51
|
Fagan SG, Campbell VA. The influence of cannabinoids on generic traits of neurodegeneration. Br J Pharmacol 2014; 171:1347-60. [PMID: 24172185 PMCID: PMC3954477 DOI: 10.1111/bph.12492] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 09/14/2013] [Accepted: 09/16/2013] [Indexed: 12/30/2022] Open
Abstract
UNLABELLED In an increasingly ageing population, the incidence of neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease and Huntington's disease are rising. While the aetiologies of these disorders are different, a number of common mechanisms that underlie their neurodegenerative components have been elucidated; namely neuroinflammation, excitotoxicity, mitochondrial dysfunction and reduced trophic support. Current therapies focus on treatment of the symptoms and attempt to delay the progression of these diseases but there is currently no cure. Modulation of the endogenous cannabinoid system is emerging as a potentially viable option in the treatment of neurodegeneration. Endocannabinoid signalling has been found to be altered in many neurodegenerative disorders. To this end, pharmacological manipulation of the endogenous cannabinoid system, as well as application of phytocannabinoids and synthetic cannabinoids have been investigated. Signalling from the CB1 and CB2 receptors are known to be involved in the regulation of Ca(2+) homeostasis, mitochondrial function, trophic support and inflammatory status, respectively, while other receptors gated by cannabinoids such as PPARγ, are gaining interest in their anti-inflammatory properties. Through multiple lines of evidence, this evolutionarily conserved neurosignalling system has shown neuroprotective capabilities and is therefore a potential target for neurodegenerative disorders. This review details the mechanisms of neurodegeneration and highlights the beneficial effects of cannabinoid treatment. LINKED ARTICLES This article is part of a themed section on Cannabinoids 2013. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue-6.
Collapse
Affiliation(s)
- S G Fagan
- Trinity College Institute of Neuroscience, Department of Physiology, School of Medicine, University of Dublin, Trinity CollegeDublin 2, Ireland
| | - V A Campbell
- Trinity College Institute of Neuroscience, Department of Physiology, School of Medicine, University of Dublin, Trinity CollegeDublin 2, Ireland
| |
Collapse
|
52
|
Abstract
The neurotrophin family is comprised of the structurally related secreted proteins nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), and neurotrophine-4 (NT-4). They bind and activate the tyrosine kinase receptors Trk A, B, and C in a ligand-specific manner and additionally bind a shared p75NTR receptor. The neurotrophins were originally defined by their ability to support the survival and maturation of embryonic neurons. However, they also control important physiological functions of the adult nervous system including learning and memory, sensation, and energy homeostasis. For example, NGF/trkA signaling is critical for normal and pathological sensation of pain. Likewise, the BDNF/trkB pathway controls feeding and metabolism, and its dysfunction leads to severe obesity. Antibodies can modulate neurotrophin signaling. Thus, NGF blocking agents can attenuate pain in several animal models, and a recombinant humanized NGF blocking antibody (Tanezumab) has shown promising results in human clinical trials for osteoarthritic pain. On the other hand trkB agonist antibodies can modulate food intake and body weight in rodents and nonhuman primates. The power of monoclonal antibodies to modulate neurotrophin signaling promises to turn the rich biological insights into novel human medicines.
Collapse
Affiliation(s)
- A Rosenthal
- Alector Inc., 953 Indiana St., San Francisco, CA, 94107, USA,
| | | |
Collapse
|
53
|
Abstract
Changes in the level and activity of brain-derived neurotrophic factor (BDNF) have been described in a number of neurodegenerative disorders since early 1990s. However, only in Huntington disease (HD) gain- and loss-of-function experiments have mechanistically linked these abnormalities with the genetic defect.In this chapter we will describe how huntingtin protein, whose mutation causes HD, is involved in the physiological control of BDNF synthesis and transport in neurons and how both processes are simultaneously disrupted in HD. We will describe the underlying molecular mechanisms and discuss pre-clinical data concerning the impact of the experimental manipulation of BDNF levels on HD progression. These studies have revealed that a major loss of BDNF protein in the brain of HD patients may contribute to the clinical manifestations of the disease. The experimental strategies under investigation to increase brain BDNF levels in animal models of HD will also be described, with a view to ultimately improving the clinical treatment of this condition.
Collapse
Affiliation(s)
- Chiara Zuccato
- Department of Biosciences and Centre for Stem cell Research, Università degli Studi di Milano, Via Viotti 3/5, 20133, Milan, Italy,
| | | |
Collapse
|
54
|
Yu SJ, Tseng KY, Shen H, Harvey BK, Airavaara M, Wang Y. Local administration of AAV-BDNF to subventricular zone induces functional recovery in stroke rats. PLoS One 2013; 8:e81750. [PMID: 24312581 PMCID: PMC3847037 DOI: 10.1371/journal.pone.0081750] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 10/16/2013] [Indexed: 12/15/2022] Open
Abstract
Migration of new neuroprogenitor cells (NPCs) from the subventricular zone (SVZ) plays an important role in neurorepair after injury. Previous studies have shown that brain derived neurotrophic factor (BDNF) enhances the migration of NPCs from SVZ explants in neonatal mice in vitro. The purpose of this study was to identify the role of BDNF in SVZ cells using AAV-BDNF in an animal model of stroke. BDNF protein production after AAV‐BDNF infection was verified in primary neuronal culture. AAV-BDNF or AAV-RFP was injected into the left SVZ region of adult rats at 14 days prior to right middle cerebral artery occlusion (MCAo). SVZ tissues were collected from the brain and placed in Metrigel cultures 1 day after MCAo. Treatment with AAV-BDNF significantly increased the migration of SVZ cells in the stroke brain in vitro. In another set of animals, AAV-GFP was co-injected with AAV-BDNF or AAV-RFP to label cells in left SVZ prior to right MCAo. Local administration of AAV-BDNF significantly enhanced recovery of locomotor function and migration of GFP-positive cells from the SVZ toward the lesioned hemisphere in stroke rats. Our data suggest that focal administration of AAV-BDNF to the SVZ increases behavioral recovery post stroke, possibly through the enhancement of migration of cells from SVZ in stroke animals. Regional manipulation of BDNF expression through AAV may be a novel approach for neurorepair in stroke brains.
Collapse
Affiliation(s)
- Seong-Jin Yu
- Center for Neuropsychiatric Research, National Health Research Institute, Zhunan, Taiwan
- Neural Protection and Regeneration Section, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland, United States of America
| | - Kuan-Yin Tseng
- Institute of Biotechnology, Viikki Biocenter, University of Helsinki, Helsinki, Finland
| | - Hui Shen
- Neural Protection and Regeneration Section, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland, United States of America
| | - Brandon K. Harvey
- Neural Protection and Regeneration Section, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland, United States of America
| | - Mikko Airavaara
- Institute of Biotechnology, Viikki Biocenter, University of Helsinki, Helsinki, Finland
| | - Yun Wang
- Center for Neuropsychiatric Research, National Health Research Institute, Zhunan, Taiwan
- Neural Protection and Regeneration Section, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
55
|
Hoban DB, Newland B, Moloney TC, Howard L, Pandit A, Dowd E. The reduction in immunogenicity of neurotrophin overexpressing stem cells after intra-striatal transplantation by encapsulation in an in situ gelling collagen hydrogel. Biomaterials 2013; 34:9420-9. [DOI: 10.1016/j.biomaterials.2013.08.073] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 08/22/2013] [Indexed: 11/17/2022]
|
56
|
Ellison SM, Trabalza A, Tisato V, Pazarentzos E, Lee S, Papadaki V, Goniotaki D, Morgan S, Mirzaei N, Mazarakis ND. Dose-dependent neuroprotection of VEGF₁₆₅ in Huntington's disease striatum. Mol Ther 2013; 21:1862-75. [PMID: 23799534 DOI: 10.1038/mt.2013.132] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 05/20/2013] [Indexed: 12/21/2022] Open
Abstract
Huntington's disease (HD) is a devastating neurodegenerative disorder caused by abnormal polyglutamine expansion in the huntingtin protein (Exp-Htt). Currently, there are no effective treatments for HD. We used bidirectional lentiviral transfer vectors to generate in vitro and in vivo models of HD and to test the therapeutic potential of vascular endothelial growth factor 165 (VEGF₁₆₅). Lentiviral-mediated expression of Exp-Htt caused cell death and aggregate formation in human neuroblastoma SH-SY5Y and rat primary striatal cultures. Lentiviral-mediated VEGF₁₆₅ expression was found to be neuroprotective in both of these models. Unilateral stereotaxic vector delivery of Exp-Htt vector in adult rat striatum led to progressive inclusion formation and striatal neuron loss at 10 weeks post-transduction. Coinjection of a lower dose VEGF₁₆₅ significantly attenuated DARPP-32(+) neuronal loss, enhanced NeuN staining and reduced Exp-Htt aggregation. A tenfold higher dose VEGF₁₆₅ led to overt neuronal toxicity marked by tissue damage, neovascularization, extensive astrogliosis, vascular leakage, chronic inflammation and distal neuronal loss. No overt behavioral phenotype was observed in these animals. Expression of VEGF₁₆₅ at this higher dose in the brain of wild-type rats led to early mortality with global neuronal loss. This report raises important safety concerns about unregulated VEGF₁₆₅ CNS applications.
