integration, explorative behaviors and gait. Similar but milder behavioral deficits were found in the unilaterally lesioned mice, with an effective compensation was observed for the auditory sensory-motor integration. Histologically, no neural loss, apoptosis, neuroinflammation and neurodegeneration were found in the substantia nigra pars compacta and caudate putamen (CPu) following PPTg lesion, while reduced neural activity was found in the dorsolateral part of the CPu and striatal indirect pathway-related structures including subthalamic nucleus, globus pallidus internus and substantia nigra pars reticular. Moreover, the neural activity was decreased for the reticular formations such as pontine reticular nucleus, parvicellular reticular nucleus and gigantocellular reticular nucleus, while deep cerebellar nuclei were spared. Conclusion In conclusion, lesion of the PPTg could elicit dystonia-like behaviors through its effect on the balance of the striatal pathways and the reticular formations.
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
| | - Yun-Ping Song
- Department of Neurology and Neurological Rehabilitation, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), 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
| | - Kai-Ge Zhou
- Department of Neurology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ling Hu
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
| | - Xin-Hua Wan
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Fei Teng
- Department of Neurology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Fei Teng
| | - Ling-Jing Jin
- Department of Neurology and Neurological Rehabilitation, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
- Ling-Jing Jin
| |
Collapse
|
6
|
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
|
7
|
Cholinesterase inhibitors for gait, balance, and fall in Parkinson disease: a meta-analysis. NPJ Parkinsons Dis 2021; 7:103. [PMID: 34824258 PMCID: PMC8617004 DOI: 10.1038/s41531-021-00251-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 11/04/2021] [Indexed: 11/08/2022] Open
Abstract
Gait disturbance and imbalance are the major symptoms of Parkinson disease (PD), with fall being the most undesirable consequence. However, few effective evidence-based treatments are available for alleviating these symptoms and preventing falls. Cholinesterase inhibitors (ChEIs) are a well-established treatment for PD dementia with possible impacts on gait, balance, and fall reduction. The present study involved a meta-analysis of randomized controlled trials (RCTs) to investigate the effects of ChEIs on gait, balance, and fall in patients with PD. We searched for studies using the PubMed, Embase, and Web of Science databases. The major outcomes were effects on gait parameters, balance, and fall. This study was registered with PROSPERO (CRD42021254733). Five RCTs were included in the present meta-analysis. ChEIs did not significantly increase gait speed in PD patients (mean difference [MD]: 0.03 m/s, 95% confidence interval [CI]: -0.02 to 0.07, p = 0.29). However, ChEI treatment significantly decreased step or stride variability during the single task (standard MD: -0.43, 95% CI = -0.79 to -0.06, p = 0.02). Regarding fall and balance, trending but nonsignificant beneficial effects were observed with ChEI treatment. In conclusion, although ChEI treatment did not significantly improve gait speed and reduce fall, it can significantly reduce step or stride variability. Considering that gait disorder is a challenging issue in patients with PD and that ChEIs are generally tolerable, the present meta-analysis may provide more evidence for the benefit of ChEIs on PD gait disturbance as an alternative treatment consideration.
Collapse
|
8
|
Özkan M, Köse B, Algın O, Oğuz S, Erden ME, Çavdar S. Non-motor connections of the pedunculopontine nucleus of the rat and human brain. Neurosci Lett 2021; 767:136308. [PMID: 34715273 DOI: 10.1016/j.neulet.2021.136308] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 10/21/2021] [Accepted: 10/21/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION The connections of the pedunculopontine nucleus (PPN) with motor areas of the central nervous system (CNS) are well described in the literature, in contrast relations with non-motor areas are lacking. Thus, the aim of the present study is to define the non-motor connections of the PPN in rats using the fluoro-gold (FG) tracer and compare the presence of these connections in healthy human adults using diffusion tensor tractography (DTI). MATERIALS AND METHODS We injected FG into the PPN of 12 rats. The non-motor connections of the PPN with cortical, subcortical, and brainstem structures were documented. The non-motor connections of the rats were compared with the DTI obtained from 35 healthy adults. RESULTS The results of the tract-tracing study in the rat showed that the PPN was connected to non-motor cortical (cingulate, somatosensory, visual, auditory, medial frontal cortices), subcortical (amygdala, hypothalamus, thalamus, habenular, and bed nucleus of stria terminalis), and brainstem (medullary reticular, trigeminal spinal, external cuneate, pontine reticular, vestibular, superior and inferior colliculus, locus ceruleus, periaqueductal gray, parabrachial, dorsal raphe, pretectal, lateral lemniscus nuclei, and the contralateral PPN) structures. The DTI obtained from healthy adults showed similar PPN non-motor connections as in rats. CONCLUSION Understanding the connections of the PPN with non-motor cortical, subcortical, and brainstem areas of the CNS will enrich our knowledge of its contribution in various circuits and the areas that PPN activity can influence. Further, it will provide insight into the role of Parkinson's disease and related disorders and explain the non-motor complications which occur subsequent to deep brain stimulation (DBS) of the PPN.
Collapse
Affiliation(s)
- Mazhar Özkan
- Department of Anatomy, Tekirdağ Namık Kemal University, School of Medicine, Istanbul, Turkey
| | - Büşra Köse
- Department of Anatomy, Koç University, School of Medicine, Rumelifener Yolu, Istanbul, Turkey
| | - Oktay Algın
- Department of Radiology, City Hospital, Yıldırım Beyazıt University, Ankara, Turkey and National MR Research Center (UMRAM), Bilkent University, Ankara, Turkey
| | - Sinem Oğuz
- Department of Anatomy, Koç University, School of Medicine, Rumelifener Yolu, Istanbul, Turkey
| | - Mert Emre Erden
- Department of Anatomy, Koç University, School of Medicine, Rumelifener Yolu, Istanbul, Turkey
| | - Safiye Çavdar
- Department of Anatomy, Koç University, School of Medicine, Rumelifener Yolu, Istanbul, Turkey.
| |
Collapse
|
9
|
King G, Veros KM, MacLaren DAA, Leigh MPK, Spernyak JA, Clark SD. Human wildtype tau expression in cholinergic pedunculopontine tegmental neurons is sufficient to produce PSP-like behavioural deficits and neuropathology. Eur J Neurosci 2021; 54:7688-7709. [PMID: 34668254 DOI: 10.1111/ejn.15496] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/30/2021] [Accepted: 10/12/2021] [Indexed: 11/30/2022]
Abstract
Progressive Supranuclear Palsy (PSP) is the most common atypical parkinsonism and exhibits hallmark symptomology including motor function impairment and dysexecutive dementia. In contrast to Parkinson's disease, the underlying pathology displays aggregation of the protein tau, which is also seen in disorders such as Alzheimer's disease. Currently, there are no pharmacological treatments for PSP, and drug discovery efforts are hindered by the lack of an animal model specific to PSP. Based on previous results and clinical pathology, it was hypothesized that viral deposition of tau in cholinergic neurons within the hindbrain would produce a tauopathy along neural connections to produce PSP-like symptomology and pathology. By using a combination of ChAT-CRE rats and CRE-dependent AAV vectors, wildtype human tau (the PSP-relevant 1N4R isoform; hTau) was expressed in hindbrain cholinergic neurons. Compared to control subjects (GFP), rats with tau expression displayed deficits in a variety of behavioural paradigms: acoustic startle reflex, marble burying, horizontal ladder and hindlimb motor reflex. Postmortem, the hTau rats had significantly reduced number of cholinergic pedunculopontine tegmentum and dopaminergic substantia nigra neurons, as well as abnormal tau deposits. This preclinical model has multiple points of convergence with the clinical features of PSP, some of which distinguish between PSP and Parkinson's disease.
Collapse
Affiliation(s)
- Gabriella King
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
| | - Kaliana M Veros
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
| | | | | | - Joseph A Spernyak
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Stewart D Clark
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
| |
Collapse
|
10
|
Dautan D, Kovács A, Bayasgalan T, Diaz-Acevedo MA, Pal B, Mena-Segovia J. Modulation of motor behavior by the mesencephalic locomotor region. Cell Rep 2021; 36:109594. [PMID: 34433068 PMCID: PMC8641693 DOI: 10.1016/j.celrep.2021.109594] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 05/05/2021] [Accepted: 08/02/2021] [Indexed: 11/09/2022] Open
Abstract
The mesencephalic locomotor region (MLR) serves as an interface between higher-order motor systems and lower motor neurons. The excitatory module of the MLR is composed of the pedunculopontine nucleus (PPN) and the cuneiform nucleus (CnF), and their activation has been proposed to elicit different modalities of movement. However, how the differences in connectivity and physiological properties explain their contributions to motor activity is not well known. Here we report that CnF glutamatergic neurons are more electrophysiologically homogeneous than PPN neurons and have mostly short-range connectivity, whereas PPN glutamatergic neurons are heterogeneous and maintain long-range connections, most notably with the basal ganglia. Optogenetic activation of CnF neurons produces short-lasting muscle activation, driving involuntary motor activity. In contrast, PPN neuron activation produces long-lasting increases in muscle tone that reduce motor activity and disrupt gait. Our results highlight biophysical and functional attributes among MLR neurons that support their differential contribution to motor behavior. Dautan et al. show key differences in the connectivity and physiological properties of neurons of the mesencephalic locomotor region. Although activation of CnF neurons elicits involuntary locomotor responses, activation of PPN neurons increases muscle tone and reduces motor activity, suggesting that PPN encodes a readiness signal that precedes locomotion.
Collapse
Affiliation(s)
- Daniel Dautan
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, NJ 07102, USA.
| | - Adrienn Kovács
- Department of Physiology, University of Debrecen, Faculty of Medicine, 4012 Debrecen, Hungary
| | - Tsogbadrakh Bayasgalan
- Department of Physiology, University of Debrecen, Faculty of Medicine, 4012 Debrecen, Hungary
| | - Miguel A Diaz-Acevedo
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, NJ 07102, USA
| | - Balazs Pal
- Department of Physiology, University of Debrecen, Faculty of Medicine, 4012 Debrecen, Hungary
| | - Juan Mena-Segovia
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, NJ 07102, USA.
| |
Collapse
|
11
|
Paparella G, Fasano A, Hallett M, Berardelli A, Bologna M. Emerging concepts on bradykinesia in non-parkinsonian conditions. Eur J Neurol 2021; 28:2403-2422. [PMID: 33793037 DOI: 10.1111/ene.14851] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/24/2021] [Accepted: 03/29/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND PURPOSE Bradykinesia is one of the cardinal motor symptoms of Parkinson's disease. However, clinical and experimental studies indicate that bradykinesia may also be observed in various neurological diseases not primarily characterized by parkinsonism. These conditions include hyperkinetic movement disorders, such as dystonia, chorea, and essential tremor. Bradykinesia may also be observed in patients with neurological conditions that are not seen as "movement disorders," including those characterized by the involvement of the cerebellum and corticospinal system, dementia, multiple sclerosis, and psychiatric disorders. METHODS We reviewed clinical reports and experimental studies on bradykinesia in non-parkinsonian conditions and discussed the major findings. RESULTS Bradykinesia is a common motor abnormality in non-parkinsonian conditions. From a pathophysiological standpoint, bradykinesia in neurological conditions not primarily characterized by parkinsonism may be explained by brain network dysfunction. CONCLUSION In addition to the pathophysiological implications, the present paper highlights important terminological issues and the need for a new, more accurate, and more widely used definition of bradykinesia in the context of movement disorders and other neurological conditions.
