1
|
Mousavi MA, Rezaei M, Pourhamzeh M, Salari M, Hossein-Khannazer N, Shpichka A, Nabavi SM, Timashev P, Vosough M. Translational Approach using Advanced Therapy Medicinal Products for Huntington's Disease. Curr Rev Clin Exp Pharmacol 2025; 20:14-31. [PMID: 38797903 DOI: 10.2174/0127724328300166240510071548] [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: 12/22/2023] [Revised: 04/02/2024] [Accepted: 04/22/2024] [Indexed: 05/29/2024]
Abstract
Current therapeutic approaches for Huntington's disease (HD) focus on symptomatic treatment. Therefore, the unavailability of efficient disease-modifying medicines is a significant challenge. Regarding the molecular etiology, targeting the mutant gene or advanced translational steps could be considered promising strategies. The evidence in gene therapy suggests various molecular techniques, including knocking down mHTT expression using antisense oligonucleotides and small interfering RNAs and gene editing with zinc finger proteins and CRISPR-Cas9-based techniques. Several post-transcriptional and post-translational modifications have also been proposed. However, the efficacy and long-term side effects of these modalities have yet to be verified. Currently, cell therapy can be employed in combination with conventional treatment and could be used for HD in which the structural and functional restoration of degenerated neurons can occur. Several animal models have been established recently to develop cell-based therapies using renewable cell sources such as embryonic stem cells, induced pluripotent stem cells, mesenchymal stromal cells, and neural stem cells. These models face numerous challenges in translation into clinics. Nevertheless, investigations in Advanced Therapy Medicinal Products (ATMPs) open a promising window for HD research and their clinical application. In this study, the ATMPs entry pathway in HD management was highlighted, and their advantages and disadvantages were discussed.
Collapse
Affiliation(s)
- Maryam Alsadat Mousavi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Maliheh Rezaei
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahsa Pourhamzeh
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Departments of Pathology and Medicine, UC San Diego, La Jolla, CA, USA
| | - Mehri Salari
- Department of Neurology, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Nikoo Hossein-Khannazer
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Anastasia Shpichka
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare," Sechenov University, Moscow, Russia
| | - Seyed Massood Nabavi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare," Sechenov University, Moscow, Russia
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, 141-83 Stockholm, Sweden
| |
Collapse
|
2
|
Iguchi Y, Fukabori R, Kato S, Takahashi K, Eifuku S, Maejima Y, Shimomura K, Mizuma H, Mawatari A, Doi H, Cui Y, Onoe H, Hikishima K, Osanai M, Nishijo T, Momiyama T, Benton R, Kobayashi K. Chemogenetic activation of mammalian brain neurons expressing insect Ionotropic Receptors by systemic ligand precursor administration. Commun Biol 2024; 7:547. [PMID: 38714803 PMCID: PMC11076466 DOI: 10.1038/s42003-024-06223-4] [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: 10/03/2023] [Accepted: 04/22/2024] [Indexed: 05/10/2024] Open
Abstract
Chemogenetic approaches employing ligand-gated ion channels are advantageous regarding manipulation of target neuronal population functions independently of endogenous second messenger pathways. Among them, Ionotropic Receptor (IR)-mediated neuronal activation (IRNA) allows stimulation of mammalian neurons that heterologously express members of the insect chemosensory IR repertoire in response to their cognate ligands. In the original protocol, phenylacetic acid, a ligand of the IR84a/IR8a complex, was locally injected into a brain region due to its low permeability of the blood-brain barrier. To circumvent this invasive injection, we sought to develop a strategy of peripheral administration with a precursor of phenylacetic acid, phenylacetic acid methyl ester, which is efficiently transferred into the brain and converted to the mature ligand by endogenous esterase activities. This strategy was validated by electrophysiological, biochemical, brain-imaging, and behavioral analyses, demonstrating high utility of systemic IRNA technology in the remote activation of target neurons in the brain.
Collapse
Affiliation(s)
- Yoshio Iguchi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Ryoji Fukabori
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Shigeki Kato
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Kazumi Takahashi
- Department of Systems Neuroscience, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Satoshi Eifuku
- Department of Systems Neuroscience, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Yuko Maejima
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Kenju Shimomura
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Hiroshi Mizuma
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Aya Mawatari
- Laboratory for Labeling Chemistry, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Hisashi Doi
- Laboratory for Labeling Chemistry, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
- Research, Institute for Drug Discovery Science, Collaborative Creation Research Center, Organization for Research Promotion, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai, 599-8531, Japan
| | - Yilong Cui
- Laboratory for Biofunction Dynamics Imaging, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Hirotaka Onoe
- Human Brain Research Center, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-Cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Keigo Hikishima
- Medical Devices Research Group, Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-2-1 Namiki, Tsukuba, 305-8564, Japan
| | - Makoto Osanai
- Department of Medical Physics and Engineering, Division of Health Sciences, Osaka University Graduate School of Medicine, 1-7 Yamadaoka, Suita, 565-0871, Japan
| | - Takuma Nishijo
- Department of Pharmacology, Jikei University School of Medicine, 3-25-8 Nishi-shinbashi, Tokyo, 105-8461, Japan
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya-cho, Kasugai, 480-0392, Japan
| | - Toshihiko Momiyama
- Department of Pharmacology, Jikei University School of Medicine, 3-25-8 Nishi-shinbashi, Tokyo, 105-8461, Japan
| | - Richard Benton
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, CH-1015, Lausanne, Switzerland
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan.
| |
Collapse
|
3
|
Binda CS, Lelos MJ, Rosser AE, Massey TH. Using gene or cell therapies to treat Huntington's disease. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:193-215. [PMID: 39341655 DOI: 10.1016/b978-0-323-90120-8.00014-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Huntington's disease is caused by a CAG repeat expansion in the first exon of the HTT gene, leading to the production of gain-of-toxic-function mutant huntingtin protein species and consequent transcriptional dysregulation and disrupted cell metabolism. The brunt of the disease process is borne by the striatum from the earliest disease stages, with striatal atrophy beginning approximately a decade prior to the onset of neurologic signs. Although the expanded CAG repeat in the HTT gene is necessary and sufficient to cause HD, other genes can influence the age at onset of symptoms and how they progress. Many of these modifier genes have roles in DNA repair and are likely to modulate the stability of the CAG repeat in somatic cells. Currently, there are no disease-modifying treatments for HD that can be prescribed to patients and few symptomatic treatments, but there is a lot of interest in therapeutics that can target the pathogenic pathways at the DNA and RNA levels, some of which have reached the stage of human studies. In contrast, cell therapies aim to replace key neural cells lost to the disease process and/or to support the host vulnerable striatum by direct delivery of cells to the brain. Ultimately it may be possible to combine gene and cell therapies to both slow disease processes and provide some level of neural repair. In this chapter we consider the current status of these therapeutic strategies along with their prospects and challenges.
Collapse
Affiliation(s)
- Caroline S Binda
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom; UK Dementia Research Institute at Cardiff, Cardiff University, Cardiff, United Kingdom
| | - Mariah J Lelos
- Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Anne E Rosser
- Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, United Kingdom; BRAIN Unit, Neuroscience and Mental Health Research Institute, Cardiff, United Kingdom.
| | - Thomas H Massey
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom; UK Dementia Research Institute at Cardiff, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
4
|
Barbuti PA, Barker RA, Brundin P, Przedborski S, Papa SM, Kalia LV, Mochizuki H. Recent Advances in the Development of Stem-Cell-Derived Dopaminergic Neuronal Transplant Therapies for Parkinson's Disease. Mov Disord 2021; 36:1772-1780. [PMID: 33963552 DOI: 10.1002/mds.28628] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 12/16/2022] Open
Abstract
The last decade has seen exciting advances in the development of potential stem cell-based therapies for Parkinson's disease (PD), which have used different types of stem cells as starting material. These cells have been developed primarily to replace dopamine-producing neurons in the substantia nigra that are progressively lost in the disease process. The aim is to largely restore lost motor functions, whilst not ever being curative. We discuss cell-based strategies that will have to fulfill important criteria to become effective and competitive therapies for PD. These criteria include reproducibly producing sufficient numbers of cells with an authentic substantia nigra dopamine neuron A9 phenotype, which can integrate into the host brain after transplantation and form synapses (considered crucial for long-term functional benefits). Furthermore, it is essential that transplanted cells exhibit no, or only very low levels of, proliferation without tumor formation at the site of grafting. Cumulative research has shown that stem cell-based approaches continue to have great potential in PD, but key questions remain to be answered. Here, we review the most recent progress in research on stem cell-based dopamine neuron replacement therapy for PD and briefly discuss what the immediate future might hold. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Peter A Barbuti
- Departments of Neurology, Pathology and Cell Biology, and Neuroscience, Columbia University, New York, New York, USA
| | - Roger A Barker
- Department of Clinical Neuroscience and WT-MRC Cambridge Stem Cell Institute, University of Cambridge and Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Patrik Brundin
- Van Andel Institute, Center for Parkinson's Disease, Department of Neurodegenerative Science, Grand Rapids, Michigan, USA
| | - Serge Przedborski
- Departments of Neurology, Pathology and Cell Biology, and Neuroscience, Columbia University, New York, New York, USA
| | - Stella M Papa
- Yerkes National Primate Research Center and Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Lorraine V Kalia
- Division of Neurology, Department of Medicine, Morton and Gloria Shulman Movement Disorders Clinic and the Edmond J. Safra Program in Parkinson's Disease, Toronto Western Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Hideki Mochizuki
- Department of Neurology, Osaka University Graduate School of Medicine, Osaka, Japan
| | | |
Collapse
|
5
|
Monk R, Connor B. Cell Replacement Therapy for Huntington's Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1266:57-69. [PMID: 33105495 DOI: 10.1007/978-981-15-4370-8_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disorder which is characterised by a triad of highly debilitating motor, cognitive, and psychiatric symptoms. While cell death occurs in many brain regions, GABAergic medium spiny neurons (MSNs) in the striatum experience preferential and extensive degeneration. Unlike most neurodegenerative disorders, HD is caused by a single genetic mutation resulting in a CAG repeat expansion and the production of a mutant Huntingtin protein (mHTT). Despite identifying the mutation causative of HD in 1993, there are currently no disease-modifying treatments for HD. One potential strategy for the treatment of HD is the development of cell-based therapies. Cell-based therapies aim to restore neuronal circuitry and function by replacing lost neurons, as well as providing neurotropic support to prevent further degeneration. In order to successfully restore basal ganglia functioning in HD, cell-based therapies would need to reconstitute the complex signalling network disrupted by extensive MSN degeneration. This chapter will discuss the potential use of foetal tissue grafts, pluripotent stem cells, neural stem cells, and somatic cell reprogramming to develop cell-based therapies for treating HD.
