201
|
Convection Enhanced Delivery: A Comparison of infusion characteristics in ex vivo and in vivo non-human primate brain tissue. Ann Neurosci 2014; 20:108-14. [PMID: 25206026 PMCID: PMC4117126 DOI: 10.5214/ans.0972.7531.200306] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 05/17/2013] [Accepted: 07/29/2013] [Indexed: 11/25/2022] Open
Abstract
Background Convection enhanced delivery (CED) is emerging as a promising infusion toolto facilitate delivery of therapeutic agents into the brain via mechanically controlled pumps. Infusion protocols and catheter design have an important impact on delivery. CED is a valid alternative for systemic administration of agents in clinical trials for cell and gene therapies. Where gel and ex vivo models are not sufficient in modeling the disease, in vivo models allow researchers to better understand the underlying mechanisms of neuron degeneration, which is helpful in finding novel approaches to control the process or reverse the progression. Determining the risks, benefits, and efficacy of new gene therapies introduced via CED will pave a way to enter human clinical trial. Purpose The objective of this study is to compare volume distribution (Vd)/ volume infused (Vi) ratios and backflow measurements following CED infusions in ex vivo versus in vivo non-human primate brain tissue, based on infusion protocols developed in vitro. Methods In ex vivo infusions, the first brain received 2 infusions using a balloon catheter at rates of 1 μL/min and 2 μL/min for 30 minutes. The second and third brains received infusions using a valve-tip (VT) catheter at 1 μL/min for 30 minutes. The fourth brain received a total of 45 μL infused at a rate of 1 μL/min for 15 minutes followed by 2 μL/min for 15 minutes. Imaging was performed (SPGR FA34) every 3 minutes. In the in vivo group, 4 subjects received a total of 8 infusions of 50 μL. Subjects 1 and 2 received infusions at 1.0 μL/min using a VT catheter in the left hemisphere and a smart-flow (SF) catheter in the right hemisphere. Subjects 3 and 4 each received 1 infusion in the left and right hemisphere at 1.0 μL/min. Results MRI calculations of Vd/Vi did not significantly differ from those obtained on post-mortem pathology. The mean measured Vd/Vi of in vivo (5.23 + /-1.67) compared to ex vivo (2.17 + /-1.39) demonstrated a significantly larger Vd/Vi for in vivo by 2.4 times (p = 0.0017). Conclusion We detected higher ratios in the in vivo subjects than in ex vivo. This difference could be explained by the extra cellular space volume fraction. Studies evaluating backflow and morphology use in vivo tissue as a medium are recommended. Further investigation is warranted to evaluate the role blood pressure and heart rate may play in human CED clinical trials.
Collapse
|
202
|
Müller J, Ossig C, Greiner JFW, Hauser S, Fauser M, Widera D, Kaltschmidt C, Storch A, Kaltschmidt B. Intrastriatal transplantation of adult human neural crest-derived stem cells improves functional outcome in parkinsonian rats. Stem Cells Transl Med 2014; 4:31-43. [PMID: 25479965 DOI: 10.5966/sctm.2014-0078] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Parkinson's disease (PD) is considered the second most frequent and one of the most severe neurodegenerative diseases, with dysfunctions of the motor system and with nonmotor symptoms such as depression and dementia. Compensation for the progressive loss of dopaminergic (DA) neurons during PD using current pharmacological treatment strategies is limited and remains challenging. Pluripotent stem cell-based regenerative medicine may offer a promising therapeutic alternative, although the medical application of human embryonic tissue and pluripotent stem cells is still a matter of ethical and practical debate. Addressing these challenges, the present study investigated the potential of adult human neural crest-derived stem cells derived from the inferior turbinate (ITSCs) transplanted into a parkinsonian rat model. Emphasizing their capability to give rise to nervous tissue, ITSCs isolated from the adult human nose efficiently differentiated into functional mature neurons in vitro. Additional successful dopaminergic differentiation of ITSCs was subsequently followed by their transplantation into a unilaterally lesioned 6-hydroxydopamine rat PD model. Transplantation of predifferentiated or undifferentiated ITSCs led to robust restoration of rotational behavior, accompanied by significant recovery of DA neurons within the substantia nigra. ITSCs were further shown to migrate extensively in loose streams primarily toward the posterior direction as far as to the midbrain region, at which point they were able to differentiate into DA neurons within the locus ceruleus. We demonstrate, for the first time, that adult human ITSCs are capable of functionally recovering a PD rat model.
Collapse
Affiliation(s)
- Janine Müller
- Molecular Neurobiology, University of Bielefeld, Bielefeld, Germany; Division of Neurodegenerative Diseases, Department of Neurology, and Center for Regenerative Therapies Dresden, Dresden University of Technology, Dresden, Germany; German Center for Neurodegenerative Diseases Dresden, Dresden, Germany; Cell Biology, University of Bielefeld, Bielefeld, Germany
| | - Christiana Ossig
- Molecular Neurobiology, University of Bielefeld, Bielefeld, Germany; Division of Neurodegenerative Diseases, Department of Neurology, and Center for Regenerative Therapies Dresden, Dresden University of Technology, Dresden, Germany; German Center for Neurodegenerative Diseases Dresden, Dresden, Germany; Cell Biology, University of Bielefeld, Bielefeld, Germany
| | - Johannes F W Greiner
- Molecular Neurobiology, University of Bielefeld, Bielefeld, Germany; Division of Neurodegenerative Diseases, Department of Neurology, and Center for Regenerative Therapies Dresden, Dresden University of Technology, Dresden, Germany; German Center for Neurodegenerative Diseases Dresden, Dresden, Germany; Cell Biology, University of Bielefeld, Bielefeld, Germany
| | - Stefan Hauser
- Molecular Neurobiology, University of Bielefeld, Bielefeld, Germany; Division of Neurodegenerative Diseases, Department of Neurology, and Center for Regenerative Therapies Dresden, Dresden University of Technology, Dresden, Germany; German Center for Neurodegenerative Diseases Dresden, Dresden, Germany; Cell Biology, University of Bielefeld, Bielefeld, Germany
| | - Mareike Fauser
- Molecular Neurobiology, University of Bielefeld, Bielefeld, Germany; Division of Neurodegenerative Diseases, Department of Neurology, and Center for Regenerative Therapies Dresden, Dresden University of Technology, Dresden, Germany; German Center for Neurodegenerative Diseases Dresden, Dresden, Germany; Cell Biology, University of Bielefeld, Bielefeld, Germany
| | - Darius Widera
- Molecular Neurobiology, University of Bielefeld, Bielefeld, Germany; Division of Neurodegenerative Diseases, Department of Neurology, and Center for Regenerative Therapies Dresden, Dresden University of Technology, Dresden, Germany; German Center for Neurodegenerative Diseases Dresden, Dresden, Germany; Cell Biology, University of Bielefeld, Bielefeld, Germany
| | - Christian Kaltschmidt
- Molecular Neurobiology, University of Bielefeld, Bielefeld, Germany; Division of Neurodegenerative Diseases, Department of Neurology, and Center for Regenerative Therapies Dresden, Dresden University of Technology, Dresden, Germany; German Center for Neurodegenerative Diseases Dresden, Dresden, Germany; Cell Biology, University of Bielefeld, Bielefeld, Germany
| | - Alexander Storch
- Molecular Neurobiology, University of Bielefeld, Bielefeld, Germany; Division of Neurodegenerative Diseases, Department of Neurology, and Center for Regenerative Therapies Dresden, Dresden University of Technology, Dresden, Germany; German Center for Neurodegenerative Diseases Dresden, Dresden, Germany; Cell Biology, University of Bielefeld, Bielefeld, Germany
| | - Barbara Kaltschmidt
- Molecular Neurobiology, University of Bielefeld, Bielefeld, Germany; Division of Neurodegenerative Diseases, Department of Neurology, and Center for Regenerative Therapies Dresden, Dresden University of Technology, Dresden, Germany; German Center for Neurodegenerative Diseases Dresden, Dresden, Germany; Cell Biology, University of Bielefeld, Bielefeld, Germany
| |
Collapse
|
203
|
Serguera C, Bemelmans AP. Gene therapy of the central nervous system: general considerations on viral vectors for gene transfer into the brain. Rev Neurol (Paris) 2014; 170:727-38. [PMID: 25459120 DOI: 10.1016/j.neurol.2014.09.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 08/13/2014] [Accepted: 09/10/2014] [Indexed: 02/04/2023]
Abstract
The last decade has nourished strong doubts on the beneficial prospects of gene therapy for curing fatal diseases. However, this climate of reservation is currently being transcended by the publication of several successful clinical protocols, restoring confidence in the appropriateness of therapeutic gene transfer. A strong sign of this present enthusiasm for gene therapy by clinicians and industrials is the market approval of the therapeutic viral vector Glybera, the first commercial product in Europe of this class of drug. This new field of medicine is particularly attractive when considering therapies for a number of neurological disorders, most of which are desperately waiting for a satisfactory treatment. The central nervous system is indeed a very compliant organ where gene transfer can be stable and successful if provided through an appropriate strategy. The purpose of this review is to present the characteristics of the most efficient virus-derived vectors used by researchers and clinicians to genetically modify particular cell types or whole regions of the brain. In addition, we discuss major issues regarding side effects, such as genotoxicity and immune response associated to the use of these vectors.
Collapse
Affiliation(s)
- C Serguera
- CEA, DSV, I(2)BM, Molecular Imaging Research Center (MIRCen) and CNRS, CEA URA 2210, 18, route du Panorama, 92265 Fontenay-aux-Roses, France
| | - A-P Bemelmans
- CEA, DSV, I(2)BM, Molecular Imaging Research Center (MIRCen) and CNRS, CEA URA 2210, 18, route du Panorama, 92265 Fontenay-aux-Roses, France.
| |
Collapse
|
204
|
Intrastriatal GDNF gene transfer by inducible lentivirus vectors protects dopaminergic neurons in a rat model of parkinsonism. Exp Neurol 2014; 261:87-96. [DOI: 10.1016/j.expneurol.2014.06.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 06/18/2014] [Accepted: 06/20/2014] [Indexed: 11/20/2022]
|
205
|
Hadaczek P, Wu G, Sharma N, Ciesielska A, Bankiewicz K, Davidow AL, Lu ZH, Forsayeth J, Ledeen RW. GDNF signaling implemented by GM1 ganglioside; failure in Parkinson's disease and GM1-deficient murine model. Exp Neurol 2014; 263:177-89. [PMID: 25448159 DOI: 10.1016/j.expneurol.2014.10.010] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 09/16/2014] [Accepted: 10/16/2014] [Indexed: 11/26/2022]
Abstract
GDNF is indispensible for adult catecholaminergic neuron survival, and failure of GDNF signaling has been linked to loss of dopaminergic neurons in Parkinson's disease (PD). This study demonstrates attenuated GDNF signaling in neurons deficient in ganglio-series gangliosides, and restoration of such signaling with LIGA20, a membrane permeable analog of GM1. GM1 is shown to associate in situ with GFRα1 and RET, the protein components of the GDNF receptor, this being necessary for assembly of the tripartite receptor complex. Mice wholly or partially deficient in GM1 due to disruption of the B4galnt1 gene developed PD symptoms based on behavioral and neuropathological criteria which were largely ameliorated by gene therapy with AAV2-GDNF and also with LIGA20 treatment. The nigral neurons of PD subjects that were severely deficient in GM1 showed subnormal levels of tyrosine phosphorylated RET. Also in PD brain, GM1 levels in the occipital cortex, a region of limited PD pathology, were significantly below age-matched controls, suggesting the possibility of systemic GM1 deficiency as a risk factor in PD. This would accord with our finding that mice with partial GM1 deficiency represent a faithful recapitulation of the human disease. Together with the previously demonstrated age-related decline of GM1 in human brain, this points to gradual development of subthreshold levels of GM1 in the brain of PD subjects below that required for effective GDNF signaling. This hypothesis offers a dramatically different explanation for the etiology of sporadic PD as a manifestation of acquired resistance to GDNF.
