1
|
Heiss JD, Ray-Chaudhury A, Kleiner DE, Ehrlich D, Scott G, Edwards NA, Goldstein DS, Hammoud DA, Hadaczek P, Van Laar VS, Graff S, Herscovitch P, Lungu C, Hallett M, Lonser RR, Zaghloul K, Bankiewicz KS. Persistent GDNF Expression 45 Months after Putaminal Infusion of AAV2-GDNF in a Patient with Parkinson's Disease. Mov Disord 2024; 39:1412-1417. [PMID: 38718138 PMCID: PMC11341257 DOI: 10.1002/mds.29820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 04/08/2024] [Indexed: 08/23/2024] Open
Abstract
OBJECTIVE Gene therapy by convection-enhanced delivery of type 2 adeno-associated virus-glial cell derived neurotrophic factor (AAV2-GDNF) to the bilateral putamina seeks to increase GDNF gene expression and treat Parkinson's disease (PD). METHODS A 63-year-old man with advanced PD received AAV2-GDNF in a clinical trial. He died from pneumonia after anterior cervical discectomy and fusion 45 months later. An autopsy included brain examination for GDNF transgene expression. Putaminal catecholamine concentrations were compared to in vivo 18F-Fluorodopa (18F-FDOPA) positron emission tomography (PET) scanning results before and 18 months after AAV2-GDNF infusion. RESULTS Parkinsonian progression stabilized clinically. Postmortem neuropathology confirmed PD. Bilateral putaminal regions previously infused with AAV2-GDNF expressed the GDNF gene. Total putaminal dopamine was 1% of control, confirming the striatal dopaminergic deficiency suggested by baseline 18F-DOPA-PET scanning. Putaminal regions responded as expected to AAV2-GDNF. CONCLUSION After AAV2-GDNF infusion, infused putaminal regions showed increased GDNF gene expression, tyrosine hydroxylase immunoreactive sprouting, catechol levels, and 18F-FDOPA-PET signal, suggesting the regenerative potential of AAV2-GDNF in PD.
Collapse
Affiliation(s)
- John D. Heiss
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Abhik Ray-Chaudhury
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - David E. Kleiner
- Laboratory of Pathology, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD 20892
| | - Debra Ehrlich
- Parkinson’s Disease Clinic, Office of the Clinical Director, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Gretchen Scott
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Nancy A. Edwards
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - David S. Goldstein
- Autonomic Medicine Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Dima A. Hammoud
- Center for Infectious Disease Imaging, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Piotr Hadaczek
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Victor S. Van Laar
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Shantelle Graff
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Peter Herscovitch
- Positron Emission Tomography Department, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Codrin Lungu
- Division of Clinical Research, and Office of the Clinical Director, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Mark Hallett
- Human Motor Control Section, Medical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Russell R. Lonser
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Kareem Zaghloul
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Krystof S. Bankiewicz
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
2
|
Kakoty V, Sarathlal KC, Kaur P, Wadhwa P, Vishwas S, Khan FR, Alhazmi AYM, Almasoudi HH, Gupta G, Chellappan DK, Paudel KR, Kumar D, Dua K, Singh SK. Unraveling the role of glial cell line-derived neurotrophic factor in the treatment of Parkinson's disease. Neurol Sci 2024; 45:1409-1418. [PMID: 38082050 DOI: 10.1007/s10072-023-07253-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/02/2023] [Indexed: 03/16/2024]
Abstract
Parkinson's disease is the second most common neurodegenerative condition with its prevalence projected to 8.9 million individuals globally in the year 2019. Parkinson's disease affects both motor and certain non-motor functions of an individual. Numerous research has focused on the neuroprotective effect of the glial cell line-derived neurotrophic factor (GDNF) in Parkinson's disease. Discovered in 1993, GDNF is a neurotrophic factor identified from the glial cells which was found to have selective effects on promoting survival and regeneration of certain populations of neurons including the dopaminergic nigrostriatal pathway. Given this property, recent studies have focused on the exogenous administration of GDNF for relieving Parkinson's disease-related symptoms both at a pre-clinical and a clinical level. This review will focus on enumerating the molecular connection between Parkinson's disease and GDNF and shed light on all the available drug delivery approaches to facilitate the selective delivery of GDNF into the brain paving the way as a potential therapeutic candidate for Parkinson's disease in the future.
Collapse
Affiliation(s)
- Violina Kakoty
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India
| | - K C Sarathlal
- Department of Non-Communicable Disease, Translational Health Science and Technology Institute, Faridabad, India
| | - Palwinder Kaur
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India
| | - Pankaj Wadhwa
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India
| | - Sukriti Vishwas
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India
| | - Farhan R Khan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| | | | - Hassan Hussain Almasoudi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran, 61441, Saudi Arabia
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- School of Pharmacy, Graphic Era Hill University, Dehradun, 248007, India
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | | | - Keshav Raj Paudel
- Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW, 2050, Australia
| | - Dileep Kumar
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Kamal Dua
- School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India.
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
3
|
Bentea E, De Pauw L, Verbruggen L, Winfrey LC, Deneyer L, Moore C, Albertini G, Sato H, Van Eeckhaut A, Meshul CK, Massie A. Aged xCT-Deficient Mice Are Less Susceptible for Lactacystin-, but Not 1-Methyl-4-Phenyl-1,2,3,6- Tetrahydropyridine-, Induced Degeneration of the Nigrostriatal Pathway. Front Cell Neurosci 2022; 15:796635. [PMID: 34975413 PMCID: PMC8718610 DOI: 10.3389/fncel.2021.796635] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 11/24/2021] [Indexed: 12/23/2022] Open
Abstract
The astrocytic cystine/glutamate antiporter system x c - (with xCT as the specific subunit) imports cystine in exchange for glutamate and has been shown to interact with multiple pathways in the brain that are dysregulated in age-related neurological disorders, including glutamate homeostasis, redox balance, and neuroinflammation. In the current study, we investigated the effect of genetic xCT deletion on lactacystin (LAC)- and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced degeneration of the nigrostriatal pathway, as models for Parkinson's disease (PD). Dopaminergic neurons of adult xCT knock-out mice (xCT-/-) demonstrated an equal susceptibility to intranigral injection of the proteasome inhibitor LAC, as their wild-type (xCT+/+) littermates. Contrary to adult mice, aged xCT-/- mice showed a significant decrease in LAC-induced degeneration of nigral dopaminergic neurons, depletion of striatal dopamine (DA) and neuroinflammatory reaction, compared to age-matched xCT+/+ littermates. Given this age-related protection, we further investigated the sensitivity of aged xCT-/- mice to chronic and progressive MPTP treatment. However, in accordance with our previous observations in adult mice (Bentea et al., 2015a), xCT deletion did not confer protection against MPTP-induced nigrostriatal degeneration in aged mice. We observed an increased loss of nigral dopaminergic neurons, but equal striatal DA denervation, in MPTP-treated aged xCT-/- mice when compared to age-matched xCT+/+ littermates. To conclude, we reveal age-related protection against proteasome inhibition-induced nigrostriatal degeneration in xCT-/- mice, while xCT deletion failed to protect nigral dopaminergic neurons of aged mice against MPTP-induced toxicity. Our findings thereby provide new insights into the role of system x c - in mechanisms of dopaminergic cell loss and its interaction with aging.
Collapse
Affiliation(s)
- Eduard Bentea
- Laboratory of Neuro-Aging and Viro-Immunotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Laura De Pauw
- Laboratory of Neuro-Aging and Viro-Immunotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lise Verbruggen
- Laboratory of Neuro-Aging and Viro-Immunotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lila C Winfrey
- Neurocytology Laboratory, Veterans Affairs Medical Center, Research Services, Portland, OR, United States
| | - Lauren Deneyer
- Laboratory of Neuro-Aging and Viro-Immunotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Cynthia Moore
- Neurocytology Laboratory, Veterans Affairs Medical Center, Research Services, Portland, OR, United States
| | - Giulia Albertini
- Laboratory of Neuro-Aging and Viro-Immunotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hideyo Sato
- Department of Medical Technology, Niigata University, Niigata, Japan
| | - Ann Van Eeckhaut
- Research Group Experimental Pharmacology, Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Vrije Universiteit Brussel, Brussels, Belgium
| | - Charles K Meshul
- Neurocytology Laboratory, Veterans Affairs Medical Center, Research Services, Portland, OR, United States.,Department of Behavioral Neuroscience and Pathology, Oregon Health and Science University, Portland, OR, United States
| | - Ann Massie
- Laboratory of Neuro-Aging and Viro-Immunotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
4
|
Behl T, Kaur I, Kumar A, Mehta V, Zengin G, Arora S. Gene Therapy in the Management of Parkinson's Disease: Potential of GDNF as a Promising Therapeutic Strategy. Curr Gene Ther 2021; 20:207-222. [PMID: 32811394 DOI: 10.2174/1566523220999200817164051] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/14/2020] [Accepted: 07/15/2020] [Indexed: 12/13/2022]
Abstract
The limitations of conventional treatment therapies in Parkinson's disorder, a common neurodegenerative disorder, lead to the development of an alternative gene therapy approach. Multiple treatment options targeting dopaminergic neuronal regeneration, production of enzymes linked with dopamine synthesis, subthalamic nucleus neurons, regulation of astrocytes and microglial cells and potentiating neurotrophic factors, were established. Viral vector-based dopamine delivery, prodrug approaches, fetal ventral mesencephalon tissue transplantation and dopamine synthesizing enzyme encoding gene delivery are significant therapies evidently supported by numerous trials. The review primarily elaborates on the significant role of glial cell-line derived neurotrophic factor in alleviating motor symptoms and the loss of dopaminergic neurons in Parkinson's disease. Neuroprotective and neuroregenerative effects of GDNF were established via preclinical and clinical study outcomes. The binding of GDNF family ligands with associated receptors leads to the formation of a receptor-ligand complex activating Ret receptor of tyrosine kinase family, which is only expressed in dopaminergic neurons, playing an important role in Parkinson's disease, via its association with the essential protein encoded genes. Furthermore, the review establishes delivery aspects, like ventricular delivery of recombinant GDNF, intraparenchymal and intraputaminal delivery using infusion catheters. The review highlights problems and challenges of GDNF delivery, and essential measures to overcome them, like gene therapy combinations, optimization of delivery vectors, newer targeting devices, motor symptoms curbing focused ultrasound techniques, modifications in patient selection criteria and development of novel delivery strategies based on liposomes and encapsulated cells, to promote safe and effective delivery of neurotrophic factor and establishment of routine treatment therapy for patients.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Ishnoor Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | | | - Gokhan Zengin
- Department of Biology, Faculty of Science, Selcuk University Campus, Konya, Turkey
| | - Sandeep Arora
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| |
Collapse
|
5
|
Zhang N, Lin J, Lin VPH, Milbreta U, Chin JS, Chew EGY, Lian MM, Foo JN, Zhang K, Wu W, Chew SY. A 3D Fiber-Hydrogel Based Non-Viral Gene Delivery Platform Reveals that microRNAs Promote Axon Regeneration and Enhance Functional Recovery Following Spinal Cord Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100805. [PMID: 34050637 PMCID: PMC8336488 DOI: 10.1002/advs.202100805] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Indexed: 05/05/2023]
Abstract
Current treatment approaches toward spinal cord injuries (SCI) have mainly focused on overcoming the inhibitory microenvironment that surrounds lesion sites. Unfortunately, the mere modulation of the cell/tissue microenvironment is often insufficient to achieve desired functional recovery. Therefore, stimulating the intrinsic growth ability of injured neurons becomes crucial. MicroRNAs (miRs) play significant roles during axon regeneration by regulating local protein synthesis at growth cones. However, one challenge of using miRs to treat SCI is the lack of efficient delivery approaches. Here, a 3D fiber-hydrogel scaffold is introduced which can be directly implanted into a spinal cord transected rat. This 3D scaffold consists of aligned electrospun fibers which provide topographical cues to direct axon regeneration, and collagen matrix which enables a sustained delivery of miRs. Correspondingly, treatment with Axon miRs (i.e., a cocktail of miR-132/miR-222/miR-431) significantly enhances axon regeneration. Moreover, administration of Axon miRs along with anti-inflammatory drug, methylprednisolone, synergistically enhances functional recovery. Additionally, this combined treatment also decreases the expression of pro-inflammatory genes and enhance gene expressions related to extracellular matrix deposition. Finally, increased Axon miRs dosage with methylprednisolone, significantly promotes functional recovery and remyelination. Altogether, scaffold-mediated Axon miR treatment with methylprednisolone is a promising therapeutic approach for SCI.
