1
|
Galgani A, Scotto M, Faraguna U, Giorgi FS. Fading Blue: Exploring the Causes of Locus Coeruleus Damage Across the Lifespan. Antioxidants (Basel) 2025; 14:255. [PMID: 40227216 PMCID: PMC11939699 DOI: 10.3390/antiox14030255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 04/15/2025] Open
Abstract
Locus Coeruleus (LC) is a brain nucleus that is involved in a variety of key functions (ranging from attention modulation to sleep-wake cycle regulation, to memory encoding); its proper function is necessary both during brain development and for brain integrity maintenance, and both at the microscale and macroscale level. Due to their specific intrinsic and extrinsic features, LC cells are considered particularly susceptible to damage concerning a variety of insults. This explains LC involvement in degenerative diseases not only in adults (in the context of neurodegenerative disease, mainly), but also in children (in relation to early hypoxic damage and Down's Syndrome, among others). In this narrative review, we dissect the potential mechanisms through which LC is affected in different diseases, with a special emphasis on the high rate of activity it is subjected to and the oxidative stress associated with it. Further research aimed at deepening our understanding of these mechanisms is needed to enable the development of potential strategies in the future that could slow down LC degeneration in subjects predisposed to specific brain disorders.
Collapse
Affiliation(s)
- Alessandro Galgani
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56125 Pisa, Italy
| | - Marco Scotto
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56125 Pisa, Italy
- Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Ugo Faraguna
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56125 Pisa, Italy
- I.R.C.C.S. Stella Maris, Calambrone, 56128 Pisa, Italy
| | - Filippo S. Giorgi
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56125 Pisa, Italy
- I.R.C.C.S. Stella Maris, Calambrone, 56128 Pisa, Italy
| |
Collapse
|
2
|
Rhoten SE, Wenger MJ, De Stefano LA. Iron deficiency negatively affects behavioral measures of learning, indirect neural measures of dopamine, and neural efficiency. COGNITIVE, AFFECTIVE & BEHAVIORAL NEUROSCIENCE 2025; 25:89-113. [PMID: 39638921 DOI: 10.3758/s13415-024-01241-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/30/2024] [Indexed: 12/07/2024]
Abstract
Iron deficiency (ID) is the most prevalent nutrient deficiency in the world, with a growing literature documenting the negative effects of ID on perception, attention, and memory. Animal models of ID suggest that dysregulation of dopamine is responsible for the deficits in memory. However, evidence that ID affects dopamine in humans is extremely limited. We report the results of a study involving college-aged women with and without ID learning two different category structures - a rule-based and an information-integration structure - selected based on the putative differential role of dopamine in learning these two structures. ID non-anemic (IDNA) and iron-sufficient (IS) women completed 1200 learning trials for each structure. EEG was collected to assess the effects of ID on features affected by dopaminergic state: error-related negativity (ERN) and positivity (Pe), feedback-related negativity (FRN), and task-related blink rate. In addition, we examined the EEG data for dynamics distinguishing IDNA from IS women, including a measure of neural efficiency. Both groups of women were able to learn both structures. However, IDNA women were initially slower and less accurate than IS women, specifically for the rule-based structure. There were large and persistent group differences in brain dynamics and neural efficiency measures. The results are discussed with respect to the selective impact of ID on initial rule-based learning and the persistent effect of ID on dopamine signaling and energetic efficiency.
Collapse
Affiliation(s)
- Stephanie E Rhoten
- Psychology and Cellular and Behavioral Neurobiology, The University of Oklahoma, 201 Stephenson Parkway, Suite 4100, Norman, OK, 73019, USA
| | - Michael J Wenger
- Psychology and Cellular and Behavioral Neurobiology, The University of Oklahoma, 201 Stephenson Parkway, Suite 4100, Norman, OK, 73019, USA.
| | - Lisa A De Stefano
- Psychology and Cellular and Behavioral Neurobiology, The University of Oklahoma, 201 Stephenson Parkway, Suite 4100, Norman, OK, 73019, USA
| |
Collapse
|
3
|
Iannitelli AF, Hassenein L, Mulvey B, Blankenship HE, Liles LC, Sharpe AL, Pare JF, Segal A, Sloan SA, Martinowich K, McCann KE, Dougherty JD, Smith Y, Beckstead MJ, Weinshenker D. Tyrosinase-induced neuromelanin accumulation triggers rapid dysregulation and degeneration of the mouse locus coeruleus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.07.530845. [PMID: 36945637 PMCID: PMC10028911 DOI: 10.1101/2023.03.07.530845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
The locus coeruleus (LC), the major source of norepinephrine (NE) in the brain, is an early site of pathology in both Alzheimer's disease (AD) and Parkinson's disease (PD), and it undergoes catastrophic degeneration later in both disorders. Dysregulation of the LC is thought to contribute to prodromal symptoms of AD and PD such as anxiety and sleep disturbances, while frank LC-NE loss promotes cognitive decline. However, the mechanisms responsible for its selective vulnerability are unknown. The LC is among the only structures in the brain that produces appreciable amounts of neuromelanin (NM), a dark cytoplasmic pigment. It has been proposed that NM initially plays a protective role by sequestering toxic catecholamine metabolites and heavy metals, but may become harmful during aging as it overwhelms cellular machinery and is released during neurodegeneration. Rodents do not naturally produce NM, limiting the study of causal relationships between NM and LC pathology. Adapting a viral-mediated approach for expression of human tyrosinase, the enzyme responsible for peripheral melanin production, we successfully promoted pigmentation in mouse LC neurons that recapitulates key ultrastructural features of endogenous NM found in primates. Pigment expression results in LC neuron hyperactivity, reduced tissue NE levels, transcriptional changes, and novelty-induced anxiety phenotypes as early as 1-week post-injection. By 6-10 weeks, NM accumulation is associated with severe LC neuron neurodegeneration and microglial engulfment of the pigment granules, while the anxiety-like behavior is abated. These phenotypes are reminiscent of LC dysfunction and cell death in AD and PD, validating this model for studying the consequences of pigment accumulation in the LC as it relates to neurodegenerative disease.
Collapse
Affiliation(s)
- Alexa F. Iannitelli
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Leslie Hassenein
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Bernard Mulvey
- Department of Psychiatry, Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Harris E. Blankenship
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| | - L. Cameron Liles
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Amanda L. Sharpe
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, 73117
| | - Jean-Francoise Pare
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Arielle Segal
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Steven A. Sloan
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Katharine E. McCann
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Joseph D. Dougherty
- Department of Psychiatry, Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yoland Smith
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Michael J. Beckstead
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| | - David Weinshenker
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
4
|
Laurencin C, Lancelot S, Brosse S, Mérida I, Redouté J, Greusard E, Lamberet L, Liotier V, Le Bars D, Costes N, Thobois S, Boulinguez P, Ballanger B. Noradrenergic alterations in Parkinson's disease: a combined 11C-yohimbine PET/neuromelanin MRI study. Brain 2024; 147:1377-1388. [PMID: 37787503 PMCID: PMC10994534 DOI: 10.1093/brain/awad338] [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] [Received: 06/27/2023] [Revised: 09/05/2023] [Accepted: 09/19/2023] [Indexed: 10/04/2023] Open
Abstract
Degeneration of the noradrenergic system is now considered a pathological hallmark of Parkinson's disease, but little is known about its consequences in terms of parkinsonian manifestations. Here, we evaluated two aspects of the noradrenergic system using multimodal in vivo imaging in patients with Parkinson's disease and healthy controls: the pigmented cell bodies of the locus coeruleus with neuromelanin sensitive MRI; and the density of α2-adrenergic receptors (ARs) with PET using 11C-yohimbine. Thirty patients with Parkinson's disease and 30 age- and sex-matched healthy control subjects were included. The characteristics of the patients' symptoms were assessed using the Movement Disorder Society Unified Parkinson's Disease Rating Scale (MDS-UPDRS). Patients showed reduced neuromelanin signal intensity in the locus coeruleus compared with controls and diminished 11C-yohimbine binding in widespread cortical regions, including the motor cortex, as well as in the insula, thalamus and putamen. Clinically, locus coeruleus neuronal loss was correlated with motor (bradykinesia, motor fluctuations, tremor) and non-motor (fatigue, apathy, constipation) symptoms. A reduction of α2-AR availability in the thalamus was associated with tremor, while a reduction in the putamen, the insula and the superior temporal gyrus was associated with anxiety. These results highlight a multifaceted alteration of the noradrenergic system in Parkinson's disease since locus coeruleus and α2-AR degeneration were found to be partly uncoupled. These findings raise important issues about noradrenergic dysfunction that may encourage the search for new drugs targeting this system, including α2-ARs, for the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Chloé Laurencin
- Lyon Neuroscience Research Center (CRNL), INSERM U1028, CNRS UMR5292, University Lyon 1, F-69000 Lyon, France
- Department of Neurology C, Expert Parkinson Centre, Hospices Civils de Lyon, Pierre Wertheimer Neurological Hospital, NS-Park/F-CRIN, 69500 Bron, France
| | - Sophie Lancelot
- Lyon Neuroscience Research Center (CRNL), INSERM U1028, CNRS UMR5292, University Lyon 1, F-69000 Lyon, France
- CERMEP-Imagerie du Vivant, PET-MRI Department, 69500 Bron, France
| | - Sarah Brosse
- Lyon Neuroscience Research Center (CRNL), INSERM U1028, CNRS UMR5292, University Lyon 1, F-69000 Lyon, France
| | - Inés Mérida
- CERMEP-Imagerie du Vivant, PET-MRI Department, 69500 Bron, France
| | - Jérôme Redouté
- CERMEP-Imagerie du Vivant, PET-MRI Department, 69500 Bron, France
| | - Elise Greusard
- CERMEP-Imagerie du Vivant, PET-MRI Department, 69500 Bron, France
| | - Ludovic Lamberet
- CERMEP-Imagerie du Vivant, PET-MRI Department, 69500 Bron, France
| | | | - Didier Le Bars
- CERMEP-Imagerie du Vivant, PET-MRI Department, 69500 Bron, France
| | - Nicolas Costes
- CERMEP-Imagerie du Vivant, PET-MRI Department, 69500 Bron, France
| | - Stéphane Thobois
- Department of Neurology C, Expert Parkinson Centre, Hospices Civils de Lyon, Pierre Wertheimer Neurological Hospital, NS-Park/F-CRIN, 69500 Bron, France
- Institut des Sciences Cognitives Marc Jeannerod, UMR 5229, CNRS, 69500 Bron, France
| | - Philippe Boulinguez
- Lyon Neuroscience Research Center (CRNL), INSERM U1028, CNRS UMR5292, University Lyon 1, F-69000 Lyon, France
| | - Bénédicte Ballanger
- Lyon Neuroscience Research Center (CRNL), INSERM U1028, CNRS UMR5292, University Lyon 1, F-69000 Lyon, France
| |
Collapse
|
5
|
Hayley S, Vahid-Ansari F, Sun H, Albert PR. Mood disturbances in Parkinson's disease: From prodromal origins to application of animal models. Neurobiol Dis 2023; 181:106115. [PMID: 37037299 DOI: 10.1016/j.nbd.2023.106115] [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: 08/26/2022] [Revised: 03/09/2023] [Accepted: 04/05/2023] [Indexed: 04/12/2023] Open
Abstract
Parkinson's disease (PD) is a complex illness with a constellation of environmental insults and genetic vulnerabilities being implicated. Strikingly, many studies only focus on the cardinal motor symptoms of the disease and fail to appreciate the major non-motor features which typically occur early in the disease process and are debilitating. Common comorbid psychiatric features, notably clinical depression, as well as anxiety and sleep disorders are thought to emerge before the onset of prominent motor deficits. In this review, we will delve into the prodromal stage of PD and how early neuropsychiatric pathology might unfold, followed by later motor disturbances. It is also of interest to discuss how animal models of PD capture the complexity of the illness, including depressive-like characteristics along with motor impairment. It remains to be determined how the underlying PD disease processes contributes to such comorbidity. But some of the environmental toxicants and microbial pathogens implicated in PD might instigate pro-inflammatory effects favoring α-synuclein accumulation and damage to brainstem neurons fueling the evolution of mood disturbances. We posit that comprehensive animal-based research approaches are needed to capture the complexity and time-dependent nature of the primary and co-morbid symptoms. This will allow for the possibility of early intervention with more novel and targeted treatments that fit with not only individual patient variability, but also with changes that occur over time with the evolution of the disease.