Collapse
Affiliation(s)
- Stuart M Ellison
- Division of Brain Sciences, Department of Gene Therapy, Faculty of Medicine, Centre for Neuroinflammation & Neurodegeneration, Imperial College London, Hammersmith Hospital Campus, London, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
57
|
Gunawardena S. Nanoparticles in the Brain: A Potential Therapeutic System Targeted to an Early Defect Observed in Many Neurodegenerative Diseases. Pharm Res 2013; 30:2459-74. [DOI: 10.1007/s11095-013-1037-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2012] [Accepted: 03/25/2013] [Indexed: 12/14/2022]
|
58
|
MAP kinase phosphatase 1 (MKP-1/DUSP1) is neuroprotective in Huntington's disease via additive effects of JNK and p38 inhibition. J Neurosci 2013; 33:2313-25. [PMID: 23392662 DOI: 10.1523/jneurosci.4965-11.2013] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
We previously demonstrated that sodium butyrate is neuroprotective in Huntington's disease (HD) mice and that this therapeutic effect is associated with increased expression of mitogen-activated protein kinase/dual-specificity phosphatase 1 (MKP-1/DUSP1). Here we show that enhancing MKP-1 expression is sufficient to achieve neuroprotection in lentiviral models of HD. Wild-type MKP-1 overexpression inhibited apoptosis in primary striatal neurons exposed to an N-terminal fragment of polyglutamine-expanded huntingtin (Htt171-82Q), blocking caspase-3 activation and significantly reducing neuronal cell death. This neuroprotective effect of MKP-1 was demonstrated to be dependent on its enzymatic activity, being ablated by mutation of its phosphatase domain and being attributed to inhibition of specific MAP kinases (MAPKs). Overexpression of MKP-1 prevented the polyglutamine-expanded huntingtin-induced activation of c-Jun N-terminal kinases (JNKs) and p38 MAPKs, whereas extracellular signal-regulated kinase (ERK) 1/2 activation was not altered by either polyglutamine-expanded Htt or MKP-1. Moreover, mutants of MKP-1 that selectively prevented p38 or JNK binding confirmed the important dual contributions of p38 and JNK regulation to MKP-1-mediated neuroprotection. These results demonstrate additive effects of p38 and JNK MAPK inhibition by MKP-1 without consequence to ERK activation in this striatal neuron-based paradigm. MKP-1 also provided neuroprotection in vivo in a lentiviral model of HD neuropathology in rat striatum. Together, these data extend previous evidence that JNK- and p38-mediated pathways contribute to HD pathogenesis and, importantly, show that therapies simultaneously inhibiting both JNK and p38 signaling pathways may lead to improved neuroprotective outcomes.
Collapse
|
59
|
Géral C, Angelova A, Lesieur S. From molecular to nanotechnology strategies for delivery of neurotrophins: emphasis on brain-derived neurotrophic factor (BDNF). Pharmaceutics 2013; 5:127-67. [PMID: 24300402 PMCID: PMC3834942 DOI: 10.3390/pharmaceutics5010127] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 01/30/2013] [Accepted: 02/05/2013] [Indexed: 01/01/2023] Open
Abstract
Neurodegenerative diseases represent a major public health problem, but beneficial clinical treatment with neurotrophic factors has not been established yet. The therapeutic use of neurotrophins has been restrained by their instability and rapid degradation in biological medium. A variety of strategies has been proposed for the administration of these leading therapeutic candidates, which are essential for the development, survival and function of human neurons. In this review, we describe the existing approaches for delivery of brain-derived neurotrophic factor (BDNF), which is the most abundant neurotrophin in the mammalian central nervous system (CNS). Biomimetic peptides of BDNF have emerged as a promising therapy against neurodegenerative disorders. Polymer-based carriers have provided sustained neurotrophin delivery, whereas lipid-based particles have contributed also to potentiation of the BDNF action. Nanotechnology offers new possibilities for the design of vehicles for neuroprotection and neuroregeneration. Recent developments in nanoscale carriers for encapsulation and transport of BDNF are highlighted.
Collapse
Affiliation(s)
- Claire Géral
- CNRS UMR8612 Institut Galien Paris-Sud, 5 rue J.-B. Clément, F-92296 Châtenay-Malabry, France; E-Mails: (C.G.); (S.L.)
- Univ Paris Sud 11, 5 rue J.-B. Clément, F-92296 Châtenay-Malabry, France
| | - Angelina Angelova
- CNRS UMR8612 Institut Galien Paris-Sud, 5 rue J.-B. Clément, F-92296 Châtenay-Malabry, France; E-Mails: (C.G.); (S.L.)
- Univ Paris Sud 11, 5 rue J.-B. Clément, F-92296 Châtenay-Malabry, France
| | - Sylviane Lesieur
- CNRS UMR8612 Institut Galien Paris-Sud, 5 rue J.-B. Clément, F-92296 Châtenay-Malabry, France; E-Mails: (C.G.); (S.L.)
- Univ Paris Sud 11, 5 rue J.-B. Clément, F-92296 Châtenay-Malabry, France
| |
Collapse
|
60
|
Levine RM, Scott CM, Kokkoli E. Peptide functionalized nanoparticles for nonviral gene delivery. SOFT MATTER 2013; 9:985-1004. [DOI: 10.1039/c2sm26633d] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
|
61
|
Mrzljak L, Munoz-Sanjuan I. Therapeutic Strategies for Huntington's Disease. Curr Top Behav Neurosci 2013; 22:161-201. [PMID: 24277342 DOI: 10.1007/7854_2013_250] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Huntington's disease (HD) is a devastating autosomal dominant neurodegenerative disease, caused by expansion of the CAG repeat in the huntingtin (HTT) gene and characterized pathologically by the loss of pyramidal neurons in several cortical areas, of striatal medium spiny neurons, and of hypothalamic neurons. Clinically, a distinguishing feature of the disease is uncontrolled involuntary movements (chorea, dyskensias) accompanied by progressive cognitive, motor, and psychiatric impairment. This review focuses on the current state of therapeutic development for the treatment of HD, including the preclinical and clinical development of small molecules and molecular therapies.
Collapse
|
62
|
Chiu CT, Wang Z, Hunsberger JG, Chuang DM. Therapeutic potential of mood stabilizers lithium and valproic acid: beyond bipolar disorder. Pharmacol Rev 2013; 65:105-42. [PMID: 23300133 PMCID: PMC3565922 DOI: 10.1124/pr.111.005512] [Citation(s) in RCA: 290] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The mood stabilizers lithium and valproic acid (VPA) are traditionally used to treat bipolar disorder (BD), a severe mental illness arising from complex interactions between genes and environment that drive deficits in cellular plasticity and resiliency. The therapeutic potential of these drugs in other central nervous system diseases is also gaining support. This article reviews the various mechanisms of action of lithium and VPA gleaned from cellular and animal models of neurologic, neurodegenerative, and neuropsychiatric disorders. Clinical evidence is included when available to provide a comprehensive perspective of the field and to acknowledge some of the limitations of these treatments. First, the review describes how action at these drugs' primary targets--glycogen synthase kinase-3 for lithium and histone deacetylases for VPA--induces the transcription and expression of neurotrophic, angiogenic, and neuroprotective proteins. Cell survival signaling cascades, oxidative stress pathways, and protein quality control mechanisms may further underlie lithium and VPA's beneficial actions. The ability of cotreatment to augment neuroprotection and enhance stem cell homing and migration is also discussed, as are microRNAs as new therapeutic targets. Finally, preclinical findings have shown that the neuroprotective benefits of these agents facilitate anti-inflammation, angiogenesis, neurogenesis, blood-brain barrier integrity, and disease-specific neuroprotection. These mechanisms can be compared with dysregulated disease mechanisms to suggest core cellular and molecular disturbances identifiable by specific risk biomarkers. Future clinical endeavors are warranted to determine the therapeutic potential of lithium and VPA across the spectrum of central nervous system diseases, with particular emphasis on a personalized medicine approach toward treating these disorders.
Collapse
Affiliation(s)
- Chi-Tso Chiu
- Molecular Neurobiology Section, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | |
Collapse
|
63
|
Developing nanotherapies for neurodegenerative diseases: ORMOSIL and its potential in axonal transport. Ther Deliv 2012; 3:1189-98. [PMID: 23116011 DOI: 10.4155/tde.12.101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In neurons, essential components packaged into vesicles are transported down microtubules to the ends of axons (synapses) where they are utilized. Components are also transported from the synapse to the cell body. This transport pathway is crucial for normal development, cell survival and plasticity. Recent work has established that defects in transport can contribute to the initiation of neurodegenerative disease, culminating in cell death and degeneration. Thus, delivering therapeutic treatments to an early defect is critical since many current strategies target pathology that occurs at later stages in the disease. Current treatments also affect the entire organism, causing side-effects that are often more deleterious than the disease. This article discusses how engineered synthetic structures can be used to directly target axonal transport--a pathway that is affected during the early stages of disease. Studies in this area will require the exchange of fundamental knowledge between biologists, chemists and engineers to effectively manufacture novel biomaterials for medical use.