Collapse
Affiliation(s)
- Ruth Monk
- Department of Pharmacology and Clinical Pharmacology, Centre for Brain Research, SMS, FMHS, University of Auckland, Auckland, New Zealand
| | - Bronwen Connor
- Department of Pharmacology and Clinical Pharmacology, Centre for Brain Research, SMS, FMHS, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
6
|
Stahl K, Rahmani S, Prydz A, Skauli N, MacAulay N, Mylonakou MN, Torp R, Skare Ø, Berg T, Leergaard TB, Paulsen RE, Ottersen OP, Amiry-Moghaddam M. Targeted deletion of the aquaglyceroporin AQP9 is protective in a mouse model of Parkinson's disease. PLoS One 2018; 13:e0194896. [PMID: 29566083 PMCID: PMC5864064 DOI: 10.1371/journal.pone.0194896] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 03/12/2018] [Indexed: 12/21/2022] Open
Abstract
More than 90% of the cases of Parkinson’s disease have unknown etiology. Gradual loss of dopaminergic neurons of substantia nigra is the main cause of morbidity in this disease. External factors such as environmental toxins are believed to play a role in the cell loss, although the cause of the selective vulnerability of dopaminergic neurons remains unknown. We have previously shown that aquaglyceroporin AQP9 is expressed in dopaminergic neurons and astrocytes of rodent brain. AQP9 is permeable to a broad spectrum of substrates including purines, pyrimidines, and lactate, in addition to water and glycerol. Here we test our hypothesis that AQP9 serves as an influx route for exogenous toxins and, hence, may contribute to the selective vulnerability of nigral dopaminergic (tyrosine hydroxylase-positive) neurons. Using Xenopus oocytes injected with Aqp9 cRNA, we show that AQP9 is permeable to the parkinsonogenic toxin 1-methyl-4-phenylpyridinium (MPP+). Stable expression of AQP9 in HEK cells increases their vulnerability to MPP+ and to arsenite—another parkinsonogenic toxin. Conversely, targeted deletion of Aqp9 in mice protects nigral dopaminergic neurons against MPP+ toxicity. A protective effect of Aqp9 deletion was demonstrated in organotypic slice cultures of mouse midbrain exposed to MPP+in vitro and in mice subjected to intrastriatal injections of MPP+in vivo. Seven days after intrastriatal MPP+ injections, the population of tyrosine hydroxylase-positive cells in substantia nigra is reduced by 48% in Aqp9 knockout mice compared with 67% in WT littermates. Our results show that AQP9 –selectively expressed in catecholaminergic neurons—is permeable to MPP+ and suggest that this aquaglyceroporin contributes to the selective vulnerability of nigral dopaminergic neurons by providing an entry route for parkinsonogenic toxins. To our knowledge this is the first evidence implicating a toxin permeable membrane channel in the pathophysiology of Parkinson’s disease.
Collapse
MESH Headings
- 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacokinetics
- Animals
- Aquaporins/genetics
- Disease Models, Animal
- Dopaminergic Neurons/drug effects
- Dopaminergic Neurons/metabolism
- Female
- Gene Deletion
- HEK293 Cells
- Humans
- MPTP Poisoning/genetics
- MPTP Poisoning/metabolism
- MPTP Poisoning/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mutagenesis, Site-Directed
- Neuroprotection/genetics
- Neuroprotective Agents/metabolism
- Parkinson Disease/genetics
- Parkinson Disease/pathology
- Parkinson Disease, Secondary/chemically induced
- Parkinson Disease, Secondary/genetics
- Parkinson Disease, Secondary/metabolism
- Parkinson Disease, Secondary/pathology
- Substantia Nigra/drug effects
- Substantia Nigra/metabolism
- Xenopus laevis
Collapse
Affiliation(s)
- Katja Stahl
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Soulmaz Rahmani
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Agnete Prydz
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Nadia Skauli
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Nanna MacAulay
- Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maria N. Mylonakou
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, Norway Biotechnology Centre, University of Oslo, Oslo, Norway
| | - Reidun Torp
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Øivind Skare
- Department of Occupational Medicine and Epidemiology, National Institute of Occupational Health, Oslo, Norway
| | - Torill Berg
- Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Trygve B. Leergaard
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Ragnhild E. Paulsen
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Ole P. Ottersen
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Karolinska Institutet, Stockholm, Sweden
| | - Mahmood Amiry-Moghaddam
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- * E-mail:
| |
Collapse
|
7
|
Sun AX, Tan EK. Towards better cellular replacement therapies in Parkinson disease. J Neurosci Res 2017; 96:219-221. [PMID: 28791710 DOI: 10.1002/jnr.24123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 07/04/2017] [Accepted: 07/05/2017] [Indexed: 11/06/2022]
Affiliation(s)
- Alfred Xuyang Sun
- National Neuroscience Institute, Singapore General Hospital, Duke NUS Medical School, Genome Institute of Singapore, Singapore
| | - Eng-King Tan
- National Neuroscience Institute, Singapore General Hospital, Duke NUS Medical School, Genome Institute of Singapore, Singapore
| |
Collapse
|
8
|
Fricker RA, Barker RA, Fawcett JW, Dunnett SB. A Comparative Study of Preparation Techniques for Improving the Viability of Striatal Grafts Using Vital Stains, in Vitro Cultures, and in Vivo Grafts. Cell Transplant 2017; 5:599-611. [PMID: 8951218 DOI: 10.1177/096368979600500603] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Cell suspension grafts from embryonic striatal primordia placed into the adult rat striatum survive well and are able to alleviate a number of behavioral deficits caused by excitotoxic lesions to this structure. However, neither the anatomical connectivity between the graft and host nor the functional recovery elicited by the grafts is completely restored. One way in which the survival and function of embryonic striatal grafts may be enhanced is by the improvement of techniques for the preparation of the cell suspension prior to implantation, an issue that has been addressed only to a limited extent. We have evaluated a number of parameters during the preparation procedure, looking at the effects on cell survival over the first 24 h from preparation using vital dyes and the numbers of surviving neurons in vitro, after 4 days in culture, in addition to graft survival and function in vivo. Factors influencing cell survival include the type of trypsinization procedure and the age of donor tissues used for suspension preparation. The presence of DNase has no effect on cell viability but aids the dissociation of the tissue to form single cells. These results have important implications for the use of embryonic striatal grafts in animal models of Huntington's disease, and in any future clinical application of this research.
Collapse
Affiliation(s)
- R A Fricker
- Department of Experimental Psychology, University of Cambridge, UK
| | | | | | | |
Collapse
|
9
|
Emerich DF, Ragozzino M, Lehman MN, Sanberg PR. Behavioral Effects of Neural Transplantation. Cell Transplant 2017; 1:401-27. [PMID: 1344313 DOI: 10.1177/096368979200100604] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Considerable evidence suggests that transplantation of fetal neural tissue ameliorates the behavioral deficits observed in a variety of animal models of CNS disorders. However, it is also becoming increasingly clear that neural transplants do not necessarily produce behavioral recovery, and in some cases have either no beneficial effects, magnify existing behavioral abnormalities, or even produce a unique constellation of deficits. Regardless, studies demonstrating the successful use of neural transplants in reducing or eliminating behavioral deficits in these animal models has led directly to their clinical application in human neurodegenerative disorders such as Parkinson's disease. This review examines the beneficial and deleterious behavioral consequences of neural transplants in different animal models of human diseases, and discusses the possible mechanisms by which neural transplants might produce behavior recovery.
Collapse
Affiliation(s)
- D F Emerich
- Cyto Therapeutics, Inc., Providence, RI 02906
| | | | | | | |
Collapse
|
10
|
Abstract
Neural transplantation has been extensively applied in Parkinson's disease, including numerous clinical studies, studies in animal models, and related basic research on cell biology. There is evidence that the clinical trials of both adrenal medulla transplantation and fetal substantia nigra transplantation have produced a detectable clinical effect, although it is not yet clear whether the clinical benefit is sufficient to justify a more widespread application of these procedures. Studies of long-term outcome and quantitative tests are important in assaying the degree of benefit produced by transplantation procedures in Parkinson's disease and for developing improved and refined procedures. Other disease-related applications of neural transplantation are beginning to be developed. These include Huntington's disease, chronic pain, epilepsy, spinal cord injury, and perhaps even demyelinating diseases and cortical ischemic injury. Although most of these applications lie in the future, it is not too soon to begin to consider the scientific justification that should be required for initiation of human clinical trials.
Collapse
Affiliation(s)
- William J. Freed
- Preclinical Neurosciences Section, Neuropsychiatry Branch, NIMH Neuroscience Center at St. Elizabeths, 2700 Martin Luther King Ave., Washington, DC 20032, USA
| |
Collapse
|
11
|
Fricker-Gates RA, Muir JA, Dunnett SB. Transplanted hNT Cells (“LBS Neurons”) in a Rat Model of Huntington's Disease: Good Survival, Incomplete Differentiation, and Limited Functional Recovery. Cell Transplant 2017; 13:123-36. [PMID: 15129758 DOI: 10.3727/000000004773301807] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
A variety of immortalized cell lines have been proposed to exhibit sufficient phenotypic plasticity to allow them to replace primary embryonic neurons for restorative cell transplantation. In the present experiments we evaluate the functional viability of one particular cell line, the hNT cells developed by Layton Bioscience, to replace lost neurons and alleviate asymmetrical motor deficits in a unilateral excitotoxic lesion model of Huntington's disease. Because the grafts involved implantation of human-derived cells into a rat host environment, all animals were immunosuppressed. Cyclosporin A and FK-506 were similar in providing effective immunoprotection of the hNT xenografts, and whereas the lesions induced a marked inflammatory response in the host brain, this was not exacerbated by the presence of xenograft cells. The presence of grafted cells was determined with the human-specific antigen HuNu, and good graft survival was demonstrated in almost all animals up to the longest survival examined, 16 weeks posttransplantation. Although the cells exhibited progressively greater maturation and differentiation at 10-day, 4- and 16-week time points, staining for the mature neuronal marker NeuN was at best very weak, and we were unable to detect unequivocal staining with any markers of mature striatal phenotype, including DARPP-32, calbindin, parvalbumin, choline acetyl transferase, or NADPH diaphorase (with in all cases positive control provided by good staining on the intact contralateral side of the brain). Nor were we able to detect any differences between rats with lesions alone and rats with grafts in the contralateral motor deficits exhibited in a test of skilled paw reaching or cylinder placing. These results suggest that further and more extensive studies should be undertaken to assess whether hNT neurons can show more extensive and appropriate maturation and be associated with recovery in appropriate behavioral models, before they may be considered a suitable replacement for primary embryonic cells for clinical application in Huntington's disease.