Collapse
Affiliation(s)
- S Hayley
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Ottawa Hospital Research Institute (Neuroscience), University of Ottawa, Canada.
| | - F Vahid-Ansari
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Ottawa Hospital Research Institute (Neuroscience), University of Ottawa, Canada
| | - H Sun
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Ottawa Hospital Research Institute (Neuroscience), University of Ottawa, Canada
| | - P R Albert
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Ottawa Hospital Research Institute (Neuroscience), University of Ottawa, Canada
| |
Collapse
|
6
|
Hosomoto K, Sasaki T, Yasuhara T, Kameda M, Sasada S, Kin I, Kuwahara K, Kawauchi S, Okazaki Y, Yabuno S, Sugahara C, Kawai K, Nagase T, Tanimoto S, Borlongan CV, Date I. Continuous vagus nerve stimulation exerts beneficial effects on rats with experimentally induced Parkinson's disease: Evidence suggesting involvement of a vagal afferent pathway. Brain Stimul 2023; 16:594-603. [PMID: 36914065 DOI: 10.1016/j.brs.2023.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 02/27/2023] [Accepted: 03/07/2023] [Indexed: 03/13/2023] Open
Abstract
BACKGROUND Vagus nerve stimulation (VNS) exerts neuroprotective and anti-inflammatory effects in preclinical models of central nervous system disorders, including Parkinson's disease (PD). VNS setting applied for experimental models is limited into single-time or intermittent short-duration stimulation. We developed a VNS device which could deliver continuous stimulation for rats. To date, the effects of vagal afferent- or efferent-selective stimulation on PD using continuous electrical stimulation remains to be determined. OBJECTIVE To investigate the effects of continuous and selective stimulation of vagal afferent or efferent fiber on Parkinsonian rats. METHODS Rats were divided into 5 group: intact VNS, afferent VNS (left VNS in the presence of left caudal vagotomy), efferent VNS (left VNS in the presence of left rostral vagotomy), sham, vagotomy. Rats underwent the implantation of cuff-electrode on left vagus nerve and 6-hydroxydopamine administration into the left striatum simultaneously. Electrical stimulation was delivered just after 6-OHDA administration and continued for 14 days. In afferent VNS and efferent VNS group, the vagus nerve was dissected at distal or proximal portion of cuff-electrode to imitate the selective stimulation of afferent or efferent vagal fiber respectively. RESULTS Intact VNS and afferent VNS reduced the behavioral impairments in cylinder test and methamphetamine-induced rotation test, which were accompanied by reduced inflammatory glial cells in substantia nigra with the increased density of the rate limiting enzyme in locus coeruleus. In contrast, efferent VNS did not exert any therapeutic effects. CONCLUSION Continuous VNS promoted neuroprotective and anti-inflammatory effect in experimental PD, highlighting the crucial role of the afferent vagal pathway in mediating these therapeutic outcomes.
Collapse
Affiliation(s)
- Kakeru Hosomoto
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Tatsuya Sasaki
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan.
| | - Takao Yasuhara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Masahiro Kameda
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan; Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Susumu Sasada
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Ittetsu Kin
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Ken Kuwahara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Satoshi Kawauchi
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Yosuke Okazaki
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Satoru Yabuno
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Chiaki Sugahara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Koji Kawai
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Takayuki Nagase
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Shun Tanimoto
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| | - Cesario V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL, 33611, USA
| | - Isao Date
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Okayama, Japan
| |
Collapse
|
7
|
Du Y, Choi S, Pilski A, Graves SM. Differential vulnerability of locus coeruleus and dorsal raphe neurons to chronic methamphetamine-induced degeneration. Front Cell Neurosci 2022; 16:949923. [PMID: 35936499 PMCID: PMC9354074 DOI: 10.3389/fncel.2022.949923] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 07/04/2022] [Indexed: 12/15/2022] Open
Abstract
Methamphetamine (meth) increases monoamine oxidase (MAO)-dependent mitochondrial stress in axons of substantia nigra pars compacta (SNc), and ventral tegmental area (VTA) dopamine neurons. Chronic administration of meth results in SNc degeneration and MAO inhibition is neuroprotective, whereas, the VTA is resistant to degeneration. This differential vulnerability is attributed, at least in part, to the presence of L-type Ca2+ channel-dependent mitochondrial stress in SNc but not VTA dopamine neurons. MAO is also expressed in other monoaminergic neurons such as noradrenergic locus coeruleus (LC) and serotonergic dorsal raphe (DR) neurons. The impact of meth on mitochondrial stress in LC and DR neurons is unknown. In the current study we used a genetically encoded redox biosensor to investigate meth-induced MAO-dependent mitochondrial stress in LC and DR neurons. Similar to SNc and VTA neurons, meth increased MAO-dependent mitochondrial stress in axonal but not somatic compartments of LC norepinephrine and DR serotonin neurons. Chronic meth administration (5 mg/kg; 28-day) resulted in degeneration of LC neurons and MAO inhibition was neuroprotective whereas DR neurons were resistant to degeneration. Activating L-type Ca2+ channels increased mitochondrial stress in LC but not DR axons and inhibiting L-type Ca2+ channels in vivo with isradipine prevented meth-induced LC degeneration. These data suggest that similar to recent findings in SNc and VTA dopamine neurons, the differential vulnerability between LC and DR neurons can be attributed to the presence of L-type Ca2+ channel-dependent mitochondrial stress. Taken together, the present study demonstrates that both meth-induced MAO- and L-type Ca2+ channel-dependent mitochondrial stress are necessary for chronic meth-induced neurodegeneration.
Collapse
|
8
|
Sola P, Krishnamurthy PT, Kumari M, Byran G, Gangadharappa HV, Garikapati KK. Neuroprotective approaches to halt Parkinson's disease progression. Neurochem Int 2022; 158:105380. [PMID: 35718278 DOI: 10.1016/j.neuint.2022.105380] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 02/07/2023]
Abstract
One of the most significant threats in Parkinson's disease (PD) is neurodegeneration. Neurodegeneration at both nigral as well as non-nigral regions of the brain is considered responsible for disease progression in PD. The key factors that initiate neurodegeneration are oxidative stress, neuroinflammation, mitochondrial complex-1 inhibition, and abnormal α-synuclein (SNCA) protein aggregations. Nigral neurodegeneration results in motor symptoms (tremor, bradykinesia, rigidity, shuffling gait, and postural instability) whereas; non-nigral neurodegeneration is responsible for non-motor symptoms (depression, cognitive dysfunctions, sleep disorders, hallucination, and psychosis). The available therapies for PD aim at increasing dopamine levels. The medications such as Monoamine oxidase B (MAO-B) inhibitors, catechol o-methyltransferase (COMT) inhibitors, Dopamine precursor (Levodopa), dopamine agonists, and dopamine reuptake inhibitors drastically improve the motor symptoms and quality of life only in the early stages of the disease. However, dopa resistant motor symptoms (abnormality in posture, speech impediment, gait, and balance problems), dopa resistant non-motor signs (sleep problems, autonomic dysfunction, mood, and cognitive impairment, pain), and drug-related side effects (motor fluctuations, psychosis, and dyskinesias) are considered responsible for the failure of these therapies. Further, none of the treatments, alone or in combination, are capable of halting the disease progression in the long run. Therefore, there is a need to develop safe and efficient neuroprotective agents, which can slow or stop the disease progression for the better management of PD. In this review, an effort has been made to discuss the various mechanisms responsible for progressive neurodegeneration (disease progression) in PD and also multiple strategies available for halting disease progression.
Collapse
Affiliation(s)
- Piyong Sola
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, 643001, India
| | - Praveen Thaggikuppe Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, 643001, India.
| | - Mamta Kumari
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, 643001, India
| | - Gowramma Byran
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, 643001, India
| | | | - Kusuma Kumari Garikapati
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, 643001, India
| |
Collapse
|
9
|
Sustained chemogenetic activation of locus coeruleus norepinephrine neurons promotes dopaminergic neuron survival in synucleinopathy. PLoS One 2022; 17:e0263074. [PMID: 35316276 PMCID: PMC8939823 DOI: 10.1371/journal.pone.0263074] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 01/11/2022] [Indexed: 01/21/2023] Open
Abstract
Dopaminergic neuron degeneration in the midbrain plays a pivotal role in motor symptoms associated with Parkinson's disease. However, non-motor symptoms of Parkinson's disease and post-mortem histopathology confirm dysfunction in other brain areas, including the locus coeruleus and its associated neurotransmitter norepinephrine. Here, we investigate the role of central norepinephrine-producing neurons in Parkinson's disease by chronically stimulating catecholaminergic neurons in the locus coeruleus using chemogenetic manipulation. We show that norepinephrine neurons send complex axonal projections to the dopaminergic neurons in the substantia nigra, confirming physical communication between these regions. Furthermore, we demonstrate that increased activity of norepinephrine neurons is protective against dopaminergic neuronal depletion in human α-syn A53T missense mutation over-expressing mice and prevents motor dysfunction in these mice. Remarkably, elevated norepinephrine neurons action fails to alleviate α-synuclein aggregation and microgliosis in the substantia nigra suggesting the presence of an alternate neuroprotective mechanism. The beneficial effects of high norepinephrine neuron activity might be attributed to the action of norepinephrine on dopaminergic neurons, as recombinant norepinephrine treatment increased primary dopaminergic neuron cultures survival and neurite sprouting. Collectively, our results suggest a neuroprotective mechanism where noradrenergic neurons activity preserves the integrity of dopaminergic neurons, which prevents synucleinopathy-dependent loss of these cells.
Collapse
|
10
|
Manto M. Is inferior olive central to the pathophysiology of essential tremor? No. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 163:167-187. [PMID: 35750362 DOI: 10.1016/bs.irn.2022.02.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Essential tremor (ET) represents one of the commonest movement disorder worldwide and is the most common tremor disorder. ET manifests with various combinations of motor and nonmotor symptoms. The clinical hallmark is a kinetic tremor of upper limbs. Historically, the pathogenesis of ET has been based on the hypothesis of an overactivity of the inferior olive (inferior olive hypothesis: IOH) where the inferior olive would act as the central pace-maker of ET, resulting in impaired electrophysiological discharges of the olivo-cerebellar tract. The absence of structural alterations in post-mortem studies of the inferior olive is a striking argument against the IOH. Furthermore, neuroimaging studies point towards the implication of the cerebello-thalamo-cerebral pathway rather than the IO, and the harmaline model which has been considered as an animal model of ET presents important weaknesses. By contrast, a series of experiments by Louis et al. have provided convincing evidence of impaired wiring of the Purkinje cell microcircuitry and progressive neurodegeneration of the cerebellar cortex. The Purkinje neuron appears as the primary culprit (Purkinjopathy). The cerebellar cortex hypothesis (CCH) has solid neuropathological signatures, unlike the purely physiological IOH. Rather than a dysregulatory electrophysiological disorder suggested by IOH, ET is a clinical-pathological entity similar to late onset neurodegenerative disorders such as Parkinson's disease or Alzheimer's disease. The CCH emphasizes the need to develop novel therapeutic strategies in order to maintain or promote the cerebellar reserve. The modern reconceptualization of ET in a genuine cerebellar disorder is cleaning the IOH to the light of histopathological studies. ET falls in the large basket of the neurodegenerative diseases and we have entered into a novel formulation of the disease pathogenesis with direct impacts on future therapies.
Collapse
Affiliation(s)
- Mario Manto
- Unité des Ataxies Cérébelleuses, Service de Neurologie, CHU-Charleroi, Belgium; Service des Neurosciences, Université de Mons, Belgium.
| |
Collapse
|
11
|
Crawford AH, Hildyard JCW, Rushing SAM, Wells DJ, Diez-Leon M, Piercy RJ. Validation of DE50-MD dogs as a model for the brain phenotype of Duchenne muscular dystrophy. Dis Model Mech 2022; 15:dmm049291. [PMID: 35019137 PMCID: PMC8906169 DOI: 10.1242/dmm.049291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/21/2021] [Indexed: 11/20/2022] Open
Abstract
Duchenne muscular dystrophy (DMD), a fatal musculoskeletal disease, is associated with neurodevelopmental disorders and cognitive impairment caused by brain dystrophin deficiency. Dog models of DMD represent key translational tools to study dystrophin biology and to develop novel therapeutics. However, characterisation of dystrophin expression and function in the canine brain is lacking. We studied the DE50-MD canine model of DMD that has a missense mutation in the donor splice site of exon 50. Using a battery of cognitive tests, we detected a neurocognitive phenotype in DE50-MD dogs, including reduced attention, problem solving and exploration of novel objects. Through a combination of capillary immunoelectrophoresis, immunolabelling, quantitative PCR and RNAScope in situ hybridisation, we show that regional dystrophin expression in the adult canine brain reflects that of humans, and that the DE50-MD dog lacks full-length dystrophin (Dp427) protein expression but retains expression of the two shorter brain-expressed isoforms, Dp140 and Dp71. Thus, the DE50-MD dog is a translationally relevant pre-clinical model to study the consequences of Dp427 deficiency in the brain and to develop therapeutic strategies for the neurological sequelae of DMD.
