1
|
Arango D, Bittar A, Esmeral NP, Ocasión C, Muñoz-Camargo C, Cruz JC, Reyes LH, Bloch NI. Understanding the Potential of Genome Editing in Parkinson's Disease. Int J Mol Sci 2021; 22:9241. [PMID: 34502143 PMCID: PMC8430539 DOI: 10.3390/ijms22179241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 01/05/2023] Open
Abstract
CRISPR is a simple and cost-efficient gene-editing technique that has become increasingly popular over the last decades. Various CRISPR/Cas-based applications have been developed to introduce changes in the genome and alter gene expression in diverse systems and tissues. These novel gene-editing techniques are particularly promising for investigating and treating neurodegenerative diseases, including Parkinson's disease, for which we currently lack efficient disease-modifying treatment options. Gene therapy could thus provide treatment alternatives, revolutionizing our ability to treat this disease. Here, we review our current knowledge on the genetic basis of Parkinson's disease to highlight the main biological pathways that become disrupted in Parkinson's disease and their potential as gene therapy targets. Next, we perform a comprehensive review of novel delivery vehicles available for gene-editing applications, critical for their successful application in both innovative research and potential therapies. Finally, we review the latest developments in CRISPR-based applications and gene therapies to understand and treat Parkinson's disease. We carefully examine their advantages and shortcomings for diverse gene-editing applications in the brain, highlighting promising avenues for future research.
Collapse
Affiliation(s)
- David Arango
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (D.A.); (A.B.); (N.P.E.); (C.M.-C.); (J.C.C.)
| | - Amaury Bittar
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (D.A.); (A.B.); (N.P.E.); (C.M.-C.); (J.C.C.)
| | - Natalia P. Esmeral
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (D.A.); (A.B.); (N.P.E.); (C.M.-C.); (J.C.C.)
| | - Camila Ocasión
- Grupo de Diseño de Productos y Procesos, Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (C.O.); (L.H.R.)
| | - Carolina Muñoz-Camargo
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (D.A.); (A.B.); (N.P.E.); (C.M.-C.); (J.C.C.)
| | - Juan C. Cruz
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (D.A.); (A.B.); (N.P.E.); (C.M.-C.); (J.C.C.)
| | - Luis H. Reyes
- Grupo de Diseño de Productos y Procesos, Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (C.O.); (L.H.R.)
| | - Natasha I. Bloch
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (D.A.); (A.B.); (N.P.E.); (C.M.-C.); (J.C.C.)
| |
Collapse
|
2
|
Rosenblad C, Li Q, Pioli EY, Dovero S, Antunes AS, Agúndez L, Bardelli M, Linden RM, Henckaerts E, Björklund A, Bezard E, Björklund T. Vector-mediated l-3,4-dihydroxyphenylalanine delivery reverses motor impairments in a primate model of Parkinson's disease. Brain 2020; 142:2402-2416. [PMID: 31243443 PMCID: PMC6658866 DOI: 10.1093/brain/awz176] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/19/2019] [Accepted: 04/24/2019] [Indexed: 02/06/2023] Open
Abstract
Ever since its introduction 40 years ago l-3,4-dihydroxyphenylalanine (l-DOPA) therapy has retained its role as the leading standard medication for patients with Parkinson's disease. With time, however, the shortcomings of oral l-DOPA treatment have become apparent, particularly the motor fluctuations and troublesome dyskinetic side effects. These side effects, which are caused by the excessive swings in striatal dopamine caused by intermittent oral delivery, can be avoided by delivering l-DOPA in a more continuous manner. Local gene delivery of the l-DOPA synthesizing enzymes, tyrosine hydroxylase and guanosine-tri-phosphate-cyclohydrolase-1, offers a new approach to a more refined dopaminergic therapy where l-DOPA is delivered continuously at the site where it is needed i.e. the striatum. In this study we have explored the therapeutic efficacy of adeno-associated viral vector-mediated l-DOPA delivery to the putamen in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated rhesus monkeys, the standard non-human primate model of Parkinson's disease. Viral vector delivery of the two enzymes, tyrosine hydroxylase and guanosine-5'-tri-phosphate-cyclohydrolase-1, bilaterally into the dopamine-depleted putamen, induced a significant, dose-dependent improvement of motor behaviour up to a level identical to that obtained with the optimal dose of peripheral l-DOPA. Importantly, this improvement in motor function was obtained without any adverse dyskinetic effects. These results provide proof-of-principle for continuous vector-mediated l-DOPA synthesis as a novel therapeutic strategy for Parkinson's disease. The constant, local supply of l-DOPA obtained with this approach holds promise as an efficient one-time treatment that can provide long-lasting clinical improvement and at the same time prevent the appearance of motor fluctuations and dyskinetic side effects associated with standard oral dopaminergic medication.