Collapse
|
12
|
Dunnett SB, Björklund A. Mechanisms and use of neural transplants for brain repair. PROGRESS IN BRAIN RESEARCH 2017; 230:1-51. [PMID: 28552225 DOI: 10.1016/bs.pbr.2016.11.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Under appropriate conditions, neural tissues transplanted into the adult mammalian brain can survive, integrate, and function so as to influence the behavior of the host, opening the prospect of repairing neuronal damage, and alleviating symptoms associated with neuronal injury or neurodegenerative disease. Alternative mechanisms of action have been postulated: nonspecific effects of surgery; neurotrophic and neuroprotective influences on disease progression and host plasticity; diffuse or locally regulated pharmacological delivery of deficient neurochemicals, neurotransmitters, or neurohormones; restitution of the neuronal and glial environment necessary for proper host neuronal support and processing; promoting local and long-distance host and graft axon growth; formation of reciprocal connections and reconstruction of local circuits within the host brain; and up to full integration and reconstruction of fully functional host neuronal networks. Analysis of neural transplants in a broad range of anatomical systems and disease models, on simple and complex classes of behavioral function and information processing, have indicated that all of these alternative mechanisms are likely to contribute in different circumstances. Thus, there is not a single or typical mode of graft function; rather grafts can and do function in multiple ways, specific to each particular context. Consequently, to develop an effective cell-based therapy, multiple dimensions must be considered: the target disease pathogenesis; the neurodegenerative basis of each type of physiological dysfunction or behavioral symptom; the nature of the repair required to alleviate or remediate the functional impairments of particular clinical relevance; and identification of a suitable cell source or delivery system, along with the site and method of implantation, that can achieve the sought for repair and recovery.
Collapse
|
13
|
Abstract
Over the past three decades, significant progress has been made in the development of potential regenerative cell-based therapies for neurodegenerative disease, with most success being seen in Parkinson's disease. Cell-based therapies face many challenges including ethical considerations, potential for immune-mediated rejection with allogeneic and xenogeneic tissue, pathological spread of protein-related disease into the grafted tissue as well as the risk of graft overgrowth and tumorigenesis in stem cell-derived transplants. Preclinical trials have looked at many tissue types of which the most successful to date have been those using fetal ventral mesencephalon grafts, which led to clinical trials, which have shown that in some cases they can work very well. With important proof-of-concept derived from these studies, there is now much interest in how dopaminergic neurons derived from stem cell sources could be used to develop cell-based therapies suitable for clinical use, with clinical trials poised to enter the clinic in the next couple of years.
Collapse
Affiliation(s)
- Thomas B Stoker
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Cambridge, CB2 0PY, UK.,Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge
| | - Roger A Barker
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Cambridge, CB2 0PY, UK.,Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge
| |
Collapse
|
14
|
Andres RH, Wallimann T, Widmer HR. Creatine supplementation improves neural progenitor cell survival in Huntington's disease. Brain Circ 2016; 2:133-137. [PMID: 30276289 PMCID: PMC6126277 DOI: 10.4103/2394-8108.192519] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 08/30/2016] [Accepted: 09/01/2016] [Indexed: 01/30/2023] Open
Abstract
Preclinical and clinical studies suggest that striatal transplantation of neural stem cells (NSCs) and neural progenitor cells (NPCs) may be an appealing and valuable system for treating Huntington's disease. Nevertheless, for a neural replacement to become an effective translational treatment for Huntington's disease, a certain number of difficulties must be addressed, including how to improve the integration of transplanted cell grafts with the host tissue, to elevate the survival rates of transplanted cells, and to ensure their directed differentiation into specific neuronal phenotypes. Research focusing on the translational applications of creatine (Cr) supplementation in NSC and NPC cell replacement therapies continues to offer promising results, pointing to Cr as a factor with the potential to improve cell graft survivability and encourage differentiation toward GABAergic phenotypes in models of striatal transplantation. Here, we evaluate research examining the outcomes of Cr supplementation and how the timing of supplementation regimes may affect their efficacy. The recent studies indicate that Cr's effects vary according to the developmental stage of the cells being treated, noting the dynamic differences in creatine kinase expression over the developmental stages of differentiating NPCs. This research continues to move Cr supplementation closer to the widespread clinical application and suggests such techniques warrant further examination.
Collapse
Affiliation(s)
- Robert H Andres
- Department of Neurosurgery, University of Berne, Inselspital, CH-3010 Berne, Switzerland
| | - Theo Wallimann
- Department of Biology, Institute of Cell Biology, Swiss Federal Institute of Technology (ETH), CH-8093 Zurich, Switzerland
| | - Hans R Widmer
- Department of Neurosurgery, University of Berne, Inselspital, CH-3010 Berne, Switzerland
| |
Collapse
|
15
|
Andres RH, Ducray AD, Andereggen L, Hohl T, Schlattner U, Wallimann T, Widmer HR. The effects of creatine supplementation on striatal neural progenitor cells depend on developmental stage. Amino Acids 2016; 48:1913-27. [PMID: 27129463 DOI: 10.1007/s00726-016-2238-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 04/12/2016] [Indexed: 01/14/2023]
Abstract
Transplantation of neural progenitor cells (NPCs) is a promising experimental therapy for Huntington's disease (HD). The variables responsible for the success of this approach, including selection of the optimal developmental stage of the grafted cells, are however largely unknown. Supporting cellular energy metabolism by creatine (Cr) supplementation is a clinically translatable method for improving cell transplantation strategies. The present study aims at investigating differences between early (E14) and late (E18) developmental stages of rat striatal NPCs in vitro. NPCs were isolated from E14 and E18 embryos and cultured for 7 days with or without Cr [5 mM]. Chronic treatment significantly increased the percentage of GABA-immunoreactive neurons as compared to untreated controls, both in the E14 (170.4 ± 4.7 %) and the E18 groups (129.3 ± 9.3 %). This effect was greater in E14 cultures (p < 0.05). Similarly, short-term treatment for 24 h resulted in increased induction (p < 0.05) of the GABA-ergic phenotype in E14 (163.0 ± 10.4 %), compared to E18 cultures (133.3 ± 9.5 %). Total neuronal cell numbers and general viability were not affected by Cr (p > 0.05). Protective effects of Cr against a metabolic insult were equal in E14 and E18 NPCs (p > 0.05). Cr exposure promoted morphological differentiation of GABA-ergic neurons, including neurite length in both groups (p < 0.05), but the number of branching points was increased only in the E18 group (p < 0.05). Our results demonstrate that the role of Cr as a GABA-ergic differentiation factor depends on the developmental stage of striatal NPCs, while Cr-mediated neuroprotection is not significantly influenced. These findings have potential implications for optimizing future cell replacement strategies in HD.
Collapse
Affiliation(s)
- Robert H Andres
- Department of Neurosurgery, University of Berne, Inselspital, Freiburgstrasse 10, 3010, Berne, Switzerland
| | - Angelique D Ducray
- Department of Neurosurgery, University of Berne, Inselspital, Freiburgstrasse 10, 3010, Berne, Switzerland
| | - Lukas Andereggen
- Department of Neurosurgery, University of Berne, Inselspital, Freiburgstrasse 10, 3010, Berne, Switzerland.,Department of Neurosurgery and F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Tabea Hohl
- Department of Neurosurgery, University of Berne, Inselspital, Freiburgstrasse 10, 3010, Berne, Switzerland
| | - Uwe Schlattner
- Laboratory of Fundamental and Applied Bioenergetics, Université Grenoble Alpes, BP53, 38041, Grenoble Cedex, France.,Inserm, U1055, BP53, 38041, Grenoble Cedex, France
| | - Theo Wallimann
- Professor emeritus, formerly at Institute of Cell Biology, Swiss Federal Institute of Technology (ETH), Schafmattstrasse 18, 8093, Zurich, Switzerland
| | - Hans R Widmer
- Department of Neurosurgery, University of Berne, Inselspital, Freiburgstrasse 10, 3010, Berne, Switzerland.
| |
Collapse
|
16
|
Reddington AE, Rosser AE, Dunnett SB. Differentiation of pluripotent stem cells into striatal projection neurons: a pure MSN fate may not be sufficient. Front Cell Neurosci 2014; 8:398. [PMID: 25520619 PMCID: PMC4251433 DOI: 10.3389/fncel.2014.00398] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 11/05/2014] [Indexed: 11/13/2022] Open
Abstract
Huntington's disease (HD) is an autosomal dominant inherited disorder leading to the loss inter alia of DARPP-32 positive medium spiny projection neurons ("MSNs") in the striatum. There is no known cure for HD but the relative specificity of cell loss early in the disease has made cell replacement by neural transplantation an attractive therapeutic possibility. Transplantation of human fetal striatal precursor cells has shown "proof-of-principle" in clinical trials; however, the practical and ethical difficulties associated with sourcing fetal tissues have stimulated the need to identify alternative source(s) of donor cells that are more readily available and more suitable for standardization. We now have available the first generation of protocols to generate DARPP-32 positive MSN-like neurons from pluripotent stem cells and these have been successfully grafted into animal models of HD. However, whether these grafts can provide stable functional recovery to the level that can regularly be achieved with primary fetal striatal grafts remains to be demonstrated. Of particular concern, primary fetal striatal grafts are not homogenous; they contain not only the MSN subpopulation of striatal projection neurons but also include all the different cell types that make up the mature striatum, such as the multiple populations of striatal interneurons and striatal glia, and which certainly contribute to normal striatal function. By contrast, present protocols for pluripotent stem cell differentiation are almost entirely targeted at specifying just neurons of an MSN lineage. So far, evidence for the functionality and integration of stem-cell derived grafts is correspondingly limited. Indeed, consideration of the features of full striatal reconstruction that is achieved with primary fetal striatal grafts suggests that optimal success of the next generations of stem cell-derived replacement therapy in HD will require that graft protocols be developed to allow inclusion of multiple striatal cell types, such as interneurons and/or glia. Almost certainly, therefore, more sophisticated differentiation protocols will be necessary, over and above replacement of a specific population of MSNs. A rational solution to this technical challenge requires that we re-address the underlying question-what constitutes a functional striatal graft?
Collapse
Affiliation(s)
- Amy E Reddington
- The Brain Repair Group, School of Biosciences, Cardiff University Cardiff, UK
| | - Anne E Rosser
- The Brain Repair Group, School of Biosciences, Cardiff University Cardiff, UK ; Department of Psychological Medicine and Neurology, Cardiff University Cardiff, UK
| | - Stephen B Dunnett
- The Brain Repair Group, School of Biosciences, Cardiff University Cardiff, UK
| |
Collapse
|
17
|
Maucksch C, Vazey EM, Gordon RJ, Connor B. Stem cell-based therapy for Huntington's disease. J Cell Biochem 2013; 114:754-63. [PMID: 23097329 DOI: 10.1002/jcb.24432] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 10/15/2012] [Indexed: 12/23/2022]
Abstract
Huntington's disease (HD) is a late-onset neurodegenerative disease characterized by a progressive loss of medium spiny neurons in the basal ganglia. The development of stem cell-based therapies for HD aims to replace lost neurons and/or to prevent cell death. This review will discuss pre-clinical studies which have utilized stem or progenitor cells for transplantation therapy using HD animal models. In several studies, neural stem and progenitor cells used as allotransplants and xenografts have been shown to be capable of surviving transplantation and differentiating into mature GABAergic neurons, resulting in behavioral improvements. Beneficial effects have also been reported for transplantation of stem cells derived from non-neural tissue, for example, mesenchymal- and adipose-derived stem cells, which have mainly been attributed to their secretion of growth and neurotrophic factors. Finally, we review studies using stem cells genetically engineered to over-express defined neurotrophic factors. While these studies prove the potential of stem cells for transplantation therapy in HD, it also becomes clear that technical and ethical issues regarding the availability of stem cells must be solved before human trials can be conducted.