Collapse
Affiliation(s)
- Na Zhang
- School of Chemical and Biomedical EngineeringNanyang Technological University62 Nanyang DriveSingapore637459Singapore
| | - Junquan Lin
- School of Chemical and Biomedical EngineeringNanyang Technological University62 Nanyang DriveSingapore637459Singapore
| | - Vincent Po Hen Lin
- School of Chemical and Biomedical EngineeringNanyang Technological University62 Nanyang DriveSingapore637459Singapore
| | - Ulla Milbreta
- School of Chemical and Biomedical EngineeringNanyang Technological University62 Nanyang DriveSingapore637459Singapore
| | - Jiah Shin Chin
- School of Chemical and Biomedical EngineeringNanyang Technological University62 Nanyang DriveSingapore637459Singapore
- Interdisciplinary Graduate SchoolNanyang Technological University61 Nanyang DriveSingapore637335Singapore
| | - Elaine Guo Yan Chew
- Human GeneticsGenome Institute of Singapore60 Biopolis StreetSingapore138672Singapore
- Lee Kong Chian School of MedicineNanyang Technological University59 Nanyang DriveSingapore636921Singapore
| | - Michelle Mulan Lian
- Human GeneticsGenome Institute of Singapore60 Biopolis StreetSingapore138672Singapore
- Lee Kong Chian School of MedicineNanyang Technological University59 Nanyang DriveSingapore636921Singapore
| | - Jia Nee Foo
- Human GeneticsGenome Institute of Singapore60 Biopolis StreetSingapore138672Singapore
- Lee Kong Chian School of MedicineNanyang Technological University59 Nanyang DriveSingapore636921Singapore
| | - Kunyu Zhang
- School of Chemical and Biomedical EngineeringNanyang Technological University62 Nanyang DriveSingapore637459Singapore
| | - Wutian Wu
- Guangdong‐Hongkong‐Macau Institute of CNS RegenerationMinistry of Education CNS Regeneration Collaborative Joint LaboratoryJinan University601 West Huangpu AvenueGuangzhou510632P. R. China
- Re‐Stem Biotechnology Co., Ltd.1463 Wuzhong AveSuzhou330520P. R. China
| | - Sing Yian Chew
- School of Chemical and Biomedical EngineeringNanyang Technological University62 Nanyang DriveSingapore637459Singapore
- Lee Kong Chian School of MedicineNanyang Technological University59 Nanyang DriveSingapore636921Singapore
| |
Collapse
|
6
|
Mah KM, Torres-Espín A, Hallworth BW, Bixby JL, Lemmon VP, Fouad K, Fenrich KK. Automation of training and testing motor and related tasks in pre-clinical behavioural and rehabilitative neuroscience. Exp Neurol 2021; 340:113647. [PMID: 33600814 PMCID: PMC10443427 DOI: 10.1016/j.expneurol.2021.113647] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/25/2021] [Accepted: 02/12/2021] [Indexed: 12/12/2022]
Abstract
Testing and training animals in motor and related tasks is a cornerstone of pre-clinical behavioural and rehabilitative neuroscience. Yet manually testing and training animals in these tasks is time consuming and analyses are often subjective. Consequently, there have been many recent advances in automating both the administration and analyses of animal behavioural training and testing. This review is an in-depth appraisal of the history of, and recent developments in, the automation of animal behavioural assays used in neuroscience. We describe the use of common locomotor and non-locomotor tasks used for motor training and testing before and after nervous system injury. This includes a discussion of how these tasks help us to understand the underlying mechanisms of neurological repair and the utility of some tasks for the delivery of rehabilitative training to enhance recovery. We propose two general approaches to automation: automating the physical administration of behavioural tasks (i.e., devices used to facilitate task training, rehabilitative training, and motor testing) and leveraging the use of machine learning in behaviour analysis to generate large volumes of unbiased and comprehensive data. The advantages and disadvantages of automating various motor tasks as well as the limitations of machine learning analyses are examined. In closing, we provide a critical appraisal of the current state of automation in animal behavioural neuroscience and a prospective on some of the advances in machine learning we believe will dramatically enhance the usefulness of these approaches for behavioural neuroscientists.
Collapse
Affiliation(s)
- Kar Men Mah
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami, Miami, FL 33136, USA
| | - Abel Torres-Espín
- Brain and Spinal Injury Center, Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Ben W Hallworth
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada; Department of Mechanical Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - John L Bixby
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami, Miami, FL 33136, USA; Department of Molecular & Cellular Pharmacology, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Vance P Lemmon
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami, Miami, FL 33136, USA
| | - Karim Fouad
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada; Department of Physical Therapy, University of Alberta, Edmonton, Alberta, Canada; Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Keith K Fenrich
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada; Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
7
|
Beta Frequency Oscillations in the Subthalamic Nucleus Are Not Sufficient for the Development of Symptoms of Parkinsonian Bradykinesia/Akinesia in Rats. eNeuro 2019; 6:ENEURO.0089-19.2019. [PMID: 31540998 PMCID: PMC6817717 DOI: 10.1523/eneuro.0089-19.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 07/30/2019] [Accepted: 09/11/2019] [Indexed: 11/21/2022] Open
Abstract
Substantial correlative evidence links the synchronized, oscillatory patterns of neural activity that emerge in Parkinson's disease (PD) in the beta (β) frequency range (13-30 Hz) with bradykinesia in PD. However, conflicting evidence exists, and whether these changes in neural activity are causal of motor symptoms in PD remains unclear. We tested the hypothesis that the synchronized β oscillations that emerge in PD are causal of symptoms of bradykinesia/akinesia. We designed patterns of stimulation that mimicked the temporal characteristics of single unit β bursting activity seen in PD animals and humans. We applied these β-patterned stimulation patterns along with continuous low-frequency and high-frequency controls to the subthalamic nucleus (STN) of intact and 6-OHDA-lesioned female Long-Evans and Sprague-Dawley rats. β-Patterned paradigms were superior to low-frequency controls at induction of β power in downstream substantia nigra reticulata (SNr) neurons and in ipsilateral motor cortex. However, we did not detect deleterious effects on motor performance across a wide battery of validated behavioral tasks. Our results suggest that β frequency oscillations (BFOs) may not be sufficient for the generation of bradykinesia/akinesia in PD.
Collapse
|
8
|
Heiss JD, Lungu C, Hammoud DA, Herscovitch P, Ehrlich DJ, Argersinger DP, Sinharay S, Scott G, Wu T, Federoff HJ, Zaghloul KA, Hallett M, Lonser RR, Bankiewicz KS. Trial of magnetic resonance-guided putaminal gene therapy for advanced Parkinson's disease. Mov Disord 2019; 34:1073-1078. [PMID: 31145831 DOI: 10.1002/mds.27724] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/26/2019] [Accepted: 05/06/2019] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE To investigate the safety and tolerability of convection-enhanced delivery of an adeno-associated virus, serotype-2 vector carrying glial cell line-derived neurotrophic factor into the bilateral putamina of PD patients. METHODS Thirteen adult patients with advanced PD underwent adeno-associated virus, serotype-2 vector carrying glial cell line-derived neurotrophic factor and gadoteridol (surrogate MRI tracer) coinfusion (450 μL/hemisphere) at escalating doses: 9 × 1010 vg (n = 6); 3 × 1011 vg (n = 6); and 9 × 1011 vg (n = 1). Intraoperative MRI monitored infusion distribution. Patients underwent UPDRS assessment and [18 F]FDOPA-PET scanning preoperatively and 6 and 18 months postoperatively. RESULTS Adeno-associated virus, serotype-2 vector carrying glial cell line-derived neurotrophic factor was tolerated without clinical or radiographic toxicity. Average putaminal coverage was 26%. UPDRS scores remained stable. Ten of thirteen and 12 of 13 patients had increased [18 F]FDOPA Kis at 6 and 18 months postinfusion (increase range: 5-274% and 8-130%; median, 36% and 54%), respectively. Ki differences between baseline and 6- and 18-month follow-up were statistically significant (P < 0.0002). CONCLUSION Adeno-associated virus, serotype-2 vector carrying glial cell line-derived neurotrophic factor infusion was safe and well tolerated. Increased [18 F]FDOPA uptake suggests a neurotrophic effect on dopaminergic neurons. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- John D Heiss
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Codrin Lungu
- Division of Clinical Research, and Office of the Clinical Director, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Dima A Hammoud
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Peter Herscovitch
- Positron Emission Tomography Department, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Debra J Ehrlich
- Parkinson's Disease Clinic, Office of the Clinical Director, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Davis P Argersinger
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Sanhita Sinharay
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Gretchen Scott
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Tianxia Wu
- Clinical Trials Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Howard J Federoff
- Department of Neurology, University of California-Irvine, Irvine, California, USA
| | - Kareem A Zaghloul
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Mark Hallett
- Human Motor Control Section, Medical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Russell R Lonser
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Krystof S Bankiewicz
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA.,Department of Neurological Surgery, University of California-San Francisco, San Francisco, California, USA
| |
Collapse
|
9
|
Penttinen AM, Parkkinen I, Voutilainen MH, Koskela M, Bäck S, Their A, Richie CT, Domanskyi A, Harvey BK, Tuominen RK, Nevalaita L, Saarma M, Airavaara M. Pre-α-pro-GDNF and Pre-β-pro-GDNF Isoforms Are Neuroprotective in the 6-hydroxydopamine Rat Model of Parkinson's Disease. Front Neurol 2018; 9:457. [PMID: 29973907 PMCID: PMC6019446 DOI: 10.3389/fneur.2018.00457] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 05/29/2018] [Indexed: 11/13/2022] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is one of the most studied neurotrophic factors. GDNF has two splice isoforms, full-length pre-α-pro-GDNF (α-GDNF) and pre-β-pro-GDNF (β-GDNF), which has a 26 amino acid deletion in the pro-region. Thus far, studies have focused solely on the α-GDNF isoform, and nothing is known about the in vivo effects of the shorter β-GDNF variant. Here we compare for the first time the effects of overexpressed α-GDNF and β-GDNF in non-lesioned rat striatum and the partial 6-hydroxydopamine lesion model of Parkinson's disease. GDNF isoforms were overexpressed with their native pre-pro-sequences in the striatum using an adeno-associated virus (AAV) vector, and the effects on motor performance and dopaminergic phenotype of the nigrostriatal pathway were assessed. In the non-lesioned striatum, both isoforms increased the density of dopamine transporter-positive fibers at 3 weeks after viral vector delivery. Although both isoforms increased the activity of the animals in cylinder assay, only α-GDNF enhanced the use of contralateral paw. Four weeks later, the striatal tyrosine hydroxylase (TH)-immunoreactivity was decreased in both α-GDNF and β-GDNF treated animals. In the neuroprotection assay, both GDNF splice isoforms increased the number of TH-immunoreactive cells in the substantia nigra but did not promote behavioral recovery based on amphetamine-induced rotation or cylinder assays. Thus, the shorter GDNF isoform, β-GDNF, and the full-length α-isoform have comparable neuroprotective efficacy on dopamine neurons of the nigrostriatal circuitry.
