101
|
Torres N, Molet J, Moro C, Mitrofanis J, Benabid AL. Neuroprotective Surgical Strategies in Parkinson's Disease: Role of Preclinical Data. Int J Mol Sci 2017; 18:ijms18102190. [PMID: 29053638 PMCID: PMC5666871 DOI: 10.3390/ijms18102190] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 10/10/2017] [Accepted: 10/13/2017] [Indexed: 12/18/2022] Open
Abstract
Although there have been many pharmacological agents considered to be neuroprotective therapy in Parkinson's disease (PD) patients, neurosurgical approaches aimed to neuroprotect or restore the degenerative nigrostriatal system have rarely been the focus of in depth reviews. Here, we explore the neuroprotective strategies involving invasive surgical approaches (NSI) using neurotoxic models 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and 6-hydroxydopamine (6-OHDA), which have led to clinical trials. We focus on several NSI approaches, namely deep brain stimulation of the subthalamic nucleus, glial neurotrophic derived factor (GDNF) administration and cell grafting methods. Although most of these interventions have produced positive results in preclinical animal models, either from behavioral or histological studies, they have generally failed to pass randomized clinical trials to validate each approach. We argue that NSI are promising approaches for neurorestoration in PD, but preclinical studies should be planned carefully in order not only to detect benefits but also to detect potential adverse effects. Further, clinical trials should be designed to be able to detect and disentangle neuroprotection from symptomatic effects. In summary, our review study evaluates the pertinence of preclinical models to study NSI for PD and how this affects their efficacy when translated into clinical trials.
Collapse
Affiliation(s)
- Napoleon Torres
- University Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, 38000 Grenoble, France.
| | - Jenny Molet
- University Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, 38000 Grenoble, France.
| | - Cecile Moro
- University Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, 38000 Grenoble, France.
| | - John Mitrofanis
- Department of Anatomy, University of Sydney; Sydney Medical School, Sydney NSW 2006, Australia.
| | - Alim Louis Benabid
- University Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, 38000 Grenoble, France.
| |
Collapse
|
102
|
Contrasting effects of selective MAGL and FAAH inhibition on dopamine depletion and GDNF expression in a chronic MPTP mouse model of Parkinson's disease. Neurochem Int 2017; 110:14-24. [PMID: 28826718 DOI: 10.1016/j.neuint.2017.08.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 06/08/2017] [Accepted: 08/03/2017] [Indexed: 01/07/2023]
Abstract
The modulation of the brain endocannabinoid system has been identified as an option to treat neurodegenerative diseases including Parkinson's disease (PD). Especially the elevation of endocannabinoid levels by inhibition of hydrolytic degradation represents a valuable approach. To evaluate whether monoacylglycerol lipase (MAGL) or fatty acid amide hydrolase (FAAH) inhibition could be beneficial for PD, we examined in parallel the therapeutic potential of the highly selective MAGL inhibitor KML29 elevating 2-arachidonoylglyerol (2-AG) levels and the highly selective FAAH inhibitor PF-3845 elevating anandamide (AEA) levels in a chronic methyl-4-phenyl-1,2,3,6-tetrahydropyridine/probenecid (MPTP/probenecid) mouse model of PD. Chronic administration of KML29 (10 mg/kg) but not PF-3845 (10 mg/kg) attenuated striatal MPTP/probenecid-induced dopamine depletion. Furthermore, KML29 induced an increase in Gdnf but not Bdnf expression, whereas PF-3845 decreased the MPTP/probenecid-induced Cnr2 expression without any effects on neurotrophin expression. Investigation of treatment-naïve striatal mRNA levels revealed a high presence of Gdnf and Mgll in contrast to Bdnf and Faah. Treatment of primary mouse microglia with 2-AG increased Gdnf but not Bdnf expression, suggesting that microglia might mediate the observed KML29-induced increase in Gdnf. In summary, pharmacological MAGL but not FAAH inhibition in the chronic MPTP/probenecid model attenuated the MPTP/probenecid-induced effects on striatal dopamine levels which were accompanied by an increase in 2-AG levels.
Collapse
|
103
|
Schwenkgrub J, Zaremba M, Joniec-Maciejak I, Cudna A, Mirowska-Guzel D, Kurkowska-Jastrzębska I. The phosphodiesterase inhibitor, ibudilast, attenuates neuroinflammation in the MPTP model of Parkinson's disease. PLoS One 2017; 12:e0182019. [PMID: 28753652 PMCID: PMC5533435 DOI: 10.1371/journal.pone.0182019] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 07/11/2017] [Indexed: 12/22/2022] Open
Abstract
Background/Aims Since the degeneration of the nigrostriatal dopaminergic pathway in Parkinson’s disease (PD) is associated with the inflammation process and decreased levels of cyclic nucleotides, inhibition of up-regulated cyclic nucleotide phosphodiesterases (PDEs) appears to be a promising therapeutic strategy. We used ibudilast (IBD), a non-selective PDE3,4,10,11 inhibitor, due to the abundant PDE 4 and 10 expression in the striatum. The present study for the first time examined the efficacy of IBD in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. Methods IBD [0, 20, 30, 40, or 50 mg/kg] was injected b.i.d. subcutaneously for nine days to three-month-old male C57Bl/10Tar mice, beginning two days prior to MPTP (60 mg/kg) intoxication. High-pressure liquid chromatography, Western blot analysis, and real time RT-PCR methods were applied. Results Our study demonstrated that chronic administration of IBD attenuated astroglial reactivity and increased glial cell-derived neurotrophic factor (GDNF) production in the striatum. Moreover, IBD reduced TNF-α, IL-6, and IL-1β expression. Conclusion IBD had a well-defined effect on astroglial activation in the mouse model of PD; however, there was no protective effect in the acute phase of injury. Diminished inflammation and an increased level of GDNF may provide a better outcome in the later stages of neurodegeneration.
Collapse
Affiliation(s)
- Joanna Schwenkgrub
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Warsaw, Poland
| | - Malgorzata Zaremba
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Warsaw, Poland
- Laboratory of Magnetic Resonance Imaging of Small Animals, Mossakowski Medical Research Centre, PAS, Warsaw, Poland
- * E-mail:
| | - Ilona Joniec-Maciejak
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Warsaw, Poland
| | - Agnieszka Cudna
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Warsaw, Poland
- 2 Department of Neurology, Institute of Psychiatry and Neurology, Warsaw, Poland
| | - Dagmara Mirowska-Guzel
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Warsaw, Poland
| | | |
Collapse
|
104
|
Mcgrath J, Lintz E, Hoffer BJ, Gerhardt GA, Quintero EM, Granholm AC. Adeno-Associated Viral Delivery of GDNF Promotes Recovery of Dopaminergic Phenotype following a Unilateral 6-Hydroxydopamine Lesion. Cell Transplant 2017. [DOI: 10.3727/096020198389988] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is a potent neurotrophic factor for dopamine neurons that has been proposed for use in the treatment of Parkinson's disease (PD). Previous studies using viral vectors to deliver GDNF in rodent models of PD have entailed administering the virus either prior to or immediately after neurotoxin-induced lesions, when the nigrostriatal pathway is largely intact, a paradigm that does not accurately reflect the clinical situation encountered with Parkinson's patients. In this study, recombinant adeno-associated virus carrying the gene encoding GDNF (rAAV-GDNF) was administered to animals bearing a maximal lesion in the nigrostriatal system, more closely resembling fully developed PD. Rats were treated with 6-hydroxydopamine into the medial forebrain bundle and assessed by apomorphine-induced rotational behavior for 5 weeks prior to virus administration. Within 4 weeks of a single intrastriatal injection of rAAV-GDNF, unilaterally lesioned animals exhibited significant behavioral recovery, which correlated with increased expression of dopaminergic markers in the substantia nigra, the medial forebrain bundle, and the striatum. Our findings demonstrate that rAAV-GDNF is capable of rescuing adult dopaminergic neurons from near complete phenotypic loss following a neurotoxic lesion, effectively restoring a functional dopaminergic pathway and diminishing motoric deficits. These data provide further support for the therapeutic potential of rAAV-GDNF-based gene therapy in the treatment of PD.
Collapse
Affiliation(s)
| | - Elishia Lintz
- University of Colorado Health Science Center, Denver, CO
| | - Barry J. Hoffer
- Intramural Research Program National Institute on Drug Abuse, Baltimore, MD
| | - Greg A. Gerhardt
- Departments of Anatomy & Neurobiology, and Neurology, and the Morris K. Udall Parkinson's Disease Research Center of Excellence, and the Center for Sensor Technology, University of Kentucky, Chandler Medical Center, Lexington, KY
| | - E. Matthew Quintero
- ¶Department of Physiology and Neuroscience, Medical University of South Carolina, Charleston, SC
| | - Ann-Charlotte Granholm
- ¶Department of Physiology and Neuroscience, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
105
|
Yurek D, Hasselrot U, Sesenoglu-Laird O, Padegimas L, Cooper M. Intracerebral injections of DNA nanoparticles encoding for a therapeutic gene provide partial neuroprotection in an animal model of neurodegeneration. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:2209-2217. [PMID: 28666950 DOI: 10.1016/j.nano.2017.06.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/02/2017] [Accepted: 06/14/2017] [Indexed: 11/18/2022]
Abstract
This study reports proof of concept for administering compacted DNA nanoparticles (DNPs) intracerebrally as a means to protect against neurotoxin-induced neurodegeneration of dopamine (DA) neurons. In this study we used DNPs that encoded for human glial cell line-derived neurotrophic factor (hGDNF); GDNF is a potent neurotrophic factor for DA neurons. Intracerebral injections of DNPs into the striatum and/or substantia nigra were performed 1 week before treatment with a neurotoxin. We observed that the number of surviving DA cells, the density of DA fiber terminals and recovery of motor function were greater in animals injected with GDNF-encoding DNPs than in control animals receiving DNPs encoding for an inert reporter gene. The results of these studies are one of the first to demonstrate that a non-viral, synthetic nanoparticle can be used to deliver therapeutic genes to cells in the brain as a means to protect cells against neurotoxin-induced neurodegeneration.
Collapse
Affiliation(s)
- David Yurek
- Department of Neurosurgery, University of Kentucky College of Medicine, Lexington, KY; University of Kentucky Nanobiotechnology Center, Lexington, KY.
| | - Ulla Hasselrot
- Department of Neurosurgery, University of Kentucky College of Medicine, Lexington, KY
| | | | | | - Mark Cooper
- Copernicus Therapeutics, Inc., Cleveland, OH
| |
Collapse
|
106
|
Chiang YH, Borlongan CV, Zhou FC, Hoffer BJ, Wang Y. Transplantation of Fetal Kidney Cells: Neuroprotection and Neuroregeneration. Cell Transplant 2017; 14:1-9. [PMID: 15789657 DOI: 10.3727/000000005783983304] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Various trophic factors in the transforming growth factor-β (TGF-β) superfamily have been reported to have neuroprotective and neuroregenerative effects. Intracerebral administration of glial cell line-derived neurotrophic factor (GDNF) or bone morphogenetic proteins (BMPs), both members of the TGF-β family, reduce ischemia- or 6-hydroxydopamine (6-OHDA)-induced injury in adult rat brain. Because BMPs and GDNF are highly expressed in fetal kidney cells, transplantation of fetal kidney tissue could serve as a cellular reservoir for such molecules and protect against neuronal injury induced by ischemia, neurotoxins, or reactive oxygen species. In this review, we discuss preclinical evidence for the efficacy of fetal kidney cell transplantation in neuroprotection and regeneration models.