Collapse
Affiliation(s)
- Christof Maucksch
- Department of Pharmacology and Clinical Pharmacology, Centre for Brain Research, School of Medical Science, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | | | | | | |
Collapse
|
18
|
Pauly MC, Piroth T, Döbrössy M, Nikkhah G. Restoration of the striatal circuitry: from developmental aspects toward clinical applications. Front Cell Neurosci 2012; 6:16. [PMID: 22529778 PMCID: PMC3329876 DOI: 10.3389/fncel.2012.00016] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 03/23/2012] [Indexed: 12/20/2022] Open
Abstract
In the basal ganglia circuitry, the striatum is a highly complex structure coordinating motor and cognitive functions and it is severely affected in Huntington's disease (HD) patients. Transplantation of fetal ganglionic eminence (GE) derived precursor cells aims to restore neural circuitry in the degenerated striatum of HD patients. Pre-clinical transplantation in genetic and lesion HD animal models has increased our knowledge of graft vs. host interactions, and clinical studies have been shown to successfully reduce motor and cognitive effects caused by the disease. Investigating the molecular mechanisms of striatal neurogenesis is a key research target, since novel strategies aim on generating striatal neurons by differentiating embryonic stem cells or by reprogramming somatic cells as alternative cell source for neural transplantation.
Collapse
Affiliation(s)
- Marie-Christin Pauly
- Division of Stereotactic Neurosurgery, Department of General Neurosurgery, University Freiburg - Medical Center Freiburg im Breisgau, Germany
| | | | | | | |
Collapse
|
19
|
Döbrössy MD, Nikkhah G. Role of experience, training, and plasticity in the functional efficacy of striatal transplants. PROGRESS IN BRAIN RESEARCH 2012. [PMID: 23195425 DOI: 10.1016/b978-0-444-59575-1.00014-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Cell-based treatments of neurodegenerative diseases have been tested clinically with partial success. In the context of Huntington's disease (HD), experimental studies show that the grafted embryonic striatal cells survive, integrate within the host brain, and reverse some functional deficits. Importantly, once transplanted, the grafted striatal neurons retain a significant level of cellular, morphological, and functional plasticity which allows the experimental modification of their character through the manipulation of environmental cues or learning protocols. Using embryonic striatal grafts in the rodent model of HD as the principal example, this chapter summarizes seminal experiments that demonstrate that environmental factors, training, and activity can tap into mechanisms that influence the development of the grafted cells and can change the profile of graft-mediated behavioral recovery. Although currently there is limited understanding of the biological rationale behind the recovery, we put forward experimental data indicating that striatal grafts can express experience-dependent physiological plasticity at the synaptic as well as at the systemic functional level.
Collapse
Affiliation(s)
- Máté D Döbrössy
- Laboratory of Molecular Neurosurgery, Division of Stereotactic Neurosurgery, Department of General Neurosurgery, University of Freiburg Medical Center, Freiburg, Germany.
| | | |
Collapse
|
20
|
Wijeyekoon R, Barker RA. The Current Status of Neural Grafting in the Treatment of Huntington's Disease. A Review. Front Integr Neurosci 2011; 5:78. [PMID: 22162966 PMCID: PMC3232470 DOI: 10.3389/fnint.2011.00078] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Accepted: 11/15/2011] [Indexed: 11/15/2022] Open
Abstract
Huntington’s disease (HD) is a devastating, fatal, autosomal dominant condition in which the abnormal gene codes for a mutant form of huntingtin that causes widespread neuronal dysfunction and death. This leads to a clinical presentation, typically in midlife, with a combination of motor, psychiatric, cognitive, metabolic, and sleep abnormalities, for which there are some effective symptomatic therapies that can produce some transient benefits. The disease, though, runs a progressive course over a 20-year period ultimately leading to death, and there are currently no proven disease modifying therapies. However whilst the neuronal dysfunction and loss affects much of the central nervous system, the striatum is affected early on in the disease and is one of the areas most affected by the pathogenic process. As a result the prospect of treating HD using neural transplants of striatal tissue has been explored and to date the clinical data is inconclusive. In this review we discuss the rationale for treating HD using this approach, before discussing the clinical trial data and what we have learnt to date using this therapeutic strategy.
Collapse
Affiliation(s)
- Ruwani Wijeyekoon
- Cambridge Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge Cambridge, UK
| | | |
Collapse
|
21
|
Dunnett SB, Rosser AE. Cell-based treatments for huntington's disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2011; 98:483-508. [PMID: 21907097 DOI: 10.1016/b978-0-12-381328-2.00017-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In experimental rats, mice, and monkeys, transplantation of embryonic striatal cells into the striatum can repair the damage and alleviate the functional deficits caused by striatal lesions. Such strategies have been translated to striatal repair by cell transplantation in small numbers of patients with progressive genetic striatal degeneration in Huntington's disease. In spite of some encouraging preliminary data, the clinical results are to date neither as reliable nor as compelling as the broad extend of recovery observed in the animal models across motor, cognitive, and skill and habit learning domains. Strategies to achieve immediate and long-term improvements in the clinical applications include identifying and limiting the causes of complications, standardization and quality control of preparation and delivery, appropriate patient selection to match the cellular repair to specific profiles of cell loss and degeneration in individual patients and different neurodegenerative diseases, and improving the availability of alternative sources of donor cells and tissues.
Collapse
Affiliation(s)
- Stephen B Dunnett
- Brain Repair Group, Schools of Biosciences and Medicine, Cardiff University, Cardiff, Wales, UK
| | | |
Collapse
|
22
|
Walker RH, Davies G, Koch RJ, Haack AK, Moore C, Meshul CK. Effects of zona incerta lesions on striatal neurochemistry and behavioral asymmetry in 6-hydroxydopamine-lesioned rats. J Neurosci Res 2010; 88:2964-75. [DOI: 10.1002/jnr.22446] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
23
|
Dunnett SB, Lelos M. Behavioral analysis of motor and non-motor symptoms in rodent models of Parkinson's disease. PROGRESS IN BRAIN RESEARCH 2010; 184:35-51. [PMID: 20887869 DOI: 10.1016/s0079-6123(10)84003-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Alongside the classical motor symptoms, non-motor symptoms are increasingly recognised to play a major role in the disability associated with Parkinson's disease in humans. Animal models based on experimental depletion of forebrain dopamine have traditionally focussed on the simple and easy to measure motor impairments, and they reproduce well the bradykinesia, rigidity and impairments in the initiation and sequencing of voluntary goal-directed movement. However, a more comprehensive analysis is now urgently required. In this chapter we summarise the predominant unilateral and bilateral dopamine lesion, toxin and genetic models of human parkinsonism, and review the consequences in more complex cognitive, motor learning and psychiatric ('behavioural') domains. Theoretical and experimental advances in our understanding of information processing and associative plasticity within the striatum are not only revolutionising our understanding of normal striatal function but also bear directly on our understanding of the processes that underlie non-motor as well as motor disability in human disease, including in Parkinson's disease.
Collapse
Affiliation(s)
- Stephen B Dunnett
- School of Biosciences, Cardiff University, Cardiff, South Wales, UK.
| | | |
Collapse
|
24
|
Karl JM, Sacrey LAR, McDonald RJ, Whishaw IQ. Intact intracortical microstimulation (ICMS) representations of rostral and caudal forelimb areas in rats with quinolinic acid lesions of the medial or lateral caudate-putamen in an animal model of Huntington's disease. Brain Res Bull 2008; 77:42-8. [DOI: 10.1016/j.brainresbull.2008.04.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2008] [Revised: 04/17/2008] [Accepted: 04/23/2008] [Indexed: 10/22/2022]
|
25
|
Whishaw IQ, Zeeb F, Erickson C, McDonald RJ. Neurotoxic lesions of the caudate-putamen on a reaching for food task in the rat: acute sensorimotor neglect and chronic qualitative motor impairment follow lateral lesions and improved success follows medial lesions. Neuroscience 2007; 146:86-97. [PMID: 17346897 DOI: 10.1016/j.neuroscience.2007.01.034] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2006] [Revised: 12/12/2006] [Accepted: 01/15/2007] [Indexed: 11/24/2022]
Abstract
Reaching for food, or skilled reaching, is used as a test of basal ganglia function in preclinical studies as well as studies of human neurological conditions. Although changes in the end-point measure of success document the effects of neurotoxic cellular damage to the caudate-putamen and its treatment in rodents, there has been no examination of the cause of change in success after neurotoxic lesions of the striatum. This objective was addressed in the present study, in which rats trained to reach for single food pellets with one forelimb, received contralateral quinolinic acid or ibotenic acid lesions of the medial and lateral caudate-putamen. Over 21 postsurgical days, reaching performance was scored for success and qualitative changes in movement elements were examined using frame-by-frame video analysis. In the acute postoperative period, extending over 3 to 4 days, the rats with lateral lesions transported their forelimb and grasped the food, but then ignored the food and did not withdraw their limb to their mouth. After recovery of the withdrawal movement, the rats displayed chronic qualitative impairments in the rotatory movements of aiming, pronating, and supinating the forepaw. Medial quinolinic lesions improved success relative to control rats and did not change qualitative aspects of limb movement. The acute dissociation between transport and withdrawal, the chronic qualitative changes in movement elements, and the differential effect of medial and lateral injury on success, support a complex contribution of the caudate-putamen to skilled reaching that includes sensorimotor neglect, and quantitative and qualitative motoric changes.
Collapse
Affiliation(s)
- I Q Whishaw
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, 4401 University Drive, Lethbridge, Alberta, Canada T1K 3M4.
| | | | | | | |
Collapse
|
26
|
Dunnett SB, Rosser AE. Stem cell transplantation for Huntington's disease. Exp Neurol 2007; 203:279-92. [PMID: 17208230 DOI: 10.1016/j.expneurol.2006.11.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2006] [Revised: 11/13/2006] [Accepted: 11/15/2006] [Indexed: 01/13/2023]
Abstract
By way of commentary on a recent report that transplanted adult neural progenitor cells can alleviate functional deficits in a rat lesion model of Huntington's disease [Vazey, E.M., Chen, K., Hughes, S.M., Connor, B., 2006. Transplanted adult neural progenitor cells survive, differentiate and reduce motor function impairment in a rodent model of Huntington's disease. Exp. Neurol. 199, 384-396], we review the current status of the field exploring the use of stem cells, progenitor cells and immortalised cell lines to repair the lesioned striatum in animal models of the human disease. A remarkably rich range of alternative cell types have been used in various animal models, several of which exhibit cell survival and incorporation in the host brain, leading to subsequent functional recovery. In comparing the alternatives with the 'gold standard' currently offered by primary tissue grafts, key issues turn out to be: cell survival, differentiation prior to and following implantation into striatal-like phenotypes, integration and connectivity with the host brain, the nature of the electrophysiological, motor and cognitive tests used to assess functional repair, and the mechanisms by which the grafts exert their function. Although none of the alternatives yet has the capacity to match primary fetal tissues for functional repair, that standard is itself limited, and the long term goal must be not just to match but to surpass present capabilities in order to achieve fully functional reconstruction reliably, flexibly, and on demand.