Collapse
Affiliation(s)
- Piotr Hadaczek
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94103-0555, USA
| | - Gusheng Wu
- Department of Neurology and Neurosciences MSB-H506, The State University of New Jersey, 185 South Orange Ave., Newark, NJ 07103, USA
| | - Nitasha Sharma
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94103-0555, USA
| | - Agnieszka Ciesielska
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94103-0555, USA
| | - Krystof Bankiewicz
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94103-0555, USA
| | - Amy L Davidow
- Department of Biostatistics/Epidemiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 South Orange Ave., Newark, NJ 07103, USA
| | - Zi-Hua Lu
- Department of Neurology and Neurosciences MSB-H506, The State University of New Jersey, 185 South Orange Ave., Newark, NJ 07103, USA
| | - John Forsayeth
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94103-0555, USA.
| | - Robert W Ledeen
- Department of Neurology and Neurosciences MSB-H506, The State University of New Jersey, 185 South Orange Ave., Newark, NJ 07103, USA.
| |
Collapse
|
206
|
dos Santos NAG, Martins NM, Silva RDB, Ferreira RS, Sisti FM, dos Santos AC. Caffeic acid phenethyl ester (CAPE) protects PC12 cells from MPP+ toxicity by inducing the expression of neuron-typical proteins. Neurotoxicology 2014; 45:131-8. [PMID: 25454720 DOI: 10.1016/j.neuro.2014.09.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 08/29/2014] [Accepted: 09/23/2014] [Indexed: 01/18/2023]
Abstract
Neurite loss is an early event in neurodegenerative diseases; therefore, the regeneration of the network of neurites constitutes an interesting strategy of treatment for such disorders. Neurotrophic factors play a critical role in neuronal regeneration, but their clinical use is limited by their inability to cross the blood brain barrier. Oxidative and inflammatory events are implicated in neurodegeneration and antioxidant compounds have been suggested as potential neuroprotectors. The protective potential of CAPE (caffeic acid phenethyl ester) has been shown in different models of neurotoxicity (in vitro and in vivo) and it has been associated with immune-modulatory, antioxidant and anti-inflammatory properties; however, other mechanisms might be involved. The present study demonstrates that CAPE protects PC12 cells from the cellular death induced by the dopaminergic neurotoxin MPP(+) by increasing the network of neurites. Results showed that CAPE induced the formation, elongation and ramification of neurites in PC12 cells non-stimulated with NGF (nerve growth factor) and inhibited the shortage of neurites induced by the dopaminergic neurotoxin. These effects were associated with increased expression of neuron-typical proteins responsible for axonal growth (GAP-43) and synaptogenesis (synaptophysin and synapsin I). It is noteworthy that, unlike neurotrophins, CAPE would be able to cross the blood brain barrier and exert its neurotrophic effects in the brain. This study corroborates the therapeutic potential of CAPE in neurodegenerative diseases while proposes the involvement of neuroplasticity in the mechanism of neuroprotection.
Collapse
Affiliation(s)
- Neife Aparecida Guinaim dos Santos
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| | - Nádia Maria Martins
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Roberto de Barros Silva
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Rafaela Scalco Ferreira
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Flávia Malvestio Sisti
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Antonio Cardozo dos Santos
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
207
|
Gervois P, Struys T, Hilkens P, Bronckaers A, Ratajczak J, Politis C, Brône B, Lambrichts I, Martens W. Neurogenic maturation of human dental pulp stem cells following neurosphere generation induces morphological and electrophysiological characteristics of functional neurons. Stem Cells Dev 2014; 24:296-311. [PMID: 25203005 DOI: 10.1089/scd.2014.0117] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Cell-based therapies are emerging as an alternative treatment option to promote functional recovery in patients suffering from neurological disorders, which are the major cause of death and permanent disability. The present study aimed to differentiate human dental pulp stem cells (hDPSCs) toward functionally active neuronal cells in vitro. hDPSCs were subjected to a two-step protocol. First, neuronal induction was acquired through the formation of neurospheres, followed by neuronal maturation, based on cAMP and neurotrophin-3 (NT-3) signaling. At the ultrastructural level, it was shown that the intra-spheral microenvironment promoted intercellular communication. hDPSCs grew out of the neurospheres in vitro and established a neurogenic differentiated hDPSC culture (d-hDPSCs) upon cAMP and NT-3 signaling. d-hDPSCs were characterized by the increased expression of neuronal markers such as neuronal nuclei, microtubule-associated protein 2, neural cell adhesion molecule, growth-associated protein 43, synapsin I, and synaptophysin compared with nondifferentiated hDPSCs. Enzyme-linked immunosorbent assay demonstrated that the secretion of brain-derived neurotrophic factor, vascular endothelial growth factor, and nerve growth factor differed between d-hDPSCs and hDPSCs. d-hDPSCs acquired neuronal features, including multiple intercommunicating cytoplasmic extensions and increased vesicular transport, as shown by the electron microscopic observation. Patch clamp analysis demonstrated the functional activity of d-hDPSCs by the presence of tetrodotoxin- and tetraethyl ammonium-sensitive voltage-gated sodium and potassium channels, respectively. A subset of d-hDPSCs was able to fire a single action potential. The results reported in this study demonstrate that hDPSCs are capable of neuronal commitment following neurosphere formation, characterized by distinct morphological and electrophysiological properties of functional neuronal cells.
Collapse
Affiliation(s)
- Pascal Gervois
- 1 Group of Morphology, Biomedical Research Institute, Hasselt University , Diepenbeek, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
208
|
Fu X, Gao H, Tian F, Gao J, Lou L, Liang Y, Ning Q, Luo X. Mechanistic effects of amino acids and glucose in a novel glutaric aciduria type 1 cell model. PLoS One 2014; 9:e110181. [PMID: 25333616 PMCID: PMC4198201 DOI: 10.1371/journal.pone.0110181] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Accepted: 09/05/2014] [Indexed: 11/19/2022] Open
Abstract
Acute neurological crises involving striatal degeneration induced by a deficiency of glutaryl-CoA dehydrogenase (GCDH) and the accumulation of glutaric (GA) and 3-hydroxyglutaric acid (3-OHGA) are considered to be the most striking features of glutaric aciduria type I (GA1). In the present study, we investigated the mechanisms of apoptosis and energy metabolism impairment in our novel GA1 neuronal model. We also explored the effects of appropriate amounts of amino acids (2 mM arginine, 2 mM homoarginine, 0.45 g/L tyrosine and 10 mM leucine) and 2 g/L glucose on these cells. Our results revealed that the novel GA1 neuronal model effectively simulates the hypermetabolic state of GA1. We found that leucine, tyrosine, arginine, homoarginine or glucose treatment of the GA1 model cells reduced the gene expression of caspase-3, caspase-8, caspase-9, bax, fos, and jun and restored the intracellular NADH and ATP levels. Tyrosine, arginine or homoarginine treatment in particular showed anti-apoptotic effects; increased α-ketoglutarate dehydrogenase complex (OGDC), fumarase (FH), and citrate synthase (CS) expression; and relieved the observed impairment in energy metabolism. To the best of our knowledge, this study is the first to investigate the protective mechanisms of amino acids and glucose in GA1 at the cellular level from the point of view of apoptosis and energy metabolism. Our data support the results of previous studies, indicating that supplementation of arginine and homoarginine as a dietary control strategy can have a therapeutic effect on GA1. All of these findings facilitate the understanding of cell apoptosis and energy metabolism impairment in GA1 and reveal new therapeutic perspectives for this disease.
Collapse
Affiliation(s)
- Xi Fu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongjie Gao
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengyan Tian
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinzhi Gao
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liping Lou
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Liang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qin Ning
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Luo
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- * E-mail:
| |
Collapse
|
209
|
Polinski NK, Gombash SE, Manfredsson FP, Lipton JW, Kemp CJ, Cole-Strauss A, Kanaan NM, Steece-Collier K, Kuhn NC, Wohlgenant SL, Sortwell CE. Recombinant adenoassociated virus 2/5-mediated gene transfer is reduced in the aged rat midbrain. Neurobiol Aging 2014; 36:1110-20. [PMID: 25457558 DOI: 10.1016/j.neurobiolaging.2014.07.047] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 07/28/2014] [Accepted: 07/30/2014] [Indexed: 12/12/2022]
Abstract
Clinical trials are examining the efficacy of viral vector-mediated gene delivery for treating Parkinson's disease. Although viral vector strategies have been successful in preclinical studies, to date clinical trials have disappointed. This may be because of the fact that preclinical studies fail to account for aging. Aging is the single greatest risk factor for developing Parkinson's disease and age alters cellular processes utilized by viral vectors. We hypothesized that the aged brain would be relatively resistant to transduction when compared with the young adult. We examined recombinant adeno-associated virus 2/5-mediated green fluorescent protein (rAAV2/5 GFP) expression in the young adult and aged rat nigrostriatal system. GFP overexpression was produced in both age groups. However, following rAAV2/5 GFP injection to the substantia nigra aged rats displayed 40%-60% less GFP protein in the striatum, regardless of rat strain or duration of expression. Furthermore, aged rats exhibited 40% fewer cells expressing GFP and 4-fold less GFP messenger RNA. rAAV2/5-mediated gene transfer is compromised in the aged rat midbrain, with deficiencies in early steps of transduction leading to significantly less messenger RNA and protein expression.
Collapse
Affiliation(s)
- Nicole K Polinski
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA; Neuroscience Program, Michigan State University, Grand Rapids, MI, USA
| | - Sara E Gombash
- Graduate Program in Neuroscience, University of Cincinnati, Cincinnati, OH, USA
| | - Fredric P Manfredsson
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Jack W Lipton
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Christopher J Kemp
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Allyson Cole-Strauss
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Nicholas M Kanaan
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Kathy Steece-Collier
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Nathan C Kuhn
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Susan L Wohlgenant
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Caryl E Sortwell
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA.
| |
Collapse
|
210
|
Xiang L, Ma L, He Y, Wei N, Gong P. Osteogenic differentiation of human periodontal ligament cells after transfection with recombinant lentiviral vector containing follicular dendritic cell secreted protein. J Periodontal Res 2014; 49:554-62. [PMID: 24138099 DOI: 10.1111/jre.12135] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2013] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND OBJECTIVE Follicular dendritic cell secreted protein (FDC-SP), has been identified in human periodontal ligament (PDL) in a recent study. It is suggested that the expression of FDC-SP might be associated with the osteogenic differentiation and mineralization of human periodontal ligament cells (hPDLCs). However, the intrinsic mechanism regarding this is still unclear. The aim of this study was to establish hPDLCs with safe and efficient overexpression of FDC-SP and to elucidate the influence of FDC-SP transfection on hPDLC osteogenesis in periodontal regeneration. MATERIAL AND METHODS We first applied a recombinant lentiviral vector containing FDC-SP to transfect hPDLCs via different multiplicity of infection (MOI) levels (1, 10, 20, 50 and 100). Western blot was performed to confirm the expression of FDC-SP. MTT assay was employed to evaluate the proliferation status of transfected cells. Then, the extent of osteogenic differentiation was investigated by simultaneous monitoring of alkaline phosphatase (ALP) activity assessment, immunofluorescent staining, the expression patterns of osteoblastic markers and mineralization staining. RESULTS We found that hPDLCs transfected via MOI 20, 50 and 100 exhibited expression of FDC-SP protein compared with MOI 1 and 10. There was no significant effect of FDC-SP transfection (at different MOI levels of 1, 10 and 20) on the proliferation of hPDLCs, whereas higher MOI levels (50 and 100) inhibited cell proliferation ability. In addition, ALP activity decreased significantly in FDC-SP-transfected hPDLCs at day 7. When stained with alizarin red, cells overexpressing FDC-SP formed less mineralized nodules at 21 d post-induction of differentiation, compared with the control cultures. Osteogenic inhibition was also confirmed by ALP immunostaining. Moreover, mRNA expression levels of osteoblastic markers decreased after FDC-SP transfection, which were in accordance with western blot results. CONCLUSION Our data suggest that MOI 20 is optimal to transfect hPDLCs, which achieves safe and efficient overexpression of FDC-SP in transfected cells. Moreover, FDC-SP overexpression inhibits osteogenic differentiation of hPDLCs. The present study contributes to a better understanding of the biological functions governing FDC-SP-induced hPDLC differentiation.