Collapse
Affiliation(s)
- Abbe H. Crawford
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London NW1 0TU, UK
| | - John C. W. Hildyard
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London NW1 0TU, UK
| | - Sophie A. M. Rushing
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London NW1 0TU, UK
| | - Dominic J. Wells
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London NW1 0TU, UK
| | - Maria Diez-Leon
- Pathobiology and Population Sciences, Royal Veterinary College, London AL9 7TA, UK
| | - Richard J. Piercy
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London NW1 0TU, UK
| |
Collapse
|
12
|
Zeng F, Fan Y, Brown RW, Drew Gill W, Price JB, Jones TC, Zhu MY. Effects of Manipulation of Noradrenergic Activities on the Expression of Dopaminergic Phenotypes in Aged Rat Brains. ASN Neuro 2021; 13:17590914211055064. [PMID: 34812056 PMCID: PMC8613899 DOI: 10.1177/17590914211055064] [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] [Indexed: 11/30/2022] Open
Abstract
This study investigated the effects of the pharmacological manipulation of noradrenergic activities on dopaminergic phenotypes in aged rats. Results showed that the administration of L-threo-3,4-dihydroxyphenylserine (L-DOPS) for 21 days significantly increased the expression of tyrosine hydroxylase (TH) and dopamine transporter (DAT) in the striatum and substantia nigra (SN) of 23-month-old rats. Furthermore, this treatment significantly increased norepinephrine/DA concentrations in the striatum and caused a deficit of sensorimotor gating as measured by prepulse inhibition (PPI). Next, old rats were injected with the α2-adrenoceptor antagonist 2-methoxy idazoxan or β2-adrenoceptor agonist salmeterol for 21 days. Both drugs produced similar changes of TH and DAT in the striatum and SN. Moreover, treatments with L-DOPS, 2-methoxy idazoxan, or salmeterol significantly increased the protein levels of phosphorylated Akt in rat striatum and SN. However, although a combination of 2-methoxy idazoxan and salmeterol resulted in a deficit of PPI in these rats, the administration of 2-methoxy idazoxan alone showed an opposite behavioral change. The in vitro experiments revealed that treatments with norepinephrine markedly increased mRNAs and proteins of ATF2 and CBP/p300 and reduced mRNA and proteins of HDAC2 and HDAC5 in MN9D cells. A ChIP assay showed that norepinephrine significantly increased CBP/p300 binding or reduced HDAC2 and HDAC5 binding on the TH promoter. The present results indicate that facilitating noradrenergic activity in the brain can improve the functions of dopaminergic neurons in aged animals. While this improvement may have biochemically therapeutic indication for the status involving the degeneration of dopaminergic neurons, it may not definitely include behavioral improvements, as indicated by using 2-methoxy idazoxan only.
Collapse
Affiliation(s)
- Fei Zeng
- Department of Neurology, Renmin Hospital of the Wuhan University, China.,Departments of Biomedical Sciences, Quillen College of Medicine, 4154East Tennessee State University, USA
| | - Yan Fan
- Departments of Biomedical Sciences, Quillen College of Medicine, 4154East Tennessee State University, USA.,Department of Biochemistry, Nantong University College of Medicine, China
| | - Russell W Brown
- Departments of Biomedical Sciences, Quillen College of Medicine, 4154East Tennessee State University, USA
| | - Wesley Drew Gill
- Departments of Biomedical Sciences, Quillen College of Medicine, 4154East Tennessee State University, USA
| | - Jennifer B Price
- Department of Biological Sciences, College of Arts and Sciences, 4154East Tennessee State University, USA
| | - Thomas C Jones
- Department of Biological Sciences, College of Arts and Sciences, 4154East Tennessee State University, USA
| | - Meng-Yang Zhu
- Departments of Biomedical Sciences, Quillen College of Medicine, 4154East Tennessee State University, USA
| |
Collapse
|
13
|
Tirassa P, Schirinzi T, Raspa M, Ralli M, Greco A, Polimeni A, Possenti R, Mercuri NB, Severini C. What substance P might tell us about the prognosis and mechanism of Parkinson's disease? Neurosci Biobehav Rev 2021; 131:899-911. [PMID: 34653503 DOI: 10.1016/j.neubiorev.2021.10.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 10/05/2021] [Indexed: 10/20/2022]
Abstract
The neuropeptide substance P (SP) plays an important role in neurodegenerative disorders, among which Parkinson's disease (PD). In the present work we have reviewed the involvement of SP and its preferred receptor (NK1-R) in motor and non-motor PD symptoms, in both PD animal models and patients. Despite PD is primarily a motor disorder, non-motor abnormalities, including olfactory deficits and gastrointestinal dysfunctions, can represent diagnostic PD predictors, according to the hypothesis that the olfactory and the enteric nervous system represent starting points of neurodegeneration, ascending to the brain via the sympathetic fibers and the vagus nerve. In PD patients, the α-synuclein aggregates in the olfactory bulb and the gastrointestinal tract, as well as in the dorsal motor nucleus of the vagus nerve often co-localize with SP, indicating SP-positive neurons as highly vulnerable sites of degeneration. Considering the involvement of the SP/NK1-R in both the periphery and specific brain areas, this system might represent a neuronal substrate for the symptom and disease progression, as well as a therapeutic target for PD.
Collapse
Affiliation(s)
- Paola Tirassa
- Institute of Biochemistry and Cell Biology, National Research Council, Sapienza University of Rome, Viale del Policlinico, 155, 00161, Rome, Italy.
| | - Tommaso Schirinzi
- Department of Systems Medicine, University of Rome "Tor Vergata", Via della Ricerca Scientifica 1, 00133, Rome, Italy
| | - Marcello Raspa
- Institute of Biochemistry and Cell Biology, National Research Council, Sapienza University of Rome, Viale del Policlinico, 155, 00161, Rome, Italy
| | - Massimo Ralli
- Department of Sense Organs, Sapienza University of Rome, Viale del Policlinico, 155, 00161, Rome, Italy
| | - Antonio Greco
- Department of Sense Organs, Sapienza University of Rome, Viale del Policlinico, 155, 00161, Rome, Italy
| | - Antonella Polimeni
- Department of Sense Organs, Sapienza University of Rome, Viale del Policlinico, 155, 00161, Rome, Italy
| | - Roberta Possenti
- Department of Systems Medicine, University of Rome "Tor Vergata", Via della Ricerca Scientifica 1, 00133, Rome, Italy
| | - Nicola Biagio Mercuri
- Department of Systems Medicine, University of Rome "Tor Vergata", Via della Ricerca Scientifica 1, 00133, Rome, Italy
| | - Cinzia Severini
- Institute of Biochemistry and Cell Biology, National Research Council, Sapienza University of Rome, Viale del Policlinico, 155, 00161, Rome, Italy.
| |
Collapse
|
14
|
Lazzeri G, Busceti CL, Biagioni F, Fabrizi C, Morucci G, Giorgi FS, Ferrucci M, Lenzi P, Puglisi-Allegra S, Fornai F. Norepinephrine Protects against Methamphetamine Toxicity through β2-Adrenergic Receptors Promoting LC3 Compartmentalization. Int J Mol Sci 2021; 22:7232. [PMID: 34281286 PMCID: PMC8269332 DOI: 10.3390/ijms22137232] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/01/2021] [Accepted: 07/03/2021] [Indexed: 01/18/2023] Open
Abstract
Norepinephrine (NE) neurons and extracellular NE exert some protective effects against a variety of insults, including methamphetamine (Meth)-induced cell damage. The intimate mechanism of protection remains difficult to be analyzed in vivo. In fact, this may occur directly on target neurons or as the indirect consequence of NE-induced alterations in the activity of trans-synaptic loops. Therefore, to elude neuronal networks, which may contribute to these effects in vivo, the present study investigates whether NE still protects when directly applied to Meth-treated PC12 cells. Meth was selected based on its detrimental effects along various specific brain areas. The study shows that NE directly protects in vitro against Meth-induced cell damage. The present study indicates that such an effect fully depends on the activation of plasma membrane β2-adrenergic receptors (ARs). Evidence indicates that β2-ARs activation restores autophagy, which is impaired by Meth administration. This occurs via restoration of the autophagy flux and, as assessed by ultrastructural morphometry, by preventing the dissipation of microtubule-associated protein 1 light chain 3 (LC3) from autophagy vacuoles to the cytosol, which is produced instead during Meth toxicity. These findings may have an impact in a variety of degenerative conditions characterized by NE deficiency along with autophagy impairment.
Collapse
Affiliation(s)
- Gloria Lazzeri
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126 Pisa, Italy; (G.L.); (G.M.); (F.S.G.); (M.F.); (P.L.)
| | - Carla L. Busceti
- I.R.C.C.S. Neuromed, via Atinense 18, 86077 Pozzilli, Italy; (C.L.B.); (F.B.); (S.P.-A.)
| | - Francesca Biagioni
- I.R.C.C.S. Neuromed, via Atinense 18, 86077 Pozzilli, Italy; (C.L.B.); (F.B.); (S.P.-A.)
| | - Cinzia Fabrizi
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, via A. Borelli 50, 00161 Rome, Italy;
| | - Gabriele Morucci
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126 Pisa, Italy; (G.L.); (G.M.); (F.S.G.); (M.F.); (P.L.)
| | - Filippo S. Giorgi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126 Pisa, Italy; (G.L.); (G.M.); (F.S.G.); (M.F.); (P.L.)
| | - Michela Ferrucci
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126 Pisa, Italy; (G.L.); (G.M.); (F.S.G.); (M.F.); (P.L.)
| | - Paola Lenzi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126 Pisa, Italy; (G.L.); (G.M.); (F.S.G.); (M.F.); (P.L.)
| | | | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126 Pisa, Italy; (G.L.); (G.M.); (F.S.G.); (M.F.); (P.L.)
- I.R.C.C.S. Neuromed, via Atinense 18, 86077 Pozzilli, Italy; (C.L.B.); (F.B.); (S.P.-A.)
| |
Collapse
|
15
|
Cui K, Yang F, Tufan T, Raza MU, Zhan Y, Fan Y, Zeng F, Brown RW, Price JB, Jones TC, Miller GW, Zhu MY. Restoration of Noradrenergic Function in Parkinson's Disease Model Mice. ASN Neuro 2021; 13:17590914211009730. [PMID: 33940943 PMCID: PMC8114769 DOI: 10.1177/17590914211009730] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Dysfunction of the central noradrenergic and dopaminergic systems is the primary neurobiological characteristic of Parkinson’s disease (PD). Importantly, neuronal loss in the locus coeruleus (LC) that occurs in early stages of PD may accelerate progressive loss of dopaminergic neurons. Therefore, restoring the activity and function of the deficient noradrenergic system may be an important therapeutic strategy for early PD. In the present study, the lentiviral constructions of transcription factors Phox2a/2b, Hand2 and Gata3, either alone or in combination, were microinjected into the LC region of the PD model VMAT2 Lo mice at 12 and 18 month age. Biochemical analysis showed that microinjection of lentiviral expression cassettes into the LC significantly increased mRNA levels of Phox2a, and Phox2b, which were accompanied by parallel increases of mRNA and proteins of dopamine β-hydroxylase (DBH) and tyrosine hydroxylase (TH) in the LC. Furthermore, there was considerable enhancement of DBH protein levels in the frontal cortex and hippocampus, as well as enhanced TH protein levels in the striatum and substantia nigra. Moreover, these manipulations profoundly increased norepinephrine and dopamine concentrations in the striatum, which was followed by a remarkable improvement of the spatial memory and locomotor behavior. These results reveal that over-expression of these transcription factors in the LC improves noradrenergic and dopaminergic activities and functions in this rodent model of PD. It provides the necessary groundwork for the development of gene therapies of PD, and expands our understanding of the link between the LC-norepinephrine and dopamine systems during the progression of PD.