Collapse
Affiliation(s)
- Yung-Hsiao Chiang
- Tri-Service General Hospital, National Defense Medical Center, Taiwan
| | | | | | | | | |
Collapse
|
107
|
Granholm AC, Henry S, Herbert MA, Eken S, Gerhardt GA, van Horne C. Kidney Cografts Enhance Fiber Outgrowth from Ventral Mesencephalic Grafts to the 6-Ohda–Lesioned Striatum, and Improve Behavioral Recovery. Cell Transplant 2017; 7:197-212. [PMID: 9588601 DOI: 10.1177/096368979800700214] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Recent studies have demonstrated the presence of many different neurotrophic factors in the developing and adult kidney. Due to its production of this mixture of neurotrophic factors, we wanted to investigate whether fetal kidney tissue could be beneficial for neuritic fiber growth and/or cell survival in intracranial transplants of fetal ventral mesencephalic tissue (VM). A retrograde lesion of nigral dopaminergic neurons was performed in adult Fischer 344 male rats by injecting 6-hydroxydopamine into the medial forebain. The animals were monitored for spontaneous locomotor activity in addition to apomorphine-induced rotations once a week. Four weeks following the lesion, animals were anesthetized and embryonic day 14 VM tissue from rat fetuses was implanted stereotaxically into the dorsal striatum. One group of animals received a cograft of kidney tissue from the same embryos in the same needle track. The animals were then monitored behaviorally for an additional 4 months. There was a significant improvement in both spontaneous locomotor activity (distance traveled) and apomorphine-induced rotations with both single VM grafts and VM–kidney cografts, with the VM–kidney double grafts enhancing the motor behaviors to a significantly greater degree. Tyrosine hydroxylase (TH) immunohistochemistry and image analysis revealed a significantly denser innervation of the host striatum from the VM–kidney cografts than from the single VM grafts. TH-positive neurons were also significantly larger in the cografts compared to the single VM grafts. In addition to the dense TH-immunoreactive innervation, the kidney portion of cografts contained a rich cholinergic innervation, as evidenced from antibodies against choline acetyltransferase (ChAT). The striatal cholinergic cell bodies surrounding the VM–kidney cografts were enlarged and had a slightly higher staining density for ChAT. Taken together, these data support the hypothesis that neurotrophic factors secreted from fetal kidney grafts stimulated both TH-positive neurons in the VM cografts and cholinergic neurons in the host striatum. Thus, these factors may be combined for treatment of degenerative diseases involving both dopaminergic and cholinergic neurons.
Collapse
Affiliation(s)
- A C Granholm
- Department of Basic Science, University of Colorado Health Sciences Center, Denver 80262, USA
| | | | | | | | | | | |
Collapse
|
108
|
Othberg AI, Willing AE, Cameron DF, Anton A, Saporta S, Freeman TB, Sanberg PR. Trophic Effect of Porcine Sertoli Cells on Rat and Human Ventral Mesencephalic Cells and Hnt Neurons in Vitro. Cell Transplant 2017; 7:157-64. [PMID: 9588597 DOI: 10.1177/096368979800700210] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The poor survival of embryonic dopaminergic (DA) neurons transplanted into patients with Parkinson's disease (PD) has encouraged researchers to search for new methods to affect the short- as well as long-term survival of these neurons after transplantation. In several previous rodent studies Sertoli cells increased survival of islet cells and chromaffin cells when cotransplanted in vivo. The aims of this study were to investigate whether porcine Sertoli cells had a positive effect on the survival and maturation of rat and human DA neurons, and whether the Sertoli cells had an effect on differentiation of neurons derived from a human teratocarcinoma cell line (hNT neurons). A significant increase of tyrosine hydroxylase (TH)-positive neurons of both rat and human ventral mesencephalic tissue was found when cocultured with Sertoli cells. Furthermore, there was a significantly increased soma size and neurite outgrowth of neurons in the coculture treated group. The Sertoli cell and hNT coculture also revealed an increased number of TH-positive cells. These results demonstrate that the wide variety of proteins and factors secreted by porcine Sertoli cells benefit the survival and maturation of embryonic DA neurons and suggest that cotransplantation of Sertoli cells and embryonic DA neurons may be useful for a cell transplantation therapy in PD.
Collapse
Affiliation(s)
- A I Othberg
- Department of Surgery, University of South Florida, College of Medicine, Tampa 33612, USA
| | | | | | | | | | | | | |
Collapse
|
109
|
Belova E, Shaffer CL, Trapa PE. Insights from mathematical modeling for convection-enhanced intraputamenal delivery of GDNF. Med Biol Eng Comput 2017; 55:2069-2077. [PMID: 28493093 PMCID: PMC5680405 DOI: 10.1007/s11517-017-1650-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 04/18/2017] [Indexed: 11/23/2022]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is a potential therapy for Parkinson’s disease (PD) promoting survival and functional recovery of dopaminergic neurons when delivered to the degenerated striatum. To study the aspects of intraputamenal delivery of GDNF, a mathematical model of recombinant methionyl human GDNF (r-metHuGDNF) convection in the human putamen has been developed. The convection-enhanced delivery infusions of r-metHuGDNF were simulated at rates up to 5 μL/min. The high-rate infusions (≥1 μL/min) permit rapid and uniform distribution of drug with up to 75% of the distribution volume having a concentration within 5% of the infusate concentration. No relevant differences in distribution at infusion rates of 3 and 5 μL/min were found. The patterns of GDNF distribution were analyzed in relation to the anatomy of the posterior dorsal putamen, and a cylindrical shape was found to be preferable considering risks of target overflow. A magnetic resonance (MR) tracer Gd-DTPA (Magnevist®) was evaluated as a surrogate in clinical studies, and the most accurate prediction of GDNF distribution was calculated immediately after infusion. The clearance of GDNF from the striatum is confirmed to be slow, with a half-life of ca. 19 h.
Collapse
Affiliation(s)
- Elena Belova
- Worldwide Research & Development, Pfizer Inc., 1 Portland Street, Cambridge, MA, 02139, USA.
| | - Christopher L Shaffer
- Worldwide Research & Development, Pfizer Inc., 1 Portland Street, Cambridge, MA, 02139, USA
| | - Patrick E Trapa
- Worldwide Research & Development, Pfizer Inc., 1 Portland Street, Cambridge, MA, 02139, USA
| |
Collapse
|
110
|
Non-human primate models of PD to test novel therapies. J Neural Transm (Vienna) 2017; 125:291-324. [PMID: 28391443 DOI: 10.1007/s00702-017-1722-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 04/04/2017] [Indexed: 12/13/2022]
Abstract
Non-human primate (NHP) models of Parkinson disease show many similarities with the human disease. They are very useful to test novel pharmacotherapies as reviewed here. The various NHP models of this disease are described with their characteristics including the macaque, the marmoset, and the squirrel monkey models. Lesion-induced and genetic models are described. There is no drug to slow, delay, stop, or cure Parkinson disease; available treatments are symptomatic. The dopamine precursor, L-3,4-dihydroxyphenylalanine (L-Dopa) still remains the gold standard symptomatic treatment of Parkinson. However, involuntary movements termed L-Dopa-induced dyskinesias appear in most patients after chronic treatment and may become disabling. Dyskinesias are very difficult to manage and there is only amantadine approved providing only a modest benefit. In this respect, NHP models have been useful to seek new drug targets, since they reproduce motor complications observed in parkinsonian patients. Therapies to treat motor symptoms in NHP models are reviewed with a discussion of their translational value to humans. Disease-modifying treatments tested in NHP are reviewed as well as surgical treatments. Many biochemical changes in the brain of post-mortem Parkinson disease patients with dyskinesias are reviewed and compare well with those observed in NHP models. Non-motor symptoms can be categorized into psychiatric, autonomic, and sensory symptoms. These symptoms are present in most parkinsonian patients and are already installed many years before the pre-motor phase of the disease. The translational usefulness of NHP models of Parkinson is discussed for non-motor symptoms.
Collapse
|
111
|
Zuo F, Xiong F, Wang X, Li X, Wang R, Ge W, Bao X. Intrastriatal Transplantation of Human Neural Stem Cells Restores the Impaired Subventricular Zone in Parkinsonian Mice. Stem Cells 2017; 35:1519-1531. [PMID: 28328168 DOI: 10.1002/stem.2616] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 02/17/2017] [Accepted: 02/26/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Fuxing Zuo
- Department of Neurosurgery; Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing China
| | - Feng Xiong
- State Key Laboratory of Medical Molecular Biology & Department of Immunology; Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; Beijing China
| | - Xia Wang
- State Key Laboratory of Medical Molecular Biology & Department of Immunology; Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; Beijing China
| | - Xueyuan Li
- Department of Neurosurgery; Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing China
| | - Renzhi Wang
- Department of Neurosurgery; Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing China
| | - Wei Ge
- State Key Laboratory of Medical Molecular Biology & Department of Immunology; Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; Beijing China
| | - Xinjie Bao
- Department of Neurosurgery; Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing China
| |
Collapse
|
112
|
Hovland DN, Boyd RB, Butt MT, Engelhardt JA, Moxness MS, Ma MH, Emery MG, Ernst NB, Reed RP, Zeller JR, Gash DM, Masterman DM, Potter BM, Cosenza ME, Lightfoot RM. Six-Month Continuous Intraputamenal Infusion Toxicity Study of Recombinant Methionyl Human Glial Cell Line-Derived Neurotrophic Factor (r-metHuGDNF) in Rhesus Monkeys. Toxicol Pathol 2017. [DOI: 10.1177/01926230701481899a] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Recombinant human glial cell line-derived neurotrophic factor (r-metHuGDNF) is a potent neuronal growth and survival factor that has been considered for clinical use in the treatment of Parkinson’s disease (PD). Here we present results of a 6-month toxicology study in rhesus monkeys conducted to support clinical evaluation of chronic intraputamenal infusion of r-metHuGDNF for PD. Monkeys (6–9/sex/group) were treated with 0 (vehicle), 15, 30, or 100 μg/day r-metHuGDNF by continuous unilateral intraputamenal infusion (150 μl/day flow rate) for 6 months; a subset of animals (2–3/sex/group) underwent a subsequent 3-month treatment-free recovery period. Notable observations included reduced food consumption and body weight at 100 μg/day and meningeal thickening underlying the medulla oblongata and/or overlying various spinal cord segments at 30 and 100 μg/day. In addition, multifocal cerebellar Purkinje cell loss (with associated atrophy of the molecular layer and, in some cases, granule cell loss) was observed in 4 monkeys in the 100-μg/day group. This cerebellar finding has not been observed in previous nonclinical studies evaluating r-metHuGDNF. The small number of affected animals precludes definitive conclusions regarding the pathogenesis of the cerebellar lesion, but the data support an association with r-metHuGDNF treatment.