Collapse
|
64
|
Thévenot E, Jordão JF, O'Reilly MA, Markham K, Weng YQ, Foust KD, Kaspar BK, Hynynen K, Aubert I. Targeted delivery of self-complementary adeno-associated virus serotype 9 to the brain, using magnetic resonance imaging-guided focused ultrasound. Hum Gene Ther 2012; 23:1144-55. [PMID: 22838844 PMCID: PMC3498907 DOI: 10.1089/hum.2012.013] [Citation(s) in RCA: 150] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 07/21/2012] [Indexed: 01/27/2023] Open
Abstract
Noninvasive drug delivery to the brain remains a major challenge for the treatment of neurological disorders. Transcranial focused ultrasound combined with lipid-coated gas microspheres injected into the bloodstream has been shown to increase the permeability of the blood-brain barrier locally and transiently. Coupled with magnetic resonance imaging, ultrasound can be guided to allow therapeutics administered in the blood to reach brain regions of interest. Using this approach, we perform gene transfer from the blood to specific regions of the mouse brain. Focused ultrasound was targeted to the right hemisphere, at multiple foci, or restricted to one focal point of the hippocampus or the striatum. Doses from 5 × 10(8) to 1.25 × 10(10) vector genomes per gram (VG/g) of self-complementary adeno-associated virus serotype 9 carrying the green fluorescent protein were injected into the tail vein. A dose of 2.5 × 10(9) VG/g was optimal to express the transgene, 12 days later, in neurons, astrocytes, and oligodendrocytes in brain regions targeted with ultrasound, while minimizing the infection of peripheral organs. In the hippocampus and striatum, predominantly neurons and astrocytes were infected, respectively. Transcranial focused ultrasound applications could fulfill a long-term goal of gene therapy: delivering vectors to diseased brain areas directly from the circulation, in a noninvasive manner.
Collapse
Affiliation(s)
- Emmanuel Thévenot
- Brain Sciences Sunnybrook Research Institute, Toronto, ON, M4N 3M5 Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8 Canada
| | - Jessica F. Jordão
- Brain Sciences Sunnybrook Research Institute, Toronto, ON, M4N 3M5 Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8 Canada
| | - Meaghan A. O'Reilly
- Imaging Research, Sunnybrook Research Institute, Toronto, ON, M4N 3M5 Canada
| | - Kelly Markham
- Brain Sciences Sunnybrook Research Institute, Toronto, ON, M4N 3M5 Canada
| | - Ying-Qi Weng
- Brain Sciences Sunnybrook Research Institute, Toronto, ON, M4N 3M5 Canada
| | - Kevin D. Foust
- Department of Neuroscience, Ohio State University, Columbus, OH 43205
| | - Brian K. Kaspar
- Center for Gene Therapy, Research Institute at Nationwide Children's Hospital, Columbus, OH 43205
| | - Kullervo Hynynen
- Imaging Research, Sunnybrook Research Institute, Toronto, ON, M4N 3M5 Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 2M9 Canada
| | - Isabelle Aubert
- Brain Sciences Sunnybrook Research Institute, Toronto, ON, M4N 3M5 Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8 Canada
| |
Collapse
|
65
|
Intrastriatal transplantation of neurotrophic factor-secreting human mesenchymal stem cells improves motor function and extends survival in R6/2 transgenic mouse model for Huntington's disease. PLOS CURRENTS 2012; 4:e4f7f6dc013d4e. [PMID: 22953237 PMCID: PMC3426086 DOI: 10.1371/4f7f6dc013d4e] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Stem cell-based treatment for Huntington's disease (HD) is an expanding field of research. Although various stem cells have been shown to be beneficial in vivo, no long standing clinical effect has been demonstrated. To address this issue, we are developing a stem cell-based therapy designed to improve the microenvironment of the diseased tissue via delivery of neurotrophic factors (NTFs). Previously, we established that bone marrow derived human mesenchymal stem cells (MSCs) can be differentiated using medium based cues into NTF-secreting cells (NTF+ cells) that express astrocytic markers. NTF+ cells were shown to alleviate neurodegeneration symptoms in several disease models in vitro and in vivo, including the model for excitotoxicity. In the present study, we explored if the timing of intrastriatal transplantation of hNTF+ cells into the R6/2 transgenic mouse model for HD influences motor function and survival. One hundred thousand cells were transplanted bilaterally into the striatum of immune-suppressed mice at 4.5, 5.5 and 6.5 weeks of age. Contrary to our expectations, early transplantation of NTF+ cells did not improve motor function or overall survival. However, late (6.5 weeks) transplantation resulted in a temporary improvement in motor function and an extension of life span relative to that observed for PBS treated mice. We conclude that late transplantation of NTF+ cells induces a beneficial effect in this transgenic model for HD. Since no transplanted NTF+ cells could be detected in vivo, we suspect that the temporary nature of the beneficial effect is due to poor survival of transplanted cells. In general, we submit that NTF+ cells should be further evaluated for the therapy of HD.
Collapse
|
66
|
Sadan O, Shemesh N, Barzilay R, Dadon-Nahum M, Blumenfeld-Katzir T, Assaf Y, Yeshurun M, Djaldetti R, Cohen Y, Melamed E, Offen D. Mesenchymal stem cells induced to secrete neurotrophic factors attenuate quinolinic acid toxicity: A potential therapy for Huntington's disease. Exp Neurol 2012; 234:417-27. [DOI: 10.1016/j.expneurol.2011.12.045] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 12/23/2011] [Accepted: 12/31/2011] [Indexed: 10/14/2022]
|
67
|
Nucleic Acid-Based Therapy Approaches for Huntington's Disease. Neurol Res Int 2012; 2012:358370. [PMID: 22288011 PMCID: PMC3263636 DOI: 10.1155/2012/358370] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 09/28/2011] [Accepted: 10/05/2011] [Indexed: 01/30/2023] Open
Abstract
Huntington's disease (HD) is caused by a dominant mutation that results in an unstable expansion of a CAG repeat in the huntingtin gene leading to a toxic gain of function in huntingtin protein which causes massive neurodegeneration mainly in the striatum and clinical symptoms associated with the disease. Since the mutation has multiple effects in the cell and the precise mechanism of the disease remains to be elucidated, gene therapy approaches have been developed that intervene in different aspects of the condition. These approaches include increasing expression of growth factors, decreasing levels of mutant huntingtin, and restoring cell metabolism and transcriptional balance. The aim of this paper is to outline the nucleic acid-based therapeutic strategies that have been tested to date.
Collapse
|
68
|
Barandeh F, Nguyen PL, Kumar R, Iacobucci GJ, Kuznicki ML, Kosterman A, Bergey EJ, Prasad PN, Gunawardena S. Organically modified silica nanoparticles are biocompatible and can be targeted to neurons in vivo. PLoS One 2012; 7:e29424. [PMID: 22238611 PMCID: PMC3250438 DOI: 10.1371/journal.pone.0029424] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Accepted: 11/28/2011] [Indexed: 11/29/2022] Open
Abstract
The application of nanotechnology in biological research is beginning to have a major impact leading to the development of new types of tools for human health. One focus of nanobiotechnology is the development of nanoparticle-based formulations for use in drug or gene delivery systems. However most of the nano probes currently in use have varying levels of toxicity in cells or whole organisms and therefore are not suitable for in vivo application or long-term use. Here we test the potential of a novel silica based nanoparticle (organically modified silica, ORMOSIL) in living neurons within a whole organism. We show that feeding ORMOSIL nanoparticles to Drosophila has no effect on viability. ORMOSIL nanoparticles penetrate into living brains, neuronal cell bodies and axonal projections. In the neuronal cell body, nanoparticles are present in the cytoplasm, but not in the nucleus. Strikingly, incorporation of ORMOSIL nanoparticles into the brain did not induce aberrant neuronal death or interfered with normal neuronal processes. Our results in Drosophila indicate that these novel silica based nanoparticles are biocompatible and not toxic to whole organisms, and has potential for the development of long-term applications.
Collapse
Affiliation(s)
- Farda Barandeh
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Phuong-Lan Nguyen
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Rajiv Kumar
- Institute of Lasers, Photonics and Biophotonics, The State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Gary J. Iacobucci
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Michelle L. Kuznicki
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Andrew Kosterman
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Earl J. Bergey
- Institute of Lasers, Photonics and Biophotonics, The State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Paras N. Prasad
- Institute of Lasers, Photonics and Biophotonics, The State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Shermali Gunawardena
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, New York, United States of America
- Institute of Lasers, Photonics and Biophotonics, The State University of New York at Buffalo, Buffalo, New York, United States of America
- * E-mail:
| |
Collapse
|
69
|
Arregui L, Benítez JA, Razgado LF, Vergara P, Segovia J. Adenoviral astrocyte-specific expression of BDNF in the striata of mice transgenic for Huntington's disease delays the onset of the motor phenotype. Cell Mol Neurobiol 2011; 31:1229-43. [PMID: 21681558 DOI: 10.1007/s10571-011-9725-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Accepted: 06/03/2011] [Indexed: 10/18/2022]
Abstract
Huntington's disease (HD) is a neurodegenerative disorder characterized by motor, cognitive, and psychiatric symptoms. The most characteristic structural feature of this disease is neurodegeneration accompanied by gliosis in the striatum. BDNF has been proposed to protect striatal neurons from degeneration, because it is an important survival factor for these neurons from development to adulthood. Considering the extensive gliosis and the survival effects of BDNF, we constructed an adenovirus to express a BDNF cDNA in astrocyte cells using a promoter of the glial fibrillary acidic protein gene. Cells stably transfected in vitro with a BDNF cDNA driven by this promoter expressed BDNF and responded to external stimuli increasing BDNF production. When the vector was applied into the striata of mice transgenic for HD, long-term expression of the transgene was observed, associated with a delay of onset of the motor phenotype of the R6/2 HD transgenic mice. The present data indicate that the striatal expression of BDNF is a potential adjuvant for the treatment of HD.