Collapse
Affiliation(s)
| | - Alfonso Fasano
- Edmond J. Safra Program in Parkinson's Disease, Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada.,Division of Neurology, University of Toronto, Toronto, Ontario, Canada.,Krembil Brain Institute, Toronto, Ontario, Canada
| | - Mark Hallett
- Human Motor Control Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Alfredo Berardelli
- IRCCS Neuromed, Pozzilli, Italy.,Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | - Matteo Bologna
- IRCCS Neuromed, Pozzilli, Italy.,Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
12
|
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: 3] [Impact Index Per Article: 1.0] [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
|
13
|
Motor Coordination Disorders Evaluated through the Grid Test and Changes in the Nigral Nrf2 mRNA Expression in Rats with Pedunculopontine Lesion. Behav Sci (Basel) 2020; 10:bs10100156. [PMID: 33066049 PMCID: PMC7600924 DOI: 10.3390/bs10100156] [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: 08/04/2020] [Revised: 09/22/2020] [Accepted: 09/30/2020] [Indexed: 11/16/2022] Open
Abstract
Neurotoxic lesion of the pedunculopontine nucleus (PPN) is known to cause subtle motor dysfunctions. However, motor coordination during advance on a discontinuous and elevated surface has not been studied. It is also not known whether there are changes in the mRNA expression of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) in nigral tissue. Methods: The effects of the unilateral neurotoxic lesion of the PPN in motor coordination evaluated through grid test and Nrf2 mRNA expression in nigral tissue were evaluated. Two experimental designs (ED) were organized: ED#1 behavioral study (7 and 30 days after PPN lesion) and ED#2 molecular biology study (24 h, 48 h and 7 days) after PPN lesion. Results: ED#1—The number of faults made with left limbs, were significant higher in the lesioned groups (p < 0.01) both 7 and 30 days post-lesion. The number of failures made by the right limbs, was also significantly higher (p < 0.05) vs. control groups. ED#2—Nrf2 mRNA expression showed an increase 24 h after PPN injury (p < 0.01), followed by a peak of expression 48 h post injury (p < 0.001). Conclusions: Disorders of motor coordination associated with PPN injury are bilateral. The increased Nrf2 mRNA expression could represent an adaptive response to oxidative stress in the nigral tissue following pontine injury.
Collapse
|
14
|
Li HQ, Spitzer NC. Exercise enhances motor skill learning by neurotransmitter switching in the adult midbrain. Nat Commun 2020; 11:2195. [PMID: 32366867 PMCID: PMC7198516 DOI: 10.1038/s41467-020-16053-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 04/07/2020] [Indexed: 02/07/2023] Open
Abstract
Physical exercise promotes motor skill learning in normal individuals and those with neurological disorders but its mechanism of action is unclear. We find that one week of voluntary wheel running enhances the acquisition of motor skills in normal adult mice. One week of running also induces switching from ACh to GABA expression in neurons in the caudal pedunculopontine nucleus (cPPN). Consistent with regulation of motor skills, we show that the switching neurons make projections to the substantia nigra (SN), ventral tegmental area (VTA) and ventrolateral-ventromedial nuclei of the thalamus (VL-VM). Use of viral vectors to override transmitter switching blocks the beneficial effect of running on motor skill learning. We suggest that neurotransmitter switching provides the basis by which sustained running benefits motor skill learning, presenting a target for clinical treatment of movement disorders.
Collapse
Affiliation(s)
- Hui-Quan Li
- Neurobiology Section, Division of Biological Sciences and Center for Neural Circuits and Behavior, La Jolla, CA, 92093-0357, USA.
- Kavli Institute for Brain and Mind, University of California San Diego, La Jolla, CA, 92093-0357, USA.
| | - Nicholas C Spitzer
- Neurobiology Section, Division of Biological Sciences and Center for Neural Circuits and Behavior, La Jolla, CA, 92093-0357, USA.
- Kavli Institute for Brain and Mind, University of California San Diego, La Jolla, CA, 92093-0357, USA.
| |
Collapse
|
15
|
Cholinergic midbrain afferents modulate striatal circuits and shape encoding of action strategies. Nat Commun 2020; 11:1739. [PMID: 32269213 PMCID: PMC7142106 DOI: 10.1038/s41467-020-15514-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 03/13/2020] [Indexed: 02/06/2023] Open
Abstract
Assimilation of novel strategies into a consolidated action repertoire is a crucial function for behavioral adaptation and cognitive flexibility. Acetylcholine in the striatum plays a pivotal role in such adaptation, and its release has been causally associated with the activity of cholinergic interneurons. Here we show that the midbrain, a previously unknown source of acetylcholine in the striatum, is a major contributor to cholinergic transmission in the striatal complex. Neurons of the pedunculopontine and laterodorsal tegmental nuclei synapse with striatal cholinergic interneurons and give rise to excitatory responses. Furthermore, they produce uniform inhibition of spiny projection neurons. Inhibition of acetylcholine release from midbrain terminals in the striatum impairs the association of contingencies and the formation of habits in an instrumental task, and mimics the effects observed following inhibition of acetylcholine release from striatal cholinergic interneurons. These results suggest the existence of two hierarchically-organized modes of cholinergic transmission in the striatum, where cholinergic interneurons are modulated by cholinergic neurons of the midbrain.
Collapse
|
16
|
Bradykinesia in Alzheimer’s disease and its neurophysiological substrates. Clin Neurophysiol 2020; 131:850-858. [DOI: 10.1016/j.clinph.2019.12.413] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 12/11/2019] [Accepted: 12/29/2019] [Indexed: 01/15/2023]
|
17
|
Ettaro R, Markovic T, Daniels D, MacLaren DA, Clark SD. Microinjection of urotensin II into the pedunculopontine tegmentum leads to an increase in the consumption of sweet tastants. Physiol Behav 2020; 215:112775. [PMID: 31843472 DOI: 10.1016/j.physbeh.2019.112775] [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: 10/31/2019] [Revised: 12/02/2019] [Accepted: 12/11/2019] [Indexed: 11/29/2022]
Abstract
The pedunculopontine tegmentum (PPTg) plays a role in processing multiple sensory inputs and innervates brain regions associated with reward-related behaviors. The urotensin II receptor, activated by the urotensin II peptide (UII), is selectively expressed by the cholinergic neurons of the PPTg. Although the exact function of cholinergic neurons of the PPTg is unknown, they are thought to contribute to the perception of reward magnitude or salience detection. We hypothesized that the activation of PPTg cholinergic neurons would alter sensory processing across multiple modalities (ex. taste and hearing). Here we had three aims: first, determine if cholinergic activation is involved in consumption behavior of palatable solutions (sucrose). Second, if so, distinguish the impact of the caloric value by using saccharin, a zero calorie sweetener. Lastly, we tested the UII-mediated effects on perception of acoustic stimuli by measuring acoustic startle reflex (ASR). Male Sprague-Dawley rats were bilaterally cannulated into the PPTg, then placed under food restriction lasting the entire consumption experiment (water ad lib.). Treatment consisted of a microinjection of either 1 μL of aCSF or 1 μL of 10 μM UII into the PPTg, and the rats were immediately given access to either sucrose or saccharin. For the remaining five days, rats were allowed one hour access per day to the same sweet solution without any further treatments. During the saccharin experiment rats were tested in a contact lickometer which recorded each individual lick to give insight into the microstructure of the consumption behavior. ASR testing consisted of a baseline (no treatment), treatment day, and two additional days (no treatment). Immediately following the microinjection of UII, consumption of both saccharin and sucrose increased compared to controls. This significant increase persisted for days after the single administration of UII, but there was no generalized arousal or increase in water consumption between testing sessions. The effects on ASR were not significant. Activating cholinergic PPTg neurons may lead to a miscalculation of the salience of external stimuli, implicating the importance of cholinergic input in modulating a variety of behaviors. The long-lasting effects seen after UII treatment support further research into the role of sensory processing on reward related-behaviors at the level of the PPTg cholinergic neurons.
Collapse
Affiliation(s)
- Robert Ettaro
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY 14214, United States
| | - Tamara Markovic
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY 14214, United States
| | - Derek Daniels
- Department of Psychology and the Center for Ingestive Behavior Research, University at Buffalo, Buffalo, NY 14214, United States
| | - Duncan Aa MacLaren
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY 14214, United States
| | - Stewart D Clark
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY 14214, United States
| |
Collapse
|
18
|
Fulop GA, Ahire C, Csipo T, Tarantini S, Kiss T, Balasubramanian P, Yabluchanskiy A, Farkas E, Toth A, Nyúl-Tóth Á, Toth P, Csiszar A, Ungvari Z. Cerebral venous congestion promotes blood-brain barrier disruption and neuroinflammation, impairing cognitive function in mice. GeroScience 2019; 41:575-589. [PMID: 31691147 PMCID: PMC6885079 DOI: 10.1007/s11357-019-00110-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 09/20/2019] [Indexed: 02/07/2023] Open
Abstract
Cognitive impairment is one of the most common co-occurring chronic conditions among elderly heart failure patients (incidence: up to ~ 80%); however, the underlying mechanisms are not completely understood. It is hypothesized that in addition to decreased cardiac output, increases in central-and consequentially, cerebral-venous pressure (backward failure) also contribute significantly to the genesis of cognitive impairment. To test this hypothesis and elucidate the specific pathogenic role of venous congestion in the brain, we have established a novel model of increased cerebral venous pressure: mice with jugular vein ligation (JVL). To test the hypothesis that increased venous pressure in the brain contributes to the development of cognitive deficits by causing blood-brain barrier disruption, dysregulation of blood flow, and/or promoting neuroinflammation, in C57BL/6 mice, the internal and external jugular veins were ligated. Cognitive function (radial arm water maze), gait function (CatWalk), and motor coordination (rotarod) were tested post-JVL. Neurovascular coupling responses were assessed by measuring changes in cerebral blood flow in the whisker barrel cortex in response to contralateral whisker stimulation by laser speckle contrast imaging through a closed cranial window. Blood-brain barrier integrity (IgG extravasation) and microglia activation (Iba1 staining) were assessed in brain slices by immunohistochemistry. Neuroinflammation-related gene expression profile was assessed by a targeted qPCR array. After jugular vein ligation, mice exhibited impaired spatial learning and memory, altered motor coordination, and impaired gait function, mimicking important aspects of altered brain function observed in human heart failure patients. JVL did not alter neurovascular coupling responses. In the brains of mice with JVL, significant extravasation of IgG was detected, indicating blood-brain barrier disruption, which was associated with histological markers of neuroinflammation (increased presence of activated microglia) and a pro-inflammatory shift in gene expression profile. Thus, cerebral venous congestion per se can cause blood-brain barrier disruption and neuroinflammation, which likely contribute to the genesis of cognitive impairment. These findings have relevance to the pathogenesis of cognitive decline associated with heart failure as well as increased cerebal venous pressure due to increased jugular venous reflux in elderly human patients.
