1
|
Swindell WR, Bojanowski K, Kindy MS, Chau RMW, Ko D. GM604 regulates developmental neurogenesis pathways and the expression of genes associated with amyotrophic lateral sclerosis. Transl Neurodegener 2018; 7:30. [PMID: 30524706 PMCID: PMC6276193 DOI: 10.1186/s40035-018-0135-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 10/21/2018] [Indexed: 12/11/2022] Open
Abstract
Background Amyotrophic lateral sclerosis (ALS) is currently an incurable disease without highly effective pharmacological treatments. The peptide drug GM604 (GM6 or Alirinetide) was developed as a candidate ALS therapy, which has demonstrated safety and good drug-like properties with a favorable pharmacokinetic profile. GM6 is hypothesized to bolster neuron survival through the multi-target regulation of developmental pathways, but mechanisms of action are not fully understood. Methods This study used RNA-seq to evaluate transcriptome responses in SH-SY5Y neuroblastoma cells following GM6 treatment (6, 24 and 48 h). Results We identified 2867 protein-coding genes with expression significantly altered by GM6 (FDR < 0.10). Early (6 h) responses included up-regulation of Notch and hedgehog signaling components, with increased expression of developmental genes mediating neurogenesis and axon growth. Prolonged GM6 treatment (24 and 48 h) altered the expression of genes contributing to cell adhesion and the extracellular matrix. GM6 further down-regulated the expression of genes associated with mitochondria, inflammatory responses, mRNA processing and chromatin organization. GM6-increased genes were located near GC-rich motifs interacting with C2H2 zinc finger transcription factors, whereas GM6-decreased genes were located near AT-rich motifs associated with helix-turn-helix homeodomain factors. Such motifs interacted with a diverse network of transcription factors encoded by GM6-regulated genes (STAT3, HOXD11, HES7, GLI1). We identified 77 ALS-associated genes with expression significantly altered by GM6 treatment (FDR < 0.10), which were known to function in neurogenesis, axon guidance and the intrinsic apoptosis pathway. Conclusions Our findings support the hypothesis that GM6 acts through developmental-stage pathways to influence neuron survival. Gene expression responses were consistent with neurotrophic effects, ECM modulation, and activation of the Notch and hedgehog neurodevelopmental pathways. This multifaceted mechanism of action is unique among existing ALS drug candidates and may be applicable to multiple neurodegenerative diseases. Electronic supplementary material The online version of this article (10.1186/s40035-018-0135-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- William R Swindell
- 1Heritage College of Osteopathic Medicine, Ohio University, Athens, OH USA
| | | | - Mark S Kindy
- 3Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL USA.,4James A. Haley VAMC, Tampa, FL USA
| | | | - Dorothy Ko
- Genervon Biopharmaceuticals LLC, Pasadena, CA USA
| |
Collapse
|
2
|
Hashikawa N, Utaka Y, Ogawa T, Tanoue R, Morita Y, Yamamoto S, Yamaguchi S, Kayano M, Zamami Y, Hashikawa-Hobara N. HSP105 prevents depression-like behavior by increasing hippocampal brain-derived neurotrophic factor levels in mice. SCIENCE ADVANCES 2017; 3:e1603014. [PMID: 28580422 PMCID: PMC5451194 DOI: 10.1126/sciadv.1603014] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 04/07/2017] [Indexed: 06/07/2023]
Abstract
Heat shock proteins (HSPs) are stress-induced chaperones that are involved in neurological disease. Although increasingly implicated in behavioral disorders, the mechanisms of HSP action, and the relevant functional pathways, are still unclear. We examined whether oral administration of geranylgeranylacetone (GGA), a known HSP inducer, produced an antidepressant effect in a social defeat stress model of depression in mice. We also investigated the possible molecular mechanisms involved, particularly focusing on hippocampal neurogenesis and neurotrophic factor expression. In stressed mice, hippocampal HSP105 expression decreased. However, administration of GGA increased HSP105 expression and improved depression-like behavior, induced hippocampal cell proliferation, and elevated brain-derived neurotrophic factor (BDNF) levels in mouse hippocampus. Co-treatment with GGA and the BDNF receptor inhibitor K252a suppressed the antidepressant effects of GGA. HSP105 knockdown decreased BDNF mRNA levels in HT22 hippocampal cell lines and hippocampal tissue and inhibited the GGA-mediated antidepressant effect. These observations suggest that GGA administration is a therapeutic candidate for depressive diseases by increasing hippocampal BDNF levels via HSP105 expression.
Collapse
Affiliation(s)
- Naoya Hashikawa
- Department of Life Science, Okayama University of Science, 1-1 Ridai-cho, Kita-ku, Okayama, Japan
| | - Yuta Utaka
- Department of Life Science, Okayama University of Science, 1-1 Ridai-cho, Kita-ku, Okayama, Japan
| | - Takumi Ogawa
- Department of Life Science, Okayama University of Science, 1-1 Ridai-cho, Kita-ku, Okayama, Japan
| | - Ryo Tanoue
- Department of Life Science, Okayama University of Science, 1-1 Ridai-cho, Kita-ku, Okayama, Japan
| | - Yuna Morita
- Department of Life Science, Okayama University of Science, 1-1 Ridai-cho, Kita-ku, Okayama, Japan
| | - Sayumi Yamamoto
- Department of Life Science, Okayama University of Science, 1-1 Ridai-cho, Kita-ku, Okayama, Japan
| | - Satoru Yamaguchi
- Department of Life Science, Okayama University of Science, 1-1 Ridai-cho, Kita-ku, Okayama, Japan
| | - Masafumi Kayano
- Department of Emergency Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, Japan
| | - Yoshito Zamami
- Department of Emergency Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, Japan
- Department of Clinical Pharmacy, Institute of Biomedical Sciences, Tokushima University Graduate School, 2-50-1 Kuramoto-cho, Tokushima, Japan
| | - Narumi Hashikawa-Hobara
- Department of Life Science, Okayama University of Science, 1-1 Ridai-cho, Kita-ku, Okayama, Japan
| |
Collapse
|
3
|
Dunbar GL, Sandstrom MI, Rossignol J, Lescaudron L. Neurotrophic Enhancers as Therapy for Behavioral Deficits in Rodent Models of Huntington's Disease: Use of Gangliosides, Substituted Pyrimidines, and Mesenchymal Stem Cells. ACTA ACUST UNITED AC 2016; 5:63-79. [PMID: 16801683 DOI: 10.1177/1534582306289367] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The interest in using neurotrophic factors as potential treatments for neurodegenerative disorders, such as Huntington's disease, has grown in the past decade. A major impediment for the clinical utility of neurotrophic factors is their inability to cross the blood-brain barrier in therapeutically significant amounts. Although several novel mechanisms for delivering exogenous neurotrophins to the brain have been developed, most of them involve invasive procedures or present significant risks. One approach to circumventing these problems is using therapeutic agents that can be administered systemically and have the ability to enhance the activity of neurotrophic factors. This review highlights the use of gangliosides, substituted pyrimidines, and mesenchymal stem cells as neurotrophic enhancers that have significant therapeutic potential while avoiding the pitfalls of delivering exogenous neurotrophic factors through the blood-brain barrier. The review focuses on the potential of these neurotrophic enhancers for treating the behavioral deficits in rodent models of Huntington's disease.
Collapse
|
4
|
Calcitonin gene-related peptide pre-administration acts as a novel antidepressant in stressed mice. Sci Rep 2015; 5:12559. [PMID: 26251188 PMCID: PMC4528222 DOI: 10.1038/srep12559] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 07/03/2015] [Indexed: 12/21/2022] Open
Abstract
Calcitonin gene-related peptide (CGRP) is a neuropeptide that has potent vasodilator properties and is involved in various behavioral disorders. The relationship between CGRP and depression-like behavior is unclear. In this study, we used chronically stressed mice to investigate whether CGRP is involved in depression-like behavior. Each mouse was exposed to restraint and water immersion stress for 15 days. After stress exposure, mice were assessed using behavioral tests: open field test, forced swim test and sucrose preference test. Serum corticosterone levels, hippocampal proliferation and mRNA expression of neurotrophins were measured. After stress exposure, mice exhibited depression-like behavior and decreased CGRP mRNA levels in the hippocampus. Although intracerebroventricular CGRP administration (0.5 nmol) did not alter depression-like behavior after 15-day stress exposure, a single CGRP administration into the brain, before the beginning of the 15-day stress exposure, normalized the behavioral dysfunctions and increased nerve growth factor (Ngf) mRNA levels in stressed mice. Furthermore, in the mouse E14 hippocampal cell line, CGRP treatment induced increased expression of Ngf mRNA. The NGF receptor inhibitor K252a inhibited CGRP's antidepressant-like effects in stressed mice. These results suggest that CGRP expression in the mouse hippocampus is associated with depression-like behavior and changes in Ngf mRNA levels.