Collapse
Affiliation(s)
- Leticia Arregui
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Av. Instituto Politécnico Nacional # 2508, 07360 Mexico, DF, Mexico
| | | | | | | | | |
Collapse
|
70
|
Combined treatment with the mood stabilizers lithium and valproate produces multiple beneficial effects in transgenic mouse models of Huntington's disease. Neuropsychopharmacology 2011; 36:2406-21. [PMID: 21796107 PMCID: PMC3194069 DOI: 10.1038/npp.2011.128] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Emerging evidence suggests that the mood stabilizers lithium and valproate (VPA) have broad neuroprotective and neurotrophic properties, and that these occur via inhibition of glycogen synthase kinase 3 (GSK-3) and histone deacetylases (HDACs), respectively. Huntington's disease (HD) is an inherited neurodegenerative disorder characterized by impaired movement, cognitive and psychiatric disturbances, and premature death. We treated N171-82Q and YAC128 mice, two mouse models of HD varying in genetic backgrounds and pathological progressions, with a diet containing therapeutic doses of lithium, VPA, or both. Untreated, these transgenic mice displayed a decrease in levels of GSK-3β serine 9 phosphorylation and histone H3 acetylation in the striatum and cerebral cortex around the onset of behavioral deficits, indicating a hyperactivity of GSK-3β and HDACs. Using multiple well-validated behavioral tests, we found that co-treatment with lithium and VPA more effectively alleviated spontaneous locomotor deficits and depressive-like behaviors in both models of HD mice. Furthermore, compared with monotherapy with either drug alone, co-treatment more successfully improved motor skill learning and coordination in N171-82Q mice, and suppressed anxiety-like behaviors in YAC128 mice. This combined treatment consistently inhibited GSK-3β and HDACs, and caused a sustained elevation in striatal as well as cortical brain-derived neurotrophic factor and heat shock protein 70. Importantly, co-treatment markedly prolonged median survival of N171-82Q mice from 31.6 to 41.6 weeks. Given that there is presently no proven treatment for HD, our results suggest that combined treatment with lithium and VPA, two mood stabilizers with a long history of safe use in humans, may have important therapeutic potential for HD patients.
Collapse
|
71
|
Abstract
Huntington's disease (HD) is an inherited, relentlessly progressive neurodegenerative disease with an invariably fatal outcome. HD is inherited in an autosomal dominant fashion, and is characterized pathologically by the loss of cortical and striatal neurons, and clinically by involuntary choreiform movements accompanied by progressive cognitive impairment and emotional lability. The disorder is caused by an expanded cystosine adenine guanine (CAG) tri-nucleotide repeat encoding polyglutamine (polyQ) in the first exon of the Huntingtin gene. There is a correlation between the number of CAG repeats and disease onset, such that in patients with CAG repeat lengths of 36 to 60, disease symptoms typically manifest after 35 years of age, whereas CAG repeat lengths >60 yield the more severe juvenile form of the disease. Even though mutant huntingtin is expressed throughout the brain, it is characterized by the selective degeneration of medium spiny neurons of the caudate and putamen, which heralds more widespread neuronal degeneration with disease progression. The mechanisms of cell dysfunction and death in HD have been the subjects of a number of studies, which have led to therapeutic strategies largely based on the amelioration of mutant huntingtin-related metabolic impairment and cellular toxicity. Each of these approaches has aimed to delay or stop the preferential degeneration of medium spiny neurons early in the disease course. Yet, in later stages of the disease, after cell death has become prominent, cell replacement therapy (whether by direct cell transplantation or by the mobilization of endogenous progenitors) may comprise a stronger potential avenue for therapy. In this review, we will consider recent progress in the transplantation of fetal striatal cells to the HD brain, as well as emerging alternative sources for human striatal progenitor cells. We will then consider the potential application of gene therapy toward the induction of striatal neurogenesis and neuronal recruitment, with an eye toward its potential therapeutic use in HD.
Collapse
Affiliation(s)
- Abdellatif Benraiss
- Department of Neurology, Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY USA
| | - Steven A. Goldman
- Department of Neurology, Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY USA
- Department of Neurosurgery, University of Rochester Medical Center, Rochester, NY 14642 USA
| |
Collapse
|
72
|
Abstract
Huntingtin, the protein that when mutated causes Huntington disease (HD), has many known interactors and participates in diverse cellular functions. Mutant Htt (mHtt) engages in a variety of aberrant interactions that lead to pathological gain of toxic functions as well as loss of normal functions. The broad symptomatology of HD, including diminished voluntary motor control, cognitive decline, and psychiatric disturbances, reflects the multifaceted neuropathology. Although currently available therapies for HD focus on symptom management, the autosomal dominant cause and the adult onset make this disease an ideal candidate for genetic intervention. A variety of gene therapy approaches have been tested in mouse models of HD, ranging from those aimed at ameliorating downstream pathology or replacing lost neuronal populations to more upstream strategies to reduce mHtt levels. Here the authors review the results of these preclinical trials.
Collapse
Affiliation(s)
- Amber L Southwell
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada.
| | | |
Collapse
|
73
|
Battaglia G, Cannella M, Riozzi B, Orobello S, Maat-Schieman ML, Aronica E, Busceti CL, Ciarmiello A, Alberti S, Amico E, Sassone J, Sipione S, Bruno V, Frati L, Nicoletti F, Squitieri F. Early defect of transforming growth factor β1 formation in Huntington's disease. J Cell Mol Med 2011; 15:555-71. [PMID: 20082658 PMCID: PMC3922377 DOI: 10.1111/j.1582-4934.2010.01011.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
A defective expression or activity of neurotrophic factors, such as brain- and glial-derived neurotrophic factors, contributes to neuronal damage in Huntington’s disease (HD). Here, we focused on transforming growth factor-β (TGF-β1), a pleiotropic cytokine with an established role in mechanisms of neuroprotection. Asymptomatic HD patients showed a reduction in TGF-β1 levels in the peripheral blood, which was related to trinucleotide mutation length and glucose hypometabolism in the caudate nucleus. Immunohistochemical analysis in post-mortem brain tissues showed that TGF-β1 was reduced in cortical neurons of asymptomatic and symptomatic HD patients. Both YAC128 and R6/2 HD mutant mice showed a reduced expression of TGF-β1 in the cerebral cortex, localized in neurons, but not in astrocytes. We examined the pharmacological regulation of TGF-β1 formation in asymptomatic R6/2 mice, where blood TGF-β1 levels were also reduced. In these R6/2 mice, both the mGlu2/3 metabotropic glutamate receptor agonist, LY379268, and riluzole failed to increase TGF-β1 formation in the cerebral cortex and corpus striatum, suggesting that a defect in the regulation of TGF-β1 production is associated with HD. Accordingly, reduced TGF-β1 mRNA and protein levels were found in cultured astrocytes transfected with mutated exon 1 of the human huntingtin gene, and in striatal knock-in cell lines expressing full-length huntingtin with an expanded glutamine repeat. Taken together, our data suggest that serum TGF-β1 levels are potential biomarkers of HD development during the asymptomatic phase of the disease, and raise the possibility that strategies aimed at rescuing TGF-β1 levels in the brain may influence the progression of HD.
Collapse
|
74
|
Huang NK, Lin JH, Lin JT, Lin CI, Liu EM, Lin CJ, Chen WP, Shen YC, Chen HM, Chen JB, Lai HL, Yang CW, Chiang MC, Wu YS, Chang C, Chen JF, Fang JM, Lin YL, Chern Y. A new drug design targeting the adenosinergic system for Huntington's disease. PLoS One 2011; 6:e20934. [PMID: 21713039 PMCID: PMC3119665 DOI: 10.1371/journal.pone.0020934] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Accepted: 05/13/2011] [Indexed: 02/01/2023] Open
Abstract
Background Huntington's disease (HD) is a neurodegenerative disease caused by a CAG trinucleotide expansion in the Huntingtin (Htt) gene. The expanded CAG repeats are translated into polyglutamine (polyQ), causing aberrant functions as well as aggregate formation of mutant Htt. Effective treatments for HD are yet to be developed. Methodology/Principal Findings Here, we report a novel dual-function compound, N6-(4-hydroxybenzyl)adenine riboside (designated T1-11) which activates the A2AR and a major adenosine transporter (ENT1). T1-11 was originally isolated from a Chinese medicinal herb. Molecular modeling analyses showed that T1-11 binds to the adenosine pockets of the A2AR and ENT1. Introduction of T1-11 into the striatum significantly enhanced the level of striatal adenosine as determined by a microdialysis technique, demonstrating that T1-11 inhibited adenosine uptake in vivo. A single intraperitoneal injection of T1-11 in wildtype mice, but not in A2AR knockout mice, increased cAMP level in the brain. Thus, T1-11 enters the brain and elevates cAMP via activation of the A2AR in vivo. Most importantly, addition of T1-11 (0.05 mg/ml) to the drinking water of a transgenic mouse model of HD (R6/2) ameliorated the progressive deterioration in motor coordination, reduced the formation of striatal Htt aggregates, elevated proteasome activity, and increased the level of an important neurotrophic factor (brain derived neurotrophic factor) in the brain. These results demonstrate the therapeutic potential of T1-11 for treating HD. Conclusions/Significance The dual functions of T1-11 enable T1-11 to effectively activate the adenosinergic system and subsequently delay the progression of HD. This is a novel therapeutic strategy for HD. Similar dual-function drugs aimed at a particular neurotransmitter system as proposed herein may be applicable to other neurotransmitter systems (e.g., the dopamine receptor/dopamine transporter and the serotonin receptor/serotonin transporter) and may facilitate the development of new drugs for other neurodegenerative diseases.