Collapse
Affiliation(s)
- Gabor A Fulop
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience, Division of Clinical Physiology, Department of Cardiology/Kálmán Laki Doctoral School of Biomedical and Clinical Sciences, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Chetan Ahire
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
| | - Tamas Csipo
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience, Division of Clinical Physiology, Department of Cardiology/Kálmán Laki Doctoral School of Biomedical and Clinical Sciences, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine, Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine, Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Tamas Kiss
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience, Theoretical Medicine Doctoral School, Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
| | - Priya Balasubramanian
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
| | - Eszter Farkas
- International Training Program in Geroscience, Theoretical Medicine Doctoral School, Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
| | - Attila Toth
- International Training Program in Geroscience, Division of Clinical Physiology, Department of Cardiology/Kálmán Laki Doctoral School of Biomedical and Clinical Sciences, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ádám Nyúl-Tóth
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience, Institute of Biophysics, Biological Research Centre, Szeged, Hungary
| | - Peter Toth
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience, Department of Neurosurgery and Szentagothai Research Center, University of Pecs, Medical School, Pecs, Hungary
- Institute for Translational Medicine, University of Pecs, Medical School , Pecs, Hungary
| | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience, Theoretical Medicine Doctoral School, Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine, Institute of Clinical Experimental Research, Semmelweis University, Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA.
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine, Department of Public Health, Semmelweis University, Budapest, Hungary.
- International Training Program in Geroscience, Theoretical Medicine Doctoral School, Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary.
| |
Collapse
|
19
|
Hormigo S, Rodriguez-Lorenzana A, Castro-Salazar E, Millian-Morell L, López DE. Subchronic use of rivastigmine increases procognitive flexibility across multimodal behavioral tasks in healthy male rats. Behav Brain Res 2019; 376:112077. [PMID: 31499090 DOI: 10.1016/j.bbr.2019.112077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/07/2019] [Accepted: 07/07/2019] [Indexed: 01/09/2023]
Abstract
Rivastigmine (RVT) is a reversible inhibitor of cholinesterase approved worldwide for the treatment of cognitive dysfunctions, especially in Alzheimer's disease. Most previous pre-clinical studies have examined the effects of RVT treatment in a wide variety of pathological research models. Nonetheless, the effects of this drug on sensorimotor gating, memory, and learning tasks in healthy subjects remains unclear. In this study, we investigate the procognitive effects of RVT treatment in healthy rats through sensorimotor gating evaluations (measured as prepulse inhibition of the acoustic startle reflex), active avoidance learning, and spatial memory learning in a radial maze. There is an increase in the amplitude of the startle reflex in RVT-treated rats compared to the control groups, whereas the latency remained constant. Sensorimotor gating values were also incremented compared to those values from controls. In active avoidance, rats treated with RVT learned faster to successfully perform the task compared to controls, but afterwards all groups exhibited virtually identical results. During the sessions in the radial maze, RVT-treated rats committed fewer errors in both the working and reference memory compared to controls. All in all, our results support the hypothesis that RVT treatment may entail procognitive effects in healthy subjects.
Collapse
Affiliation(s)
- Sebastian Hormigo
- Institute for Neuroscience of Castilla y León (INCYL), University of Salamanca, Salamanca, Spain.
| | - Alberto Rodriguez-Lorenzana
- Institute for Neuroscience of Castilla y León (INCYL), University of Salamanca, Salamanca, Spain; Escuela de Psicologia, Universidad de Las Americas; Quito, Ecuador
| | - E Castro-Salazar
- Institute for Neuroscience of Castilla y León (INCYL), University of Salamanca, Salamanca, Spain
| | - Lymarie Millian-Morell
- Institute for Neuroscience of Castilla y León (INCYL), University of Salamanca, Salamanca, Spain; Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - Dolores E López
- Institute for Neuroscience of Castilla y León (INCYL), University of Salamanca, Salamanca, Spain; Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain; Department of Cell Biology and Pathology, University of Salamanca, Salamanca, Spain.
| |
Collapse
|
20
|
Gut NK, Mena-Segovia J. Dichotomy between motor and cognitive functions of midbrain cholinergic neurons. Neurobiol Dis 2019; 128:59-66. [PMID: 30213733 PMCID: PMC7176324 DOI: 10.1016/j.nbd.2018.09.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 07/18/2018] [Accepted: 09/09/2018] [Indexed: 12/15/2022] Open
Abstract
Cholinergic neurons of the pedunculopontine nucleus (PPN) are interconnected with all the basal ganglia structures, as well as with motor centers in the brainstem and medulla. Recent theories put into question whether PPN cholinergic neurons form part of a locomotor region that directly regulates the motor output, and rather suggest a modulatory role in adaptive behavior involving both motor and cognitive functions. In support of this, experimental studies in animals suggest that cholinergic neurons reinforce actions by signaling reward prediction and shape adaptations in behavior during changes of environmental contingencies. This is further supported by clinical studies proposing that decreased cholinergic transmission originated in the PPN is associated with impaired sensorimotor integration and perseverant behavior, giving rise to some of the symptoms observed in Parkinson's disease and progressive supranuclear palsy. Altogether, the evidence suggests that cholinergic neurons of the PPN, mainly through their interactions with the basal ganglia, have a leading role in action control.
Collapse
Affiliation(s)
- Nadine K Gut
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, NJ, USA
| | - Juan Mena-Segovia
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, NJ, USA.
| |
Collapse
|
21
|
Tarantini S, Valcarcel-Ares MN, Toth P, Yabluchanskiy A, Tucsek Z, Kiss T, Hertelendy P, Kinter M, Ballabh P, Süle Z, Farkas E, Baur JA, Sinclair DA, Csiszar A, Ungvari Z. Nicotinamide mononucleotide (NMN) supplementation rescues cerebromicrovascular endothelial function and neurovascular coupling responses and improves cognitive function in aged mice. Redox Biol 2019; 24:101192. [PMID: 31015147 PMCID: PMC6477631 DOI: 10.1016/j.redox.2019.101192] [Citation(s) in RCA: 193] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/02/2019] [Accepted: 04/07/2019] [Indexed: 01/17/2023] Open
Abstract
Adjustment of cerebral blood flow (CBF) to neuronal activity via neurovascular coupling (NVC) has an essential role in maintenance of healthy cognitive function. In aging increased oxidative stress and cerebromicrovascular endothelial dysfunction impair NVC, contributing to cognitive decline. There is increasing evidence showing that a decrease in NAD+ availability with age plays a critical role in a range of age-related cellular impairments but its role in impaired NVC responses remains unexplored. The present study was designed to test the hypothesis that restoring NAD+ concentration may exert beneficial effects on NVC responses in aging. To test this hypothesis 24-month-old C57BL/6 mice were treated with nicotinamide mononucleotide (NMN), a key NAD+ intermediate, for 2 weeks. NVC was assessed by measuring CBF responses (laser Doppler flowmetry) evoked by contralateral whisker stimulation. We found that NVC responses were significantly impaired in aged mice. NMN supplementation rescued NVC responses by increasing endothelial NO-mediated vasodilation, which was associated with significantly improved spatial working memory and gait coordination. These findings are paralleled by the sirtuin-dependent protective effects of NMN on mitochondrial production of reactive oxygen species and mitochondrial bioenergetics in cultured cerebromicrovascular endothelial cells derived from aged animals. Thus, a decrease in NAD+ availability contributes to age-related cerebromicrovascular dysfunction, exacerbating cognitive decline. The cerebromicrovascular protective effects of NMN highlight the preventive and therapeutic potential of NAD+ intermediates as effective interventions in patients at risk for vascular cognitive impairment (VCI).
Collapse
Affiliation(s)
- Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Marta Noa Valcarcel-Ares
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter Toth
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Neurosurgery, Medical School, University of Pecs, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zsuzsanna Tucsek
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Tamas Kiss
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter Hertelendy
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael Kinter
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Praveen Ballabh
- Division of Neonatology, Department of Pediatrics, Albert Einstein College of Medicine, USA
| | - Zoltán Süle
- Department of Anatomy, University of Szeged, Szeged, Hungary
| | - Eszter Farkas
- Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
| | - Joseph A Baur
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary; Department of Public Health, Semmelweis University, Budapest, Hungary; Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
22
|
MacLaren DAA, Ljungberg TL, Griffin ME, Clark SD. Pedunculopontine tegmentum cholinergic loss leads to a progressive decline in motor abilities and neuropathological changes resembling progressive supranuclear palsy. Eur J Neurosci 2018; 48:3477-3497. [PMID: 30339310 DOI: 10.1111/ejn.14212] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 08/28/2018] [Accepted: 09/27/2018] [Indexed: 11/28/2022]
Abstract
Progressive supranuclear palsy (PSP) is the most common atypical Parkinsonism. Although PSP shares some symptomology with Parkinson's disease (PD), PSP has a different underlying pathology characterized by tau aggregation. Furthermore, PSP sufferers respond poorly to PD medications and there are no effective alternative therapeutics. The development of both palliative and disease altering therapeutics has been hampered by the lack of an animal model that displays relevant PSP-like pathology and behavioral deficits. Previously, our lab found that in rats the selective removal of cholinergic pedunculopontine neurons (whose axonal projections overlap with areas of PSP pathology), mimics the extensive loss of cholinergic pedunculopontine neurons seen in PSP, and produces a unique PSP-like combination of deficits in: startle reflex, attention, and motor function. The present study extends those findings by allowing the lesion to incubate for over a year and compares behavioral and post-mortem pathology of pedunculopontine-cholinergic-lesioned and sham-lesioned rats. There was an early startle reflex deficit which did not improve over time. Progressive declines in motor function developed over the course of the year, including an increase in the number of "slips" while navigating various beams and poorly coordinated transitions from an elevated platform into homecages. Histological analysis discovered that the loss off cholinergic pedunculopontine neurons precipitated a significant loss of substantia nigra tyrosine hydroxylase-positive neurons and a significant enlargement of the lateral ventricles. The latter is a distinguishing feature between PSP and PD. This preclinical animal model of PSP has the potential to further our understanding of PSP and aid in the testing of potential therapeutic agents.