Collapse
|
5
|
Hashikawa N, Ogawa T, Sakamoto Y, Ogawa M, Matsuo Y, Zamami Y, Hashikawa-Hobara N. Time Course of Behavioral Alteration and mRNA Levels of Neurotrophic Factor Following Stress Exposure in Mouse. Cell Mol Neurobiol 2015; 35:807-17. [PMID: 25820756 PMCID: PMC11486325 DOI: 10.1007/s10571-015-0174-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 03/07/2015] [Indexed: 10/23/2022]
Abstract
Stress is known to affect neurotrophic factor expression, which induces depression-like behavior. However, whether there are time-dependent changes in neurotrophic factor mRNA expression following stress remains unclear. In the present study, we tested whether chronic stress exposure induces long-term changes in depression-related behavior, serum corticosterone, and hippocampal proliferation as well as neurotrophic factor family mRNA levels, such as brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), neurotrophin-3 (NT-3), and ciliary neurotrophic factor (CNTF), in the mouse hippocampus. The mRNA level of neurotrophic factors (BDNF, NGF, NT-3, and CNTF) was measured using the real-time PCR. The serum corticosterone level was evaluated by enzyme-linked immunosorbent assay, and, for each subject, the hippocampal proliferation was examined by 5-bromo-2-deoxyuridine immunostaining. Mice exhibited depression-like behavior in the forced-swim test (FST) and decreased BDNF mRNA and hippocampal proliferation in the middle of the stress exposure. After 15 days of stress exposure, we observed increased immobility in the FST, serum corticosterone levels, and BDNF mRNA levels and degenerated hippocampal proliferation, maintained for at least 2 weeks. Anhedonia-like behavior in the sucrose preference test and NGF mRNA levels were decreased following 15 days of stress. NGF mRNA levels were significantly higher 1 week after stress exposure. The current data demonstrate that chronic stress exposure induces prolonged BDNF and NGF mRNA changes and increases corticosterone levels and depression-like behavior in the FST, but does not alter other neurotrophic factors or performance in the sucrose preference test.
Collapse
Affiliation(s)
- Naoya Hashikawa
- Department of Life Science, Okayama University of Science, 1-1 Ridai-cho, Kita-ku, Okayama, 700-0005 Japan
| | - Takumi Ogawa
- Department of Life Science, Okayama University of Science, 1-1 Ridai-cho, Kita-ku, Okayama, 700-0005 Japan
| | - Yusuke Sakamoto
- Department of Life Science, Okayama University of Science, 1-1 Ridai-cho, Kita-ku, Okayama, 700-0005 Japan
| | - Mami Ogawa
- Department of Life Science, Okayama University of Science, 1-1 Ridai-cho, Kita-ku, Okayama, 700-0005 Japan
| | - Yumi Matsuo
- Department of Life Science, Okayama University of Science, 1-1 Ridai-cho, Kita-ku, Okayama, 700-0005 Japan
| | - Yoshito Zamami
- Medicinal Drug Design, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama, Japan
| | - Narumi Hashikawa-Hobara
- Department of Life Science, Okayama University of Science, 1-1 Ridai-cho, Kita-ku, Okayama, 700-0005 Japan
| |
Collapse
|
6
|
Emerich DF, Orive G, Thanos C, Tornoe J, Wahlberg LU. Encapsulated cell therapy for neurodegenerative diseases: from promise to product. Adv Drug Deliv Rev 2014; 67-68:131-41. [PMID: 23880505 DOI: 10.1016/j.addr.2013.07.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 05/31/2013] [Accepted: 07/12/2013] [Indexed: 12/27/2022]
Abstract
Delivering therapeutic molecules, including trophic factor proteins, across the blood brain barrier to the brain parenchyma to treat chronic neurodegenerative diseases remains one of the great challenges in biology. To be effective, delivery needs to occur in a long-term and stable manner at sufficient quantities directly to the target region in a manner that is selective but yet covers enough of the target site to be efficacious. One promising approach uses cellular implants that produce and deliver therapeutic molecules directly to the brain region of interest. Implanted cells can be precisely positioned into the desired region and can be protected from host immunological attack by encapsulating them and by surrounding them within an immunoisolatory, semipermeable capsule. In this approach, cells are enclosed within a semiporous capsule with a perm selective membrane barrier that admits oxygen and required nutrients and releases bioactive cell secretions while restricting passage of larger cytotoxic agents from the host immune defense system. Recent advances in human cell line development have increased the levels of secreted therapeutic molecules from encapsulated cells, and membrane extrusion techniques have led to the first ever clinical demonstrations of long-term survival and function of encapsulated cells in the brain parenchyma. As such, cell encapsulation is capable of providing a targeted, continuous, de novo synthesized source of very high levels of therapeutic molecules that can be distributed over significant portions of the brain.
Collapse
|
7
|
Russmann V, Seeger N, Zellinger C, Hadamitzky M, Pankratova S, Wendt H, Bock E, Berezin V, Potschka H. The CNTF-derived peptide mimetic Cintrofin attenuates spatial-learning deficits in a rat post-status epilepticus model. Neurosci Lett 2013; 556:170-5. [DOI: 10.1016/j.neulet.2013.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 09/10/2013] [Accepted: 10/01/2013] [Indexed: 12/24/2022]
|
8
|
Quertainmont R, Cantinieaux D, Botman O, Sid S, Schoenen J, Franzen R. Mesenchymal stem cell graft improves recovery after spinal cord injury in adult rats through neurotrophic and pro-angiogenic actions. PLoS One 2012; 7:e39500. [PMID: 22745769 PMCID: PMC3380009 DOI: 10.1371/journal.pone.0039500] [Citation(s) in RCA: 161] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Accepted: 05/21/2012] [Indexed: 12/13/2022] Open
Abstract
Numerous strategies have been managed to improve functional recovery after spinal cord injury (SCI) but an optimal strategy doesn't exist yet. Actually, it is the complexity of the injured spinal cord pathophysiology that begets the multifactorial approaches assessed to favour tissue protection, axonal regrowth and functional recovery. In this context, it appears that mesenchymal stem cells (MSCs) could take an interesting part. The aim of this study is to graft MSCs after a spinal cord compression injury in adult rat to assess their effect on functional recovery and to highlight their mechanisms of action. We found that in intravenously grafted animals, MSCs induce, as early as 1 week after the graft, an improvement of their open field and grid navigation scores compared to control animals. At the histological analysis of their dissected spinal cord, no MSCs were found within the host despite their BrdU labelling performed before the graft, whatever the delay observed: 7, 14 or 21 days. However, a cytokine array performed on spinal cord extracts 3 days after MSC graft reveals a significant increase of NGF expression in the injured tissue. Also, a significant tissue sparing effect of MSC graft was observed. Finally, we also show that MSCs promote vascularisation, as the density of blood vessels within the lesioned area was higher in grafted rats. In conclusion, we bring here some new evidences that MSCs most likely act throughout their secretions and not via their own integration/differentiation within the host tissue.
Collapse
Affiliation(s)
- Renaud Quertainmont
- GIGA Neurosciences, Axonal Regeneration and Cephalic Pain Unit, University of Liege, Liege, Belgium
| | - Dorothée Cantinieaux
- GIGA Neurosciences, Axonal Regeneration and Cephalic Pain Unit, University of Liege, Liege, Belgium
| | - Olivier Botman
- GIGA Neurosciences, Axonal Regeneration and Cephalic Pain Unit, University of Liege, Liege, Belgium
| | - Selim Sid
- GIGA Neurosciences, Axonal Regeneration and Cephalic Pain Unit, University of Liege, Liege, Belgium
| | - Jean Schoenen
- GIGA Neurosciences, Axonal Regeneration and Cephalic Pain Unit, University of Liege, Liege, Belgium
| | - Rachelle Franzen
- GIGA Neurosciences, Axonal Regeneration and Cephalic Pain Unit, University of Liege, Liege, Belgium
- * E-mail:
| |
Collapse
|
9
|
Ciliary neurotrophic factor cell-based delivery prevents synaptic impairment and improves memory in mouse models of Alzheimer's disease. J Neurosci 2010; 30:7516-27. [PMID: 20519526 DOI: 10.1523/jneurosci.4182-09.2010] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The development of novel therapeutic strategies for Alzheimer's disease (AD) represents one of the biggest unmet medical needs today. Application of neurotrophic factors able to modulate neuronal survival and synaptic connectivity is a promising therapeutic approach for AD. We aimed to determine whether the loco-regional delivery of ciliary neurotrophic factor (CNTF) could prevent amyloid-beta (Abeta) oligomer-induced synaptic damages and associated cognitive impairments that typify AD. To ensure long-term administration of CNTF in the brain, we used recombinant cells secreting CNTF encapsulated in alginate polymers. The implantation of these bioreactors in the brain of Abeta oligomer-infused mice led to a continuous secretion of recombinant CNTF and was associated with the robust improvement of cognitive performances. Most importantly, CNTF led to full recovery of cognitive functions associated with the stabilization of synaptic protein levels in the Tg2576 AD mouse model. In vitro as well as in vivo, CNTF activated a Janus kinase/signal transducer and activator of transcription-mediated survival pathway that prevented synaptic and neuronal degeneration. These preclinical studies suggest that CNTF and/or CNTF receptor-associated pathways may have AD-modifying activity through protection against progressive Abeta-related memory deficits. Our data also encourage additional exploration of ex vivo gene transfer for the prevention and/or treatment of AD.
