1
|
Nagaeva E, Turconi G, Mätlik K, Segerstråle M, Olfat S, Iivanainen V, Taira T, Andressoo JO. Motor learning is regulated by postnatal GDNF levels in Purkinje cells. Neuroscience 2025; 576:27-41. [PMID: 40254124 DOI: 10.1016/j.neuroscience.2025.04.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 04/08/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
Purkinje cells (PCs), the sole output neurons of the cerebellar cortex, are crucial for cerebellum-dependent motor learning. In cerebellar ataxia, reduction in motor function and learning associates with decreased spontaneous activity of PCs. Thus, understanding what molecules regulate PCs activity is important. Previously, we demonstrated that a ubiquitous 2-fold increase of endogenous glial cell line-derived neurotrophic factor (GDNF) improves motor function in adult mice and motor learning and coordination in aged mice. However, since GDNF impacts many organ systems the underlying mechanism remained elusive. Here we utilize GDNF Hypermorphic, conditional GDNF Hypermorphic and conditional knock-out mouse models to reveal that up to a 2-fold increase in endogenous GDNF, specifically in PCs postnatally, is sufficient to enhance motor learning. We find that improved motor learning associates with increased glutamatergic input to PCs and with elevated spontaneous firing rate of PCs, opposite to cerebellar ataxia where reduction in motor function and learning associates with decreased spontaneous activity of PCs. Analysis of the human cerebellum revealed that normal interindividual variation in GDNF expression levels falls in the same variation range as studied in the mouse models, suggesting that interindividual variation in PC GDNF levels may contribute to interindividual variation in PC function. Collectively, our findings reveal how a relatively small change in postnatal GDNF expression level within the physiological range in one cell type, the PCs, affects motor learning. Thus, drugs enhancing postnatal GDNF expression in PCs or cerebellar GDNF signaling may have potential in treating cerebellar ataxias, making an interesting topic for future studies.
Collapse
Affiliation(s)
- Elina Nagaeva
- Department of Pharmacology, Faculty of Medicine & Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Giorgio Turconi
- Department of Pharmacology, Faculty of Medicine & Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Kärt Mätlik
- Department of Pharmacology, Faculty of Medicine & Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Mikael Segerstråle
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Soophie Olfat
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden
| | - Vilma Iivanainen
- Department of Pharmacology, Faculty of Medicine & Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Tomi Taira
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Finland
| | - Jaan-Olle Andressoo
- Department of Pharmacology, Faculty of Medicine & Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland; Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
2
|
Antonijevic M, Dallemagne P, Rochais C. Indirect influence on the BDNF/TrkB receptor signaling pathway via GPCRs, an emerging strategy in the treatment of neurodegenerative disorders. Med Res Rev 2025; 45:274-310. [PMID: 39180386 DOI: 10.1002/med.22075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/06/2022] [Accepted: 08/04/2024] [Indexed: 08/26/2024]
Abstract
Neuronal survival depends on neurotrophins and their receptors. There are two types of neurotrophin receptors: a nonenzymatic, trans-membrane protein of the tumor necrosis factor receptor (TNFR) family-p75 receptor and the tyrosine kinase receptors (TrkR) A, B, and C. Activation of the TrkBR by brain-derived neurotrophic factor (BDNF) or neurotrophin 4/5 (NT-4/5) promotes neuronal survival, differentiation, and synaptic function. It is shown that in the pathogenesis of several neurodegenerative conditions (Alzheimer's disease, Parkinson's disease, Huntington's disease) the BDNF/TrkBR signaling pathway is impaired. Since it is known that GPCRs and TrkR are regulating several cell functions by interacting with each other and generating a cross-communication in this review we have focused on the interaction between different GPCRs and their ligands on BDNF/TrkBR signaling pathway.
Collapse
|
3
|
Fursa GA, Andretsova SS, Shishkina VS, Voronova AD, Karsuntseva EK, Chadin AV, Reshetov IV, Stepanova OV, Chekhonin VP. The Use of Neurotrophic Factors as a Promising Strategy for the Treatment of Neurodegenerative Diseases (Review). Bull Exp Biol Med 2024:10.1007/s10517-024-06218-5. [PMID: 39266924 DOI: 10.1007/s10517-024-06218-5] [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: 08/07/2023] [Indexed: 09/14/2024]
Abstract
The review considers the use of exogenous neurotrophic factors in the treatment of neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, multiple sclerosis, and others. This group of diseases is associated with the death of neurons and dysfunction of the nervous tissue. Currently, there is no effective therapy for neurodegenerative diseases, and their treatment remains a serious problem of modern medicine. A promising strategy is the use of exogenous neurotrophic factors. Targeted delivery of these factors to the nervous tissue can improve survival of neurons during the development of neurodegenerative processes and ensure neuroplasticity. There are methods of direct injection of neurotrophic factors into the nervous tissue, delivery using viral vectors, as well as the use of gene cell products. The effectiveness of these approaches has been studied in numerous experimental works and in a number of clinical trials. Further research in this area could provide the basis for the creation of an alternative treatment for neurodegenerative diseases.
Collapse
Affiliation(s)
- G A Fursa
- V. Serbsky National Medical Research Centre for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia.
- Pirogov Russian National Research Medical University, Moscow, Russia.
- National Medical Research Centre of Cardiology named after academician E. I. Chazov, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - S S Andretsova
- V. Serbsky National Medical Research Centre for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - V S Shishkina
- V. Serbsky National Medical Research Centre for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - A D Voronova
- V. Serbsky National Medical Research Centre for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia
- National Medical Research Centre of Cardiology named after academician E. I. Chazov, Ministry of Health of the Russian Federation, Moscow, Russia
| | - E K Karsuntseva
- V. Serbsky National Medical Research Centre for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - A V Chadin
- V. Serbsky National Medical Research Centre for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - I V Reshetov
- University Clinical Hospital No. 1, I. M. Sechenov First Moscow State Medical University, Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
- Academy of Postgraduate Education, Federal Research and Clinical Center of Specialized Types of Health Care and Medical Technology of the Federal Medical and Biological Agency, Moscow, Russia
| | - O V Stepanova
- V. Serbsky National Medical Research Centre for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia
- National Medical Research Centre of Cardiology named after academician E. I. Chazov, Ministry of Health of the Russian Federation, Moscow, Russia
| | - V P Chekhonin
- V. Serbsky National Medical Research Centre for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
4
|
Kasanga EA, Soto I, Centner A, McManus R, Shifflet MK, Navarrete W, Han Y, Lisk J, Ehrhardt T, Wheeler K, Mhatre-Winters I, Richardson JR, Bishop C, Nejtek VA, Salvatore MF. Moderate intensity aerobic exercise alleviates motor deficits in 6-OHDA lesioned rats and reduces serum levels of biomarkers of Parkinson's disease severity without recovery of striatal dopamine or tyrosine hydroxylase. Exp Neurol 2024; 379:114875. [PMID: 38944332 DOI: 10.1016/j.expneurol.2024.114875] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/11/2024] [Accepted: 06/20/2024] [Indexed: 07/01/2024]
Abstract
Alleviation of motor impairment by aerobic exercise (AE) in Parkinson's disease (PD) patients points to activation of neurobiological mechanisms that may be targetable by therapeutic approaches. However, evidence for AE-related recovery of striatal dopamine (DA) signaling or tyrosine hydroxylase (TH) loss has been inconsistent in rodent studies. This ambiguity may be related to the timing of AE intervention in relation to the status of nigrostriatal neuron loss. Here, we replicated human PD at diagnosis by establishing motor impairment with >80% striatal DA and TH loss prior to initiating AE, and assessed its potential to alleviate motor decline and restore DA and TH loss. We also evaluated if serum levels of neurofilament light (NfL) and glial fibrillary acidic protein (GFAP), biomarkers of human PD severity, changed in response to AE. 6-hydroxydopamine (6-OHDA) was infused unilaterally into rat medial forebrain bundle to induce progressive nigrostriatal neuron loss over 28 days. Moderate intensity AE (3× per week, 40 min/session), began 8-10 days post-lesion following establishment of impaired forelimb use. Striatal tissue DA, TH protein and mRNA, and serum levels of NfL/GFAP were determined 3-wks after AE began. Despite severe striatal DA depletion at AE initiation, forelimb use deficits and hypokinesia onset were alleviated by AE, without recovery of striatal DA or TH protein loss, but reduced NfL and GFAP serum levels. This proof-of-concept study shows AE alleviates motor impairment when initiated with >80% striatal DA loss without obligate recovery of striatal DA or TH protein. Moreover, the AE-related reduction of NfL and GFAP serum levels may serve as objective blood-based biomarkers of AE efficacy.
Collapse
Affiliation(s)
- Ella A Kasanga
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States of America
| | - Isabel Soto
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States of America
| | - Ashley Centner
- Department of Psychology, Binghamton University, Binghamton, NY, United States of America
| | - Robert McManus
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States of America
| | - Marla K Shifflet
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States of America
| | - Walter Navarrete
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States of America
| | - Yoonhee Han
- Department of Environmental Health Sciences, Robert Stempel School of Public Health & Social Work, Florida International University, Miami, FL, United States of America; Isakson Center for Neurological Disease Research, College of Veterinary Medicine, University of Georgia, Athens, GA, United States of America
| | - Jerome Lisk
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States of America
| | - Travis Ehrhardt
- Clearcut Ortho Rehab & Diagnostics, Fort Worth, TX, United States of America
| | - Ken Wheeler
- Clearcut Ortho Rehab & Diagnostics, Fort Worth, TX, United States of America
| | - Isha Mhatre-Winters
- Department of Environmental Health Sciences, Robert Stempel School of Public Health & Social Work, Florida International University, Miami, FL, United States of America; Isakson Center for Neurological Disease Research, College of Veterinary Medicine, University of Georgia, Athens, GA, United States of America
| | - Jason R Richardson
- Department of Environmental Health Sciences, Robert Stempel School of Public Health & Social Work, Florida International University, Miami, FL, United States of America; Isakson Center for Neurological Disease Research, College of Veterinary Medicine, University of Georgia, Athens, GA, United States of America
| | - Christopher Bishop
- Department of Psychology, Binghamton University, Binghamton, NY, United States of America
| | - Vicki A Nejtek
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States of America
| | - Michael F Salvatore
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States of America.
| |
Collapse
|
5
|
Wegman E, Wosiski-Kuhn M, Luo Y. The dual role of striatal interneurons: circuit modulation and trophic support for the basal ganglia. Neural Regen Res 2024; 19:1277-1283. [PMID: 37905876 PMCID: PMC11467944 DOI: 10.4103/1673-5374.382987] [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: 02/28/2023] [Revised: 06/26/2023] [Accepted: 07/30/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT Striatal interneurons play a key role in modulating striatal-dependent behaviors, including motor activity and reward and emotional processing. Interneurons not only provide modulation to the basal ganglia circuitry under homeostasis but are also involved in changes to plasticity and adaptation during disease conditions such as Parkinson's or Huntington's disease. This review aims to summarize recent findings regarding the role of striatal cholinergic and GABAergic interneurons in providing circuit modulation to the basal ganglia in both homeostatic and disease conditions. In addition to direct circuit modulation, striatal interneurons have also been shown to provide trophic support to maintain neuron populations in adulthood. We discuss this interesting and novel role of striatal interneurons, with a focus on the maintenance of adult dopaminergic neurons from interneuron-derived sonic-hedgehog.
Collapse
Affiliation(s)
- Elliot Wegman
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
| | - Marlena Wosiski-Kuhn
- Department of Emergency Medicine at the School of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Yu Luo
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
6
|
Salvatore MF. Dopamine Signaling in Substantia Nigra and Its Impact on Locomotor Function-Not a New Concept, but Neglected Reality. Int J Mol Sci 2024; 25:1131. [PMID: 38256204 PMCID: PMC10815979 DOI: 10.3390/ijms25021131] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/11/2024] [Accepted: 01/13/2024] [Indexed: 01/24/2024] Open
Abstract
The mechanistic influences of dopamine (DA) signaling and impact on motor function are nearly always interpreted from changes in nigrostriatal neuron terminals in striatum. This is a standard practice in studies of human Parkinson's disease (PD) and aging and related animal models of PD and aging-related parkinsonism. However, despite dozens of studies indicating an ambiguous relationship between changes in striatal DA signaling and motor phenotype, this perseverating focus on striatum continues. Although DA release in substantia nigra (SN) was first reported almost 50 years ago, assessment of nigral DA signaling changes in relation to motor function is rarely considered. Whereas DA signaling has been well-characterized in striatum at all five steps of neurotransmission (biosynthesis and turnover, storage, release, reuptake, and post-synaptic binding) in the nigrostriatal pathway, the depth of such interrogations in the SN, outside of cell counts, is sparse. However, there is sufficient evidence that these steps in DA neurotransmission in the SN are operational and regulated autonomously from striatum and are present in human PD and aging and related animal models. To complete our understanding of how nigrostriatal DA signaling affects motor function, it is past time to include interrogation of nigral DA signaling. This brief review highlights evidence that changes in nigral DA signaling at each step in DA neurotransmission are autonomous from those in striatum and changes in the SN alone can influence locomotor function. Accordingly, for full characterization of how nigrostriatal DA signaling affects locomotor activity, interrogation of DA signaling in SN is essential.