Collapse
Affiliation(s)
- Duncan A A MacLaren
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Trisha L Ljungberg
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Meghan E Griffin
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Stewart D Clark
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA
| |
Collapse
|
23
|
Sébille SB, Rolland AS, Faillot M, Perez-Garcia F, Colomb-Clerc A, Lau B, Dumas S, Vidal SF, Welter ML, Francois C, Bardinet E, Karachi C. Normal and pathological neuronal distribution of the human mesencephalic locomotor region. Mov Disord 2018; 34:218-227. [PMID: 30485555 DOI: 10.1002/mds.27578] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 06/10/2018] [Accepted: 10/22/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Deep brain stimulation of the pedunculopontine nucleus has been performed to treat dopamine-resistant gait and balance disorders in patients with degenerative diseases. The outcomes, however, are variable, which may be the result of the lack of a well-defined anatomical target. OBJECTIVES The objectives of this study were to identify the main neuronal populations of the pedunculopontine and the cuneiform nuclei that compose the human mesencephalic locomotor region and to compare their 3-dimensional distribution with those found in patients with Parkinson's disease and progressive supranuclear palsy. METHODS We used high-field MRI, immunohistochemistry, and in situ hybridization to characterize the distribution of the different cell types, and we developed software to merge all data within a common 3-dimensional space. RESULTS We found that cholinergic, GABAergic, and glutamatergic neurons comprised the main cell types of the mesencephalic locomotor region, with the peak densities of cholinergic and GABAergic neurons similarly located within the rostral pedunculopontine nucleus. Cholinergic and noncholinergic neuronal losses were homogeneous in the mesencephalic locomotor region of patients, with the peak density of remaining neurons at the same location as in controls. The degree of denervation of the pedunculopontine nucleus was highest in patients with progressive supranuclear palsy, followed by Parkinson's disease patients with falls. CONCLUSIONS The peak density of cholinergic and GABAergic neurons was located similarly within the rostral pedunculopontine nucleus not only in controls but also in pathological cases. The neuronal loss was homogeneously distributed and highest in the pedunculopontine nucleus of patients with falls, which suggests a potential pathophysiological link. © 2018 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Sophie B Sébille
- Sorbonne University, Univ. Pierre & Marie Curie Paris 06, Cnrs, Inserm, AP-HP Pitié-Salpêtrière hospital, Brain and Spinal Cord Institute, Paris, France.,Centre de Neuro-Imagerie de Recherche, Paris, France
| | - Anne-Sophie Rolland
- Sorbonne University, Univ. Pierre & Marie Curie Paris 06, Cnrs, Inserm, AP-HP Pitié-Salpêtrière hospital, Brain and Spinal Cord Institute, Paris, France
| | - Matthieu Faillot
- Sorbonne University, Univ. Pierre & Marie Curie Paris 06, Cnrs, Inserm, AP-HP Pitié-Salpêtrière hospital, Brain and Spinal Cord Institute, Paris, France.,Neurosurgical Department, La Pitié-Salpêtrière University Hospital, Paris, France
| | | | - Antoine Colomb-Clerc
- Sorbonne University, Univ. Pierre & Marie Curie Paris 06, Cnrs, Inserm, AP-HP Pitié-Salpêtrière hospital, Brain and Spinal Cord Institute, Paris, France
| | - Brian Lau
- Sorbonne University, Univ. Pierre & Marie Curie Paris 06, Cnrs, Inserm, AP-HP Pitié-Salpêtrière hospital, Brain and Spinal Cord Institute, Paris, France
| | | | | | - Marie-Laure Welter
- Sorbonne University, Univ. Pierre & Marie Curie Paris 06, Cnrs, Inserm, AP-HP Pitié-Salpêtrière hospital, Brain and Spinal Cord Institute, Paris, France.,Neurosurgical Department, La Pitié-Salpêtrière University Hospital, Paris, France
| | - Chantal Francois
- Sorbonne University, Univ. Pierre & Marie Curie Paris 06, Cnrs, Inserm, AP-HP Pitié-Salpêtrière hospital, Brain and Spinal Cord Institute, Paris, France
| | - Eric Bardinet
- Sorbonne University, Univ. Pierre & Marie Curie Paris 06, Cnrs, Inserm, AP-HP Pitié-Salpêtrière hospital, Brain and Spinal Cord Institute, Paris, France.,Centre de Neuro-Imagerie de Recherche, Paris, France
| | - Carine Karachi
- Sorbonne University, Univ. Pierre & Marie Curie Paris 06, Cnrs, Inserm, AP-HP Pitié-Salpêtrière hospital, Brain and Spinal Cord Institute, Paris, France.,Neurosurgical Department, La Pitié-Salpêtrière University Hospital, Paris, France
| |
Collapse
|
24
|
Pappas SS, Li J, LeWitt TM, Kim JK, Monani UR, Dauer WT. A cell autonomous torsinA requirement for cholinergic neuron survival and motor control. eLife 2018; 7:36691. [PMID: 30117805 PMCID: PMC6115190 DOI: 10.7554/elife.36691] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 08/16/2018] [Indexed: 12/14/2022] Open
Abstract
Cholinergic dysfunction is strongly implicated in dystonia pathophysiology. Previously (Pappas et al., 2015;4:e08352), we reported that Dlx5/6-Cre mediated forebrain deletion of the DYT1 dystonia protein torsinA (Dlx-CKO) causes abnormal twisting and selective degeneration of dorsal striatal cholinergic interneurons (ChI) (Pappas et al., 2015). A central question raised by that work is whether the ChI loss is cell autonomous or requires torsinA loss from neurons synaptically connected to ChIs. Here, we addressed this question by using ChAT-Cre mice to conditionally delete torsinA from cholinergic neurons ('ChAT-CKO'). ChAT-CKO mice phenocopy the Dlx-CKO phenotype of selective dorsal striatal ChI loss and identify an essential requirement for torsinA in brainstem and spinal cholinergic neurons. ChAT-CKO mice are tremulous, weak, and exhibit trunk twisting and postural abnormalities. These findings are the first to demonstrate a cell autonomous requirement for torsinA in specific populations of cholinergic neurons, strengthening the connection between torsinA, cholinergic dysfunction and dystonia pathophysiology.
Collapse
Affiliation(s)
- Samuel S Pappas
- Department of Neurology, University of Michigan, Ann Arbor, United States
| | - Jay Li
- Department of Neurology, University of Michigan, Ann Arbor, United States.,Cell and Molecular Biology Program, University of Michigan, Ann Arbor, United States
| | - Tessa M LeWitt
- Department of Neurology, University of Michigan, Ann Arbor, United States
| | - Jeong-Ki Kim
- Department of Cell Biology, Columbia University Medical Center, New York, United States.,Center for Motor Neuron Biology and Disease, Columbia University Medical Center, New York, United States.,Department of Pathology, Columbia University Medical Center, New York, United States
| | - Umrao R Monani
- Department of Cell Biology, Columbia University Medical Center, New York, United States.,Center for Motor Neuron Biology and Disease, Columbia University Medical Center, New York, United States.,Department of Pathology, Columbia University Medical Center, New York, United States
| | - William T Dauer
- Department of Neurology, University of Michigan, Ann Arbor, United States.,Cell and Molecular Biology Program, University of Michigan, Ann Arbor, United States.,Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, United States
| |
Collapse
|
25
|
Manfré G, Clemensson EKH, Kyriakou EI, Clemensson LE, van der Harst JE, Homberg JR, Nguyen HP. The BACHD Rat Model of Huntington Disease Shows Specific Deficits in a Test Battery of Motor Function. Front Behav Neurosci 2017; 11:218. [PMID: 29163089 PMCID: PMC5675855 DOI: 10.3389/fnbeh.2017.00218] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 10/19/2017] [Indexed: 02/01/2023] Open
Abstract
Rationale: Huntington disease (HD) is a progressive neurodegenerative disorder characterized by motor, cognitive and neuropsychiatric symptoms. HD is usually diagnosed by the appearance of motor deficits, resulting in skilled hand use disruption, gait abnormality, muscle wasting and choreatic movements. The BACHD transgenic rat model for HD represents a well-established transgenic rodent model of HD, offering the prospect of an in-depth characterization of the motor phenotype. Objective: The present study aims to characterize different aspects of motor function in BACHD rats, combining classical paradigms with novel high-throughput behavioral phenotyping. Methods: Wild-type (WT) and transgenic animals were tested longitudinally from 2 to 12 months of age. To measure fine motor control, rats were challenged with the pasta handling test and the pellet reaching test. To evaluate gross motor function, animals were assessed by using the holding bar and the grip strength tests. Spontaneous locomotor activity and circadian rhythmicity were assessed in an automated home-cage environment, namely the PhenoTyper. We then integrated existing classical methodologies to test motor function with automated home-cage assessment of motor performance. Results: BACHD rats showed strong impairment in muscle endurance at 2 months of age. Altered circadian rhythmicity and locomotor activity were observed in transgenic animals. On the other hand, reaching behavior, forepaw dexterity and muscle strength were unaffected. Conclusions: The BACHD rat model exhibits certain features of HD patients, like muscle weakness and changes in circadian behavior. We have observed modest but clear-cut deficits in distinct motor phenotypes, thus confirming the validity of this transgenic rat model for treatment and drug discovery purposes.