Collapse
|
10
|
Dey ND, Bombard MC, Roland BP, Davidson S, Lu M, Rossignol J, Sandstrom MI, Skeel RL, Lescaudron L, Dunbar GL. Genetically engineered mesenchymal stem cells reduce behavioral deficits in the YAC 128 mouse model of Huntington's disease. Behav Brain Res 2010; 214:193-200. [PMID: 20493905 DOI: 10.1016/j.bbr.2010.05.023] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2010] [Revised: 05/07/2010] [Accepted: 05/12/2010] [Indexed: 12/13/2022]
Abstract
The purpose of this study was to evaluate the therapeutic effects of the transplantation of bone-marrow mesenchymal stem cells (MSCs), genetically engineered to over-express brain-derived neurotrophic factor (BDNF) or nerve growth factor (NGF) on motor deficits and neurodegeneration in YAC 128 transgenic mice. MSCs, harvested from mouse femurs, were genetically engineered to over-express BDNF and/or NGF and these cells, or the vehicle solution, were injected into the striata of four-month old YAC 128 transgenic and wild-type mice. Assessments of motor ability on the rotarod and the severity of clasping were made one day prior to transplantation and once monthly, thereafter, to determine the effects of the transplanted cells on motor function. The mice were sacrificed at 13-months of age for immunohistological examination. All YAC 128 mice receiving transplants had reduced clasping, relative to vehicle-treated YAC 128 mice, while YAC 128 mice that were transplanted with MSCs which were genetically engineered to over-express BDNF, had the longest latencies on the rotarod and the least amount of neuronal loss within the striatum of the YAC 128 mice. These results indicate that intrastriatal transplantation of MSCs that over-express BDNF may create an environment within the striatum that slows neurodegenerative processes and provides behavioral sparing in the YAC 128 mouse model of HD. Further research on the long-term safety and efficacy of this approach is needed before its potential clinical utility can be comprehensively assessed.
Collapse
Affiliation(s)
- Nicholas D Dey
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI 48858, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Skinner SJM, Geaney MS, Lin H, Muzina M, Anal AK, Elliott RB, Tan PLJ. Encapsulated living choroid plexus cells: potential long-term treatments for central nervous system disease and trauma. J Neural Eng 2009; 6:065001. [PMID: 19850973 DOI: 10.1088/1741-2560/6/6/065001] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
In neurodegenerative disease and in acute brain injury, there is often local up-regulation of neurotrophin production close to the site of the lesion. Treatment by direct injection of neurotrophins and growth factors close to these lesion sites has repeatedly been demonstrated to improve recovery. It has therefore been proposed that transplanting viable neurotrophin-producing cells close to the trauma lesion, or site of degenerative disease, might provide a novel means for continuous delivery of these molecules directly to the site of injury or to a degenerative region. The aim of this paper is to summarize recent published information and present new experimental data that indicate that long-lasting therapeutic implants of choroid plexus (CP) neuroepithelium may be used to treat brain disease. CP produces and secretes numerous biologically active neurotrophic factors (NT). New gene microarray and proteomics data presented here indicate that many other anti-oxidant, anti-toxin and neuronal support proteins are also produced and secreted by CP cells. In the healthy brain, these circulate in the cerebrospinal fluid through the brain and spinal cord, maintaining neuronal networks and associated cells. Recent publications describe how transplanted CP cells and tissue, either free or in an immunoprotected encapsulated form, can effectively deliver therapeutic molecules when placed near the lesion or site of degenerative disease in animal models. Using simple techniques, CP neuroepithelial cell clusters in suspension culture were very durable, remaining viable for 6 months or more in vitro. The cell culture conditions had little effect on the wide range and activity of genes expressed and proteins secreted. Recently, completed experiments show that implanting CP within alginate-poly-ornithine capsules effectively protected these xenogeneic cells from the host immune system and allowed their survival for 6 months or more in the brains of rats, causing no adverse effects. Previously reported evidence demonstrated that CP cells support the survival and differentiation of neuronal cells in vitro and effectively treat acute brain injury and disease in rodents and non-human primates in vivo. The accumulated preclinical data together with the long-term survival of implanted encapsulated cells in vivo provide a sound base for the investigation of these treatments for chronic inherited and established neurodegenerative conditions.
Collapse
Affiliation(s)
- S J M Skinner
- Living Cell Technologies NZ Ltd, PO Box 23 566, Hunters Corner, Manukau 2025, New Zealand
| | | | | | | | | | | | | |
Collapse
|
12
|
Emerich DF, Mooney DJ, Storrie H, Babu RS, Kordower JH. Injectable hydrogels providing sustained delivery of vascular endothelial growth factor are neuroprotective in a rat model of Huntington's disease. Neurotox Res 2009; 17:66-74. [PMID: 19588214 DOI: 10.1007/s12640-009-9079-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Revised: 06/19/2009] [Accepted: 06/19/2009] [Indexed: 11/26/2022]
Abstract
Vascular endothelial growth factor (VEGF) is a potent peptide with well-documented pro-angiogenic effects. Recently, it has also become clear that exogenous administration of VEGF is neuroprotective in animal models of central nervous system diseases. In the present study, VEGF was incorporated into a sustained release hydrogel delivery system to examine its potential benefits in a rat model of Huntington's disease (HD). The VEGF-containing hydrogel was stereotaxically injected into the striatum of adult rats. Three days later, quinolinic acid (QA; 225 nmol) was injected into the ipsilateral striatum to produce neuronal loss and behavioral deficits that mimic those observed in HD. Two weeks after surgery, animals were tested for motor function using the placement and cylinder tests. Control animals received either QA alone or QA plus empty hydrogel implants. Behavioral testing confirmed that the QA lesion resulted in significant deficits in the ability of the control animals to use their contralateral forelimb. In contrast, the performance of those animals receiving VEGF was significantly improved relative to controls with only modest motor impairments observed. Stereological counts of NeuN-positive neurons throughout the striatum demonstrated that VEGF implants significantly protected against the loss of striatal neurons induced by QA. These data are the first to demonstrate that VEGF can be used to protect striatal neurons from excitotoxic damage in a rat model of HD.
Collapse
Affiliation(s)
- Dwaine F Emerich
- InCytu, Inc, 701 George Washington Highway, Lincoln, RI, 02865, USA.
| | | | | | | | | |
Collapse
|
13
|
Barchet TM, Amiji MM. Challenges and opportunities in CNS delivery of therapeutics for neurodegenerative diseases. Expert Opin Drug Deliv 2009; 6:211-25. [PMID: 19290842 DOI: 10.1517/17425240902758188] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
With an increase in lifespan and changing population demographics, the incidence of central nervous system (CNS) diseases is expected to increase significantly in the 21st century. The most challenging of the CNS diseases are neurodegenerative diseases, characterized by age-related gradual decline in neurological function, often accompanied by neuronal death. Alzheimer's disease, Parkinson's disease and Huntington's disease are some examples of neurodegenerative diseases and have been well described in terms of disease mechanisms and pathology. However, successful treatment strategies for neurodegenerative diseases have so far been limited. Delivery of drugs into the CNS is one of the most challenging problems faced in the treatment of neurodegeneration. In this review, we describe the difficulties with CNS therapy, especially with the use of biological macromolecules, such as proteins and nucleic acid constructs. CNS therapeutics also represents a huge opportunity and examples of strategies that can enhance therapeutic delivery for the treatment of neurodegenerative diseases are emphasized. It is anticipated that with an increase in biological understanding of neurodegenerative diseases, there will be even more therapeutic opportunities. As such, these delivery strategies have a very important role to play in the future in the translation of CNS therapeutics from bench to bedside.