Collapse
Affiliation(s)
- Michael F Salvatore
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
7
|
Barker RA, Buttery PC. Disease-specific interventions: The use of cell and gene therapies for Parkinson disease. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:171-191. [PMID: 39341654 DOI: 10.1016/b978-0-323-90120-8.00003-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Approaches to repair the brain around the loss of the nigrostriatal dopaminergic pathways in Parkinson disease (PD) are not new and have been attempted over many years. However, of late, the situation has moved forward in two main ways. In the case of cell therapies, the ability to make large numbers of authentic midbrain dopaminergic neuroblasts from human pluripotent stem cell sources has turned what was an interesting avenue of research into a major area of investment and trialing, by academics in conjunction with Pharma. In the case of gene therapies, their use around dopamine replacement has waned, as the interest in using them for disease modification targeting PD-specific pathways has grown. In this chapter, we discuss all these developments and the current status of cell and gene therapies for PD.
Collapse
Affiliation(s)
- Roger A Barker
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom.
| | - Philip C Buttery
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
8
|
Jazaeri SZ, Taghizadeh G, Babaei JF, Goudarzi S, Saadatmand P, Joghataei MT, Khanahmadi Z. Aquaporin 4 beyond a water channel; participation in motor, sensory, cognitive and psychological performances, a comprehensive review. Physiol Behav 2023; 271:114353. [PMID: 37714320 DOI: 10.1016/j.physbeh.2023.114353] [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: 06/05/2023] [Revised: 08/15/2023] [Accepted: 09/13/2023] [Indexed: 09/17/2023]
Abstract
Aquaporin 4 (AQP4) is a protein highly expressed in the central nervous system (CNS) and peripheral nervous system (PNS) as well as various other organs, whose different sites of action indicate its importance in various functions. AQP4 has a variety of essential roles beyond water homeostasis. In this article, we have for the first time summarized different roles of AQP4 in motor and sensory functions, besides cognitive and psychological performances, and most importantly, possible physiological mechanisms by which AQP4 can exert its effects. Furthermore, we demonstrated that AQP4 participates in pathology of different neurological disorders, various effects depending on the disease type. Since neurological diseases involve a spectrum of dysfunctions and due to the difficulty of obtaining a treatment that can simultaneously affect these deficits, it is therefore suggested that future studies consider the role of this protein in different functional impairments related to neurological disorders simultaneously or separately by targeting AQP4 expression and/or polarity modulation.
Collapse
Affiliation(s)
- Seyede Zohreh Jazaeri
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ghorban Taghizadeh
- Department of Occupational Therapy, School of Rehabilitation Sciences, Iran University of Medical Sciences, Tehran, Iran.
| | - Javad Fahanik Babaei
- Electrophysiology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Goudarzi
- Experimental Medicine Research Center, Tehran University of medical Sciences, Tehran, Iran
| | - Pegah Saadatmand
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Taghi Joghataei
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Department of Innovation in Medical Education, Faculty of Medicine, Ottawa University, Ottawa, Canada.
| | - Zohreh Khanahmadi
- Department of Occupational Therapy, School of Rehabilitation Services, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
9
|
Kasanga EA, Han Y, Shifflet MK, Navarrete W, McManus R, Parry C, Barahona A, Nejtek VA, Manfredsson FP, Kordower JH, Richardson JR, Salvatore MF. Nigral-specific increase in ser31 phosphorylation compensates for tyrosine hydroxylase protein and nigrostriatal neuron loss: Implications for delaying parkinsonian signs. Exp Neurol 2023; 368:114509. [PMID: 37634696 DOI: 10.1016/j.expneurol.2023.114509] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/08/2023] [Accepted: 08/23/2023] [Indexed: 08/29/2023]
Abstract
Compensatory mechanisms that augment dopamine (DA) signaling are thought to mitigate onset of hypokinesia prior to major loss of tyrosine hydroxylase (TH) in striatum that occurs in Parkinson's disease. However, the identity of such mechanisms remains elusive. In the present study, the rat nigrostriatal pathway was unilaterally-lesioned with 6-hydroxydopamine (6-OHDA) to determine whether differences in DA content, TH protein, TH phosphorylation, or D1 receptor expression in striatum or substantia nigra (SN) aligned with hypokinesia onset and severity at two time points. In striatum, DA and TH loss reached its maximum (>90%) 7 days after lesion induction. However, in SN, no DA loss occurred, despite ∼60% TH loss. Hypokinesia was established at 21 days post-lesion and maintained at 28 days. At this time, DA loss was ∼60% in the SN, but still of lesser magnitude than TH loss. At day 7 and 28, ser31 TH phosphorylation increased only in SN, corresponding to less DA versus TH protein loss. In contrast, ser40 TH phosphorylation was unaffected in either region. Despite DA loss in both regions at day 28, D1 receptor expression increased only in lesioned SN. These results support the concept that augmented components of DA signaling in the SN, through increased ser31 TH phosphorylation and D1 receptor expression, contribute as compensatory mechanisms against progressive nigrostriatal neuron and TH protein loss, and may mitigate hypokinesia severity.
Collapse
Affiliation(s)
- Ella A Kasanga
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76117, USA
| | - Yoonhee Han
- Robert Stempel School of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| | - Marla K Shifflet
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76117, USA
| | - Walter Navarrete
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76117, USA
| | - Robert McManus
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76117, USA
| | - Caleb Parry
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76117, USA
| | - Arturo Barahona
- Robert Stempel School of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| | - Vicki A Nejtek
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76117, USA
| | - Fredric P Manfredsson
- Parkinson's Disease Research Unit, Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Jeffrey H Kordower
- ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ 85287, USA
| | - Jason R Richardson
- Robert Stempel School of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| | - Michael F Salvatore
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76117, USA.
| |
Collapse
|
10
|
Casserly L, Garton DR, Montaño-Rodriguez A, Andressoo JO. Analysis of Acute and Chronic Methamphetamine Treatment in Mice on Gdnf System Expression Reveals a Potential Mechanism of Schizophrenia Susceptibility. Biomolecules 2023; 13:1428. [PMID: 37759827 PMCID: PMC10526418 DOI: 10.3390/biom13091428] [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: 08/11/2023] [Revised: 09/11/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
The increase in presynaptic striatal dopamine is the main dopaminergic abnormality in schizophrenia (SCZ). SCZ is primarily treated by modulating the activity of monoamine systems, with a focus on dopamine and serotonin receptors. Glial cell line-derived neurotrophic factor (GDNF) is a strong dopaminergic factor, that recently was shown to correlate with SCZ in human CSF and in striatal tissue. A 2-3-fold increase in GDNF in the brain was sufficient to induce SCZ-like dopaminergic and behavioural changes in mice. Here, we analysed the effect of acute, chronic, and embryonic methamphetamine, a drug known to enhance the risk of psychosis, on Gdnf and its receptors, Gfra1 and Ret, as well as on monoamine metabolism-related gene expression in the mouse brain. We found that acute methamphetamine application increases Gdnf expression in the striatum and chronic methamphetamine decreases the striatal expression of GDNF receptors Gfra1 and Ret. Both chronic and acute methamphetamine treatment upregulated the expression of genes related to dopamine and serotonin metabolism in the striatum, prefrontal cortex, and substantia nigra. Our results suggest a potential mechanism as to how methamphetamine elicits individual psychosis risk in young adults-variation in initial striatal GDNF induction and subsequent GFRα1 and RET downregulation may determine individual susceptibility to psychosis. Our results may guide future experiments and precision medicine development for methamphetamine-induced psychosis using GDNF/GFRa1/RET antagonists.
Collapse
Affiliation(s)
- Laoise Casserly
- Department of Pharmacology, Faculty of Medicine, Helsinki Institute of Life Science, University of Helsinki, 00290 Helsinki, Finland
| | - Daniel R. Garton
- Department of Pharmacology, Faculty of Medicine, Helsinki Institute of Life Science, University of Helsinki, 00290 Helsinki, Finland
| | - Ana Montaño-Rodriguez
- Department of Pharmacology, Faculty of Medicine, Helsinki Institute of Life Science, University of Helsinki, 00290 Helsinki, Finland
| | - Jaan-Olle Andressoo
- Department of Pharmacology, Faculty of Medicine, Helsinki Institute of Life Science, University of Helsinki, 00290 Helsinki, Finland
- Division of Neurogeriatrics, Department of Neurobiology, Care Science and Society (NVS), Karolinska Institutet, 17177 Stockholm, Sweden
| |
Collapse
|
11
|
Du W, Wang T, Hu S, Luan J, Tian F, Ma G, Xue J. Engineering of electrospun nanofiber scaffolds for repairing brain injury. ENGINEERED REGENERATION 2023; 4:289-303. [DOI: 10.1016/j.engreg.2023.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/14/2023] Open
|
12
|
Kasanga EA, Soto I, Centner A, McManus R, Shifflet MK, Navarrete W, Han Y, Lisk J, Wheeler K, Mhatre-Winters I, Richardson JR, Bishop C, Nejtek VA, Salvatore MF. Moderate intensity aerobic exercise in 6-OHDA-lesioned rats alleviates established motor deficits and reduces neurofilament light and glial fibrillary acidic protein serum levels without increased striatal dopamine or tyrosine hydroxylase protein. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.11.548638. [PMID: 37502851 PMCID: PMC10369940 DOI: 10.1101/2023.07.11.548638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Background Alleviation of motor impairment by aerobic exercise (AE) in Parkinson's disease (PD) points to a CNS response that could be targeted by therapeutic approaches, but recovery of striatal dopamine (DA) or tyrosine hydroxylase (TH) has been inconsistent in rodent studies. Objective To increase translation of AE, 3 components were implemented into AE design to determine if recovery of established motor impairment, concomitant with >80% striatal DA and TH loss, was possible. We also evaluated if serum levels of neurofilament light (NfL) and glial fibrillary acidic protein (GFAP), blood-based biomarkers of disease severity in human PD, were affected. Methods We used a 6-OHDA hemiparkinson rat model featuring progressive nigrostriatal neuron loss over 28 days, with impaired forelimb use 7 days post-lesion, and hypokinesia onset 21 days post-lesion. After establishing forelimb use deficits, moderate intensity AE began 1-3 days later, 3x per week, for 40 min/session. Motor assessments were conducted weekly for 3 wks, followed by determination of striatal DA, TH protein and mRNA, and NfL and GFAP serum levels. Results Seven days after 6-OHDA lesion, recovery of depolarization-stimulated extracellular DA and DA tissue content was <10%, representing severity of DA loss in human PD, concomitant with 50% reduction in forelimb use. Despite severe DA loss, recovery of forelimb use deficits and alleviation of hypokinesia progression began after 2 weeks of AE and was maintained. Increased NfLand GFAP levels from lesion were reduced by AE. Despite these AE-driven changes, striatal DA tissue and TH protein levels were unaffected. Conclusions This proof-of-concept study shows AE, using exercise parameters within the capabilities most PD patients, promotes recovery of established motor deficits in a rodent PD model, concomitant with reduced levels of blood-based biomarkers associated with PD severity, without commensurate increase in striatal DA or TH protein.
Collapse
|
13
|
Alarcón TA, Presti-Silva SM, Simões APT, Ribeiro FM, Pires RGW. Molecular mechanisms underlying the neuroprotection of environmental enrichment in Parkinson's disease. Neural Regen Res 2023; 18:1450-1456. [PMID: 36571341 PMCID: PMC10075132 DOI: 10.4103/1673-5374.360264] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Parkinson's disease is the most common movement disorder, affecting about 1% of the population over the age of 60 years. Parkinson's disease is characterized clinically by resting tremor, bradykinesia, rigidity and postural instability, as a result of the progressive loss of nigrostriatal dopaminergic neurons. In addition to this neuronal cell loss, Parkinson's disease is characterized by the accumulation of intracellular protein aggregates, Lewy bodies and Lewy neurites, composed primarily of the protein α-synuclein. Although it was first described almost 200 years ago, there are no disease-modifying drugs to treat patients with Parkinson's disease. In addition to conventional therapies, non-pharmacological treatment strategies are under investigation in patients and animal models of neurodegenerative disorders. Among such strategies, environmental enrichment, comprising physical exercise, cognitive stimulus, and social interactions, has been assessed in preclinical models of Parkinson's disease. Environmental enrichment can cause structural and functional changes in the brain and promote neurogenesis and dendritic growth by modifying gene expression, enhancing the expression of neurotrophic factors and modulating neurotransmission. In this review article, we focus on the current knowledge about the molecular mechanisms underlying environmental enrichment neuroprotection in Parkinson's disease, highlighting its influence on the dopaminergic, cholinergic, glutamatergic and GABAergic systems, as well as the involvement of neurotrophic factors. We describe experimental pre-clinical data showing how environmental enrichment can act as a modulator in a neurochemical and behavioral context in different animal models of Parkinson's disease, highlighting the potential of environmental enrichment as an additional strategy in the management and prevention of this complex disease.