Collapse
Affiliation(s)
- Nai-Kuei Huang
- National Research Institute of Chinese Medicine, Taipei, Taiwan
| | - Jung-Hsin Lin
- Division of Mechanics, Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| | - Jiun-Tsai Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chia-I Lin
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Eric Minwei Liu
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| | - Chun-Jung Lin
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| | - Wan-Ping Chen
- National Research Institute of Chinese Medicine, Taipei, Taiwan
| | - Yuh-Chiang Shen
- National Research Institute of Chinese Medicine, Taipei, Taiwan
| | - Hui-Mei Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jhih-Bin Chen
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Hsing-Lin Lai
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chieh-Wen Yang
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Ming-Chang Chiang
- Graduate Institute of Biotechnology, Chinese Culture University, Taipei, Taiwan
| | - Yu-Shuo Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chen Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jiang-Fan Chen
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Jim-Min Fang
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
- The Genomics Research Center, Academia Sinica, Taipei, Taiwan
- * E-mail: (YC); (YLL); (JMF)
| | - Yun-Lian Lin
- National Research Institute of Chinese Medicine, Taipei, Taiwan
- * E-mail: (YC); (YLL); (JMF)
| | - Yijuang Chern
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- * E-mail: (YC); (YLL); (JMF)
| |
Collapse
|
75
|
Abstract
It has been more than 17 years since the causative mutation for Huntington's disease was discovered as the expansion of the triplet repeat in the N-terminal portion of the Huntingtin (HTT) gene. In the intervening time, researchers have discovered a great deal about Huntingtin's involvement in a number of cellular processes. However, the role of Huntingtin in the key pathogenic mechanism leading to neurodegeneration in the disease process has yet to be discovered. Here, we review the body of knowledge that has been uncovered since gene discovery and include discussions of the HTT gene, CAG triplet repeat expansion, HTT expression, protein features, posttranslational modifications, and many of its known protein functions and interactions. We also highlight potential pathogenic mechanisms that have come to light in recent years.
Collapse
Affiliation(s)
- Karen N McFarland
- Department of Neurology, University of Florida, Gainesville, FL 32610-0236, USA.
| | | |
Collapse
|
76
|
Ribeiro FM, Pires RGW, Ferguson SSG. Huntington's disease and Group I metabotropic glutamate receptors. Mol Neurobiol 2010; 43:1-11. [PMID: 21153060 DOI: 10.1007/s12035-010-8153-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Accepted: 11/15/2010] [Indexed: 12/21/2022]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder characterized by involuntary body movement, cognitive impairment and psychiatric disturbance. A polyglutamine expansion in the amino-terminal region of the huntingtin (htt) protein is the genetic cause of HD. Htt protein interacts with a wide variety of proteins, and htt mutation causes cell signaling alterations in various neurotransmitter systems, including dopaminergic, glutamatergic, and cannabinoid systems, as well as trophic factor systems. This review will overview recent findings concerning htt-promoted alterations in cell signaling that involve different neurotransmitters and trophic factor systems, especially involving mGluR1/5, as glutamate plays a crucial role in neuronal cell death. The neuronal cell death that takes place in the striatum and cortex of HD patients is the most important factor underlying HD progression. Metabotropic glutamate receptors (mGluR1 and mGluR5) have a very controversial role in neuronal cell death and it is not clear whether mGluR1/5 activation either protects or exacerbates neuronal death. Thus, understanding how mutant htt protein affects glutamatergic receptor signaling will be essential to further establish a role for glutamate receptors in HD and develop therapeutic strategies to treat HD.
Collapse
Affiliation(s)
- Fabiola M Ribeiro
- Departamento de Bioquimica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | |
Collapse
|
77
|
Saylor AJ, McGinty JF. An intrastriatal brain-derived neurotrophic factor infusion restores striatal gene expression in Bdnf heterozygous mice. Brain Struct Funct 2010; 215:97-104. [PMID: 20938680 DOI: 10.1007/s00429-010-0282-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Accepted: 09/24/2010] [Indexed: 11/24/2022]
Abstract
Reduction in the amount of brain-derived neurotrophic factor (BDNF) in corticostriatal afferents is thought to contribute to the vulnerability of medium spiny striatal neurons in Huntington's disease. In young Bdnf heterozygous ((+/-)) mice, striatal medium spiny neurons (MSNs) express less preprodynorphin (PPD), preproenkephalin (PPE), and D(3) receptor mRNA than wildtype mice. Further, in aged Bdnf (+/-) mice, opioid, trkB receptor, and glutamic acid decarboxylase gene expression, and the number of dendritic spines on MSNs are more affected than in wildtype or younger Bdnf (+/-) mice. In this study, the possibility that intrastriatal infusions of BDNF would elevate gene expression in the striatum of Bdnf (+/-) mice was investigated. Wildtype and Bdnf (+/-) mice received a single, bilateral microinjection of BDNF or PBS into the dorsal striatum. Mice were killed 24 h later and semi-quantitative in situ hybridization histochemical analysis confirmed that PPD, PPE, and D(3) receptor mRNA was less in the caudate-putamen (CPu) and nucleus accumbens (NAc) core of Bdnf (+/-) mice than in wildtype mice. A BDNF infusion increased PPD mRNA in the CPu and NAc core of wildtype mice and restored PPD mRNA levels in the NAc core of Bdnf (+/-) mice. BDNF also restored the gene expression of PPE in the CPu of Bdnf (+/-) mice to wildtype levels; however, PPE mRNA in the NAc did not differ among groups. Furthermore, BDNF increased D(3) receptor mRNA in the NAc core of wildtype and Bdnf (+/-) mice. These results demonstrate that exogenous BDNF restores striatal opioid and D(3)R gene expression in mice with genetically reduced levels of endogenous BDNF.
Collapse
Affiliation(s)
- Alicia J Saylor
- Department of Neurosciences, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| | | |
Collapse
|
78
|
Zuccato C, Valenza M, Cattaneo E. Molecular Mechanisms and Potential Therapeutical Targets in Huntington's Disease. Physiol Rev 2010; 90:905-81. [DOI: 10.1152/physrev.00041.2009] [Citation(s) in RCA: 626] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder caused by a CAG repeat expansion in the gene encoding for huntingtin protein. A lot has been learned about this disease since its first description in 1872 and the identification of its causative gene and mutation in 1993. We now know that the disease is characterized by several molecular and cellular abnormalities whose precise timing and relative roles in pathogenesis have yet to be understood. HD is triggered by the mutant protein, and both gain-of-function (of the mutant protein) and loss-of-function (of the normal protein) mechanisms are involved. Here we review the data that describe the emergence of the ancient huntingtin gene and of the polyglutamine trait during the last 800 million years of evolution. We focus on the known functions of wild-type huntingtin that are fundamental for the survival and functioning of the brain neurons that predominantly degenerate in HD. We summarize data indicating how the loss of these beneficial activities reduces the ability of these neurons to survive. We also review the different mechanisms by which the mutation in huntingtin causes toxicity. This may arise both from cell-autonomous processes and dysfunction of neuronal circuitries. We then focus on novel therapeutical targets and pathways and on the attractive option to counteract HD at its primary source, i.e., by blocking the production of the mutant protein. Strategies and technologies used to screen for candidate HD biomarkers and their potential application are presented. Furthermore, we discuss the opportunities offered by intracerebral cell transplantation and the likely need for these multiple routes into therapies to converge at some point as, ideally, one would wish to stop the disease process and, at the same time, possibly replace the damaged neurons.
Collapse
Affiliation(s)
- Chiara Zuccato
- Department of Pharmacological Sciences and Centre for Stem Cell Research, Università degli Studi di Milano, Milan, Italy
| | - Marta Valenza
- Department of Pharmacological Sciences and Centre for Stem Cell Research, Università degli Studi di Milano, Milan, Italy
| | - Elena Cattaneo
- Department of Pharmacological Sciences and Centre for Stem Cell Research, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
79
|
Tan PLJ. Company profile: Tissue regeneration for diabetes and neurological diseases at Living Cell Technologies. Regen Med 2010; 5:181-7. [PMID: 20210578 DOI: 10.2217/rme.10.4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Living Cell Technologies’ (LCT’s) cell-based therapeutic for Type 1 diabetes, DIABECELL®, comprises encapsulated porcine insulin-producing cells. DIABECELL is presently in a Phase II clinical trial in New Zealand following positive early results. The cells are implanted into the abdomen to replace the patient’s pancreatic β-islet cells that have been lost as a result of autoimmune disease. LCT is also developing brain choroid plexus cells for the treatment of neurologic diseases. The aim is to enhance the brain’s natural repair mechanism by implanting cells releasing neurotrophins. Choroid plexus cell implants alleviate disease in animal models of Parkinson’s disease, Huntington’s disease and stroke. LCT encapsulates all cells in alginate, permitting implantation without using immunosuppressive drugs.
Collapse
|
80
|
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: 118] [Impact Index Per Article: 7.9] [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
|
81
|
Giralt A, Friedman HC, Caneda-Ferrón B, Urbán N, Moreno E, Rubio N, Blanco J, Peterson A, Canals JM, Alberch J. BDNF regulation under GFAP promoter provides engineered astrocytes as a new approach for long-term protection in Huntington's disease. Gene Ther 2010; 17:1294-308. [PMID: 20463759 DOI: 10.1038/gt.2010.71] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is the main candidate for neuroprotective therapeutic strategies for Huntington's disease. However, the administration system and the control over the dosage are still important problems to be solved. Here we generated transgenic mice overexpressing BDNF under the promoter of the glial fibrillary acidic protein (GFAP) (pGFAP-BDNF mice). These mice are viable and have a normal phenotype. However, intrastriatal administration of quinolinate increased the number of reactive astrocytes and enhanced the release of BDNF in pGFAP-BDNF mice compared with wild-type mice. Coincidentally, pGFAP-BDNF mice are more resistant to quinolinate than wild-type mice, suggesting a protective effect of astrocyte-derived BDNF. To verify this, we next cultured astrocytes from pGFAP-BDNF and wild-type mice for grafting. Wild-type and pGFAP-BDNF-derived astrocytes behave similarly in nonlesioned mice. However, pGFAP-BDNF-derived astrocytes showed higher levels of BDNF and larger neuroprotective effects than the wild-type ones when quinolinate was injected 30 days after grafting. Interestingly, mice grafted with pGFAP-BDNF astrocytes showed important and sustained behavioral improvements over time after quinolinate administration as compared with mice grafted with wild-type astrocytes. These findings show that astrocytes engineered to release BDNF can constitute a therapeutic approach for Huntington's disease.