Collapse
Affiliation(s)
- Thomas M Barchet
- Department of Pharmaceutical Sciences, Northeastern University, School of Pharmacy, 110 Mugar Life Sciences Building, Boston, Massachussets, MA 02115, USA
| | | |
Collapse
|
14
|
Heberlein A, Bleich S, Bayerlein K, Frieling H, Gröschl M, Kornhuber J, Hillemacher T. NGF plasma levels increase due to alcohol intoxication and decrease during withdrawal. Psychoneuroendocrinology 2008; 33:999-1003. [PMID: 18639986 DOI: 10.1016/j.psyneuen.2008.05.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2008] [Revised: 03/18/2008] [Accepted: 05/01/2008] [Indexed: 11/19/2022]
Abstract
Recent studies show that alcohol dependence is associated with alterations in plasma levels of nerve growth factor (NGF). The aim of this study was to further elucidate reported alterations in NGF plasma levels during alcohol intoxication and withdrawal. Therefore, we assessed NGF plasma levels by enzyme-linked immunosorbent assay (ELISA) on admission (day 0) and day 7 of alcohol withdrawal in male alcohol dependent patients (n=75) in comparison to healthy controls (n=44). We found significant higher (U=1005.0, p<0.001) NGF plasma levels in the alcohol-dependent patients. Subgroup analysis showed significant higher (U=-2.934, p=0.003) NGF plasma levels in patients suffering from acute alcohol intoxication (group A) than in early abstinent patients (group B). From day 0 to day 7 of alcohol withdrawal NGF plasma levels decreased significantly in both groups (group A: Z=-3.118, p=0.002, group B: Z=-2.103, p=0.035). Our results suggest that acute alcohol intoxication is associated with an increase in NGF plasma levels, which decrease during alcohol withdrawal. These results suggest that NGF plasma levels increase as part of a regulation mechanism that counteracts alcohol intoxication.
Collapse
Affiliation(s)
- Annemarie Heberlein
- Department of Psychiatry and Psychotherapy, University Hospital Erlangen-Nuremberg, Schwabachanlage 6, D-91054 Erlangen, Germany.
| | | | | | | | | | | | | |
Collapse
|
15
|
Ormerod BK, Palmer TD, Caldwell MA. Neurodegeneration and cell replacement. Philos Trans R Soc Lond B Biol Sci 2008; 363:153-70. [PMID: 17331894 PMCID: PMC2605492 DOI: 10.1098/rstb.2006.2018] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The past decade has witnessed ground-breaking advances in human stem cell biology with scientists validating adult neurogenesis and establishing methods to isolate and propagate stem cell populations suitable for transplantation. These advances have forged promising strategies against human neurodegenerative diseases. For example, growth factor administration could stimulate intrinsic repair from endogenous neural stem cells, and cultured stem cells engineered into biopumps could be transplanted to deliver neuroprotective or restorative agents. Stem cells could also be transplanted to generate new neural elements that augment and potentially replace degenerating central nervous system (CNS) circuitry. Early efforts in neural tissue transplantation have shown that these strategies can improve functional outcome, but the ultimate success of clinical stem cell-based strategies will depend on detailed understanding of stem cell biology in the degenerating brain and detailed evaluation of their functional efficacy and safety in preclinical animal models.
Collapse
Affiliation(s)
- Brandi K Ormerod
- Department of Neurosurgery, Stanford University300 Pasteur Drive, Boswell Building, A301, Stanford, CA 94305-5327, USA
| | - Theo D Palmer
- Department of Neurosurgery, Stanford University300 Pasteur Drive, Boswell Building, A301, Stanford, CA 94305-5327, USA
| | - Maeve A Caldwell
- Centre for Brain Repair, University of Cambridge School of Clinical MedicineAddenbrooke's Hospital, Box 111, Hills Road, Cambridge CB2 2SP, UK
- Author and address for correspondence: Laboratory for Integrative Neuroscience and Endocrinology, Dorothy Hodgkin Building, University of Bristol, Whitson Street, Bristol BS1 3NY, UK ()
| |
Collapse
|
16
|
Abstract
The mainstays of Parkinson's disease (PD) treatment remain symptomatic, including initial dopamine replacement and subsequent deep brain stimulation, however, neither of these approaches is neuroprotective. Neurotrophic factors - proteins that activate cell signalling pathways regulating neuronal survival, differentiation, growth and regeneration - represent an alternative for treating dopaminergic neurons in PD but are difficult to administer clinically because they do not pass through the blood-brain barrier. Glial cell line-derived neurotrophic factor (GDNF) has potent neurotrophic effects particularly but not exclusively on dopaminergic neurons; in animal models of PD, it has consistently demonstrated both neuroprotective and neuroregenerative effects when provided continuously, either by means of a viral vector or through continuous infusion either into the cerebral ventricles (ICV) or directly into the denervated putamen. This led to a human PD study in which GDNF was administered by monthly bolus intracerebroventricular injections, however, no clinical benefit resulted, probably because of the limited penetration to the target brain areas, and instead significant side effects occurred. In an open-label study of continuous intraputamenal GDNF infusion in five patients (one unilaterally and four bilaterally), we reported excellent tolerance, few side effects and clinical benefit evident within three months of the commencement of treatment. The clinical improvement was sustained and progressive, and by 24-months patients demonstrated a 57 and 63% improvement in their off-medication motor and activities of daily living UPDRS subscores, respectively, with clear benefit in dyskinesias. The benefit was associated with a significant increase in putamenal 18F-dopa uptake on positron emission tomography (PET), and in one patient coming to autopsy after 43 months of unilateral infusion there was evident increased tyrosine hydroxylase immunopositive nerve fibres in the infused putamen. A second open trial in 10 patients using unilateral intraputamenal GDNF infusions has also demonstrated a greater than 30% bilateral benefit in both on- and off-medication scores at 24 weeks. Based on our 6-month results, a randomized controlled clinical trial was conducted to confirm the open-label results, however, GDNF infusion over 6-months did not confer the predetermined level of clinical benefit to patients with PD despite increased 18F-dopa uptake surrounding the catheter tip. It is possible that technical differences between this trial and the positive open label studies contributed to this negative outcome.
Collapse
Affiliation(s)
- N K Patel
- Institute of Neurosciences, Frenchay Hospital, Bristol, UK
| | | |
Collapse
|
17
|
Abstract
Neurotrophic factors (NTFs) have the unique potential to support neuronal survival and to augment neuronal function in the injured and diseased nervous system. Numerous studies conducted over the last 20 years have provided evidence for the potent therapeutic potential of NTFs in animal models of neurodegenerative diseases. However, major obstacles for the therapeutic use of NTFs are the inability to deliver proteins across the blood-brain-barrier, and dose-limiting adverse effects resulting from the broad exposure of nontargeted structures to NTFs. Two recent developments have allowed NTFs' promise to be truly tested for the first time: first, recent improvements in viral vectors that allow the targeted delivery of NTFs while providing a long-lasting supply and sufficient therapeutic doses of NTFs; and second, improved animal models developed in recent years. In this review, we will discuss some of the potential therapeutic applications of NTFs in neurodegenerative diseases and the potential contribution of disturbed neurotrophic factor signaling to neurodegenerative diseases.
Collapse
Affiliation(s)
- Armin Blesch
- Department of Neurosciences-0626, Center for Neural Repair, University of California, San Diego, La Jolla, California 92093-0626, USA.
| |
Collapse
|
18
|
Abstract
Neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease trigger neuronal cell death through endogenous suicide pathways. Surprisingly, although the cell death itself may occur relatively late in the course of the degenerative process, the mediators of the underlying cell-death pathways have shown promise as potential therapeutic targets.
Collapse
Affiliation(s)
- Dale E Bredesen
- Buck Institute for Age Research, 8001 Redwood Boulevard, Novato, California 94945, USA.
| | | | | |
Collapse
|
19
|
Emerich DF, Thanos CG, Goddard M, Skinner SJM, Geany MS, Bell WJ, Bintz B, Schneider P, Chu Y, Babu RS, Borlongan CV, Boekelheide K, Hall S, Bryant B, Kordower JH. Extensive neuroprotection by choroid plexus transplants in excitotoxin lesioned monkeys. Neurobiol Dis 2006; 23:471-80. [PMID: 16777422 DOI: 10.1016/j.nbd.2006.04.014] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2006] [Revised: 04/04/2006] [Accepted: 04/07/2006] [Indexed: 11/20/2022] Open
Abstract
Huntington's disease (HD) results from degeneration of striatal neurons. Choroid plexus (CP) cells secrete neurotrophic factors, and CP transplants are neuroprotective in rat models of HD. To determine if similar neuroprotective effects could be obtained in primates, porcine CP was encapsulated in alginate capsules. PCR confirmed that the CP cells expressed transthyretin and immunocytochemistry demonstrated typical ZO-1 and tubulin staining. In vitro, CP conditioned media enhanced the survival and preserved neurite number and length on serum deprived neurons. Cynomolgus primates were transplanted with CP-loaded capsules into the caudate and putamen followed by quinolinic acid (QA) lesions 1 week later. Control monkeys received empty capsules plus QA. Choroid plexus transplants significantly protected striatal neurons as revealed by stereological counts of NeuN-positive neurons (8% loss vs. 43% in controls) and striatum volume (10% decrease vs. 40% in controls). These data indicate that CP transplants might be useful for preventing the degeneration of neurons in HD.