Collapse
Affiliation(s)
| | - Robert B. Boyd
- Northern Biomedical Research, Inc., Muskegon, Michigan 49441
| | - Mark T. Butt
- Pathology Associates, Charles River Laboratories, Frederick, Maryland 21701
| | | | | | - Mark H. Ma
- Amgen Inc., Thousand Oaks, California 91320
| | | | | | - Randall P. Reed
- Northern Biomedical Research, Inc., Muskegon, Michigan 49441
| | | | - Don M. Gash
- Anatomy and Neurobiology, University of Kentucky, Lexington, Kentucky 40536
| | | | | | | | | |
Collapse
|
113
|
Pignataro D, Sucunza D, Rico AJ, Dopeso-Reyes IG, Roda E, Rodríguez-Perez AI, Labandeira-Garcia JL, Broccoli V, Kato S, Kobayashi K, Lanciego JL. Gene therapy approaches in the non-human primate model of Parkinson's disease. J Neural Transm (Vienna) 2017; 125:575-589. [PMID: 28130586 DOI: 10.1007/s00702-017-1681-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 01/16/2017] [Indexed: 12/23/2022]
Abstract
The field of gene therapy has recently witnessed a number of major conceptual changes. Besides the traditional thinking that comprises the use of viral vectors for the delivery of a given therapeutic gene, a number of original approaches have been recently envisaged, focused on using vectors carrying genes to further modify basal ganglia circuits of interest. It is expected that these approaches will ultimately induce a therapeutic potential being sustained by gene-induced changes in brain circuits. Among others, at present, it is technically feasible to use viral vectors to (1) achieve a controlled release of neurotrophic factors, (2) conduct either a transient or permanent silencing of any given basal ganglia circuit of interest, (3) perform an in vivo cellular reprogramming by promoting the conversion of resident cells into dopaminergic-like neurons, and (4) improving levodopa efficacy over time by targeting aromatic L-amino acid decarboxylase. Furthermore, extensive research efforts based on viral vectors are currently ongoing in an attempt to better replicate the dopaminergic neurodegeneration phenomena inherent to the progressive intraneuronal aggregation of alpha-synuclein. Finally, a number of incoming strategies will soon emerge over the horizon, these being sustained by the underlying goal of promoting alpha-synuclein clearance, such as, for instance, gene therapy initiatives based on increasing the activity of glucocerebrosidase. To provide adequate proof-of-concept on safety and efficacy and to push forward true translational initiatives based on these different types of gene therapies before entering into clinical trials, the use of non-human primate models undoubtedly plays an instrumental role.
Collapse
Affiliation(s)
- D Pignataro
- Department of Neurosciences, Center for Applied Medical Research (CIMA), University of Navarra, Pio XII Ave 55, Edificio CIMA, 31008, Pamplona, Navarra, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - D Sucunza
- Department of Neurosciences, Center for Applied Medical Research (CIMA), University of Navarra, Pio XII Ave 55, Edificio CIMA, 31008, Pamplona, Navarra, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - A J Rico
- Department of Neurosciences, Center for Applied Medical Research (CIMA), University of Navarra, Pio XII Ave 55, Edificio CIMA, 31008, Pamplona, Navarra, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - I G Dopeso-Reyes
- Department of Neurosciences, Center for Applied Medical Research (CIMA), University of Navarra, Pio XII Ave 55, Edificio CIMA, 31008, Pamplona, Navarra, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - E Roda
- Department of Neurosciences, Center for Applied Medical Research (CIMA), University of Navarra, Pio XII Ave 55, Edificio CIMA, 31008, Pamplona, Navarra, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - A I Rodríguez-Perez
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - J L Labandeira-Garcia
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - V Broccoli
- Division of Neuroscience, Ospedale San Raffaele, 20132, Milan, Italy
- CNR Institute of Neuroscience, 20129, Milan, Italy
| | - S Kato
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - K Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - José L Lanciego
- Department of Neurosciences, Center for Applied Medical Research (CIMA), University of Navarra, Pio XII Ave 55, Edificio CIMA, 31008, Pamplona, Navarra, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.
| |
Collapse
|
114
|
Ibáñez CF, Andressoo JO. Biology of GDNF and its receptors — Relevance for disorders of the central nervous system. Neurobiol Dis 2017; 97:80-89. [DOI: 10.1016/j.nbd.2016.01.021] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 01/14/2016] [Accepted: 01/25/2016] [Indexed: 01/15/2023] Open
|
115
|
Jia Y, Deng J, Zhang W, Sun Z, Yang J, Yu Y, Gong X, Jia J, Wang X. The Role of Group II Metabotropic Glutamate Receptors in the Striatum in Electroacupuncture Treatment of Parkinsonian Rats. CNS Neurosci Ther 2017; 23:23-32. [PMID: 27412260 PMCID: PMC6492692 DOI: 10.1111/cns.12587] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 06/11/2016] [Accepted: 06/12/2016] [Indexed: 12/21/2022] Open
Abstract
AIMS Glutamatergic transmission may play a critical role in the pathogenesis of Parkinson's disease (PD). Electroacupuncture (EA) has been demonstrated to effectively alleviate PD symptoms. In this study, a potential glutamate-dependent mechanism underlying the therapeutic action of EA was investigated. METHODS The effects of EA stimulation on motor behaviors, dopamine contents, glutamate release, and group II metabotropic glutamate receptor (mGluR2/3) expression in unilateral 6-hydroxydopamine (6-OHDA)-lesioned rats were examined. RESULTS Unilateral 6-OHDA lesions of the nigrostriatal system caused a marked increase in glutamate content in the ipsilateral cortex and striatum. mGluR2/3 protein expression and mGluR3 mRNA expression were reduced in the striatum. Noticeably, prolonged EA stimulation at 100 Hz significantly reversed these changes in the striatal glutamate system. Behaviorally, EA improved the motor deficits induced by 6-OHDA lesions. Intrastriatal infusion of an mGluR2/3 antagonist APICA blocked the improving effect of EA. CONCLUSIONS These data collectively demonstrate that the group II mGluR-mediated glutamatergic transmission in the striatum is sensitive to dopamine depletion and may serve as a substrate of EA for mediating the therapeutic effect of EA in a rat model of PD.
Collapse
Affiliation(s)
- Yan‐Jun Jia
- Departments of Neurobiology and PhysiologyKey Laboratory for Neurodegenerative Disorders of the Ministry of EducationBeijing Key Laboratory for Parkinson's DiseaseCapital Medical UniversityBeijing Institute for Brain DisordersBeijingChina
| | - Jia‐Hui Deng
- Departments of Neurobiology and PhysiologyKey Laboratory for Neurodegenerative Disorders of the Ministry of EducationBeijing Key Laboratory for Parkinson's DiseaseCapital Medical UniversityBeijing Institute for Brain DisordersBeijingChina
| | - Wen‐Zhong Zhang
- Departments of Neurobiology and PhysiologyKey Laboratory for Neurodegenerative Disorders of the Ministry of EducationBeijing Key Laboratory for Parkinson's DiseaseCapital Medical UniversityBeijing Institute for Brain DisordersBeijingChina
| | - Zuo‐Li Sun
- Departments of Neurobiology and PhysiologyKey Laboratory for Neurodegenerative Disorders of the Ministry of EducationBeijing Key Laboratory for Parkinson's DiseaseCapital Medical UniversityBeijing Institute for Brain DisordersBeijingChina
| | - Jian Yang
- Departments of Neurobiology and PhysiologyKey Laboratory for Neurodegenerative Disorders of the Ministry of EducationBeijing Key Laboratory for Parkinson's DiseaseCapital Medical UniversityBeijing Institute for Brain DisordersBeijingChina
| | - Yan Yu
- Departments of Neurobiology and PhysiologyKey Laboratory for Neurodegenerative Disorders of the Ministry of EducationBeijing Key Laboratory for Parkinson's DiseaseCapital Medical UniversityBeijing Institute for Brain DisordersBeijingChina
| | - Xiao‐Li Gong
- Departments of Neurobiology and PhysiologyKey Laboratory for Neurodegenerative Disorders of the Ministry of EducationBeijing Key Laboratory for Parkinson's DiseaseCapital Medical UniversityBeijing Institute for Brain DisordersBeijingChina
| | - Jun Jia
- Departments of Neurobiology and PhysiologyKey Laboratory for Neurodegenerative Disorders of the Ministry of EducationBeijing Key Laboratory for Parkinson's DiseaseCapital Medical UniversityBeijing Institute for Brain DisordersBeijingChina
| | - Xiao‐Min Wang
- Departments of Neurobiology and PhysiologyKey Laboratory for Neurodegenerative Disorders of the Ministry of EducationBeijing Key Laboratory for Parkinson's DiseaseCapital Medical UniversityBeijing Institute for Brain DisordersBeijingChina
| |
Collapse
|
116
|
Kaur G, Baino F, Mauro JC, Kumar V, Pickrell G, Sriranganathan N, Waldrop SG. Biomaterials for Cell Encapsulation: Progress Toward Clinical Applications. CLINICAL APPLICATIONS OF BIOMATERIALS 2017:425-458. [DOI: 10.1007/978-3-319-56059-5_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
117
|
Kirik D, Cederfjäll E, Halliday G, Petersén Å. Gene therapy for Parkinson's disease: Disease modification by GDNF family of ligands. Neurobiol Dis 2017; 97:179-188. [DOI: 10.1016/j.nbd.2016.09.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 08/24/2016] [Accepted: 09/06/2016] [Indexed: 10/21/2022] Open
|
118
|
Garbayo E, Ansorena E, Lana H, Carmona-Abellan MDM, Marcilla I, Lanciego JL, Luquin MR, Blanco-Prieto MJ. Brain delivery of microencapsulated GDNF induces functional and structural recovery in parkinsonian monkeys. Biomaterials 2016; 110:11-23. [DOI: 10.1016/j.biomaterials.2016.09.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 09/19/2016] [Accepted: 09/21/2016] [Indexed: 01/03/2023]
|
119
|
Bezdjian A, Kraaijenga VJC, Ramekers D, Versnel H, Thomeer HGXM, Klis SFL, Grolman W. Towards Clinical Application of Neurotrophic Factors to the Auditory Nerve; Assessment of Safety and Efficacy by a Systematic Review of Neurotrophic Treatments in Humans. Int J Mol Sci 2016; 17:ijms17121981. [PMID: 27898033 PMCID: PMC5187781 DOI: 10.3390/ijms17121981] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/11/2016] [Accepted: 11/21/2016] [Indexed: 01/31/2023] Open
Abstract
Animal studies have evidenced protection of the auditory nerve by exogenous neurotrophic factors. In order to assess clinical applicability of neurotrophic treatment of the auditory nerve, the safety and efficacy of neurotrophic therapies in various human disorders were systematically reviewed. Outcomes of our literature search included disorder, neurotrophic factor, administration route, therapeutic outcome, and adverse event. From 2103 articles retrieved, 20 randomized controlled trials including 3974 patients were selected. Amyotrophic lateral sclerosis (53%) was the most frequently reported indication for neurotrophic therapy followed by diabetic polyneuropathy (28%). Ciliary neurotrophic factor (50%), nerve growth factor (24%) and insulin-like growth factor (21%) were most often used. Injection site reaction was a frequently occurring adverse event (61%) followed by asthenia (24%) and gastrointestinal disturbances (20%). Eighteen out of 20 trials deemed neurotrophic therapy to be safe, and six out of 17 studies concluded the neurotrophic therapy to be effective. Positive outcomes were generally small or contradicted by other studies. Most non-neurodegenerative diseases treated by targeted deliveries of neurotrophic factors were considered safe and effective. Hence, since local delivery to the cochlea is feasible, translation from animal studies to human trials in treating auditory nerve degeneration seems promising.