Collapse
Affiliation(s)
- A Giralt
- Facultat de Medicina, Departament de Biologia Cel·lular, Immunologia i Neurociències, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Universitat de Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Puerta E, Hervias I, Barros-Miñones L, Jordan J, Ricobaraza A, Cuadrado-Tejedor M, García-Osta A, Aguirre N. Sildenafil protects against 3-nitropropionic acid neurotoxicity through the modulation of calpain, CREB, and BDNF. Neurobiol Dis 2010; 38:237-45. [DOI: 10.1016/j.nbd.2010.01.013] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2009] [Revised: 12/24/2009] [Accepted: 01/17/2010] [Indexed: 11/26/2022] Open
|
83
|
Massa SM, Yang T, Xie Y, Shi J, Bilgen M, Joyce JN, Nehama D, Rajadas J, Longo FM. Small molecule BDNF mimetics activate TrkB signaling and prevent neuronal degeneration in rodents. J Clin Invest 2010; 120:1774-85. [PMID: 20407211 DOI: 10.1172/jci41356] [Citation(s) in RCA: 308] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Accepted: 02/17/2010] [Indexed: 02/01/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) activates the receptor tropomyosin-related kinase B (TrkB) with high potency and specificity, promoting neuronal survival, differentiation, and synaptic function. Correlations between altered BDNF expression and/or function and mechanism(s) underlying numerous neurodegenerative conditions, including Alzheimer disease and traumatic brain injury, suggest that TrkB agonists might have therapeutic potential. Using in silico screening with a BDNF loop-domain pharmacophore, followed by low-throughput in vitro screening in mouse fetal hippocampal neurons, we have efficiently identified small molecules with nanomolar neurotrophic activity specific to TrkB versus other Trk family members. Neurotrophic activity was dependent on TrkB and its downstream targets, although compound-induced signaling activation kinetics differed from those triggered by BDNF. A selected prototype compound demonstrated binding specificity to the extracellular domain of TrkB. In in vitro models of neurodegenerative disease, it prevented neuronal degeneration with efficacy equal to that of BDNF, and when administered in vivo, it caused hippocampal and striatal TrkB activation in mice and improved motor learning after traumatic brain injury in rats. These studies demonstrate the utility of loop modeling in drug discovery and reveal what we believe to be the first reported small molecules derived from a targeted BDNF domain that specifically activate TrkB.We propose that these compounds constitute a novel group of tools for the study of TrkB signaling and may provide leads for developing new therapeutic agents for neurodegenerative diseases.
Collapse
Affiliation(s)
- Stephen M Massa
- Department of Neurology and Laboratory for Computational Neurochemistry and Drug Discovery, UCSF, San Francisco, California, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Zinc sulfate could be a potential agent for the treatment of. Huntington's disease through activating central TrkB signaling. CNS Spectr 2010; 15:56-7. [PMID: 20394186 DOI: 10.1017/s1092852900000316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
85
|
Cazzin C, Ring CJA. Recent advances in the manipulation of murine gene expression and its utility for the study of human neurological disease. Biochim Biophys Acta Mol Basis Dis 2009; 1802:796-807. [PMID: 20004244 DOI: 10.1016/j.bbadis.2009.11.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Revised: 11/24/2009] [Accepted: 11/25/2009] [Indexed: 12/11/2022]
Abstract
Transgenic mouse models have vastly contributed to our knowledge of the genetic and molecular pathways underlying the pathogenesis of neurological disorders that affect millions of people worldwide. Not only have they allowed the generation of disease models mimicking the human pathological state but they have also permitted the exploration of the pathological role of specific genes through the generation of knock-out and knock-in models. Classical constitutive transgenic mice have several limitations however, due to behavioral adaptation process occurring and conditional mouse models are time-consuming and often lack extensive spatial or temporal control of gene manipulation. These limitations could be overcome by means of innovative methods that are now available such as RNAi, viral vectors and large cloning DNA vectors. These tools have been extensively used for the generation of mouse models and are characterized by the superior control of transgene expression that has been proven invaluable in the assessment of novel treatments for neurological diseases and to further investigate the molecular processes underlying the etiopathology of neurological disorders. Furthermore, in association with classical transgenic mouse models, they have allowed the validation of innovative therapeutic strategies for the treatment of human neurological disorders. This review describes how these tools have overcome the limitations of classical transgenic mouse models and how they have been of value for the study of human neurological diseases.
Collapse
Affiliation(s)
- Chiara Cazzin
- Biology Department A&S DPU, Neuroscience CEDD, GlaxoSmithKline, Medicines Research Center, Verona, Italy.
| | | |
Collapse
|
86
|
Lim ST, Airavaara M, Harvey BK. Viral vectors for neurotrophic factor delivery: a gene therapy approach for neurodegenerative diseases of the CNS. Pharmacol Res 2009; 61:14-26. [PMID: 19840853 DOI: 10.1016/j.phrs.2009.10.002] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2009] [Revised: 10/11/2009] [Accepted: 10/11/2009] [Indexed: 01/11/2023]
Abstract
The clinical manifestation of most diseases of the central nervous system results from neuronal dysfunction or loss. Diseases such as stroke, epilepsy and neurodegeneration (e.g. Alzheimer's disease and Parkinson's disease) share common cellular and molecular mechanisms (e.g. oxidative stress, endoplasmic reticulum stress, mitochondrial dysfunction) that contribute to the loss of neuronal function. Neurotrophic factors (NTFs) are secreted proteins that regulate multiple aspects of neuronal development including neuronal maintenance, survival, axonal growth and synaptic plasticity. These properties of NTFs make them likely candidates for preventing neurodegeneration and promoting neuroregeneration. One approach to delivering NTFs to diseased cells is through viral vector-mediated gene delivery. Viral vectors are now routinely used as tools for studying gene function as well as developing gene-based therapies for a variety of diseases. Currently, many clinical trials using viral vectors in the nervous system are underway or completed, and seven of these trials involve NTFs for neurodegeneration. In this review, we discuss viral vector-mediated gene transfer of NTFs to treat neurodegenerative diseases of the central nervous system.
Collapse
Affiliation(s)
- Seung T Lim
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057, United States
| | | | | |
Collapse
|
87
|
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: 23] [Impact Index Per Article: 1.4] [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
|
88
|
Almeida S, Laço M, Cunha-Oliveira T, Oliveira CR, Rego AC. BDNF regulates BIM expression levels in 3-nitropropionic acid-treated cortical neurons. Neurobiol Dis 2009; 35:448-56. [PMID: 19555760 DOI: 10.1016/j.nbd.2009.06.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2009] [Revised: 06/03/2009] [Accepted: 06/11/2009] [Indexed: 01/09/2023] Open
Abstract
3-Nitropropionic acid (3-NP) is an irreversible inhibitor of succinate dehydrogenase that has been used to explore the primary mechanisms of cell death associated with mitochondrial dysfunction and neurodegeneration in Huntington's disease. In this study we investigated the ability of brain-derived neurotrophic factor (BDNF) to suppress mitochondrial-dependent cell death induced by 3-NP in primary cortical neurons. This neurotrophin prevented 3-NP-induced release of cytochrome c and Smac/Diablo, caspase-3-like activity and nuclear condensation/fragmentation. Furthermore, it greatly increased phosphorylation of Akt and MAPK, suggesting the involvement of these signalling pathways in BDNF neuroprotection. Interestingly, BDNF decreased the levels of the pro-apoptotic protein Bim in mitochondrial and total cell lysates through the activation of the MEK1/2 pathway. This effect was due to an increase in the degradation rates of Bim. Our data support an important role for BDNF, in protecting cortical neurons against apoptotic cell death caused by inhibition of mitochondrial complex II.
Collapse
Affiliation(s)
- Sandra Almeida
- Institute of Biochemistry, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal
| | | | | | | | | |
Collapse
|
89
|
Gagliardi C, Bunnell BA. Large animal models of neurological disorders for gene therapy. ILAR J 2009; 50:128-43. [PMID: 19293458 DOI: 10.1093/ilar.50.2.128] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
he development of therapeutic interventions for genetic disorders and diseases that affect the central nervous system (CNS) has proven challenging. There has been significant progress in the development of gene therapy strategies in murine models of human disease, but gene therapy outcomes in these models do not always translate to the human setting. Therefore, large animal models are crucial to the development of diagnostics, treatments, and eventual cures for debilitating neurological disorders. This review focuses on the description of large animal models of neurological diseases such as lysosomal storage diseases, Parkinsons disease, Huntingtons disease, and neuroAIDS. The review also describes the contributions of these models to progress in gene therapy research.