Collapse
Affiliation(s)
- Stephen B Dunnett
- The Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, Wales, UK.
| | | |
Collapse
|
27
|
Johann V, Schiefer J, Sass C, Mey J, Brook G, Krüttgen A, Schlangen C, Bernreuther C, Schachner M, Dihné M, Kosinski CM. Time of transplantation and cell preparation determine neural stem cell survival in a mouse model of Huntington’s disease. Exp Brain Res 2006; 177:458-70. [PMID: 17013619 DOI: 10.1007/s00221-006-0689-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2006] [Accepted: 08/24/2006] [Indexed: 10/24/2022]
Abstract
Cell replacement therapies for neurodegenerative diseases, using multipotent neural stem cells (NSCs), require above all, a good survival of the graft. In this study, we unilaterally injected quinolinic acid (QA) into the striatum of adult mice and transplanted syngeneic NSCs of enhanced green fluorescent protein-transgenic mice into the lesioned striatum. The injection of QA leads to an excitotoxic lesion with selective cell death of the medium sized spiny neurons, the same cells that are affected in Huntington's disease. In order to investigate the best timing of transplantation for the survival of donor cells, we transplanted the stem cells at 2, 7 and 14 days after injury. In addition, the influence of graft preparation prior to transplantation, i.e., intact neurospheres versus dissociated cell suspension on graft survival was investigated. By far the best survival was found with the combination of early transplantation (i.e., 2 days after QA-lesion) with the use of neurospheres instead of dissociated cell suspension. This might be due to the different states of host's astrocytic and microglia activation which we found to be moderate at 2, but pronounced at 7 and 14 days after QA-lesion. We also investigated brain derived neurotrophic factor (BDNF)-expression in the striatum after QA-lesion and found no significant change in BDNF protein-level. We conclude that already the method of graft preparation of NSCs for transplantation, as well as the timing of the transplantation procedure strongly affects the survival of the donor cells when grafted into the QA-lesioned striatum of adult mice.
Collapse
Affiliation(s)
- Verena Johann
- Department of Neurology, University Hospital RWTH, Pauwelsstr. 30, 52074 Aachen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
McBride JL, Ramaswamy S, Gasmi M, Bartus RT, Herzog CD, Brandon EP, Zhou L, Pitzer MR, Berry-Kravis EM, Kordower JH. Viral delivery of glial cell line-derived neurotrophic factor improves behavior and protects striatal neurons in a mouse model of Huntington's disease. Proc Natl Acad Sci U S A 2006; 103:9345-50. [PMID: 16751280 PMCID: PMC1482612 DOI: 10.1073/pnas.0508875103] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Huntington's disease (HD) is a fatal, genetic, neurological disorder resulting from a trinucleotide repeat expansion in the gene that encodes for the protein huntingtin. These excessive repeats confer a toxic gain of function on huntingtin, which leads to the degeneration of striatal and cortical neurons and a devastating motor, cognitive, and psychological disorder. Trophic factor administration has emerged as a compelling potential therapy for a variety of neurodegenerative disorders, including HD. We previously demonstrated that viral delivery of glial cell line-derived neurotrophic factor (GDNF) provides structural and functional neuroprotection in a rat neurotoxin model of HD. In this report we demonstrate that viral delivery of GDNF into the striatum of presymptomatic mice ameliorates behavioral deficits on the accelerating rotorod and hind limb clasping tests in transgenic HD mice. Behavioral neuroprotection was associated with anatomical preservation of the number and size of striatal neurons from cell death and cell atrophy. Additionally, GDNF-treated mice had a lower percentage of neurons containing mutant huntingtin-stained inclusion bodies, a hallmark of HD pathology. These data further support the concept that viral vector delivery of GDNF may be a viable treatment for patients suffering from HD.
Collapse
Affiliation(s)
- Jodi L. McBride
- *Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison Street, Suite 300, Chicago, IL 60612
| | - Shilpa Ramaswamy
- *Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison Street, Suite 300, Chicago, IL 60612
| | - Mehdi Gasmi
- Ceregene Inc., 9381 Judicial Drive, Suite 130, San Diego, CA 92121; and
| | - Raymond T. Bartus
- Ceregene Inc., 9381 Judicial Drive, Suite 130, San Diego, CA 92121; and
| | | | - Eugene P. Brandon
- Ceregene Inc., 9381 Judicial Drive, Suite 130, San Diego, CA 92121; and
| | - Lili Zhou
- *Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison Street, Suite 300, Chicago, IL 60612
| | - Mark R. Pitzer
- Department of Psychology, Grinnell College, 1116 Eighth Avenue, Grinnell, IA 50112
| | - Elizabeth M. Berry-Kravis
- *Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison Street, Suite 300, Chicago, IL 60612
| | - Jeffrey H. Kordower
- *Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison Street, Suite 300, Chicago, IL 60612
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
29
|
Dunnett SB, White A. Striatal grafts alleviate bilateral striatal lesion deficits in operant delayed alternation in the rat. Exp Neurol 2006; 199:479-89. [PMID: 16516889 DOI: 10.1016/j.expneurol.2006.01.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2005] [Revised: 01/03/2006] [Accepted: 01/12/2006] [Indexed: 11/27/2022]
Abstract
In order to assess the capacity of striatal grafts to alleviate cognitive deficits of the frontal type that arise following bilateral striatal lesions, control, lesion and grafted rats were tested in an operant test of delayed alternation. Bilateral striatal lesions induced a marked impairment in choice accuracy, and signal detection analysis indicated that the lesion animals were reliably impaired on both parametric and non-parametric indices of discriminative sensitivity but not of response bias. The impairment was apparent at all intertrial interval delays, including the very shortest, suggesting the deficit is one of frontal-type executive function rather than of short-term memory. The grafted animals exhibited a significant alleviation of the deficit, again apparent at all delays. Histological analyses indicated good graft survival, and injections of a dextran amine anterograde tracer bilaterally into the host prefrontal cortex indicated reformation of extensive projections into the grafted tissues. Since performance of the operant delayed alternation task is dependent upon the integrity of corticostriatal connections, which is disrupted bilaterally by the lesions and restored to the grafts in the transplanted animals, the results corroborate the hypothesis that striatal grafts can alleviate complex cognitive functions of the frontal type by a mechanism that involves functional integration of the grafted neurons into the neural circuits of the host brain.
Collapse
Affiliation(s)
- Stephen B Dunnett
- School of Biosciences, Cardiff University, Museum Avenue Box 911, Cardiff CF10 3US, Wales, UK.
| | | |
Collapse
|
30
|
Döbrössy MD, Dunnett SB. The effects of lateralized training on spontaneous forelimb preference, lesion deficits, and graft-mediated functional recovery after unilateral striatal lesions in rats. Exp Neurol 2006; 199:373-83. [PMID: 16499910 DOI: 10.1016/j.expneurol.2005.12.033] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2005] [Revised: 11/25/2005] [Accepted: 12/03/2005] [Indexed: 10/25/2022]
Abstract
The ability of striatal embryonic grafts to promote functional recovery on complex behavioral tasks depends on various factors, including the amount of striatal-like tissue within the grafts and the duration of post-graft training. However, how the innate paw bias of animals is affected by experience, or influences recovery following injury, is less known. Here, we have examined the effects of intrinsic side bias and lateralized limb use training on spontaneous forelimb preference and graft-mediated functional recovery in a skilled reaching task in a rodent model of Huntington's disease. Naïve rats were assessed on their baseline paw preferences when reaching between the bars of their cage to retrieve sugar pellets from a tray attached outside. Next, rats were lesioned unilaterally in the lateral dorsal striatum with quinolinic acid, and 7-10 days later, half of the animals were given suspension grafts prepared from E15 whole ganglionic eminence implanted into the lesioned striatum. The animals then received extensive unilateral training, either ipsi- or contralateral to the side of the lesion and graft in separate subgroups, on the 'staircase' task until asymptotic performance was obtained. As reported previously, the grafts alleviated lesion-induced deficits in retrieving pellets from the contralateral staircase. Spontaneous biases were then reassessed in the cage-reaching task. Irrespective of whether the animal received ipsilateral or contralateral staircase training, the unilateral lesions induced a significant shift in spontaneous bias towards the ipsilateral paw. Grafted animals showed a similar shift in bias if staircase training was given to the ipsilateral paw but showed no change in spontaneous bias (similar to controls) if they had received contralateral training during the post-transplantation period. The results suggest that striatal grafts can alleviate lesion-induced changes in their spontaneous side preferences, but only if they receive extensive training in the use of the contralateral limb, compatible with the notion that recovery is use-dependent.
Collapse
Affiliation(s)
- Máté D Döbrössy
- Brain Repair Group, School of Biosciences, Cardiff University, Museum Avenue Box 911, Cardiff CF10 3US, UK.
| | | |
Collapse
|
31
|
Pollock K, Stroemer P, Patel S, Stevanato L, Hope A, Miljan E, Dong Z, Hodges H, Price J, Sinden JD. A conditionally immortal clonal stem cell line from human cortical neuroepithelium for the treatment of ischemic stroke. Exp Neurol 2006; 199:143-55. [PMID: 16464451 DOI: 10.1016/j.expneurol.2005.12.011] [Citation(s) in RCA: 207] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2005] [Accepted: 12/05/2005] [Indexed: 01/19/2023]
Abstract
Transplantation of neural stem cells into the brain is a novel approach to the treatment of chronic stroke disability. For clinical application, safety and efficacy of defined, stable cell lines produced under GMP conditions are required. To this end, a human neural stem cell line, CTX0E03, was derived from human somatic stem cells following genetic modification with a conditional immortalizing gene, c-mycER(TAM). This transgene generates a fusion protein that stimulates cell proliferation in the presence of a synthetic drug 4-hydroxy-tamoxifen (4-OHT). The cell line is clonal, expands rapidly in culture (doubling time 50-60 h) and has a normal human karyotype (46 XY). In the absence of growth factors and 4-OHT, the cells undergo growth arrest and differentiate into neurons and astrocytes. Transplantation of CTX0E03 in a rat model of stroke (MCAo) caused statistically significant improvements in both sensorimotor function and gross motor asymmetry at 6-12 weeks post-grafting. In addition, cell migration and long-term survival in vivo were not associated with significant cell proliferation. These data indicate that CTX0E03 has the appropriate biological and manufacturing characteristics necessary for development as a therapeutic cell line.