Collapse
Affiliation(s)
- L Xiang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Dental Implant Center, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | | | | | | | | |
Collapse
|
211
|
Abstract
The last decade has seen major progress at all levels of neuroscience, from genes and molecules up to integrated systems-level models of brain function. In particular, there have been advances in the understanding of cell-type-specific contributions to function, together with a clearer account of how these contributions are coordinated from moment to moment to organise behavior. A major current endeavor is to leverage this knowledge to develop new therapeutic approaches. In Parkinson's disease, there are a number of promising emerging treatments. Here, we will highlight three ambitious novel therapeutic approaches for this condition, each robustly driven by primary neuroscience. Pharmacogenetics genetically re-engineers neurons to produce neurotrophins that are neuroprotective to vulnerable dopaminergic cells or to directly replace dopamine through enzyme transduction. Deep brain stimulation (DBS) is undergoing a transformation, with adaptive DBS controlled by neural signals resulting in better motor outcomes and significant reductions in overall stimulation that could reduce side effects. Finally, optogenetics presents the opportunity to achieve cell-type-specific control with a high temporal specification on a large enough scale to effectively repair network-level dysfunction.
Collapse
Affiliation(s)
- S Little
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, OX3 9DU, UK
| | - P Brown
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, OX3 9DU, UK.
| |
Collapse
|
212
|
Qian S, Wang Q, Zuo Z. Improved brain uptake of peptide-based CNS drugs via alternative routes of administrations of its nanocarrier delivery systems: a promising strategy for CNS targeting delivery of peptides. Expert Opin Drug Metab Toxicol 2014; 10:1491-508. [DOI: 10.1517/17425255.2014.956080] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
213
|
Jang W, Park HH, Lee KY, Lee YJ, Kim HT, Koh SH. 1,25-dyhydroxyvitamin D3 Attenuates l-DOPA-Induced Neurotoxicity in Neural Stem Cells. Mol Neurobiol 2014; 51:558-70. [DOI: 10.1007/s12035-014-8835-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 07/25/2014] [Indexed: 11/27/2022]
|
214
|
Chan AWS. Progress and prospects for genetic modification of nonhuman primate models in biomedical research. ILAR J 2014; 54:211-23. [PMID: 24174443 DOI: 10.1093/ilar/ilt035] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The growing interest of modeling human diseases using genetically modified (transgenic) nonhuman primates (NHPs) is a direct result of NHPs (rhesus macaque, etc.) close relation to humans. NHPs share similar developmental paths with humans in their anatomy, physiology, genetics, and neural functions; and in their cognition, emotion, and social behavior. The NHP model within biomedical research has played an important role in the development of vaccines, assisted reproductive technologies, and new therapies for many diseases. Biomedical research has not been the primary role of NHPs. They have mainly been used for safety evaluation and pharmacokinetics studies, rather than determining therapeutic efficacy. The development of the first transgenic rhesus macaque (2001) revolutionized the role of NHP models in biomedicine. Development of the transgenic NHP model of Huntington's disease (2008), with distinctive clinical features, further suggested the uniqueness of the model system; and the potential role of the NHP model for human genetic disorders. Modeling human genetic diseases using NHPs will continue to thrive because of the latest advances in molecular, genetic, and embryo technologies. NHPs rising role in biomedical research, specifically pre-clinical studies, is foreseeable. The path toward the development of transgenic NHPs and the prospect of transgenic NHPs in their new role in future biomedicine needs to be reviewed. This article will focus on the advancement of transgenic NHPs in the past decade, including transgenic technologies and disease modeling. It will outline new technologies that may have significant impact in future NHP modeling and will conclude with a discussion of the future prospects of the transgenic NHP model.
Collapse
|
215
|
Kanaan NM, Manfredsson FP. Loss of functional alpha-synuclein: a toxic event in Parkinson's disease? JOURNAL OF PARKINSONS DISEASE 2014; 2:249-67. [PMID: 23938255 PMCID: PMC4736738 DOI: 10.3233/jpd-012138] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The discovery that alpha-synuclein (α-syn) is the primary component of the neuropathological hallmarks of Parkinson's disease (PD) and the identification of α-syn mutations in numerous inherited forms of PD has positioned α-syn at the top of the list of important factors in the pathogenesis of PD. Based on the pathological accumulation of α-syn in the brains of patients, the field is currently focused on therapeutic strategies that aim to reduce or eliminate α-syn. However, recent evidence suggests α-syn is a critical protein in neuron (i.e. dopamine neurons) survival and that maintaining a certain level of biologically functional α-syn is an important consideration in targeting α-syn for therapies. Despite the widespread interest in α-syn, the normal biological functions remain elusive, but a large body of work is focused on addressing this issue. In this review, we will discuss the current evidence related to α-syn function, α-syn folding and aggregation, and α-syn's role in disease. Finally, we will propose a relatively novel hypothesis on the pathogenesis of PD that hinges upon the premises that functional α-syn is critical to cell survival and that a reduction in biologically functional α-syn, whether through aggregation or reduced expression, may lead to the neurodegeneration in PD.
Collapse
Affiliation(s)
- Nicholas M Kanaan
- Department of Translational Science & Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | | |
Collapse
|
216
|
Potts LF, Wu H, Singh A, Marcilla I, Luquin MR, Papa SM. Modeling Parkinson's disease in monkeys for translational studies, a critical analysis. Exp Neurol 2014; 256:133-43. [PMID: 24070854 PMCID: PMC3962841 DOI: 10.1016/j.expneurol.2013.09.014] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 09/07/2013] [Accepted: 09/16/2013] [Indexed: 01/03/2023]
Abstract
The non-human primate MPTP model of Parkinson's disease is an essential tool for translational studies. However, the currently used methodologies to produce parkinsonian monkeys do not follow unified criteria, and the applied models may often fall short of reproducing the characteristics of patients in clinical trials. Pooling of data from the parkinsonian monkeys produced in our Centers provided the opportunity to evaluate thoroughly the behavioral outcomes that may be considered for appropriate modeling in preclinical studies. We reviewed records from 108 macaques including rhesus and cynomolgus species used to model moderate to advanced parkinsonism with systemic MPTP treatment. The attained motor disability and the development of levodopa-induced dyskinesias, as primary outcomes, and the occurrence of clinical complications and instability of symptoms were all analyzed for correlations with the parameters of MPTP administration and for estimation of sample sizes. Results showed that frequently the MPTP-treated macaque can recapitulate the phenotype of patients entering clinical trials, but to produce this model consistently it is important to adapt the MPTP exposure tightly according to individual animal responses. For studies of reduced animal numbers it is also important to produce stable models, and stability of parkinsonism in macaques critically depends on reaching "marked" motor disability. The analyzed data also led to put forward recommendations for successfully producing the primate MPTP model of Parkinson's disease for translational studies.
Collapse
Affiliation(s)
- Lisa F Potts
- Yerkes National Primate Research Center, Emory University School of Medicine, Neuroscience Building, Atlanta, GA 30329, USA
| | - Hao Wu
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Arun Singh
- Yerkes National Primate Research Center, Emory University School of Medicine, Neuroscience Building, Atlanta, GA 30329, USA
| | - Irene Marcilla
- Laboratory of Regenerative Therapy, Department of Neurology and Neuroscience Division, Centre for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Maria R Luquin
- Laboratory of Regenerative Therapy, Department of Neurology and Neuroscience Division, Centre for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Stella M Papa
- Yerkes National Primate Research Center, Emory University School of Medicine, Neuroscience Building, Atlanta, GA 30329, USA; Department of Neurology, Emory University School of Medicine, 6000 WMRC, 101 Woodruff Circle, Atlanta, GA 30322, USA.
| |
Collapse
|
217
|
Cass WA, Peters LE, Fletcher AM, Yurek DM. Calcitriol promotes augmented dopamine release in the lesioned striatum of 6-hydroxydopamine treated rats. Neurochem Res 2014; 39:1467-76. [PMID: 24858239 DOI: 10.1007/s11064-014-1331-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 05/06/2014] [Accepted: 05/09/2014] [Indexed: 10/25/2022]
Abstract
Current therapies for Parkinson's disease (PD) offer symptomatic relief but do not provide a cure or slow the disease process. Treatments that could halt progression of the disease or help restore function to damaged neurons would be of substantial benefit. Calcitriol, the active metabolite of vitamin D, has been shown to have significant effects on the brain. These effects include upregulating trophic factor levels, and reducing the severity of some central nervous system lesions. While previous studies have shown that calcitriol can be neuroprotective in 6-hydroxydopamine (6-OHDA) rodent models of PD, the present experiments were designed to examine the ability of calcitriol to promote restoration of extracellular dopamine (DA) levels and tissue content of DA in animals previously lesioned with 6-OHDA. Male Fischer-344 rats were given a single injection of 12 µg 6-OHDA into the right striatum. Four weeks later the animals were administered vehicle or calcitriol (0.3 or 1.0 µg/kg, s.c.) once a day for eight consecutive days. Three weeks after the calcitriol treatments in vivo microdialysis experiments were conducted to measure potassium and amphetamine evoked overflow of DA from both the left and right striata. In control animals treated with 6-OHDA and vehicle there were significant reductions in both potassium and amphetamine evoked overflow of DA on the lesioned side of the brain compared to the contralateral side. In animals treated with 6-OHDA followed by calcitriol there was significantly greater potassium and amphetamine evoked overflow of DA from the lesioned striatum compared to that from the control animals. The calcitriol treatments also led to increases in postmortem tissue levels of DA in the striatum and substantia nigra. These results suggest that calcitriol may help promote recovery of dopaminergic functioning in injured nigrostriatal neurons.