Collapse
Affiliation(s)
- Aren Bezdjian
- Department of Otorhinolaryngology and Head & Neck Surgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands.
| | - Véronique J C Kraaijenga
- Department of Otorhinolaryngology and Head & Neck Surgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands.
| | - Dyan Ramekers
- Department of Otorhinolaryngology and Head & Neck Surgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands.
| | - Huib Versnel
- Department of Otorhinolaryngology and Head & Neck Surgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands.
| | - Hans G X M Thomeer
- Department of Otorhinolaryngology and Head & Neck Surgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands.
| | - Sjaak F L Klis
- Department of Otorhinolaryngology and Head & Neck Surgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands.
| | - Wilko Grolman
- Department of Otorhinolaryngology and Head & Neck Surgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands.
| |
Collapse
|
120
|
Pramanik S, Sulistio YA, Heese K. Neurotrophin Signaling and Stem Cells-Implications for Neurodegenerative Diseases and Stem Cell Therapy. Mol Neurobiol 2016; 54:7401-7459. [PMID: 27815842 DOI: 10.1007/s12035-016-0214-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 10/11/2016] [Indexed: 02/07/2023]
Abstract
Neurotrophins (NTs) are members of a neuronal growth factor protein family whose action is mediated by the tropomyosin receptor kinase (TRK) receptor family receptors and the p75 NT receptor (p75NTR), a member of the tumor necrosis factor (TNF) receptor family. Although NTs were first discovered in neurons, recent studies have suggested that NTs and their receptors are expressed in various types of stem cells mediating pivotal signaling events in stem cell biology. The concept of stem cell therapy has already attracted much attention as a potential strategy for the treatment of neurodegenerative diseases (NDs). Strikingly, NTs, proNTs, and their receptors are gaining interest as key regulators of stem cells differentiation, survival, self-renewal, plasticity, and migration. In this review, we elaborate the recent progress in understanding of NTs and their action on various stem cells. First, we provide current knowledge of NTs, proNTs, and their receptor isoforms and signaling pathways. Subsequently, we describe recent advances in the understanding of NT activities in various stem cells and their role in NDs, particularly Alzheimer's disease (AD) and Parkinson's disease (PD). Finally, we compile the implications of NTs and stem cells from a clinical perspective and discuss the challenges with regard to transplantation therapy for treatment of AD and PD.
Collapse
Affiliation(s)
- Subrata Pramanik
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 133-791, Republic of Korea
| | - Yanuar Alan Sulistio
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 133-791, Republic of Korea
| | - Klaus Heese
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 133-791, Republic of Korea.
| |
Collapse
|
121
|
Olanow CW, Bartus RT, Volpicelli-Daley LA, Kordower JH. Trophic factors for Parkinson's disease: To live or let die. Mov Disord 2016; 30:1715-24. [PMID: 26769457 DOI: 10.1002/mds.26426] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 08/06/2015] [Accepted: 08/19/2015] [Indexed: 12/23/2022] Open
Abstract
Trophic factors show great promise in laboratory studies as potential therapies for PD. However, multiple double-blind, clinical trials have failed to show benefits in comparison to a placebo control. This article will review the scientific rationale for testing trophic factors in PD, the results of the different clinical trials that have been performed to date, and the possible explanations for these failed outcomes. We will also consider future directions and the likelihood that trophic factors will become a viable therapy for patients with PD.
Collapse
Affiliation(s)
- C Warren Olanow
- Department of Neurology, Mount Sinai School of Medicine, New York, New York, USA
| | | | | | | |
Collapse
|
122
|
Li W, Tong HI, Gorantla S, Poluektova LY, Gendelman HE, Lu Y. Neuropharmacologic Approaches to Restore the Brain's Microenvironment. J Neuroimmune Pharmacol 2016; 11:484-94. [PMID: 27352074 PMCID: PMC4985494 DOI: 10.1007/s11481-016-9686-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 05/25/2016] [Indexed: 12/15/2022]
Abstract
Maintaining the central nervous system microenvironment after injury, infection, inflammatory and degenerative diseases is contingent upon adequate control of glial homeostatic functions. Disease is caused by microbial, environmental and endogenous factors that compromise ongoing nervous system functions. The final result is neuronal injury, dropout and nerve connection loss, and these underlie the pathobiology of Alzheimer's and Parkinson's disease, amyotrophic lateral sclerosis, stroke, and bacterial, parasitic and viral infections. However, what promotes disease are homeostatic changes in the brain's microenvironment affected by innate glial immune pro-inflammatory and adaptive immune responses. These events disturb the brain's metabolic activities and communication abilities. How the process affects the brain's regulatory functions that can be harnessed for therapeutic gain is the subject at hand. Specific examples are provided that serve to modulate inflammation and improve disease outcomes specifically for HIV-associated neurocognitive disorders.
Collapse
Affiliation(s)
- Weizhe Li
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Hsin-I Tong
- Department of Public Health Sciences, Environmental Health Laboratory, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Santhi Gorantla
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Larisa Y Poluektova
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Yuanan Lu
- Department of Public Health Sciences, Environmental Health Laboratory, University of Hawaii at Manoa, Honolulu, HI, 96822, USA.
| |
Collapse
|
123
|
Sidorova YA, Saarma M. Glial cell line-derived neurotrophic factor family ligands and their therapeutic potential. Mol Biol 2016. [DOI: 10.1134/s0026893316040105] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
124
|
Newland B, Dunnett SB, Dowd E. Targeting delivery in Parkinson's disease. Drug Discov Today 2016; 21:1313-20. [DOI: 10.1016/j.drudis.2016.06.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 05/23/2016] [Accepted: 06/06/2016] [Indexed: 01/24/2023]
|
125
|
Lin CY, Hsieh HY, Chen CM, Wu SR, Tsai CH, Huang CY, Hua MY, Wei KC, Yeh CK, Liu HL. Non-invasive, neuron-specific gene therapy by focused ultrasound-induced blood-brain barrier opening in Parkinson's disease mouse model. J Control Release 2016; 235:72-81. [DOI: 10.1016/j.jconrel.2016.05.052] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 04/28/2016] [Accepted: 05/24/2016] [Indexed: 02/01/2023]
|
126
|
Wang Y, Yang H, Yang Q, Yang J, Wang H, Xu H, Gao WQ. Chemical conversion of mouse fibroblasts into functional dopaminergic neurons. Exp Cell Res 2016; 347:283-92. [PMID: 27485858 DOI: 10.1016/j.yexcr.2016.07.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 07/26/2016] [Accepted: 07/29/2016] [Indexed: 01/03/2023]
Abstract
Ectopic expression of lineage-specific transcription factors facilitates the conversion of mammalian somatic cells into dopaminergic (DA) neurons, which is a promising strategy for cell therapy of Parkinson's disease (PD). However, this approach still has some drawbacks limiting its clinical application due to the potential risks of integrating vectors into the host genome. Therefore, it is critical to seek a more desired approach to generate DA neurons derived from mammalian somatic cells. Here, we report that mouse embryonic fibroblasts (MEFs) can be efficiently converted into DA neurons by using small molecules along with specific growth factors. These neuron-like cells generate DA neuronal morphology, and acquire immunocytochemical and calcium imaging special for neuronal electrophysiological profile. More importantly, these converted cells can secrete dopamine, indicating that they are functionally similar to DA neurons. Taken together, our study might provide a promising cell source for treating PD by using chemical approach without introduction of exogenous transcription factors.
Collapse
Affiliation(s)
- Yonghui Wang
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Hao Yang
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Qiong Yang
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Junhua Yang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology (Ministry of Health of China), Key Laboratory of Neurobiology of Zhejiang Province, Zhejiang School of Medicine, Hangzhou, China
| | - Hao Wang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology (Ministry of Health of China), Key Laboratory of Neurobiology of Zhejiang Province, Zhejiang School of Medicine, Hangzhou, China
| | - Huiming Xu
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China.
| | - Wei-Qiang Gao
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China; Collaborative Innovative Research Center for Systems Biomedicine, Shanghai, China.
| |
Collapse
|
127
|
Abstract
Parkinson disease (PD) is one of the most widespread neurodegenerative disorders. In North America alone it affects 1 million people. It is a multifactorial disorder caused by genetic, various biological and environmental factors. One of the important features of PD is the dementia, which is believed to be due to the loss of dopaminergic neurons. In some cases the disease can be inherited as an autosomal dominant or recessive trait but in the majority of cases it is acquired. The biological causes of the disorder are unknown. The identification of mutations in the parkin gene in the autosomal recessive case and alpha-synuclein gene in autosomal dominant cases has opened a new avenue for studies to understand the basic biochemical mechanisms of pathogenesis. Although several types of treatments such as transplantation of cells that produce L-Dopa and direct gene delivery using adeno-associated viral vectors may correct animal models of PD, their usefulness in the human is not yet clear. A better understanding of the causes of neurodegeneration may lead to better therapies in the future.