Collapse
Affiliation(s)
- Tamara Andrea Alarcón
- Department of Physiological Sciences; Laboratory of Molecular and Behavioral Neurobiology, Health Science Center, Universidade Federal do Espirito Santo, Vitoria, Brazil
| | - Sarah Martins Presti-Silva
- Laboratory of Molecular and Behavioral Neurobiology, Health Science Center, Universidade Federal do Espirito Santo, Vitoria; Department of Biochemistry and Immunology, Institute o Biological Sciences, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, Belo Horizonte, Brazil
| | - Ana Paula Toniato Simões
- Department of Physiological Sciences; Laboratory of Molecular and Behavioral Neurobiology, Health Science Center, Universidade Federal do Espirito Santo, Vitoria, Brazil
| | - Fabiola Mara Ribeiro
- Department of Biochemistry and Immunology, Institute o Biological Sciences, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, Belo Horizonte, Brazil
| | - Rita Gomes Wanderley Pires
- Department of Physiological Sciences; Laboratory of Molecular and Behavioral Neurobiology, Health Science Center, Universidade Federal do Espirito Santo, Vitoria, Brazil
| |
Collapse
|
14
|
Kasanga EA, Han Y, Navarrete W, McManus R, Shifflet MK, Parry C, Barahona A, Manfredsson FP, Nejtek VA, Richardson JR, Salvatore MF. Differential expression of RET and GDNF family receptor, GFR-α1, between striatum and substantia nigra following nigrostriatal lesion: a case for diminished GDNF-signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.01.530671. [PMID: 36909534 PMCID: PMC10002742 DOI: 10.1101/2023.03.01.530671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Although glial cell line-derived neurotrophic factor (GDNF) showed efficacy in preclinical and early clinical studies to alleviate parkinsonian signs in Parkinson's disease (PD), later trials did not meet primary endpoints, giving pause to consider further investigation. While GDNF dose and delivery methods may have contributed to diminished efficacy, one crucial aspect of these clinical studies is that GDNF treatment across all studies began ∼8 years after PD diagnosis; a time point representing several years after near 100% depletion of nigrostriatal dopamine markers in striatum and at least 50% in substantia nigra (SN), and is later than the timing of GDNF treatment in preclinical studies. With nigrostriatal terminal loss exceeding 70% at PD diagnosis, we utilized hemi-parkinsonian rats to determine if expression of GDNF family receptor, GFR-α1, and receptor tyrosine kinase, RET, differed between striatum and SN at 1 and 4 weeks following a 6-hydroxydopamine (6-OHDA) lesion. Whereas GDNF expression changed minimally, GFR-α1 expression decreased progressively in striatum and in tyrosine hydroxylase positive (TH+) cells in SN, correlating with reduced TH cell number. However, in nigral astrocytes, GFR-α1 expression increased. RET expression decreased maximally in striatum by 1 week, whereas in the SN, a transient bilateral increase occurred that returned to control levels by 4 weeks. Expression of brain-derived neurotrophic factor (BDNF) or its receptor, TrkB, were unchanged throughout lesion progression. Together, these results reveal that differential GFR-α1 and RET expression between the striatum and SN, and cell-specific differences in GFR-α1 expression in SN, occur during nigrostriatal neuron loss. Targeting loss of GDNF receptors appears critical to enhance GDNF therapeutic efficacy against nigrostriatal neuron loss. Significance Statement Although preclinical evidence supports that GDNF provides neuroprotection and improves locomotor function in preclinical studies, clinical data supporting its efficacy to alleviate motor impairment in Parkinson's disease patients remains uncertain. Using the established 6-OHDA hemi-parkinsonian rat model, we determined whether expression of its cognate receptors, GFR-α1 and RET, were differentially affected between striatum and substantia nigra in a timeline study. In striatum, there was early and significant loss of RET, but a gradual, progressive loss of GFR-α1. In contrast, RET transiently increased in lesioned substantia nigra, but GFR-α1 progressively decreased only in nigrostriatal neurons and correlated with TH cell loss. Our results indicate that direct availability of GFR-α1 may be a critical element that determines GDNF efficacy following striatal delivery. Highlights GDNF expression was minimally affected by nigrostriatal lesionGDNF family receptor, GFR-α1, progressively decreased in striatum and in TH neurons in SN.GFR-α1 expression decreased along with TH neurons as lesion progressedGFR-α1 increased bilaterally in GFAP+ cells suggesting an inherent response to offset TH neuron lossRET expression was severely reduced in striatum, whereas it increased in SN early after lesion induction.
Collapse
|
15
|
AAV vectors applied to the treatment of CNS disorders: Clinical status and challenges. J Control Release 2023; 355:458-473. [PMID: 36736907 DOI: 10.1016/j.jconrel.2023.01.067] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023]
Abstract
In recent years, adeno-associated virus (AAV) has become the most important vector for central nervous system (CNS) gene therapy. AAV has already shown promising results in the clinic, for several CNS diseases that cannot be treated with drugs, including neurodegenerative diseases, neuromuscular diseases, and lysosomal storage disorders. Currently, three of the four commercially available AAV-based drugs focus on neurological disorders, including Upstaza for aromatic l-amino acid decarboxylase deficiency, Luxturna for hereditary retinal dystrophy, and Zolgensma for spinal muscular atrophy. All these studies have provided paradigms for AAV-based therapeutic intervention platforms. AAV gene therapy, with its dual promise of targeting disease etiology and enabling 'long-term correction' of disease processes, has the advantages of immune privilege, high delivery efficiency, tissue specificity, and cell tropism in the CNS. Although AAV-based gene therapy has been shown to be effective in most CNS clinical trials, limitations have been observed in its clinical applications, which are often associated with side effects. In this review, we summarized the therapeutic progress, challenges, limitations, and solutions for AAV-based gene therapy in 14 types of CNS diseases. We focused on viral vector technologies, delivery routes, immunosuppression, and other relevant clinical factors. We also attempted to integrate several hurdles faced in clinical and preclinical studies with their solutions, to seek the best path forward for the application of AAV-based gene therapy in the context of CNS diseases. We hope that these thoughtful recommendations will contribute to the efficient translation of preclinical studies and wide application of clinical trials.
Collapse
|
16
|
Kong C, Chang WS. Preclinical Research on Focused Ultrasound-Mediated Blood-Brain Barrier Opening for Neurological Disorders: A Review. Neurol Int 2023; 15:285-300. [PMID: 36810473 PMCID: PMC9944161 DOI: 10.3390/neurolint15010018] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/02/2023] [Accepted: 02/10/2023] [Indexed: 02/17/2023] Open
Abstract
Several therapeutic agents for neurological disorders are usually not delivered to the brain owing to the presence of the blood-brain barrier (BBB), a special structure present in the central nervous system (CNS). Focused ultrasound (FUS) combined with microbubbles can reversibly and temporarily open the BBB, enabling the application of various therapeutic agents in patients with neurological disorders. In the past 20 years, many preclinical studies on drug delivery through FUS-mediated BBB opening have been conducted, and the use of this method in clinical applications has recently gained popularity. As the clinical application of FUS-mediated BBB opening expands, it is crucial to understand the molecular and cellular effects of FUS-induced microenvironmental changes in the brain so that the efficacy of treatment can be ensured, and new treatment strategies established. This review describes the latest research trends in FUS-mediated BBB opening, including the biological effects and applications in representative neurological disorders, and suggests future directions.
Collapse
Affiliation(s)
| | - Won Seok Chang
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| |
Collapse
|
17
|
Olfat S, Mätlik K, Kopra JJ, Garton DR, Iivanainen VH, Bhattacharya D, Jakobsson J, Piepponen TP, Andressoo JO. Increased Physiological GDNF Levels Have No Effect on Dopamine Neuron Protection and Restoration in a Proteasome Inhibition Mouse Model of Parkinson's Disease. eNeuro 2023; 10:ENEURO.0097-22.2023. [PMID: 36690469 PMCID: PMC9910577 DOI: 10.1523/eneuro.0097-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 12/23/2022] [Accepted: 01/04/2023] [Indexed: 01/24/2023] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease that comprises a range of motor and nonmotor symptoms. Glial cell line-derived neurotrophic factor (GDNF) promotes the survival of dopamine neurons in vitro and in vivo, and intracranial delivery of GDNF has been tested in six clinical trials for treating PD. However, clinical trials with ectopic GDNF have yielded variable results, which could in part result from abnormal expression site and levels caused by ectopic overexpression. Therefore, an important open question is whether an increase in endogenous GDNF expression could be potent in reversing PD progression. Here, we tested the therapeutic potential of endogenous GDNF using mice in which endogenous GDNF can be conditionally upregulated specifically in cells that express GDNF naturally (conditional GDNF hypermorphic mice; GdnfcHyper ). We analyzed the impact of endogenous GDNF upregulation in both neuroprotection and neurorestoration procedures, and for both motor and nonmotor symptoms in the proteasome inhibitor lactacystin (LC) model of PD. Our results showed that upregulation of endogenous GDNF in the adult striatum is not protective in LC-induced PD model in mice. Since age is the largest risk factor for PD, we also analyzed the effect of deletion of endogenous GDNF in aged Gdnf conditional knock-out mice. We found that GDNF deletion does not increase susceptibility to LC-induced damage. We conclude that endogenous GDNF does not impact the outcome in the LC-induced proteasome inhibition mouse model of Parkinson's disease.
Collapse
Affiliation(s)
- Soophie Olfat
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm 17177, Sweden
- Department of Pharmacology, Faculty of Medicine, Neuroscience Center & Helsinki Institute of Life Science, University of Helsinki, Helsinki 00290, Finland
| | - Kärt Mätlik
- Department of Pharmacology, Faculty of Medicine, Neuroscience Center & Helsinki Institute of Life Science, University of Helsinki, Helsinki 00290, Finland
| | - Jaakko J Kopra
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
| | - Daniel R Garton
- Department of Pharmacology, Faculty of Medicine, Neuroscience Center & Helsinki Institute of Life Science, University of Helsinki, Helsinki 00290, Finland
| | - Vilma H Iivanainen
- Department of Pharmacology, Faculty of Medicine, Neuroscience Center & Helsinki Institute of Life Science, University of Helsinki, Helsinki 00290, Finland
| | - Dipabarna Bhattacharya
- Department of Pharmacology, Faculty of Medicine, Neuroscience Center & Helsinki Institute of Life Science, University of Helsinki, Helsinki 00290, Finland
| | - Johan Jakobsson
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, Lund 221 84, Sweden
| | - T Petteri Piepponen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
| | - Jaan-Olle Andressoo
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm 17177, Sweden
- Department of Pharmacology, Faculty of Medicine, Neuroscience Center & Helsinki Institute of Life Science, University of Helsinki, Helsinki 00290, Finland
| |
Collapse
|
18
|
Modulation of nigral dopamine signaling mitigates parkinsonian signs of aging: evidence from intervention with calorie restriction or inhibition of dopamine uptake. GeroScience 2023; 45:45-63. [PMID: 35635679 PMCID: PMC9886753 DOI: 10.1007/s11357-022-00583-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 04/29/2022] [Indexed: 02/03/2023] Open
Abstract
Identifying neurobiological mechanisms of aging-related parkinsonism, and lifestyle interventions that mitigate them, remain critical knowledge gaps. No aging study, from rodent to human, has reported loss of any dopamine (DA) signaling marker near the magnitude associated with onset of parkinsonian signs in Parkinson's disease (PD). However, in substantia nigra (SN), similar loss of DA signaling markers in PD or aging coincide with parkinsonian signs. Alleviation of these parkinsonian signs may be possible by interventions such as calorie restriction (CR), which augment DA signaling markers like tyrosine hydroxylase (TH) expression in the SN, but not striatum. Here, we interrogated respective contributions of nigral and striatal DA mechanisms to aging-related parkinsonian signs in aging (18 months old) rats in two studies: by the imposition of CR for 6 months, and inhibition of DA uptake within the SN or striatum by cannula-directed infusion of nomifensine. Parkinsonian signs were mitigated within 12 weeks after CR and maintained until 24 months old, commensurate with increased D1 receptor expression in the SN alone, and increased GDNF family receptor, GFR-α1, in the striatum, suggesting increased GDNF signaling. Nomifensine infusion into the SN or striatum selectively increased extracellular DA. However, only nigral infusion increased locomotor activity. These results indicate mechanisms that increase components of DA signaling in the SN alone mitigate parkinsonian signs in aging, and are modifiable by interventions, like CR, to offset parkinsonian signs, even at advanced age. Moreover, these results give evidence that changes in nigral DA signaling may modulate some parameters of locomotor activity autonomously from striatal DA signaling.