Collapse
Affiliation(s)
- Wayne A Cass
- Department of Anatomy and Neurobiology, MN-225 Chandler Medical Center, University of Kentucky College of Medicine, Lexington, KY, 40536-0298, USA,
| | | | | | | |
Collapse
|
218
|
Tarazi FI, Sahli ZT, Wolny M, Mousa SA. Emerging therapies for Parkinson's disease: from bench to bedside. Pharmacol Ther 2014; 144:123-33. [PMID: 24854598 DOI: 10.1016/j.pharmthera.2014.05.010] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 05/01/2014] [Indexed: 02/08/2023]
Abstract
The prevalence of Parkinson's disease (PD) increases with age and is projected to increase in parallel to the rising average age of the population. The disease can have significant health-related, social, and financial implications not only for the patient and the caregiver, but for the health care system as well. While the neuropathology of this neurodegenerative disorder is fairly well understood, its etiology remains a mystery, making it difficult to target therapy. The currently available drugs for treatment provide only symptomatic relief and do not control or prevent disease progression, and as a result patient compliance and satisfaction are low. Several emerging pharmacotherapies for PD are in different stages of clinical development. These therapies include adenosine A2A receptor antagonists, glutamate receptor antagonists, monoamine oxidase inhibitors, anti-apoptotic agents, and antioxidants such as coenzyme Q10, N-acetyl cysteine, and edaravone. Other emerging non-pharmacotherapies include viral vector gene therapy, microRNAs, transglutaminases, RTP801, stem cells and glial derived neurotrophic factor (GDNF). In addition, surgical procedures including deep brain stimulation, pallidotomy, thalamotomy and gamma knife surgery have emerged as alternative interventions for advanced PD patients who have completely utilized standard treatments and still suffer from persistent motor fluctuations. While several of these therapies hold much promise in delaying the onset of the disease and slowing its progression, more pharmacotherapies and surgical interventions need to be investigated in different stages of PD. It is hoped that these emerging therapies and surgical procedures will strengthen our clinical armamentarium for improved treatment of PD.
Collapse
Affiliation(s)
- F I Tarazi
- Department of Psychiatry and Neuroscience Program, Harvard Medical School, McLean Hospital, Belmont, MA 02478, USA.
| | - Z T Sahli
- Department of Psychiatry and Neuroscience Program, Harvard Medical School, McLean Hospital, Belmont, MA 02478, USA; School of Medicine, American University of Beirut, Beirut, Lebanon
| | - M Wolny
- The Pharmaceutical Research Institute at Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA
| | - S A Mousa
- The Pharmaceutical Research Institute at Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA
| |
Collapse
|
219
|
Neurotrophic and neuroprotective efficacy of intranasal GDNF in a rat model of Parkinson's disease. Neuroscience 2014; 274:11-23. [PMID: 24845869 DOI: 10.1016/j.neuroscience.2014.05.019] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 05/05/2014] [Accepted: 05/10/2014] [Indexed: 01/27/2023]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) exerts neurotrophic and neuroprotective effects on substantia nigra (SN) dopamine neurons and has great therapeutic potential for Parkinson's disease (PD). Hindering this potential is the fact that GDNF cannot cross the blood-brain barrier. The aim of this study was to assess the effects of GDNF administered by the intranasal route in normal rats, and in the unilateral 6-hydroxydopamine (6-OHDA) model of PD. In the first study, rats received single intranasal doses of 50-μg GDNF in phosphate-buffered saline (PBS) or cationic liposomes, but no 6-OHDA. In the second study, rats were nasally administered 10, 50 or 150 μg of GDNF in PBS or cationic liposomes 1h before injection of 6-OHDA. All groups were sacrificed 3-4 weeks later. Both intranasal GDNF treatments induced a neurotrophic effect in the SN insofar as the number of tyrosine hydroxylase (TH)-positive neurons was significantly higher than in controls given intranasal PBS liposomes. Dopamine cell counts were also higher in the intact SN of 6-OHDA-lesioned rats compared to controls given PBS liposomes. Most importantly, intranasal GDNF provided significant neuroprotective efficacy indicated by greater TH immunostaining density in the lesioned versus intact SN of rats given single 50-μg doses of GDNF in PBS, or 150-μg doses of liposomal GDNF, compared to lesioned rats given PBS liposomes. Three 50-μg doses given at daily intervals (1 day before, 1h before, and 1 day after 6-OHDA) provided even greater protection than single 150-μg doses. Multiple doses at short intervals may therefore provide greater neuroprotection than single bolus doses. These results demonstrate both a neurotrophic effect of intranasal GDNF in the intact SN as well as neuroprotective efficacy in the unilateral 6-OHDA model, supporting pursuit of this approach as a potential treatment for PD.
Collapse
|
220
|
Navarro-Yepes J, Zavala-Flores L, Anandhan A, Wang F, Skotak M, Chandra N, Li M, Pappa A, Martinez-Fong D, Del Razo LM, Quintanilla-Vega B, Franco R. Antioxidant gene therapy against neuronal cell death. Pharmacol Ther 2014; 142:206-30. [PMID: 24333264 PMCID: PMC3959583 DOI: 10.1016/j.pharmthera.2013.12.007] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Accepted: 11/26/2013] [Indexed: 12/21/2022]
Abstract
Oxidative stress is a common hallmark of neuronal cell death associated with neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, as well as brain stroke/ischemia and traumatic brain injury. Increased accumulation of reactive species of both oxygen (ROS) and nitrogen (RNS) has been implicated in mitochondrial dysfunction, energy impairment, alterations in metal homeostasis and accumulation of aggregated proteins observed in neurodegenerative disorders, which lead to the activation/modulation of cell death mechanisms that include apoptotic, necrotic and autophagic pathways. Thus, the design of novel antioxidant strategies to selectively target oxidative stress and redox imbalance might represent important therapeutic approaches against neurological disorders. This work reviews the evidence demonstrating the ability of genetically encoded antioxidant systems to selectively counteract neuronal cell loss in neurodegenerative diseases and ischemic brain damage. Because gene therapy approaches to treat inherited and acquired disorders offer many unique advantages over conventional therapeutic approaches, we discussed basic research/clinical evidence and the potential of virus-mediated gene delivery techniques for antioxidant gene therapy.
Collapse
Affiliation(s)
- Juliana Navarro-Yepes
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; Department of Toxicology, CINVESTAV-IPN, Mexico City, Mexico
| | - Laura Zavala-Flores
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Annadurai Anandhan
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Fang Wang
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Maciej Skotak
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Namas Chandra
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Ming Li
- Department of Psychology, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Aglaia Pappa
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus, Dragana, Alexandroupolis, Greece
| | - Daniel Martinez-Fong
- Department of Physiology, Biophysics and Neurosciences, CINVESTAV-IPN, Mexico City, Mexico
| | | | | | - Rodrigo Franco
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States.
| |
Collapse
|
221
|
Chen X, Tang Y, Zhang Y, Zhuo M, Tang Z, Yu Y, Zang G. Tapasin modification on the intracellular epitope HBcAg18-27 enhances HBV-specific CTL immune response and inhibits hepatitis B virus replication in vivo. J Transl Med 2014; 94:478-90. [PMID: 24614195 DOI: 10.1038/labinvest.2014.6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 11/23/2013] [Accepted: 12/23/2013] [Indexed: 01/16/2023] Open
Abstract
HBV-specific cytotoxic T-lymphocyte (CTL) activity has a very important role in hepatitis B virus clearance. Present studies suggest that Tapasin, a endoplasmic reticulum (ER) chaperone, stabilizes the peptide-receptive MHC I conformation, allowing peptide exchange and increasing more peptides to be translocated into the ER. We have previously testified that cytoplasmic transduction peptide (CTP)-HBcAg(18-27)-Tapasin fusion protein could enter cytoplasm of dendritic cells, and enhance T cells' response to generate specific CTLs efficiently in vitro. In the present study, we evaluated specific immune responses of CTP-HBcAg(18-27)-Tapasin fusion protein in HLA-A2 transgenic mice (H-2K(b)) and anti-viral ability in HBV transgenic mice, and explored the mechanisms probably involved in. The studies showed that CTP-HBcAg(18-27)-Tapasin not only increased production of cytokine IFN-γ and interleukin-2 (IL-2), compared with CTP-HBcAg(18-27), HBcAg(18-27)-Tapasin, and PBS, but also significantly induced the higher percentages of IFN-γ+CD8(+) T cells and specific CTL responses in HLA-A2 transgenic mice. Moreover, enhancement of specific CTL activity induced by the fusion protein reduced HBV DNA and hepatitis B surface antigen (HBsAg) levels and decreased the expression of HBsAg and hepatitis B core antigen (HBcAg) in liver tissue of HBV transgenic mice. In addition, CTP-HBcAg(18-27)-Tapasin could upregulate the expression of JAK2, Tyk2, STAT1, and STAT4 in T lymphocytes in HLA-A2 transgenic mice splenocytes. However, there was no significant difference on the expressions of JAK1, JAK3, and STAT6 between each group. In conclusion, CTP-HBcAg(18-27)-Tapasin fusion protein could enhance not only the percentages of CTLs but also induce robust specific CTL activity and inhibits hepatitis B virus replication in vivo, which was associated with activation of the JAK/STAT signaling pathway.
Collapse
Affiliation(s)
- Xiaohua Chen
- Department of Infectious Disease, Shanghai JiaoTong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yuyan Tang
- Department of Infectious Disease, Shanghai JiaoTong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yi Zhang
- Department of Infectious Disease, Shanghai JiaoTong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Meng Zhuo
- Department of Infectious Disease, Shanghai JiaoTong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Zhenghao Tang
- Department of Infectious Disease, Shanghai JiaoTong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yongsheng Yu
- Department of Infectious Disease, Shanghai JiaoTong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Guoqing Zang
- Department of Infectious Disease, Shanghai JiaoTong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
222
|
Wakeman DR, Redmond DE, Dodiya HB, Sladek JR, Leranth C, Teng YD, Samulski RJ, Snyder EY. Human neural stem cells survive long term in the midbrain of dopamine-depleted monkeys after GDNF overexpression and project neurites toward an appropriate target. Stem Cells Transl Med 2014; 3:692-701. [PMID: 24744393 DOI: 10.5966/sctm.2013-0208] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Transplanted multipotent human fetal neural stem cells (hfNSCs) significantly improved the function of parkinsonian monkeys in a prior study primarily by neuroprotection, with only 3%-5% of cells expressing a dopamine (DA) phenotype. In this paper, we sought to determine whether further manipulation of the neural microenvironment by overexpression of a developmentally critical molecule, glial cell-derived neurotrophic factor (GDNF), in the host striatum could enhance DA differentiation of hfNSCs injected into the substantia nigra and elicit growth of their axons to the GDNF-expressing target. hfNSCs were transplanted into the midbrain of 10 green monkeys exposed to 1-methyl-4-phenyl-1,2,3,6-tetrahydro-pyridine. GDNF was delivered concomitantly to the striatum via an adeno-associated virus serotype 5 vector, and the fate of grafted cells was assessed after 11 months. Donor cells remained predominantly within the midbrain at the injection site and sprouted numerous neurofilament-immunoreactive fibers that appeared to course rostrally toward the striatum in parallel with tyrosine hydroxylase-immunoreactive fibers from the host substantia nigra but did not mature into DA neurons. This work suggests that hfNSCs can generate neurons that project long fibers in the adult primate brain. However, in the absence of region-specific signals and despite GDNF overexpression, hfNSCs did not differentiate into mature DA neurons in large numbers. It is encouraging, however, that the adult primate brain appeared to retain axonal guidance cues. We believe that transplantation of stem cells, specifically instructed ex vivo to yield DA neurons, could lead to reconstruction of some portion of the nigrostriatal pathway and prove beneficial for the parkinsonian condition.