Collapse
Affiliation(s)
- Giuseppe Manfré
- Donders Institute for Brain, Cognition and Behaviour, Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, Netherlands.,Noldus Information Technology BV, Wageningen, Netherlands.,Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Erik K H Clemensson
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany.,Centre of Rare Diseases, University of Tübingen, Tübingen, Germany
| | - Elisavet I Kyriakou
- Donders Institute for Brain, Cognition and Behaviour, Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, Netherlands.,Noldus Information Technology BV, Wageningen, Netherlands.,Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Laura E Clemensson
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany.,Centre of Rare Diseases, University of Tübingen, Tübingen, Germany
| | - Johanneke E van der Harst
- Donders Institute for Brain, Cognition and Behaviour, Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, Netherlands.,Noldus Information Technology BV, Wageningen, Netherlands
| | - Judith R Homberg
- Donders Institute for Brain, Cognition and Behaviour, Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, Netherlands
| | - Huu Phuc Nguyen
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany.,Centre of Rare Diseases, University of Tübingen, Tübingen, Germany
| |
Collapse
|
26
|
Selective cholinergic depletion of pedunculopontine tegmental nucleus aggravates freezing of gait in parkinsonian rats. Neurosci Lett 2017; 659:92-98. [DOI: 10.1016/j.neulet.2017.08.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 07/21/2017] [Accepted: 08/07/2017] [Indexed: 12/13/2022]
|
27
|
Sanjari Moghaddam H, Zare-Shahabadi A, Rahmani F, Rezaei N. Neurotransmission systems in Parkinson’s disease. Rev Neurosci 2017; 28:509-536. [DOI: 10.1515/revneuro-2016-0068] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 01/10/2017] [Indexed: 12/17/2022]
Abstract
AbstractParkinson’s disease (PD) is histologically characterized by the accumulation of α-synuclein particles, known as Lewy bodies. The second most common neurodegenerative disorder, PD is widely known because of the typical motor manifestations of active tremor, rigidity, and postural instability, while several prodromal non-motor symptoms including REM sleep behavior disorders, depression, autonomic disturbances, and cognitive decline are being more extensively recognized. Motor symptoms most commonly arise from synucleinopathy of nigrostriatal pathway. Glutamatergic, γ-aminobutyric acid (GABA)ergic, cholinergic, serotoninergic, and endocannabinoid neurotransmission systems are not spared from the global cerebral neurodegenerative assault. Wide intrabasal and extrabasal of the basal ganglia provide enough justification to evaluate network circuits disturbance of these neurotransmission systems in PD. In this comprehensive review, English literature in PubMed, Science direct, EMBASE, and Web of Science databases were perused. Characteristics of dopaminergic and non-dopaminergic systems, disturbance of these neurotransmitter systems in the pathophysiology of PD, and their treatment applications are discussed.
Collapse
Affiliation(s)
- Hossein Sanjari Moghaddam
- Research Center for Immunodeficiencies, Children’s Medical Center Hospital, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, Tehran 14194, Iran
- NeuroImmunology Research Association (NIRA), Universal Scientific Education and Research Network (USERN), Tehran 1419783151, Iran
- Student Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Ameneh Zare-Shahabadi
- Research Center for Immunodeficiencies, Children’s Medical Center Hospital, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, Tehran 14194, Iran
- NeuroImmunology Research Association (NIRA), Universal Scientific Education and Research Network (USERN), Tehran 1419783151, Iran
- Psychiatry and Psychology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzaneh Rahmani
- Research Center for Immunodeficiencies, Children’s Medical Center Hospital, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, Tehran 14194, Iran
- NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children’s Medical Center Hospital, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, Tehran 14194, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1419783151, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Boston, MA, USA
| |
Collapse
|
28
|
Noga BR, Sanchez FJ, Villamil LM, O'Toole C, Kasicki S, Olszewski M, Cabaj AM, Majczyński H, Sławińska U, Jordan LM. LFP Oscillations in the Mesencephalic Locomotor Region during Voluntary Locomotion. Front Neural Circuits 2017; 11:34. [PMID: 28579945 PMCID: PMC5437718 DOI: 10.3389/fncir.2017.00034] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 04/28/2017] [Indexed: 11/28/2022] Open
Abstract
Oscillatory rhythms in local field potentials (LFPs) are thought to coherently bind cooperating neuronal ensembles to produce behaviors, including locomotion. LFPs recorded from sites that trigger locomotion have been used as a basis for identification of appropriate targets for deep brain stimulation (DBS) to enhance locomotor recovery in patients with gait disorders. Theta band activity (6–12 Hz) is associated with locomotor activity in locomotion-inducing sites in the hypothalamus and in the hippocampus, but the LFPs that occur in the functionally defined mesencephalic locomotor region (MLR) during locomotion have not been determined. Here we record the oscillatory activity during treadmill locomotion in MLR sites effective for inducing locomotion with electrical stimulation in rats. The results show the presence of oscillatory theta rhythms in the LFPs recorded from the most effective MLR stimulus sites (at threshold ≤60 μA). Theta activity increased at the onset of locomotion, and its power was correlated with the speed of locomotion. In animals with higher thresholds (>60 μA), the correlation between locomotor speed and theta LFP oscillations was less robust. Changes in the gamma band (previously recorded in vitro in the pedunculopontine nucleus (PPN), thought to be a part of the MLR) were relatively small. Controlled locomotion was best achieved at 10–20 Hz frequencies of MLR stimulation. Our results indicate that theta and not delta or gamma band oscillation is a suitable biomarker for identifying the functional MLR sites.
Collapse
Affiliation(s)
- Brian R Noga
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of MedicineMiami, FL, United States
| | - Francisco J Sanchez
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of MedicineMiami, FL, United States
| | - Luz M Villamil
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of MedicineMiami, FL, United States
| | - Christopher O'Toole
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of MedicineMiami, FL, United States
| | - Stefan Kasicki
- Department of Neurophysiology, Nencki Institute of Experimental BiologyWarsaw, Poland
| | - Maciej Olszewski
- Department of Neurophysiology, Nencki Institute of Experimental BiologyWarsaw, Poland
| | - Anna M Cabaj
- Department of Neurophysiology, Nencki Institute of Experimental BiologyWarsaw, Poland
| | - Henryk Majczyński
- Department of Neurophysiology, Nencki Institute of Experimental BiologyWarsaw, Poland
| | - Urszula Sławińska
- Department of Neurophysiology, Nencki Institute of Experimental BiologyWarsaw, Poland
| | - Larry M Jordan
- Department of Physiology, Spinal Cord Research Centre, University of ManitobaWinnipeg, MB, Canada
| |
Collapse
|
29
|
Seke Etet PF, Farahna M, Satti GMH, Bushara YM, El-Tahir A, Hamza MA, Osman SY, Dibia AC, Vecchio L. Garcinia kola seeds may prevent cognitive and motor dysfunctions in a type 1 diabetes mellitus rat model partly by mitigating neuroinflammation. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2017; 14:/j/jcim.2017.14.issue-3/jcim-2016-0167/jcim-2016-0167.xml. [PMID: 28889733 DOI: 10.1515/jcim-2016-0167] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 02/21/2017] [Indexed: 01/06/2023]
Abstract
Background We reported recently that extracts of seeds of Garcinia kola, a plant with established hypoglycemic properties, prevented the loss of inflammation-sensible neuronal populations like Purkinje cells in a rat model of type 1 diabetes mellitus (T1DM). Here, we assessed G. kola extract ability to prevent the early cognitive and motor dysfunctions observed in this model. Methods Rats made diabetic by single injection of streptozotocin were treated daily with either vehicle solution (diabetic control group), insulin, or G. kola extract from the first to the 6th week post-injection. Then, cognitive and motor functions were assessed using holeboard and vertical pole behavioral tests, and animals were sacrificed. Brains were dissected out, cut, and processed for Nissl staining and immunohistochemistry. Results Hyperglycemia (209.26 %), body weight loss (-12.37 %), and T1DM-like cognitive and motor dysfunctions revealed behavioral tests in diabetic control animals were not observed in insulin and extract-treated animals. Similar, expressions of inflammation markers tumor necrosis factor (TNF), iba1 (CD68), and Glial fibrillary acidic protein (GFAP), as well as decreases of neuronal density in regions involved in cognitive and motor functions (-49.56 % motor cortex, -33.24 % medial septal nucleus, -41.8 % /-37.34 % cerebellar Purkinje /granular cell layers) were observed in diabetic controls but not in animals treated with insulin or G. kola. Conclusions Our results indicate that T1DM-like functional alterations are mediated, at least partly, by neuroinflammation and neuronal loss in this model. The prevention of the development of such alterations by early treatment with G. kola confirms the neuroprotective properties of the plant and warrant further mechanistic studies, considering the potential for human disease.
Collapse
|
30
|
Blanco-Lezcano L, Jimenez-Martin J, Díaz-Hung ML, Alberti-Amador E, Wong-Guerra M, González-Fraguela ME, Estupiñán-Díaz B, Serrano-Sánchez T, Francis-Turner L, Delgado-Ocaña S, Núñez-Figueredo Y, Vega-Hurtado Y, Fernández-Jiménez I. Motor dysfunction and alterations in glutathione concentration, cholinesterase activity, and BDNF expression in substantia nigra pars compacta in rats with pedunculopontine lesion. Neuroscience 2017; 348:83-97. [DOI: 10.1016/j.neuroscience.2017.02.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 01/19/2017] [Accepted: 02/07/2017] [Indexed: 12/18/2022]
|
31
|
Role of the pedunculopontine nucleus in controlling gait and sleep in normal and parkinsonian monkeys. J Neural Transm (Vienna) 2017; 125:471-483. [PMID: 28084536 DOI: 10.1007/s00702-017-1678-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 01/07/2017] [Indexed: 12/20/2022]
Abstract
Patients with Parkinson's disease (PD) develop cardinal motor symptoms, including akinesia, rigidity, and tremor, that are alleviated by dopaminergic medication and/or subthalamic deep brain stimulation. Over the time course of the disease, gait and balance disorders worsen and become resistant to pharmacological and surgical treatments. These disorders generate debilitating motor symptoms leading to increased dependency, morbidity, and mortality. PD patients also experience sleep disturbance that raise the question of a common physiological basis. An extensive experimental and clinical body of work has highlighted the crucial role of the pedunculopontine nucleus (PPN) in the control of gait and sleep, and its potential major role in PD. Here, we summarise our investigations in the monkey PPN in the normal and parkinsonian states. We first examined the anatomy and connectivity of the PPN and the cuneiform nucleus which both belong to the mesencephalic locomotor region. Second, we conducted experiments to demonstrate the specific effects of PPN cholinergic lesions on locomotion in the normal and parkinsonian monkey. Third, we aimed to understand how PPN cholinergic lesions impair sleep in parkinsonian monkeys. Our final goal was to develop a novel model of advanced PD with gait and sleep disorders. We believe that this monkey model, even if it does not attempt to reproduce the exact human disease with all its complexities, represents a good biomedical model to characterise locomotion and sleep in the context of PD.