Collapse
Affiliation(s)
- Anna-Maija Penttinen
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Ilmari Parkkinen
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Merja H Voutilainen
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Maryna Koskela
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Susanne Bäck
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Anna Their
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Christopher T Richie
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, United States
| | - Andrii Domanskyi
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Brandon K Harvey
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, United States
| | - Raimo K Tuominen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Liina Nevalaita
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Mikko Airavaara
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
10
|
Treatment with the noradrenaline re-uptake inhibitor atomoxetine alone and in combination with the α2-adrenoceptor antagonist idazoxan attenuates loss of dopamine and associated motor deficits in the LPS inflammatory rat model of Parkinson's disease. Brain Behav Immun 2018; 69:456-469. [PMID: 29339319 DOI: 10.1016/j.bbi.2018.01.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/04/2018] [Accepted: 01/11/2018] [Indexed: 01/14/2023] Open
Abstract
The impact of treatment with the noradrenaline (NA) re-uptake inhibitor atomoxetine and the α2-adrenoceptor (AR) antagonist idazoxan in an animal model of Parkinson's disease (PD) was assessed. Concurrent systemic treatment with atomoxetine and idazoxan, a combination which serves to enhance the extra-synaptic availability of NA, exerts anti-inflammatory and neuroprotective effects following delivery of an inflammatory stimulus, the bacterial endotoxin, lipopolysaccharide (LPS) into the substantia nigra. Lesion-induced deficits in motor function (akinesia, forelimb-use asymmetry) and striatal dopamine (DA) loss were rescued to varying degrees depending on the treatment. Treatment with atomoxetine following LPS-induced lesion to the substantia nigra, yielded a robust anti-inflammatory effect, suppressing microglial activation and expression of the pro-inflammatory cytokine TNF-α whilst increasing the expression of neurotrophic factors. Furthermore atomoxetine treatment prevented loss of tyrosine hydroxylase (TH) positive nigral dopaminergic neurons and resulted in functional improvements in motor behaviours. Atomoxetine alone was sufficient to achieve most of the observed effects. In combination with idazoxan, an additional improvement in the impairment of contralateral limb use 7 days post lesion and a reduction in amphetamine-mediated rotational asymmetry 14 days post-lesion was observed, compared to atomoxetine or idazoxan treatments alone. The results indicate that increases in central NA tone has the propensity to regulate the neuroinflammatory phenotype in vivo and may act as an endogenous neuroprotective mechanism where inflammation contributes to the progression of DA loss. In accordance with this, the clinical use of agents such as NA re-uptake inhibitors and α2-AR antagonists may prove useful in enhancing the endogenous neuroimmunomodulatory potential of NA in conditions associated with brain inflammation.
Collapse
|
11
|
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: 4.0] [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
|
12
|
Tenenbaum L, Humbert-Claude M. Glial Cell Line-Derived Neurotrophic Factor Gene Delivery in Parkinson's Disease: A Delicate Balance between Neuroprotection, Trophic Effects, and Unwanted Compensatory Mechanisms. Front Neuroanat 2017; 11:29. [PMID: 28442998 PMCID: PMC5385337 DOI: 10.3389/fnana.2017.00029] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 03/23/2017] [Indexed: 12/14/2022] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) and Neurturin (NRTN) bind to a receptor complex consisting of a member of the GDNF family receptor (GFR)-α and the Ret tyrosine kinase. Both factors were shown to protect nigro-striatal dopaminergic neurons and reduce motor symptoms when applied terminally in toxin-induced Parkinson's disease (PD) models. However, clinical trials based on intraputaminal GDNF protein administration or recombinant adeno-associated virus (rAAV)-mediated NRTN gene delivery have been disappointing. In this review, several factors that could have limited the clinical benefits are discussed. Retrograde transport of GDNF/NRTN to the dopaminergic neurons soma is thought to be necessary for NRTN/GFR-α/Ret signaling mediating the pro-survival effect. Therefore, the feasibility of treating advanced patients with neurotrophic factors is questioned by recent data showing that: (i) tyrosine hydroxylase-positive putaminal innervation has almost completely disappeared at 5 years post-diagnosis and (ii) in patients enrolled in the rAAV-NRTN trial more than 5 years post-diagnosis, NRTN was almost not transported to the substantia nigra pars compacta. In addition to its anti-apoptotic and neurotrophic properties, GDNF also interferes with dopamine homeostasis via time and dose-dependent effects such as: stimulation of dopamine neuron excitability, inhibition of dopamine transporter activity, tyrosine hydroxylase phosphorylation, and inhibition of tyrosine hydroxylase transcription. Depending on the delivery parameters, the net result of this intricate network of regulations could be either beneficial or deleterious. In conclusion, further unraveling of the mechanism of action of GDNF gene delivery in relevant animal models is still needed to optimize the clinical benefits of this new therapeutic approach. Recent developments in the design of regulated viral vectors will allow to finely adjust the GDNF dose and period of administration. Finally, new clinical studies in less advanced patients are warranted to evaluate the potential of AAV-mediated neurotrophic factors gene delivery in PD. These will be facilitated by the demonstration of the safety of rAAV administration into the human brain.
Collapse
Affiliation(s)
- Liliane Tenenbaum
- Laboratory of Cellular and Molecular Neurotherapies, Clinical Neuroscience Department, Center for Neuroscience Research, Lausanne University HospitalLausanne, Switzerland
| | - Marie Humbert-Claude
- Laboratory of Cellular and Molecular Neurotherapies, Clinical Neuroscience Department, Center for Neuroscience Research, Lausanne University HospitalLausanne, Switzerland
| |
Collapse
|
13
|
GDNF and AADC Gene Therapy for Parkinson’s Disease. Transl Neurosci 2016. [DOI: 10.1007/978-1-4899-7654-3_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
14
|
The transfection of BDNF to dopamine neurons potentiates the effect of dopamine D3 receptor agonist recovering the striatal innervation, dendritic spines and motor behavior in an aged rat model of Parkinson's disease. PLoS One 2015; 10:e0117391. [PMID: 25693197 PMCID: PMC4332861 DOI: 10.1371/journal.pone.0117391] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 12/21/2014] [Indexed: 01/28/2023] Open
Abstract
The progressive degeneration of the dopamine neurons of the pars compacta of substantia nigra and the consequent loss of the dopamine innervation of the striatum leads to the impairment of motor behavior in Parkinson's disease. Accordingly, an efficient therapy of the disease should protect and regenerate the dopamine neurons of the substantia nigra and the dopamine innervation of the striatum. Nigral neurons express Brain Derived Neurotropic Factor (BDNF) and dopamine D3 receptors, both of which protect the dopamine neurons. The chronic activation of dopamine D3 receptors by their agonists, in addition, restores, in part, the dopamine innervation of the striatum. Here we explored whether the over-expression of BDNF by dopamine neurons potentiates the effect of the activation of D3 receptors restoring nigrostriatal innervation. Twelve-month old Wistar rats were unilaterally injected with 6-hydroxydopamine into the striatum. Five months later, rats were treated with the D3 agonist 7-hydroxy-N,N-di-n-propy1-2-aminotetralin (7-OH-DPAT) administered i.p. during 4½ months via osmotic pumps and the BDNF gene transfection into nigral cells using the neurotensin-polyplex nanovector (a non-viral transfection) that selectively transfect the dopamine neurons via the high-affinity neurotensin receptor expressed by these neurons. Two months after the withdrawal of 7-OH-DPAT when rats were aged (24 months old), immunohistochemistry assays were made. The over-expression of BDNF in rats receiving the D3 agonist normalized gait and motor coordination; in addition, it eliminated the muscle rigidity produced by the loss of dopamine. The recovery of motor behavior was associated with the recovery of the nigral neurons, the dopamine innervation of the striatum and of the number of dendritic spines of the striatal neurons. Thus, the over-expression of BDNF in dopamine neurons associated with the chronic activation of the D3 receptors appears to be a promising strategy for restoring dopamine neurons in Parkinson's disease.
Collapse
|
15
|
Gene Therapy for Parkinson’s Disease: AAV5-Mediated Delivery of Glial Cell Line-Derived Neurotrophic Factor (GDNF). NEUROMETHODS 2015. [DOI: 10.1007/978-1-4939-2306-9_3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
|
16
|
Zubedat S, Aga-Mizrachi S, Cymerblit-Sabba A, Ritter A, Nachmani M, Avital A. Methylphenidate and environmental enrichment ameliorate the deleterious effects of prenatal stress on attention functioning. Stress 2015; 18:280-8. [PMID: 25783195 DOI: 10.3109/10253890.2015.1023790] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Either pre- or post-natal environmental factors seem to play a key role in brain and behavioral development and to exert long-term effects. Increasing evidence suggests that exposure to prenatal stress (PS) leads to motor and learning deficits and elevated anxiety, while enriched environment (EE) shows protective effects. The dopaminergic system is also sensitive to environmental life circumstances and affects attention functioning, which serves as the preliminary gate to cognitive processes. However, the effects of methylphenidate (MPH) on the dopaminergic system and attentional functioning, in the context of these life experiences, remain unclear. Therefore, we aimed to examine the effects of EE or PS on distinct types of attention, along with possible effects of MPH exposure. We found that PS impaired selective attention as well as partial sustained attention, while EE had beneficial effects. Both EE and MPH ameliorated the deleterious effects of PS on attention functioning. Considering the possible psychostimulant effect of MPH, we examined both anxiety-like behavior as well as motor learning. We found that PS had a clear anxiogenic effect, whereas EE had an anxiolytic effect. Nevertheless, the treatment with both MPH and/or EE recovered the deleterious effects of PS. In the motor-learning task, the PS group showed superior performance while MPH led to impaired motor learning. Performance decrements were prevented in both the PS + MPH and EE + MPH groups. This study provides evidence that peripubertal exposure to EE (by providing enhanced sensory, motor, and social opportunities) or MPH treatments might be an optional therapeutic intervention in preventing the PS long-term adverse consequences.