Collapse
|
19
|
The Effects of Dietary Interventions on Brain Aging and Neurological Diseases. Nutrients 2022; 14:nu14235086. [PMID: 36501116 PMCID: PMC9740746 DOI: 10.3390/nu14235086] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/17/2022] [Accepted: 11/23/2022] [Indexed: 12/02/2022] Open
Abstract
Dietary interventions can ameliorate age-related neurological decline. Decades of research of in vitro studies, animal models, and clinical trials support their ability and efficacy to improve behavioral outcomes by inducing biochemical and physiological changes that lead to a more resilient brain. Dietary interventions including calorie restriction, alternate day fasting, time restricted feeding, and fasting mimicking diets not only improve normal brain aging but also slow down, or even reverse, the progression of neurological diseases. In this review, we focus on the effects of intermittent and periodic fasting on improving phenotypic outcomes, such as cognitive and motor-coordination decline, in the normal aging brain through an increase in neurogenesis and synaptic plasticity, and decrease in neuroinflammation, mitochondrial dysfunction, and oxidative stress. We summarize the results of various dietary interventions in animal models of age-related neurological diseases such as Alzheimer's disease, Parkinson's disease, epilepsy, and Multiple Sclerosis and discuss the results of clinical trials that explore the feasibility of dietary interventions in the prevention and treatment of these diseases.
Collapse
|
20
|
Li SH, Abd-Elrahman KS, Ferguson SS. Targeting mGluR2/3 for treatment of neurodegenerative and neuropsychiatric diseases. Pharmacol Ther 2022; 239:108275. [DOI: 10.1016/j.pharmthera.2022.108275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/23/2022] [Accepted: 08/23/2022] [Indexed: 10/15/2022]
|
21
|
Neuropathology of the Basal Ganglia in SNCA Transgenic Rat Model of Parkinson's Disease: Involvement of Parvalbuminergic Interneurons and Glial-Derived Neurotropic Factor. Int J Mol Sci 2022; 23:ijms231710126. [PMID: 36077524 PMCID: PMC9456397 DOI: 10.3390/ijms231710126] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/19/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disease characterized by the accumulation of alpha-synuclein, encoded by the SNCA gene. The main neuropathological hallmark of PD is the degeneration of dopaminergic neurons leading to striatal dopamine depletion. Trophic support by a neurotrophin called glial-derived neurotrophic factor (GDNF) is also lacking in PD. We performed immunohistochemical studies to investigate neuropathological changes in the basal ganglia of a rat transgenic model of PD overexpressing alfa-synuclein. We observed that neuronal loss also occurs in the dorsolateral part of the striatum in the advanced stages of the disease. Moreover, along with the degeneration of the medium spiny projection neurons, we found a dramatic loss of parvalbumin interneurons. A marked decrease in GDNF, which is produced by parvalbumin interneurons, was observed in the striatum and in the substantia nigra of these animals. This confirmed the involvement of the striatum in the pathophysiology of PD and the importance of GDNF in maintaining the health of the substantia nigra.
Collapse
|
22
|
Gordon J, Lockard G, Monsour M, Alayli A, Choudhary H, Borlongan CV. Sequestration of Inflammation in Parkinson's Disease via Stem Cell Therapy. Int J Mol Sci 2022; 23:ijms231710138. [PMID: 36077534 PMCID: PMC9456021 DOI: 10.3390/ijms231710138] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 12/02/2022] Open
Abstract
Parkinson’s disease is the second most common neurodegenerative disease. Insidious and progressive, this disorder is secondary to the gradual loss of dopaminergic signaling and worsening neuroinflammation, affecting patients’ motor capabilities. Gold standard treatment includes exogenous dopamine therapy in the form of levodopa–carbidopa, or surgical intervention with a deep brain stimulator to the subcortical basal ganglia. Unfortunately, these therapies may ironically exacerbate the already pro-inflammatory environment. An alternative approach may involve cell-based therapies. Cell-based therapies, whether endogenous or exogenous, often have anti-inflammatory properties. Alternative strategies, such as exercise and diet modifications, also appear to play a significant role in facilitating endogenous and exogenous stem cells to induce an anti-inflammatory response, and thus are of unique interest to neuroinflammatory conditions including Parkinson’s disease. Treating patients with current gold standard therapeutics and adding adjuvant stem cell therapy, alongside the aforementioned lifestyle modifications, may ideally sequester inflammation and thus halt neurodegeneration.
Collapse
Affiliation(s)
- Jonah Gordon
- Morsani College of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Gavin Lockard
- Morsani College of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Molly Monsour
- Morsani College of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Adam Alayli
- Morsani College of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Hassan Choudhary
- Morsani College of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Cesario V. Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Correspondence:
| |
Collapse
|
23
|
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: 12] [Impact Index Per Article: 4.0] [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
|
24
|
Wianny F, Dzahini K, Fifel K, Wilson CRE, Bernat A, Dolmazon V, Misery P, Lamy C, Giroud P, Cooper HM, Knoblauch K, Procyk E, Kennedy H, Savatier P, Dehay C, Vezoli J. Induced Cognitive Impairments Reversed by Grafts of Neural Precursors: A Longitudinal Study in a Macaque Model of Parkinson's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103827. [PMID: 35137562 PMCID: PMC8981458 DOI: 10.1002/advs.202103827] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/14/2022] [Indexed: 05/10/2023]
Abstract
Parkinson's disease (PD) evolves over an extended and variable period in humans; years prior to the onset of classical motor symptoms, sleep and biological rhythm disorders develop, significantly impacting the quality-of-life of patients. Circadian-rhythm disorders are accompanied by mild cognitive deficits that progressively worsen with disease progression and can constitute a severe burden for patients at later stages. The gold-standard 6-methyl-1-methyl-4-phenyl-1,2,3,6-tetrahydropyridin (MPTP) macaque model of PD recapitulates the progression of motor and nonmotor symptoms over contracted periods of time. Here, this multidisciplinary/multiparametric study follows, in five animals, the steady progression of motor and nonmotor symptoms and describes their reversal following grafts of neural precursors in diverse functional domains of the basal ganglia. Results show unprecedented recovery from cognitive symptoms in addition to a strong clinical motor recuperation. Both motor and cognitive recovery and partial circadian rhythm recovery correlate with the degree of graft integration, and in a subset of animals, with in vivo levels of striatal dopaminergic innervation and function. The present study provides empirical evidence that integration of neural precursors following transplantation efficiently restores function at multiple levels in parkinsonian nonhuman primates and, given interindividuality of disease progression and recovery, underlines the importance of longitudinal multidisciplinary assessments in view of clinical translation.
Collapse
Affiliation(s)
- Florence Wianny
- Univ Lyon, Université Claude Bernard Lyon 1Inserm U1208Stem Cell and Brain Research InstituteBron69500France
- PrimastemBron69500France
| | - Kwamivi Dzahini
- Univ Lyon, Université Claude Bernard Lyon 1Inserm U1208Stem Cell and Brain Research InstituteBron69500France
- PrimastemBron69500France
| | - Karim Fifel
- Univ Lyon, Université Claude Bernard Lyon 1Inserm U1208Stem Cell and Brain Research InstituteBron69500France
- Present address: International Institute for Integrative Sleep Medicine (WPI‐IIIS)University of TsukubaTsukubaIbaraki305‐8575Japan
| | - Charles Robert Eden Wilson
- Univ Lyon, Université Claude Bernard Lyon 1Inserm U1208Stem Cell and Brain Research InstituteBron69500France
| | - Agnieszka Bernat
- Univ Lyon, Université Claude Bernard Lyon 1Inserm U1208Stem Cell and Brain Research InstituteBron69500France
- Present address: Laboratory of Molecular DiagnosticsDepartment of BiotechnologyInter‐collegiate Faculty of BiotechnologyUniversity of Gdańsk and Medical University of GdańskGdańsk80‐307Poland
- Present address: Laboratory of Experimental EmbryologyInstitute of Genetics and Animal BiotechnologyPolish Academy of SciencesWarsaw05‐552Poland
| | - Virginie Dolmazon
- Univ Lyon, Université Claude Bernard Lyon 1Inserm U1208Stem Cell and Brain Research InstituteBron69500France
| | - Pierre Misery
- Univ Lyon, Université Claude Bernard Lyon 1Inserm U1208Stem Cell and Brain Research InstituteBron69500France
| | - Camille Lamy
- Univ Lyon, Université Claude Bernard Lyon 1Inserm U1208Stem Cell and Brain Research InstituteBron69500France
| | - Pascale Giroud
- Univ Lyon, Université Claude Bernard Lyon 1Inserm U1208Stem Cell and Brain Research InstituteBron69500France
| | - Howard Michael Cooper
- Univ Lyon, Université Claude Bernard Lyon 1Inserm U1208Stem Cell and Brain Research InstituteBron69500France
| | - Kenneth Knoblauch
- Univ Lyon, Université Claude Bernard Lyon 1Inserm U1208Stem Cell and Brain Research InstituteBron69500France
- National Centre for OpticsVision and Eye CareFaculty of Health and Social SciencesUniversity College of Southeast NorwayKongsbergN‐3603Norway
| | - Emmanuel Procyk
- Univ Lyon, Université Claude Bernard Lyon 1Inserm U1208Stem Cell and Brain Research InstituteBron69500France
| | - Henry Kennedy
- Univ Lyon, Université Claude Bernard Lyon 1Inserm U1208Stem Cell and Brain Research InstituteBron69500France
- Institute of NeuroscienceState Key Laboratory of NeuroscienceChinese Academy of Sciences (CAS) Key Laboratory of Primate NeurobiologyShanghai200031China
| | - Pierre Savatier
- Univ Lyon, Université Claude Bernard Lyon 1Inserm U1208Stem Cell and Brain Research InstituteBron69500France
- PrimastemBron69500France
| | - Colette Dehay
- Univ Lyon, Université Claude Bernard Lyon 1Inserm U1208Stem Cell and Brain Research InstituteBron69500France
- PrimastemBron69500France
| | - Julien Vezoli
- Univ Lyon, Université Claude Bernard Lyon 1Inserm U1208Stem Cell and Brain Research InstituteBron69500France
- Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck SocietyFrankfurt60528Germany
| |
Collapse
|
25
|
Stefani A, Pierantozzi M, Cardarelli S, Stefani L, Cerroni R, Conti M, Garasto E, Mercuri NB, Marini C, Sucapane P. Neurotrophins as Therapeutic Agents for Parkinson’s Disease; New Chances From Focused Ultrasound? Front Neurosci 2022; 16:846681. [PMID: 35401084 PMCID: PMC8990810 DOI: 10.3389/fnins.2022.846681] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 01/31/2022] [Indexed: 01/02/2023] Open
Abstract
Magnetic Resonance–guided Focused Ultrasound (MRgFUS) represents an effective micro-lesioning approach to target pharmaco-resistant tremor, mostly in patients afflicted by essential tremor (ET) and/or Parkinson’s disease (PD). So far, experimental protocols are verifying the clinical extension to other facets of the movement disorder galaxy (i.e., internal pallidus for disabling dyskinesias). Aside from those neurosurgical options, one of the most intriguing opportunities of this technique relies on its capability to remedy the impermeability of blood–brain barrier (BBB). Temporary BBB opening through low-intensity focused ultrasound turned out to be safe and feasible in patients with PD, Alzheimer’s disease, and amyotrophic lateral sclerosis. As a mere consequence of the procedures, some groups described even reversible but significant mild cognitive amelioration, up to hippocampal neurogenesis partially associated to the increased of endogenous brain-derived neurotrophic factor (BDNF). A further development elevates MRgFUS to the status of therapeutic tool for drug delivery of putative neurorestorative therapies. Since 2012, FUS-assisted intravenous administration of BDNF or neurturin allowed hippocampal or striatal delivery. Experimental studies emphasized synergistic modalities. In a rodent model for Huntington’s disease, engineered liposomes can carry glial cell line–derived neurotrophic factor (GDNF) plasmid DNA (GDNFp) to form a GDNFp-liposome (GDNFp-LPs) complex through pulsed FUS exposures with microbubbles; in a subacute MPTP-PD model, the combination of intravenous administration of neurotrophic factors (either through protein or gene delivery) plus FUS did curb nigrostriatal degeneration. Here, we explore these arguments, focusing on the current, translational application of neurotrophins in neurodegenerative diseases.