Collapse
|
128
|
Hovland DN, Boyd RB, Butt MT, Engelhardt JA, Moxness MS, Ma MH, Emery MG, Ernst NB, Reed RP, Zeller JR, Gash DM, Masterman DM, Potter BM, Cosenza ME, Lightfoot RM. Reprint: Six-Month Continuous Intraputamenal Infusion Toxicity Study of Recombinant Methionyl Human Glial Cell Line-Derived Neurotrophic Factor (r-metHuGDNF) in Rhesus Monkeys. Toxicol Pathol 2016; 35:1013-29. [PMID: 18098052 DOI: 10.1177/01926230701481899] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Recombinant human glial cell line-derived neurotrophic factor (r-metHuGDNF) is a potent neuronal growth and survival factor that has been considered for clinical use in the treatment of Parkinson’s disease (PD). Here we present results of a 6-month toxicology study in rhesus monkeys conducted to support clinical evaluation of chronic intraputamenal infusion of r-metHuGDNF for PD. Monkeys (6–9/sex/group) were treated with 0 (vehicle), 15, 30, or 100 μg/day r-metHuGDNF by continuous unilateral intraputamenal infusion (150 μl/day flow rate) for 6 months; a subset of animals (2–3/sex/group) underwent a subsequent 3-month treatment-free recovery period. Notable observations included reduced food consumption and body weight at 100 μg/day and meningeal thickening underlying the medulla oblongata and/or overlying various spinal cord segments at 30 and 100 μg/day. In addition, multifocal cerebellar Purkinje cell loss (with associated atrophy of the molecular layer and, in some cases, granule cell loss) was observed in 4 monkeys in the 100-μg/day group. This cerebellar finding has not been observed in previous nonclinical studies evaluating r-metHuGDNF. The small number of affected animals precludes definitive conclusions regarding the pathogenesis of the cerebellar lesion, but the data support an association with r-metHuGDNF treatment.
Collapse
Affiliation(s)
| | - Robert B. Boyd
- Northern Biomedical Research, Inc., Muskegon, Michigan 49441
| | - Mark T. Butt
- Pathology Associates, Charles River Laboratories, Frederick, Maryland 21701
| | | | | | - Mark H. Ma
- Amgen Inc., Thousand Oaks, California 91320
| | | | | | - Randall P. Reed
- Northern Biomedical Research, Inc., Muskegon, Michigan 49441
| | | | - Don M. Gash
- Anatomy and Neurobiology, University of Kentucky, Lexington, Kentucky 40536
| | | | | | | | | |
Collapse
|
129
|
Voigt A, Berlemann LA, Winklhofer KF. The mitochondrial kinase PINK1: functions beyond mitophagy. J Neurochem 2016; 139 Suppl 1:232-239. [PMID: 27251035 DOI: 10.1111/jnc.13655] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 04/05/2016] [Accepted: 04/22/2016] [Indexed: 11/29/2022]
Abstract
Mutations in the genes encoding the mitochondrial kinase PINK1 and the E3 ubiquitin ligase Parkin cause autosomal recessive Parkinson's disease (PD). Pioneering work in Drosophila melanogaster revealed that the loss of PINK1 or Parkin function causes similar phenotypes including dysfunctional mitochondria. Further research showed that PINK1 can act upstream of Parkin in a mitochondrial quality control pathway to induce removal of damaged mitochondria in a process called mitophagy. Albeit the PINK1/Parkin-induced mitophagy pathway is well established and has recently been elucidated in great detail, its pathophysiological relevance is being debated. Mounting evidence indicates that PINK1 has additional functions, for example, in regulating complex I activity and maintaining neuronal viability in response to stress. Here, we discuss mitophagy-dependent and -independent functions of PINK1 and their possible role in PD pathogenesis. Mutations in the PINK1 gene, encoding a mitochondrial kinase, are associated with autosomal recessive Parkinson's disease. In this review, we summarize and discuss the functional roles of PINK1 in maintaining mitochondrial integrity, eliminating damaged mitochondria, and promoting cell survival. This article is part of a special issue on Parkinson disease.
Collapse
Affiliation(s)
- Aaron Voigt
- Department of Neurology, University Hospital, RWTH Aachen University, Aachen, Germany.
| | - Lena A Berlemann
- Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Germany
| | - Konstanze F Winklhofer
- Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Germany.
| |
Collapse
|
130
|
Jaumotte JD, Wyrostek SL, Zigmond MJ. Protection of cultured dopamine neurons from MPP(+) requires a combination of neurotrophic factors. Eur J Neurosci 2016; 44:1691-9. [PMID: 27098376 DOI: 10.1111/ejn.13252] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 03/15/2016] [Accepted: 04/01/2016] [Indexed: 11/30/2022]
Abstract
Parkinson's disease is a progressive neurodegenerative disorder, caused in part by the loss of dopamine (DA) neurons in the substantia nigra (SN). Neurotrophic factors have been shown to increase the basal survival of DA neurons in vitro, as well as to protect the neurons from some toxins under certain in vitro conditions and in animal models. Although these factors have often been tested individually, they have rarely been studied in combinations. We therefore examined the effect of such combinations after acute exposure to the toxin 1-methyl-4-phenylpyridinium (MPP(+) ) using dissociated postnatal rat midbrain cultures isolated from SN and ventral tegmental area (VTA). We found that significant loss of DA neurons in the SN occurred with an LC50 of between 1 and 10 μm, whereas the LC50 of DA neurons from the VTA was approximately 1000-fold higher. We did not observe neuroprotection against MPP(+) by individual exposure to glial cell-line derived neurotrophic factor (GDNF), brain derived neurotrophic factor (BDNF), transforming growth factor beta (TGFβ), basic fibroblast growth factor (FGF-2) or growth/differentiation factor 5 (GDF5) at concentrations of 100 or 500 ng/mL. Combinations of two, three or four neurotrophic factors were also ineffective. However, when the SN cultures were exposed to a combination of all five neurotrophic factors, each at a concentration of 100 ng/mL, we observed a 30% increase in DA neuron survival in the presence of 10 and 500 μm MPP(+) . These results may be relevant to the use of neurotrophic factors as therapeutic treatments for Parkinson's disease.
Collapse
Affiliation(s)
- Juliann D Jaumotte
- Pittsburgh Institute for Neurodegenerative Disease, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Stephanie L Wyrostek
- Pittsburgh Institute for Neurodegenerative Disease, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Michael J Zigmond
- Pittsburgh Institute for Neurodegenerative Disease, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
131
|
Xu H, Wang Y, He Z, Yang H, Gao WQ. Direct conversion of mouse fibroblasts to GABAergic neurons with combined medium without the introduction of transcription factors or miRNAs. Cell Cycle 2016; 14:2451-60. [PMID: 26114472 DOI: 10.1080/15384101.2015.1060382] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Degeneration or loss of GABAergic neurons frequently may lead to many neuropsychiatric disorders such as epilepsy and autism spectrum disorders. So far no clinically effective therapies can slow and halt the progression of these diseases. Cell-replacement therapy is a promising strategy for treatment of these neuropsychiatric diseases. Although increasing evidence showed that mammalian somatic cells can be directly converted into functional neurons using specific transcription factors or miRNAs via virus delivery, the application of these induced neurons is potentially problematic, due to integration of vectors into the host genome, which results in the disruption or dysfunction of nearby genes. Here, we show that mouse fibroblasts could be efficiently reprogrammed into GABAergic neurons in a combined medium composed of conditioned medium from neurotrophin-3 modified Olfactory Ensheathing Cells (NT3-OECs) plus SB431542, GDNF and RA. Following 3 weeks of induction, these cells derived from fibroblasts acquired the morphological and phenotypical GABAerigic neuronal properties, as demonstrated by the expression of neuronal markers including Tuj1, NeuN, Neurofilament-L, GABA, GABA receptors and GABA transporter 1. More importantly, these converted cells acquired neuronal functional properties such as synapse formation and increasing intracellular free calcium influx when treated with BayK, a specific activator of L-type calcium channel. Therefore, our findings demonstrate for the first time that fibroblasts can be directly converted into GABAergic neurons without ectopic expression of specific transcription factors or miRNA. This study may provide a promising cell source for the application of cell replacement therapy in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Huiming Xu
- a State Key Laboratory of Oncogenes and Related Genes; Renji-MedX Clinical Stem Cell Research Center; Ren Ji Hospital; School of Medicine; Shanghai Jiao Tong University ; Shangha , China
| | | | | | | | | |
Collapse
|
132
|
van Horne CG, Quintero JE, Gurwell JA, Wagner RP, Slevin JT, Gerhardt GA. Implantation of autologous peripheral nerve grafts into the substantia nigra of subjects with idiopathic Parkinson's disease treated with bilateral STN DBS: a report of safety and feasibility. J Neurosurg 2016; 126:1140-1147. [PMID: 27153166 DOI: 10.3171/2016.2.jns151988] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE One avenue of intense efforts to treat Parkinson's disease (PD) involves the delivery of neurotrophic factors to restore dopaminergic cell function. A source of neurotrophic factors that could be used is the Schwann cell from the peripheral nervous system. The authors have begun an open-label safety study to examine the safety and feasibility of implanting an autologous peripheral nerve graft into the substantia nigra of PD patients undergoing deep brain stimulation (DBS) surgery. METHODS Multistage DBS surgery targeting the subthalamic nucleus was performed using standard procedures in 8 study participants. After the DBS leads were implanted, a section of sural nerve containing Schwann cells was excised and unilaterally delivered into the area of the substantia nigra. Adverse events were continuously monitored. RESULTS Eight of 8 participants were implanted with DBS systems and grafts. Adverse event profiles were comparable to those of standard DBS surgery. Postoperative MR images did not reveal edema, hemorrhage, or significant signal changes in the graft target region. Three participants reported a patch of numbness on the outside of the foot below the sural nerve harvest site. CONCLUSIONS Based on the safety outcome of the procedure, targeted peripheral nerve graft delivery to the substantia nigra at the time of DBS surgery is feasible and may provide a means to deliver neurorestorative therapy. Clinical trial registration no.: NCT01833364 ( clinicaltrials.gov ).
Collapse
Affiliation(s)
- Craig G van Horne
- Brain Restoration Center, and.,Departments of 2 Neurosurgery.,Anatomy & Neurobiology, and
| | | | - Julie A Gurwell
- Brain Restoration Center, and.,Neurology, University of Kentucky, Lexington, Kentucky
| | | | - John T Slevin
- Brain Restoration Center, and.,Neurology, University of Kentucky, Lexington, Kentucky
| | - Greg A Gerhardt
- Brain Restoration Center, and.,Departments of 2 Neurosurgery.,Anatomy & Neurobiology, and.,Neurology, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
133
|
Gao J, Kang XY, Sun S, Li L, Zhang BL, Li YQ, Gao DS. Transcription factor Six2 mediates the protection of GDNF on 6-OHDA lesioned dopaminergic neurons by regulating Smurf1 expression. Cell Death Dis 2016; 7:e2217. [PMID: 27148690 PMCID: PMC4917658 DOI: 10.1038/cddis.2016.120] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 04/06/2016] [Accepted: 04/06/2016] [Indexed: 01/19/2023]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) has strong neuroprotective and neurorestorative effects on dopaminergic (DA) neurons in the substantia nigra (SN); however, the underlying molecular mechanisms remain to be fully elucidated. In this study, we found that the expression level of transcription factor Six2 was increased in damaged DA neurons after GDNF rescue in vivo and in vitro. Knockdown of Six2 resulted in decreased cell viability and increased the apoptosis of damaged DA neurons after GDNF treatment in vitro. In contrast, Six2 overexpression increased cell viability and decreased cell apoptosis. Furthermore, genome-wide chromatin immunoprecipitation sequencing (ChIP-seq) indicated that Six2 directly bound to the promoter CAGCTG sequence of smad ubiquitylation regulatory factor 1 (Smurf1). ChIP-quantitative polymerase chain reaction (qPCR) analysis showed that Smurf1 expression was significantly upregulated after GDNF rescue. Moreover, knockdown of Six2 decreased Smurf1 expression, whereas overexpression of Six2 increased Smurf1 expression in damaged DA neurons after GDNF rescue. Meanwhile, knockdown and overexpression of Smurf1 increased and decreased p53 expression, respectively. Taken together, our results from in vitro and in vivo analysis indicate that Six2 mediates the protective effects of GDNF on damaged DA neurons by regulating Smurf1 expression, which could be useful in identifying potential drug targets for injured DA neurons.