Collapse
|
32
|
Janickova H, Rosborough K, Al-Onaizi M, Kljakic O, Guzman MS, Gros R, Prado MAM, Prado VF. Deletion of the vesicular acetylcholine transporter from pedunculopontine/laterodorsal tegmental neurons modifies gait. J Neurochem 2017; 140:787-798. [DOI: 10.1111/jnc.13910] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 11/04/2016] [Accepted: 11/24/2016] [Indexed: 11/27/2022]
Affiliation(s)
- Helena Janickova
- Robarts Research Institute; Schulich School of Medicine & Dentistry; University of Western Ontario; London Ontario Canada
| | - Kaie Rosborough
- Robarts Research Institute; Schulich School of Medicine & Dentistry; University of Western Ontario; London Ontario Canada
- Department of Anatomy and Cell Biology; Schulich School of Medicine & Dentistry; University of Western Ontario; London Ontario Canada
| | - Mohammed Al-Onaizi
- Robarts Research Institute; Schulich School of Medicine & Dentistry; University of Western Ontario; London Ontario Canada
- Department of Anatomy and Cell Biology; Schulich School of Medicine & Dentistry; University of Western Ontario; London Ontario Canada
| | - Ornela Kljakic
- Robarts Research Institute; Schulich School of Medicine & Dentistry; University of Western Ontario; London Ontario Canada
- Department of Anatomy and Cell Biology; Schulich School of Medicine & Dentistry; University of Western Ontario; London Ontario Canada
| | - Monica S. Guzman
- Robarts Research Institute; Schulich School of Medicine & Dentistry; University of Western Ontario; London Ontario Canada
- Department of Anatomy and Cell Biology; Schulich School of Medicine & Dentistry; University of Western Ontario; London Ontario Canada
| | - Robert Gros
- Robarts Research Institute; Schulich School of Medicine & Dentistry; University of Western Ontario; London Ontario Canada
- Department of Physiology and Pharmacology; Schulich School of Medicine & Dentistry; University of Western Ontario; London Ontario Canada
| | - Marco A. M. Prado
- Robarts Research Institute; Schulich School of Medicine & Dentistry; University of Western Ontario; London Ontario Canada
- Department of Anatomy and Cell Biology; Schulich School of Medicine & Dentistry; University of Western Ontario; London Ontario Canada
- Department of Physiology and Pharmacology; Schulich School of Medicine & Dentistry; University of Western Ontario; London Ontario Canada
| | - Vania F. Prado
- Robarts Research Institute; Schulich School of Medicine & Dentistry; University of Western Ontario; London Ontario Canada
- Department of Anatomy and Cell Biology; Schulich School of Medicine & Dentistry; University of Western Ontario; London Ontario Canada
- Department of Physiology and Pharmacology; Schulich School of Medicine & Dentistry; University of Western Ontario; London Ontario Canada
| |
Collapse
|
33
|
Pienaar IS, Vernon A, Winn P. The Cellular Diversity of the Pedunculopontine Nucleus: Relevance to Behavior in Health and Aspects of Parkinson's Disease. Neuroscientist 2016; 23:415-431. [PMID: 27932591 DOI: 10.1177/1073858416682471] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The pedunculopontine nucleus (PPN) is a rostral brainstem structure that has extensive connections with basal ganglia nuclei and the thalamus. Through these the PPN contributes to neural circuits that effect cortical and hippocampal activity. The PPN also has descending connections to nuclei of the pontine and medullary reticular formations, deep cerebellar nuclei, and the spinal cord. Interest in the PPN has increased dramatically since it was first suggested to be a novel target for treating patients with Parkinson's disease who are refractory to medication. However, application of frequency-specific electrical stimulation of the PPN has produced inconsistent results. A central reason for this is that the PPN is not a heterogeneous structure. In this article, we review current knowledge of the neurochemical identity and topographical distribution of neurons within the PPN of both humans and experimental animals, focusing on studies that used neuronally selective targeting strategies to ascertain how the neurochemical heterogeneity of the PPN relates to its diverse functions in relation to movement and cognitive processes. If the therapeutic potential of the PPN is to be realized, it is critical to understand the complex structure-function relationships that exist here.
Collapse
Affiliation(s)
- Ilse S Pienaar
- 1 Centre for Neuroinflammation & Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Cane Road, London, UK.,2 Faculty of Health and Life Sciences, Department of Applied Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - Anthony Vernon
- 3 Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Philip Winn
- 4 Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
34
|
Okada KI, Kobayashi Y. Reward and Behavioral Factors Contributing to the Tonic Activity of Monkey Pedunculopontine Tegmental Nucleus Neurons during Saccade Tasks. Front Syst Neurosci 2016; 10:94. [PMID: 27891082 PMCID: PMC5104745 DOI: 10.3389/fnsys.2016.00094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 11/03/2016] [Indexed: 01/24/2023] Open
Abstract
The pedunculopontine tegmental nucleus (PPTg) in the brainstem plays a role in controlling reinforcement learning and executing conditioned behavior. We previously examined the activity of PPTg neurons in monkeys during a reward-conditioned, visually guided saccade task, and reported that a population of these neurons exhibited tonic responses throughout the task period. These tonic responses might depend on prediction of the upcoming reward, successful execution of the task, or both. Here, we sought to further distinguish these factors and to investigate how each contributes to the tonic neuronal activity of the PPTg. In our normal visually guided saccade task, the monkey initially fixated on the central fixation target (FT), then made saccades to the peripheral saccade target and received a juice reward after the saccade target disappeared. Most of the tonic activity terminated shortly after the reward delivery, when the monkey broke fixation. To distinguish between reward and behavioral epochs, we then changed the task sequence for a block of trials, such that the saccade target remained visible after the reward delivery. Under these visible conditions, the monkeys tended to continue fixating on the saccade target even after the reward delivery. Therefore, the prediction of the upcoming reward and the end of an individual trial were separated in time. Regardless of the task conditions, half of the tonically active PPTg neurons terminated their activity around the time of the reward delivery, consistent with the view that PPTg neurons might send reward prediction signals until the time of reward delivery, which is essential for computing reward prediction error in reinforcement learning. On the other hand, the other half of the tonically active PPTg neurons changed their activity dependent on the task condition. In the normal condition, the tonic responses terminated around the time of the reward delivery, while in the visible condition, the activity continued until the disappearance of the saccade target (ST) after reward delivery. Thus, for these neurons, the tonic activity might be related to maintaining attention to complete fixation behavior. These results suggest that, in addition to the reward value information, some PPTg neurons might contribute to the execution of conditioned task behavior.
Collapse
Affiliation(s)
- Ken-Ichi Okada
- Laboratories for Neuroscience, Visual Neuroscience Group, Graduate School of Frontier Biosciences, Osaka UniversityOsaka, Japan; Center for Information and Neural Networks, National Institute of Information and Communications Technology, Osaka UniversityOsaka, Japan
| | - Yasushi Kobayashi
- Laboratories for Neuroscience, Visual Neuroscience Group, Graduate School of Frontier Biosciences, Osaka UniversityOsaka, Japan; Center for Information and Neural Networks, National Institute of Information and Communications Technology, Osaka UniversityOsaka, Japan; Research Center for Behavioral Economics, Osaka UniversityOsaka, Japan
| |
Collapse
|
35
|
Perez-Lloret S, Peralta MC, Barrantes FJ. Pharmacotherapies for Parkinson's disease symptoms related to cholinergic degeneration. Expert Opin Pharmacother 2016; 17:2405-2415. [PMID: 27785919 DOI: 10.1080/14656566.2016.1254189] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Dopamine depletion is one of the most important features of Parkinson's Disease (PD). However, insufficient response to dopaminergic replacement therapy suggests the involvement of other neurotransmitter systems in the pathophysiology of PD. Cholinergic degeneration contributes to gait impairments, cognitive impairment, psychosis, and REM-sleep disturbances, among other symptoms. Areas covered: In this review, we explore the idea that enhancing cholinergic tone by pharmacological or neurosurgical procedures could be a first-line therapeutic strategy for the treatment of symptoms derived from cholinergic degeneration in PD. Expert opinion: Rivastigmine, a drug that increases cholinergic tone by inhibiting the enzyme cholinesterase, is effective for dementia, whereas the use of Donepezil is still in the realm of investigation. Interesting results suggest the efficacy of these drugs in the treatment of gait dysfunction. Evidence on the clinical effects of these drugs for psychosis and REM-sleep disturbances is still weak. Stimulation of the pedunculo-pontine tegmental nuclei (which provide cholinergic innervation to the brain stem and subcortical nuclei) has also been used with some success for the treatment of gait dysfunction. Anticholinergic drugs should be used with caution in PD, as they may aggravate cholinergic symptoms. Notwithstanding, in some patients they might help control parkinsonian motor symptoms.
Collapse
Affiliation(s)
- Santiago Perez-Lloret
- a Institute of Cardiology Research , University of Buenos Aires, National Research Council (CONICET-ININCA) , Buenos Aires , Argentina
| | - María Cecilia Peralta
- b Parkinson's Disease and Movement Disorders Clinic, Neurology Department , CEMIC University Hospital , Buenos Aires , Argentina
| | - Francisco J Barrantes
- c Laboratory of Molecular Neurobiology , Institute for Biomedical Research, UCA-CONICET, Faculty of Medical Sciences , Buenos Aires , Argentina
| |
Collapse
|
36
|
Mena-Segovia J. Structural and functional considerations of the cholinergic brainstem. J Neural Transm (Vienna) 2016; 123:731-736. [PMID: 26945862 DOI: 10.1007/s00702-016-1530-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 02/19/2016] [Indexed: 12/24/2022]
Abstract
Cholinergic neurons of the brainstem have traditionally been associated with a role in wakefulness as part of the reticular activating system, but their function cannot be explained solely on the basis of their modulation of the brain state. Recent findings about their connectivity and functional heterogeneity suggest a wider role in behavior, where basal ganglia is at the center of their influence. This review focuses on recent findings that suggest an intrinsic functional organization of the cholinergic brainstem that is closely correlated with its connectivity with midbrain and forebrain circuits. Furthermore, recent evidence on the temporal structure of the activation of brainstem cholinergic neurons reveals fundamental aspects about the nature of cholinergic signaling. Consideration of the cholinergic brainstem complex in the context of wider brain circuits is critical to understand its contribution to normal behavior.
Collapse
Affiliation(s)
- Juan Mena-Segovia
- Center for Molecular and Behavioral Neuroscience, Aidekman Research Center, Rutgers University, Newark, NJ, 07102, USA.
| |
Collapse
|
37
|
Perez-Lloret S, Barrantes FJ. Deficits in cholinergic neurotransmission and their clinical correlates in Parkinson's disease. NPJ PARKINSONS DISEASE 2016; 2:16001. [PMID: 28725692 PMCID: PMC5516588 DOI: 10.1038/npjparkd.2016.1] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/10/2015] [Accepted: 12/21/2015] [Indexed: 01/04/2023]
Abstract
In view of its ability to explain the most frequent motor symptoms of Parkinson’s Disease (PD), degeneration of dopaminergic neurons has been considered one of the disease’s main pathophysiological features. Several studies have shown that neurodegeneration also affects noradrenergic, serotoninergic, cholinergic and other monoaminergic neuronal populations. In this work, the characteristics of cholinergic deficits in PD and their clinical correlates are reviewed. Important neurophysiological processes at the root of several motor and cognitive functions remit to cholinergic neurotransmission at the synaptic, pathway, and circuital levels. The bulk of evidence highlights the link between cholinergic alterations and PD motor symptoms, gait dysfunction, levodopa-induced dyskinesias, cognitive deterioration, psychosis, sleep abnormalities, autonomic dysfunction, and altered olfactory function. The pathophysiology of these symptoms is related to alteration of the cholinergic tone in the striatum and/or to degeneration of cholinergic nuclei, most importantly the nucleus basalis magnocellularis and the pedunculopontine nucleus. Several results suggest the clinical usefulness of antimuscarinic drugs for treating PD motor symptoms and of inhibitors of the enzyme acetylcholinesterase for the treatment of dementia. Data also suggest that these inhibitors and pedunculopontine nucleus deep-brain stimulation might also be effective in preventing falls. Finally, several drugs acting on nicotinic receptors have proved efficacious for treating levodopa-induced dyskinesias and cognitive impairment and as neuroprotective agents in PD animal models. Results in human patients are still lacking.