Collapse
Affiliation(s)
- Alessandro Stefani
- Department of System Medicine, Parkinson Center, University Tor Vergata, Rome, Italy
- *Correspondence: Alessandro Stefani,
| | | | - Silvia Cardarelli
- Department of System Medicine, Parkinson Center, University Tor Vergata, Rome, Italy
| | - Lucrezia Stefani
- Department of System Medicine, Parkinson Center, University Tor Vergata, Rome, Italy
| | - Rocco Cerroni
- Department of System Medicine, Parkinson Center, University Tor Vergata, Rome, Italy
| | - Matteo Conti
- Department of System Medicine, UOC Neurology, University Tor Vergata, Rome, Italy
| | - Elena Garasto
- Department of System Medicine, UOC Neurology, University Tor Vergata, Rome, Italy
| | - Nicola B. Mercuri
- Department of System Medicine, UOC Neurology, University Tor Vergata, Rome, Italy
| | - Carmine Marini
- UOC Neurology and Stroke Unit, University of L’Aquila, L’Aquila, Italy
| | | |
Collapse
|
26
|
Wang Y, Luan M, Xue L, Jin J, Xie A. Evaluation of the relationship between SORL1 gene polymorphism and Parkinson's disease in the Chinese population. Neurosci Lett 2022; 778:136602. [DOI: 10.1016/j.neulet.2022.136602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 03/26/2022] [Indexed: 11/29/2022]
|
27
|
Lin MS, Chen SM, Hua KF, Chen WJ, Hsieh CC, Lin CC. Freshwater Clam Extract Mitigates Neuroinflammation and Amplifies Neurotrophic Activity of Glia: Insights from In Vitro Model of Neurodegenerative Pathomechanism. J Clin Med 2022; 11:jcm11030553. [PMID: 35160004 PMCID: PMC8836940 DOI: 10.3390/jcm11030553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/16/2022] [Accepted: 01/20/2022] [Indexed: 11/28/2022] Open
Abstract
Background. An extensive body of research suggests that brain inflammation and oxidative stress are the underlying causes of Parkinson’s disease (PD), for which no potent therapeutic approach exists to mitigate the degradation of dopamine neurons. Freshwater clams, an ancient health food of Chinese origin, have been documented to exhibit anti-inflammatory and antioxidant effects. We previously reported that freshwater clam extract (FCE) can attenuate astrocytic activation and subsequent proinflammatory cytokine production from substantia nigra in an MPTP-induced PD mouse model. This article provides insight into the potential mechanisms through which FCE regulates neuroinflammation in a glia model of injury. Materials and methods. In total, 1 μg/mL lipopolysaccharide (LPS) and 200 μM rotenone were conducted in primary glial cell cultures to mimic the respective neuroinflammation and oxidative stress during injury-induced glial cell reactivation, which is relevant to the pathological process of PD. Results. FCE markedly reduced LPS-induced neuroinflammation by suppressing NO and TNF-α production and the expression of pro-inflammatory cytokines. In addition, FCE was effective at reducing rotenone-induced toxicity by diminishing ROS production, promoting antioxidant enzymes (SOD, catalase, and GPx) and minimizing the decline in glial-cell-secreted neurotrophic factors (GDNF, BDNF). These impacts ultimately led to a decrease in glial apoptosis. Conclusions. Evidence reveals that FCE is capable of stabilizing reactive glia, as demonstrated by reduced neuroinflammation, oxidative stress, the increased release of neurotrophic factors and the inhibition of apoptosis, which provides therapeutic insight into neurodegenerative diseases, including PD.
Collapse
Affiliation(s)
- Muh-Shi Lin
- Division of Neurosurgery, Department of Surgery, Kuang Tien General Hospital, Taichung 43303, Taiwan;
- Department of Biotechnology and Animal Science, College of Bioresources, National Ilan University, Yilan 26047, Taiwan; (K.-F.H.); (W.-J.C.); (C.-C.H.)
- Department of Biotechnology, College of Medical and Health Care, Hung Kuang University, Taichung 43302, Taiwan
- Department of Health Business Administration, College of Medical and Health Care, Hung Kuang University, Taichung 43302, Taiwan
| | - Shu-Mei Chen
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei Medical University, Taipei 110, Taiwan;
- Department of Surgery, School of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Kuo-Feng Hua
- Department of Biotechnology and Animal Science, College of Bioresources, National Ilan University, Yilan 26047, Taiwan; (K.-F.H.); (W.-J.C.); (C.-C.H.)
| | - Wei-Jung Chen
- Department of Biotechnology and Animal Science, College of Bioresources, National Ilan University, Yilan 26047, Taiwan; (K.-F.H.); (W.-J.C.); (C.-C.H.)
| | - Cho-Chen Hsieh
- Department of Biotechnology and Animal Science, College of Bioresources, National Ilan University, Yilan 26047, Taiwan; (K.-F.H.); (W.-J.C.); (C.-C.H.)
| | - Chai-Ching Lin
- Department of Biotechnology and Animal Science, College of Bioresources, National Ilan University, Yilan 26047, Taiwan; (K.-F.H.); (W.-J.C.); (C.-C.H.)
- Correspondence: ; Tel.: +886-3-9310592; Fax: +886-3-9280609
| |
Collapse
|
28
|
Sveinsdóttir HS, Decker A, Christensen C, Lucena PB, Þorsteinsson H, Richert E, Maier VH, Cornell R, Karlsson KÆ. Motility phenotype in a zebrafish vmat2 mutant. PLoS One 2022; 17:e0259753. [PMID: 34986152 PMCID: PMC8730441 DOI: 10.1371/journal.pone.0259753] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/20/2021] [Indexed: 01/22/2023] Open
Abstract
In the present study, we characterize a novel zebrafish mutant of solute carrier 18A2 (slc18a2), also known as vesicular monoamine transporter 2 (vmat2), that exhibits a behavioural phenotype partially consistent with human Parkinson´s disease. At six days-post-fertilization, behaviour was analysed and demonstrated that vmat2 homozygous mutant larvae, relative to wild types, show changes in motility in a photomotor assay, altered sleep parameters, and reduced dopamine cell number. Following an abrupt lights-off stimulus mutant larvae initiate larger movements but subsequently inhibit them to a lesser extent in comparison to wild-type larvae. Conversely, during a lights-on period, the mutant larvae are hypomotile. Thigmotaxis, a preference to avoid the centre of a behavioural arena, was increased in homozygotes over heterozygotes and wild types, as was daytime sleep ratio. Furthermore, incubating mutant larvae in pramipexole or L-Dopa partially rescued the motor phenotypes, as did injecting glial cell-derived neurotrophic factor (GDNF) into their brains. This novel vmat2 model represents a tool for high throughput pharmaceutical screens for novel therapeutics, in particular those that increase monoamine transport, and for studies of the function of monoamine transporters.
Collapse
Affiliation(s)
| | - Amanda Decker
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa, United States of America
| | | | | | | | - Elena Richert
- School of Science and Engineering, Reykjavik University, Reykjavik, Iceland
- Department of Psychology, University of Oldenburg, Oldenburg, Germany
| | | | - Robert Cornell
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa, United States of America
| | - Karl Ægir Karlsson
- 3Z, Reykjavik, Iceland
- School of Science and Engineering, Reykjavik University, Reykjavik, Iceland
- Biomedical Center, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
29
|
Costas C, Faro LR. Do Naturally Occurring Antioxidants Protect Against Neurodegeneration of the Dopaminergic System? A Systematic Revision in Animal Models of Parkinson's Disease. Curr Neuropharmacol 2022; 20:432-459. [PMID: 33882808 PMCID: PMC9413795 DOI: 10.2174/1570159x19666210421092725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/18/2021] [Accepted: 04/16/2021] [Indexed: 11/22/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease and is characterized by a significant decrease in dopamine levels, caused by progressive degeneration of the dopaminergic neurons in the nigrostriatal pathway. Multiple mechanisms have been implicated in its pathogenesis, including oxidative stress, neuroinflammation, protein aggregation, mitochondrial dysfunction, insufficient support for neurotrophic factors and cell apoptosis. The absence of treatments capable of slowing or stopping the progression of PD has increased the interest in the natural antioxidant substances present in the diet, since they have multiple beneficial properties and it is possible that they can influence the mechanisms responsible for the dysfunction and death of dopaminergic neurons. Thus, the purpose of this systematic review is to analyze the results obtained in a set of studies carried out in the last years, which describe the neuroprotective, antioxidant and regenerative functions of some naturally occurring antioxidants in experimental models of PD. The results show that the exogenous no enzymatic antioxidants can significantly modify the biochemical and behavioral mechanisms that contribute to the pathophysiology of Parkinsonism in experimental animals. Therefore, it is possible that they may contribute to effective neuroprotection by providing a significant improvement in neuropathological markers. In conclusion, the results of this review suggest that exogenous antioxidants can be promising therapeutic candidates for the prevention and treatment of PD.
Collapse
Affiliation(s)
- Carmen Costas
- Department of Functional Biology and Health Sciences, Faculty of Biology, University of Vigo, Campus Lagoas-Marcosende, 36310, Vigo, Spain
| | - Lilian R.F. Faro
- Department of Functional Biology and Health Sciences, Faculty of Biology, University of Vigo, Campus Lagoas-Marcosende, 36310, Vigo, Spain
| |
Collapse
|
30
|
Grosso Jasutkar H, Oh SE, Mouradian MM. Therapeutics in the Pipeline Targeting α-Synuclein for Parkinson's Disease. Pharmacol Rev 2022; 74:207-237. [PMID: 35017177 PMCID: PMC11034868 DOI: 10.1124/pharmrev.120.000133] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/21/2021] [Indexed: 02/06/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder and the fastest growing neurologic disease in the world, yet no disease-modifying therapy is available for this disabling condition. Multiple lines of evidence implicate the protein α-synuclein (α-Syn) in the pathogenesis of PD, and as such, there is intense interest in targeting α-Syn for potential disease modification. α-Syn is also a key pathogenic protein in other synucleionpathies, most commonly dementia with Lewy bodies. Thus, therapeutics targeting this protein will have utility in these disorders as well. Here we discuss the various approaches that are being investigated to prevent and mitigate α-Syn toxicity in PD, including clearing its pathologic aggregates from the brain using immunization strategies, inhibiting its misfolding and aggregation, reducing its expression level, enhancing cellular clearance mechanisms, preventing its cell-to-cell transmission within the brain and perhaps from the periphery, and targeting other proteins associated with or implicated in PD that contribute to α-Syn toxicity. We also discuss the therapeutics in the pipeline that harness these strategies. Finally, we discuss the challenges and opportunities for the field in the discovery and development of therapeutics for disease modification in PD. SIGNIFICANCE STATEMENT: PD is the second most common neurodegenerative disorder, for which disease-modifying therapies remain a major unmet need. A large body of evidence points to α-synuclein as a key pathogenic protein in this disease as well as in dementia with Lewy bodies, making it of leading therapeutic interest. This review discusses the various approaches being investigated and progress made to date toward discovering and developing therapeutics that would slow and stop progression of these disabling diseases.
Collapse
Affiliation(s)
- Hilary Grosso Jasutkar
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, New Jersey
| | - Stephanie E Oh
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, New Jersey
| | - M Maral Mouradian
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, New Jersey
| |
Collapse
|
31
|
Elevated endogenous GDNF induces altered dopamine signalling in mice and correlates with clinical severity in schizophrenia. Mol Psychiatry 2022; 27:3247-3261. [PMID: 35618883 PMCID: PMC9708553 DOI: 10.1038/s41380-022-01554-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 11/08/2022]
Abstract
Presynaptic increase in striatal dopamine is the primary dopaminergic abnormality in schizophrenia, but the underlying mechanisms are not understood. Here, we hypothesized that increased expression of endogenous GDNF could induce dopaminergic abnormalities that resemble those seen in schizophrenia. To test the impact of GDNF elevation, without inducing adverse effects caused by ectopic overexpression, we developed a novel in vivo approach to conditionally increase endogenous GDNF expression. We found that a 2-3-fold increase in endogenous GDNF in the brain was sufficient to induce molecular, cellular, and functional changes in dopamine signalling in the striatum and prefrontal cortex, including increased striatal presynaptic dopamine levels and reduction of dopamine in prefrontal cortex. Mechanistically, we identified adenosine A2a receptor (A2AR), a G-protein coupled receptor that modulates dopaminergic signalling, as a possible mediator of GDNF-driven dopaminergic abnormalities. We further showed that pharmacological inhibition of A2AR with istradefylline partially normalised striatal GDNF and striatal and cortical dopamine levels in mice. Lastly, we found that GDNF levels are increased in the cerebrospinal fluid of first episode psychosis patients, and in post-mortem striatum of schizophrenia patients. Our results reveal a possible contributor for increased striatal dopamine signalling in a subgroup of schizophrenia patients and suggest that GDNF-A2AR crosstalk may regulate dopamine function in a therapeutically targetable manner.