Collapse
Affiliation(s)
- Dustin R Wakeman
- Graduate Program in Biomedical Sciences, University of California at San Diego, La Jolla, California, USA; Program in Stem Cell and Regenerative Biology, Sanford-Burnham Medical Research Institute, La Jolla, California, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA; Departments of Psychiatry, Neurosurgery, and Obstetrics and Gynecology, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, University of Colorado School of Medicine, Denver, Colorado, USA; Department of Neurosurgery and Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA; Gene Therapy Center and Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - D Eugene Redmond
- Graduate Program in Biomedical Sciences, University of California at San Diego, La Jolla, California, USA; Program in Stem Cell and Regenerative Biology, Sanford-Burnham Medical Research Institute, La Jolla, California, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA; Departments of Psychiatry, Neurosurgery, and Obstetrics and Gynecology, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, University of Colorado School of Medicine, Denver, Colorado, USA; Department of Neurosurgery and Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA; Gene Therapy Center and Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Hemraj B Dodiya
- Graduate Program in Biomedical Sciences, University of California at San Diego, La Jolla, California, USA; Program in Stem Cell and Regenerative Biology, Sanford-Burnham Medical Research Institute, La Jolla, California, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA; Departments of Psychiatry, Neurosurgery, and Obstetrics and Gynecology, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, University of Colorado School of Medicine, Denver, Colorado, USA; Department of Neurosurgery and Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA; Gene Therapy Center and Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - John R Sladek
- Graduate Program in Biomedical Sciences, University of California at San Diego, La Jolla, California, USA; Program in Stem Cell and Regenerative Biology, Sanford-Burnham Medical Research Institute, La Jolla, California, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA; Departments of Psychiatry, Neurosurgery, and Obstetrics and Gynecology, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, University of Colorado School of Medicine, Denver, Colorado, USA; Department of Neurosurgery and Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA; Gene Therapy Center and Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Csaba Leranth
- Graduate Program in Biomedical Sciences, University of California at San Diego, La Jolla, California, USA; Program in Stem Cell and Regenerative Biology, Sanford-Burnham Medical Research Institute, La Jolla, California, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA; Departments of Psychiatry, Neurosurgery, and Obstetrics and Gynecology, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, University of Colorado School of Medicine, Denver, Colorado, USA; Department of Neurosurgery and Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA; Gene Therapy Center and Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Yang D Teng
- Graduate Program in Biomedical Sciences, University of California at San Diego, La Jolla, California, USA; Program in Stem Cell and Regenerative Biology, Sanford-Burnham Medical Research Institute, La Jolla, California, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA; Departments of Psychiatry, Neurosurgery, and Obstetrics and Gynecology, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, University of Colorado School of Medicine, Denver, Colorado, USA; Department of Neurosurgery and Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA; Gene Therapy Center and Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - R Jude Samulski
- Graduate Program in Biomedical Sciences, University of California at San Diego, La Jolla, California, USA; Program in Stem Cell and Regenerative Biology, Sanford-Burnham Medical Research Institute, La Jolla, California, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA; Departments of Psychiatry, Neurosurgery, and Obstetrics and Gynecology, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, University of Colorado School of Medicine, Denver, Colorado, USA; Department of Neurosurgery and Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA; Gene Therapy Center and Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Evan Y Snyder
- Graduate Program in Biomedical Sciences, University of California at San Diego, La Jolla, California, USA; Program in Stem Cell and Regenerative Biology, Sanford-Burnham Medical Research Institute, La Jolla, California, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA; Departments of Psychiatry, Neurosurgery, and Obstetrics and Gynecology, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, University of Colorado School of Medicine, Denver, Colorado, USA; Department of Neurosurgery and Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA; Gene Therapy Center and Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
223
|
Löw K, Aebischer P, Schneider BL. Direct and retrograde transduction of nigral neurons with AAV6, 8, and 9 and intraneuronal persistence of viral particles. Hum Gene Ther 2014; 24:613-29. [PMID: 23600720 DOI: 10.1089/hum.2012.174] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Recombinant adeno-associated viral (AAV) vectors of serotypes 6, 8, and 9 were characterized as tools for gene delivery to dopaminergic neurons in the substantia nigra for future gene therapeutic applications in Parkinson's disease. While vectors of all three serotypes transduced nigral dopaminergic neurons with equal efficiency when directly injected to the substantia nigra, AAV6 was clearly superior to AAV8 and AAV9 for retrograde transduction of nigral neurons after striatal delivery. For sequential transduction of nigral dopaminergic neurons, the combination of AAV9 with AAV6 proved to be more powerful than AAV8 with AAV6 or repeated AAV6 administration. Surprisingly, single-stranded viral genomes persisted in nigral dopaminergic neurons within cell bodies and axon terminals in the striatum, and intact assembled AAV capsid was enriched in nuclei of nigral neurons, 4 weeks after virus injections to the substantia nigra. 6-Hydroxydopamine (6-OHDA)-induced degeneration of dopaminergic neurons in the substantia nigra reduced the number of viral genomes in the striatum, in line with viral genome persistence in axon terminals. However, 6-OHDA-induced axonal degeneration did not induce any transsynaptic spread of AAV infection in the striatum. Therefore, the potential presence of viral particles in axons may not represent an important safety issue for AAV gene therapy applications in neurodegenerative diseases.
Collapse
Affiliation(s)
- Karin Löw
- Swiss Federal Institute of Technology Lausanne, CH-1015 Lausanne, Switzerland.
| | | | | |
Collapse
|
224
|
Chermenina M, Schouten P, Nevalainen N, Johansson F, Orädd G, Strömberg I. GDNF is important for striatal organization and maintenance of dopamine neurons grown in the presence of the striatum. Neuroscience 2014; 270:1-11. [PMID: 24726488 DOI: 10.1016/j.neuroscience.2014.04.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 04/02/2014] [Accepted: 04/02/2014] [Indexed: 11/17/2022]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) exerts neuroprotective and neurorestorative effects on neurons and GDNF plays a significant role in maintenance of the dopamine neurons utilizing grafting to create a nigrostriatal microcircuit of Gdnf knockout (Gdnf(-/-)) tissue. To further evaluate the role of GDNF on organization of the nigrostriatal system, single or double grafts of ventral mesencephalon (VM) and lateral ganglionic eminence (LGE) with mismatches in Gdnf genotypes were performed. The survival of single grafts was monitored utilizing magnetic resonance imaging (MRI) and cell survival and graft organization were evaluated with immunohistochemistry. The results revealed that the size of VM single grafts did not change over time independent of genotype, while the size of the LGE transplants was significantly reduced already at 2 weeks postgrafting when lacking GDNF. Lack of GDNF did not significantly affect the survival of tyrosine hydroxylase (TH)-positive neurons in single VM grafts. However, the survival of TH-positive neurons was significantly reduced in VM derived from Gdnf(+/+) when co-grafted with LGE from the Gdnf(-/-) tissue. In contrast, lack of GDNF in the VM portion of co-grafts had no effect on the survival of TH-positive neurons when co-grafted with LGE from Gdnf(+/+) mice. The TH-positive innervation of co-grafts was sparse when the striatal co-grafts were derived from the Gdnf(-/-) tissue while dense and patchy when innervating LGE producing GDNF. The TH-positive innervation overlapped with the organization of dopamine and cyclic AMP-regulated phosphoprotein-relative molecular mass 32,000 (DARPP-32)-positive neurons, that was disorganized in LGE lacking GDNF production. In conclusion, GDNF is important for a proper striatal organization and for survival of TH-positive neurons in the presence of the striatal tissue.
Collapse
Affiliation(s)
- M Chermenina
- Integrative Medical Biology, Umeå University, 901 87 Umeå, Sweden
| | - P Schouten
- Integrative Medical Biology, Umeå University, 901 87 Umeå, Sweden
| | - N Nevalainen
- Integrative Medical Biology, Umeå University, 901 87 Umeå, Sweden
| | - F Johansson
- Integrative Medical Biology, Umeå University, 901 87 Umeå, Sweden
| | - G Orädd
- Integrative Medical Biology, Umeå University, 901 87 Umeå, Sweden
| | - I Strömberg
- Integrative Medical Biology, Umeå University, 901 87 Umeå, Sweden.
| |
Collapse
|
225
|
Wickersham IR, Sullivan HA, Seung HS. Axonal and subcellular labelling using modified rabies viral vectors. Nat Commun 2014; 4:2332. [PMID: 23945836 PMCID: PMC5939574 DOI: 10.1038/ncomms3332] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 07/19/2013] [Indexed: 01/03/2023] Open
Abstract
An important aspect of any neural circuit is the placement of its output synapses, at levels ranging from macroscopic to subcellular. The many new molecular tools for locating and manipulating synapses are limited by the viral vectors available for delivering them. Adeno-associated viruses are the best current means of labeling and manipulating axons and synapses, but they have never expressed more than one transgene highly enough to label fine axonal structure while also labeling or perturbing synapses. Their slow expression also makes them incompatible with retrograde and transsynaptic vectors, preventing powerful combinatorial experiments. Here we show that deletion-mutant rabies virus can be specifically targeted to cells local to an injection site, brightly labeling axons even when coexpressing two other transgenes. We demonstrate several novel capabilities: simultaneously labeling axons and presynaptic terminals, labeling both dendrites and postsynaptic densities, and simultaneously labeling a region’s inputs and outputs using coinjected vectors.
Collapse
Affiliation(s)
- Ian R Wickersham
- Department of Brain & Cognitive Sciences, Massachusetts Institute of Technology, 43 Vassar Street, Cambridge, Massachusetts 02139, USA
| | | | | |
Collapse
|
226
|
Buttery PC, Barker RA. Treating Parkinson's disease in the 21st century: can stem cell transplantation compete? J Comp Neurol 2014; 522:2802-16. [PMID: 24610597 PMCID: PMC4233918 DOI: 10.1002/cne.23577] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Revised: 08/07/2013] [Accepted: 10/08/2013] [Indexed: 12/25/2022]
Abstract
The characteristic and selective degeneration of a unique population of cells—the nigrostriatal dopamine (DA) neurons—that occurs in Parkinson’s disease (PD) has made the condition an iconic target for cell replacement therapies. Indeed, transplantation of fetal ventral mesencephalic cells into the DA-deficient striatum was first trialled nearly 30 years ago, at a time when other treatments for the disease were less well developed. Over recent decades standard treatments for PD have advanced, and newer biological therapies are now emerging. In the 21st century, stem cell technology will have to compete alongside other sophisticated treatments, including deep brain stimulation and gene therapies. In this review we examine how stem cell–based transplantation therapies compare with these novel and emerging treatments in the management of this common condition. J. Comp. Neurol. 522:2802–2816, 2014.
Collapse
Affiliation(s)
- Philip C Buttery
- John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
| | | |
Collapse
|
227
|
Emerich DF, Orive G, Thanos C, Tornoe J, Wahlberg LU. Encapsulated cell therapy for neurodegenerative diseases: from promise to product. Adv Drug Deliv Rev 2014; 67-68:131-41. [PMID: 23880505 DOI: 10.1016/j.addr.2013.07.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 05/31/2013] [Accepted: 07/12/2013] [Indexed: 12/27/2022]
Abstract
Delivering therapeutic molecules, including trophic factor proteins, across the blood brain barrier to the brain parenchyma to treat chronic neurodegenerative diseases remains one of the great challenges in biology. To be effective, delivery needs to occur in a long-term and stable manner at sufficient quantities directly to the target region in a manner that is selective but yet covers enough of the target site to be efficacious. One promising approach uses cellular implants that produce and deliver therapeutic molecules directly to the brain region of interest. Implanted cells can be precisely positioned into the desired region and can be protected from host immunological attack by encapsulating them and by surrounding them within an immunoisolatory, semipermeable capsule. In this approach, cells are enclosed within a semiporous capsule with a perm selective membrane barrier that admits oxygen and required nutrients and releases bioactive cell secretions while restricting passage of larger cytotoxic agents from the host immune defense system. Recent advances in human cell line development have increased the levels of secreted therapeutic molecules from encapsulated cells, and membrane extrusion techniques have led to the first ever clinical demonstrations of long-term survival and function of encapsulated cells in the brain parenchyma. As such, cell encapsulation is capable of providing a targeted, continuous, de novo synthesized source of very high levels of therapeutic molecules that can be distributed over significant portions of the brain.