Collapse
Affiliation(s)
- Santiago Perez-Lloret
- Institute of Cardiologic Research, National Scientific and Research Council (ININCA-CONICET), Faculty of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Francisco J Barrantes
- Laboratory of Molecular Neurobiology, Institute for Biomedical Research, UCA-CONICET, Faculty of Medical Sciences, Buenos Aires, Argentina
| |
Collapse
|
38
|
Gut NK, Winn P. The pedunculopontine tegmental nucleus-A functional hypothesis from the comparative literature. Mov Disord 2016; 31:615-24. [PMID: 26880095 PMCID: PMC4949639 DOI: 10.1002/mds.26556] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 12/22/2015] [Accepted: 12/28/2015] [Indexed: 12/21/2022] Open
Abstract
We present data from animal studies showing that the pedunculopontine tegmental nucleus-conserved through evolution, compartmentalized, and with a complex pattern of inputs and outputs-has functions that involve formation and updates of action-outcome associations, attention, and rapid decision making. This is in contrast to previous hypotheses about pedunculopontine function, which has served as a basis for clinical interest in the pedunculopontine in movement disorders. Current animal literature points to it being neither a specifically motor structure nor a master switch for sleep regulation. The pedunculopontine is connected to basal ganglia circuitry but also has primary sensory input across modalities and descending connections to pontomedullary, cerebellar, and spinal motor and autonomic control systems. Functional and anatomical studies in animals suggest strongly that, in addition to the pedunculopontine being an input and output station for the basal ganglia and key regulator of thalamic (and consequently cortical) activity, an additional major function is participation in the generation of actions on the basis of a first-pass analysis of incoming sensory data. Such a function-rapid decision making-has very high adaptive value for any vertebrate. We argue that in developing clinical strategies for treating basal ganglia disorders, it is necessary to take an account of the normal functions of the pedunculopontine. We believe that it is possible to use our hypothesis to explain why pedunculopontine deep brain stimulation used clinically has had variable outcomes in the treatment of parkinsonism motor symptoms and effects on cognitive processing. © 2016 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Nadine K Gut
- Biozentrum, University of Basel, Basel, Switzerland
| | - Philip Winn
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| |
Collapse
|
39
|
Jin X, Schwabe K, Krauss JK, Alam M. The anterior and posterior pedunculopontine tegmental nucleus are involved in behavior and neuronal activity of the cuneiform and entopeduncular nuclei. Neuroscience 2016; 322:39-53. [PMID: 26880033 DOI: 10.1016/j.neuroscience.2016.02.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 01/13/2016] [Accepted: 02/06/2016] [Indexed: 10/22/2022]
Abstract
Loss of cholinergic neurons in the mesencephalic locomotor region, comprising the pedunculopontine nucleus (PPN) and the cuneiform nucleus (CnF), is related to gait disturbances in late stage Parkinson's disease (PD). We investigate the effect of anterior or posterior cholinergic lesions of the PPN on gait-related motor behavior, and on neuronal network activity of the PPN area and basal ganglia (BG) motor loop in rats. Anterior PPN lesions, posterior PPN lesions or sham lesions were induced by stereotaxic microinjection of the cholinergic toxin AF64-A or vehicle in male Sprague-Dawley rats. First, locomotor activity (open field), postural disturbances (Rotarod) and gait asymmetry (treadmill test) were assessed. Thereafter, single-unit and oscillatory activities were measured in the non-lesioned area of the PPN, the CnF and the entopeduncular nucleus (EPN), the BG output region, with microelectrodes under urethane anesthesia. Additionally, ECoG was recorded in the motor cortex. Injection of AF64-A into the anterior and posterior PPN decreased cholinergic cell counts as compared to naive controls (P<0.001) but also destroyed non-cholinergic cells. Only anterior PPN lesions decreased the front limb swing time of gait in the treadmill test, while not affecting other gait-related parameters tested. Main electrophysiological findings were that anterior PPN lesions increased the firing activity in the CnF (P<0.001). Further, lesions of either PPN region decreased the coherence of alpha (8-12 Hz) band between CnF and motor cortex (MCx), and increased the beta (12-30 Hz) oscillatory synchronization between EPN and the MCx. Lesions of the PPN in rats had complex effects on oscillatory neuronal activity of the CnF and the BG network, which may contribute to the understanding of the pathophysiology of gait disturbance in PD.
Collapse
Affiliation(s)
- X Jin
- Department of Neurosurgery, Hannover Medical School, Hannover, Germany
| | - K Schwabe
- Department of Neurosurgery, Hannover Medical School, Hannover, Germany
| | - J K Krauss
- Department of Neurosurgery, Hannover Medical School, Hannover, Germany
| | - M Alam
- Department of Neurosurgery, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
40
|
Pharmacologically-induced neurovascular uncoupling is associated with cognitive impairment in mice. J Cereb Blood Flow Metab 2015; 35:1871-81. [PMID: 26174328 PMCID: PMC4635246 DOI: 10.1038/jcbfm.2015.162] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 05/23/2015] [Accepted: 05/29/2015] [Indexed: 01/15/2023]
Abstract
There is increasing evidence that vascular risk factors, including aging, hypertension, diabetes mellitus, and obesity, promote cognitive impairment; however, the underlying mechanisms remain obscure. Cerebral blood flow (CBF) is adjusted to neuronal activity via neurovascular coupling (NVC) and this mechanism is known to be impaired in the aforementioned pathophysiologic conditions. To establish a direct relationship between impaired NVC and cognitive decline, we induced neurovascular uncoupling pharmacologically in mice by inhibiting the synthesis of vasodilator mediators involved in NVC. Treatment of mice with the epoxygenase inhibitor N-(methylsulfonyl)-2-(2-propynyloxy)-benzenehexanamide (MSPPOH), the NO synthase inhibitor l-NG-Nitroarginine methyl ester (L-NAME), and the COX inhibitor indomethacin decreased NVC by over 60% mimicking the aging phenotype, which was associated with significantly impaired spatial working memory (Y-maze), recognition memory (Novel object recognition), and impairment in motor coordination (Rotarod). Blood pressure (tail cuff) and basal cerebral perfusion (arterial spin labeling perfusion MRI) were unaffected. Thus, selective experimental disruption of NVC is associated with significant impairment of cognitive and sensorimotor function, recapitulating neurologic symptoms and signs observed in brain aging and pathophysiologic conditions associated with accelerated cerebromicrovascular aging.
Collapse
|
41
|
Pienaar IS, Gartside SE, Sharma P, De Paola V, Gretenkord S, Withers D, Elson JL, Dexter DT. Pharmacogenetic stimulation of cholinergic pedunculopontine neurons reverses motor deficits in a rat model of Parkinson's disease. Mol Neurodegener 2015; 10:47. [PMID: 26394842 PMCID: PMC4580350 DOI: 10.1186/s13024-015-0044-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 09/08/2015] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Patients with advanced Parkinson's disease (PD) often present with axial symptoms, including postural- and gait difficulties that respond poorly to dopaminergic agents. Although deep brain stimulation (DBS) of a highly heterogeneous brain structure, the pedunculopontine nucleus (PPN), improves such symptoms, the underlying neuronal substrate responsible for the clinical benefits remains largely unknown, thus hampering optimization of DBS interventions. Choline acetyltransferase (ChAT)::Cre(+) transgenic rats were sham-lesioned or rendered parkinsonian through intranigral, unihemispheric stereotaxic administration of the ubiquitin-proteasomal system inhibitor, lactacystin, combined with designer receptors exclusively activated by designer drugs (DREADD), to activate the cholinergic neurons of the nucleus tegmenti pedunculopontine (PPTg), the rat equivalent of the human PPN. We have previously shown that the lactacystin rat model accurately reflects aspects of PD, including a partial loss of PPTg cholinergic neurons, similar to what is seen in the post-mortem brains of advanced PD patients. RESULTS In this manuscript, we show that transient activation of the remaining PPTg cholinergic neurons in the lactacystin rat model of PD, via peripheral administration of the cognate DREADD ligand, clozapine-N-oxide (CNO), dramatically improved motor symptoms, as was assessed by behavioral tests that measured postural instability, gait, sensorimotor integration, forelimb akinesia and general motor activity. In vivo electrophysiological recordings revealed increased spiking activity of PPTg putative cholinergic neurons during CNO-induced activation. c-Fos expression in DREADD overexpressed ChAT-immunopositive (ChAT+) neurons of the PPTg was also increased by CNO administration, consistent with upregulated neuronal activation in this defined neuronal population. CONCLUSIONS Overall, these findings provide evidence that functional modulation of PPN cholinergic neurons alleviates parkinsonian motor symptoms.
Collapse
Affiliation(s)
- Ilse S Pienaar
- Centre for Neuroinflammation and Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, London, W12 ONN, UK.
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, NE1 8ST, UK.
| | - Sarah E Gartside
- Institute of Neuroscience, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Puneet Sharma
- Centre for Neuroinflammation and Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, London, W12 ONN, UK
| | - Vincenzo De Paola
- Medical Research Council Clinical Sciences Centre, Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - Sabine Gretenkord
- Institute of Neuroscience, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Dominic Withers
- Medical Research Council Clinical Sciences Centre, Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - Joanna L Elson
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, NE1 3BZ, UK
- Centre for Human Metabonomics, North-West University, Potchefstroom, South Africa
| | - David T Dexter
- Centre for Neuroinflammation and Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, London, W12 ONN, UK
| |
Collapse
|
42
|
Deep brain stimulation of different pedunculopontine targets in a novel rodent model of parkinsonism. J Neurosci 2015; 35:4792-803. [PMID: 25810510 DOI: 10.1523/jneurosci.3646-14.2015] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The pedunculopontine tegmental nucleus (PPTg) has been proposed as a target for deep brain stimulation (DBS) in parkinsonian patients, particularly for symptoms such as gait and postural difficulties refractory to dopaminergic treatments. Several patients have had electrodes implanted aimed at the PPTg, but outcomes have been disappointing, with little evidence that gait and posture are improved. The PPTg is a heterogeneous structure. Consequently, exact target sites in PPTg, possible DBS mechanisms, and potential benefits still need systematic investigation in good animal models. We have investigated the role of PPTg in gait, developed a refined model of parkinsonism including partial loss of the PPTg with bilateral destruction of nigrostriatal dopamine neurons that mimics human pathophysiology, and investigated the effect of DBS at different PPTg locations on gait and posture using a wireless device that lets rats move freely while receiving stimulation. Neither partial nor complete lesions of PPTg caused gait deficits, underlining questions raised previously about the status of PPTg as a motor control structure. The effect of DBS in the refined and standard model of parkinsonism were very different despite minimal behavioral differences in nonstimulation control conditions. Anterior PPTg DBS caused severe episodes of freezing and worsened gait, whereas specific gait parameters were mildly improved by stimulation of posterior PPTg. These results emphasize the critical importance of intra-PPTg DBS location and highlight the need to take PPTg degeneration into consideration when modeling parkinsonian symptoms. They also further implicate a role for PPTg in the pathophysiology of parkinsonism.