Collapse
Affiliation(s)
- Kui Cui
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, United States
| | - Fan Yang
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, United States.,Hong Kong Institute, Asia Metropolitan University, Hong Kong, China
| | - Turan Tufan
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, United States
| | - Muhammad U Raza
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, United States
| | - Yanqiang Zhan
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, United States.,Department of Neurology, Renmin Hospital of the Wuhan University, Wuhan, China
| | - Yan Fan
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, United States.,Department of Biochemistry, Nantong University College of Medicine, Nantong, China
| | - Fei Zeng
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, United States.,Department of Neurology, Renmin Hospital of the Wuhan University, Wuhan, China
| | - Russell W Brown
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, United States
| | - Jennifer B Price
- Department of Biological Sciences, College of Arts and Sciences; East Tennessee State University, Johnson City, United States
| | - Thomas C Jones
- Department of Biological Sciences, College of Arts and Sciences; East Tennessee State University, Johnson City, United States
| | - Gary W Miller
- Department of Environmental Health Sciences, Mailmen School of Public Health, Columbia University, New York, New York, United States
| | - Meng-Yang Zhu
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, United States
| |
Collapse
|
16
|
Perkins JE, Janzen A, Bernhard FP, Wilhelm K, Brien DC, Huang J, Coe BC, Vadasz D, Mayer G, Munoz DP, Oertel WH. Saccade, Pupil, and Blink Responses in Rapid Eye Movement Sleep Behavior Disorder. Mov Disord 2021; 36:1720-1726. [PMID: 33754406 PMCID: PMC8359943 DOI: 10.1002/mds.28585] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 01/30/2021] [Accepted: 03/02/2021] [Indexed: 02/03/2023] Open
Abstract
Background Parkinson's disease (PD) patients exhibit deficits in saccade performance, pupil function, and blink rate. Isolated REM (rapid eye movement) Sleep Behavior Disorder (RBD) is a harbinger to PD making them candidates to investigate for early oculomotor abnormalities as PD biomarkers. Objectives We tested whether saccade, pupillary, and blink responses in RBD were similar to PD. Methods RBD (n = 22), PD (n = 22) patients, and healthy controls (CTRL) (n = 74) were studied with video‐based eye‐tracking. Results RBD patients did not have significantly different saccadic behavior compared to CTRL, but PD patients differed from CTRL and RBD. Both patient groups had significantly lower blink rates, dampened pupil constriction, and dilation responses compared to CTRL. Conclusion RBD and PD patients had altered pupil and blink behavior compared to CTRL. Because RBD saccade parameters were comparable to CTRL, pupil and blink brain areas may be impacted before saccadic control areas, making them potential prodromal PD biomarkers. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society
Collapse
Affiliation(s)
- Julia E Perkins
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| | - Annette Janzen
- Department of Neurology, Philipps-University, Marburg, Germany
| | | | - Karén Wilhelm
- Department of Neurology, Philipps-University, Marburg, Germany
| | - Donald C Brien
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| | - Jeff Huang
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| | - Brian C Coe
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| | - David Vadasz
- Department of Neurology, Philipps-University, Marburg, Germany
| | - Geert Mayer
- Department of Neurology, Philipps-University, Marburg, Germany
| | - Douglas P Munoz
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada.,Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | | |
Collapse
|
17
|
Kinnerup MB, Sommerauer M, Damholdt MF, Schaldemose JL, Ismail R, Terkelsen AJ, Stær K, Hansen A, Fedorova TD, Knudsen K, Skjærbæk C, Borghammer P, Pavese N, Brooks DJ, Nahimi A. Preserved noradrenergic function in Parkinson's disease patients with rest tremor. Neurobiol Dis 2021; 152:105295. [PMID: 33549722 DOI: 10.1016/j.nbd.2021.105295] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 01/27/2021] [Accepted: 02/03/2021] [Indexed: 11/28/2022] Open
Abstract
Noradrenergic neurotransmission may play an important role in tremor modulation through its innervation of key structures of the central tremor circuits. Here, Parkinson's disease (PD) patients with (PDT+) or without (PDT-) rest tremor had 11C-methylreboxetine(11C-MeNER) positron emission tomography (PET) to test the hypothesis that noradrenaline terminal function was relatively preserved in PDT+ compared to PDT-. METHODS Sixty-five PD patients and 28 healthy controls (HC) were scanned with 11C-MeNER PET. Patients were categorized as PDT+ if subscores in UPDRS-III item 3 or MDS-UPDRS-III item 17 was ≥2; remaining were categorized as PDT-. Simplified reference tissue model 2 distribution volume ratios (DVR) for 11C-MeNER were calculated for thalamus, dorsal and median raphe, locus coeruleus (LC) and red nucleus using time activity curves (TACs) obtained from volumes of interest (VOI). Data were statistically interrogated with a general linear mixed model using 'region', and 'group' as factors and the interaction of 'region x group' was examined. RESULTS Tremor positive PD patients had a significantly higher mean 11C-MeNER DVR compared to PDT- in LC and thalamus. The PDT+ mean LC DVR was similar to that of HC. PDT+ mean 11C-MeNER DVRs were significantly lower than HC in the dorsal raphe while the PDT- group showed significantly lower mean 11C-MeNER DVR across all regions compared to HC. CONCLUSION While both PD T+ and PD T- groups showed a significant loss of noradrenaline terminal function compared to controls, noradrenergic neurons were relatively preserved in PDT+ in LC and thalamus. The greater loss of noradrenergic transporters in PDT- in LC and thalamus compared with PDT+ is in line with earlier in-vitro studies and could potentially contribute to their tremor negative phenotype.
Collapse
Affiliation(s)
- Martin B Kinnerup
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, 8200 Aarhus N, Denmark.
| | - Michael Sommerauer
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, 8200 Aarhus N, Denmark; Department of Neurology, University Hospital Cologne and Faculty of Medicine, University of Cologne, 50937 Köln, Germany
| | - Malene F Damholdt
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, 8200 Aarhus N, Denmark; Department for Psychology and Behavioural Sciences, Aarhus University, 8000 Aarhus, Denmark
| | - Jeppe L Schaldemose
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Rola Ismail
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Astrid J Terkelsen
- Department of Neurology, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Kristian Stær
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Allan Hansen
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Tatyana D Fedorova
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Karoline Knudsen
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Casper Skjærbæk
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Per Borghammer
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Nicola Pavese
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, 8200 Aarhus N, Denmark; Translational and Clinical Research Institute, Newcastle University, Newcastle NE1 7RU, United Kingdom
| | - David J Brooks
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, 8200 Aarhus N, Denmark; Translational and Clinical Research Institute, Newcastle University, Newcastle NE1 7RU, United Kingdom; Department of Brain Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Adjmal Nahimi
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, 8200 Aarhus N, Denmark; Department of Neurology, Restorative Parkinson Unit, Universitetssjukhuset, 22184 Lund, Sweden
| |
Collapse
|
18
|
Galgani A, Lombardo F, Della Latta D, Martini N, Bonuccelli U, Fornai F, Giorgi FS. Locus Coeruleus Magnetic Resonance Imaging in Neurological Diseases. Curr Neurol Neurosci Rep 2020; 21:2. [PMID: 33313963 PMCID: PMC7732795 DOI: 10.1007/s11910-020-01087-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2020] [Indexed: 01/01/2023]
Abstract
PURPOSE OF REVIEW Locus coeruleus (LC) is the main noradrenergic nucleus of the brain, and its degeneration is considered to be key in the pathogenesis of neurodegenerative diseases. In the last 15 years,MRI has been used to assess LC in vivo, both in healthy subjects and in patients suffering from neurological disorders. In this review, we summarize the main findings of LC-MRI studies, interpreting them in light of preclinical and histopathological data, and discussing its potential role as diagnostic and experimental tool. RECENT FINDINGS LC-MRI findings were largely in agreement with neuropathological evidences; LC signal showed to be not significantly affected during normal aging and to correlate with cognitive performances. On the contrary, a marked reduction of LC signal was observed in patients suffering from neurodegenerative disorders, with specific features. LC-MRI is a promising tool, which may be used in the future to explore LC pathophysiology as well as an early biomarker for degenerative diseases.
Collapse
Affiliation(s)
| | - Francesco Lombardo
- U.O.C. "Risonanza Magnetica Specialistica e Neuroradiologia", Fondazione "G. Monasterio"- National Research Council/Tuscany Region, Pisa, Italy
| | - Daniele Della Latta
- Deep Health Unit, Fondazione "G. Monasterio"- National Research Council/Tuscany Region, Pisa, Italy
| | - Nicola Martini
- Deep Health Unit, Fondazione "G. Monasterio"- National Research Council/Tuscany Region, Pisa, Italy
| | | | - Francesco Fornai
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Filippo Sean Giorgi
- Neurology Unit, Pisa University Hospital, Pisa, Italy.
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy.
| |
Collapse
|
19
|
Locus Coeruleus Modulates Neuroinflammation in Parkinsonism and Dementia. Int J Mol Sci 2020; 21:ijms21228630. [PMID: 33207731 PMCID: PMC7697920 DOI: 10.3390/ijms21228630] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 12/11/2022] Open
Abstract
Locus Coeruleus (LC) is the main noradrenergic nucleus of the central nervous system, and its neurons widely innervate the whole brain. LC is severely degenerated both in Alzheimer’s disease (AD) and in Parkinson’s disease (PD), years before the onset of clinical symptoms, through mechanisms that differ among the two disorders. Several experimental studies have shown that noradrenaline modulates neuroinflammation, mainly by acting on microglia/astrocytes function. In the present review, after a brief introduction on the anatomy and physiology of LC, we provide an overview of experimental data supporting a pathogenetic role of LC degeneration in AD and PD. Then, we describe in detail experimental data, obtained in vitro and in vivo in animal models, which support a potential role of neuroinflammation in such a link, and the specific molecules (i.e., released cytokines, glial receptors, including pattern recognition receptors and others) whose expression is altered by LC degeneration and might play a key role in AD/PD pathogenesis. New imaging and biochemical tools have recently been developed in humans to estimate in vivo the integrity of LC, the degree of neuroinflammation, and pathology AD/PD biomarkers; it is auspicable that these will allow in the near future to test the existence of a link between LC-neuroinflammation and neurodegeneration directly in patients.
Collapse
|
20
|
Fan Y, Zeng F, Brown RW, Price JB, Jones TC, Zhu MY. Transcription Factors Phox2a/2b Upregulate Expression of Noradrenergic and Dopaminergic Phenotypes in Aged Rat Brains. Neurotox Res 2020; 38:793-807. [PMID: 32617854 PMCID: PMC7484387 DOI: 10.1007/s12640-020-00250-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/30/2020] [Accepted: 06/25/2020] [Indexed: 12/14/2022]
Abstract
The present study investigated the effects of forced overexpression of Phox2a/2b, two transcription factors, in the locus coeruleus (LC) of aged rats on noradrenergic and dopaminergic phenotypes in brains. Results showed that a significant increase in Phox2a/2b mRNA levels in the LC region was paralleled by marked enhancement in expression of DBH and TH per se. Furthermore, similar increases in TH protein levels were observed in the substantial nigra and striatum, as well as in the hippocampus and frontal cortex. Overexpression of Phox2 genes also significantly increased BrdU-positive cells in the hippocampal dentate gyrus and NE levels in the striatum. Moreover, this manipulation significantly improved the cognition behavior. The in vitro experiments revealed that norepinephrine treatments may increase the transcription of TH gene through the epigenetic action on the TH promoter. The results indicate that Phox2 genes may play an important role in improving the function of the noradrenergic and dopaminergic neurons in aged animals, and regulation of Phox2 gene expression may have therapeutic utility in aging or disorders involving degeneration of noradrenergic neurons.
Collapse
Affiliation(s)
- Yan Fan
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
- Department of Biochemistry, Nantong University College of Medicine, Nantong, China
| | - Fei Zeng
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
- Department of Neurology, Remin Hospital of the Wuhan University, Wuhan, China
| | - Russell W Brown
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Jennifer B Price
- Department of Biological Sciences, College of Arts and Sciences, East Tennessee State University, Johnson City, TN, USA
| | - Thomas C Jones
- Department of Biological Sciences, College of Arts and Sciences, East Tennessee State University, Johnson City, TN, USA
| | - Meng-Yang Zhu
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.
| |
Collapse
|
21
|
Butkovich LM, Houser MC, Chalermpalanupap T, Porter-Stransky KA, Iannitelli AF, Boles JS, Lloyd GM, Coomes AS, Eidson LN, De Sousa Rodrigues ME, Oliver DL, Kelly SD, Chang J, Bengoa-Vergniory N, Wade-Martins R, Giasson BI, Joers V, Weinshenker D, Tansey MG. Transgenic Mice Expressing Human α-Synuclein in Noradrenergic Neurons Develop Locus Ceruleus Pathology and Nonmotor Features of Parkinson's Disease. J Neurosci 2020; 40:7559-7576. [PMID: 32868457 PMCID: PMC7511194 DOI: 10.1523/jneurosci.1468-19.2020] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 08/02/2020] [Accepted: 08/09/2020] [Indexed: 12/11/2022] Open
Abstract
Degeneration of locus ceruleus (LC) neurons and dysregulation of noradrenergic signaling are ubiquitous features of Parkinson's disease (PD). The LC is among the first brain regions affected by α-synuclein (asyn) pathology, yet how asyn affects these neurons remains unclear. LC-derived norepinephrine (NE) can stimulate neuroprotective mechanisms and modulate immune cells, while dysregulation of NE neurotransmission may exacerbate disease progression, particularly nonmotor symptoms, and contribute to the chronic neuroinflammation associated with PD pathology. Although transgenic mice overexpressing asyn have previously been developed, transgene expression is usually driven by pan-neuronal promoters and thus has not been selectively targeted to LC neurons. Here we report a novel transgenic mouse expressing human wild-type asyn under control of the noradrenergic-specific dopamine β-hydroxylase promoter (DBH-hSNCA). These mice developed oligomeric and conformation-specific asyn in LC neurons, alterations in hippocampal and LC microglial abundance, upregulated GFAP expression, degeneration of LC fibers, decreased striatal DA metabolism, and age-dependent behaviors reminiscent of nonmotor symptoms of PD that were rescued by adrenergic receptor antagonists. These mice provide novel insights into how asyn pathology affects LC neurons and how central noradrenergic dysfunction may contribute to early PD pathophysiology.SIGNIFICANCE STATEMENT ɑ-Synuclein (asyn) pathology and loss of neurons in the locus ceruleus (LC) are two of the most ubiquitous neuropathologic features of Parkinson's disease (PD). Dysregulated norepinephrine (NE) neurotransmission is associated with the nonmotor symptoms of PD, including sleep disturbances, emotional changes such as anxiety and depression, and cognitive decline. Importantly, the loss of central NE may contribute to the chronic inflammation in, and progression of, PD. We have generated a novel transgenic mouse expressing human asyn in LC neurons to investigate how increased asyn expression affects the function of the central noradrenergic transmission and associated behaviors. We report cytotoxic effects of oligomeric and conformation-specific asyn, astrogliosis, LC fiber degeneration, disruptions in striatal dopamine metabolism, and age-dependent alterations in nonmotor behaviors without inclusions.