Collapse
Affiliation(s)
- Carl Rosenblad
- Division of Neurology, Department of Clinical Sciences, Lund University, Skane University Hospital, 221 84 Lund, Sweden.,Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden
| | - Qin Li
- Motac Neuroscience, Manchester, UK
| | | | - Sandra Dovero
- Université de Bordeaux, Institut des Maladies Neurodégénératives, Bordeaux, France.,Centre National de la Recherche Scientifique Unité Mixte de Recherche 5293, Institut des Maladies Neurodégénératives, Bordeaux, France
| | - André Slm Antunes
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Leticia Agúndez
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Martino Bardelli
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - R Michael Linden
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Els Henckaerts
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Anders Björklund
- Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden
| | - Erwan Bezard
- Motac Neuroscience, Manchester, UK.,Université de Bordeaux, Institut des Maladies Neurodégénératives, Bordeaux, France.,Centre National de la Recherche Scientifique Unité Mixte de Recherche 5293, Institut des Maladies Neurodégénératives, Bordeaux, France
| | - Tomas Björklund
- Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden.,Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK
| |
Collapse
|
3
|
Melin E, Nanobashvili A, Avdic U, Gøtzsche CR, Andersson M, Woldbye DPD, Kokaia M. Disease Modification by Combinatorial Single Vector Gene Therapy: A Preclinical Translational Study in Epilepsy. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 15:179-193. [PMID: 31660420 PMCID: PMC6807261 DOI: 10.1016/j.omtm.2019.09.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 09/09/2019] [Indexed: 12/31/2022]
Abstract
Gene therapy has been suggested as a plausible novel approach to achieve seizure control in patients with focal epilepsy that do not adequately respond to pharmacological treatment. We investigated the seizure-suppressant potential of combinatorial neuropeptide Y and Y2 receptor single vector gene therapy based on adeno-associated virus serotype 1 (AAV1) in rats. First, a dose-response study in the systemic kainate-induced acute seizure model was performed, whereby the 1012 genomic particles (gp)/mL titer of the vector was selected as an optimal concentration. Second, an efficacy study was performed in the intrahippocampal kainate chronic model of spontaneous recurrent seizures (SRSs), designed to reflect a likely clinical scenario, with magnetic resonance image (MRI)-guided focal unilateral administration of the vector in the hippocampus during the chronic stage of the disease. The efficacy study demonstrated a favorable outcome of the gene therapy, with a 31% responder rate (more than 50% reduction in SRS frequency) and 13% seizure-freedom rate, whereas no such effects were observed in the control animals. The inter-SRS and SRS cluster intervals were also significantly prolonged in the treated group compared to controls. In addition, the SRS duration was significantly reduced in the treated group but not in the controls. This study establishes the SRS-suppressant ability of the single vector combinatorial neuropeptide Y/Y2 receptor gene therapy in a clinically relevant chronic model of epilepsy.