Collapse
Affiliation(s)
- J Gao
- Department of Anatomy and Histology, The Fourth Military Medical University, Xian 710003, Shanxi, China.,Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical College, Xuzhou 221004, Jiangsu, China
| | - X-Y Kang
- Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical College, Xuzhou 221004, Jiangsu, China
| | - S Sun
- Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical College, Xuzhou 221004, Jiangsu, China
| | - L Li
- Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical College, Xuzhou 221004, Jiangsu, China
| | - B-L Zhang
- Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical College, Xuzhou 221004, Jiangsu, China
| | - Y-Q Li
- Department of Anatomy and Histology, The Fourth Military Medical University, Xian 710003, Shanxi, China
| | - D-S Gao
- Department of Anatomy and Histology, The Fourth Military Medical University, Xian 710003, Shanxi, China.,Department of Neurobiology and Anatomy, Xuzhou Key Laboratory of Neurobiology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical College, Xuzhou 221004, Jiangsu, China
| |
Collapse
|
134
|
Bruno V, Caraci F, Copani A, Matrisciano F, Nicoletti F, Battaglia G. The impact of metabotropic glutamate receptors into active neurodegenerative processes: A "dark side" in the development of new symptomatic treatments for neurologic and psychiatric disorders. Neuropharmacology 2016; 115:180-192. [PMID: 27140693 DOI: 10.1016/j.neuropharm.2016.04.044] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 04/22/2016] [Accepted: 04/28/2016] [Indexed: 12/17/2022]
Abstract
Metabotropic glutamate (mGlu) receptor ligands are under clinical development for the treatment of CNS disorders with high social and economic burden, such as schizophrenia, major depressive disorder (MDD), and Parkinson's disease (PD), and are promising drug candidates for the treatment of Alzheimer's disease (AD). So far, clinical studies have shown symptomatic effects of mGlu receptor ligands, but it is unknown whether these drugs act as disease modifiers or, at the opposite end, they accelerate disease progression by enhancing neurodegeneration. This is a fundamental issue in the treatment of PD and AD, and is also an emerging theme in the treatment of schizophrenia and MDD, in which neurodegeneration is also present and contribute to disease progression. Moving from in vitro data and preclinical studies, we discuss the potential impact of drugs targeting mGlu2, mGlu3, mGlu4 and mGlu5 receptor ligands on active neurodegeneration associated with AD, PD, schizophrenia, and MDD. We wish to highlight that our final comments on the best drug candidates are not influenced by commercial interests or by previous or ongoing collaborations with drug companies. This article is part of the Special Issue entitled 'Metabotropic Glutamate Receptors, 5 years on'.
Collapse
Affiliation(s)
- Valeria Bruno
- Department of Physiology and Pharmacology, University Sapienza, 00185 Rome, Italy; I.R.C.C.S. Neuromed, 86077 Pozzilli, Italy.
| | - Filippo Caraci
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy; I.R.C.C.S. Associazione Oasi Maria S.S., Institute for Research on Mental Retardation and Brain Aging, 94018 Troina, Italy
| | - Agata Copani
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy; National Research Council, Institute of Biostructure and Bioimaging (IBB-CNR), 95126 Catania, Italy
| | - Francesco Matrisciano
- Department of Psychiatry and Behavioral Sciences, Northwestern Feinberg School of Medicine, Chicago, USA
| | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology, University Sapienza, 00185 Rome, Italy; I.R.C.C.S. Neuromed, 86077 Pozzilli, Italy
| | | |
Collapse
|
135
|
Eid L, Parent M. Chemical anatomy of pallidal afferents in primates. Brain Struct Funct 2016; 221:4291-4317. [PMID: 27028222 DOI: 10.1007/s00429-016-1216-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 03/15/2016] [Indexed: 12/24/2022]
Abstract
Neurons of the globus pallidus receive massive inputs from the striatum and the subthalamic nucleus, but their activity, as well as those of their striatal and subthalamic inputs, are modulated by brainstem afferents. These include serotonin (5-HT) projections from the dorsal raphe nucleus, cholinergic (ACh) inputs from the pedunculopontine tegmental nucleus, and dopamine (DA) afferents from the substantia nigra pars compacta. This review summarizes our recent findings on the distribution, quantitative and ultrastructural aspects of pallidal 5-HT, ACh and DA innervations. These results have led to the elaboration of a new model of the pallidal neuron based on a precise knowledge of the hierarchy and chemical features of the various synaptic inputs. The dense 5-HT, ACh and DA innervations disclosed in the associative and limbic pallidal territories suggest that these brainstem inputs contribute principally to the planification of motor behaviors and the regulation of attention and mood. Although 5-HT, ACh and DA inputs were found to modulate pallidal neurons and their afferents mainly through asynaptic (volume) transmission, genuine synaptic contacts occur between these chemospecific axon varicosities and pallidal dendrites, revealing that these brainstem projections have a direct access to pallidal neurons, in addition to their indirect input through the striatum and subthalamic nucleus. Altogether, these findings reveal that the brainstem 5-HT, ACh and DA pallidal afferents act in concert with the more robust GABAergic inhibitory striatopallidal and glutamatergic excitatory subthalamopallidal inputs. We hypothesize that a fragile equilibrium between forebrain and brainstem pallidal afferents plays a key role in the functional organization of the primate basal ganglia, in both health and disease.
Collapse
Affiliation(s)
- Lara Eid
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Centre de recherche de l'Institut universitaire en santé mentale de Québec (CRIUSMQ), Université Laval, F-6530-1, 2601, de la Canardière, Quebec, QC, G1J 2G3, Canada
| | - Martin Parent
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Centre de recherche de l'Institut universitaire en santé mentale de Québec (CRIUSMQ), Université Laval, F-6530-1, 2601, de la Canardière, Quebec, QC, G1J 2G3, Canada.
| |
Collapse
|
136
|
Yu-Wai-Man P. Genetic manipulation for inherited neurodegenerative diseases: myth or reality? Br J Ophthalmol 2016; 100:1322-31. [PMID: 27002113 PMCID: PMC5050284 DOI: 10.1136/bjophthalmol-2015-308329] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 02/28/2016] [Indexed: 12/22/2022]
Abstract
Rare genetic diseases affect about 7% of the general population and over 7000 distinct clinical syndromes have been described with the majority being due to single gene defects. This review will provide a critical overview of genetic strategies that are being pioneered to halt or reverse disease progression in inherited neurodegenerative diseases. This field of research covers a vast area and only the most promising treatment paradigms will be discussed with a particular focus on inherited eye diseases, which have paved the way for innovative gene therapy paradigms, and mitochondrial diseases, which are currently generating a lot of debate centred on the bioethics of germline manipulation.
Collapse
Affiliation(s)
- Patrick Yu-Wai-Man
- Wellcome Trust Centre for Mitochondrial Research, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK Newcastle Eye Centre, Royal Victoria Infirmary, Newcastle upon Tyne, UK NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology, London, UK
| |
Collapse
|
137
|
Comparative Analysis of the Effects of Neurotrophic Factors CDNF and GDNF in a Nonhuman Primate Model of Parkinson's Disease. PLoS One 2016; 11:e0149776. [PMID: 26901822 PMCID: PMC4763937 DOI: 10.1371/journal.pone.0149776] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 02/04/2016] [Indexed: 11/19/2022] Open
Abstract
Cerebral dopamine neurotrophic factor (CDNF) belongs to a newly discovered family of evolutionarily conserved neurotrophic factors. We demonstrate for the first time a therapeutic effect of CDNF in a unilateral 6-hydroxydopamine (6-OHDA) lesion model of Parkinson’s disease in marmoset monkeys. Furthermore, we tested the impact of high chronic doses of human recombinant CDNF on unlesioned monkeys and analyzed the amino acid sequence of marmoset CDNF. The severity of 6-OHDA lesions and treatment effects were monitored in vivo using 123I-FP-CIT (DaTSCAN) SPECT. Quantitative analysis of 123I-FP-CIT SPECT showed a significant increase of dopamine transporter binding activity in lesioned animals treated with CDNF. Glial cell line-derived neurotrophic factor (GDNF), a well-characterized and potent neurotrophic factor for dopamine neurons, served as a control in a parallel comparison with CDNF. By contrast with CDNF, only single animals responded to the treatment with GDNF, but no statistical difference was observed in the GDNF group. However, increased numbers of tyrosine hydroxylase immunoreactive neurons, observed within the lesioned caudate nucleus of GDNF-treated animals, indicate a strong bioactive potential of GDNF.
Collapse
|
138
|
Arnold JC, Salvatore MF. Exercise-Mediated Increase in Nigral Tyrosine Hydroxylase Is Accompanied by Increased Nigral GFR-α1 and EAAC1 Expression in Aging Rats. ACS Chem Neurosci 2016; 7:227-39. [PMID: 26599339 PMCID: PMC4926611 DOI: 10.1021/acschemneuro.5b00282] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Exercise may alleviate locomotor impairment in Parkinson's disease (PD) or aging. Identifying molecular responses immediately engaged by exercise in the nigrostriatal pathway and allied tissue may reveal critical targets associated with its long-term benefits. In aging, there is loss of tyrosine hydroxylase (TH) and the glial cell line-derived neurotrophic factor (GDNF) receptor, GFR-α1, in the substantia nigra (SN). Exercise can increase GDNF expression, but its effect on GFR-α1 expression is unknown. Infusion of GDNF into striatum or GFR-α1 in SN, respectively, can increase locomotor activity and TH function in SN but not striatum in aged rats. GDNF may also increase glutamate transporter expression, which attenuates TH loss in PD models. We utilized a footshock-free treadmill exercise regimen to determine the immediate impact of short-term exercise on GFR-α1 expression, dopamine regulation, glutamate transporter expression, and glutamate uptake in 18 month old male Brown-Norway/Fischer 344 F1 hybrid rats. GFR-α1 and TH expression significantly increased in SN but not striatum. This exercise regimen did not affect glutamate uptake or glutamate transporter expression in striatum. However, EAAC1 expression increased in SN. These results indicate that nigral GFR-α1 and EAAC1 expression increased in conjunction with increased nigral TH expression following short-term exercise.