Collapse
Affiliation(s)
- Salman Zubedat
- a Behavioral Neuroscience Lab, The Rappaport Faculty of Medicine , Technion - Israel Institute of Technology , Haifa , Israel and
| | - Shlomit Aga-Mizrachi
- a Behavioral Neuroscience Lab, The Rappaport Faculty of Medicine , Technion - Israel Institute of Technology , Haifa , Israel and
| | - Adi Cymerblit-Sabba
- a Behavioral Neuroscience Lab, The Rappaport Faculty of Medicine , Technion - Israel Institute of Technology , Haifa , Israel and
| | - Ami Ritter
- a Behavioral Neuroscience Lab, The Rappaport Faculty of Medicine , Technion - Israel Institute of Technology , Haifa , Israel and
| | - Maayan Nachmani
- a Behavioral Neuroscience Lab, The Rappaport Faculty of Medicine , Technion - Israel Institute of Technology , Haifa , Israel and
| | - Avi Avital
- a Behavioral Neuroscience Lab, The Rappaport Faculty of Medicine , Technion - Israel Institute of Technology , Haifa , Israel and
- b Emek Medical Center , Afula , Israel
| |
Collapse
|
17
|
Tai Y, Chen L, Huang E, Liu C, Yang X, Qiu P, Wang H. Protective effect of alpha-synuclein knockdown on methamphetamine-induced neurotoxicity in dopaminergic neurons. Neural Regen Res 2014; 9:951-8. [PMID: 25206917 PMCID: PMC4146216 DOI: 10.4103/1673-5374.133146] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2014] [Indexed: 12/04/2022] Open
Abstract
The over-expression of α-synuclein is a major factor in the death of dopaminergic neurons in a methamphetamine-induced model of Parkinson's disease. In the present study, α-synuclein knockdown rats were created by injecting α-synuclein-shRNA lentivirus stereotaxically into the right striatum of experimental rats. At 2 weeks post-injection, the rats were injected intraperitoneally with methamphetamine to establish the model of Parkinson's disease. Expression of α-synuclein mRNA and protein in the right striatum of the injected rats was significantly downregulated. Food intake and body weight were greater in α-synuclein knockdown rats, and water intake and stereotyped behavior score were lower than in model rats. Striatal dopamine and tyrosine hydroxylase levels were significantly elevated in α-synuclein knockdown rats. Moreover, superoxide dismutase activity was greater in α-synuclein knockdown rat striatum, but the levels of reactive oxygen species, malondialdehyde, nitric oxide synthase and nitrogen monoxide were lower compared with model rats. We also found that α-synuclein knockdown inhibited methamphetamine-induced neuronal apoptosis. These results suggest that α-synuclein has the capacity to reverse methamphetamine-induced apoptosis of dopaminergic neurons in the rat striatum by inhibiting oxidative stress and improving dopaminergic system function.
Collapse
Affiliation(s)
- Yunchun Tai
- Department of Forensic Medicine, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Ling Chen
- Department of Forensic Medicine, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Enping Huang
- Department of Forensic Medicine, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Chao Liu
- Department of Forensic Medicine, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China ; Guangzhou Forensic Science Institute, Guangzhou, Guangdong Province, China
| | - Xingyi Yang
- Department of Forensic Medicine, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Pingming Qiu
- Department of Forensic Medicine, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Huijun Wang
- Department of Forensic Medicine, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
18
|
Glavaski-Joksimovic A, Bohn MC. Mesenchymal stem cells and neuroregeneration in Parkinson's disease. Exp Neurol 2013; 247:25-38. [DOI: 10.1016/j.expneurol.2013.03.016] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 03/14/2013] [Indexed: 02/06/2023]
|
19
|
Ho DXK, Tan YC, Tan J, Too HP, Ng WH. High-frequency stimulation of the globus pallidus interna nucleus modulates GFRα1 gene expression in the basal ganglia. J Clin Neurosci 2013; 21:657-60. [PMID: 24291478 DOI: 10.1016/j.jocn.2013.05.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 05/26/2013] [Indexed: 01/20/2023]
Abstract
Deep brain stimulation (DBS) is an established therapy for movement disorders such as Parkinson's disease (PD). Although the efficacy of DBS is clear, its precise molecular mechanism remains unknown. The glial cell line derived factor (GDNF) family of ligands has been shown to confer neuroprotective effects on dopaminergic neurons, and putaminal infusion of GDNF have been investigated in PD patients with promising results. Despite the potential therapeutic role of GDNF in alleviating motor symptoms, there is no data on the effects of electrical stimulation on GDNF-family receptor (GFR) expression in the basal ganglia structures. Here, we report the effects of electrical stimulation on GFRα1 isoforms, particularly GFRα1a and GFRα1b. Wistar rats underwent 2 hours of high frequency stimulation (HFS) at the globus pallidus interna nucleus. A control group was subjected to a similar procedure but without stimulation. The HFS group, sacrificed 24 hours after treatment, had a threefold decrease in mRNA expression level of GFRα1b (p=0.037), but the expression level reverted to normal 72 hours after stimulation. Our preliminary data reveal the acute effects of HFS on splice isoforms of GFRα1, and suggest that HFS may modulate the splice isoforms of GFRα1a and GFRα1b to varying degrees. Going forward, elucidating the interactions between HFS and GFR may shed new insights into the complexity of GDNF signaling in the nervous system and lead to better design of clinical trials using these signaling pathways to halt disease progression in PD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Duncun Xun Kiat Ho
- Department of Neurosurgery, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore 308433, Singapore.
| | - Yong Chee Tan
- Department of Neurosurgery, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore 308433, Singapore
| | - Jiayi Tan
- Department of Neurosurgery, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore 308433, Singapore
| | - Heng Phon Too
- Department of Biochemistry, National University of Singapore, Singapore
| | - Wai Hoe Ng
- Department of Neurosurgery, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore 308433, Singapore; Duke-NUS Graduate Medical School, Singapore.
| |
Collapse
|
20
|
Fleming SM, Ekhator OR, Ghisays V. Assessment of sensorimotor function in mouse models of Parkinson's disease. J Vis Exp 2013. [PMID: 23851663 PMCID: PMC3727502 DOI: 10.3791/50303] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Sensitive and reliable behavioral outcome measures are essential to the evaluation of potential therapeutic treatments in preclinical trials for many neurodegenerative diseases. In Parkinson's disease, sensorimotor tests sensitive to varying degrees of nigrostriatal dysfunction are fundamental for testing the efficacy of potential therapeutics. Reliable and quite elegant sensorimotor measures exist for rats, however many of these tests measure sensorimotor asymmetry within the rat and are not entirely suitable for the newer genetic mouse models of PD. We have put together a battery of sensorimotor tests inspired by the sensitive tests in rats and adapted for mice. The test battery highlighted in this study is chosen for a) its sensitivity in a wide variety of mouse models of PD, b) its ease in implementing into a study, and c) its low expense. These tests have proven useful in characterizing novel genetic mouse models of PD as well as in testing potential disease-modifying therapies.
Collapse
|
21
|
Herzog CD, Brown L, Kruegel BR, Wilson A, Tansey MG, Gage FH, Johnson EM, Bartus RT. Enhanced neurotrophic distribution, cell signaling and neuroprotection following substantia nigral versus striatal delivery of AAV2-NRTN (CERE-120). Neurobiol Dis 2013; 58:38-48. [PMID: 23631873 DOI: 10.1016/j.nbd.2013.04.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 03/29/2013] [Accepted: 04/18/2013] [Indexed: 11/18/2022] Open
Abstract
This paper reassesses the currently accepted viewpoint that targeting the terminal fields (i.e. striatum) of degenerating nigrostriatal dopamine neurons with neurotrophic factors in Parkinson's disease (PD) is sufficient for achieving an optimal neurotrophic response. Recent insight indicating that PD is an axonopathy characterized by axonal transport deficits prompted this effort. We tested whether a significantly greater neurotrophic response might be induced in SN neurons when the neurotrophic factor neurturin (NRTN) is also targeted to the substantia nigra (SN), compared to the more conventional, striatum-only target. While recognizing the importance of maintaining the integrity of nigrostriatal fibers and terminals (especially for achieving optimal function), we refocused attention to the fate of SN neurons. Under conditions of axonal degeneration and neuronal transport deficits, this component of the nigrostriatal system is most vulnerable to the lack of neurotrophic exposure following striatal-only delivery. Given the location of repair genes induced by neurotrophic factors, achieving adequate neurotrophic exposure to the SN neurons is essential for an optimal neurotrophic response, while the survival of these neurons is essential to the very survival of the fibers. Two separate studies were performed using the 6-OHDA model of nigrostriatal degeneration, in conjunction with delivery of the viral vector AAV2-NRTN (CERE-120) to continuously express NRTN to either striatum or nigra alone or combined striatal/nigral exposure, including conditions of ongoing axonopathy. These studies provide additional insight for reinterpreting past animal neurotrophic/6-OHDA studies conducted under conditions where axon transport deficiencies were generally not accounted for, which suggested that targeting the striatum was both necessary and sufficient. The current data demonstrate that delivering NRTN directly to the SN produces 1) expanded NRTN distribution within the terminal field and cell bodies of targeted nigrostriatal neurons, 2) enhanced intracellular neurotrophic factor signaling in the nigrostriatal neurons, and 3) produced greater numbers of surviving dopamine neurons against 6-OHDA-induced toxicity, particularly under the conditions of active axonopathy. Thus, these data provide empirical support that targeting the SN with neurotrophic factors (in addition to striatum) may help enhance the neurotrophic response in midbrain neurons, particularly under conditions of active neurodegeneration which occurs in PD patients.
Collapse
|
22
|
Li J, Xia Z, Sun X, Zhang R, Huang G, Hickling R, Xia Z, Hu Y, Zhang Y. Reversal of dopamine neurons and locomotor ability degeneration in aged rats with smilagenin. Neuroscience 2013; 245:90-8. [PMID: 23624370 DOI: 10.1016/j.neuroscience.2013.04.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 04/08/2013] [Accepted: 04/09/2013] [Indexed: 11/19/2022]
Abstract
The purpose of this paper is to study the effect of smilagenin (SMI) (PYM50028), a sapogenin compound originally identified from Chinese medicinal herb, on dopamine neurons and locomotor ability in aged rats. Experiments were carried out on young and aged Sprague-Dawley rats, which were daily administered with either SMI (18mg/kg/day) or vehicle (0.5% sodium carboxymethycellulose [CMCNa]). Open-field and rotarod performance tests revealed that behavioral ability was impaired in aged rats and was improved by oral administration of smilagenin. Immunohistochemical data showed that tyrosine hydroxylase (TH)-positive neuron numbers in the substantia nigra pars compacta (unbiased stereological counting) were altered with aging and were increased by smilagenin treatment. Likewise, the dopamine receptor density and the striatal dopamine transporter (DAT) density ((125)I-2β-carbomethoxy-3β-(4-iodophenyl)-N-(3-fluoropropyl) nortropane [(125)I-FP-CIT] autoradiography) were significantly lowered in aged rats as compared to young rats, and treatment with smilagenin significantly elevated the dopamine receptor and DAT density in aged rats. Furthermore, smilagenin enhances glial cell-derived neurotrophic factor (GDNF) release both in the striatum and midbrain. These results indicate a possible role of smilagenin in the treatment of age-related extrapyramidal disorders.