Collapse
|
20
|
Janak PH, Wolf FW, Heberlein U, Pandey SC, Logrip ML, Ron D. BIG news in alcohol addiction: new findings on growth factor pathways BDNF, insulin, and GDNF. Alcohol Clin Exp Res 2006; 30:214-21. [PMID: 16441270 DOI: 10.1111/j.1530-0277.2006.00026.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In recent years, it has become clear that growth factors are not only critical for the development of the central nervous system (CNS) but may also be important contributors to other neuronal functions in the adult brain. This symposium, presented at the 2005 RSA meeting, discussed evidence to support the hypothesis that alterations in growth factor pathways produce dramatic changes in the effects of alcohol on the CNS. The 4 speakers showed that the behavioral effects of alcohol in the adult are regulated by 3 growth factors, insulin, glial cell line-derived neurotrophic factor (GDNF), and brain-derived neurotrophic factor (BDNF). Dr. Wolf from the Heberlein laboratory presented findings obtained from genetic manipulations in Drosophila melanogaster, demonstrating that the insulin pathway controls sensitivity to the intoxicating effects of alcohol. Marian Logrip from the Ron and Janak laboratories presented evidence obtained in rodents that low concentrations of alcohol increase the expression of BDNF in the brain to regulate alcohol consumption. Dr. Pandey showed that amygdalar BDNF regulates alcohol's anxiolytic effects and preference. Finally, Dr. Janak presented evidence that increases in the expression of GDNF in the midbrain reduce alcohol self-administration in rats.
Collapse
Affiliation(s)
- Patricia H Janak
- Ernest Gallo Clinic and Research Center, University of California, San Francisco, California, USA
| | | | | | | | | | | |
Collapse
|
21
|
Skinner SJM, Geaney MS, Rush R, Rogers ML, Emerich DF, Thanos CG, Vasconcellos AV, Tan PLJ, Elliott RB. Choroid plexus transplants in the treatment of brain diseases. Xenotransplantation 2006; 13:284-8. [PMID: 16768721 DOI: 10.1111/j.1399-3089.2006.00310.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The choroid plexus (CP) produces and secretes numerous biologically active neurotrophic factors into the cerebrospinal fluid (CSF). These circulate throughout the brain and spinal cord, maintaining neuronal networks and associated cells. In neurodegenerative disease and in acute brain injury there is local up-regulation of neurotrophin production close to the site of the lesion. Treatment by direct injection of neurotrophins and growth factors close to these lesion sites has repeatedly been demonstrated to improve recovery. It has therefore been proposed that transplanting viable choroid plexus cells close to the lesion might provide a novel means for continuous delivery of these molecules directly to the site of injury. Recent publications describe how transplanted CP, either free or in an immunoprotected encapsulated form, deliver therapeutic molecules to the desired site. This review briefly describes the accumulated evidence that CP cells support neuronal cells in vitro and have therapeutic properties when transplanted to treat acute and chronic brain disease and injury in animal models.
Collapse
|
22
|
Handley OJ, Naji JJ, Dunnett SB, Rosser AE. Pharmaceutical, cellular and genetic therapies for Huntington's disease. Clin Sci (Lond) 2005; 110:73-88. [PMID: 16336206 DOI: 10.1042/cs20050148] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
HD (Huntington's disease) is a devastating neurodegenerative disorder caused by a polyglutamine expansion in the gene encoding the huntingtin protein. Presently, there is no known cure for HD and existing symptomatic treatments are limited. However, recent advances have identified multiple pathological mechanisms involved in HD, some of which have now become the focus of therapeutic intervention. In this review, we consider progress made towards developing safe and effective pharmaceutical-, cell- and genetic-based therapies, and discuss the extent to which some of these therapies have been successfully translated into clinical trials. These new prospects offer hope for delaying and possibly halting this debilitating disease.
Collapse
Affiliation(s)
- Olivia J Handley
- The Brain Repair Group, School of Biosciences, Cardiff University, Cardiff CF10 3US, UK.
| | | | | | | |
Collapse
|
23
|
|
24
|
Sonkusare SK, Kaul CL, Ramarao P. Dementia of Alzheimer's disease and other neurodegenerative disorders--memantine, a new hope. Pharmacol Res 2005; 51:1-17. [PMID: 15519530 DOI: 10.1016/j.phrs.2004.05.005] [Citation(s) in RCA: 185] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/19/2004] [Indexed: 11/28/2022]
Abstract
Alzheimer's disease is the fourth largest cause of death for people over 65 years of age. Dementia of Alzheimer's type is the commonest form of dementia, the other two forms being vascular dementia and mixed dementia. At present, the therapy of Alzheimer's disease is aimed at improving both, cognitive and behavioural symptoms and thereby, quality of life for the patients. Since the discovery of Alzheimer's disease by Alois Alzheimer, many pathological mechanisms have been proposed which led to the testing of various new treatments. Until recently the available drugs for the treatment of Alzheimer's disease are cholinesterase inhibitors, which have limited success because these drugs improve cognitive functions only in mild dementia and cannot stop the process of neurodegeneration. Moreover, drugs of this category show gastrointestinal side effects. As the cells of central and peripheral nervous system cannot regenerate, newer strategies are aimed at preserving the surviving neurons by preventing their degeneration. NMDA-receptor-mediated glutamate excitotoxicity plays a major role in Abeta-induced neuronal death. Hence, it was thought that NMDA receptors could be a promising target for preventing the progression of Alzheimer's disease. All the compounds synthesized initially in this category showed toxicity mainly because of their high affinity for NMDA receptors. Memantine (1-amino adamantane derivative), NMDA-receptor antagonist was reported to be effective therapeutically in Alzheimer's disease. It was available in Germany as well as European Union and has been approved for moderate to severe dementia in United States of America recently. It is an uncompetitive, moderate affinity antagonist of NMDA receptors that inhibits the pathological functions of NMDA receptors while physiological processes in learning and memory are unaffected. Memantine is also reported to have beneficial effects in other CNS disorders viz., Parkinson's disease (PD), stroke, epilepsy, CNS trauma, amyotrophic lateral sclerosis (ALS), drug dependence and chronic pain. Mechanisms of neuroprotection, preclinical and clinical evidence for effectiveness of memantine have been provided. Pharmacology and pharmacokinetics of memantine and other NMDA-receptor antagonists in comparison with currently approved drugs for dementia treatment have been discussed. The focus is on 'glutamate excitotoxicity' and glutamate receptors as drug target. Various other novel strategies for the treatment of dementia of neurodegenerative disorders have also been discussed.
Collapse
Affiliation(s)
- S K Sonkusare
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Mohali 160 062, India
| | | | | |
Collapse
|
25
|
Bloch J, Bachoud-Lévi AC, Déglon N, Lefaucheur JP, Winkel L, Palfi S, Nguyen JP, Bourdet C, Gaura V, Remy P, Brugières P, Boisse MF, Baudic S, Cesaro P, Hantraye P, Aebischer P, Peschanski M. Neuroprotective gene therapy for Huntington's disease, using polymer-encapsulated cells engineered to secrete human ciliary neurotrophic factor: results of a phase I study. Hum Gene Ther 2005; 15:968-75. [PMID: 15585112 DOI: 10.1089/hum.2004.15.968] [Citation(s) in RCA: 177] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Huntington's disease (HD) is a monogenic neurodegenerative disease that affects the efferent neurons of the striatum. The protracted evolution of the pathology over 15 to 20 years, after clinical onset in adulthood, underscores the potential of therapeutic tools that would aim at protecting striatal neurons. Proteins with neuroprotective effects in the adult brain have been identified, among them ciliary neurotrophic factor (CNTF), which protected striatal neurons in animal models of HD. Accordingly, we have carried out a phase I study evaluating the safety of intracerebral administration of this protein in subjects with HD, using a device formed by a semipermeable membrane encapsulating a BHK cell line engineered to synthesize CNTF. Six subjects with stage 1 or 2 HD had one capsule implanted into the right lateral ventricle; the capsule was retrieved and exchanged for a new one every 6 months, over a total period of 2 years. No sign of CNTF-induced toxicity was observed; however, depression occurred in three subjects after removal of the last capsule, which may have correlated with the lack of any future therapeutic option. All retrieved capsules were intact but contained variable numbers of surviving cells, and CNTF release was low in 13 of 24 cases. Improvements in electrophysiological results were observed, and were correlated with capsules releasing the largest amount of CNTF. This phase I study shows the safety, feasibility, and tolerability of this gene therapy procedure. Heterogeneous cell survival, however, stresses the need for improving the technique.