Collapse
Affiliation(s)
- Kenneth Pollock
- ReNeuron Ltd., 10 Nugent Road, Surrey Research Park, Guildford, Surrey GU2 7AF, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Mátyás F, Yanovsky Y, Mackie K, Kelsch W, Misgeld U, Freund TF. Subcellular localization of type 1 cannabinoid receptors in the rat basal ganglia. Neuroscience 2005; 137:337-61. [PMID: 16289348 DOI: 10.1016/j.neuroscience.2005.09.005] [Citation(s) in RCA: 131] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2005] [Revised: 09/08/2005] [Accepted: 09/09/2005] [Indexed: 11/29/2022]
Abstract
Endocannabinoids, acting via type 1 cannabinoid receptors (CB1), are known to be involved in short-term synaptic plasticity via retrograde signaling. Strong depolarization of the postsynaptic neurons is followed by the endocannabinoid-mediated activation of presynaptic CB1 receptors, which suppresses GABA and/or glutamate release. This phenomenon is termed depolarization-induced suppression of inhibition (DSI) or excitation (DSE), respectively. Although both phenomena have been reported to be present in the basal ganglia, the anatomical substrate for these actions has not been clearly identified. Here we investigate the high-resolution subcellular localization of CB1 receptors in the nucleus accumbens, striatum, globus pallidus and substantia nigra, as well as in the internal capsule, where the striato-nigral and pallido-nigral pathways are located. In all examined nuclei of the basal ganglia, we found that CB1 receptors were located on the membrane of axon terminals and preterminal axons. Electron microscopic examination revealed that the majority of these axon terminals were GABAergic, giving rise to mostly symmetrical synapses. Interestingly, preterminal axons showed far more intense staining for CB1, especially in the globus pallidus and substantia nigra, whereas their terminals were only faintly stained. Non-varicose, thin unmyelinated fibers in the internal capsule also showed strong CB1-labeling, and were embedded in bundles of myelinated CB1-negative axons. The majority of CB1 receptors labeled by immunogold particles were located in the axonal plasma membrane (92.3%), apparently capable of signaling cannabinoid actions. CB1 receptors in this location cannot directly modulate transmitter release, because the release sites are several hundred micrometers away. Interestingly, both the CB1 agonist, WIN55,212-2, as well as its antagonist, AM251, were able to block action potential generation, but via a CB1 independent mechanism, since the effects remained intact in CB1 knockout animals. Thus, our electrophysiological data suggest that these receptors are unable to influence action potential propagation, thus they may not be functional at these sites, but are likely being transported to the terminal fields. The present data are consistent with a role of endocannabinoids in the control of GABA, but not glutamate, release in the basal ganglia via presynaptic CB1 receptors, but also call the attention to possible non-CB1-mediated effects of widely used cannabinoid ligands on action potential generation.
Collapse
Affiliation(s)
- F Mátyás
- Institute of Experimental Medicine, Hungarian Academy of Sciences, PO Box 67, Budapest H-1450, Hungary
| | | | | | | | | | | |
Collapse
|
33
|
Nichols CM, Myckatyn TM, Rickman SR, Fox IK, Hadlock T, Mackinnon SE. Choosing the correct functional assay: A comprehensive assessment of functional tests in the rat. Behav Brain Res 2005; 163:143-58. [PMID: 15979168 DOI: 10.1016/j.bbr.2005.05.003] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2005] [Revised: 05/16/2005] [Accepted: 05/18/2005] [Indexed: 11/19/2022]
Abstract
While there are several ways to quantify peripheral nerve regeneration; the true measure of successful outcome is functional recovery. Functional tests are relatively easily conducted in human subjects; however it is more difficult in a laboratory animal. The laboratory rat is an excellent animal model of peripheral nerve injury and has been used extensively in the field of peripheral nerve research. Due to the intense interest in the rat as an experimental model, functional assays have been reported. In an effort to provide a resource to which investigators can refer when considering the most appropriate functional assay for a given experiment, the authors have compiled and tabulated the available functional tests applicable to various models of rat nerve injury.
Collapse
Affiliation(s)
- Chris M Nichols
- Washington University School of Medicine, Division of Plastic and Reconstructive Surgery, Campus Box 8238, St. Louis, MO 63110, USA
| | | | | | | | | | | |
Collapse
|
34
|
Andres RH, Ducray AD, Huber AW, Pérez-Bouza A, Krebs SH, Schlattner U, Seiler RW, Wallimann T, Widmer HR. Effects of creatine treatment on survival and differentiation of GABA-ergic neurons in cultured striatal tissue. J Neurochem 2005; 95:33-45. [PMID: 16045451 DOI: 10.1111/j.1471-4159.2005.03337.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder, characterized by a prominent loss of GABA-ergic medium-sized spiny neurons in the caudate putamen. There is evidence that impaired energy metabolism contributes to neuronal death in HD. Creatine is an endogenous substrate for creatine kinases and thereby supports cellular ATP levels. This study investigated the effects of creatine supplementation (5 mm) on cell survival and neuronal differentiation in striatal cultures. Chronic creatine treatment resulted in significant increased densities of GABA-immunoreactive (-ir) neurons, although total neuronal cell number and general viability were not affected. Similar effects were seen after short-term treatment, suggesting that creatine acted as a differentiation factor. Inhibitors of transcription or translation did not abolish the creatine-mediated effects, nor did omission of extracellular calcium, whereas inhibition of mitogen-activated protein kinase and phosphatidylinositol-3-kinase significantly attenuated the creatine induced increase in GABA-ir cell densities. Creatine exhibited significant neuroprotection against toxicity instigated either by glucose- and serum deprivation or addition of 3-nitropropionic acid. In sum, the neuroprotective properties in combination with promotion of neuronal differentiation suggest that creatine has potential as a therapeutic drug in the treatment of neurodegenerative diseases, like HD.
Collapse
Affiliation(s)
- R H Andres
- Department of Neurosurgery, University Hospital, Berne, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Döbrössy MD, Dunnett SB. Environmental enrichment affects striatal graft morphology and functional recovery. Eur J Neurosci 2004; 19:159-68. [PMID: 14750974 DOI: 10.1111/j.1460-9568.2004.03105.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Environmental conditions and behavioural experience can affect neuronal function and morphology. It is less well known whether such factors also influence the growth, integration and functional recovery provided by neural grafts placed within the damaged brain. Here we report on the effects of differential housing conditions on striatal graft morphology and functional recovery after striatal lesions. Rats were pretrained on a skilled bilateral forelimb task, the staircase test, and lesioned unilaterally in the lateral dorsal striatum with quinolinic acid. One group of lesioned animals was given suspension grafts of E15 whole ganglionic eminence implanted into the lesioned striatum. Following transplantation, the animals were housed either in standard cages (four per cage) or in enriched environment housing conditions (10 per cage) with tunnels, ladders and increased living space available for exploration, social interaction and play. The differentially housed animals were retested on the skilled staircase test at two separate time points. Repeated testing, environmental enrichment and transplantation positively influenced behavioural recovery. Partial recovery was observed bilaterally amongst the grafted animals in both housing conditions. Nevertheless, the grafted animals housed in the enriched environment performed significantly better in the final test compared with all of the other experimental groups. The grafts survived equally well under both housing conditions but the grafts of animals housed in the enriched environment contained larger projection neurons and were somewhat better reinnervated by dopaminergic afferents. An increased level of striatal brain-derived neurotrophic factor was observed in the control animals housed under the enriched compared with the standard conditions. The results indicate that an enriched environment can affect both graft function and graft morphology through as yet unknown mechanisms.
Collapse
Affiliation(s)
- Máté D Döbrössy
- Brain Repair Group, School of Biosciences, Cardiff University, Museum Avenue Box 911, Cardiff CF10 3US, UK.
| | | |
Collapse
|
36
|
McBride JL, Behrstock SP, Chen EY, Jakel RJ, Siegel I, Svendsen CN, Kordower JH. Human neural stem cell transplants improve motor function in a rat model of Huntington's disease. J Comp Neurol 2004; 475:211-9. [PMID: 15211462 DOI: 10.1002/cne.20176] [Citation(s) in RCA: 177] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The present study investigated the neuroanatomical and behavioral effects of human stem cell transplants into the striatum of quinolinic acid (QA)-lesioned rats. Twenty-four rats received unilateral QA (200 nM/microl) injections into the striatum. One week later, rats were transplanted with stem cells derived from human fetal cortex (12 weeks postconception) that were either 1) pretreated in culture media with the differentiating cytokine ciliary neurotrophic factor (CNTF; n = 9) or 2) allowed to grow in culture media alone (n=7). Each rat was injected with a total of 200,000 cells. A third group of rats (n=8) was given a sham injection of vehicle. Rats transplanted with human stem cells performed significantly better over the 8 weeks of testing on the cylinder test compared with those treated with vehicle (P < or = 0.001). Stereological striatal volume analyses performed on Nissl-stained sections revealed that rats transplanted with CNTF-treated neurospheres had a 22% greater striatal volume on the lesioned side compared with those receiving transplants of untreated neurospheres (P = 0.0003) and a 26% greater striatal volume compared with rats injected with vehicle (P < or = 0.0001). Numerous human nuclei-positive cells were visualized in the striatum in both transplantation groups. Grafted cells were also observed in the globus pallidus, entopeduncular nucleus, and substantia nigra pars reticulata, areas of the basal ganglia receiving striatal projections. Some of the human nuclei-positive cells coexpressed glial fibrillary acidic protein and NeuN, suggesting that they had differentiated into neurons and astrocytes. Taken together, these data demonstrate that striatal transplants of human fetal stem cells elicit behavioral and anatomical recovery in a rodent model of Huntington's disease.
Collapse
Affiliation(s)
- Jodi L McBride
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois 60612, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
The gene for Huntington's disease was identified in 1993 as being a CAG repeat expansion in exon 1 of a gene now known as huntingtin on chromosome 4. Although many of the downstream effects of this mutant gene were identified in the subsequent years, a more detailed understanding of these events will be necessary in order to design specific interventions to interfere with the disease process and slow disease progression. In parallel, a number of groups have been investigating alternative approaches to treatment of Huntington's disease, including cell and tissue transplantation. As the brunt of cell dysfunction and loss is borne by the striatum, at least in the early to mid-stages of disease, the goal is to identify methods for replacing lost cells with fetal neuroblasts that can develop, integrate into the host circuitry and thereby restore lost function. Clinical studies in which primary fetal neuroblasts were transplanted into the brains of patients with advanced Parkinson's disease have demonstrated benefit when the transplant methodology closely follows the biological principles established in animal experiments. On the basis of demonstrated benefit following striatal cell transplantation in animal models of Huntington's disease, a small number of studies have now commenced in patients with Huntington's disease. To date, these clinical studies have demonstrated the feasibility and safety of transplantation in this condition, but it will require several more years yet before the efficacy of the procedure can be confidently established.
Collapse
Affiliation(s)
- Anne E Rosser
- School of Biosciences, Cardiff University, Cardiff, UK.
| | | |
Collapse
|
38
|
Döbrössy MD, Dunnett SB. Motor training effects on recovery of function after striatal lesions and striatal grafts. Exp Neurol 2003; 184:274-84. [PMID: 14637098 DOI: 10.1016/s0014-4886(03)00028-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Environment, training, and experience can influence plasticity and recovery of function after brain damage. However, it is less well known whether, and how, such factors influence the growth, integration, and functional recovery provided by neural grafts placed within the brain. To explore this process, rats were pretrained on the skilled staircase test, then lesioned unilaterally in the lateral dorsal striatum with quinolinic acid. Half of the animals were given suspension grafts prepared from E15 whole ganglionic eminence implanted into the lesioned striatum. For the following 5 months, half of the animals in each group were trained daily in a bilateral manual dexterity task. Then, 23 weeks after surgery, all animals were retested on the staircase test. The grafts promoted recovery in the reaching task, irrespective of the additional dexterity training, and within the trained group recovery was proportional to the volume of the striatal-like tissue in the graft, suggesting that training influenced the pattern of graft-induced functional recovery. The additional training also benefited the rats with lesions alone, raising their performance close to level of the grafted groups. In separate tests of rotation, the grafts reduced drug-induced ipsilateral turning in response to both amphetamine and apomorphine, an effect that was greater in the grafted rats given extra training. The results suggest that both nonspecific motor training and cell transplantation can contribute to recovery of lost function in tests of spontaneous and skilled lateralized motor function after striatal damage, and that these two factors interact in a task-specific manner.