Collapse
Affiliation(s)
| | | | - Termpanit Chalermpalanupap
- Laney Graduate School, Emory University, Atlanta, Georgia 30322
- Department of Human Genetics, Emory School of Medicine, Atlanta, Georgia 30322
| | - Kirsten A Porter-Stransky
- Department of Human Genetics, Emory School of Medicine, Atlanta, Georgia 30322
- Department of Biomedical Sciences, Homer Stryker M.D. School of Medicine, Western Michigan University, Kalamazoo, Michigan 49008
| | - Alexa F Iannitelli
- Department of Human Genetics, Emory School of Medicine, Atlanta, Georgia 30322
| | - Jake S Boles
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - Grace M Lloyd
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - Alexandra S Coomes
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - Lori N Eidson
- Department of Physiology, Emory School of Medicine, Atlanta, Georgia 30322
| | | | | | - Sean D Kelly
- Laney Graduate School, Emory University, Atlanta, Georgia 30322
| | - Jianjun Chang
- Laney Graduate School, Emory University, Atlanta, Georgia 30322
| | - Nora Bengoa-Vergniory
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, United Kingdom
| | - Richard Wade-Martins
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, United Kingdom
| | - Benoit I Giasson
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - Valerie Joers
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - David Weinshenker
- Department of Human Genetics, Emory School of Medicine, Atlanta, Georgia 30322
| | - Malú Gámez Tansey
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida 32610
- Susan and Normal Fixel Chair in Parkinson's Disease, Normal Fixel Institute for Neurological Diseases, University of Florida Health, Gainesville, Florida 32610
| |
Collapse
|
22
|
Andersen KB, Hansen AK, Sommerauer M, Fedorova TD, Knudsen K, Vang K, Van Den Berge N, Kinnerup M, Nahimi A, Pavese N, Brooks DJ, Borghammer P. Altered sensorimotor cortex noradrenergic function in idiopathic REM sleep behaviour disorder – A PET study. Parkinsonism Relat Disord 2020; 75:63-69. [DOI: 10.1016/j.parkreldis.2020.05.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 02/27/2020] [Accepted: 05/08/2020] [Indexed: 12/15/2022]
|
23
|
Paredes-Rodriguez E, Vegas-Suarez S, Morera-Herreras T, De Deurwaerdere P, Miguelez C. The Noradrenergic System in Parkinson's Disease. Front Pharmacol 2020; 11:435. [PMID: 32322208 PMCID: PMC7157437 DOI: 10.3389/fphar.2020.00435] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 03/20/2020] [Indexed: 12/16/2022] Open
Abstract
Nowadays it is well accepted that in Parkinson’s disease (PD), the neurodegenerative process occurs in stages and that damage to other areas precedes the neuronal loss in the substantia nigra pars compacta, which is considered a pathophysiological hallmark of PD. This heterogeneous and progressive neurodegeneration may explain the diverse symptomatology of the disease, including motor and non-motor alterations. In PD, one of the first areas undergoing degeneration is the locus coeruleus (LC). This noradrenergic nucleus provides extensive innervation throughout the brain and plays a fundamental neuromodulator role, participating in stress responses, emotional memory, and control of motor, sensory, and autonomic functions. Early in the disease, LC neurons suffer modifications that can condition the effectiveness of pharmacological treatments, and importantly, can lead to the appearance of common non-motor symptomatology. The noradrenergic system also exerts anti-inflammatory and neuroprotective effect on the dopaminergic degeneration and noradrenergic damage can consequently condition the progress of the disease. From the pharmacological point of view, it is also important to understand how the noradrenergic system performs in PD, since noradrenergic medication is often used in these patients, and drug interactions can take place when combining them with the gold standard drug therapy in PD, L-3,4-dihydroxyphenylalanine (L-DOPA). This review provides an overview about the functional status of the noradrenergic system in PD and its contribution to the efficacy of pharmacological-based treatments. Based on preclinical and clinical publications, a special attention will be dedicated to the most prevalent non-motor symptoms of the disease.
Collapse
Affiliation(s)
- Elena Paredes-Rodriguez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.,Autonomic and Movement Disorders Unit, Neurodegenerative Diseases, Biocruces Health Research Institute, Barakaldo, Spain
| | - Sergio Vegas-Suarez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.,Autonomic and Movement Disorders Unit, Neurodegenerative Diseases, Biocruces Health Research Institute, Barakaldo, Spain
| | - Teresa Morera-Herreras
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.,Autonomic and Movement Disorders Unit, Neurodegenerative Diseases, Biocruces Health Research Institute, Barakaldo, Spain
| | - Philippe De Deurwaerdere
- Centre National de la Recherche scientifique, Institut des Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA UMR 5287), Bordeaux, France
| | - Cristina Miguelez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.,Autonomic and Movement Disorders Unit, Neurodegenerative Diseases, Biocruces Health Research Institute, Barakaldo, Spain
| |
Collapse
|
24
|
Atomoxetine Does Not Improve Complex Attention in Idiopathic Parkinson's Disease Patients with Cognitive Deficits: A Meta-Analysis. PARKINSONS DISEASE 2020; 2020:4853590. [PMID: 32211146 PMCID: PMC7049416 DOI: 10.1155/2020/4853590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022]
Abstract
Objectives To evaluate the effects of atomoxetine on complex attention and other neurocognitive domains in idiopathic Parkinson's disease (PD). Methods Interventional trials reporting changes in complex attention and other neurocognitive functions (Diagnostic and Statistical Manual of Mental Disorders-5) following administration of atomoxetine for at least 8 weeks in adults with idiopathic PD were included. Effect sizes (Cohen's d), the standardized mean difference in the scores of each cognitive domain, were compared using a random-effects model (MetaXL version 5.3). Results Three studies were included in the final analysis. For a change in complex attention in PD with mild cognitive impairment (MCI), the estimated effect size was small and nonsignificant (0.16 (95% CI: −0.09, 0.42), n = 42). For changes in executive function, perceptual-motor function, language, social cognition, and learning and memory, the estimated effect sizes were small and medium, but nonsignificant. A deteriorative trend in executive function was observed after atomoxetine treatment in PD with MCI. For a change in global cognitive function in PD without MCI, the estimated effect size was large and significant. Conclusion In idiopathic PD with MCI, atomoxetine does not improve complex attention. Also, a deteriorative trend in the executive function was noted.
Collapse
|
25
|
MPTP-Induced Impairment of Cardiovascular Function. Neurotox Res 2020; 38:27-37. [PMID: 32198706 DOI: 10.1007/s12640-020-00182-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 02/10/2020] [Accepted: 02/14/2020] [Indexed: 02/07/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the accumulation of Lewy bodies and loss of dopaminergic neurons in the substantia nigra pars compacta (SNpC). MPTP is widely used to generate murine PD model. In addition to classical motor disorders, PD patients usually have non-motor symptoms related to autonomic impairment, which precedes decades before the motor dysfunction. This study's objective is to examine the effects of MPTP on noradrenergic neurons in the hindbrain, thereby on the cardiovascular function in mice. Adult mice received 10 mg/kg/day of MPTP (4 consecutive days) to generate PD model. Systolic blood pressure was measured by tail cuff system in conscious mice, and baroreflex sensitivity was evaluated by heart rate alteration in response to a transient increase or decrease in blood pressure induced by intravenous infusion of phenylalanine (PE) or sodium nitroprusside (SNP) in anesthetized condition, respectively. Baseline heart rate and heart rate variability were analyzed in both sham and MPTP-treated mice. Dopamine, norepinephrine, and related metabolites in the plasma and brain tissues including SNpC, locus coeruleus (LC), rostroventrolateral medulla (RVLM), and nucleus tractus solitarii (NTS) were measured by liquid chromatography-mass spectrometry (LC-MS). Tyrosine hydroxylase-positive (TH+) neurons in above nuclei were quantified by immunoreactivities. We found that in addition to the loss of TH+ neurons in SNpC, MPTP treatment induced a dramatic reduction of TH+ cell counts in the LC, RVLM, and NTS. These are associated with significant decreases of dopamine, norepinephrine, and epinephrine in above nuclei. Meanwhile, MPTP induced a lasting effect of baroreflex desensitization, tachycardia, and decreased heart rate variability compared to the sham mice. Notably, MPTP treatment elevated sympathetic outflow and suppressed parasympathetic tonicity according to the heart rate power spectrum analysis. Our results indicate that the loss of TH+ neurons in the brainstem by MPTP treatment led to impaired autonomic cardiovascular function. These results suggest that MPTP treatment can be used to study the autonomic dysfunction in murine model.
Collapse
|
26
|
Gonzalez‐Lopez E, Vrana KE. Dopamine beta‐hydroxylase and its genetic variants in human health and disease. J Neurochem 2019; 152:157-181. [DOI: 10.1111/jnc.14893] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 09/18/2019] [Accepted: 09/26/2019] [Indexed: 12/12/2022]
Affiliation(s)
| | - Kent E. Vrana
- Department of Pharmacology Penn State College of Medicine Hershey PA USA
| |
Collapse
|
27
|
Abstract
Noradrenergic system of brain supplies the neurotransmitter noradrenalin throughout the brain through widespread efferent projections and play pivotal role in cognitive activities and could be involve in motor and non-motor symptoms of Parkinson's disease (PD) pathology. Profound loss of noradrenergic pathways has been reported in both Parkinson's and Alzheimer's disease (AD) pathology however their employment in therapeutics is still scarce. Therefore the present review is providing the various aspects for involvement on noradrenergic pathways in PD and AD pathology as well as the imaging of locus coeruleus as indicative diagnostic marker for disease. The present review is describing about the role of tiny nucleus locus coeruleus located noradrenergic pathways in said pathologies and discussing the past research as well as lacunas in this regard.
Collapse
Affiliation(s)
- Sarika Singh
- Toxicology and Experimental Medicine Division, CDRI-CSIR, Lucknow, UP, India
| |
Collapse
|
28
|
Hassani OK, Rymar VV, Nguyen KQ, Huo L, Cloutier JF, Miller FD, Sadikot AF. The noradrenergic system is necessary for survival of vulnerable midbrain dopaminergic neurons: implications for development and Parkinson's disease. Neurobiol Aging 2019; 85:22-37. [PMID: 31734438 DOI: 10.1016/j.neurobiolaging.2019.09.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 09/17/2019] [Accepted: 09/19/2019] [Indexed: 12/22/2022]
Abstract
The cause of midbrain dopaminergic (mDA) neuron loss in sporadic Parkinson's disease (PD) is multifactorial, involving cell autonomous factors, cell-cell interactions, and the effects of environmental toxins. Early loss of neurons in the locus coeruleus (LC), the main source of ascending noradrenergic (NA) projections, is an important feature of PD and other neurodegenerative disorders. We hypothesized that NA afferents provide trophic support for vulnerable mDA neurons. We demonstrate that depriving mDA neurons of NA input increases postnatal apoptosis and decreases cell survival in young adult rodents, with relative sparing of calbindin-positive subpopulations known to be resistant to degeneration in PD. As a mechanism, we propose that the neurotrophin brain-derived neurotrophic factor (BDNF) modulates anterograde survival effects of LC inputs to mDA neurons. We demonstrate that the LC is rich in BDNF mRNA in postnatal and young adult brains. Early postnatal NA denervation reduces both BDNF protein and activation of TrkB receptors in the ventral midbrain. Furthermore, overexpression of BDNF in NA afferents in transgenic mice increases mDA neuronal survival. Finally, increasing NA activity in primary cultures of mDA neurons improves survival, an effect that is additive or synergistic in the presence of different concentrations of BDNF. Taken together, our results point to a novel mechanism whereby LC afferents couple BDNF effects and NA activity to provide anterograde trophic support for vulnerable mDA neurons. Early loss of NA activity and anterograde neurotrophin support may contribute to degeneration of vulnerable neurons in PD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Oum Kaltoum Hassani
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Vladimir V Rymar
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Khanh Q Nguyen
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Lia Huo
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Jean-François Cloutier
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Freda D Miller
- Departments of Medical Genetics, Microbiology & Physiology, The Hospital for Sick Children Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Abbas F Sadikot
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
29
|
Af Bjerkén S, Stenmark Persson R, Barkander A, Karalija N, Pelegrina-Hidalgo N, Gerhardt GA, Virel A, Strömberg I. Noradrenaline is crucial for the substantia nigra dopaminergic cell maintenance. Neurochem Int 2019; 131:104551. [PMID: 31542295 DOI: 10.1016/j.neuint.2019.104551] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 09/03/2019] [Accepted: 09/19/2019] [Indexed: 12/19/2022]
Abstract
In Parkinson's disease, degeneration of substantia nigra dopaminergic neurons is accompanied by damage on other neuronal systems. A severe denervation is for example seen in the locus coerulean noradrenergic system. Little is known about the relation between noradrenergic and dopaminergic degeneration, and the effects of noradrenergic denervation on the function of the dopaminergic neurons of substantia nigra are not fully understood. In this study, N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP4) was injected in rats, whereafter behavior, striatal KCl-evoked dopamine and glutamate releases, and immunohistochemistry were monitored at 3 days, 3 months, and 6 months. Quantification of dopamine-beta-hydroxylase-immunoreactive nerve fiber density in the cortex revealed a tendency towards nerve fiber regeneration at 6 months. To sustain a stable noradrenergic denervation throughout the experimental timeline, the animals in the 6-month time point received an additional DSP4 injection (2 months after the first injection). Behavioral examinations utilizing rotarod revealed that DSP4 reduced the time spent on the rotarod at 3 but not at 6 months. KCl-evoked dopamine release was significantly increased at 3 days and 3 months, while the concentrations were normalized at 6 months. DSP4 treatment prolonged both time for onset and reuptake of dopamine release over time. The dopamine degeneration was confirmed by unbiased stereology, demonstrating significant loss of tyrosine hydroxylase-immunoreactive neurons in the substantia nigra. Furthermore, striatal glutamate release was decreased after DSP4. In regards of neuroinflammation, reactive microglia were found over the substantia nigra after DSP4 treatment. In conclusion, long-term noradrenergic denervation reduces the number of dopaminergic neurons in the substantia nigra and affects the functionality of the nigrostriatal system. Thus, locus coeruleus is important for maintenance of nigral dopaminergic neurons.