Collapse
Affiliation(s)
- J Bloch
- Swiss Federal Institute of Technology Lausanne, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Ohta K, Fujinami A, Kuno S, Sakakimoto A, Matsui H, Kawahara Y, Ohta M. Cabergoline stimulates synthesis and secretion of nerve growth factor, brain-derived neurotrophic factor and glial cell line-derived neurotrophic factor by mouse astrocytes in primary culture. Pharmacology 2004; 71:162-8. [PMID: 15161999 DOI: 10.1159/000077451] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2003] [Accepted: 12/10/2003] [Indexed: 11/19/2022]
Abstract
Neuroprotection is the primary concern in patients with newly diagnosed Parkinson's disease. The D2/weak D1 dopamine agonist cabergoline elicits neuroprotection by antioxidation and scavenging free radicals, and may protect neurons by up-regulating endogenous neurotrophic factors synthesis in the brain. In primary cultured mouse astrocytes, cabergoline 37 micromol/l immediately elevated concentrations of nerve growth factor, brain-derived neurotrophic factor, and glial cell line-derived neurotrophic factor (GDNF) in culture medium, reaching 9.9-, 2.6- and 30-fold, respectively, of control levels at 16 h. Relative mRNA levels were 3.0-, 1.5- and 1.9-fold, respectively, of controls at 3 h. These effects may be mediated partly by the dopamine D2 receptor. Cabergoline may be a good candidate for an inducer of GDNF, which may have neuroprotective and neurorestorative properties in dopaminergic nigral neurons.
Collapse
Affiliation(s)
- Kiyoe Ohta
- Clinical Research Center, Utano National Hospital, Kyoto, Japan
| | | | | | | | | | | | | |
Collapse
|
27
|
Vergara C, Ramirez B. CNTF, a pleiotropic cytokine: emphasis on its myotrophic role. ACTA ACUST UNITED AC 2004; 47:161-73. [PMID: 15572170 DOI: 10.1016/j.brainresrev.2004.07.010] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2004] [Indexed: 11/19/2022]
Abstract
Ciliary neurotrophic factor (CNTF) is a cytokine whose neurotrophic and differentiating effects over cells in the central nervous system (CNS) have been clearly demonstrated. This article summarizes the general characteristics of CNTF, its receptor and the signaling pathway that it activates and focuses on its effects over skeletal muscle, one of its major target tissues outside the central nervous system. The evidence for the existence of other molecules that signal through the same complex as CNTF is also reviewed.
Collapse
Affiliation(s)
- Cecilia Vergara
- Biology Department, Faculty of Sciences, University of Chile, Casilla 653, Santiago, Chile.
| | | |
Collapse
|
28
|
Borlongan CV, Skinner SJM, Geaney M, Vasconcellos AV, Elliott RB, Emerich DF. Neuroprotection by encapsulated choroid plexus in a rodent model of Huntington’s disease. Neuroreport 2004; 15:2521-5. [PMID: 15538187 DOI: 10.1097/00001756-200411150-00018] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Choroid plexus from neonatal pigs was encapsulated in alginate microcapsules and transplanted into the rat striatum. Three days later, the same animals received unilateral injections of quinolinic acid (225 nmol) into the ipsilateral striatum. Choroid plexus transplants ameliorated the weight loss and motor impairments resulting from QA. Histological analysis demonstrated that choroid plexus transplants reduced the volume of striatal damage and protected ChAT-, but not NADPH-diaphorase-positive neurons. These data are the first to demonstrate that transplanted choroid plexus cells can protect striatal neurons from excitotoxic damage and that this strategy may ultimately prove relevant for the treatment of Huntington's disease.
Collapse
Affiliation(s)
- Cesario V Borlongan
- Neurology Department, Institute of Mol Med and Genetics and School of Graduate Studies, Medical College, 1120 15th Street, Augusta, GA 30912-3200, USA.
| | | | | | | | | | | |
Collapse
|
29
|
Seo H, Sonntag KC, Isacson O. Generalized brain and skin proteasome inhibition in Huntington's disease. Ann Neurol 2004; 56:319-328. [PMID: 15349858 DOI: 10.1002/ana.20207] [Citation(s) in RCA: 135] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Mutated intracellular huntingtin is widely expressed in tissues of Huntington's disease (HD) patients. Intraneuronal nuclear protein aggregates of mutant huntingtin are present in HD brains, suggesting a dysfunction of the ubiquitin proteasome system (UPS). Because many cells and tissues can cope with the abnormal gene effects while others dysfunction and die, we determined gene-induced effects and considered the hypothesis that the gene causes multiple intracellular problems, but severe pathology is seen only in selected brain regions. In this study, we found inhibition of UPS function in both early (0-1, with no or little neuronal loss) and late (3-4, with more severe neuronal loss) stage HD patients' cerebellum, cortex, substantia nigra and caudate-putamen brain regions. Late HD stage increases in ubiquitin levels were unique to caudate-putamen. HD patients' skin fibroblasts also had UPS inhibition similar to brain despite increases in proteasome beta-subunit expression. Gene delivery and expression of proteasome activator PA28 increased UPS function in normal but not HD fibroblasts. These generalized UPS problems are associated with severe neuronal pathology only when coupled with decreases in brain-derived neurotrophic factor levels, mitochondrial complex II/III activity, and increases of ubiquitin levels particularly as seen in the caudate-putamen of HD patients.
Collapse
Affiliation(s)
- Hyemyung Seo
- Neuroregeneration Laboratories, Departments of Neurology and Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478, USA
| | | | | |
Collapse
|
30
|
Alberch J, Pérez-Navarro E, Canals JM. Neurotrophic factors in Huntington's disease. PROGRESS IN BRAIN RESEARCH 2004; 146:195-229. [PMID: 14699966 DOI: 10.1016/s0079-6123(03)46014-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Huntington's disease is a neurodegenerative disorder characterized by the selective loss of striatal neurons and, to a lesser extent, cortical neurons. The neurodegenerative process is caused by the mutation of huntingtin gene. Recent studies have established a link between mutant huntingtin, excitotoxicity and neurotrophic factors. Neurotrophic factors prevent cell death in degenerative processes but they can also enhance growth and function of neurons that are affected in Huntington's disease. The endogenous regulation of the expression of neurotrophic factors and their receptors in the striatum and its connections can be important to protect striatal cells and maintains basal ganglia connectivity. The administration of exogenous neurotrophic factors, in animal models of Huntington's disease, has been used to characterize the trophic requirements of striatal and cortical neurons. Neurotrophins, glial cell line-derived neurotrophic factor family members and ciliary neurotrophic factor have shown a potent neuroprotective effects on different neuronal populations of the striatum. Furthermore, they are also useful to maintain the integrity of the corticostriatal pathway. Thus, these neurotrophic factors may be suitable for the development of a neuroprotective therapy for neurodegenerative disorders of the basal ganglia.
Collapse
Affiliation(s)
- Jordi Alberch
- Department of Cell Biology and Pathology, Medical School, IDIBAPS, University of Barcelona, Casanova 143, E-08036 Barcelona, Spain.
| | | | | |
Collapse
|
31
|
Böttcher T, Mix E, Koczan D, Bauer P, Pahnke J, Peters S, Weinelt S, Knoblich R, Strauss U, Cattaneo E, Thiesen HJ, Rolfs A. Gene expression profiling of ciliary neurotrophic factor-overexpressing rat striatal progenitor cells (ST14A) indicates improved stress response during the early stage of differentiation. J Neurosci Res 2003; 73:42-53. [PMID: 12815707 DOI: 10.1002/jnr.10624] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Neuronal progenitor cells delivering neurotrophic factors are a promising therapeutic tool for treatment of neurodegenerative diseases. Although several promising results have come from studies in different animal models, detailed knowledge of the action of neurotrophic factors in the CNS is still lacking. A clonally derived, immortalized rat striatal cell line (ST14A) expressing ciliary neurotrophic factor (CNTF) offers a stable and controlled background with which to analyze CNTF actions on the transcriptional level in CNS progenitor cells. To identify early transcriptional changes induced by CNTF expression, we transfected the CNTF gene into ST14A cells, which differentiate at the nonpermissive temperature of 39 degrees C via suppression of the immortalizing SV40 large T antigen. This shows a CNTF-dependent hypoxic/ischemic stress response during the earliest stage of differentiation, with expression of specific transcripts and evidence of translational repression leading to decreased protein synthesis in the transfected cells. This process is mediated by the Ras/MAP kinase pathway and is accompanied by impaired proliferation and metabolism as well as signs of neuronal differentiation. The stress-like response in the early stage of differentiation improves the ability of the transfected cells to respond to and cope with a stressful environment in vivo. The present data indicate higher viability, longer life, and greater differentiation capacity of CNTF-ST14A cells if they are used for transplantation. We conclude that the stress-like response during the early stage of differentiation improves the ability of the CNTF-ST14A cells to respond and adapt to a stressful environment, which renders them useful candidate cells for in vivo trials of treatment for neurodegenerative diseases in animal models, e.g., of Huntington's disease.