Collapse
Affiliation(s)
- Máté D Döbrössy
- Brain Repair Group, School of Biosciences, Cardiff University, Museum Avenue Box 911, Cardiff CF10 3US, Wales, UK.
| | | |
Collapse
|
39
|
Ghorayeb I, Fernagut PO, Hervier L, Labattu B, Bioulac B, Tison F. A 'single toxin-double lesion' rat model of striatonigral degeneration by intrastriatal 1-methyl-4-phenylpyridinium ion injection: a motor behavioural analysis. Neuroscience 2003; 115:533-46. [PMID: 12421620 DOI: 10.1016/s0306-4522(02)00401-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Previous attempts to reproduce striatonigral degeneration, the core pathology underlying Parkinsonism in multiple system atrophy, have been impeded by interactions in the neurotoxins used to replicate striatal and nigral degeneration in rodents. To overcome these interactions, we have developed a new model of striatonigral degeneration which uses a single unilateral administration of 1-methyl-4-phenylpyridinium ion (MPP(+)) into the rat striatum. Spontaneous and drug-induced rotational behaviour, thigmotactic scanning, stepping adjusting steps and paw reaching deficits were compared in four groups of animals: group 1 (control), group 2 (20 microg quinolinic acid), group 3 (20 microg 6-hydroxydopamine), and group 4 (90 nmol MPP(+)). MPP(+) administration resulted in the absence of the amphetamine-induced ipsilateral bias observed in the 6-hydroxydopamine group and of the apomorphine-induced ipsilateral bias observed in the quinolinic acid group. There was no thigmotactic scanning asymmetry in the MPP(+)-injected rats compared to the quinolinic acid- and the 6-hydroxydopamine-injected rats. MPP(+) elicited a bilateral stepping adjustment deficit similar to that found in the quinolinic acid group when compared to controls. MPP(+) also elicited a more severe and significant contralateral deficit in paw reaching compared to controls, 6-hydroxydopamine and quinolinic acid groups. Histopathology revealed a significant reduction of the lesioned striatal surface (-47.53%) with neuronal loss and increased astrogliosis in the MPP(+) group grossly similar to that found in the quinolinic acid group. Contrary to the latter group, however, loss of intrastriatal and striatal-crossing fibre bundles was observed in the MPP(+) group as there was also some retrograde degeneration in the ipsilateral thalamic parafascicular nucleus. The mean loss of dopaminergic cells in the ipsilateral substantia nigra pars compacta in MPP(+) rats was less marked (-48.8%) than in the 6-hydroxydopamine rats (-63.6%) and was not significant in quinolinic acid rats (-5.2%). This study shows that a single unilateral intrastriatal administration of MPP(+) induces a unique motor behaviour resulting from both nigral and striatal degeneration, but also from possible extrastriatal damage. This 'single toxin-double lesion' paradigm may thus serve as a rat model of striatonigral degeneration.
Collapse
Affiliation(s)
- I Ghorayeb
- Laboratoire de Neurophysiologie, CNRS-UMR 5543, Université Victor Segalen-Bordeaux 2, 146 rue Léo-Saignat, 33076 Bordeaux Cedex, France
| | | | | | | | | | | |
Collapse
|
40
|
A novel skilled-reaching impairment in paw supination on the "good" side of the hemi-Parkinson rat improved with rehabilitation. J Neurosci 2003. [PMID: 12533618 DOI: 10.1523/jneurosci.23-02-00579.2003] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Parkinson's disease is characterized by tremor, rigidity, bradykinesia, and postural abnormalities ascribed to the loss of nigrostriatal dopamine (DA). Symptoms similar to the human condition can be produced in the rat by DA-depleting 6-hydroxydopamine injections made into the nigrostriatal system. After a unilateral lesion, the rat symptoms include sensory and motor impairments and turning biases reflecting motor abnormalities to the contralateral-to-depletion side of the body. In addition, a number of studies on skilled reaching report impairments in the use of the ipsilateral limb. It is suggested that the ipsilateral deficit is secondary to the contralateral motor impairments however. Here we re-examine how rats with unilateral DA depletion use their ipsilateral limb for skilled reaching for food. We provide the first description of an impairment on the ipsilateral-to-depletion side of the body of the rat and the first demonstration of amelioration of the defect using behavioral therapy. Video analysis of rats reaching for single pellets of food with the ipsilateral limb revealed that, although limb advancement and food grasping were normal, paw supination and food release to the mouth were impaired. Consequently, the animals were unable to transport a grasped food pellet to the mouth. Behavioral therapy, consisting of training in a simpler reaching task, strikingly lessened the impairment and improved reaching movements to the point that the rats could transport the food to the mouth. The results are discussed in relation to possible causes of the ipsilateral impairment, its treatment, and to relevant research on human Parkinson patients, indicating that they display bilateral improvements after unilateral treatments.
Collapse
|
41
|
Mayer E, Brown VJ, Dunnett SB, Robbins TW. Striatal graft-associated recovery of a lesion-induced performance deficit in the rat requires learning to use the transplant. Eur J Neurosci 2002; 4:119-26. [PMID: 12106374 DOI: 10.1111/j.1460-9568.1992.tb00858.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Peformance in a prelearned choice reaction time task was studied 6 months after surgery in rats with ibotenate-induced lesions of the striatum either with or without striatal grafts, and in sham-operated controls. The long postoperative interval allowed full transplant maturation and the establishment of appropriate connections by the transplants. The animals were trained prior to surgery on a visual choice reaction time task which requires that a movement is made away from stimuli signalling reward. The use of multiple measures allowed a thorough analysis of several aspects of the animals' performance. Whilst sham-operated control animals recovered normal (preoperative) performance rapidly, the lesioned animals had a severe performance deficit. Although the transplanted animals were initially at least as deficient in performance as the lesioned group, repeated testing led to an amelioration of the lesion-induced deficit according to two distinct measures of spatial bias and reaction time. On a third measure, latency to complete the lateralized movement, the grafted group were initially worse than the lesioned group but repeated testing resulted in significant recovery. These results suggest that postoperative training may help to optimize the efficacy of graft-induced recovery, and that animals may need to learn to use a transplant in order for it to confer functional benefit in complex prelearned tasks.
Collapse
Affiliation(s)
- E Mayer
- MRC Brain Repair Centre and Department of Experimental Psychology, University of Cambridge, Downing Street, Cambridge CB2 3EB, UK
| | | | | | | |
Collapse
|
42
|
Biomedical vignette. J Biomed Sci 2002. [DOI: 10.1007/bf02256582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
43
|
Moore AE, Cicchetti F, Hennen J, Isacson O. Parkinsonian motor deficits are reflected by proportional A9/A10 dopamine neuron degeneration in the rat. Exp Neurol 2001; 172:363-76. [PMID: 11716560 DOI: 10.1006/exnr.2001.7823] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In a model of Parkinson's disease (PD), amphetamine, a dopamine (DA)-releasing drug, fails to induce ipsilateral drug rotations in a proportion of rats with complete unilateral 6-hydroxydopamine (6-OHDA) lesions of the medial forebrain bundle and DA neurons of the substantia nigra. To investigate this phenomenon, individual 6-OHDA lesions (measured by tyrosine hydroxylase immunohistochemistry) in the substantia nigra pars compacta (A9), ventral tegmental area (A10), and striatum were examined in conjunction with outcomes of four behavioral tests. The behavioral tests were skilled paw reaching, a head-turning test, and apomorphine (0.05 mg/kg) and amphetamine (4 mg/kg) drug-induced rotations. Four weeks postlesion, ipsilateral side bias measured by the head-turning test correlated strongly with extent of A9 DA neuronal lesion. Additional A10 neuronal DA lesions did not substantially improve the model fit, indicating that the head-turning bias was primarily A9 dependent. In contrast, total head-turning activity increased monotonically with lesions of A10 striatal DA fibers. Skilled paw-reaching accuracy decreased with increased lesion of both A9 and A10 DA neuronal systems. Associating amphetamine-induced rotations with extent of A9 DA lesion generated a second-order polynomial model, y = -11.1x + 0.20 x(2) + 208.7 (R(2) = 0.73), with an overall F ratio (df = 2,21) of 28.4 (P < 0.0001). This model predicts that an A9 DA lesion of about 50% is required to induce an ipsilateral turning bias, after which rotations increase with the degree of A9 DA neuronal lesion. No further change in rotational behavior was seen until an additional A10 DA lesion reached 60%, after which the rotational response decreased. This analysis provides tests that differentiate between A9 DA degeneration and combined A9/A10 lesions in animal models and in addition allows predictive testing of PD therapeutic intervention at a preclinical level.
Collapse
Affiliation(s)
- A E Moore
- Neuroregeneration Laboratory, Harvard Medical School/McLean Hospital, Belmont, Massachusetts 02478, USA
| | | | | | | |
Collapse
|
44
|
Jeyasingham RA, Baird AL, Meldrum A, Dunnett SB. Differential effects of unilateral striatal and nigrostriatal lesions on grip strength, skilled paw reaching and drug-induced rotation in the rat. Brain Res Bull 2001; 55:541-8. [PMID: 11543955 DOI: 10.1016/s0361-9230(01)00557-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Lateralised motor deficits associated with basal ganglia dysfunction were compared in separate groups of rats receiving unilateral 6-hydroxydopamine-induced lesions of the dopaminergic nigrostriatal pathway, quinolinic acid-induced lesions of the striatum, or sham control injections. Amphetamine induced ipsilateral rotation in both lesion groups, whereas a low ('supersensitive') dose of apomorphine induced rotation only in the nigrostriatal lesion group. Both lesions induced impairments in skilled paw reaching with the contralateral paw in the 'staircase' test; by contrast the striatal lesions also induced a marked impairment with the ipsilateral paw, which was unaffected by the nigrostriatal lesion. A previously reported increase in grip strength with the contralateral paw after nigrostriatal lesion was replicated, whereas striatal lesions induced only minor bilateral deficits in this test. The results are discussed in the context of the utility, reliability and validity of alternative tests of motor deficit in animal models of Parkinson's and Huntington's diseases.