Collapse
Affiliation(s)
- Sara Af Bjerkén
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden; Department of Clinical Science, Neurosciences, Umeå University, Umeå, Sweden.
| | - Rasmus Stenmark Persson
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden; Department of Clinical Science, Neurosciences, Umeå University, Umeå, Sweden
| | - Anna Barkander
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Nina Karalija
- Department of Radiation Sciences, Umeå University, Umeå, Sweden; Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
| | | | - Greg A Gerhardt
- Department of Anatomy and Neurobiology, University of Kentucky, Center for Microelectrode Technology, Lexington, KY, USA
| | - Ana Virel
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Ingrid Strömberg
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| |
Collapse
|
30
|
Zhu MY, Raza MU, Zhan Y, Fan Y. Norepinephrine upregulates the expression of tyrosine hydroxylase and protects dopaminegic neurons against 6-hydrodopamine toxicity. Neurochem Int 2019; 131:104549. [PMID: 31539561 DOI: 10.1016/j.neuint.2019.104549] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/27/2019] [Accepted: 09/17/2019] [Indexed: 10/26/2022]
Abstract
As a classic neurotransmitter in the brain, norepinephrine (NE) also is an important modulator to other neuronal systems. Using primary cultures from rat ventral mesencephalon (VM) and dopaminergic cell line MN9D, the present study examined the neuroprotective effects of NE and its effects on the expression of tyrosine hydroxylase (TH). The results showed that NE protected both VM cultures and MN9D cells against 6-hydroxydopamine-caused apoptosis, with possible involvement of adrenal receptors. In addition, treatment with NE upregulated TH protein levels in dose- and time-dependent manner. Further experiments to investigate the potential mechanisms underlying this NE-induced upregulation of TH demonstrated a marked increase in protein levels of the brain-derived neurotrophic factor (BDNF) and the phosphorylated extracellular signal-regulated protein kinase 1 and 2 (pERK1/2) in VM cultures treated with NE. In MN9D cells, a significantly increase of TH and pERK1/2 protein levels were observed after their transfection with BDNF cDNA or exposure to BDNF peptides. Treatment of VM cultures with K252a, an antagonist of the tropomyosin-related kinase B, blocked the upregulatory effects of NE on TH, BDNF and pERK1/2. Administration of MEK1 & MEK2 inhibitors also reversed NE-induced upregulation of TH and pERK1/2. Moreover, ChIP assay showed that treatment with NE or BDNF increased H4 acetylation in the TH promoter. These results suggest that the neuroprotection and modulation of NE on dopaminergic neurons are mediated via BDNF and MAPK/ERK pathways, as well as through epigenetic histone modification, which may have implications for the improvement of therapeutic strategies for Parkinson's disease.
Collapse
Affiliation(s)
- Meng-Yang Zhu
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.
| | - Muhammad U Raza
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Yanqiang Zhan
- Department of Neurology, Remin Hospital of the Wuhan University, Wuhan, China
| | - Yan Fan
- Department of Biochemistry, Nantong University College of Medicine, Nantong, China
| |
Collapse
|
31
|
Receptor Ligands as Helping Hands to L-DOPA in the Treatment of Parkinson's Disease. Biomolecules 2019; 9:biom9040142. [PMID: 30970612 PMCID: PMC6523988 DOI: 10.3390/biom9040142] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/05/2019] [Accepted: 04/06/2019] [Indexed: 12/12/2022] Open
Abstract
Levodopa (LD) is the most effective drug in the treatment of Parkinson’s disease (PD). However, although it represents the “gold standard” of PD therapy, LD can cause side effects, including gastrointestinal and cardiovascular symptoms as well as transient elevated liver enzyme levels. Moreover, LD therapy leads to LD-induced dyskinesia (LID), a disabling motor complication that represents a major challenge for the clinical neurologist. Due to the many limitations associated with LD therapeutic use, other dopaminergic and non-dopaminergic drugs are being developed to optimize the treatment response. This review focuses on recent investigations about non-dopaminergic central nervous system (CNS) receptor ligands that have been identified to have therapeutic potential for the treatment of motor and non-motor symptoms of PD. In a different way, such agents may contribute to extending LD response and/or ameliorate LD-induced side effects.
Collapse
|
32
|
Stimulation of noradrenergic transmission by reboxetine is beneficial for a mouse model of progressive parkinsonism. Sci Rep 2019; 9:5262. [PMID: 30918302 PMCID: PMC6437187 DOI: 10.1038/s41598-019-41756-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 03/14/2019] [Indexed: 01/23/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder and is characterized by motor deficits such as tremor, rigidity and bradykinesia. These symptoms are directly caused by the loss of dopaminergic neurons. However, a wealth of clinical evidence indicates that the dopaminergic system is not the only system affected in PD. Postmortem studies of brains from PD patients have revealed the degeneration of noradrenergic neurons in the locus coeruleus (LC) to the same or even greater extent than that observed in the dopaminergic neurons of substantia nigra (SN) and ventral tegmental area (VTA). Moreover, studies performed on rodent models suggest that enhancement of noradrenergic transmission may attenuate the PD-like phenotype induced by MPTP administration, a neurotoxin-based PD model. The aim of this study was to investigate whether chronic treatment with either of two compounds targeting the noradrenergic system (reboxetine or atipamezole) possess the ability to reduce the progression of a PD-like phenotype in a novel mouse model of progressive dopaminergic neurodegeneration induced by the genetic inhibition of rRNA synthesis in dopaminergic neurons, mimicking a PD-like phenotype. The results showed that reboxetine improved the parkinsonian phenotype associated with delayed progression of SN/VTA dopaminergic neurodegeneration and higher dopamine content in the striatum. Moreover, the alpha1-adrenergic agonist phenylephrine enhanced survival of TH+ neurons in primary cell cultures, supporting the putative neuroprotective effects of noradrenergic stimulation. Our results provide new insights regarding the possible influence of the noradrenergic system on dopaminergic neuron survival and strongly support the hypothesis regarding the neuroprotective role of noradrenaline.
Collapse
|
33
|
Butkovich LM, Houser MC, Tansey MG. α-Synuclein and Noradrenergic Modulation of Immune Cells in Parkinson's Disease Pathogenesis. Front Neurosci 2018; 12:626. [PMID: 30258347 PMCID: PMC6143806 DOI: 10.3389/fnins.2018.00626] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 08/21/2018] [Indexed: 12/29/2022] Open
Abstract
α-synuclein (α-syn) pathology and loss of noradrenergic neurons in the locus coeruleus (LC) are among the most ubiquitous features of Parkinson's disease (PD). While noradrenergic dysfunction is associated with non-motor symptoms of PD, preclinical research suggests that the loss of LC norepinephrine (NE), and subsequently its immune modulatory and neuroprotective actions, may exacerbate or even accelerate disease progression. In this review, we discuss the mechanisms by which α-syn pathology and loss of central NE may directly impact brain health by interrupting neurotrophic factor signaling, exacerbating neuroinflammation, and altering regulation of innate and adaptive immune cells.
Collapse
Affiliation(s)
| | | | - Malú G. Tansey
- Tansey Laboratory, Department of Physiology, School of Medicine, Emory University, Atlanta, GA, United States
| |
Collapse
|
34
|
Sommerauer M, Hansen AK, Parbo P, Fedorova TD, Knudsen K, Frederiksen Y, Nahimi A, Barbe MT, Brooks DJ, Borghammer P. Decreased noradrenaline transporter density in the motor cortex of Parkinson's disease patients. Mov Disord 2018; 33:1006-1010. [DOI: 10.1002/mds.27411] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 03/01/2018] [Accepted: 03/08/2018] [Indexed: 01/10/2023] Open
Affiliation(s)
- Michael Sommerauer
- Aarhus University Hospital, Department of Nuclear Medicine and PET Centre; Aarhus Denmark
- Department of Neurology; University Hospital Cologne; Cologne Germany
| | - Allan K Hansen
- Aarhus University Hospital, Department of Nuclear Medicine and PET Centre; Aarhus Denmark
| | - Peter Parbo
- Aarhus University Hospital, Department of Nuclear Medicine and PET Centre; Aarhus Denmark
| | - Tatyana D. Fedorova
- Aarhus University Hospital, Department of Nuclear Medicine and PET Centre; Aarhus Denmark
| | - Karoline Knudsen
- Aarhus University Hospital, Department of Nuclear Medicine and PET Centre; Aarhus Denmark
| | - Yoon Frederiksen
- Aarhus University, Department of Clinical Medicine & Department of Psychology; Aarhus Denmark
| | - Adjmal Nahimi
- Aarhus University Hospital, Department of Nuclear Medicine and PET Centre; Aarhus Denmark
| | - Michael T. Barbe
- Department of Neurology; University Hospital Cologne; Cologne Germany
| | - David J. Brooks
- Aarhus University Hospital, Department of Nuclear Medicine and PET Centre; Aarhus Denmark
- Division of Neuroscience, Department of Medicine; Imperial College London; London UK
- Division of Neuroscience; Newcastle University; Newcastle UK
| | - Per Borghammer
- Aarhus University Hospital, Department of Nuclear Medicine and PET Centre; Aarhus Denmark
| |
Collapse
|
35
|
Sescousse G, Ligneul R, van Holst RJ, Janssen LK, de Boer F, Janssen M, Berry AS, Jagust WJ, Cools R. Spontaneous eye blink rate and dopamine synthesis capacity: preliminary evidence for an absence of positive correlation. Eur J Neurosci 2018. [PMID: 29514419 PMCID: PMC5969266 DOI: 10.1111/ejn.13895] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Dopamine is central to a number of cognitive functions and brain disorders. Given the cost of neurochemical imaging in humans, behavioural proxy measures of dopamine have gained in popularity in the past decade, such as spontaneous eye blink rate (sEBR). Increased sEBR is commonly associated with increased dopamine function based on pharmacological evidence and patient studies. Yet, this hypothesis has not been validated using in vivo measures of dopamine function in humans. To fill this gap, we measured sEBR and striatal dopamine synthesis capacity using [18F]DOPA PET in 20 participants (nine healthy individuals and 11 pathological gamblers). Our results, based on frequentist and Bayesian statistics, as well as region‐of‐interest and voxel‐wise analyses, argue against a positive relationship between sEBR and striatal dopamine synthesis capacity. They show that, if anything, the evidence is in favour of a negative relationship. These results, which complement findings from a recent study that failed to observe a relationship between sEBR and dopamine D2 receptor availability, suggest that caution and nuance are warranted when interpreting sEBR in terms of a proxy measure of striatal dopamine.