Collapse
Affiliation(s)
- J Li
- Research Laboratory of Cell Regulation, School of Medicine, Shanghai Jiaotong University, 280 South Chongqing Road, Shanghai 200025, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Antipova V, Hawlitschka A, Mix E, Schmitt O, Dräger D, Benecke R, Wree A. Behavioral and structural effects of unilateral intrastriatal injections of botulinum neurotoxin a in the rat model of Parkinson's disease. J Neurosci Res 2013; 91:838-47. [PMID: 23553727 DOI: 10.1002/jnr.23210] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 10/29/2012] [Accepted: 01/07/2013] [Indexed: 12/25/2022]
Abstract
Botulinum neurotoxin (BoNT) inhibits the release of acetylcholine from presynaptic vesicles through its proteinase activity cleaving the SNARE complex. Parkinson's disease (PD) is associated with locally increased cholinergic activity in the striatum. Therefore, the present study investigates the effect of unilateral intrastriatal BoNT-A injection in naïve rats on striatal morphology; i.e., the total number of Nissl-stained neurons and the volume of caudate-putamen (CPu) were estimated. Furthermore, stainings for markers of gliosis (glial fibrillary acidic protein) and microglia (Iba1) were performed. In addition, the potential beneficial effects of a unilateral intrastriatal injection of BoNT-A on motor activity in the rat model of hemi-PD were evaluated. Hemi-PD was induced by unilateral injection of 6-hydroxydopamine (6-OHDA) into the right medial forebrain bundle. Six weeks later, rats received an ipsilateral intrastriatal injection of BoNT-A. Behaviorally, motor performance was tested. The total number of CPu neurons and the striatal volume were not significantly different between the BoNT-A-injected right and the intact left hemispheres of naïve rats. In hemi-PD rats, intrastriatal BoNT-A abolished apomorphine-induced rotations, increased amphetamine-induced rotations, and tended to improve left forelimb usage. Forced motor function in the accelerod test was not significantly changed by BoNT-A, and open field activity was also unaltered compared with sham treatment. Thus, intrastriatal BoNT-A affects spontaneous motor activity of hemi-PD rats to a minor degree compared with drug-induced motor function. In the future, tests assessing the cognitive and emotional performance should be performed to ascertain finally the potential therapeutic usefulness of intrastriatal BoNT-A for PD.
Collapse
|
24
|
Fleming SM, Schallert T, Ciucci MR. Cranial and related sensorimotor impairments in rodent models of Parkinson's disease. Behav Brain Res 2012; 231:317-22. [PMID: 22394540 DOI: 10.1016/j.bbr.2012.02.034] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Revised: 02/17/2012] [Accepted: 02/20/2012] [Indexed: 12/21/2022]
Abstract
In Parkinson's disease both limb and cranial sensorimotor functions are impaired, leading to a profound diminished quality of life for many patients. Toxin and genetic animal models of Parkinson's disease are likely essential for understanding the pathology associated with these impairments as well as for the development and testing of potential therapeutics. Here we describe useful novel and established behavioral outcome measures for assessing limb and cranial sensorimotor functions in toxin and genetic models of parkinsonism in rats and mice.
Collapse
Affiliation(s)
- Sheila M Fleming
- Departments of Psychology and Neurology, University of Cincinnati, Cincinnati, OH 45221-0376, USA.
| | | | | |
Collapse
|
25
|
Kells AP, Forsayeth J, Bankiewicz KS. Glial-derived neurotrophic factor gene transfer for Parkinson's disease: anterograde distribution of AAV2 vectors in the primate brain. Neurobiol Dis 2011; 48:228-35. [PMID: 22019719 DOI: 10.1016/j.nbd.2011.10.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Revised: 08/26/2011] [Accepted: 10/06/2011] [Indexed: 01/08/2023] Open
Abstract
Delivery of neurotrophic factors to treat neurodegenerative diseases has not been efficacious in clinical trials despite their known potency for promoting neuronal growth and survival. Direct gene delivery to the brain offers an approach for establishing sustained expression of neurotrophic factors but is dependent on accurate surgical procedures to target specific anatomical regions of the brain. Serotype-2 adeno-associated viral (AAV2) vectors have been investigated in multiple clinical studies for neurological diseases without adverse effects; however the absence of significant clinical efficacy after neurotrophic factor gene transfer has been largely attributed to insufficient coverage of the target region. Our pre-clinical development of AAV2-glial-derived neurotrophic factor (GDNF) for Parkinson's disease involved real-time image guided delivery and optimization of delivery techniques to maximize gene transfer in the putamen. We have demonstrated that AAV2 vectors are anterogradely transported in the primate brain with GDNF expression observed in the substantia nigra after putaminal delivery in both intact and nigrostriatal lesioned primates. Direct midbrain delivery of AAV2-GDNF resulted in extensive anterograde transport to multiple brain regions and significant weight loss.
Collapse
Affiliation(s)
- Adrian P Kells
- University of California San Francisco, Department of Neurological Surgery, Box 0555, San Francisco, CA 94143, USA
| | | | | |
Collapse
|
26
|
Bartus RT, Brown L, Wilson A, Kruegel B, Siffert J, Johnson EM, Kordower JH, Herzog CD. Properly scaled and targeted AAV2-NRTN (neurturin) to the substantia nigra is safe, effective and causes no weight loss: support for nigral targeting in Parkinson's disease. Neurobiol Dis 2011; 44:38-52. [PMID: 21704161 DOI: 10.1016/j.nbd.2011.05.026] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Revised: 05/06/2011] [Accepted: 05/28/2011] [Indexed: 10/18/2022] Open
Abstract
Recent analyses of autopsied brains from subjects previously administered AAV2-neurturin (NRTN) gene transfer argues that optimizing the effects of neurotrophic factors in Parkinson's disease (PD) likely requires delivery to both the degenerating cell bodies (in substantia nigra) and their terminals (in striatum). Prior to implementing this novel dosing paradigm in humans, we conducted eight nonclinical experiments with three general objectives: (1) evaluate the feasibility, safety and effectiveness of targeting the substantia nigra (SN) with AAV2-NRTN, (2) better understand and appraise recent warnings of serious weight loss that might occur with targeting the SN with neurotrophic factors, and (3) define an appropriate dose of AAV2-NRTN that should safely and effectively cover the SN in PD patients. Toward these ends, we first determined SN volume for rats, monkeys and humans, and employed these values to calculate comparable dose equivalents for each species by scaling each dose, based on relative SN volume. Using this information, we next injected AAV2-GFP to monkey SN to quantify AAV2-vector distribution and confirm reasonable SN coverage. We then selected and administered a ~200-fold range of AAV2-NRTN doses (and a single AAV2-GDNF dose) to rat SN, producing a wide range of protein expression. In contrast to recent warnings regarding nigra targeting, no dose produced any serious side effects or toxicity, though we replicated the modest reduction in weight gain reported by others with the highest AAV2-NRTN and the AAV2-GDNF dose. A dose-related increase in NRTN expression was seen, with the lower doses limiting NRTN to the peri-SN and the highest dose producing mistargeted NRTN well outside the SN. We then demonstrated that the reduction in weight gain following excessive-doses can be dissociated from NRTN in the targeted SN, and is linked to mistargeted NRTN in the diencephalon. We also showed that prior destruction of the dopaminergic SN neurons via 6-OHDA had no impact on the weight loss phenomenon, further dissociating neurotrophic exposure to the SN as the culprit for weight changes. Finally, low AAV2-NRTN doses provided significant neuroprotection against 6-OHDA toxicity, establishing a wide therapeutic index for nigral targeting. These data support targeting the SN with AAV2-NRTN in PD patients, demonstrating that properly targeted and scaled AAV2-NRTN provides safe and effective NRTN expression. They also provided the means to define an appropriate human-equivalent dose for proceeding into an ongoing clinical trial, using empirically-based scaling to account for marked differences in SN volume between species.
Collapse
|
27
|
Degeorge ML, Marlowe D, Werner E, Soderstrom KE, Stock M, Mueller A, Bohn MC, Kozlowski DA. Combining glial cell line-derived neurotrophic factor gene delivery (AdGDNF) with L-arginine decreases contusion size but not behavioral deficits after traumatic brain injury. Brain Res 2011; 1403:45-56. [PMID: 21672665 DOI: 10.1016/j.brainres.2011.05.058] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 05/20/2011] [Accepted: 05/25/2011] [Indexed: 01/09/2023]
Abstract
Our laboratory has previously demonstrated that viral administration of glial cell line-derived neurotrophic factor (AdGDNF), one week prior to a controlled cortical impact (CCI) over the forelimb sensorimotor cortex of the rat (FL-SMC) is neuroprotective, but does not significantly enhance recovery of sensorimotor function. One possible explanation for this discrepancy is that although protected, neurons may not have been functional due to enduring metabolic deficiencies. Additionally, metabolic events following TBI may interfere with expression of therapeutic proteins administered to the injured brain via gene therapy. The current study focused on enhancing the metabolic function of the brain by increasing cerebral blood flow (CBF) with l-arginine in conjunction with administration of AdGDNF immediately following CCI. An adenoviral vector harboring human GDNF was injected unilaterally into FL-SMC of the rat immediately following a unilateral CCI over the FL-SMC. Within 30min of the CCI and AdGDNF injections, some animals were injected with l-arginine (i.v.). Tests of forelimb function and asymmetry were administered for 4weeks post-injury. Animals were sacrificed and contusion size and GDNF protein expression measured. This study demonstrated that rats treated with AdGDNF and l-arginine post-CCI had a significantly smaller contusion than injured rats who did not receive any treatment, or injured rats treated with either AdGDNF or l-arginine alone. Nevertheless, no amelioration of behavioral deficits was seen. These findings suggest that AdGDNF alone following a CCI was not therapeutic and although combining it with l-arginine decreased contusion size, it did not enhance behavioral recovery.
Collapse
Affiliation(s)
- M L Degeorge
- DePaul University, Department of Biological Sciences, Chicago, IL 60614, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Neurotrophic factors for the treatment of Parkinson's disease. Cytokine Growth Factor Rev 2011; 22:157-65. [DOI: 10.1016/j.cytogfr.2011.05.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Accepted: 05/20/2011] [Indexed: 11/20/2022]
|
29
|
Wakeman DR, Dodiya HB, Kordower JH. Cell transplantation and gene therapy in Parkinson's disease. ACTA ACUST UNITED AC 2011; 78:126-58. [PMID: 21259269 DOI: 10.1002/msj.20233] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Parkinson's disease is a progressive neurodegenerative disorder affecting, in part, dopaminergic motor neurons of the ventral midbrain and their terminal projections that course to the striatum. Symptomatic strategies focused on dopamine replacement have proven effective at remediating some motor symptoms during the course of disease but ultimately fail to deliver long-term disease modification and lose effectiveness due to the emergence of side effects. Several strategies have been experimentally tested as alternatives for Parkinson's disease, including direct cell replacement and gene transfer through viral vectors. Cellular transplantation of dopamine-secreting cells was hypothesized as a substitute for pharmacotherapy to directly provide dopamine, whereas gene therapy has primarily focused on restoration of dopamine synthesis or neuroprotection and restoration of spared host dopaminergic circuitry through trophic factors as a means to enhance sustained controlled dopamine transmission. This seems now to have been verified in numerous studies in rodents and nonhuman primates, which have shown that grafts of fetal dopamine neurons or gene transfer through viral vector delivery can lead to improvements in biochemical and behavioral indices of dopamine deficiency. However, in clinical studies, the improvements in parkinsonism have been rather modest and variable and have been plagued by graft-induced dyskinesias. New developments in stem-cell transplantation and induced patient-derived cells have opened the doors for the advancement of cell-based therapeutics. In addition, viral-vector-derived therapies have been developed preclinically with excellent safety and efficacy profiles, showing promise in clinical trials thus far. Further progress and optimization of these therapies will be necessary to ensure safety and efficacy before widespread clinical use is deemed appropriate.