Collapse
Affiliation(s)
- R A Jeyasingham
- Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, UK
| | | | | | | |
Collapse
|
45
|
Metz GA, Farr T, Ballermann M, Whishaw IQ. Chronic levodopa therapy does not improve skilled reach accuracy or reach range on a pasta matrix reaching task in 6-OHDA dopamine-depleted (hemi-Parkinson analogue) rats. Eur J Neurosci 2001; 14:27-37. [PMID: 11488946 DOI: 10.1046/j.0953-816x.2001.01615.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
L-dopa therapy reverses some but not all of the motor deficits in human Parkinson patients. Although a number rat analogues of human Parkinson's disease have been developed for evaluating the efficacy of drug therapies, it is not known whether L-dopa has a similar selective action on the motor symptoms in the rat models. To examine the effectiveness of L-dopa in reversing the motor deficits in rats, we administered 6-OHDA unilaterally to produce hemi-Parkinson rats, which were then trained to reach for food using either their impaired (contralateral to the lesion) limb or their good (ipsilateral to the lesion) limb. To assess the skill, accuracy and range of limb movement, rats reached for pasta from a horizontal array of 260 vertically orientated pieces of pasta. The number and location of pasta pieces taken from this matrix was calculated and the qualitative aspects of the reaching movements were rated. The quantitative data on pasta sticks retrieved indicated that forelimb extension and movement radius around the shoulder joint was reduced by 6-OHDA treatment and did not improve after chronic L-dopa treatment. The qualitative analysis showed that grasping patterns, paw movements and body movements impaired by the lesion were also not improved by L-dopa treatment. These findings are the first in the rat to suggest that whereas L-dopa has a general activating effect on the rat's whole-body movements, as displayed in contralateral rotation, its effectiveness does not extend to skilled forelimb movements. The results are discussed in relationship to the idea that the restoration of some skilled movements may require normal synaptic function.
Collapse
Affiliation(s)
- G A Metz
- Department of Psychology and Neuroscience, University of Lethbridge, 4401 University Drive, Lethbridge, AB, Canada, T1K 3M4.
| | | | | | | |
Collapse
|
46
|
Waldner R, Puschban Z, Scherfler C, Seppi K, Jellinger K, Poewe W, Wenning GK. No functional effects of embryonic neuronal grafts on motor deficits in a 3-nitropropionic acid rat model of advanced striatonigral degeneration (multiple system atrophy). Neuroscience 2001; 102:581-92. [PMID: 11226695 DOI: 10.1016/s0306-4522(00)00500-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Intrastriatal injection of 3-nitropropionic acid results in secondary excitotoxic local damage and retrograde neuronal cell loss in substantia nigra pars compacta, thus mimicking salient features of striatonigral degeneration, the core pathology underlying Parkinsonism associated with multiple system atrophy. We used 3-nitropropionic acid to create a rat model of advanced striatonigral degeneration in order to assess the effects of embryonic allografts upon rotational and complex-motor behavioural abnormalities. Following stereotaxic intrastriatal administration of 500nmol 3-nitropropionic acid in male Wistar rats we observed consistent amphetamine- and apomorphine-induced ipsiversive rotation. Furthermore, there were marked deficits of contralateral paw reaching. Subsequently, animals received intrastriatal implantations of either E14 mesencephalic or striatal or mixed embryonic cell suspensions. In addition, one group received sham injections. Grafted rats were followed for up to 21 weeks and repeated behavioural tests were obtained during this period. Drug-induced rotation asymmetries and complex motor deficits measured by paw reaching tests were not compensated by embryonic grafts. Persistence of drug-induced rotations and of paw reaching deficits following transplantation probably reflects severe atrophy of adult striatum, additional nigral degeneration as well as glial demarcation of embryonic grafts. We suggest that dopamine rich embryonic grafts fail to induce functional recovery in a novel 3-nitropropionic acid rat model of advanced striatonigral degeneration (multiple system atrophy).
Collapse
Affiliation(s)
- R Waldner
- Neurological Research Laboratory, Department of Neurology, University Hospital Innsbruck, Anichstrasse 35, A-6020, Innsbruck, Austria
| | | | | | | | | | | | | |
Collapse
|
47
|
Dunnett SB, Nathwani F, Björklund A. The integration and function of striatal grafts. PROGRESS IN BRAIN RESEARCH 2001; 127:345-80. [PMID: 11142035 DOI: 10.1016/s0079-6123(00)27017-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- S B Dunnett
- School of Biosciences, Cardiff University, Cardiff CF1 3US, Wales, UK.
| | | | | |
Collapse
|
48
|
Kendall AL, Hantraye P, Palfi S. Striatal tissue transplantation in non-human primates. PROGRESS IN BRAIN RESEARCH 2001; 127:381-404. [PMID: 11142037 DOI: 10.1016/s0079-6123(00)27018-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The caudate nucleus and putamen form part of a complex but topographically connected circuitry that links the cortex, the basal ganglia and the thalamus. Within this complex system lie a series of functionally and anatomically segregated loops that allow the concurrent processing of a wide range of cognitive and motor information (Alexander et al., 1986; Alexander and Crutcher, 1990). As a constituent of these loops it has been shown that the striatum is involved in movement initiation, response selection and attentional processes (Robbins and Brown, 1990; Alexander, 1994; Lawrence et al., 1998). Although it is the medium spiny GABAergic projection neurones that are primarily lost in HD, it is not sufficient merely to replace the GABA. Instead it is crucial for striatal tissue transplants to integrate with the host tissue in such a way that the cortico-striatal-thalamic circuitry is restored and is functional. Rodent studies have progressed a long way in establishing the principle that striatal grafts can, at least partially, restore function and integrate appropriately with the host (Dunnett and Svendsen, 1993; Björklund et al., 1994; Sanberg et al., 1998) but the limited behavioural repertoire and the undifferentiated striatum meant that it was inevitable that studies should progress into primate models. Anatomical tracing studies have demonstrated that motor, premotor and somatosensory cortical areas send corticostriatal projections primarily to the putamen region in primates, whereas the head and body of the caudate nucleus mostly receive efferent input from associative cortical areas (Kemp and Powell, 1970; Kunzle, 1975, 1977, 1978; Selemon and Goldman-Rakic, 1985). Based on such anatomical, and functional, studies Alexander and colleagues have proposed the existence of at least five cortico-striatal-thalamic loops including a motor, a dorsolateral-prefrontal and an orbito-frontal loop (Alexander et al., 1986). The concentration of motor inputs to the putamen region suggests a particular involvement of this structure in the motor loop. Indeed, unilateral lesions of the putamen disrupt motor performance in the marmoset and generate apomorphine-induced dyskinesias in larger primates (Burns et al., 1995; Kendall et al., 2000). The implantation of striatal grafts into marmosets that had previously received unilateral putamen lesions ameliorated some of the motor impairments, which suggested at least partial restoration of the motor loop. In support of this we found direct evidence of host-graft cortico-striatal connectivity using an anterograde tracer injected in the primary motor cortical region (Kendall et al., 1998a). In larger primates, with lesions of the caudate and putamen, striatal [figure: see text] allografts and xenografts have been shown to reduce apomorphine-induced dyskinesias (Isacson et al., 1989; Hantraye et al., 1992; Palfi et al., 1998). The mechanism by which dyskinesias are elicited is not fully understood but alterations in firing patterns within both segments of the globus pallidus have been identified during dyskinetic movements (Matsumura et al., 1995). It seems likely that it would actually require re-establishment of afferent connections between the implanted putamen and the globus pallidus as well as of functioning dopamine receptors within the graft for the reduction in the dyskinetic profile to be observed. Certainly there is evidence, from rodent studies and the marmoset study described here, that close proximity of the graft to the globus pallidus yields better functional recovery (Isacson et al., 1986). In addition, anatomical tracing studies in rats have demonstrated connections between the implanted tissue and the host globus pallidus (Wictorin et al., 1989b, 1990) However, the relationship between graft placement and functional recovery remains to be fully substantiated.
Collapse
Affiliation(s)
- A L Kendall
- Department of Experimental Psychology, University of Cambridge, Downing Street, Cambridge CB2 3EB, UK.
| | | | | |
Collapse
|
49
|
Abstract
The "staircase" test has become established for measurement of side-specific deficits in coordinated paw reaching in rats, and has been shown to reveal impairments on the contralateral side following unilateral lesions in a wide range of motor structures of the brain. As mice become more widely used in behavioural neuroscience, we have scaled down the staircase reaching test for application to this latter species. We here validate the test in C57BL/6J mice by (a) establishing the optimal dimensions of the apparatus, (b) comparing the effects of test parameters including sex, test duration, levels of deprivation and alternative reward pellets, and (c) demonstrating contralateral deficits after aspirative lesions of the motor cortex. Differences between mice and rats in normal performance of the task are noted. The staircase test provides a simple objective test of skilled motor function that allows measurement of lateralised effects without unduly constraining the animal, and which may prove as useful for mice as has previously been demonstrated in rats.
Collapse
Affiliation(s)
- A L Baird
- School of Biosciences, Cardiff University, Cardiff, Wales, UK
| | | | | |
Collapse
|
50
|
Ghorayeb I, Puschban Z, Fernagut PO, Scherfler C, Rouland R, Wenning GK, Tison F. Simultaneous intrastriatal 6-hydroxydopamine and quinolinic acid injection: a model of early-stage striatonigral degeneration. Exp Neurol 2001; 167:133-47. [PMID: 11161601 DOI: 10.1006/exnr.2000.7535] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Animal models reproducing early stages of striatonigral degeneration (SND), the core pathology underlying parkinsonism in multiple system atrophy, are lacking. We have developed a new model of early-stage SND by using a simultaneous unilateral administration of quinolinic acid (QA) and 6-hydroxydopamine (6-OHDA) into the putaminal equivalent of the rat striatum. Spontaneous and drug-induced behavior, thigmotactic scanning, paw reaching deficits, and histopathology were studied in rat groups: group 1 (control), group 2 (QA), group 3 (6-OHDA), and group 4 (QA + 6-OHDA). The double toxin administration resulted in reduction of the spontaneous and the amphetamine-induced ipsiversive bias in the 6-OHDA group and in a reduction of the apomorphine-induced ipsiversive rotations in the QA group. Simultaneous QA and 6-OHDA also reduced the thigmotactic bias observed in the 6-OHDA rats. Combined toxin elicited a nonsignificant contralateral deficit in paw reaching but a significant deficit on the ipsilateral side. Histopathology revealed a significant reduction of the lesioned striatal surface (-27%) with neuronal loss and increased astrogliosis in group 4 compared to group 2, consistent with an exacerbation of QA toxicity by additional 6-OHDA. By contrast, the mean loss of the TH-positive neurons in the ipsilateral substantia nigra pars compacta (SNc) of group 4 was less marked (-15%) than in the 6-OHDA group (-36%), indicating a possible protective action of intrastriatal QA upon 6-OHDA retrograde SNc degeneration. This study shows that a combined unilateral intrastriatal administration of QA and 6-OHDA may serve as a model of early stage SND which is more suitable for early therapeutic interventions.
Collapse
Affiliation(s)
- I Ghorayeb
- Laboratoire de Neurophysiologie, CNRS-UMR 5543, Université Victor Segalen Bordeaux 2, Bordeaux Cedex, 33076, France
| | | | | | | | | | | | | |
Collapse
|