Collapse
Affiliation(s)
- Esbjörn Melin
- Experimental Epilepsy Group, Epilepsy Centre, Lund University Hospital, Sölvegatan 17, 221 84 Lund, Sweden
| | - Avtandil Nanobashvili
- Experimental Epilepsy Group, Epilepsy Centre, Lund University Hospital, Sölvegatan 17, 221 84 Lund, Sweden.,CombiGene AB, Medicon Village, Scheelevägen 2, 223 81 Lund, Sweden
| | - Una Avdic
- Experimental Epilepsy Group, Epilepsy Centre, Lund University Hospital, Sölvegatan 17, 221 84 Lund, Sweden
| | - Casper R Gøtzsche
- CombiGene AB, Medicon Village, Scheelevägen 2, 223 81 Lund, Sweden.,Laboratory of Neural Plasticity, Center for Neuroscience, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - My Andersson
- Experimental Epilepsy Group, Epilepsy Centre, Lund University Hospital, Sölvegatan 17, 221 84 Lund, Sweden
| | - David P D Woldbye
- Laboratory of Neural Plasticity, Center for Neuroscience, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Merab Kokaia
- Experimental Epilepsy Group, Epilepsy Centre, Lund University Hospital, Sölvegatan 17, 221 84 Lund, Sweden
| |
Collapse
|
4
|
Petty A, Cui X, Tesiram Y, Kirik D, Howes O, Eyles D. Enhanced Dopamine in Prodromal Schizophrenia (EDiPS): a new animal model of relevance to schizophrenia. NPJ SCHIZOPHRENIA 2019; 5:6. [PMID: 30926827 PMCID: PMC6441087 DOI: 10.1038/s41537-019-0074-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 03/12/2019] [Indexed: 12/13/2022]
Abstract
One of the most robust neurochemical abnormalities reported in patients living with schizophrenia is an increase in dopamine (DA) synthesis and release in the dorsal striatum (DS). Importantly, it appears that this increase progresses as a patient transitions from a prodromal stage to the clinical diagnosis of schizophrenia. Here we have recreated this pathophysiology in an animal model by increasing the capacity for DA synthesis preferentially within the DS. To achieve this we administer a genetic construct containing the rate-limiting enzymes in DA synthesis-tyrosine hydroxylase (TH), and GTP cyclohydrolase 1 (GCH1) (packaged within an adeno-associated virus)-into the substantia nigra pars compacta (SNpc) of adolescent animals. We refer to this model as "Enhanced Dopamine in Prodromal Schizophrenia" (EDiPS). We first confirmed that the TH enzyme is preferentially increased in the DS. As adults, EDiPS animals release significantly more DA in the DS following a low dose of amphetamine (AMPH), have increased AMPH-induced hyperlocomotion and show deficits in pre-pulse inhibition (PPI). The glutamatergic response to AMPH is also altered, again in the DS. EDiPS represents an ideal experimental platform to (a) understand how a preferential increase in DA synthesis capacity in the DS relates to "positive" symptoms in schizophrenia; (b) understand how manipulation of DS DA may influence other neurotransmitter systems shown to be altered in patients with schizophrenia; (c) allow researchers to follow an "at risk"-like disease course from adolescence to adulthood; and (d) ultimately allow trials of putative prophylactic agents to prevent disease onset in vulnerable populations.
Collapse
Affiliation(s)
- Alice Petty
- Queensland Brain Institute, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Xiaoying Cui
- Queensland Brain Institute, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Yasvir Tesiram
- Centre for Advanced Imaging, University of Queensland, QLD, Brisbane, 4072, Australia
| | - Deniz Kirik
- BRAINS Unit, Department of Experimental Medical Science, Lund University, 22184, Lund, Sweden
| | - Oliver Howes
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- MRS London Institute of Medical Sciences, Hammersmith Hospital, London, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Darryl Eyles
- Queensland Brain Institute, University of Queensland, Brisbane, QLD, 4072, Australia.
- Queensland Centre for Mental Health Research, Wacol, QLD, 4076, Australia.