Collapse
Affiliation(s)
- Jennifer C. Arnold
- Department of Pharmacology, Toxicology & Neuroscience, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130, United States
| | - Michael F. Salvatore
- Department of Pharmacology, Toxicology & Neuroscience, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130, United States
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas 76107, United States
| |
Collapse
|
139
|
Lewis O, Woolley M, Johnson D, Rosser A, Barua NU, Bienemann AS, Gill SS, Evans S. Chronic, intermittent convection-enhanced delivery devices. J Neurosci Methods 2016; 259:47-56. [DOI: 10.1016/j.jneumeth.2015.11.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 10/28/2015] [Accepted: 11/06/2015] [Indexed: 12/11/2022]
|
140
|
Fan CH, Ting CY, Lin CY, Chan HL, Chang YC, Chen YY, Liu HL, Yeh CK. Noninvasive, Targeted, and Non-Viral Ultrasound-Mediated GDNF-Plasmid Delivery for Treatment of Parkinson's Disease. Sci Rep 2016; 6:19579. [PMID: 26786201 PMCID: PMC4726227 DOI: 10.1038/srep19579] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 12/16/2015] [Indexed: 01/30/2023] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) supports the growth and survival of dopaminergic neurons. CNS gene delivery currently relies on invasive intracerebral injection to transit the blood-brain barrier. Non-viral gene delivery via systematic transvascular route is an attractive alternative because it is non-invasive, but a high-yield and targeted gene-expressed method is still lacking. In this study, we propose a novel non-viral gene delivery approach to achieve targeted gene transfection. Cationic microbubbles as gene carriers were developed to allow the stable formation of a bubble-GDNF gene complex, and transcranial focused ultrasound (FUS) exposure concurrently interacting with the bubble-gene complex allowed transient gene permeation and induced local GDNF expression. We demonstrate that the focused ultrasound-triggered GDNFp-loaded cationic microbubbles platform can achieve non-viral targeted gene delivery via a noninvasive administration route, outperform intracerebral injection in terms of targeted GDNF delivery of high-titer GDNF genes, and has a neuroprotection effect in Parkinson’s disease (PD) animal models to successfully block PD syndrome progression and to restore behavioral function. This study explores the potential of using FUS and bubble-gene complexes to achieve noninvasive and targeted gene delivery for the treatment of neurodegenerative disease.
Collapse
Affiliation(s)
- Ching-Hsiang Fan
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013 Taiwan
| | - Chien-Yu Ting
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013 Taiwan
| | - Chung-Yin Lin
- Medical Imaging Research Center, Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital, Taoyuan, 33302 Taiwan
| | - Hong-Lin Chan
- Department of Medical Science and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 30013 Taiwan
| | - Yuan-Chih Chang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 11529 Taiwan
| | - You-Yin Chen
- Department of Biomedical Engineering, National Yang Ming University, Taipei, 11221 Taiwan
| | - Hao-Li Liu
- Medical Imaging Research Center, Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital, Taoyuan, 33302 Taiwan.,Department of Electrical Engineering, Chang-Gung University, Taoyuan, 33302 Taiwan
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013 Taiwan
| |
Collapse
|
141
|
Saghazadeh A, Rezaei N. MicroRNA machinery in Parkinson's disease: a platform for neurodegenerative diseases. Expert Rev Neurother 2015; 22:427-453. [PMID: 26574782 DOI: 10.1586/14737175.2015.1114886] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
MicroRNAs (miRNAs) are noncoding RNAs that recognize their protein-coding target genes and whereby subjugate them after transcription. Despite the infancy of this field of science, the role of miRNAs in neurodegeneration is well-acknowledged. This review was conducted to indicate that Parkinson's disease (PD) is not excluded from this rule. To this end, we evaluated the existing literature and arranged PD-associated miRNAs according to their mechanism of action, particularly apoptosis, autophagy, inflammation, mitochondrial dysfunction and oxidative stress. According to this arrangement, a majority of PD-associated miRNAs were indicated to influence autophagic/apoptotic pathways. We also categorized PD-associated miRNAs according to that they could exert detrimental or beneficial or both into three sets, activator, inhibitor, and double-edged, correspondingly. Considering this criterion, a majority of PD-associated miRNAs were included in the activator category. In addition, evidences from genetic association studies investigating genetic variants of or related to miRNAs in PD patients are presented. Finally, possible applications of the miRNA machinery in PD, including mechanistic networks, diagnostic, prognostic and therapeutic potentials, are discussed. But there may be additional miRNAs involved in the pathogenesis of PD which have hitherto remained unknown and thus further studies are needed to explore the issue and to extend this platform.
Collapse
Affiliation(s)
- Amene Saghazadeh
- a Molecular Immunology Research Center and Department of Immunology, School of Medicine , Tehran University of Medical Sciences , Tehran , Iran
| | - Nima Rezaei
- a Molecular Immunology Research Center and Department of Immunology, School of Medicine , Tehran University of Medical Sciences , Tehran , Iran.,b Research Center for Immunodeficiencies, Children's Medical Center , Tehran University of Medical Sciences , Tehran , Iran.,c Universal Scientific Education and Research Network (USERN) , Tehran , Iran
| |
Collapse
|
142
|
GDNF-induced cerebellar toxicity: A brief review. Neurotoxicology 2015; 52:46-56. [PMID: 26535469 DOI: 10.1016/j.neuro.2015.10.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 09/22/2015] [Accepted: 10/24/2015] [Indexed: 12/19/2022]
Abstract
Recombinant-methionyl human glial cell line-derived neurotrophic factor (GDNF) is known for its neurorestorative and neuroprotective effects in rodent and primate models of Parkinson's disease (PD). When administered locally into the putamen of Parkinsonian subjects, early clinical studies showed its potential promise as a disease-modifying agent. However, the development of GDNF for the treatment of PD has been significantly clouded by findings of cerebellar toxicity after continuous intraputamenal high-dose administration in a 6-month treatment/3-month recovery toxicology study in rhesus monkeys. Specifically, multifocal cerebellar Purkinje cell loss affecting 1-21% of the cerebellar cortex was observed in 4 of 15 (26.7%; 95% confidence interval [CI]: 10.5-52.4%) animals treated at the highest dose level tested (3000μg/month). No cerebellar toxicity was observed at lower doses (450 and 900μg/month) in the same study, or at similar or higher doses (up to 10,000μg/month) in subchronic or chronic toxicology studies testing intermittent intracerebroventricular administration. While seemingly associated with the use of GDNF, the pathogenesis of the cerebellar lesions has not been fully understood to date. This review integrates available information to evaluate potential pathogenic mechanisms and provide a consolidated assessment of the findings. While other explanations are considered, the existing evidence is most consistent with the hypothesis that leakage of GDNF into cerebrospinal fluid during chronic infusions into the putamen down-regulates GDNF receptors on Purkinje cells, and that subsequent acute withdrawal of GDNF generates the observed lesions. The implications of these findings for clinical studies with GDNF are discussed.
Collapse
|
143
|
Staudt MD, Di Sebastiano AR, Xu H, Jog M, Schmid S, Foster P, Hebb MO. Advances in Neurotrophic Factor and Cell-Based Therapies for Parkinson's Disease: A Mini-Review. Gerontology 2015; 62:371-80. [PMID: 26330171 DOI: 10.1159/000438701] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 07/14/2015] [Indexed: 12/28/2022] Open
Abstract
Parkinson's disease (PD) affects an estimated 7-10 million people worldwide and remains without definitive or disease-modifying treatment. There have been many recent developments in cell-based therapy (CBT) to replace lost circuitry and provide chronic biological sources of therapeutic agents to the PD-affected brain. Early neural transplantation studies underscored the challenges of immune compatibility, graft integration and the need for renewable, autologous graft sources. Neurotrophic factors (NTFs) offer a potential class of cytoprotective pharmacotherapeutics that may complement dopamine (DA) replacement and CBT strategies in PD. Chronic NTF delivery may be an integral goal of CBT, with grafts consisting of autologous drug-producing (e.g., DA, NTF) cells that are capable of integration and function in the host brain. In this mini-review, we outline the past experience and recent advances in NTF technology and CBT as promising and integrated approaches for the treatment of PD.
Collapse
Affiliation(s)
- Michael D Staudt
- Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western University, London, Ont., Canada
| | | | | | | | | | | | | |
Collapse
|
144
|
Eid L, Parent M. Morphological evidence for dopamine interactions with pallidal neurons in primates. Front Neuroanat 2015; 9:111. [PMID: 26321923 PMCID: PMC4531254 DOI: 10.3389/fnana.2015.00111] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 07/27/2015] [Indexed: 12/04/2022] Open
Abstract
The external (GPe) and internal (GPi) segments of the primate globus pallidus receive dopamine (DA) axonal projections arising mainly from the substantia nigra pars compacta and this innervation is here described based on tyrosine hydroxylase (TH) immunohistochemical observations gathered in the squirrel monkey (Saimiri sciureus). At the light microscopic level, unbiased stereological quantification of TH positive (+) axon varicosities reveals a similar density of innervation in the GPe (0.19 ± 0.02 × 106 axon varicosities/mm3 of tissue) and GPi (0.17 ± 0.01 × 106), but regional variations occur in the anteroposterior and dorsoventral axes in both GPe and GPi and along the mediolateral plane in the GPe. Estimation of the neuronal population in the GPe (3.47 ± 0.15 × 103 neurons/mm3) and GPi (2.69 ± 0.18 × 103) yields a mean ratio of, respectively, 28 ± 3 and 68 ± 15 TH+ axon varicosities/pallidal neuron. At the electron microscopic level, TH+ axon varicosities in the GPe appear significantly smaller than those in the GPi and very few TH+ axon varicosities are engaged in synaptic contacts in the GPe (17 ± 3%) and the GPi (15 ± 4%) compared to their unlabeled counterparts (77 ± 6 and 50 ± 12%, respectively). Genuine synaptic contacts made by TH+ axon varicosities in the GPe and GPi are of the symmetrical and asymmetrical type. Such synaptic contacts together with the presence of numerous synaptic vesicles in all TH+ axon varicosities observed in the GPe and GPi support the functionality of the DA pallidal innervation. By virtue of its predominantly volumic mode of action, DA appears to exert a key modulatory effect upon pallidal neurons in concert with the more direct GABAergic inhibitory and glutamatergic excitatory actions of the striatum and subthalamic nucleus. We argue that the DA pallidal innervation plays a major role in the functional organization of the primate basal ganglia under both normal and pathological conditions.