Collapse
|
32
|
Dai H, Wang L, Li L, Huang Z, Ye L. Metallothionein 1: A New Spotlight on Inflammatory Diseases. Front Immunol 2021; 12:739918. [PMID: 34804020 PMCID: PMC8602684 DOI: 10.3389/fimmu.2021.739918] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/18/2021] [Indexed: 01/15/2023] Open
Abstract
MT1 has been demonstrated to be an essential stress protein in maintaining physiological balance and regulating immune homeostasis. While the immunological involvement of MT1 in central nervous system disorders and cancer has been extensively investigated, mounting evidence suggests that MT1 has a broader role in inflammatory diseases and can shape innate and adaptive immunity. In this review, we will first summarize the biological features of MT1 and the regulators that influence MT1 expression, emphasizing metal, inflammation, and immunosuppressive factors. We will then focus on the immunoregulatory function of MT1 on diverse immune cells and the signaling pathways regulated by MT1. Finally, we will discuss recent advances in our knowledge of the biological role of MT1 in several inflammatory diseases to develop novel therapeutic strategies.
Collapse
Affiliation(s)
- Hanying Dai
- Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Lu Wang
- Respiratory Medicine Department, Shenzhen University General Hospital, Shenzhen, China
| | - Lingyun Li
- Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Zhong Huang
- Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Liang Ye
- Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| |
Collapse
|
33
|
Mechanistic Insight from Preclinical Models of Parkinson's Disease Could Help Redirect Clinical Trial Efforts in GDNF Therapy. Int J Mol Sci 2021; 22:ijms222111702. [PMID: 34769132 PMCID: PMC8583859 DOI: 10.3390/ijms222111702] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 10/23/2021] [Accepted: 10/25/2021] [Indexed: 11/16/2022] Open
Abstract
Parkinson’s disease (PD) is characterized by four pathognomonic hallmarks: (1) motor and non-motor deficits; (2) neuroinflammation and oxidative stress; (3) pathological aggregates of the α-synuclein (α-syn) protein; (4) neurodegeneration of the nigrostriatal system. Recent evidence sustains that the aggregation of pathological α-syn occurs in the early stages of the disease, becoming the first trigger of neuroinflammation and subsequent neurodegeneration. Thus, a therapeutic line aims at striking back α-synucleinopathy and neuroinflammation to impede neurodegeneration. Another therapeutic line is restoring the compromised dopaminergic system using neurotrophic factors, particularly the glial cell-derived neurotrophic factor (GDNF). Preclinical studies with GDNF have provided encouraging results but often lack evaluation of anti-α-syn and anti-inflammatory effects. In contrast, clinical trials have yielded imprecise results and have reported the emergence of severe side effects. Here, we analyze the discrepancy between preclinical and clinical outcomes, review the mechanisms of the aggregation of pathological α-syn, including neuroinflammation, and evaluate the neurorestorative properties of GDNF, emphasizing its anti-α-syn and anti-inflammatory effects in preclinical and clinical trials.
Collapse
|
34
|
Wang Y, Hu LF, Zhou TJ, Qi LY, Xing L, Lee J, Wang FZ, Oh YK, Jiang HL. Gene therapy strategies for rare monogenic disorders with nuclear or mitochondrial gene mutations. Biomaterials 2021; 277:121108. [PMID: 34478929 DOI: 10.1016/j.biomaterials.2021.121108] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 08/19/2021] [Accepted: 08/26/2021] [Indexed: 12/26/2022]
Abstract
Rare monogenic disorders are a group of single-gene-mutated diseases that have a low incidence rate (less than 0.5‰) and eventually lead to patient disability and even death. Due to the relatively low number of people affected, these diseases typically fail to attract a great deal of commercial investment and research interest, and the affected patients thus have unmet medical needs. Advances in genomics biology, gene editing, and gene delivery can now offer potentially effective options for treating rare monogenic diseases. Herein, we review the application of gene therapy strategies (traditional gene therapy and gene editing) against various rare monogenic diseases with nuclear or mitochondrial gene mutations, including eye, central nervous system, pulmonary, systemic, and blood cell diseases. We summarize their pathologic features, address the barriers to gene delivery for these diseases, discuss available therapies in the clinic and in clinical trials, and sum up in-development gene delivery systems for various rare monogenic disorders. Finally, we elaborate the possible directions and outlook of gene therapy for rare monogenic disorders.
Collapse
Affiliation(s)
- Yi Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Li-Fan Hu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Tian-Jiao Zhou
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Lian-Yu Qi
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Lei Xing
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China; State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, 830054, China
| | - Jaiwoo Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Feng-Zhen Wang
- Department of Clinical Pharmacy, The First Clinical School of Xuzhou Medical University, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, China.
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Hu-Lin Jiang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China; State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, 830054, China; Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
35
|
Mitra S, Turconi G, Darreh-Shori T, Mätlik K, Aquilino M, Eriksdotter M, Andressoo JO. Increased Endogenous GDNF in Mice Protects Against Age-Related Decline in Neuronal Cholinergic Markers. Front Aging Neurosci 2021; 13:714186. [PMID: 34475820 PMCID: PMC8406776 DOI: 10.3389/fnagi.2021.714186] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/14/2021] [Indexed: 12/12/2022] Open
Abstract
Gradual decline in cholinergic transmission and cognitive function occurs during normal aging, whereas pathological loss of cholinergic function is a hallmark of different types of dementia, including Alzheimer’s disease (AD), Lewy body dementia (LBD), and Parkinson’s disease dementia (PDD). Glial cell line-derived neurotrophic factor (GDNF) is known to modulate and enhance the dopamine system. However, how endogenous GDNF influences brain cholinergic transmission has remained elusive. In this study, we explored the effect of a twofold increase in endogenous GDNF (Gdnf hypermorphic mice, Gdnfwt/hyper) on cholinergic markers and cognitive function upon aging. We found that Gdnfwt/hyper mice resisted an overall age-associated decline in the cholinergic index observed in the brain of Gdnfwt/wt animals. Biochemical analysis revealed that the level of nerve growth factor (NGF), which is important for survival and function of central cholinergic neurons, was significantly increased in several brain areas of old Gdnfwt/hyper mice. Analysis of expression of genes involved in cholinergic transmission in the cortex and striatum confirmed modulation of cholinergic pathways by GDNF upon aging. In line with these findings, Gdnfwt/hyper mice did not undergo an age-related decline in cognitive function in the Y-maze test, as observed in the wild type littermates. Our results identify endogenous GDNF as a potential modulator of cholinergic transmission and call for future studies on endogenous GDNF function in neurodegenerative disorders characterized by cognitive impairments, including AD, LBD, and PDD.
Collapse
Affiliation(s)
- Sumonto Mitra
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Huddinge, Sweden
| | - Giorgio Turconi
- Department of Pharmacology, Faculty of Medicine and Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Taher Darreh-Shori
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Huddinge, Sweden
| | - Kärt Mätlik
- Department of Pharmacology, Faculty of Medicine and Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Matilde Aquilino
- Department of Pharmacology, Faculty of Medicine and Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Maria Eriksdotter
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Huddinge, Sweden.,Theme Inflammation and Aging, Karolinska University Hospital, Huddinge, Sweden
| | - Jaan-Olle Andressoo
- Department of Pharmacology, Faculty of Medicine and Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland.,Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
36
|
Arango D, Bittar A, Esmeral NP, Ocasión C, Muñoz-Camargo C, Cruz JC, Reyes LH, Bloch NI. Understanding the Potential of Genome Editing in Parkinson's Disease. Int J Mol Sci 2021; 22:9241. [PMID: 34502143 PMCID: PMC8430539 DOI: 10.3390/ijms22179241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 01/05/2023] Open
Abstract
CRISPR is a simple and cost-efficient gene-editing technique that has become increasingly popular over the last decades. Various CRISPR/Cas-based applications have been developed to introduce changes in the genome and alter gene expression in diverse systems and tissues. These novel gene-editing techniques are particularly promising for investigating and treating neurodegenerative diseases, including Parkinson's disease, for which we currently lack efficient disease-modifying treatment options. Gene therapy could thus provide treatment alternatives, revolutionizing our ability to treat this disease. Here, we review our current knowledge on the genetic basis of Parkinson's disease to highlight the main biological pathways that become disrupted in Parkinson's disease and their potential as gene therapy targets. Next, we perform a comprehensive review of novel delivery vehicles available for gene-editing applications, critical for their successful application in both innovative research and potential therapies. Finally, we review the latest developments in CRISPR-based applications and gene therapies to understand and treat Parkinson's disease. We carefully examine their advantages and shortcomings for diverse gene-editing applications in the brain, highlighting promising avenues for future research.
Collapse
Affiliation(s)
- David Arango
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (D.A.); (A.B.); (N.P.E.); (C.M.-C.); (J.C.C.)
| | - Amaury Bittar
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (D.A.); (A.B.); (N.P.E.); (C.M.-C.); (J.C.C.)
| | - Natalia P. Esmeral
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (D.A.); (A.B.); (N.P.E.); (C.M.-C.); (J.C.C.)
| | - Camila Ocasión
- Grupo de Diseño de Productos y Procesos, Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (C.O.); (L.H.R.)
| | - Carolina Muñoz-Camargo
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (D.A.); (A.B.); (N.P.E.); (C.M.-C.); (J.C.C.)
| | - Juan C. Cruz
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (D.A.); (A.B.); (N.P.E.); (C.M.-C.); (J.C.C.)
| | - Luis H. Reyes
- Grupo de Diseño de Productos y Procesos, Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (C.O.); (L.H.R.)
| | - Natasha I. Bloch
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (D.A.); (A.B.); (N.P.E.); (C.M.-C.); (J.C.C.)
| |
Collapse
|
37
|
Behl T, Kaur I, Kumar A, Mehta V, Zengin G, Arora S. Gene Therapy in the Management of Parkinson's Disease: Potential of GDNF as a Promising Therapeutic Strategy. Curr Gene Ther 2021; 20:207-222. [PMID: 32811394 DOI: 10.2174/1566523220999200817164051] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/14/2020] [Accepted: 07/15/2020] [Indexed: 12/13/2022]
Abstract
The limitations of conventional treatment therapies in Parkinson's disorder, a common neurodegenerative disorder, lead to the development of an alternative gene therapy approach. Multiple treatment options targeting dopaminergic neuronal regeneration, production of enzymes linked with dopamine synthesis, subthalamic nucleus neurons, regulation of astrocytes and microglial cells and potentiating neurotrophic factors, were established. Viral vector-based dopamine delivery, prodrug approaches, fetal ventral mesencephalon tissue transplantation and dopamine synthesizing enzyme encoding gene delivery are significant therapies evidently supported by numerous trials. The review primarily elaborates on the significant role of glial cell-line derived neurotrophic factor in alleviating motor symptoms and the loss of dopaminergic neurons in Parkinson's disease. Neuroprotective and neuroregenerative effects of GDNF were established via preclinical and clinical study outcomes. The binding of GDNF family ligands with associated receptors leads to the formation of a receptor-ligand complex activating Ret receptor of tyrosine kinase family, which is only expressed in dopaminergic neurons, playing an important role in Parkinson's disease, via its association with the essential protein encoded genes. Furthermore, the review establishes delivery aspects, like ventricular delivery of recombinant GDNF, intraparenchymal and intraputaminal delivery using infusion catheters. The review highlights problems and challenges of GDNF delivery, and essential measures to overcome them, like gene therapy combinations, optimization of delivery vectors, newer targeting devices, motor symptoms curbing focused ultrasound techniques, modifications in patient selection criteria and development of novel delivery strategies based on liposomes and encapsulated cells, to promote safe and effective delivery of neurotrophic factor and establishment of routine treatment therapy for patients.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Ishnoor Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | | | - Gokhan Zengin
- Department of Biology, Faculty of Science, Selcuk University Campus, Konya, Turkey
| | - Sandeep Arora
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| |
Collapse
|
38
|
Renko JM, Mahato AK, Visnapuu T, Valkonen K, Karelson M, Voutilainen MH, Saarma M, Tuominen RK, Sidorova YA. Neuroprotective Potential of a Small Molecule RET Agonist in Cultured Dopamine Neurons and Hemiparkinsonian Rats. JOURNAL OF PARKINSONS DISEASE 2021; 11:1023-1046. [PMID: 34024778 PMCID: PMC8461720 DOI: 10.3233/jpd-202400] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Parkinson's disease (PD) is a progressive neurological disorder where loss of dopamine neurons in the substantia nigra and dopamine depletion in the striatum cause characteristic motor symptoms. Currently, no treatment is able to halt the progression of PD. Glial cell line-derived neurotrophic factor (GDNF) rescues degenerating dopamine neurons both in vitro and in animal models of PD. When tested in PD patients, however, the outcomes from intracranial GDNF infusion paradigms have been inconclusive, mainly due to poor pharmacokinetic properties. OBJECTIVE We have developed drug-like small molecules, named BT compounds that activate signaling through GDNF's receptor, the transmembrane receptor tyrosine kinase RET, both in vitro and in vivo and are able to penetrate through the blood-brain barrier. Here we evaluated the properties of BT44, a second generation RET agonist, in immortalized cells, dopamine neurons and rat 6-hydroxydopamine model of PD. METHODS We used biochemical, immunohistochemical and behavioral methods to evaluate the effects of BT44 on dopamine system in vitro and in vivo. RESULTS BT44 selectively activated RET and intracellular pro-survival AKT and MAPK signaling pathways in immortalized cells. In primary midbrain dopamine neurons cultured in serum-deprived conditions, BT44 promoted the survival of the neurons derived from wild-type, but not from RET knockout mice. BT44 also protected cultured wild-type dopamine neurons from MPP+-induced toxicity. In a rat 6-hydroxydopamine model of PD, BT44 reduced motor imbalance and seemed to protect dopaminergic fibers in the striatum. CONCLUSION BT44 holds potential for further development into a novel, possibly disease-modifying, therapy for PD.