Collapse
|
30
|
Khodr CE, Sapru MK, Pedapati J, Han Y, West NC, Kells AP, Bankiewicz KS, Bohn MC. An α-synuclein AAV gene silencing vector ameliorates a behavioral deficit in a rat model of Parkinson's disease, but displays toxicity in dopamine neurons. Brain Res 2011; 1395:94-107. [PMID: 21565333 DOI: 10.1016/j.brainres.2011.04.036] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Accepted: 04/19/2011] [Indexed: 12/16/2022]
Abstract
Effects of silencing ectopically expressed hSNCA in rat substantia nigra (SN) were examined as a novel therapeutic approach to Parkinson's disease (PD). AAV-hSNCA with or without an AAV harboring a short-hairpin (sh)RNA targeting hSNCA or luciferase was injected into one SN. At 9weeks, hSNCA-expressing rats had reduced SN dopamine (DA) neurons and exhibited a forelimb deficit. AAV-shRNA-SNCA silenced hSNCA and protected against the forelimb deficit. However, AAV-shRNA-SNCA also led to DA neuron loss suggesting undesirable effects of chronic shRNA expression. Effects on nigrostriatal-projecting neurons were examined using a retrograde tract tracer. Loss of striatal-projecting DA neurons was evident in the vector injection site, whereas DA neurons outside this site were lost in hSNCA-expressing rats, but not in hSNCA-silenced rats. These observations suggest that high levels of shRNA-SNCA were toxic to DA neurons, while neighboring neurons exposed to lower levels were protected by hSNCA gene silencing. Also, data collected on DA levels suggest that neurons other than or in addition to nigrostriatal DA neurons contributed to protection of forelimb use. Our observations suggest that while hSNCA gene silencing in DA neurons holds promise as a novel PD therapy, further development of silencing technology is required.
Collapse
Affiliation(s)
- Christina E Khodr
- Department of Pediatrics, Neurobiology Program, Children's Memorial Research Center, Feinberg School of Medicine, Northwestern University, 2300 Children's Plaza, Box 209, Chicago, IL 60614, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Glavaski-Joksimovic A, Virag T, Mangatu TA, McGrogan M, Wang XS, Bohn MC. Glial cell line-derived neurotrophic factor-secreting genetically modified human bone marrow-derived mesenchymal stem cells promote recovery in a rat model of Parkinson's disease. J Neurosci Res 2011; 88:2669-81. [PMID: 20544825 DOI: 10.1002/jnr.22435] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by progressive degeneration of nigrostriatal dopaminergic (DA) neurons. The therapeutic potential of glial cell line-derived neurotrophic factor (GDNF), the most potent neurotrophic factor for DA neurons, has been demonstrated in many experimental models of PD. However, chronic delivery of GDNF to DA neurons in the brain remains an unmet challenge. Here, we report the effects of GDNF-releasing Notch-induced human bone marrow-derived mesenchymal stem cells (MSC) grafted into striatum of the 6-hydroxydopamine (6-OHDA) progressively lesioned rat model of PD. Human MSC, obtained from bone marrow aspirates of young, healthy adult volunteers, were transiently transfected with the intracellular domain of the Notch1 gene (NICD) to generate SB623 cells. SB623 cells expressing GDNF and/or humanized Renilla green fluorescent protein (hrGFP) following lentiviral transduction or nontransduced cells were stereotaxically placed into rat striatum 1 week after a unilateral partial 6-OHDA striatal lesion. At 4 weeks, rats that had received GDNF-transduced SB623 cells had significantly decreased amphetamine-induced rotation compared with control rats, although this effect was not observed in rats that received GFP-transduced or nontransduced SB623 cells. At 5 weeks, rejuvenated tyrosine hydroxylase-immunoreactive (TH-IR) fibers that appeared to be host DA axons were observed in and around grafts. This effect was more prominent in rats that received GDNF-secreting cells and was not observed in controls. These observations suggest that human bone-marrow derived MSC, genetically modified to secrete GDNF, hold potential as an allogeneic or autologous stem cell therapy for PD.
Collapse
Affiliation(s)
- Aleksandra Glavaski-Joksimovic
- Department of Pediatrics, Neurobiology Program, Children's Memorial Research Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60614, USA
| | | | | | | | | | | |
Collapse
|
32
|
Voutilainen MH, Bäck S, Peränen J, Lindholm P, Raasmaja A, Männistö PT, Saarma M, Tuominen RK. Chronic infusion of CDNF prevents 6-OHDA-induced deficits in a rat model of Parkinson's disease. Exp Neurol 2010; 228:99-108. [PMID: 21185834 DOI: 10.1016/j.expneurol.2010.12.013] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Revised: 12/10/2010] [Accepted: 12/12/2010] [Indexed: 10/18/2022]
Abstract
Cerebral dopamine neurotrophic factor (CDNF) and mesencephalic astrocyte-derived neurotrophic factor (MANF) constitute a novel, evolutionarily conserved family of neurotrophic factors (NTF) expressed in vertebrates and invertebrates. The effects of two-week infusions of CDNF, MANF and glial cell line-derived neurotrophic factor (GDNF) were studied in a rat 6-hydroxydopamine (6-OHDA) hemiparkinsonian model. Degeneration of nigrostriatal dopamine nerve tract after toxin injection was assessed by measuring amphetamine-induced rotational behavior, and at the end of the experiment by quantifying tyrosine hydroxylase (TH)-positive neurons in the substantia nigra pars compacta (SNpc) and TH-positive fibers in the striatum. The diffusion of the NTFs into the brain tissue following chronic infusion was also studied. Finally, we examined the transportation of intrastriatally injected (125)I-CDNF within the brain. The amphetamine-induced rotational behavior was gradually normalized in rats treated with CDNF for two weeks following the intrastriatal 6-OHDA injection. CDNF was also able to inhibit 6-OHDA-induced loss of TH-immunoreactive cells of the SNpc and TH-positive fibers in the striatum. MANF and GDNF had no statistically significant effect in any of the above measures. The volume of distribution for MANF in the striatum was significantly larger than that of GDNF after 3-day infusions. Both (125)I-CDNF and (125)I-GDNF were retrogradely transported from the striatum to the SN. No behavioral signs of toxicity were observed during treatment with the three NTFs. These results imply that CDNF may have potential as a neuroprotective or even neurorestorative therapy of PD.
Collapse
Affiliation(s)
- Merja H Voutilainen
- Division of Pharmacology and Toxicology, Faculty of Pharmacy, P.O. Box 56, Viikki Biocenter, University of Helsinki, FI-00014, Helsinki, Finland
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Morel GR, Sosa YE, Bellini MJ, Carri NG, Rodriguez SS, Bohn MC, Goya RG. Glial cell line-derived neurotrophic factor gene therapy ameliorates chronic hyperprolactinemia in senile rats. Neuroscience 2010; 167:946-53. [PMID: 20219648 PMCID: PMC2854198 DOI: 10.1016/j.neuroscience.2010.02.053] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Revised: 02/02/2010] [Accepted: 02/19/2010] [Indexed: 11/17/2022]
Abstract
Progressive dysfunction of hypothalamic tuberoinfundibular dopaminergic (TIDA) neurons during normal aging is associated in the female rat with chronic hyperprolactinemia. We assessed the effectiveness of glial cell line-derived neurotrophic factor (GDNF) gene therapy to restore TIDA neuron function in senile female rats and reverse their chronic hyperprolactinemia. Young (2.5 months) and senile (29 months) rats received a bilateral intrahypothalamic injection (10(10) pfu) of either an adenoviral vector expressing the gene for beta-galactosidase; (Y-betagal and S-betagal, respectively) or a vector expressing rat GDNF (Y-GDNF and S-GDNF, respectively). Transgenic GDNF levels in supernatants of GDNF adenovector-transduced N2a neuronal cell cultures were 25+/-4 ng/ml, as determined by bioassay. In the rats, serum prolactin (PRL) was measured at regular intervals. On day 17 animals were sacrificed and neuronal nuclear antigen (NeuN) and tyrosine hydroxylase (TH) immunoreactive cells counted in the arcuate-periventricular hypothalamic region. The S-GDNF but not the S-betagal rats, showed a significant reduction in body weight. The chronic hyperprolactinemia of the senile females was significantly ameliorated in the S-GDNF rats (P<0.05) but not in the S-betagal rats. Neither age nor GDNF induced significant changes in the number of NeuN and TH neurons. We conclude that transgenic GDNF ameliorates chronic hyperprolactinemia in aging female rats, probably by restoring TIDA neuron function.
Collapse
Affiliation(s)
- Gustavo R. Morel
- INIBIOLP-CONICET-Histology B, School of Medicine, University of La Plata, Argentina
| | - Yolanda E. Sosa
- INIBIOLP-CONICET-Histology B, School of Medicine, University of La Plata, Argentina
| | - Maria J. Bellini
- INIBIOLP-CONICET-Histology B, School of Medicine, University of La Plata, Argentina
| | | | - Silvia S. Rodriguez
- INIBIOLP-CONICET-Histology B, School of Medicine, University of La Plata, Argentina
| | - Martha C. Bohn
- Children’s Memorial Research Center (CMRC), Northwestern University , Chicago, IL
| | - Rodolfo G. Goya
- INIBIOLP-CONICET-Histology B, School of Medicine, University of La Plata, Argentina
| |
Collapse
|
34
|
Abstract
Parkinson's disease (PD) is a chronic, progressive neurodegenerative movement disorder for which there is currently no effective therapy. Over the past several decades, there has been a considerable interest in neuroprotective therapies using trophic factors to alleviate the symptoms of PD. Neurotrophic factors (NTFs) are a class of molecules that influence a number of neuronal functions, including cell survival and axonal growth. Experimental studies in animal models suggest that members of neurotrophin family and GDNF family of ligands (GFLs) have the potent ability to protect degenerating dopamine neurons as well as promote regeneration of the nigrostriatal dopamine system. In clinical trials, although no serious adverse events related to the NTF therapy has been reported in patients, they remain inconclusive. In this chapter, we attempt to give a brief overview on several different growth factors that have been explored for use in animal models of PD and those already used in PD patients.