Collapse
|
43
|
MacLaren DAA, Wilson DIG, Winn P. Selective lesions of the cholinergic neurons within the posterior pedunculopontine do not alter operant learning or nicotine sensitization. Brain Struct Funct 2015; 221:1481-97. [PMID: 25586659 DOI: 10.1007/s00429-014-0985-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 11/30/2014] [Indexed: 02/02/2023]
Abstract
Cholinergic neurons within the pedunculopontine tegmental nucleus have been implicated in a range of functions, including behavioral state control, attention, and modulation of midbrain and basal ganglia systems. Previous experiments with excitotoxic lesions have found persistent learning impairment and altered response to nicotine following lesion of the posterior component of the PPTg (pPPTg). These effects have been attributed to disrupted input to midbrain dopamine systems, particularly the ventral tegmental area. The pPPTg contains a dense collection of cholinergic neurons and also large numbers of glutamatergic and GABAergic neurons. Because these interdigitated populations of neurons are all susceptible to excitotoxins, the effects of such lesions cannot be attributed to one neuronal population. We wished to assess whether the learning impairments and altered responses to nicotine in excitotoxic PPTg-lesioned rats were due to loss of cholinergic neurons within the pPPTg. Selective depletion of cholinergic pPPTg neurons is achievable with the fusion toxin Dtx-UII, which targets UII receptors expressed only by cholinergic neurons in this region. Rats bearing bilateral lesions of cholinergic pPPTg neurons (>90% ChAT+ neuronal loss) displayed no deficits in the learning or performance of fixed and variable ratio schedules of reinforcement for pellet reward. Separate rats with the same lesions had a normal locomotor response to nicotine and furthermore sensitized to repeated administration of nicotine at the same rate as sham controls. Previously seen changes in these behaviors following excitotoxic pPPTg lesions cannot be attributed solely to loss of cholinergic neurons. These findings indicate that non-cholinergic neurons within the pPPTg are responsible for the learning deficits and altered responses to nicotine seen after excitotoxic lesions. The functions of cholinergic neurons may be related to behavioral state control and attention rather than learning.
Collapse
Affiliation(s)
- Duncan A A MacLaren
- Strathclyde Institute of Pharmacy and Biomedical Sciences, 161 Cathedral Street, Glasgow, G4 0RE, UK. .,School of Psychology and Neuroscience, University of St Andrews, St Andrews, Fife, KY16 9JP, UK.
| | - David I G Wilson
- School of Psychology and Neuroscience, University of St Andrews, St Andrews, Fife, KY16 9JP, UK
| | - Philip Winn
- Strathclyde Institute of Pharmacy and Biomedical Sciences, 161 Cathedral Street, Glasgow, G4 0RE, UK
| |
Collapse
|
44
|
MacLaren DAA, Markovic T, Clark SD. Assessment of sensorimotor gating following selective lesions of cholinergic pedunculopontine neurons. Eur J Neurosci 2014; 40:3526-37. [PMID: 25208852 DOI: 10.1111/ejn.12716] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 07/23/2014] [Accepted: 08/08/2014] [Indexed: 12/21/2022]
Abstract
Sensorimotor gating is the state-dependent transfer of sensory information into a motor system. When this occurs at an early stage of the processing stream it enables stimuli to be filtered out or partially ignored, thereby reducing the demands placed on advanced systems. Prepulse inhibition (PPI) of the acoustic startle reflex (ASR) is the standard measure of sensorimotor gating. A brainstem-midbrain circuitry is widely viewed as mediating both PPI and ASR. In this circuitry, the pedunculopontine tegmental nucleus (PPTg) integrates sensory input and cortico-basal ganglia output and, via presumed cholinergic signaling, inhibits ASR-generating neurons within the reticular formation. Non-selective damage to all neuronal types within PPTg reduces PPI. We assessed whether this effect originates in the loss of cholinergic signaling by examining ASR and PPI in rats bearing non-selective (excitotoxic) or selective cholinergic (Dtx-UII) lesions of PPTg. Excitotoxic lesions had no effect on ASR but reduced PPI at all prepulse levels tested. In contrast, selective depletion of cholinergic neurons reduced ASR to the extent that PPI was not measurable with standard (10-20 s) inter-trial intervals. Subsequent testing revealed appreciable ASRs could be generated when the inter-trial interval was increased (180 s). Under these conditions, PPI was assessed and no deficits were found after lesions of cholinergic PPTg neurons. These results show that cholinergic output from PPTg is essential for rapidly regenerating the ASR, but has no influence on PPI. Results are discussed in terms of sensorimotor integration circuitry and psychiatric disorders that feature disrupted ASR and PPI.
Collapse
Affiliation(s)
- Duncan A A MacLaren
- Department of Pharmacology and Toxicology, School of Medicine and Biomedical Sciences, University at Buffalo, SUNY, Buffalo, NY, USA
| | | | | |
Collapse
|
45
|
Enhanced consumption of salient solutions following pedunculopontine tegmental lesions. Neuroscience 2014; 284:381-399. [PMID: 25305665 DOI: 10.1016/j.neuroscience.2014.09.075] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 09/26/2014] [Accepted: 09/30/2014] [Indexed: 12/19/2022]
Abstract
Rats with lesions of the pedunculopontine tegmental nucleus (PPTg) reliably overconsume high concentration sucrose solution. This effect is thought to be indicative of response-perseveration or loss of behavioral control in conditions of high excitement. While these theories have anatomical and behavioral support, they have never been explicitly tested. Here, we used a contact lickometer to examine the microstructure of drinking behavior to gain insight into the behavioral changes during overconsumption. Rats received either excitotoxic (ibotenic acid) damage to all PPTg neuronal subpopulations or selective depletion of the cholinergic neuronal sub-population (diphtheria toxin-urotensin II (Dtx-UII) lesions). We offered rats a variety of pleasant, neutral and aversive tastants to assess the generalizability and specificity of the overconsumption effect. Ibotenic-lesioned rats consumed significantly more 20% sucrose than sham controls, and did so through licking significantly more times. However, the behavioral microstructure during overconsumption was unaffected by the lesion and showed no indications of response-perseveration. Furthermore, the overconsumption effect did not generalize to highly consumed saccharin. In contrast, while only consuming small amounts of quinine solution, ibotenic-lesioned rats had significantly more licks and bursts for this tastant. Selective depletion of cholinergic PPTg neurons had no effect on consumption of any tastant. We then assessed whether it is the salience of the solution which determines overconsumption by ibotenic-lesioned rats. While maintained on free-food, ibotenic-lesioned rats had normal consumption of sucrose and hypertonic saline. After mild food deprivation ibotenic PPTg-lesioned rats overconsumed 20% sucrose. Subsequently, after dietary-induced sodium deficiency, lesioned rats consumed significantly more saline than controls. These results establish that it is the salience of the solution which is the determining factor leading to overconsumption following excitotoxic PPTg lesion. They also find no support for response-perseveration contributing to this effect. Results are discussed in terms of altered dopamine (DA) and salience signaling.
Collapse
|
46
|
Cyr M, Parent MJ, Mechawar N, Rosa-Neto P, Soucy JP, Clark SD, Aghourian M, Bedard MA. Deficit in sustained attention following selective cholinergic lesion of the pedunculopontine tegmental nucleus in rat, as measured with both post-mortem immunocytochemistry and in vivo PET imaging with [¹⁸F]fluoroethoxybenzovesamicol. Behav Brain Res 2014; 278:107-14. [PMID: 25257103 DOI: 10.1016/j.bbr.2014.09.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Revised: 09/12/2014] [Accepted: 09/15/2014] [Indexed: 12/28/2022]
Abstract
Cholinergic neurons of the pedunculopontine tegmental nucleus (PPTg) are thought to be involved in cognitive functions such as sustained attention, and lesions of these cells have been documented in patients showing fluctuations of attention such as in Parkinson's disease or dementia with Lewy Body. Animal studies have been conducted to support the role of these cells in attention, but the lesions induced in these animals were not specific to the cholinergic PPTg system, and were assessed by post-mortem methods remotely performed from the in vivo behavioral assessments. Moreover, sustained attention have not been directly assessed in these studies, but rather deduced from indirect measurements. In the present study, rats were assessed on the 5-Choice Serial Reaction Time Task (5-CSRTT), and a specific measure of variability in response latency was created. Animals were observed both before and after selective lesion of the PPTg cholinergic neurons. Brain cholinergic denervation was assessed both in vivo and ex vivo, using PET imaging with [(18)F]fluoroethoxybenzovesamicol ([(18)F]FEOBV) and immunocytochemistry respectively. Results showed that the number of correct responses and variability in response latency in the 5-CSRTT were the only behavioral measures affected following the lesions. These measures were found to correlate significantly with the number of PPTg cholinergic cells, as measured with both [(18)F]FEOBV and immunocytochemistry. This suggests the primary role of the PPTg cholinergic cells in sustained attention. It also allows to reliably use the PET imaging with [(18)F]FEOBV for the purpose of assessing the relationship between behavior and cholinergic innervation in living animals.
Collapse
Affiliation(s)
- Marilyn Cyr
- Université du Québec à Montréal (UQAM), Canada; Douglas Mental Health University Institute, Canada
| | - Maxime J Parent
- Université du Québec à Montréal (UQAM), Canada; Douglas Mental Health University Institute, Canada
| | | | - Pedro Rosa-Neto
- Douglas Mental Health University Institute, Canada; Montreal Neurological Institute (MNI), Canada
| | | | | | | | - Marc-Andre Bedard
- Université du Québec à Montréal (UQAM), Canada; Douglas Mental Health University Institute, Canada; Montreal Neurological Institute (MNI), Canada.
| |
Collapse
|