Collapse
Affiliation(s)
- Juho-Matti Renko
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Arun Kumar Mahato
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Tanel Visnapuu
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Konsta Valkonen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Mati Karelson
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Merja H Voutilainen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Raimo K Tuominen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Yulia A Sidorova
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
39
|
Li D, Zhang J, Li X, Chen Y, Yu F, Liu Q. Insights into lncRNAs in Alzheimer's disease mechanisms. RNA Biol 2021; 18:1037-1047. [PMID: 32605500 PMCID: PMC8216181 DOI: 10.1080/15476286.2020.1788848] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 06/19/2020] [Accepted: 06/22/2020] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the most common dementia among the elderly. The pathophysiology of AD is characterized by two hallmarks: amyloid plaques, produced by amyloid β (Aβ) aggregation, and neurofibrillary tangle (NFT), produced by accumulation of phosphorylated tau. The regulatory roles of non-coding RNAs (ncRNAs), particularly long noncoding RNAs (lncRNAs), have been widely recognized in gene expression at the transcriptional and posttranscriptional levels. Mounting evidence shows that lncRNAs are aberrantly expressed in AD progression. Here, we review the lncRNAs that implicated in the regulation of Aβ peptide, tau, inflammation, cell death, and other aspects which are the main mechanisms of AD pathology. We also discuss the possible clinical or therapeutic utility of lncRNA detection or targeting to help diagnose or possibly combat AD.
Collapse
Affiliation(s)
- Dingfeng Li
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
- Neurodegenerative Disease Research Center, University of Science and Technology of China, Hefei, China
- National Synchrotron Radiation Laboratory, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei, China
| | - Juan Zhang
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
- Neurodegenerative Disease Research Center, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei, China
| | - Xiaohui Li
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
- Neurodegenerative Disease Research Center, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei, China
| | - Yuhua Chen
- Department of Neurology, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Feng Yu
- Department of Neurology, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Qiang Liu
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
- Neurodegenerative Disease Research Center, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei, China
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
40
|
Sung CY, Chiang PK, Tsai CW, Yang FY. Low-Intensity Pulsed Ultrasound Enhances Neurotrophic Factors and Alleviates Neuroinflammation in a Rat Model of Parkinson's Disease. Cereb Cortex 2021; 32:176-185. [PMID: 34196669 DOI: 10.1093/cercor/bhab201] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/04/2021] [Accepted: 06/07/2021] [Indexed: 12/30/2022] Open
Abstract
Low-intensity pulsed ultrasound (LIPUS) has also been reported to improve behavioral functions in Parkinson's disease (PD) animal models; however, the effect of LIPUS stimulation on the neurotrophic factors and neuroinflammation has not yet been addressed. PD rat model was built by injection of 6-hydroxydopamine (6-OHDA) in 2 sites in the right striatum. The levels of neurotrophic factors and lipocalin-2 (LCN2)-induced neuroinflammation were quantified using a western blot. Rotational test and cylinder test were conducted biweekly for 8 weeks. When the 6-OHDA + LIPUS and 6-OHDA groups were compared, the locomotor function of the 6-OHDA + LIPUS rats was significantly improved. After LIPUS stimulation, the tyrosine hydroxylase staining density was significantly increased in the striatum and substantia nigra pars compacta (SNpc) of lesioned rats. Unilateral LIPUS stimulation did not increase brain-derived neurotrophic factor in the striatum and SNpc of lesioned rats. In contrast, unilateral LIPUS stimulation increased glial cell line-derived neurotrophic factor (GDNF) protein 1.98-fold unilaterally in the SNpc. Additionally, LCN2-induced neuroinflammation can be attenuated following LIPUS stimulation. Our data indicated that LIPUS stimulation may be a potential therapeutic tool against PD via enhancement of GDNF level and inhibition of inflammatory responses in the SNpc of the brain.
Collapse
Affiliation(s)
- Chen-Yu Sung
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Pai-Kai Chiang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Urology, Mackay Memorial Hospital, Taipei, Taiwan
| | - Che-Wen Tsai
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Feng-Yi Yang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
41
|
Welleford AS, Quintero JE, Seblani NE, Blalock E, Gunewardena S, Shapiro SM, Riordan SM, Huettl P, Guduru Z, Stanford JA, van Horne CG, Gerhardt GA. RNA Sequencing of Human Peripheral Nerve in Response to Injury: Distinctive Analysis of the Nerve Repair Pathways. Cell Transplant 2021; 29:963689720926157. [PMID: 32425114 PMCID: PMC7563818 DOI: 10.1177/0963689720926157] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The development of regenerative therapies for central nervous system diseases can likely benefit from an understanding of the peripheral nervous system repair process, particularly in identifying potential gene pathways involved in human nerve repair. This study employed RNA sequencing (RNA-seq) technology to analyze the whole transcriptome profile of the human peripheral nerve in response to an injury. The distal sural nerve was exposed, completely transected, and a 1 to 2 cm section of nerve fascicles was collected for RNA-seq from six participants with Parkinson’s disease, ranging in age between 53 and 70 yr. Two weeks after the initial injury, another section of the nerve fascicles of the distal and pre-degenerated stump of the nerve was dissected and processed for RNA-seq studies. An initial analysis between the pre-lesion status and the postinjury gene expression revealed 3,641 genes that were significantly differentially expressed. In addition, the results support a clear transdifferentiation process that occurred by the end of the 2-wk postinjury. Gene ontology (GO) and hierarchical clustering were used to identify the major signaling pathways affected by the injury. In contrast to previous nonclinical studies, important changes were observed in molecular pathways related to antiapoptotic signaling, neurotrophic factor processes, cell motility, and immune cell chemotactic signaling. The results of our current study provide new insights regarding the essential interactions of different molecular pathways that drive neuronal repair and axonal regeneration in humans.
Collapse
Affiliation(s)
- Andrew S Welleford
- Department of Neuroscience, University of Kentucky Medical Center, Lexington, KY, USA.,Brain Restoration Center, University of Kentucky, Lexington, KY, USA.,* These are co-first authors and have contributed equally to this article
| | - Jorge E Quintero
- Department of Neuroscience, University of Kentucky Medical Center, Lexington, KY, USA.,Brain Restoration Center, University of Kentucky, Lexington, KY, USA.,Department of Neurosurgery, University of Kentucky Medical Center, Lexington, KY, USA.,* These are co-first authors and have contributed equally to this article
| | - Nader El Seblani
- Department of Neuroscience, University of Kentucky Medical Center, Lexington, KY, USA.,Brain Restoration Center, University of Kentucky, Lexington, KY, USA.,Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, USA.,* These are co-first authors and have contributed equally to this article
| | - Eric Blalock
- Department of Neuroscience, University of Kentucky Medical Center, Lexington, KY, USA.,Brain Restoration Center, University of Kentucky, Lexington, KY, USA
| | - Sumedha Gunewardena
- Kansas Intellectual and Developmental Disabilities Research Center, University of Kansas Medical Center, KS, USA
| | - Steven M Shapiro
- Division of Neurology, Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO, USA.,Department of Molecular and Integrative Physiology, University of Kansas Medical Center, KS, USA
| | - Sean M Riordan
- Division of Neurology, Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO, USA
| | - Peter Huettl
- Department of Neuroscience, University of Kentucky Medical Center, Lexington, KY, USA.,Brain Restoration Center, University of Kentucky, Lexington, KY, USA
| | - Zain Guduru
- Department of Neurology, University of Kentucky Medical Center, Lexington, KY, USA
| | - John A Stanford
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, KS, USA
| | - Craig G van Horne
- Department of Neuroscience, University of Kentucky Medical Center, Lexington, KY, USA.,Brain Restoration Center, University of Kentucky, Lexington, KY, USA.,Department of Neurosurgery, University of Kentucky Medical Center, Lexington, KY, USA
| | - Greg A Gerhardt
- Department of Neuroscience, University of Kentucky Medical Center, Lexington, KY, USA.,Brain Restoration Center, University of Kentucky, Lexington, KY, USA.,Department of Neurosurgery, University of Kentucky Medical Center, Lexington, KY, USA.,Department of Neurology, University of Kentucky Medical Center, Lexington, KY, USA
| |
Collapse
|
42
|
Burns TC, Quinones-Hinojosa A. Regenerative medicine for neurological diseases-will regenerative neurosurgery deliver? BMJ 2021; 373:n955. [PMID: 34162530 DOI: 10.1136/bmj.n955] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Regenerative medicine aspires to transform the future practice of medicine by providing curative, rather than palliative, treatments. Healing the central nervous system (CNS) remains among regenerative medicine's most highly prized but formidable challenges. "Regenerative neurosurgery" provides access to the CNS or its surrounding structures to preserve or restore neurological function. Pioneering efforts over the past three decades have introduced cells, neurotrophins, and genes with putative regenerative capacity into the CNS to combat neurodegenerative, ischemic, and traumatic diseases. In this review we critically evaluate the rationale, paradigms, and translational progress of regenerative neurosurgery, harnessing access to the CNS to protect, rejuvenate, or replace cell types otherwise irreversibly compromised by neurological disease. We discuss the evidence surrounding fetal, somatic, and pluripotent stem cell derived implants to replace endogenous neuronal and glial cell types and provide trophic support. Neurotrophin based strategies via infusions and gene therapy highlight the motivation to preserve neuronal circuits, the complex fidelity of which cannot be readily recreated. We specifically highlight ongoing translational efforts in Parkinson's disease, amyotrophic lateral sclerosis, stroke, and spinal cord injury, using these to illustrate the principles, challenges, and opportunities of regenerative neurosurgery. Risks of associated procedures and novel neurosurgical trials are discussed, together with the ethical challenges they pose. After decades of efforts to develop and refine necessary tools and methodologies, regenerative neurosurgery is well positioned to advance treatments for refractory neurological diseases. Strategic multidisciplinary efforts will be critical to harness complementary technologies and maximize mechanistic feedback, accelerating iterative progress toward cures for neurological diseases.
Collapse
Affiliation(s)
- Terry C Burns
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
43
|
Kotliarova A, Sidorova YA. Glial Cell Line-Derived Neurotrophic Factor Family Ligands, Players at the Interface of Neuroinflammation and Neuroprotection: Focus Onto the Glia. Front Cell Neurosci 2021; 15:679034. [PMID: 34220453 PMCID: PMC8250866 DOI: 10.3389/fncel.2021.679034] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/21/2021] [Indexed: 12/25/2022] Open
Abstract
Well-known effects of neurotrophic factors are related to supporting the survival and functioning of various neuronal populations in the body. However, these proteins seem to also play less well-documented roles in glial cells, thus, influencing neuroinflammation. This article summarizes available data on the effects of glial cell line derived neurotrophic factor (GDNF) family ligands (GFLs), proteins providing trophic support to dopaminergic, sensory, motor and many other neuronal populations, in non-neuronal cells contributing to the development and maintenance of neuropathic pain. The paper also contains our own limited data describing the effects of small molecules targeting GFL receptors on the expression of the satellite glial marker IBA1 in dorsal root ganglia of rats with surgery- and diabetes-induced neuropathy. In our experiments activation of GFLs receptors with either GFLs or small molecule agonists downregulated the expression of IBA1 in this tissue of experimental animals. While it can be a secondary effect due to a supportive role of GFLs in neuronal cells, growing body of evidence indicates that GFL receptors are expressed in glial and peripheral immune system cells. Thus, targeting GFL receptors with either proteins or small molecules may directly suppress the activation of glial and immune system cells and, therefore, reduce neuroinflammation. As neuroinflammation is considered to be an important contributor to the process of neurodegeneration these data further support research efforts to modulate the activity of GFL receptors in order to develop disease-modifying treatments for neurodegenerative disorders and neuropathic pain that target both neuronal and glial cells.