Collapse
|
35
|
Lim ST, Airavaara M, Harvey BK. Viral vectors for neurotrophic factor delivery: a gene therapy approach for neurodegenerative diseases of the CNS. Pharmacol Res 2009; 61:14-26. [PMID: 19840853 DOI: 10.1016/j.phrs.2009.10.002] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2009] [Revised: 10/11/2009] [Accepted: 10/11/2009] [Indexed: 01/11/2023]
Abstract
The clinical manifestation of most diseases of the central nervous system results from neuronal dysfunction or loss. Diseases such as stroke, epilepsy and neurodegeneration (e.g. Alzheimer's disease and Parkinson's disease) share common cellular and molecular mechanisms (e.g. oxidative stress, endoplasmic reticulum stress, mitochondrial dysfunction) that contribute to the loss of neuronal function. Neurotrophic factors (NTFs) are secreted proteins that regulate multiple aspects of neuronal development including neuronal maintenance, survival, axonal growth and synaptic plasticity. These properties of NTFs make them likely candidates for preventing neurodegeneration and promoting neuroregeneration. One approach to delivering NTFs to diseased cells is through viral vector-mediated gene delivery. Viral vectors are now routinely used as tools for studying gene function as well as developing gene-based therapies for a variety of diseases. Currently, many clinical trials using viral vectors in the nervous system are underway or completed, and seven of these trials involve NTFs for neurodegeneration. In this review, we discuss viral vector-mediated gene transfer of NTFs to treat neurodegenerative diseases of the central nervous system.
Collapse
Affiliation(s)
- Seung T Lim
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057, United States
| | | | | |
Collapse
|
36
|
Mijatovic J, Piltonen M, Alberton P, Männistö PT, Saarma M, Piepponen TP. Constitutive Ret signaling is protective for dopaminergic cell bodies but not for axonal terminals. Neurobiol Aging 2009; 32:1486-94. [PMID: 19767128 DOI: 10.1016/j.neurobiolaging.2009.08.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2009] [Revised: 08/03/2009] [Accepted: 08/22/2009] [Indexed: 12/27/2022]
Abstract
Ret is the canonical signaling receptor for glial cell line-derived neurotrophic factor (GDNF), which has been shown to have neuroprotective effects when administered prior to neurotoxic challenge. A missense Meth918Thr mutation causes the constitutive activation of Ret, resulting in multiple endocrine neoplasia type 2 B (MEN2B). To clarify the role of Ret signaling in neuroprotection, we studied the effects of the neurotoxins 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and 6-hydroxydopamine (6-OHDA) on the dopaminergic system of mice carrying the MEN2B mutation. We found that MEN2B mice were significantly more resistant to nigral tyrosine hydroxylase (TH)-positive cell loss induced by unilateral striatal 6-OHDA than Wt mice. However, 6-OHDA caused profound dopamine (DA) depletion in the striatum of both MEN2B and Wt mice. Systemic MPTP caused similar DA depletion and a decrease in TH-immunostaining in the striatum of MEN2B and Wt mice. Neither neurotoxin induced a compensatory increase in striatal metabolite/DA ratios in the MEN2B mice, possibly contributing to an increased amphetamine-induced turning behavior observed in behavioral assessments of these mice. Thus, our data suggest that activated Ret protects DA cell bodies in the substantia nigra pars compacta, but does not protect DA axons in the striatum.
Collapse
Affiliation(s)
- Jelena Mijatovic
- Division of Pharmacology and Toxicology, Faculty of Pharmacy, University of Helsinki, Finland.
| | | | | | | | | | | |
Collapse
|
37
|
Glavaski-Joksimovic A, Virag T, Chang QA, West NC, Mangatu TA, McGrogan MP, Dugich-Djordjevic M, Bohn MC. Reversal of Dopaminergic Degeneration in a Parkinsonian Rat following Micrografting of Human Bone Marrow-Derived Neural Progenitors. Cell Transplant 2009; 18:801-14. [DOI: 10.3727/096368909x470801] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease characterized by the selective loss of dopaminergic (DA) neurons in the midbrain. Various types of stem cells that have potential to differentiate into DA neurons are being investigated as cellular therapies for PD. Stem cells also secrete growth factors and therefore also may have therapeutic effects in promoting the health of diseased DA neurons in the PD brain. To address this possibility in an experimental model of PD, bone marrow-derived neuroprogenitor-like cells were generated from bone marrow procured from healthy human adult volunteers and their potential to elicit recovery of damaged DA axons was studied in a partial lesion rat model of PD. Following collection of bone marrow, mesenchymal stem cells (MSC) were isolated and then genetically modified to create SB623 cells by transient transfection with the intracellular domain of the Notch1 gene (NICD), a modification that upregulates expression of certain neuroprogenitor markers. Ten deposits of 0.5 μl of SB623 cell suspension adjusted from 6,000 to 21,000 cells/μl in PBS or PBS alone were stereotaxically placed in the striatum 1 week after the nigrostriatal projection had been partially lesioned in adult F344 rats by injection of 6-hydroxydopamine (6-OHDA) into the striatum. At 3 weeks, a small number of grafted SB623 cells survived in the lesioned striatum as visualized by expression of the human specific nuclear matrix protein (hNuMA). In rats that received SB623 cells, but not in control rats, dense tyrosine hydroxylase immunoreactive (TH-ir) fibers were observed around the grafts. These fibers appeared to be rejuvenated host DA axons because no TH-ir in soma of surviving SB623 cells or coexpression of TH and hNuMA-ir were observed. In addition, dense serotonin immunoreactive (5-HT-ir) fibers were observed around grafted SB623 cells and these fibers also appeared to be of the host origin. Also, in some SB623 grafted rats that were sacrificed within 2 h of dl-amphetamine injection, hot spots of c-Fos-positive nuclei that coincided with rejuvenated dense TH fibers around the grafted SB623 cells were observed, suggesting increased availability of DA in these locations. Our observations suggest that NICD-transfected MSC hold potential as a readily available autologous or allogenic cellular therapy for ameliorating the degeneration of DA and 5-HT neurons in PD patients.
Collapse
Affiliation(s)
- Aleksandra Glavaski-Joksimovic
- Department of Pediatrics, Neurobiology Program, Children's Memorial Research Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Tamas Virag
- Department of Pediatrics, Neurobiology Program, Children's Memorial Research Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Qin A. Chang
- Department of Pediatrics, Neurobiology Program, Children's Memorial Research Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Neva C. West
- Department of Pediatrics, Neurobiology Program, Children's Memorial Research Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Thomas A. Mangatu
- Department of Pediatrics, Neurobiology Program, Children's Memorial Research Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | | | - Martha C. Bohn
- Department of Pediatrics, Neurobiology Program, Children's Memorial Research Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
38
|
Louboutin JP, Agrawal L, Reyes BAS, Van Bockstaele EJ, Strayer DS. HIV-1 gp120 neurotoxicity proximally and at a distance from the point of exposure: protection by rSV40 delivery of antioxidant enzymes. Neurobiol Dis 2009; 34:462-76. [PMID: 19327399 DOI: 10.1016/j.nbd.2009.03.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2008] [Revised: 02/26/2009] [Accepted: 03/04/2009] [Indexed: 01/21/2023] Open
Abstract
Toxicity of HIV-1 envelope glycoprotein (gp120) for substantia nigra (SN) neurons may contribute to the Parkinsonian manifestations often seen in HIV-1-associated dementia (HAD). We studied the neurotoxicity of gp120 for dopaminergic neurons and potential neuroprotection by antioxidant gene delivery. Rats were injected stereotaxically into their caudate-putamen (CP); CP and (substantia nigra) SN neuron loss was quantified. The area of neuron loss extended several millimeters from the injection site, approximately 35% of the CP area. SN neurons, outside of this area of direct neurotoxicity, were also severely affected. Dopaminergic SN neurons (expressing tyrosine hydroxylase, TH, in the SN and dopamine transporter, DAT, in the CP) were mostly affected: intra-CP gp120 caused approximately 50% DAT+ SN neuron loss. Prior intra-CP gene delivery of Cu/Zn superoxide dismutase (SOD1) or glutathione peroxidase (GPx1) protected SN neurons from intra-CP gp120. Thus, SN dopaminergic neurons are highly sensitive to HIV-1 gp120-induced neurotoxicity, and antioxidant gene delivery, even at a distance, is protective.
Collapse
Affiliation(s)
- Jean-Pierre Louboutin
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | | | | | | | | |
Collapse
|
39
|
Scientific rationale for the development of gene therapy strategies for Parkinson's disease. Biochim Biophys Acta Mol Basis Dis 2009; 1792:703-13. [PMID: 19254760 DOI: 10.1016/j.bbadis.2009.02.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2008] [Revised: 02/16/2009] [Accepted: 02/18/2009] [Indexed: 12/31/2022]
Abstract
The ever-evolving understanding of the neuronal systems involved in Parkinson's disease together with the recent advances in recombinant viral vector technology has led to the development of several gene therapy applications that are now entering into clinical testing phase. To date, four fundamentally different approaches have been pursued utilizing recombinant adeno-associated virus and lentiviruses as vectors for delivery. These strategies aim either to restore the lost brain functions by substitution of enzymes critical for synthesis of neurotransmitters or neurotrophic factors as a means to boost the function of remaining neurons in the diseased brain. In this review we discuss the differences in mechanism of action and describe the scientific rationale behind the currently tested gene therapy approaches for Parkinson's disease in some detail and pinpoint their individual unique strengths and weaknesses.
Collapse
|
40
|
Isacson O, Kordower JH. Future of cell and gene therapies for Parkinson's disease. Ann Neurol 2009; 64 Suppl 2:S122-38. [PMID: 19127583 DOI: 10.1002/ana.21473] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The experimental field of restorative neurology continues to advance with implantation of cells or transfer of genes to treat patients with neurological disease. Both strategies have generated a consensus that demonstrates their capacity for structural and molecular brain modification in the adult brain. However, both approaches have yet to successfully address the complexities to make such novel therapeutic modalities work in the clinic. Prior experimental cell transplantation to patients with PD utilized dissected pieces of fetal midbrain tissue, containing mixtures of cells and neuronal types, as donor cells. Stem cell and progenitor cell biology provide new opportunities for selection and development of large batches of specific therapeutic cells. This may allow for cell composition analysis and dosing to optimize the benefit to an individual patient. The biotechnology used for cell and gene therapy for treatment of neurological disease may eventually be as advanced as today's pharmaceutical drug-related design processes. Current gene therapy phase 1 safety trials for PD include the delivery of a growth factor (neurturin via the glial cell line-derived neurotrophic factor receptor) and a transmitter enzyme (glutamic acid decarboxylase and aromatic acid decarboxylase). Many new insights from cell biological and molecular studies provide opportunities to selectively express or suppress factors relevant to neuroprotection and improved function of neurons involved in PD. Future gene and cell therapies are likely to coexist with classic pharmacological therapies because their use can be tailored to individual patients' underlying disease process and need for neuroprotective or restorative interventions.
Collapse
Affiliation(s)
- Ole Isacson
- Department of Neurology (Neuroscience), Center for Neuroregeneration Research and National Institute of Neurological Disorders and Stroke Udall Parkinson's Disease Research Center of Excellence, Harvard Medical School/McLean Hospital, Belmont, MA, USA
| | | |
Collapse
|
41
|
Behavioral outcome measures for the assessment of sensorimotor function in animal models of movement disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2009; 89:57-65. [PMID: 19900615 DOI: 10.1016/s0074-7742(09)89003-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Animal models have and continue to contribute to our understanding of the neurobiology many types of disorders. In movement disorders such as Parkinson's disease (PD), animal models have directly led to various therapeutic treatments such as deep brain stimulation. To facilitate the development of potential therapeutics, sensitive and reliable outcome measures in animal models are necessary to maximize their benefit. In this chapter, behavioral outcome measures, sensitive to varying degrees of sensorimotor dysfunction, are reviewed in rats and mice.