Collapse
Affiliation(s)
- Guillaume Sescousse
- Donders Institute for Brain, Cognition and Behaviour, Donders Centre for Cognitive Neuroimaging, Radboud University, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Romain Ligneul
- Donders Institute for Brain, Cognition and Behaviour, Donders Centre for Cognitive Neuroimaging, Radboud University, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Ruth J van Holst
- Donders Institute for Brain, Cognition and Behaviour, Donders Centre for Cognitive Neuroimaging, Radboud University, PO Box 9101, Nijmegen, 6500 HB, The Netherlands.,Department of Psychiatry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Lieneke K Janssen
- Donders Institute for Brain, Cognition and Behaviour, Donders Centre for Cognitive Neuroimaging, Radboud University, PO Box 9101, Nijmegen, 6500 HB, The Netherlands.,Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Femke de Boer
- Donders Institute for Brain, Cognition and Behaviour, Donders Centre for Cognitive Neuroimaging, Radboud University, PO Box 9101, Nijmegen, 6500 HB, The Netherlands.,Social, Health, and Organizational Psychology, Utrecht University, Utrecht, The Netherlands
| | - Marcel Janssen
- Department of Nuclear Medicine, Radboudumc, Nijmegen, The Netherlands
| | - Anne S Berry
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| | - William J Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Roshan Cools
- Donders Institute for Brain, Cognition and Behaviour, Donders Centre for Cognitive Neuroimaging, Radboud University, PO Box 9101, Nijmegen, 6500 HB, The Netherlands.,Department of Psychiatry, Radboudumc, Nijmegen, The Netherlands
| |
Collapse
|
36
|
Zhu MY. Noradrenergic Modulation on Dopaminergic Neurons. Neurotox Res 2018; 34:848-859. [DOI: 10.1007/s12640-018-9889-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 03/05/2018] [Accepted: 03/08/2018] [Indexed: 12/24/2022]
|
37
|
Striatal norepinephrine efflux in l-DOPA-induced dyskinesia. Neurochem Int 2018; 114:85-98. [DOI: 10.1016/j.neuint.2018.01.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 01/12/2018] [Accepted: 01/17/2018] [Indexed: 11/23/2022]
|
38
|
Treatment with the noradrenaline re-uptake inhibitor atomoxetine alone and in combination with the α2-adrenoceptor antagonist idazoxan attenuates loss of dopamine and associated motor deficits in the LPS inflammatory rat model of Parkinson's disease. Brain Behav Immun 2018; 69:456-469. [PMID: 29339319 DOI: 10.1016/j.bbi.2018.01.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/04/2018] [Accepted: 01/11/2018] [Indexed: 01/14/2023] Open
Abstract
The impact of treatment with the noradrenaline (NA) re-uptake inhibitor atomoxetine and the α2-adrenoceptor (AR) antagonist idazoxan in an animal model of Parkinson's disease (PD) was assessed. Concurrent systemic treatment with atomoxetine and idazoxan, a combination which serves to enhance the extra-synaptic availability of NA, exerts anti-inflammatory and neuroprotective effects following delivery of an inflammatory stimulus, the bacterial endotoxin, lipopolysaccharide (LPS) into the substantia nigra. Lesion-induced deficits in motor function (akinesia, forelimb-use asymmetry) and striatal dopamine (DA) loss were rescued to varying degrees depending on the treatment. Treatment with atomoxetine following LPS-induced lesion to the substantia nigra, yielded a robust anti-inflammatory effect, suppressing microglial activation and expression of the pro-inflammatory cytokine TNF-α whilst increasing the expression of neurotrophic factors. Furthermore atomoxetine treatment prevented loss of tyrosine hydroxylase (TH) positive nigral dopaminergic neurons and resulted in functional improvements in motor behaviours. Atomoxetine alone was sufficient to achieve most of the observed effects. In combination with idazoxan, an additional improvement in the impairment of contralateral limb use 7 days post lesion and a reduction in amphetamine-mediated rotational asymmetry 14 days post-lesion was observed, compared to atomoxetine or idazoxan treatments alone. The results indicate that increases in central NA tone has the propensity to regulate the neuroinflammatory phenotype in vivo and may act as an endogenous neuroprotective mechanism where inflammation contributes to the progression of DA loss. In accordance with this, the clinical use of agents such as NA re-uptake inhibitors and α2-AR antagonists may prove useful in enhancing the endogenous neuroimmunomodulatory potential of NA in conditions associated with brain inflammation.
Collapse
|
39
|
Cerebrospinal fluid and serum MHPG improve Alzheimer's disease versus dementia with Lewy bodies differential diagnosis. ALZHEIMER'S & DEMENTIA: DIAGNOSIS, ASSESSMENT & DISEASE MONITORING 2018; 10:172-181. [PMID: 29552632 PMCID: PMC5852321 DOI: 10.1016/j.dadm.2018.01.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction Given the challenges concerning the differential diagnosis of dementia, we investigated the possible added value of monoaminergic compounds to the standard cerebrospinal fluid (CSF) Alzheimer's disease (AD) biomarkers. Particularly, regarding the AD versus dementia with Lewy bodies (DLB) comparison, monoamines or their metabolites might have added discriminative value as there is a more severe neuropathological burden in the locus coeruleus of DLB patients, the principal site of noradrenaline synthesis. Methods We applied enzyme-linked immunosorbent assay (ELISA) to analyze CSF amyloid β peptide of 42 amino acids, total tau, and tau phosphorylated at threonine 181, in patients with AD, frontotemporal dementia, DLB/Parkinson's disease dementia, and controls. Reversed-phase high-performance liquid chromatography with electrochemical detection was implemented to study monoamine and metabolite levels in CSF and serum. Stepwise forward conditional logistic regression and receiver operating characteristic (ROC) curve analyses were performed to assess the diagnostic accuracy of these newly fitted models containing the most discriminative indicators of disease status. Results Most significant differences in CSF and serum were confined to the noradrenergic system. More specifically, CSF 3-methoxy-4-hydroxyphenylglycol (MHPG) levels were higher, whereas serum MHPG levels were lower, in DLB patients compared with all other groups. Addition of CSF and serum MHPG levels to the CSF AD biomarker panel significantly increased diagnostic accuracy between DLB/Parkinson's disease dementia and AD. Interestingly, a model only including CSF and serum MHPG without the classic AD biomarker panel reached similar area under the curve values. Discussion We hypothesize that varying degrees of neuronal loss in the locus coeruleus of DLB/Parkinson's disease dementia versus AD patients result in differentially altered MHPG levels, making this metabolite a valuable biomarker.
Collapse
|
40
|
Huot P, Sgambato-Faure V, Fox SH, McCreary AC. Serotonergic Approaches in Parkinson's Disease: Translational Perspectives, an Update. ACS Chem Neurosci 2017; 8:973-986. [PMID: 28460160 DOI: 10.1021/acschemneuro.6b00440] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Parkinson's disease (PD) has long been seen as a disorder caused by degeneration of the dopaminergic system, leading to the classic motor manifestations of the disease. However, there is now overwhelming evidence that PD is more than a disease merely caused by dopamine depletion. It is well-known that a myriad of other neurotransmitters are affected by the disease process. One such neurotransmitter is serotonin (5-HT). 5-HT has been shown to play a role in several motor and nonmotor manifestations of PD, including tremor, cognition, depression and psychosis. 5-HT also seems to play a critical role in L-3,4-dihydroxyphenylalanine (L-DOPA)-induced dyskinesia. A breadth of preclinical studies and clinical trials have been conducted that aimed at modulating the 5-HT system in order to alleviate depression, cognitive deficits, psychosis, and dyskinesia. In this Review, we summarize recent advances in the 5-HT field in PD, but with a translational emphasis. We start by presenting a novel nonhuman primate model of PD that presents with dual dopamine and 5-HT lesions. We then present preclinical and clinical data that introduce new concepts, such as the use of biased and partial agonists, as well as molecules recently introduced to the field of PD, such as eltoprazine, pimavanserin, nelotanserin, and SYN-120, to enhance therapeutic benefit while minimizing adverse events, notably on parkinsonian disability.
Collapse
Affiliation(s)
- Philippe Huot
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montreal, QC H2X 0A9, Canada
- Department
of Pharmacology, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Unité
des Troubles du Mouvement André Barbeau, Centre Hospitalier de l’Université de Montréal, Montreal, QC H2L 4M1, Canada
- Division
of Neurology, Centre Hospitalier de l’Université de Montréal, Montreal, QC, Canada
| | - Véronique Sgambato-Faure
- Institute of Cognitive
Neuroscience Marc Jeannerod, UMR 5229 CNRS, 69 675 Cedex Bron, France
- University Lyon 1, 69100 Villeurbanne, France
| | - Susan H. Fox
- Movement
Disorder Clinic, Toronto Western Hospital, University of Toronto, Toronto, ON M5T2S8, Canada
| | - Andrew C. McCreary
- Janssen Vaccines & Prevention B.V., Archimedesweg 4, 2333 CN Leiden, The Netherlands
| |
Collapse
|
41
|
Noradrenergic Modulation of Cognition in Health and Disease. Neural Plast 2017; 2017:6031478. [PMID: 28596922 PMCID: PMC5450174 DOI: 10.1155/2017/6031478] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 04/18/2017] [Indexed: 12/15/2022] Open
Abstract
Norepinephrine released by the locus coeruleus modulates cellular processes and synaptic transmission in the central nervous system through its actions at a number of pre- and postsynaptic receptors. This transmitter system facilitates sensory signal detection and promotes waking and arousal, processes which are necessary for navigating a complex and dynamic sensory environment. In addition to its effects on sensory processing and waking behavior, norepinephrine is now recognized as a contributor to various aspects of cognition, including attention, behavioral flexibility, working memory, and long-term mnemonic processes. Two areas of dense noradrenergic innervation, the prefrontal cortex and the hippocampus, are particularly important with regard to these functions. Due to its role in mediating normal cognitive function, it is reasonable to expect that noradrenergic transmission becomes dysfunctional in a number of neuropsychiatric and neurodegenerative diseases characterized by cognitive deficits. In this review, we summarize the unique role that norepinephrine plays in prefrontal cortical and hippocampal function and how its interaction with its various receptors contribute to cognitive behaviors. We further assess the changes that occur in the noradrenergic system in Alzheimer's disease, Parkinson's disease, attention-deficit/hyperactivity disorder, and schizophrenia and how these changes contribute to cognitive decline in these pathologies.
Collapse
|
42
|
Vermeiren Y, De Deyn PP. Targeting the norepinephrinergic system in Parkinson's disease and related disorders: The locus coeruleus story. Neurochem Int 2017; 102:22-32. [DOI: 10.1016/j.neuint.2016.11.009] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 11/10/2016] [Accepted: 11/23/2016] [Indexed: 01/12/2023]
|
43
|
Masilamoni GJ, Groover O, Smith Y. Reduced noradrenergic innervation of ventral midbrain dopaminergic cell groups and the subthalamic nucleus in MPTP-treated parkinsonian monkeys. Neurobiol Dis 2016; 100:9-18. [PMID: 28042095 DOI: 10.1016/j.nbd.2016.12.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 12/22/2016] [Accepted: 12/28/2016] [Indexed: 02/07/2023] Open
Abstract
There is anatomical and functional evidence that ventral midbrain dopaminergic (DA) cell groups and the subthalamic nucleus (STN) receive noradrenergic innervation in rodents, but much less is known about these interactions in primates. Degeneration of NE neurons in the locus coeruleus (LC) and related brainstem NE cell groups is a well-established pathological feature of Parkinson's disease (PD), but the development of such pathology in animal models of PD has been inconsistent across species and laboratories. We recently demonstrated 30-40% neuronal loss in the LC, A5 and A6 NE cell groups of rhesus monkeys rendered parkinsonian by chronic administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). In this study, we used dopamine-beta-hydroxylase (DβH) immunocytochemistry to assess the impact of this neuronal loss on the number of NE terminal-like varicosities in the substantia nigra pars compacta (SNC), ventral tegmental area (VTA), retrorubral field (RRF) and STN of MPTP-treated parkinsonian monkeys. Our findings reveal that the NE innervation of the ventral midbrain and STN of normal monkeys is heterogeneously distributed being far more extensive in the VTA, RRF and dorsal tier of the SNC than in the ventral SNC and STN. In parkinsonian monkeys, all regions underwent a significant (~50-70%) decrease in NE innervation. At the electron microscopic level, some DβH-positive terminals formed asymmetric axo-dendritic synapses in VTA and STN. These findings demonstrate that the VTA, RRF and SNCd are the main ventral midbrain targets of ascending NE inputs, and that these connections undergo a major break-down in chronically MPTP-treated parkinsonian monkeys. This severe degeneration of the ascending NE system may contribute to the pathophysiology of ventral midbrain and STN neurons in PD.
Collapse
Affiliation(s)
- Gunasingh Jeyaraj Masilamoni
- Yerkes National Primate Research Center, Emory University, 954, Gatewood Rd NE, Atlanta, GA 30322, USA; Udall Center of Excellence for Parkinson's Disease, Emory University, 954, Gatewood Rd NE, Atlanta, GA 30322, USA.
| | - Olivia Groover
- Yerkes National Primate Research Center, Emory University, 954, Gatewood Rd NE, Atlanta, GA 30322, USA; Department of Neurology, Emory University, 954, Gatewood Rd NE, Atlanta, GA 30322, USA.
| | - Yoland Smith
- Yerkes National Primate Research Center, Emory University, 954, Gatewood Rd NE, Atlanta, GA 30322, USA; Department of Neurology, Emory University, 954, Gatewood Rd NE, Atlanta, GA 30322, USA; Udall Center of Excellence for Parkinson's Disease, Emory University, 954, Gatewood Rd NE, Atlanta, GA 30322, USA.
| |
Collapse
|
44
|
Hinson VK, Delambo A, Elm J, Turner T. A Randomized Clinical Trial of Atomoxetine for Mild Cognitive Impairment in Parkinson's Disease. Mov Disord Clin Pract 2016; 4:416-423. [PMID: 30363371 DOI: 10.1002/mdc3.12455] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 09/13/2016] [Accepted: 09/24/2016] [Indexed: 01/18/2023] Open
Abstract
Background Mild cognitive impairment in Parkinson's disease (PD-MCI) is associated with diminished norepinephrine from the locus coeruleus to the prefrontal cortex. Atomoxetine is a specific norepinephrine reuptake inhibitor that has been approved by the US Food and Drug Administration to treat attention deficit hyperactivity disorder in adults. The authors hypothesized that atomoxetine would improve attention and executive functioning in patients with PD-MCI. Methods Thirty participants who met Movement Disorder Society Task Force Level I criteria for PD-MCI were enrolled in a randomized controlled trial of atomoxetine. Cognitive evaluations were performed at baseline and after 10 weeks of treatment or placebo. A safety visit was performed at Week 12. A global statistical test was used to examine treatment effects on standardized tests of attention, working memory, processing speed, and set shifting (primary outcome measure). Secondary outcomes included cognitive measures hypothesized to be insensitive to atomoxetine, the Conners Adult Attention Deficit Hyperactivity Disorder Rating Scale, and safety measures. Results Fifteen participants were randomized to each arm. Groups were similar on medical and demographic variables and baseline cognition. Three serious adverse events occurred; 2 on atomoxetine (syncope, isolated episode of atrial fibrillation) and 1 on placebo (atrial fibrillation). The global statistical test of primary outcome measures did not reveal a significant difference between groups. However, significant improvements were observed for atomoxetine but not placebo on subjective measures of attention and impulsivity (Conners Adult Attention Deficit Hyperactivity Disorder Rating Scale). Conclusions Atomoxetine treatment produced subjective, but not objective, improvements in PD-MCI. Failure to detect objective differences may be due to insensitivity of cognitive tests or severity of cognitive deficits in the study participants.