Collapse
Affiliation(s)
- Tobias Böttcher
- Department of Neurology, University of Rostock, Rostock, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Gill SS, Patel NK, Hotton GR, O'Sullivan K, McCarter R, Bunnage M, Brooks DJ, Svendsen CN, Heywood P. Direct brain infusion of glial cell line-derived neurotrophic factor in Parkinson disease. Nat Med 2003; 9:589-95. [PMID: 12669033 DOI: 10.1038/nm850] [Citation(s) in RCA: 924] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2002] [Accepted: 03/07/2003] [Indexed: 12/11/2022]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is a potent neurotrophic factor with restorative effects in a wide variety of rodent and primate models of Parkinson disease, but penetration into brain tissue from either the blood or the cerebro-spinal fluid is limited. Here we delivered GDNF directly into the putamen of five Parkinson patients in a phase 1 safety trial. One catheter needed to be repositioned and there were changes in the magnetic resonance images that disappeared after lowering the concentration of GDNF. After one year, there were no serious clinical side effects, a 39% improvement in the off-medication motor sub-score of the Unified Parkinson's Disease Rating Scale (UPDRS) and a 61% improvement in the activities of daily living sub-score. Medication-induced dyskinesias were reduced by 64% and were not observed off medication during chronic GDNF delivery. Positron emission tomography (PET) scans of [(18)F]dopamine uptake showed a significant 28% increase in putamen dopamine storage after 18 months, suggesting a direct effect of GDNF on dopamine function. This study warrants careful examination of GDNF as a treatment for Parkinson disease.
Collapse
Affiliation(s)
- Steven S Gill
- Frenchay Hospital, Institute of Neurosciences, Bristol, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
The mainstay of current therapy for pediatric movement disorders is oral symptomatic medication, unless a reversible etiology can be found. However, this approach is apt to pale in comparison with innovative strategies on the clinical forefront. Classical pharmacotherapy is restricted by the blood-brain barrier, which prevents access to the brain of potentially therapeutic molecules. Recent developments in molecular biotechnology include antibody-mediated drug release, feedback-responsive delivery systems, carrier-mediated transport, microspheres composed of polymers and liposomes, permeabilizers, and selective delivery to localized sites and vectors. Neuroprotective strategies for delivering neurotrophic factors and antiapoptotic and antioxidant molecules in neurodegenerative disorders are currently under study in clinical trials. Stem cell transplantation has great potential for tissue engineering and also as a carrier for gene therapy, although its use raises complex societal issues. These approaches, together with a plethora of transgenic knockout animal models of neurodegenerative disorders, offer real promise for a previously untreatable group of movement disorders.
Collapse
Affiliation(s)
- Michael R Pranzatelli
- Department of Neurology, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
| |
Collapse
|
34
|
Eisel ULM. Cytokines in degenerative brain diseases: lessons from transgenic animals. Curr Top Microbiol Immunol 2002; 265:49-62. [PMID: 12014195 DOI: 10.1007/978-3-662-09525-6_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Affiliation(s)
- U L M Eisel
- University of Stuttgart, Institute of Cell Biology and Immunology, Allmandring 31, 70569 Stuttgart, Germany
| |
Collapse
|
35
|
Affiliation(s)
- N Déglon
- Division of Surgical Research and Gene Therapy Center, Lausanne University Medical School, Lausanne, Switzerland
| | | |
Collapse
|
36
|
Andsberg G, Kokaia Z, Klein RL, Muzyczka N, Lindvall O, Mandel RJ. Neuropathological and behavioral consequences of adeno-associated viral vector-mediated continuous intrastriatal neurotrophin delivery in a focal ischemia model in rats. Neurobiol Dis 2002; 9:187-204. [PMID: 11895371 DOI: 10.1006/nbdi.2001.0456] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) were continuously delivered to the striatum at biologically active levels via recombinant adeno-associated viral (rAAV) gene transfer 4-5 weeks prior to 30 min of middle cerebral artery occlusion (MCAO). The magnitude of the deficits in a battery of behavioral tests designed to assess striatal function was highly correlated to the extent of ischemic damage determined by unbiased stereological estimations of striatal neuron numbers. The delivery of neurotrophins lead to mild functional improvements in the ischemia-induced motor impairments assessed 3-5 weeks after the insult, in agreement with a small but significant increase of the survival of dorsolateral striatal neurons. Detailed phenotypic analysis demonstrated that the parvalbumin-containing interneurons were spared to a greater extent by the neurotrophin treatment as compared to the projection neurons, which agreed with the specificity for interneuron transduction by the rAAV vector. These data show the advantage of the never previously performed combination of precise quantification of the ischemia-induced neuropathology along with detailed behavioural analysis for assessing neuroprotection after stroke. We observe that intrastriatal delivery of NGF and BDNF using a viral vector system can mitigate, albeit only moderately, neuronal death following stroke, which leads to detectable functional sparing.
Collapse
Affiliation(s)
- Gunnar Andsberg
- Section of Restorative Neurology, Wallenberg Neuroscience Center, University Hospital, Lund, Sweden
| | | | | | | | | | | |
Collapse
|
37
|
Scorer CA. Preclinical and clinical challenges in the development of disease-modifying therapies for Alzheimer's disease. Drug Discov Today 2001; 6:1207-1219. [PMID: 11722873 DOI: 10.1016/s1359-6446(01)02042-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The neurodegenerative disease first described almost 100 years ago by Alois Alzheimer is predicted to be one of the major health problems of the 21st century. Alzheimer's disease (AD) is a progressive dementia characterized by global cognitive decline and is defined pathologically by amyloid plaques and neurofibrillary tangles. Major unmet medical need has encouraged pharmaceutical companies to invest in AD drug development. Promising novel approaches are under way, assisted by recent advances in animal models and an increased understanding of pathophysiology. However, demonstration of disease modification and identification of at-risk individuals are among the significant challenges facing those working in AD drug development.
Collapse
Affiliation(s)
- Carol A. Scorer
- Psychiatry Centre of Excellence for Drug Discovery, GlaxoSmithKline, New Frontiers Science Park, Third Avenue, CM19 5AW tel: +44 1279 875227 fax: +44 1279 622660, Essex, UK
| |
Collapse
|
38
|
Bensadoun JC, de Almeida LP, Dréano M, Aebischer P, Déglon N. Neuroprotective effect of interleukin-6 and IL6/IL6R chimera in the quinolinic acid rat model of Huntington's syndrome. Eur J Neurosci 2001; 14:1753-61. [PMID: 11860469 DOI: 10.1046/j.0953-816x.2001.01802.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Ciliary neurotrophic factor prevents behavioural deficits and striatal degeneration in rat and primate models of Huntington's disease. Interleukin-6, another member of the cytokine family, and the chimeric molecule (IL6/IL6R) in which interleukin-6 and its soluble receptor are fused, have been shown to exert trophic action on various neuronal populations in the central nervous system. Therefore, we investigated the neuroprotective effect of these two molecules in the quinolinic acid model of Huntington's disease. LacZ-, interleukin-6- and IL6/IL6R-expressing lentiviral vectors were stereotaxically injected into the striatum of Wistar rats. Three weeks later the animals were lesioned through the intrastriatal injection of 180 nmol of quinolinic acid. The extent of the striatal damage was significantly diminished in the rats that had been treated with interleukin-6 or IL6/IL6R. The neuroprotective effect was, however, more pronounced with the IL6/IL6R chimera than with interleukin-6 as indicated by the volume of the lesions (38.6 +/- 10% in the IL6/IL6R group, 63.3 +/- 3.6% in the IL-6 group and 84.3 +/-2.9% in the control group). Quantitative analysis of striatal interneurons further demonstrated that the IL6/IL6R chimera is more neuroprotective than IL-6 on ChAT- and NADPH-d-immunoreactive neurons. These results suggest that the IL6/IL6R chimera is a potential treatment for Huntington's disease.