Collapse
|
42
|
Abstract
With an increase in the aging population, the incidence of Parkinson's disease (PD), a disabling neurodegenerative disorder mainly affecting motor function, will inevitably present a challenge to an already overburdened healthcare system. Current medical and surgical therapies offer symptomatic relief but do not provide a cure. Experimental studies suggest that GDNF has the ability to protect degenerating dopamine neurons in PD as well as promote regeneration of the nigrostriatal dopamine system. However, clinical trials of GDNF infusion to date remain inconclusive. This review will examine the experimental and clinical evidence of GDNF use in PD with particular focus on its potential as an effective therapy in the treatment of PD.
Collapse
Affiliation(s)
- Murray Hong
- Cell Restoration Laboratory, Departments of Anatomy & Neurobiology & Surgery (Neurosurgery), Dalhousie University, Room 12H1, 5850 College Street, Halifax, Nova Scotia, Canada B3H 1X5
| | | | | |
Collapse
|
43
|
Pertusa M, García-Matas S, Mammeri H, Adell A, Rodrigo T, Mallet J, Cristòfol R, Sarkis C, Sanfeliu C. Expression of GDNF transgene in astrocytes improves cognitive deficits in aged rats. Neurobiol Aging 2008; 29:1366-79. [PMID: 17399854 DOI: 10.1016/j.neurobiolaging.2007.02.026] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2006] [Revised: 02/14/2007] [Accepted: 02/20/2007] [Indexed: 01/25/2023]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) was assayed for its neurotrophic effects against the neuronal atrophy that causes cognitive deficits in old age. Aged Fisher 344 rats with impairment in the Morris water maze received intrahippocampal injections at the dorsal CA1 area of either a lentiviral vector encoding human GDNF or the same vector encoding human green fluorescent protein as a control. Recombinant lentiviral vectors constructed with human cytomegalovirus promotor and pseudotyped with lyssavirus Mokola glycoprotein specifically transduced the astrocytes in vivo. Astrocyte-secreted GDNF enhanced neuron function as shown by local increases in synthesis of the neurotransmitters acetylcholine, dopamine and serotonin. This neurotrophic effect led to cognitive improvement of the rats as early as 2 weeks after gene transduction. Spatial learning and memory testing showed a significant gain in cognitive abilities due to GDNF exposure, whereas control-transduced rats kept their performance at the chance level. These results confirm the broad spectrum of the neurotrophic action of GDNF and open new gene therapy possibilities for reducing age-related neurodegeneration.
Collapse
Affiliation(s)
- M Pertusa
- Departament de Farmacologia i Toxicologia, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC-IDIBAPS, Rosselló 161, 08036 Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Manciocco A, Chiarotti F, Vitale A, Calamandrei G, Laviola G, Alleva E. The application of Russell and Burch 3R principle in rodent models of neurodegenerative disease: the case of Parkinson's disease. Neurosci Biobehav Rev 2008; 33:18-32. [PMID: 18771685 DOI: 10.1016/j.neubiorev.2008.08.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2007] [Revised: 07/10/2008] [Accepted: 08/07/2008] [Indexed: 12/21/2022]
Abstract
Currently, the accepted ethical standards for the regulation of animal experimentation are provided by the 3R principle (Replacement, Reduction and Refinement). The development of alternative methods to the use of animals (Replacement), the design of adequate experimental protocols to reduce the number of animals (Reduction), the application of refinement practices (Refinement) are all aspects to be considered to ensure ethical and scientific validity to animal experimentation. This review intends to address these issues, using experimental research on Parkinson's disease (PD) as a paradigmatic example of the use of animal models to improve knowledge on a devastating human pathology. In particular, current rodent models of PD and their validity are reviewed and discussed, and methodologies that may ultimately reduce animal's suffering emphasized. Although procedures referring to with 3R principle can be traced in the literature reviewed, they are not considered yet an important part of the methodological information. The formal inclusion in scientific papers of a section devoted to 3Rs may increase knowledge and eventually adherence to this principle by scientists.
Collapse
Affiliation(s)
- Arianna Manciocco
- Section of Behavioral Neuroscience, Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Rome I-00161, Italy
| | | | | | | | | | | |
Collapse
|
45
|
Emerging restorative treatments for Parkinson's disease. Prog Neurobiol 2008; 85:407-32. [PMID: 18586376 DOI: 10.1016/j.pneurobio.2008.05.001] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2007] [Revised: 04/03/2008] [Accepted: 05/06/2008] [Indexed: 01/18/2023]
Abstract
Several exciting approaches for restorative therapy in Parkinson's disease have emerged over the past two decades. This review initially describes experimental and clinical data regarding growth factor administration. We focus on glial cell line-derived neurotrophic factor (GDNF), particularly its role in neuroprotection and in regeneration in Parkinson's disease. Thereafter, we discuss the challenges currently facing cell transplantation in Parkinson's disease and briefly consider the possibility to continue testing intrastriatal transplantation of fetal dopaminergic progenitors clinically. We also give a more detailed overview of the developmental biology of dopaminergic neurons and the potential of certain stem cells, i.e. neural and embryonic stem cells, to differentiate into dopaminergic neurons. Finally, we discuss adult neurogenesis as a potential tool for restoring lost dopamine neurons in patients suffering from Parkinson's disease.
Collapse
|
46
|
Aponso P, Faull R, Connor B. Increased progenitor cell proliferation and astrogenesis in the partial progressive 6-hydroxydopamine model of Parkinson’s disease. Neuroscience 2008; 151:1142-53. [DOI: 10.1016/j.neuroscience.2007.11.036] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2007] [Revised: 11/20/2007] [Accepted: 11/28/2007] [Indexed: 02/01/2023]
|
47
|
RET signaling does not modulate MPTP toxicity but is required for regeneration of dopaminergic axon terminals. Proc Natl Acad Sci U S A 2007; 104:20049-54. [PMID: 18056810 DOI: 10.1073/pnas.0706177104] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Activation of the RET (rearranged during transfection) receptor by glial cell-line-derived neurotrophic factor (GDNF) has been identified as an important differentiation and survival factor for dopaminergic neurons of the midbrain in preclinical experiments. These encouraging results have led to clinical trials of GDNF in patients with Parkinson's disease, which have resulted in conflicting findings. To investigate the potential benefit of Ret-dependent signaling on the challenged dopaminergic system, we tested the effect of tissue-selective ablation of the Ret gene on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) toxicity in mice, the most widely used animal model for Parkinson's disease. Ablation of Ret did not modify the MPTP-induced loss of dopaminergic neurons in the substantia nigra pars compacta and the dopaminergic innervation of the striatum at 14 days. However, Ret ablation abolished the regeneration of dopaminergic fibers and terminals, as well as the partial recovery of striatal dopamine concentrations, that was observed in control mice between days 14 and 90 after MPTP treatment. We therefore conclude that RET signaling has no influence on the survival of dopaminergic neurons in the MPTP model of Parkinson's disease but rather facilitates the regeneration of dopaminergic axon terminals.
Collapse
|
48
|
Maguire-Zeiss KA, Mhyre TR, Federoff HJ. Gazing into the future: Parkinson's disease gene therapeutics to modify natural history. Exp Neurol 2007; 209:101-13. [PMID: 18035353 DOI: 10.1016/j.expneurol.2007.09.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2007] [Revised: 09/19/2007] [Accepted: 09/24/2007] [Indexed: 12/21/2022]
Abstract
PD gene therapy clinical trials have primarily focused on increasing the production of dopamine (DA) through supplemental amino acid decarboxylase (AADC) expression, neurotrophic support for surviving dopaminergic neurons (DAN) or altering brain circuitry to compensate for DA neuron loss. The future of PD gene therapy will depend upon resolving a number of important issues that are discussed in this special issue. Of particular importance is the identification of novel targets that are amenable to early intervention prior to the substantial loss of DAN. However, for the most part the etiopathogenesis of PD is unknown making early intervention a challenge and the development of early biomarker diagnostics imperative.
Collapse
|
49
|
Borgal L, Hong M, Sadi D, Mendez I. Differential effects of glial cell line-derived neurotrophic factor on A9 and A10 dopamine neuron survival in vitro. Neuroscience 2007; 147:712-9. [PMID: 17583436 DOI: 10.1016/j.neuroscience.2007.03.057] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2006] [Revised: 03/08/2007] [Accepted: 03/30/2007] [Indexed: 10/23/2022]
Abstract
Glial cell-line derived neurotrophic factor (GDNF) enhances dopamine (DA) cell survival and fiber outgrowth, and may be beneficial in enhancing cell restorative strategies for Parkinson's disease (PD). However, GDNF may have different roles for transplanted DA cell sub-types. The present in vitro study investigated the effect of GDNF on the survival of rat DA cells displaying a phenotype consistent with either the substantia nigra [A9 cells immunopositive for tyrosine hydroxylase (TH) and G-protein-gated inwardly rectifying potassium channel subunit 2 (GIRK2)] or with the ventral tegmental area [A10 cells immunopositive for TH and calbindin]. It was found that a single exposure of GDNF enhanced the number of DA cells of an A9 phenotype, without affecting DA cells of an A10 phenotype. Conversely, repeated GDNF exposure did not alter the survival of A9 phenotypic cells, but doubled the percentage of A10 cells. It was concluded that GDNF administration may affect dopaminergic cells differently depending on time and degree of GDNF exposure. For cell transplantation in PD, long-term GDNF administration may result in detrimental effects for transplanted A9 TH+ cells as this may introduce competition with A10 TH+ cells for survival and fiber outgrowth into the host striatum. These results may have important implications for clinical neural transplantation in PD.
Collapse
Affiliation(s)
- L Borgal
- Cell Restoration Laboratory, Brain Repair Centre, Dalhousie University, Halifax, NS, Canada
| | | | | | | |
Collapse
|
50
|
Fleming SM, Chesselet MF. Behavioral phenotypes and pharmacology in genetic mouse models of Parkinsonism. Behav Pharmacol 2007; 17:383-91. [PMID: 16940759 DOI: 10.1097/00008877-200609000-00004] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Prior to the discovery of genes associated with familial forms of Parkinson's disease, animal models of Parkinson's disease mainly consisted of toxin models based exclusively on the degeneration of nigrostriatal dopamine neurons. These traditional models have provided valuable insight into symptomatic treatments for Parkinson's disease; however, they lack the broad extra-nigral pathology and the progression that is observed in the disease. The novel genetic mouse models recently generated are advantageous because they have mutations that are known to cause familial Parkinson's disease and thus they have good construct validity. To maximize the utility of these models, a thoughtful phenotypical characterization is important. Our laboratory has assembled a battery of behavioral tests to assess sensorimotor function in genetic mouse models of Parkinsonism. This review discusses the sensitivity of these tests in different genetic mice in addition to their behavioral response to dopamine agonists.
Collapse
Affiliation(s)
- Sheila M Fleming
- Department of Neurology, The David Geffen School of Medicine at UCLA, Los Angeles, California 90095-1769, USA
| | | |
Collapse
|