Collapse
Affiliation(s)
- Vanessa K Hinson
- Department of Neurology Medical University of South Carolina Charleston South Carolina USA
| | - Amy Delambo
- Department of Neurology Medical University of South Carolina Charleston South Carolina USA
| | - Jordan Elm
- Division of Biostatistics and Epidemiology Medical University of South Carolina Charleston South Carolina USA
| | - Travis Turner
- Department of Neurology Medical University of South Carolina Charleston South Carolina USA
| |
Collapse
|
45
|
Spontaneous eye blink rate as predictor of dopamine-related cognitive function-A review. Neurosci Biobehav Rev 2016; 71:58-82. [PMID: 27555290 DOI: 10.1016/j.neubiorev.2016.08.020] [Citation(s) in RCA: 159] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/29/2016] [Accepted: 08/16/2016] [Indexed: 12/18/2022]
Abstract
An extensive body of research suggests the spontaneous eye blink rate (EBR) is a non-invasive indirect marker of central dopamine (DA) function, with higher EBR predicting higher DA function. In the present review we provide a comprehensive overview of this literature. We broadly divide the available research in studies that aim to disentangle the dopaminergic underpinnings of EBR, investigate its utility in diagnosis of DA-related disorders and responsivity to drug treatment, and, lastly, investigate EBR as predictor of individual differences in DA-related cognitive performance. We conclude (i) EBR can reflect both DA receptor subtype D1 and D2 activity, although baseline EBR might be most strongly related to the latter, (ii) EBR can predict hypo- and hyperdopaminergic activity as well as normalization of this activity following treatment, and (iii) EBR can reliably predict individual differences in performance on many cognitive tasks, in particular those related to reward-driven behavior and cognitive flexibility. In sum, this review establishes EBR as a useful predictor of DA in a wide variety of contexts.
Collapse
|
46
|
Ledreux A, Boger HA, Hinson VK, Cantwell K, Granholm AC. BDNF levels are increased by aminoindan and rasagiline in a double lesion model of Parkinson׳s disease. Brain Res 2016; 1631:34-45. [PMID: 26607251 PMCID: PMC11354023 DOI: 10.1016/j.brainres.2015.11.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 11/13/2015] [Accepted: 11/16/2015] [Indexed: 01/12/2023]
Abstract
The anti-Parkinsonian drug rasagiline is a selective, irreversible inhibitor of monoamine oxidase and is used in the treatment of Parkinson׳s disease (PD). Its postulated neuroprotective effects may be attributed to MAO inhibition, or to its propargylamine moiety. The major metabolite of rasagiline, aminoindan, has shown promising neuroprotective properties in vitro but there is a paucity of studies investigating in vivo effects of this compound. Therefore, we examined neuroprotective effects of rasagiline and its metabolite aminoindan in a double lesion model of PD. Male Fisher 344 rats received i.p. injections of the noradrenergic neurotoxin DSP-4 and intra-striatal stereotaxic microinjections of the dopamine neurotoxin 6-OHDA. Saline, rasagiline or aminoindan (3mg/kg/day s.c.) were delivered via Alzet minipumps for 4 weeks. Rats were then tested for spontaneous locomotion and a novel object recognition task. Following behavioral testing, brain tissue was processed for ELISA measurements of growth factors and immunohistochemistry. Double-lesioned rats treated with rasagiline or aminoindan had reduced behavioral deficits, both in motor and cognitive tasks compared to saline-treated double-lesioned rats. BDNF levels were significantly increased in the hippocampus and striatum of the rasagiline- and aminoindan-lesioned groups compared to the saline-treated lesioned group. Double-lesioned rats treated with rasagiline or aminoindan exhibited a sparing in the mitochondrial marker Hsp60, suggesting mitochondrial involvement in neuroprotection. Tyrosine hydroxylase (TH) immunohistochemistry revealed a sparing of TH-immunoreactive terminals in double-lesioned rats treated with rasagiline or aminoindan in the striatum, hippocampus, and substantia nigra. These data provide evidence of neuroprotection by aminoindan and rasagiline via their ability to enhance BDNF levels.
Collapse
Affiliation(s)
- Aurélie Ledreux
- Department of Neurosciences and the Center on Aging, Medical University of South Carolina, Charleston, SC, USA
| | - Heather A Boger
- Department of Neurosciences and the Center on Aging, Medical University of South Carolina, Charleston, SC, USA
| | - Vanessa K Hinson
- Department of Neurology, Medical University of South Carolina, Charleston, SC, USA; Neurology Service, Ralph H. Johnson VA Medical Center, Charleston, SC, USA
| | - Kelsey Cantwell
- Psychology and Program in Neuroscience, College of Charleston, Charleston, SC, USA
| | - Ann-Charlotte Granholm
- Department of Neurosciences and the Center on Aging, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
47
|
Loavenbruck A, Sandroni P. Neurogenic orthostatic hypotension: roles of norepinephrine deficiency in its causes, its treatment, and future research directions. Curr Med Res Opin 2015; 31:2095-104. [PMID: 26373628 DOI: 10.1185/03007995.2015.1087988] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Although a diversity of neurotransmitters and hormones participate in controlling blood pressure, norepinephrine released from postganglionic sympathetic nerve terminals is an important mediator of the rapid regulation of cardiovascular function required for homeostasis of cerebral perfusion. Hence, neurogenic orthostatic hypotension (NOH) often represents a deficiency of noradrenergic responsiveness to postural change. RESEARCH DESIGN AND METHODS PubMed searches with 'orthostatic hypotension' and 'norepinephrine' as conjoint search terms and no restriction on language or date, so as to survey the pathophysiologic and clinical relevance of norepinephrine deficiency for current NOH interventions and for future directions in treatment and research. RESULTS Norepinephrine deficiency in NOH can arise peripherally, due to cardiovascular sympathetic denervation (as in pure autonomic failure, Parkinson's disease, and a variety of neuropathies), or centrally, due to a failure of viscerosensory signals to generate adequate sympathetic traffic to intact sympathetic nerve endings (as in multiple system atrophy). Nonpharmacologic countermeasures such as pre-emptive water intake may yield blood-pressure increases exceeding those achieved pharmacologically. For patients with symptomatic NOH unresponsive to such strategies, a variety of pharmacologic interventions have been administered off-label on the basis of drug mechanisms expected to increase blood pressure via blood-volume expansion or vasoconstriction. Two pressor agents have received FDA approval: the sympathomimetic midodrine and more recently the norepinephrine prodrug droxidopa. CONCLUSIONS Pressor agents are important for treating symptomatic NOH in patients unresponsive to lifestyle changes alone. However, the dysautonomia underlying NOH often permits blood-pressure excursions toward both hypotension and hypertension. Future research should aim to shed light on the resulting management issues, and should also explore the possibility of pharmacotherapy selectively targeting orthostatic blood-pressure decreases.
Collapse
Affiliation(s)
- Adam Loavenbruck
- a a Department of Neurology , University of Minnesota , Minneapolis , MN , USA
| | - Paola Sandroni
- b b Department of Neurology , Mayo Clinic , Rochester , MN , USA
| |
Collapse
|
48
|
Niccolini F, Rocchi L, Politis M. Molecular imaging of levodopa-induced dyskinesias. Cell Mol Life Sci 2015; 72:2107-17. [PMID: 25681866 PMCID: PMC11113208 DOI: 10.1007/s00018-015-1854-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 02/06/2015] [Accepted: 02/09/2015] [Indexed: 12/15/2022]
Abstract
Levodopa-induced dyskinesias (LIDs) occur in the majority of patients with Parkinson's disease (PD) following years of levodopa treatment. The pathophysiology underlying LIDs in PD is poorly understood, and current treatments generate only minor benefits for the patients. Studies with positron emission tomography (PET) molecular imaging have demonstrated that in advanced PD patients, levodopa administration induces sharp increases in striatal dopamine levels, which correlate with LIDs severity. Fluctuations in striatal dopamine levels could be the result of the attenuated buffering ability in the dopaminergically denervated striatum. Lines of evidence from PET studies indicate that serotonergic terminals could also be responsible for the development of LIDs in PD by aberrantly processing exogenous levodopa and by releasing dopamine in a dysregulated manner from the serotonergic terminals. Additionally, other downstream mechanisms involving glutamatergic, cannabinoid, opioid, cholinergic, adenosinergic, and noradrenergic systems may contribute in the development of LIDs. In this article, we review the findings from preclinical, clinical, and molecular imaging studies, which have contributed to our understanding the pathophysiology of LIDs in PD.
Collapse
Affiliation(s)
- Flavia Niccolini
- Neurodegeneration Imaging Group, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, SE5 8AF UK
| | - Lorenzo Rocchi
- Neurodegeneration Imaging Group, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, SE5 8AF UK
| | - Marios Politis
- Neurodegeneration Imaging Group, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, SE5 8AF UK
| |
Collapse
|
49
|
Wang Y, Chen X, Wang T, Sun YN, Han LN, Li LB, Zhang L, Wu ZH, Huang C, Liu J. Additional noradrenergic depletion aggravates forelimb akinesia and abnormal subthalamic nucleus activity in a rat model of Parkinson's disease. Life Sci 2014; 119:18-27. [PMID: 25445222 DOI: 10.1016/j.lfs.2014.10.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 09/18/2014] [Accepted: 10/09/2014] [Indexed: 11/25/2022]
Abstract
AIMS This study aims to identify the contribution of additional noradrenergic depletion to forelimb akinesia and abnormal subthalamic nucleus (STN) firing activity in Parkinson's disease (PD). MAIN METHODS Forelimb akinesia behaviors were tested in awake rats with noradrenergic N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP-4) lesions, unilateral 6-hydroxydopamine (6-OHDA) lesions in the substantia nigra pars compacta (SNc) and with combined 6-OHDA and DSP-4 lesions. STN extracellular single-unit and local field potential (LFP) activities were examined in the animals that were anesthetized with urethane. KEY FINDINGS The adjusting steps and the contralateral touches of rats in the forelimb akinesia behavior tests were markedly inhibited by a further noradrenergic lesion with DSP-4 in 6-OHDA+DSP-4-lesioned group when compared with those of 6-OHDA-lesioned animals (P<0.05 for all comparisons). Meanwhile, the neuronal firing pattern of STN also changed significantly towards more bursty in 6-OHDA + DSP-4-lesioned group (P <0 .05). Compared with 6-OHDA-lesioned animals, an additional noradrenergic lesion increased the 0.3-2.5 Hz oscillatory activity and the spike power of STN neurons (P < 0.01 for both comparisons), and strengthened the synchronized oscillation between subthalamic neuronal firing and LFP activity in 6-OHDA + DSP-4-lesioned group (P < 0.01). SIGNIFICANCE The results provide evidence to support the correlation between noradrenergic depletion and the further exaggerated dysfunction of STN electrical activity in PD and suggest that an aberrant noradrenergic system might play a specific role in the motor deficits of PD.
Collapse
Affiliation(s)
- Yong Wang
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiang Chen
- The Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Tao Wang
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, Xi'an 710061, China
| | - Yi Na Sun
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, Xi'an 710061, China
| | - Ling Na Han
- The Key Laboratory of Environment and Disease-Related Genes, Ministry of Education, Xi'an 710061, China
| | - Li Bo Li
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, Xi'an 710061, China
| | - Li Zhang
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, Xi'an 710061, China
| | - Zhong Heng Wu
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, Xi'an 710061, China
| | - Chen Huang
- The Key Laboratory of Environment and Disease-Related Genes, Ministry of Education, Xi'an 710061, China
| | - Jian Liu
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
50
|
Espay AJ, LeWitt PA, Kaufmann H. Norepinephrine deficiency in Parkinson's disease: The case for noradrenergic enhancement. Mov Disord 2014; 29:1710-9. [PMID: 25297066 DOI: 10.1002/mds.26048] [Citation(s) in RCA: 179] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 09/02/2014] [Accepted: 09/11/2014] [Indexed: 12/28/2022] Open
Affiliation(s)
- Alberto J. Espay
- Gardner Family Center for Parkinson's Disease and Movement Disorders; Department of Neurology; University of Cincinnati; Cincinnati Ohio USA
| | - Peter A. LeWitt
- Departments of Neurology; Henry Ford Hospital and Wayne State University School of Medicine; West Bloomfield Michigan USA
| | - Horacio Kaufmann
- Dysautonomia Center; Department of Neurology; NYU School of Medicine; New York New York USA
| |
Collapse
|