Collapse
|
228
|
Palfi S, Gurruchaga JM, Ralph GS, Lepetit H, Lavisse S, Buttery PC, Watts C, Miskin J, Kelleher M, Deeley S, Iwamuro H, Lefaucheur JP, Thiriez C, Fenelon G, Lucas C, Brugières P, Gabriel I, Abhay K, Drouot X, Tani N, Kas A, Ghaleh B, Le Corvoisier P, Dolphin P, Breen DP, Mason S, Guzman NV, Mazarakis ND, Radcliffe PA, Harrop R, Kingsman SM, Rascol O, Naylor S, Barker RA, Hantraye P, Remy P, Cesaro P, Mitrophanous KA. Long-term safety and tolerability of ProSavin, a lentiviral vector-based gene therapy for Parkinson's disease: a dose escalation, open-label, phase 1/2 trial. Lancet 2014; 383:1138-46. [PMID: 24412048 DOI: 10.1016/s0140-6736(13)61939-x] [Citation(s) in RCA: 310] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Parkinson's disease is typically treated with oral dopamine replacement therapies; however, long-term treatment leads to motor complications and, occasionally, impulse control disorders caused by intermittent stimulation of dopamine receptors and off-target effects, respectively. We aimed to assess the safety, tolerability, and efficacy of bilateral, intrastriatal delivery of ProSavin, a lentiviral vector-based gene therapy aimed at restoring local and continuous dopamine production in patients with advanced Parkinson's disease. METHODS We undertook a phase 1/2 open-label trial with 12-month follow-up at two study sites (France and UK) to assess the safety and efficacy of ProSavin after bilateral injection into the putamen of patients with Parkinson's disease. All patients were then enrolled in a separate open-label follow-up study of long-term safety. Three doses were assessed in separate cohorts: low dose (1·9×10(7) transducing units [TU]); mid dose (4·0×10(7) TU); and high dose (1×10(8) TU). Inclusion criteria were age 48-65 years, disease duration 5 years or longer, motor fluctuations, and 50% or higher motor response to oral dopaminergic therapy. The primary endpoints of the phase 1/2 study were the number and severity of adverse events associated with ProSavin and motor responses as assessed with Unified Parkinson's Disease Rating Scale (UPDRS) part III (off medication) scores, at 6 months after vector administration. Both trials are registered at ClinicalTrials.gov, NCT00627588 and NCT01856439. FINDINGS 15 patients received ProSavin and were followed up (three at low dose, six mid dose, six high dose). During the first 12 months of follow-up, 54 drug-related adverse events were reported (51 mild, three moderate). Most common were increased on-medication dyskinesias (20 events, 11 patients) and on-off phenomena (12 events, nine patients). No serious adverse events related to the study drug or surgical procedure were reported. A significant improvement in mean UPDRS part III motor scores off medication was recorded in all patients at 6 months (mean score 38 [SD 9] vs 26 [8], n=15, p=0·0001) and 12 months (38 vs 27 [8]; n=15, p=0·0001) compared with baseline. INTERPRETATION ProSavin was safe and well tolerated in patients with advanced Parkinson's disease. Improvement in motor behaviour was observed in all patients. FUNDING Oxford BioMedica.
Collapse
Affiliation(s)
- Stéphane Palfi
- AP-HP, Groupe Hospitalier Henri-Mondor, DHU PePsy, UF Neurochirurgie Fonctionnelle, Neurologie, Neurophysiologie, Anesthésie, Centre d'Investigation Clinique 006, Plateforme de Ressources Biologiques, Créteil, France; Université Paris 12, Faculté de Médecine, Créteil, France.
| | - Jean Marc Gurruchaga
- AP-HP, Groupe Hospitalier Henri-Mondor, DHU PePsy, UF Neurochirurgie Fonctionnelle, Neurologie, Neurophysiologie, Anesthésie, Centre d'Investigation Clinique 006, Plateforme de Ressources Biologiques, Créteil, France; Université Paris 12, Faculté de Médecine, Créteil, France
| | | | - Helene Lepetit
- AP-HP, Groupe Hospitalier Henri-Mondor, DHU PePsy, UF Neurochirurgie Fonctionnelle, Neurologie, Neurophysiologie, Anesthésie, Centre d'Investigation Clinique 006, Plateforme de Ressources Biologiques, Créteil, France; Université Paris 12, Faculté de Médecine, Créteil, France
| | - Sonia Lavisse
- CEA, DSV I(2)BM, MIRCen and CNRS URA2210, Fontenay-aux-Roses, France
| | - Philip C Buttery
- John van Geest Centre for Brain Repair and Addenbrooke's Hospital, Cambridge, UK
| | - Colin Watts
- John van Geest Centre for Brain Repair and Addenbrooke's Hospital, Cambridge, UK
| | | | | | | | - Hirokazu Iwamuro
- AP-HP, Groupe Hospitalier Henri-Mondor, DHU PePsy, UF Neurochirurgie Fonctionnelle, Neurologie, Neurophysiologie, Anesthésie, Centre d'Investigation Clinique 006, Plateforme de Ressources Biologiques, Créteil, France; Université Paris 12, Faculté de Médecine, Créteil, France
| | - Jean Pascal Lefaucheur
- AP-HP, Groupe Hospitalier Henri-Mondor, DHU PePsy, UF Neurochirurgie Fonctionnelle, Neurologie, Neurophysiologie, Anesthésie, Centre d'Investigation Clinique 006, Plateforme de Ressources Biologiques, Créteil, France; Université Paris 12, Faculté de Médecine, Créteil, France
| | - Claire Thiriez
- AP-HP, Groupe Hospitalier Henri-Mondor, DHU PePsy, UF Neurochirurgie Fonctionnelle, Neurologie, Neurophysiologie, Anesthésie, Centre d'Investigation Clinique 006, Plateforme de Ressources Biologiques, Créteil, France; Université Paris 12, Faculté de Médecine, Créteil, France
| | - Gilles Fenelon
- AP-HP, Groupe Hospitalier Henri-Mondor, DHU PePsy, UF Neurochirurgie Fonctionnelle, Neurologie, Neurophysiologie, Anesthésie, Centre d'Investigation Clinique 006, Plateforme de Ressources Biologiques, Créteil, France; Université Paris 12, Faculté de Médecine, Créteil, France; INSERM U955, E01, Institut de Recherche Biomédicale, Créteil, France
| | | | - Pierre Brugières
- AP-HP, Groupe Hospitalier Henri-Mondor, DHU PePsy, UF Neurochirurgie Fonctionnelle, Neurologie, Neurophysiologie, Anesthésie, Centre d'Investigation Clinique 006, Plateforme de Ressources Biologiques, Créteil, France; Université Paris 12, Faculté de Médecine, Créteil, France
| | - Inanna Gabriel
- AP-HP, Groupe Hospitalier Henri-Mondor, DHU PePsy, UF Neurochirurgie Fonctionnelle, Neurologie, Neurophysiologie, Anesthésie, Centre d'Investigation Clinique 006, Plateforme de Ressources Biologiques, Créteil, France; Université Paris 12, Faculté de Médecine, Créteil, France
| | - Kou Abhay
- AP-HP, Groupe Hospitalier Henri-Mondor, DHU PePsy, UF Neurochirurgie Fonctionnelle, Neurologie, Neurophysiologie, Anesthésie, Centre d'Investigation Clinique 006, Plateforme de Ressources Biologiques, Créteil, France; Université Paris 12, Faculté de Médecine, Créteil, France
| | - Xavier Drouot
- AP-HP, Groupe Hospitalier Henri-Mondor, DHU PePsy, UF Neurochirurgie Fonctionnelle, Neurologie, Neurophysiologie, Anesthésie, Centre d'Investigation Clinique 006, Plateforme de Ressources Biologiques, Créteil, France; Université Paris 12, Faculté de Médecine, Créteil, France
| | - Naoki Tani
- AP-HP, Groupe Hospitalier Henri-Mondor, DHU PePsy, UF Neurochirurgie Fonctionnelle, Neurologie, Neurophysiologie, Anesthésie, Centre d'Investigation Clinique 006, Plateforme de Ressources Biologiques, Créteil, France; Université Paris 12, Faculté de Médecine, Créteil, France
| | - Aurelie Kas
- CEA, DSV I(2)BM, MIRCen and CNRS URA2210, Fontenay-aux-Roses, France
| | - Bijan Ghaleh
- AP-HP, Groupe Hospitalier Henri-Mondor, DHU PePsy, UF Neurochirurgie Fonctionnelle, Neurologie, Neurophysiologie, Anesthésie, Centre d'Investigation Clinique 006, Plateforme de Ressources Biologiques, Créteil, France; Université Paris 12, Faculté de Médecine, Créteil, France
| | - Philippe Le Corvoisier
- AP-HP, Groupe Hospitalier Henri-Mondor, DHU PePsy, UF Neurochirurgie Fonctionnelle, Neurologie, Neurophysiologie, Anesthésie, Centre d'Investigation Clinique 006, Plateforme de Ressources Biologiques, Créteil, France; Université Paris 12, Faculté de Médecine, Créteil, France
| | - Patrice Dolphin
- AP-HP, Groupe Hospitalier Henri-Mondor, DHU PePsy, UF Neurochirurgie Fonctionnelle, Neurologie, Neurophysiologie, Anesthésie, Centre d'Investigation Clinique 006, Plateforme de Ressources Biologiques, Créteil, France; Université Paris 12, Faculté de Médecine, Créteil, France
| | - David P Breen
- John van Geest Centre for Brain Repair and Addenbrooke's Hospital, Cambridge, UK
| | - Sarah Mason
- John van Geest Centre for Brain Repair and Addenbrooke's Hospital, Cambridge, UK
| | - Natalie Valle Guzman
- John van Geest Centre for Brain Repair and Addenbrooke's Hospital, Cambridge, UK
| | - Nicholas D Mazarakis
- Gene Therapy, Centre of Neuroinflammation and Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Hammersmith Hospital Campus, London, UK
| | | | | | | | - Olivier Rascol
- CIC9302 and UMR 825, INSERM and Department of Pharmacology and Neurosciences, University Hospital and University of Toulouse III, Toulouse, France
| | | | - Roger A Barker
- John van Geest Centre for Brain Repair and Addenbrooke's Hospital, Cambridge, UK
| | - Philippe Hantraye
- CEA, DSV I(2)BM, MIRCen and CNRS URA2210, Fontenay-aux-Roses, France
| | - Philippe Remy
- AP-HP, Groupe Hospitalier Henri-Mondor, DHU PePsy, UF Neurochirurgie Fonctionnelle, Neurologie, Neurophysiologie, Anesthésie, Centre d'Investigation Clinique 006, Plateforme de Ressources Biologiques, Créteil, France; Université Paris 12, Faculté de Médecine, Créteil, France; CEA, DSV I(2)BM, MIRCen and CNRS URA2210, Fontenay-aux-Roses, France
| | - Pierre Cesaro
- AP-HP, Groupe Hospitalier Henri-Mondor, DHU PePsy, UF Neurochirurgie Fonctionnelle, Neurologie, Neurophysiologie, Anesthésie, Centre d'Investigation Clinique 006, Plateforme de Ressources Biologiques, Créteil, France; Université Paris 12, Faculté de Médecine, Créteil, France; INSERM U955, E01, Institut de Recherche Biomédicale, Créteil, France
| | | |
Collapse
|
229
|
Roles for the TGFβ superfamily in the development and survival of midbrain dopaminergic neurons. Mol Neurobiol 2014; 50:559-73. [PMID: 24504901 DOI: 10.1007/s12035-014-8639-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 01/02/2014] [Indexed: 12/29/2022]
Abstract
The adult midbrain contains 75% of all dopaminergic neurons in the CNS. Within the midbrain, these neurons are divided into three anatomically and functionally distinct clusters termed A8, A9 and A10. The A9 group plays a functionally non-redundant role in the control of voluntary movement, which is highlighted by the motor syndrome that results from their progressive degeneration in the neurodegenerative disorder, Parkinson's disease. Despite 50 years of investigation, treatment for Parkinson's disease remains symptomatic, but an intensive research effort has proposed delivering neurotrophic factors to the brain to protect the remaining dopaminergic neurons, or using these neurotrophic factors to differentiate dopaminergic neurons from stem cell sources for cell transplantation. Most neurotrophic factors studied in this context have been members of the transforming growth factor β (TGFβ) superfamily. In recent years, an intensive research effort has focused on understanding the function of these proteins in midbrain dopaminergic neuron development and their role in the molecular architecture that regulates the development of this brain region, with the goal of applying this knowledge to develop novel therapies for Parkinson's disease. In this review, the current evidence showing that TGFβ superfamily members play critical roles in the regulation of midbrain dopaminergic neuron induction, differentiation, target innervation and survival during embryonic and postnatal development is analysed, and the implications of these findings are discussed.