Collapse
|
90
|
Fumagalli F, Molteni R, Calabrese F, Maj PF, Racagni G, Riva MA. Neurotrophic factors in neurodegenerative disorders : potential for therapy. CNS Drugs 2009; 22:1005-19. [PMID: 18998739 DOI: 10.2165/0023210-200822120-00004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Finding an effective therapy to treat chronic neurodegenerative disorders still represents an unmet and elusive goal, mainly because so many pathogenic variables come into play in these diseases. Recent emphasis has been placed on the role of neurotrophic factors in the aetiology of such disorders because of their role in the survival of different cell phenotypes under various adverse conditions, including neurodegeneration.This review summarizes the current status and the efforts to treat neurodegenerative disorders by the exogenous administration of neurotrophic factors in an attempt to replenish trophic supply, the paucity of which may contribute to the development of the illness. Although promising results have been seen in animal models, this approach still meets disparate and often insurmountable problems in clinical settings, presumably related to the unique nature of the human being.
Collapse
Affiliation(s)
- Fabio Fumagalli
- Department of Pharmacological Sciences, Center of Neuropharmacology, University of Milan, Milan, Italy
| | | | | | | | | | | |
Collapse
|
91
|
Nanou A, Azzouz M. Gene therapy for neurodegenerative diseases based on lentiviral vectors. PROGRESS IN BRAIN RESEARCH 2009; 175:187-200. [DOI: 10.1016/s0079-6123(09)17513-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
92
|
Duan W, Peng Q, Masuda N, Ford E, Tryggestad E, Ladenheim B, Zhao M, Cadet JL, Wong J, Ross CA. Sertraline slows disease progression and increases neurogenesis in N171-82Q mouse model of Huntington's disease. Neurobiol Dis 2008; 30:312-322. [PMID: 18403212 PMCID: PMC3683653 DOI: 10.1016/j.nbd.2008.01.015] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2007] [Revised: 01/09/2008] [Accepted: 01/31/2008] [Indexed: 12/12/2022] Open
Abstract
Huntington's disease (HD) is an inherited progressive neurodegenerative disorder resulting from CAG repeat expansion in the gene that encodes for the protein huntingtin. To identify neuroprotective compound (s) that can slow down disease progression and can be administered long term with few side effects in Huntington's disease, we investigated the effect of sertraline, a selective serotonin reuptake inhibitor (SSRI) which has been shown to upregulate BDNF levels in rodent brains. We report here that in HD mice sertraline increased BDNF levels, preserved chaperone protein HSP70 and Bcl-2 levels in brains, attenuated the progression of brain atrophy and behavioral abnormalities and thereby increased survival. Sertraline also enhanced neurogenesis, which appeared to be responsible for mediating the beneficial effects of sertraline in HD mice. Additionally, the effective levels of sertraline are comparable to the safe levels achievable in humans. The findings suggest that sertraline is a potential candidate for treatment of HD patients.
Collapse
Affiliation(s)
- Wenzhen Duan
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, CMSC 8-121, 600 North Wolfe Street, Baltimore, MD 21287, USA.
| | - Qi Peng
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, CMSC 8-121, 600 North Wolfe Street, Baltimore, MD 21287, USA
| | - Naoki Masuda
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, CMSC 8-121, 600 North Wolfe Street, Baltimore, MD 21287, USA
| | - Eric Ford
- Department of Radiology, Johns Hopkins University School of Medicine, USA
| | - Erik Tryggestad
- Department of Radiology, Johns Hopkins University School of Medicine, USA
| | - Bruce Ladenheim
- Molecular Neuropsychiatry Branch, National Institute on Drug Abuse, 5500 Nathan Shock Drive, Baltimore, MD 21224, USA
| | - Ming Zhao
- Oncology Analytical Pharmacology Core, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Branch, National Institute on Drug Abuse, 5500 Nathan Shock Drive, Baltimore, MD 21224, USA
| | - John Wong
- Department of Radiology, Johns Hopkins University School of Medicine, USA
| | - Christopher A Ross
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, CMSC 8-121, 600 North Wolfe Street, Baltimore, MD 21287, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
93
|
Bergen JM, Pun SH. Analysis of the intracellular barriers encountered by nonviral gene carriers in a model of spatially controlled delivery to neurons. J Gene Med 2008; 10:187-97. [PMID: 18064730 DOI: 10.1002/jgm.1137] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Neuron-specific, nonviral gene delivery vehicles are useful tools for the potential treatment of neurological disease and spinal cord injury. For minimally invasive, peripheral administration, gene carriers must efficiently mediate uptake at axon terminals, retrograde axonal transport, vesicular escape, and nuclear entry. The design of improved vehicles will benefit from an understanding of the barriers that limit nonviral delivery to neurons. Here, we demonstrate a detailed analysis of intracellular trafficking of both a lipid-based and a polymer-based delivery vehicle following site-specific exposure to neuron-like cells. METHODS Site-specific exposure of gene carriers to soma or neurites of neuron-like PC-12 cells was accomplished using a microfluidic, compartmented culture chamber. Binding and internalization of vehicles at neurites and soma were quantified using an environmentally sensitive fluorescent marker. The intracellular transport of gene carriers was analyzed by time-lapse particle tracking in live cells, and transfection efficiencies were measured using green fluorescent protein (GFP) as a reporter gene. RESULTS While the lipid-based carrier mediated measurable transfection when delivered to neuronal soma, neuritic delivery of this formulation failed to produce reporter gene expression due to limited internalization and transport. In contrast, the polymeric nanoparticles displayed active retrograde transport toward neuronal soma, but failed to produce measurable reporter gene expression. CONCLUSIONS These results highlight distinct intracellular barriers preventing efficient neuronal transfection by the nonviral carriers examined, and provide a basis for the rational improvement of existing nonviral systems.
Collapse
Affiliation(s)
- Jamie M Bergen
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | | |
Collapse
|
94
|
Abstract
After nearly 20 years of preclinical experimentation with various gene delivery approaches in animal models of Parkinson's disease (PD), clinical trials are finally underway. The risk/benefit ratio for these procedures is now generally considered acceptable under approved protocols. The current vehicle for gene delivery to the human brain is recombinant adeno-associated viral vector, which is nonpathogenic and non-self-amplifying. Candidate genes tested in PD patients encode 1) glutamic acid decarboxylase, which is injected into the subthalamic nucleus to catalyze biosynthesis of the inhibitory neurotransmitter gamma-aminobutyric acid and so essentially mimic deep brain stimulation of this nucleus; 2) aromatic l-amino acid decarboxylase, which converts l-dopa to dopamine; and 3) neurturin, a member of the glial cell line-derived neurotrophic factor family. Unraveling the genetic underpinnings of PD could allow gene therapy to go beyond modulating neurotransmission or providing trophic effects to dopaminergic neurons by delivering a specific missing or defective gene. For example, the parkin gene (PARK2) is linked to recessively inherited PD due to loss of function mutations; it prevents alpha-synuclein-induced degeneration of nigral dopaminergic neurons in rats and nonhuman primates. On the other hand, for dominantly inherited Huntington's disease (HD), in which an expanded polyglutamine tract imparts to the protein huntingtin a toxic gain of function, repressing expression of the mutant allele in the striatum using RNA interference technology mitigates pathology and delays the phenotype in a mouse model. Here we review the current state of preclinical and clinical gene therapy studies conducted in PD and HD.
Collapse
Affiliation(s)
- Hideki Mochizuki
- grid.258269.20000000417622738Research Institute for Diseases of Old Age, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyoku, 113-8421 Tokyo, Japan
- grid.258269.20000000417622738Department of Neurology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyoku, 113-8421 Tokyo, Japan
| | - Toru Yasuda
- grid.258269.20000000417622738Research Institute for Diseases of Old Age, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyoku, 113-8421 Tokyo, Japan
| | - M. Maral Mouradian
- grid.430387.b0000000419368796Center for Neurodegenerative and Neuroimmunologic Diseases, Department of Neurology, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, 08854 Piscataway, New Jersey
| |
Collapse
|
95
|
Kells AP, Connor B. AAV-mediated expression of Bcl-xL or XIAP fails to induce neuronal resistance against quinolinic acid-induced striatal lesioning. Neurosci Lett 2008; 436:326-30. [PMID: 18406531 DOI: 10.1016/j.neulet.2008.03.051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2007] [Revised: 03/04/2008] [Accepted: 03/17/2008] [Indexed: 11/26/2022]
Abstract
Apoptotic mechanisms have been proposed to contribute to the selective loss of medium spiny striatal projection neurons in Huntington's disease (HD). This raises the question as to whether enhancing the expression of anti-apoptotic factors in vulnerable striatal projection neurons can reduce their susceptibility to neurotoxic processes occurring in the HD brain. In this study AAV 1/2 vectors encoding either the anti-apoptotic factor Bcl-xL or XIAP were used to transduce striatal neurons prior to an intrastriatal injection of the excitotoxic glutamate analogue quinolinic acid (QA). AAV 1/2 vector treated rats were observed in behavioural tests undertaken to assess whether anti-apoptotic factor expression provided amelioration of motor function impairment following unilateral QA-induced striatal lesioning. AAV-XIAP treated rats displayed complete amelioration of an ipsilateral forelimb use bias relative to control animals. However, neither AAV-XIAP nor AAV-Bcl-xL treated rats demonstrated an improvement in sensorimotor neglect compared to control animals. Furthermore, we did not observe a significant reduction of QA-induced pathology in assessed neuronal populations of the basal ganglia. These results indicate that sole enhancement of XIAP or Bcl-xL is not sufficient to counteract QA-induced excitotoxic insult of striatal neurons.