Collapse
Affiliation(s)
- J C Bensadoun
- Division of Surgical Research and Gene Therapy Center, Lausanne Medical School, Pavillon 4, CHUV, 1011 Lausanne, Switzerland
| | | | | | | | | |
Collapse
|
39
|
Castro M, Hurtado-Lorenzo A, Umana P, Smith-Arica JR, Zermansky A, Abordo-Adesida E, Löwenstein PR. Regulatable and cell-type specific transgene expression in glial cells: prospects for gene therapy for neurological disorders. PROGRESS IN BRAIN RESEARCH 2001; 132:655-81. [PMID: 11545027 DOI: 10.1016/s0079-6123(01)32109-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Affiliation(s)
- M Castro
- Molecular Medicine and Gene Therapy Unit, Room 1.302, Stopford Building, School of Medicine, University of Manchester, Oxford Road, Manchester M13 9PT, UK.
| | | | | | | | | | | | | |
Collapse
|
40
|
Mizuta I, Ohta M, Ohta K, Nishimura M, Mizuta E, Kuno S. Riluzole stimulates nerve growth factor, brain-derived neurotrophic factor and glial cell line-derived neurotrophic factor synthesis in cultured mouse astrocytes. Neurosci Lett 2001; 310:117-20. [PMID: 11585581 DOI: 10.1016/s0304-3940(01)02098-5] [Citation(s) in RCA: 110] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Riluzole is an antiexcitotoxic agent used for the treatment of amyotrophic lateral sclerosis, and reported to have neuroprotective effects in animal models of Parkinson's disease, Huntington's disease and brain ischemia. We investigated the effects of riluzole on synthesis of nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF) in cultured mouse astrocytes. The protein and mRNA levels were measured by enzyme-linked immunosorbent assay and semiquantitative reverse transcription-polymerase chain reaction, respectively. Treatment with riluzole at 100 microg/ml (426 microM) for 24 h increased the contents of NGF, BDNF, and GDNF in the culture medium 109-fold, 2.0-fold and 3.1-fold over the control, respectively. The drug-induced relative mRNA levels of NGF, BDNF, and GDNF were 7.3-fold at 2 h, 2.1-fold at 4 h, and 1.9-fold at 2 h, respectively. These results indicate that riluzole stimulates synthesis of NGF, BDNF and GDNF in cultured astrocytes. Riluzole might exert neuroprotective effects, at least in part, via stimulation of neurotrophic factors.
Collapse
Affiliation(s)
- I Mizuta
- Clinical Research Center, Utano National Hospital, Narutaki, Ukyo-ku, Kyoto 616-8255, Japan
| | | | | | | | | | | |
Collapse
|
41
|
Gratacòs E, Pérez-Navarro E, Tolosa E, Arenas E, Alberch J. Neuroprotection of striatal neurons against kainate excitotoxicity by neurotrophins and GDNF family members. J Neurochem 2001; 78:1287-96. [PMID: 11579137 DOI: 10.1046/j.1471-4159.2001.00538.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Neurotrophic factors are regarded as potential therapeutic tools in neurodegenerative disorders. Here, we analysed the protective effects of brain-derived neurotrophic factor, neurotrophin-3, glial cell line-derived neurotrophic factor and neurturin against the excitotoxic damage induced by kainate in striatal neurons in vitro and in vivo. Our results show that the decrease in the number of cultured striatal calbindin-positive neurons induced by kainate was prevented by treatment with any of these factors. To characterize their protective effects in vivo, cell lines overexpressing brain-derived neurotrophic factor, neurotrophin-3, glial cell line-derived neurotrophic factor or neurturin were grafted into the striatum. We found that the numbers of striatal projection neurons (calbindin-positive) and striatal interneurons (parvalbumin- or choline acetyltransferase-positive) were differentially decreased after kainate lesion. These neurotrophic factors prevented the loss of striatal projection neurons and interneurons with differing efficiency: brain-derived neurotrophic factor was the most efficient, whereas neurturin was the least. Our findings show that brain-derived neurotrophic factor, neurotrophin-3, glial cell line-derived neurotrophic factor and neurturin have specific neuroprotective profiles in striatal neurons and indicate that they are specific modulators of the survival of distinct subsets of striatal neurons in pathophysiological conditions.
Collapse
Affiliation(s)
- E Gratacòs
- Departament de Biologia Cellular i Anatomia Patològica, Facultat de Medicina, Universitat de Barcelona, IDIBAPS, Barcelona, Spain
| | | | | | | | | |
Collapse
|
42
|
de Almeida LP, Zala D, Aebischer P, Déglon N. Neuroprotective effect of a CNTF-expressing lentiviral vector in the quinolinic acid rat model of Huntington's disease. Neurobiol Dis 2001; 8:433-46. [PMID: 11442352 DOI: 10.1006/nbdi.2001.0388] [Citation(s) in RCA: 119] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neurodegenerative diseases represent promising targets for gene therapy approaches provided effective transfer vectors. In the present study, we evaluated the effectiveness of LacZ-expressing lentiviral vectors with two different internal promoters, the mouse phosphoglycerate kinase 1 (PGK) and cytomegalovirus (CMV), to infect striatal cells. The intrastriatal injection of lenti-beta-Gal vectors lead to 207, 400 +/- 11,500 and 303,100 +/- 4,300 infected cells in adult rats, respectively. Importantly, the beta-galactosidase activity was higher in striatal extracts from PGK-LacZ-injected animals as compared to CMV-LacZ animals. The efficacy of the system was further examined with a potential therapeutic gene for the treatment of Huntington's disease, the human ciliary neurotrophic factor (CNTF). PGK-LacZ- or PGK-CNTF-expressing viruses were stereotaxically injected into the striatum of rats, 3 weeks later the animals were unilaterally lesioned with 180 nmol of quinolinic acid (QA). Control animals displayed 148 +/- 43 apomorphine-induced rotations ipsilateral to the lesion 5 days postlesion as compared to 26 +/- 22 turns/45 min in the CNTF-treated group. The extent of the striatal damage was significantly diminished in the CNTF-treated rats as indicated by the 52 +/- 9.7% decrease of the lesion volume and the sparing of DARPP-32, ChAT and NADPH-d neuronal populations. These results further establish that lentiviruses may represent an efficient gene delivery system for the screening of therapeutic molecules in Huntington's disease.
Collapse
Affiliation(s)
- L P de Almeida
- Division of Surgical Research and Gene Therapy Center, Lausanne Medical School, Switzerland
| | | | | | | |
Collapse
|
43
|
Affiliation(s)
- J E Carter
- Department of Neuroscience and Section of Old Age Psychiatry, Institute of Psychiatry, King's College, London, UK.
| | | |
Collapse
|
44
|
Freeman TB, Cicchetti F, Hauser RA, Deacon TW, Li XJ, Hersch SM, Nauert GM, Sanberg PR, Kordower JH, Saporta S, Isacson O. Transplanted fetal striatum in Huntington's disease: phenotypic development and lack of pathology. Proc Natl Acad Sci U S A 2000; 97:13877-82. [PMID: 11106399 PMCID: PMC17669 DOI: 10.1073/pnas.97.25.13877] [Citation(s) in RCA: 162] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Neural and stem cell transplantation is emerging as a potential treatment for neurodegenerative diseases. Transplantation of specific committed neuroblasts (fetal neurons) to the adult brain provides such scientific exploration of these new potential therapies. Huntington's disease (HD) is a fatal, incurable autosomal dominant (CAG repeat expansion of huntingtin protein) neurodegenerative disorder with primary neuronal pathology within the caudate-putamen (striatum). In a clinical trial of human fetal striatal tissue transplantation, one patient died 18 months after transplantation from cardiovascular disease, and postmortem histological analysis demonstrated surviving transplanted cells with typical morphology of the developing striatum. Selective markers of both striatal projection and interneurons such as dopamine and c-AMP-related phosphoprotein, calretinin, acetylcholinesterase, choline acetyltransferase, tyrosine hydroxylase, calbindin, enkephalin, and substance P showed positive transplant regions clearly innervated by host tyrosine hydroxylase fibers. There was no histological evidence of immune rejection including microglia and macrophages. Notably, neuronal protein aggregates of mutated huntingtin, which is typical HD neuropathology, were not found within the transplanted fetal tissue. Thus, although there is a genetically predetermined process causing neuronal death within the HD striatum, implanted fetal neural cells lacking the mutant HD gene may be able to replace damaged host neurons and reconstitute damaged neuronal connections. This study demonstrates that grafts derived from human fetal striatal tissue can survive, develop, and are unaffected by the disease process, at least for 18 months, after transplantation into a patient with HD.
Collapse
Affiliation(s)
- T B Freeman
- Department of Neurosurgery, Department of Pharmacology and Experimental Therapeutics, and The Neuroscience Program, University of South Florida, Tampa, FL 33606, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Gene therapy for neurological disorder is currently an experimental concept. The goals for clinical utilization are the relief of symptoms, slowing of disease progression, and correction of genetic abnormalities. Experimental studies are realizing these goals in the development of gene therapies in animal models. Discoveries of the molecular basis of neurological disease and advances in gene transfer systems have allowed focal and global delivery of therapeutic genes for a wide variety of CNS disorders. Limitations are still apparent, such as stability and regulation of transgene expression, and safety of both vector and expressed transgene. In addition, the brain adds several challenges not seen in peripheral gene therapy paradigms, such as post-mitotic cells, heterogeneity of cell types and circuits, and limited access. Moreover, it is likely that several modes of gene delivery will be necessary for successful gene therapies of the CNS. Collaborative efforts between clinicians and basic researchers will likely yield effective gene therapy in the CNS.
Collapse
Affiliation(s)
- L C Costantini
- Neuroregeneration Laboratory, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | | | | | | |
Collapse
|