Collapse
|
230
|
Kasinathan N, Jagani HV, Alex AT, Volety SM, Rao JV. Strategies for drug delivery to the central nervous system by systemic route. Drug Deliv 2014; 22:243-57. [DOI: 10.3109/10717544.2013.878858] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
231
|
Controlled release of glial cell line-derived neurotrophic factor from poly(ε-caprolactone) microspheres. Drug Deliv Transl Res 2014; 4:159-70. [DOI: 10.1007/s13346-013-0189-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
232
|
Linazasoro GJ. Neuroprotection in Parkinson’s disease: love story or mission impossible? Expert Rev Neurother 2014; 2:403-16. [DOI: 10.1586/14737175.2.3.403] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
233
|
O’Neill MJ, Siemers ER. Pharmacological approaches to disease-modifying therapies in Parkinson’s disease. Expert Rev Neurother 2014; 2:819-34. [DOI: 10.1586/14737175.2.6.819] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
234
|
Yasuda T, Mochizuki H. Use of growth factors for the treatment of Parkinson’s disease. Expert Rev Neurother 2014; 10:915-24. [DOI: 10.1586/ern.10.55] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
235
|
Douglas MR. Gene therapy for Parkinson's disease: state-of-the-art treatments for neurodegenerative disease. Expert Rev Neurother 2014; 13:695-705. [PMID: 23739006 DOI: 10.1586/ern.13.58] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Pharmacological and surgical treatments offer symptomatic benefits to patients with Parkinson's disease; however, as the condition progresses, patients experience gradual worsening in symptom control, with the development of a range of disabling complications. In addition, none of the currently available therapies have convincingly shown disease-modifying effects - either in slowing or reversing the disease. These problems have led to extensive research into the possible use of gene therapy as a treatment for Parkinson's disease. Several treatments have reached human clinical trial stages, providing important information on the risks and benefits of this novel therapeutic approach, and the tantalizing promise of improved control of this currently incurable neurodegenerative disorder.
Collapse
Affiliation(s)
- Michael R Douglas
- School of Clinical and Experimental Medicine, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
236
|
Wang Y, Fang Q, Gong N. Motor assessment of developing common marmosets. Neurosci Bull 2014; 30:387-93. [PMID: 24395453 DOI: 10.1007/s12264-013-1395-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 06/03/2013] [Indexed: 11/26/2022] Open
Abstract
Motor development has been extensively studied in human infants and children, with several established scales for the evaluation of motor functions. However, the study of the neuronal mechanisms underlying human motor development is hampered by the lack of good animal models. The common marmoset (Callithrix jacchus), a small New World monkey, has recently attracted much attention as a potential nonhuman primate model for understanding human physiology and diseases. However, little is known about its gross motor development. In the present study, we found that marmosets have a critical period for motor development in postnatal weeks 2 to 5, and acquire most of their motor skills by 8 weeks of age. We also developed methods to assess their motor functions, which will be useful for the evaluation of motor performance in marmoset models of human diseases. In addition, we found that marmosets exhibit a "head-to-tail" sequence of motor development similar to that found in humans, further supporting the notion that they provide a good animal model for studying the neuronal mechanisms underlying human motor development.
Collapse
Affiliation(s)
- Yiwen Wang
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | | | | |
Collapse
|
237
|
Pabon M, Tamboli C, Tamboli S, Acosta S, De La Pena I, Sanberg PR, Tajiri N, Kaneko Y, Borlongan CV. ESTROGEN REPLACEMENT THERAPY FOR STROKE. CELL MEDICINE 2014; 6:111-122. [PMID: 24999442 DOI: 10.3727/215517913x672263] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Stroke is the third most common cause of death and severe disability among Western populations. Overall, the incidence of stroke is uniformly higher in men than in women. Stroke is rare in women during the reproductive years, and rapidly increases after menopause, strongly suggesting that estrogen (E2) plays an important role in the prevention of stroke. Ongoing studies are currently evaluating both the benefits and risks associated with E2 replacement therapy and hormone replacement therapy in stroke. Equally important is the role of E2 receptor (ER), as studies indicate that ER populations in several tissue sites may significantly change during stress and aging. Such changes may affect the patient's susceptibility to neurological disorders including stroke, and greatly affect the response to selective E2 receptor modulators (SERMs). Replacement therapies may be inefficient with low ER levels. The goal of this review paper is to discuss an animal model that will allow investigations of the potential therapeutic effects of E2 and its derivatives in stroke. We hypothesize that E2 neuroprotection is, in part, receptor mediated. This hypothesis is a proof of principle approach to demonstrate a role for specific ER subtypes in E2 neuroprotection. To accomplish this, we use a retroviral mediated gene transfer strategy that express subtypes of the ER gene in regions of the rat brain most susceptible to neuronal damage, namely the striatum and cortex. The animal model is exposed to experimental stroke conditions involving middle cerebral artery occlusion (MCAo) method, and eventually the extent of neuronal damage will be evaluated. A reduction in neuronal damage is expected when E2 is administered with specific ER subtypes. From this animal model, an optimal E2 dose and treatment regimen can be determined. The animal model can help identify potential E2-like therapeutics in stroke, and screen for beneficial or toxic additives present in commercial E2 preparations that are currently available. Such studies will be informative in designing drug therapies for stroke.
Collapse
Affiliation(s)
- Mibel Pabon
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Cyrus Tamboli
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Sarosh Tamboli
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Sandra Acosta
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Ike De La Pena
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Paul R Sanberg
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Naoki Tajiri
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Yuji Kaneko
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| |
Collapse
|
238
|
|
239
|
Kantor B, Bailey RM, Wimberly K, Kalburgi SN, Gray SJ. Methods for gene transfer to the central nervous system. ADVANCES IN GENETICS 2014; 87:125-97. [PMID: 25311922 DOI: 10.1016/b978-0-12-800149-3.00003-2] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gene transfer is an increasingly utilized approach for research and clinical applications involving the central nervous system (CNS). Vectors for gene transfer can be as simple as an unmodified plasmid, but more commonly involve complex modifications to viruses to make them suitable gene delivery vehicles. This chapter will explain how tools for CNS gene transfer have been derived from naturally occurring viruses. The current capabilities of plasmid, retroviral, adeno-associated virus, adenovirus, and herpes simplex virus vectors for CNS gene delivery will be described. These include both focal and global CNS gene transfer strategies, with short- or long-term gene expression. As is described in this chapter, an important aspect of any vector is the cis-acting regulatory elements incorporated into the vector genome that control when, where, and how the transgene is expressed.
Collapse
Affiliation(s)
- Boris Kantor
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina, Columbia, SC, USA
| | - Rachel M Bailey
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Keon Wimberly
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sahana N Kalburgi
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Steven J Gray
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Ophthalmology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
240
|
Wang G, Qi J. Direct estimation of kinetic parametric images for dynamic PET. Theranostics 2013; 3:802-15. [PMID: 24396500 PMCID: PMC3879057 DOI: 10.7150/thno.5130] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 08/04/2013] [Indexed: 12/25/2022] Open
Abstract
Dynamic positron emission tomography (PET) can monitor spatiotemporal distribution of radiotracer in vivo. The spatiotemporal information can be used to estimate parametric images of radiotracer kinetics that are of physiological and biochemical interests. Direct estimation of parametric images from raw projection data allows accurate noise modeling and has been shown to offer better image quality than conventional indirect methods, which reconstruct a sequence of PET images first and then perform tracer kinetic modeling pixel-by-pixel. Direct reconstruction of parametric images has gained increasing interests with the advances in computing hardware. Many direct reconstruction algorithms have been developed for different kinetic models. In this paper we review the recent progress in the development of direct reconstruction algorithms for parametric image estimation. Algorithms for linear and nonlinear kinetic models are described and their properties are discussed.
Collapse
|
241
|
Hua WK, Shiau YH, Lee OK, Lin WJ. Elevation of protein kinase Cα stimulates osteogenic differentiation of mesenchymal stem cells through the TAT-mediated protein transduction system. Biochem Cell Biol 2013; 91:443-8. [PMID: 24219286 DOI: 10.1139/bcb-2013-0035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) can differentiate toward various lineages, including the osteogenic lineage, and thus hold great potential for clinic purposes. By using pharmacological inhibitors, protein kinase C (PKC) signaling has been shown to either negatively or positively regulate differentiation of bone, however, due to the low transfection efficiency in MSCs, the role of individual PKC isoforms is still not fully understood. In this study, we established a TAT peptide-mediated transduction system that efficiently delivered PKCα proteins into MSCs in a non-invasive fashion. The increased PKCα protein levels significantly promoted osteogenic differentiation in the murine mesenchymal C3H10T1/2 cells and in primary MSCs from both human and mouse, as demonstrated by the enhanced activity of the osteoblast marker, alkaline phosphatase, and the enhanced expression of the key transcription factor runx2. Mineralization is an important functional indication for bone differentiation. Our results further showed that PKCα promoted expression of the important osteocalcin gene and the accumulation of calcium minerals. Taken together, this study provides direct evidence showing that PKCα positively regulates osteogenic differentiation and demonstrates that the TAT peptide-mediated method enables functional study of specific PKC isoforms in MSCs without using viral infection. This may promote the application of PKCs in therapeutic treatment.
Collapse
Affiliation(s)
- Wei-Kai Hua
- a Institute of Biopharmaceutical Sciences, National Yang-Ming University, No. 155, Sec. 2, Linong St., Taipei 112, Taiwan
| | | | | | | |
Collapse
|
242
|
Chtarto A, Bockstael O, Tshibangu T, Dewitte O, Levivier M, Tenenbaum L. A next step in adeno-associated virus-mediated gene therapy for neurological diseases: regulation and targeting. Br J Clin Pharmacol 2013; 76:217-32. [PMID: 23331189 DOI: 10.1111/bcp.12065] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 12/07/2012] [Indexed: 02/04/2023] Open
Abstract
Recombinant adeno-associated virus (rAAV) vectors mediating long term transgene expression are excellent gene therapy tools for chronic neurological diseases. While rAAV2 was the first serotype tested in the clinics, more efficient vectors derived from the rh10 serotype are currently being evaluated and other serotypes are likely to be tested in the near future. In addition, aside from the currently used stereotaxy-guided intraparenchymal delivery, new techniques for global brain transduction (by intravenous or intra-cerebrospinal injections) are very promising. Various strategies for therapeutic gene delivery to the central nervous system have been explored in human clinical trials in the past decade. Canavan disease, a genetic disease caused by an enzymatic deficiency, was the first to be approved. Three gene transfer paradigms for Parkinson's disease have been explored: converting L-dopa into dopamine through AADC gene delivery in the putamen; synthesizing GABA through GAD gene delivery in the overactive subthalamic nucleus and providing neurotrophic support through neurturin gene delivery in the nigro-striatal pathway. These pioneer clinical trials demonstrated the safety and tolerability of rAAV delivery in the human brain at moderate doses. Therapeutic effects however, were modest, emphasizing the need for higher doses of the therapeutic transgene product which could be achieved using more efficient vectors or expression cassettes. This will require re-addressing pharmacological aspects, with attention to which cases require either localized and cell-type specific expression or efficient brain-wide transgene expression, and when it is necessary to modulate or terminate the administration of transgene product. The ongoing development of targeted and regulated rAAV vectors is described.