Collapse
Affiliation(s)
- Lara Eid
- Department of Psychiatry and Neuroscience, Centre de Recherche de l'Institut Universitaire en Santé Mentale de Québec, Université Laval Quebec City, QC, Canada
| | - Martin Parent
- Department of Psychiatry and Neuroscience, Centre de Recherche de l'Institut Universitaire en Santé Mentale de Québec, Université Laval Quebec City, QC, Canada
| |
Collapse
|
145
|
Aron Badin R, Vadori M, Cozzi E, Hantraye P. Translational research for Parkinson׳s disease: The value of pre-clinical primate models. Eur J Pharmacol 2015; 759:118-26. [DOI: 10.1016/j.ejphar.2015.03.038] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 03/10/2015] [Accepted: 03/12/2015] [Indexed: 12/15/2022]
|
146
|
Warren Olanow C, Bartus RT, Baumann TL, Factor S, Boulis N, Stacy M, Turner DA, Marks W, Larson P, Starr PA, Jankovic J, Simpson R, Watts R, Guthrie B, Poston K, Henderson JM, Stern M, Baltuch G, Goetz CG, Herzog C, Kordower JH, Alterman R, Lozano AM, Lang AE. Gene delivery of neurturin to putamen and substantia nigra in Parkinson disease: A double-blind, randomized, controlled trial. Ann Neurol 2015; 78:248-57. [PMID: 26061140 DOI: 10.1002/ana.24436] [Citation(s) in RCA: 183] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 04/27/2015] [Accepted: 05/04/2015] [Indexed: 11/09/2022]
Abstract
OBJECTIVE A 12-month double-blind sham-surgery-controlled trial assessing adeno-associated virus type 2 (AAV2)-neurturin injected into the putamen bilaterally failed to meet its primary endpoint, but showed positive results for the primary endpoint in the subgroup of subjects followed for 18 months and for several secondary endpoints. Analysis of postmortem tissue suggested impaired axonal transport of neurturin from putamen to substantia nigra. In the present study, we tested the safety and efficacy of AAV2-neurturin delivered to putamen and substantia nigra. METHODS We performed a 15- to 24-month, multicenter, double-blind trial in patients with advanced Parkinson disease (PD) who were randomly assigned to receive bilateral AAV2-neurturin injected bilaterally into the substantia nigra (2.0 × 10(11) vector genomes) and putamen (1.0 × 10(12) vector genomes) or sham surgery. The primary endpoint was change from baseline to final visit performed at the time the last enrolled subject completed the 15-month evaluation in the motor subscore of the Unified Parkinson's Disease Rating Scale in the practically defined off state. RESULTS Fifty-one patients were enrolled in the trial. There was no significant difference between groups in the primary endpoint (change from baseline: AAV2-neurturin, -7.0 ± 9.92; sham, -5.2 ± 10.01; p = 0.515) or in most secondary endpoints. Two subjects had cerebral hemorrhages with transient symptoms. No clinically meaningful adverse events were attributed to AAV2-neurturin. INTERPRETATION AAV2-neurturin delivery to the putamen and substantia nigra bilaterally in PD was not superior to sham surgery. The procedure was well tolerated, and there were no clinically significant adverse events related to AAV2-neurturin.
Collapse
Affiliation(s)
- C Warren Olanow
- Departments of Neurology and Neuroscience, Mount Sinai School of Medicine, New York, NY
| | | | | | - Stewart Factor
- Department of Neurology, Emory University School of Medicine, Atlanta, GA
| | - Nicholas Boulis
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA
| | - Mark Stacy
- Department of Neurology, Duke University School of Medicine, Durham, NC
| | - Dennis A Turner
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC
| | - William Marks
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Paul Larson
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Phillip A Starr
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Joseph Jankovic
- Department of Neurology, Baylor College of Medicine, Houston, TX
| | - Richard Simpson
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX
| | - Ray Watts
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL
| | - Barton Guthrie
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL
| | - Kathleen Poston
- Department of Neurology and Neurological Sciences, Stanford University Medical Center, Stanford, CA
| | - Jaimie M Henderson
- Department of Neurosurgery, Stanford University Medical Center, Stanford, CA
| | - Matthew Stern
- Department of Neurology, University of Pennsylvania, Philadelphia, PA
| | - Gordon Baltuch
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA
| | - Christopher G Goetz
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL
| | | | - Jeffrey H Kordower
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL
| | - Ron Alterman
- Beth Israel-Deaconess Medical Center, Department of Neurosurgery, Boston, MA
| | - Andres M Lozano
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Anthony E Lang
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
147
|
Abstract
ABSTRACT
Midbrain dopaminergic (mDA) neuron development has been an intense area of research during recent years. This is due in part to a growing interest in regenerative medicine and the hope that treatment for diseases affecting mDA neurons, such as Parkinson's disease (PD), might be facilitated by a better understanding of how these neurons are specified, differentiated and maintained in vivo. This knowledge might help to instruct efforts to generate mDA neurons in vitro, which holds promise not only for cell replacement therapy, but also for disease modeling and drug discovery. In this Primer, we will focus on recent developments in understanding the molecular mechanisms that regulate the development of mDA neurons in vivo, and how they have been used to generate human mDA neurons in vitro from pluripotent stem cells or from somatic cells via direct reprogramming. Current challenges and future avenues in the development of a regenerative medicine for PD will be identified and discussed.
Collapse
Affiliation(s)
- Ernest Arenas
- Laboratory of Molecular Neurobiology, Dept. Medical Biochemistry and Biophysics, Center of Developmental Biology for Regenerative Medicine, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Mark Denham
- Laboratory of Molecular Neurobiology, Dept. Medical Biochemistry and Biophysics, Center of Developmental Biology for Regenerative Medicine, Karolinska Institutet, Stockholm 171 77, Sweden
- Danish Research Institute of Translational Neuroscience, Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus 8000, Denmark
| | - J. Carlos Villaescusa
- Laboratory of Molecular Neurobiology, Dept. Medical Biochemistry and Biophysics, Center of Developmental Biology for Regenerative Medicine, Karolinska Institutet, Stockholm 171 77, Sweden
- Institute of Experimental Biology, Faculty of Science, Masaryk University, Brno 61137, Czech Republic
| |
Collapse
|
148
|
Stenslik MJ, Potts LF, Sonne JWH, Cass WA, Turchan-Cholewo J, Pomerleau F, Huettl P, Ai Y, Gash DM, Gerhardt GA, Bradley LH. Methodology and effects of repeated intranasal delivery of DNSP-11 in a rat model of Parkinson's disease. J Neurosci Methods 2015; 251:120-9. [PMID: 25999268 DOI: 10.1016/j.jneumeth.2015.05.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 05/11/2015] [Accepted: 05/11/2015] [Indexed: 01/21/2023]
Abstract
BACKGROUND To circumvent the challenges associated with delivering large compounds directly to the brain for the treatment of Parkinson's disease (PD), non-invasive procedures utilizing smaller molecules with protective and/or restorative actions on dopaminergic neurons are needed. NEW METHOD We developed a methodology for evaluating the effects of a synthetic neuroactive peptide, DNSP-11, on the nigrostriatal system using repeated intranasal delivery in both normal and a unilateral 6-hydroxydopamine (6-OHDA) lesion rat model of PD. RESULTS Normal rats repeatedly administered varying doses of DNSP-11 intranasally for 3 weeks exhibited a significant increase in dopamine (DA) turnover in both the striatum and substantia nigra (SN) at 300μg, suggestive of a stimulative effect of the dopaminergic system. Additionally, a protective effect was observed following repeated intranasal administration in 6-OHDA lesioned rats, as suggested by: a significant decrease in d-amphetamine-induced rotation at 2 weeks; a decrease in DA turnover in the lesioned striatum; and an increased sparing of tyrosine hydroxylase (TH) positive (+) neurons in a specific sub-region of the lesioned substantia nigra pars compacta (SNpc). Finally, tracer studies showed (125)I-DNSP-11 distributed diffusely throughout the brain, including the striatum and SN, as quickly as 30min after a single intranasal dose. COMPARISON WITH EXISTING METHODS The results of bilateral intranasal administration of DNSP-11 are compared to our unilateral single infusion studies to the brain in rats. CONCLUSIONS These studies support that DNSP-11 can be delivered intranasally and maintain its neuroactive properties in both normal rats and in a unilateral 6-OHDA rat model of PD.
Collapse
Affiliation(s)
- Mallory J Stenslik
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Lisa F Potts
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - James W H Sonne
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Wayne A Cass
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Jadwiga Turchan-Cholewo
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Francois Pomerleau
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Peter Huettl
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Yi Ai
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Don M Gash
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Greg A Gerhardt
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Luke H Bradley
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA; Department of Molecular & Cellular Biochemistry and Center of Structural Biology, University of Kentucky College of Medicine, Lexington, KY, USA.
| |
Collapse
|
149
|
Viral vector delivery of neurotrophic factors for Parkinson's disease therapy. Expert Rev Mol Med 2015; 17:e8. [DOI: 10.1017/erm.2015.6] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterised by the progressive loss of midbrain dopaminergic neurons, which causes motor impairments. Current treatments involve dopamine replacement to address the disease symptoms rather than its cause. Factors that promote the survival of dopaminergic neurons have been proposed as novel therapies for PD. Several dopaminergic neurotrophic factors (NTFs) have been examined for their ability to protect and/or restore degenerating dopaminergic neurons, both in animal models and in clinical trials. These include glial cell line-derived neurotrophic factor, neurturin, cerebral dopamine neurotrophic factor and growth/differentiation factor 5. Delivery of these NTFs via injection or infusion to the brain raises several practical problems. A new delivery approach for NTFs involves the use of recombinant viral vectors to enable long-term expression of these factors in brain cells. Vectors used include those based on adenoviruses, adeno-associated viruses and lentiviruses. Here we review progress to date on the potential of each of these four NTFs as novel therapeutic strategies for PD, as well as the challenges that have arisen, from pre-clinical analysis to clinical trials. We conclude by discussing recently-developed approaches to optimise the delivery of NTF-carrying viral vectors to the brain.
Collapse
|
150
|
Sano H, Murata M, Nambu A. Zonisamide reduces nigrostriatal dopaminergic neurodegeneration in a mouse genetic model of Parkinson's disease. J Neurochem 2015; 134:371-81. [DOI: 10.1111/jnc.13116] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 02/26/2015] [Accepted: 04/07/2015] [Indexed: 12/15/2022]
Affiliation(s)
- Hiromi Sano
- Division of System Neurophysiology; National Institute for Physiological Sciences; Okazaki Aichi Japan
- Department of Physiological Sciences; SOKENDAI (The Graduate University for Advanced Studies); Okazaki Aichi Japan
| | - Miho Murata
- Department of Neurology; National Center Hospital; National Center of Neurology and Psychiatry; Kodaira Tokyo Japan
| | - Atsushi Nambu
- Division of System Neurophysiology; National Institute for Physiological Sciences; Okazaki Aichi Japan
- Department of Physiological Sciences; SOKENDAI (The Graduate University for Advanced Studies); Okazaki Aichi Japan
| |
Collapse
|