Collapse
Affiliation(s)
- Anastasiia Kotliarova
- Laboratory of Molecular Neuroscience, Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Yulia A Sidorova
- Laboratory of Molecular Neuroscience, Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
44
|
Sun S, Zhang Q, Li M, Gao P, Huang K, Beejadhursing R, Jiang W, Lei T, Zhu M, Shu K. GDNF Promotes Survival and Therapeutic Efficacy of Human Adipose-Derived Mesenchymal Stem Cells in a Mouse Model of Parkinson's Disease. Cell Transplant 2021; 29:963689720908512. [PMID: 32292068 PMCID: PMC7444207 DOI: 10.1177/0963689720908512] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cell (MSC)-based regenerative therapy is regarded as a promising strategy for the treatment of Parkinson's disease (PD). However, MSC components may exhibit poor intracranial survivability, particularly in the later stages following cell transplantation, limiting their potential curative effect and also clinical applications. Glial cell line-derived neurotrophic factor (GDNF), which encompasses a variety of transforming growth factor beta super family members, has been reported to enhance motor function and exert neuroprotective effects. However, no previous studies have investigated the effects of GDNF on human primary adipose-derived MSCs (hAMSCs), despite its potential for enhancing stem cell survival and promoting therapeutic efficacy in the treatment of PD. In the present study, we proposed a novel approach for enhancing the proliferative capacity and improving the efficacy of hAMSC treatment. Pre-exposure of engineered hAMSCs to GDNF enhanced the proliferation and differentiation of these stem cells in vitro. In addition, in 6-hydroxydopamine-lesioned mice, a common PD model, intracranial injection of hAMSCs-GDNF was associated with greater performance on behavioral tests, larger graft volumes 5 weeks after transplantation, and higher levels of Nestin, glial fibrillary acidic protein, and Tuj-1 differentiation than those treated with hAMSCs-Vector. Following transplantation of hAMSCs-GDNF into the striatum of lesioned models, we observed significant increases in tyrosine hydroxylase- and NeuN-positive staining. These findings highlight the therapeutic potential of hAMSCs-GDNF for patients with PD, as well as an efficient method for promoting therapeutic efficacy of these delivery vehicles.
Collapse
Affiliation(s)
- Shoujia Sun
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.,Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, People's Republic of China.,* Both the authors contributed equally to this article
| | - Quan Zhang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.,* Both the authors contributed equally to this article
| | - Man Li
- Department of Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Pan Gao
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Kuan Huang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Rajluxmee Beejadhursing
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Wei Jiang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Ting Lei
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Mingxin Zhu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Kai Shu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
45
|
Lonser RR, Akhter AS, Zabek M, Elder JB, Bankiewicz KS. Direct convective delivery of adeno-associated virus gene therapy for treatment of neurological disorders. J Neurosurg 2021; 134:1751-1763. [PMID: 32915526 DOI: 10.3171/2020.4.jns20701] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/16/2020] [Indexed: 11/06/2022]
Abstract
Molecular biological insights have led to a fundamental understanding of the underlying genomic mechanisms of nervous system disease. These findings have resulted in the identification of therapeutic genes that can be packaged in viral capsids for the treatment of a variety of neurological conditions, including neurodegenerative, metabolic, and enzyme deficiency disorders. Recent data have demonstrated that gene-carrying viral vectors (most often adeno-associated viruses) can be effectively distributed by convection-enhanced delivery (CED) in a safe, reliable, targeted, and homogeneous manner across the blood-brain barrier. Critically, these vectors can be monitored using real-time MRI of a co-infused surrogate tracer to accurately predict vector distribution and transgene expression at the perfused site. The unique properties of CED of adeno-associated virus vectors allow for cell-specific transgene manipulation of the infused anatomical site and/or widespread interconnected sites via antero- and/or retrograde transport. The authors review the convective properties of viral vectors, associated technology, and clinical applications.
Collapse
Affiliation(s)
- Russell R Lonser
- 1Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio; and
| | - Asad S Akhter
- 1Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio; and
| | - Mirosław Zabek
- 2Department of Neurological Surgery, Bródno Hospital, Warsaw, Poland
| | - J Bradley Elder
- 1Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio; and
| | - Krystof S Bankiewicz
- 1Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio; and
| |
Collapse
|
46
|
Chu Y, Kordower JH. GDNF signaling in subjects with minimal motor deficits and Parkinson's disease. Neurobiol Dis 2021; 153:105298. [PMID: 33684514 DOI: 10.1016/j.nbd.2021.105298] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/18/2021] [Accepted: 02/09/2021] [Indexed: 12/11/2022] Open
Abstract
The failure of glial cell derived neurotropic factor to be efficacious in blinded clinical trials for Parkinson's disease may be due to alterations in signaling receptors and downstream signaling molecules. To test this hypothesis, brain sections were obtained from older adults with no motor deficit (n = 6), minimal motor deficits (n = 10), and clinical diagnosis of Parkinson's disease (n = 10) who underwent motor examination proximate to death. Quantitative unbiased stereology and densitometry were performed to analyze RET and phosphorylated ribosomal protein S6 expression in nigral neurons. Individuals with no motor deficit had extensive and intense RET and phosphorylated ribosomal protein S6 immunoreactive neurons in substantia nigra. The number and staining intensity of RET-immunoreactive neurons were reduced moderately in subjects with minimal motor deficits and severely reduced in Parkinson's disease relative to no motor deficit group. The number and staining intensity of phosphorylated ribosomal protein S6 was more markedly reduced in both subjects with minimal motor deficits and Parkinson's disease. Reductions in levels of RET and phosphorylated ribosomal protein S6 were recapitulated in a non-human primate genetic Parkinson's disease model based on over-expression of human mutant α-synuclein (A53T). These data indicate that for neurotrophic factors to be effective in patients with minimal motor deficits or PD, these factors would likely have to upregulate RET and phosphorylated ribosomal protein S6 immunoreactive neurons in substantia nigra .
Collapse
Affiliation(s)
- Yaping Chu
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, United States of America
| | - Jeffrey H Kordower
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, United States of America.; ASU-Banner Neurodgenerative Disease Research Center, Arizona State University, Tempe, Arizona 85287, United States of America..
| |
Collapse
|
47
|
Ntetsika T, Papathoma PE, Markaki I. Novel targeted therapies for Parkinson's disease. Mol Med 2021; 27:17. [PMID: 33632120 PMCID: PMC7905684 DOI: 10.1186/s10020-021-00279-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/03/2021] [Accepted: 02/08/2021] [Indexed: 02/07/2023] Open
Abstract
Parkinson’s disease (PD) is the second more common neurodegenerative disease with increasing incidence worldwide associated to the population ageing. Despite increasing awareness and significant research advancements, treatment options comprise dopamine repleting, symptomatic therapies that have significantly increased quality of life and life expectancy, but no therapies that halt or reverse disease progression, which remain a great, unmet goal in PD research. Large biomarker development programs are undertaken to identify disease signatures that will improve patient selection and outcome measures in clinical trials. In this review, we summarize PD-related mechanisms that can serve as targets of therapeutic interventions aiming to slow or modify disease progression, as well as previous and ongoing clinical trials in each field, and discuss future perspectives.
Collapse
Affiliation(s)
- Theodora Ntetsika
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Center of Neurology, Academic Specialist Center, Solnavägen 1E, 113 65, Stockholm, Sweden
| | - Paraskevi-Evita Papathoma
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Neurology, Danderyd Hospital Stockholm, Stockholm, Sweden
| | - Ioanna Markaki
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden. .,Center of Neurology, Academic Specialist Center, Solnavägen 1E, 113 65, Stockholm, Sweden.
| |
Collapse
|
48
|
Kambey PA, Kanwore K, Ayanlaja AA, Nadeem I, Du Y, Buberwa W, Liu W, Gao D. Failure of Glial Cell-Line Derived Neurotrophic Factor (GDNF) in Clinical Trials Orchestrated By Reduced NR4A2 (NURR1) Transcription Factor in Parkinson's Disease. A Systematic Review. Front Aging Neurosci 2021; 13:645583. [PMID: 33716718 PMCID: PMC7943926 DOI: 10.3389/fnagi.2021.645583] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 01/29/2021] [Indexed: 12/23/2022] Open
Abstract
Parkinson’s disease (PD) is one of the most common neurodegenerative maladies with unforeseen complex pathologies. While this neurodegenerative disorder’s neuropathology is reasonably well known, its etiology remains a mystery, making it challenging to aim therapy. Glial cell-line derived neurotrophic factor (GDNF) remains an auspicious therapeutic molecule for treating PD. Neurotrophic factor derived from glial cell lines is effective in rodents and nonhuman primates, but clinical findings have been equivocal. Laborious exertions have been made over the past few decades to improve and assess GDNF in treating PD (clinical studies). Definitive clinical trials have, however, failed to demonstrate a survival advantage. Consequently, there seemed to be a doubt as to whether GDNF has merit in the potential treatment of PD. The purpose of this cutting edge review is to speculate as to why the clinical trials have failed to meet the primary endpoint. We introduce a hypothesis, “Failure of GDNF in clinical trials succumbed by nuclear receptor-related factor 1 (Nurr1) shortfall.” We demonstrate how Nurr1 binds to GDNF to induce dopaminergic neuron synthesis. Due to its undisputable neuro-protection aptitude, we display Nurr1 (also called Nr4a2) as a promising therapeutic target for PD.
Collapse
Affiliation(s)
- Piniel Alphayo Kambey
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Kouminin Kanwore
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Abiola Abdulrahman Ayanlaja
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Iqra Nadeem
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| | - YinZhen Du
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| | | | - WenYa Liu
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Dianshuai Gao
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
49
|
Kakoty V, K C S, Dubey SK, Yang CH, Kesharwani P, Taliyan R. Lentiviral mediated gene delivery as an effective therapeutic approach for Parkinson disease. Neurosci Lett 2021; 750:135769. [PMID: 33636285 DOI: 10.1016/j.neulet.2021.135769] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/07/2021] [Accepted: 02/19/2021] [Indexed: 11/28/2022]
Abstract
Continual strategies to devise a complete therapeutic cure for neurodegenerative conditions has been a challenge, majorly due to the presence of blood brain barrier. Lack of targeted delivery in order to minimize loss of dopamine (DA) neurones has been a major challenge to overcome anomalies in Parkinson Disease (PD). PD is a neuromotor degenerative disorder deteriorating motor coordination in affected individuals. Recent research has highlighted the use of lentiviral vectors (LVs) for selective delivery of neuroprotective substance for complete halt of disease progression in PD. LVs have the ability to infect both dividing and non-dividing cells along with non-encoding capability of viral protein that might elicit an immune response. This review will mainly focus on understanding the basic mechanism of action of LVs and its therapeutic aid in PD.
Collapse
Affiliation(s)
- Violina Kakoty
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani, India
| | - Sarathlal K C
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani, India
| | - Sunil Kumar Dubey
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani, India; R&D Healthcare Division, Emami Ltd, Kolkatta, India
| | - Chih Hao Yang
- Department of Pharmacology, Taipei Medical University, Taiwan
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India.
| | - Rajeev Taliyan
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani, India.
| |
Collapse
|
50
|
Christensen C, Þorsteinsson H, Maier VH, Karlsson KÆ. Multi-parameter Behavioral Phenotyping of the MPP+ Model of Parkinson's Disease in Zebrafish. Front Behav Neurosci 2021; 14:623924. [PMID: 33390914 PMCID: PMC7775599 DOI: 10.3389/fnbeh.2020.623924] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 11/30/2020] [Indexed: 12/18/2022] Open
Abstract
Parkinson's disease (PD) has been modeled in several animal species using the neurotoxins 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and its oxidized product 1-methyl-4-phenylpyridinium (MPP+). MPP+ selectively kills dopaminergic neurons in pars compacta of the substantia nigra, inducing parkinsonian symptoms in animals. Typically, neurotoxicity models of PD in zebrafish assess acute drug effects on locomotion. In the present study, we examined the lasting effects of MPP+ exposure and drug treatment in zebrafish larvae. Larvae were incubated in 500 μM MPP+, from 1 to 5 days post fertilization (dpf), followed by 24 h drug-free acclimation. At 6 dpf, the behavior was analyzed for locomotion, thigmotaxis, and sleep. Next, in separate assays we assessed the drug effects of brain injected glial cell-derived neurotrophic factor (GDNF) and 4-phenylbutyrate (PBA), co-incubated with MPP+. We show that MPP+ exposure consistently reduces swim distance, movement frequency, and cumulative time of movement; thus mimicking a parkinsonian phenotype of reduced movement. In contrast, MPP+ exposed larvae demonstrate reduced anxiety-like behavior and exhibit a sleep phenotype inconsistent with human PD: the larvae display longer sleep bouts, less sleep fragmentation, and more sleep. Previously reported rescuing effects of PBA were not replicated in this study. Moreover, whereas GDNF attenuated the sleep phenotype induced by MPP+, PBA augmented it. The current data suggest that MPP+ exposure generates a multifaceted phenotype in zebrafish and highlights that analyzing a narrow window of data can reveal effects that may be inconsistent with longer multi-parameter approaches. It further indicates that the model generally captures motor symptoms more faithfully than non-motor symptoms.
Collapse
Affiliation(s)
| | | | | | - Karl Ægir Karlsson
- 3Z Ehf, Reykjavik, Iceland.,Biomedical Center, University of Iceland, Reykjavik, Iceland.,Department of Engineering, School of Technology, Reykjavik University, Reykjavik, Iceland
| |
Collapse
|