Collapse
Affiliation(s)
- Abdelwahed Chtarto
- Laboratory of Experimental Neurosurgery, Free University of Brussels (ULB), Brussels, Belgium
| | | | | | | | | | | |
Collapse
|
243
|
Zhang C, Jin Y, Ziemba KS, Fletcher AM, Ghosh B, Truit E, Yurek DM, Smith GM. Long distance directional growth of dopaminergic axons along pathways of netrin-1 and GDNF. Exp Neurol 2013; 250:156-64. [PMID: 24099728 DOI: 10.1016/j.expneurol.2013.09.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 09/26/2013] [Accepted: 09/26/2013] [Indexed: 12/20/2022]
Abstract
Different experimental and clinical strategies have been used to promote survival of transplanted embryonic ventral mesencephalic (VM) neurons. However, few studies have focused on the long-distance growth of dopaminergic axons from VM transplants. The aim of this study is to identify some of the growth and guidance factors that support directed long-distance growth of dopaminergic axons from VM transplants. Lentivirus encoding either glial cell line-derived neurotrophic factor (GDNF) or netrin-1, or a combination of lenti-GDNF with either lenti-GDNF family receptor α1 (GFRα-1) or lenti-netrin-1 was injected to form a gradient along the corpus callosum. Two weeks later, a piece of embryonic day 14 VM tissue was transplanted into the corpus callosum adjacent to the low end of the gradient. Results showed that tyrosine hydroxylase (TH(+)) axons grew a very short distance from the VM transplants in control groups, with few axons reaching the midline. In GDNF or netrin-1 expressing groups, more TH(+) axons grew out of transplants and reached the midline. Pathways co-expressing GDNF with either GFRα-1 or netrin-1 showed significantly increased axonal outgrowth. Interestingly, only the GDNF/netrin-1 combination resulted in the majority of axons reaching the distal target (80%), whereas along the GDNF/GFRα-1 pathway only 20% of the axons leaving the transplant reached the distal target. This technique of long-distance axon guidance may prove to be a useful strategy in reconstructing damaged neuronal circuits, such as the nigrostriatal pathway in Parkinson's disease.
Collapse
Affiliation(s)
- C Zhang
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA; Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY 40536, USA
| | | | | | | | | | | | | | | |
Collapse
|
244
|
Continuous DOPA synthesis from a single AAV: dosing and efficacy in models of Parkinson's disease. Sci Rep 2013; 3:2157. [PMID: 23831692 PMCID: PMC3703607 DOI: 10.1038/srep02157] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 06/18/2013] [Indexed: 11/09/2022] Open
Abstract
We used a single adeno-associated viral (AAV) vector co-expressing tyrosine hydroxylase (TH) and GTP cyclohydrolase 1 (GCH1) to investigate the relationship between vector dose, and the magnitude and rate of recovery in hemi-parkinsonian rats. Intrastriatal injections of >1E10 genomic copies (gc) of TH-GCH1 vector resulted in complete recovery in drug-naïve behavior tests. Lower vector dose gave partial to no functional improvement. Stereological quantification revealed no striatal NeuN+ cell loss in any of the groups, whereas a TH-GCH1 dose of >1E11 gc resulted in cell loss in globus pallidus. Thus, a TH-GCH1 dose of 1E10 gc gave complete recovery without causing neuronal loss. Safety and efficacy was also studied in non-human primates where the control vector resulted in co-expression of the transgenes in caudate-putamen. In the TH-GCH1 group, GCH1 expression was robust but TH was not detectable. Moreover, TH-GCH1 treatment did not result in functional improvement in non-human primates.
Collapse
|
245
|
Autologous transplantation of GDNF-expressing mesenchymal stem cells protects against MPTP-induced damage in cynomolgus monkeys. Sci Rep 2013; 3:2786. [PMID: 24071770 PMCID: PMC4070584 DOI: 10.1038/srep02786] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 09/11/2013] [Indexed: 12/27/2022] Open
Abstract
Glial cell-derived neurotrophic factor (GDNF) has shown beneficial effects in models of Parkinson's disease. The mild results observed in the double-blind clinical trial by intraputamenal infusion of recombinant GDNF proteins warrant a search for alternative delivery methods. In this study, we investigated the function of autologous mesenchymal stem cells (MSCs) expressing GDNF (GDNF-MSCs) for protection against MPTP-induced injury in cynomolgus monkeys. MSCs were obtained from the bone marrow of individual monkeys and gene-modified to express GDNF. Following unilateral engraftment of GDNF-MSCs into the striatum and substantia nigra, the animals were challenged with MPTP to induce a stable systemic Parkinsonian state. The motor functions were spared in the contralateral limbs of monkeys receiving GDNF-MSCs, but not in those receiving MSCs alone. In the striatum of the grafted hemisphere, dopamine levels were higher and dopamine uptake was enhanced. The results suggest that autologous MSCs may be a safe vehicle to deliver GDNF for enhancing nigro-striatum functions.
Collapse
|
246
|
Glavaski-Joksimovic A, Bohn MC. Mesenchymal stem cells and neuroregeneration in Parkinson's disease. Exp Neurol 2013; 247:25-38. [DOI: 10.1016/j.expneurol.2013.03.016] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 03/14/2013] [Indexed: 02/06/2023]
|
247
|
Kordower JH, Bjorklund A. Trophic factor gene therapy for Parkinson's disease. Mov Disord 2013; 28:96-109. [PMID: 23390096 DOI: 10.1002/mds.25344] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 12/05/2012] [Accepted: 12/13/2012] [Indexed: 11/06/2022] Open
Abstract
Parkinson's disease (PD) is a chronic and progressive neurodegenerative movement disorder for which there is presently no cure. Pharmacological remedies targeting the dopaminergic network are relatively effective at ameliorating motor deficits, especially in the early stages of the disease, but none of these therapies are curative and many generate their own problems. Recent advances in PD research have demonstrated that gene delivery of trophic factors, glial cell line-derived neurotrophic factor (GDNF) and neurturin, in particular, can provide structural and functional recovery in rodent and nonhuman primate models of PD. Similar success has been gleaned in open-label clinical trials, although this has yet to be realized in double-blinded analyses. This work reviews the field of trophic factor gene delivery for PD.
Collapse
Affiliation(s)
- Jeffrey H Kordower
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois 60612, USA.
| | | |
Collapse
|
248
|
Geraniol ameliorates the motor behavior and neurotrophic factors inadequacy in MPTP-induced mice model of Parkinson's disease. J Mol Neurosci 2013; 51:851-62. [PMID: 23943375 PMCID: PMC3824202 DOI: 10.1007/s12031-013-0074-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Accepted: 07/11/2013] [Indexed: 11/01/2022]
Abstract
Many experiments affirm the notion that augmentation of neurotrophic factors (NTFs) activity, especially brain-derived neurotrophic factors and glial cell-derived neurotrophic factors, could prevent or halt the progress of neurodegeneration in Parkinson's disease (PD). In this study, we investigated the therapeutic accomplishment of geraniol (GE 100 mg/kg) on 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP)-induced mice model of PD. Current investigation proved that pretreatment with GE ameliorates the MPTP-induced alterations in behavioral, biochemical, immunohistochemical, and immunoblotting manifestations in mice. Systematically, the loss of dopaminergic neurons and reduced NTFs mRNA expressions induced by MPTP was ameliorated to a significant extent by pretreatment with GE. We found that GE confers a potent neuroprotective agent against MPTP-induced dopaminergic denervation and may become a potential therapeutic agent for PD and/or its progression.
Collapse
|
249
|
The proform of glia cell line-derived neurotrophic factor: a potentially biologically active protein. Mol Neurobiol 2013; 49:234-50. [PMID: 23934644 DOI: 10.1007/s12035-013-8515-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 07/10/2013] [Indexed: 12/24/2022]
Abstract
Growing evidences have revealed that the proforms of several neurotrophins including nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and neurotrophin-3 (NT3), by binding to p75 neurotrophin receptor and sortilin, could induce neuronal apoptosis and are implicated in the pathogenesis of various neurodegenerative diseases. The glial cell line-derived neurotrophic factor (GDNF), one of the most potent useful neurotrophic factors for the treatment of Parkinson's disease (PD), is firstly synthesized as the proform (proGDNF) like other neurotrophin NGF, BDNF, and NT3. However, little is known about proGDNF expression and secretion under physiological as well as pathological states in vivo or in vitro. In this study, we investigated the expression profile and dynamic changes of proGDNF in brains of aging and PD animal models, with the interesting finding that proGDNF was a predominant form of GDNF with molecular weight of about 36 kDa by reducing and nonreducing immunoblots in adult brains and was unregulated in the aging, lipopolysaccharide (LPS), and 1-methyl-4-phenyl- 1,2,3,6-tetrahydropyridine (MPTP) insult. We further provided direct evidence that accompanied activation of primary astrocytes as well as C6 cell line induced by LPS stimulation, proGDNF was increasingly synthesized and released as the uncleaved form in cell culture. Taken together, our results strongly suggest that proGDNF may be a biologically active protein and has specific effects on the cells close to its secreting site, and a potentially important role of proGDNF signaling in the brains, in the glia-neuronal interaction or in the pathogenesis of PD, should merit further investigation.
Collapse
|
250
|
Redmond DE, McEntire CRS, Kingsbery JP, Leranth C, Elsworth JD, Bjugstad KB, Roth RH, Samulski RJ, Sladek JR. Comparison of fetal mesencephalic grafts, AAV-delivered GDNF, and both combined in an MPTP-induced nonhuman primate Parkinson's model. Mol Ther 2013; 21:2160-8. [PMID: 23913185 DOI: 10.1038/mt.2013.180] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Accepted: 07/20/2013] [Indexed: 02/08/2023] Open
Abstract
We combined viral vector delivery of human glial-derived neurotrophic factor (GDNF) with the grafting of dopamine (DA) precursor cells from fetal ventral mesencephalon (VM) to determine whether these strategies would improve the anti-Parkinson's effects in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated monkeys, an animal model for Parkinson's disease (PD). Both strategies have been reported as individually beneficial in animal models of PD, leading to clinical studies. GDNF delivery has also been reported to augment VM tissue implants, but no combined studies have been done in monkeys. Monkeys were treated with MPTP and placed into four balanced treatment groups receiving only recombinant adeno-associated virus serotype 5 (rAAV5)/hu-GDNF, only fetal DA precursor cells, both together, or a buffered saline solution (control). The combination of fetal precursors with rAAV5/hu-GDNF showed significantly higher striatal DA concentrations compared with the other treatments, but did not lead to greater functional improvement in this study. For the first time under identical conditions in primates, we show that all three treatments lead to improvement compared with control animals.
Collapse
Affiliation(s)
- D Eugene Redmond
- 1] Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA [2] Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | | | | | | | | | | | | | |
Collapse
|