Collapse
Affiliation(s)
- Adrian P Kells
- Department of Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, New Zealand.
| | | |
Collapse
|
96
|
Kells AP, Henry RA, Connor B. AAV-BDNF mediated attenuation of quinolinic acid-induced neuropathology and motor function impairment. Gene Ther 2008; 15:966-77. [PMID: 18323792 DOI: 10.1038/gt.2008.23] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Maintenance and plasticity of striatal neurons is dependent on brain-derived neurotrophic factor (BDNF), which is depleted in the Huntington's disease striatum due to reduced expression and disrupted corticostriatal transportation. In this study we demonstrate that overexpression of BDNF in the striatum attenuates motor impairment and reduces the extent of striatal damage following quinolinic acid lesioning. Transfer of the BDNF gene to striatal neurons using serotype 1/2 adeno-associated viral vectors enhanced BDNF protein levels in the striatum, but induced weight loss and seizure activity following long-term high-level expression. Lower concentration BDNF expression supported striatal neurons against excitotoxic insult, as demonstrated by enhanced krox-24 immunopositive neuron survival, reduction of striatal atrophy and maintenance of the patch/matrix organization. Additionally, BDNF expression attenuated motor impairment in the forelimb use cylinder test, sensorimotor neglect in the corridor food selection task and reversed apomorphine-induced rotational behaviour. Direct correlations were shown for the first time between BDNF-mediated attenuation of behavioural impairment and the integrity of the globus pallidus, seemingly independent from the severity of striatal lesioning. These results demonstrate that BDNF holds considerable therapeutic potential for alleviating both neuropathological and motor function deficits in the Huntington's disease brain, and the critical role of pallidal neurons in facilitating motor performance.
Collapse
Affiliation(s)
- A P Kells
- Department of Pharmacology and Clinical Pharmacology, Neural Repair and Neurogenesis Laboratory, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | | | | |
Collapse
|
97
|
Wang B, Zou X, Zhang H, Duan D, Ju L, Jiang X, Sun X, Zhao C, Zhao H, Guo J, Xu C, Gao E, Xu Q. Establishment of an immortalized GABAergic neuronal progenitor cell line from embryonic ventral mesencephalon in the rat. Brain Res 2008; 1210:63-75. [PMID: 18407253 DOI: 10.1016/j.brainres.2008.02.062] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2007] [Revised: 02/15/2008] [Accepted: 02/15/2008] [Indexed: 11/29/2022]
Abstract
Effective cell replacement therapies for neurological disease require neuron-restricted precursors as grafted cells. The problem of obtaining sufficient grafts for transplantation can be resolved by creating an appropriate immortalized cell line. In the present study, a thermally controlled immortalized GABAergic neuronal progenitor cell line (RMNE6) was established from E13 rat ventral mesencephalon cells immortalized using the temperature-sensitive mutant of SV40 large T antigen (ts-TAg). RMNE6 cells proliferated rapidly and expressed a neuron-like phenotype at the permissive temperature (33 degrees C), but eventually stopped growing at the non-permissive temperature (39 degrees C). Expression of the neuronal markers PSA-NCAM, beta-tubulin III and MAP2 by RMNE6 cells was confirmed by RT-PCR or immunocytochemistry. Furthermore, these cells exhibited functional GABAergic neuron properties, as evidenced by the expression of glutamate decarboxylase (GAD) as well as the synthesis and release of the neurotransmitter GABA in a calcium-dependent manner. Moreover, RMNE6 cells spontaneously expressed and secreted several neurotrophic factors, such as NGF, BDNF, NT-3, NT-4/5, and GDNF. The cells survived well and kept expression of SV40 Tag, GAD65/67 and GABA in the striatum, at least 28 days after being transplanted in the rat brain. Tumorigenesis assays confirmed the safety of the immortalized cell line in vivo. Taken together, the results support the use of RMNE6 cells as an ideal cell model for transplantation research aimed at the treatment and prevention of neurodegenerative disease.
Collapse
Affiliation(s)
- Beibei Wang
- Beijing Institute for Neuroscience, Capital Medical University, Beijing Center of Neural Regeneration and Repair, Key Laboratory of Neurodegenerative diseases of Ministry of Education of China, Beijing 100069, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Peng Q, Masuda N, Jiang M, Li Q, Zhao M, Ross CA, Duan W. The antidepressant sertraline improves the phenotype, promotes neurogenesis and increases BDNF levels in the R6/2 Huntington's disease mouse model. Exp Neurol 2007; 210:154-63. [PMID: 18096160 DOI: 10.1016/j.expneurol.2007.10.015] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2007] [Revised: 10/02/2007] [Accepted: 10/18/2007] [Indexed: 02/08/2023]
Abstract
Huntington's disease (HD) is an inherited progressive neurodegenerative disorder characterized by progressive movement, psychiatric and cognitive disturbances. Previous studies have indicated that HD pathogenesis may be mediated in part by loss of brain derived neurotrophic factor (BDNF). Antidepressants selectively blocking serotonin reuptake can increase BDNF levels, and also may increase neurogenesis. Here we report that an SSRI antidepressant, sertraline, prolongs survival, improves motor performance, and ameliorates brain atrophy in the R6/2 HD mouse model. Six-week-old R6/2 mice and nontransgenic control mice were administered either sertraline or vehicle daily. Motor function was assessed in an accelerating rotarod test and evaluated at 10 weeks. R6/2 mice exhibited reduced time on the rod. Sertraline treatment improved the motor performance in R6/2 mice, but did not affect nontransgenic mice. R6/2 mice showed significant striatal atrophy which was reduced by sertraline treatment. These beneficial effects of sertraline are associated with enhanced neurogenesis and increased BDNF levels in brain treated with sertraline. The effective serum and brain levels of sertraline are comparable to the levels achieved in human antidepressant treatment. Our findings provide evidence that sertraline is neuroprotective in this HD model. Successful treatment with sertraline in depressed HD patients has been reported; moreover, sertraline is safe and well-tolerated for long-term administration, including in HD patients. Our findings suggest that a clinical trial of SSRI treatment in order to retard disease progression in human HD may be warranted.
Collapse
Affiliation(s)
- Qi Peng
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | | | | | | | | | | |
Collapse
|
99
|
Park IK, Lasiene J, Chou SH, Horner PJ, Pun SH. Neuron-specific delivery of nucleic acids mediated by Tet1-modified poly(ethylenimine). J Gene Med 2007; 9:691-702. [PMID: 17582226 PMCID: PMC2633605 DOI: 10.1002/jgm.1062] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The development of minimally invasive, non-viral gene delivery vehicles for the central nervous system (CNS) is an important technology goal in the advancement of molecular therapies for neurological diseases. One approach is to deliver materials peripherally that are recognized and retrogradely transported by motor neurons toward the CNS. Tet1 is a peptide identified by Boulis and coworkers to possess the binding characteristics of tetanus toxin, which interacts specifically with motor neurons and undergoes fast, retrograde delivery to cell soma. In this work, Tet1-poly(ethylenimine) (Tet1-PEI) was synthesized and evaluated as a neurontargeted delivery vehicle. METHODS Tet1-PEI and NT-PEI (neurotensin-PEI) were synthesized and complexed with plasmid DNA to form polyplexes. Polyplexes were assessed for binding and uptake in differentiated neuron-like PC-12 cells by flow cytometry and confocal microscopy. In order to determine gene delivery efficiency, polyplexes were exposed to PC-12 cells at various stages of differentiation. Targeted binding of polyplexes with primary neurons was studied using dorsal root ganglion cells. RESULTS Tet1-PEI and NT-PEI polyplexes bound specifically to differentiated PC-12 cells. The specificity of the interaction was confirmed by delivery to non-neuronal cells and by competition studies with free ligands. Tet1-PEI polyplexes preferentially transfected PC-12 cells undergoing NGF-induced differentiation. Finally, neuron-specific binding of Tet1-PEI polyplexes was confirmed in primary neurons. CONCLUSIONS These studies demonstrate the potential of Tet1-PEI as a neuron-targeted material for non-invasive CNS delivery. Tet1-PEI binds specifically and is internalized by neuron-like PC-12 cells and primary dorsal root ganglion. Future work will include evaluation of siRNA delivery with these vectors.
Collapse
Affiliation(s)
- In-Kyu Park
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Jurate Lasiene
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA
| | - Shinn-Huey Chou
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Philip J. Horner
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA
| | - Suzie H. Pun
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Correspondence to: Suzie H. Pun, Department of Bioengineering, University of Washington, Seattle, WA 98195, USA. E-mail:
| |
Collapse
|
100
|
Bergen JM, Park IK, Horner PJ, Pun SH. Nonviral approaches for neuronal delivery of nucleic acids. Pharm Res 2007; 25:983-98. [PMID: 17932730 PMCID: PMC2292496 DOI: 10.1007/s11095-007-9439-5] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2007] [Accepted: 08/20/2007] [Indexed: 12/23/2022]
Abstract
The delivery of therapeutic nucleic acids to neurons has the potential to treat neurological disease and spinal cord injury. While select viral vectors have shown promise as gene carriers to neurons, their potential as therapeutic agents is limited by their toxicity and immunogenicity, their broad tropism, and the cost of large-scale formulation. Nonviral vectors are an attractive alternative in that they offer improved safety profiles compared to viruses, are less expensive to produce, and can be targeted to specific neuronal subpopulations. However, most nonviral vectors suffer from significantly lower transfection efficiencies than neurotropic viruses, severely limiting their utility in neuron-targeted delivery applications. To realize the potential of nonviral delivery technology in neurons, vectors must be designed to overcome a series of extra- and intracellular barriers. In this article, we describe the challenges preventing successful nonviral delivery of nucleic acids to neurons and review strategies aimed at overcoming these challenges.
Collapse
Affiliation(s)
- Jamie M Bergen
- Bioengineering, University of Washington, Seattle, WA 98195, USA
| | | | | | | |
Collapse
|