| |
Collapse
|
5
|
Chatterjee D, Bhatt M, Butler D, De Genst E, Dobson CM, Messer A, Kordower JH. Proteasome-targeted nanobodies alleviate pathology and functional decline in an α-synuclein-based Parkinson's disease model. NPJ PARKINSONS DISEASE 2018; 4:25. [PMID: 30155513 PMCID: PMC6105584 DOI: 10.1038/s41531-018-0062-4] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 07/10/2018] [Accepted: 07/11/2018] [Indexed: 01/02/2023]
Abstract
Therapeutics designed to target α-synuclein (α-syn) aggregation may be critical in halting the progression of pathology in Parkinson's disease (PD) patients. Nanobodies are single-domain antibody fragments that bind with antibody specificity, but allow readier genetic engineering and delivery. When expressed intracellularly as intrabodies, anti-α-syn nanobodies fused to a proteasome-targeting proline, aspartate or glutamate, serine, and threonine (PEST) motif can modulate monomeric concentrations of target proteins. Here we aimed to validate and compare the in vivo therapeutic potential of gene therapy delivery of two proteasome-directed nanobodies selectively targeting α-syn in a synuclein overexpression-based PD model: VH14*PEST (non-amyloid component region) and NbSyn87*PEST (C-terminal region). Stereotaxic injections of adeno-associated viral 5-α-syn (AAV5-α-syn) into the substantia nigra (SN) were performed in Sprague-Dawley rats that were sorted into three cohorts based on pre-operative behavioral testing. Rats were treated with unilateral SN injections of vectors for VH14*PEST, NbSyn87*PEST, or injected with saline 3 weeks post lesion. Post-mortem assessments of the SN showed that both nanobodies markedly reduced the level of phosphorylated Serine-129 α-syn labeling relative to saline-treated animals. VH14*PEST showed considerable maintenance of striatal dopaminergic tone in comparison to saline-treated and NbSyn87*PEST-treated animals as measured by tyrosine hydroxylase immunoreactivity (optical density), DAT immunoreactivity (optical density), and dopamine concentration (high-performance liquid chromatography). Microglial accumulation and inflammatory response, assessed by stereological counts of Iba-1-labeled cells, was modestly increased in NbSyn87*PEST-injected rats but not in VH14*PEST-treated or saline-treated animals. Modest behavioral rescue was also observed, although there was pronounced variability among individual animals. These data validate in vivo therapeutic efficacy of vector-delivered intracellular nanobodies targeting α-syn misfolding and aggregation in synucleinopathies such as PD.
Collapse
Affiliation(s)
- Diptaman Chatterjee
- 1Department of Neurological Sciences, Rush University Medical Center, Chicago, IL c60612 USA
| | - Mansi Bhatt
- 1Department of Neurological Sciences, Rush University Medical Center, Chicago, IL c60612 USA
| | - David Butler
- 2Neural Stem Cell Institute, Regenerative Research Foundation, Rensselaer, NY 12144 USA.,3Department of Biomedical Sciences, University at Albany, Albany, NY 12208 USA
| | - Erwin De Genst
- 4Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW UK
| | - Christopher M Dobson
- 4Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW UK
| | - Anne Messer
- 2Neural Stem Cell Institute, Regenerative Research Foundation, Rensselaer, NY 12144 USA.,3Department of Biomedical Sciences, University at Albany, Albany, NY 12208 USA
| | - Jeffrey H Kordower
- 1Department of Neurological Sciences, Rush University Medical Center, Chicago, IL c60612 USA.,5Van Andel Research Institute, Grand Rapids, MI 49503 USA
| |
Collapse
|
6
|
|
7
|
Golebiowski D, van der Bom IMJ, Kwon CS, Miller AD, Petrosky K, Bradbury AM, Maitland S, Kühn AL, Bishop N, Curran E, Silva N, GuhaSarkar D, Westmoreland SV, Martin DR, Gounis MJ, Asaad WF, Sena-Esteves M. Direct Intracranial Injection of AAVrh8 Encoding Monkey β-N-Acetylhexosaminidase Causes Neurotoxicity in the Primate Brain. Hum Gene Ther 2017; 28:510-522. [PMID: 28132521 DOI: 10.1089/hum.2016.109] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
GM2 gangliosidoses, including Tay-Sachs disease and Sandhoff disease, are lysosomal storage disorders caused by deficiencies in β-N-acetylhexosaminidase (Hex). Patients are afflicted primarily with progressive central nervous system (CNS) dysfunction. Studies in mice, cats, and sheep have indicated safety and widespread distribution of Hex in the CNS after intracranial vector infusion of AAVrh8 vectors encoding species-specific Hex α- or β-subunits at a 1:1 ratio. Here, a safety study was conducted in cynomolgus macaques (cm), modeling previous animal studies, with bilateral infusion in the thalamus as well as in left lateral ventricle of AAVrh8 vectors encoding cm Hex α- and β-subunits. Three doses (3.2 × 1012 vg [n = 3]; 3.2 × 1011 vg [n = 2]; or 1.1 × 1011 vg [n = 2]) were tested, with controls infused with vehicle (n = 1) or transgene empty AAVrh8 vector at the highest dose (n = 2). Most monkeys receiving AAVrh8-cmHexα/β developed dyskinesias, ataxia, and loss of dexterity, with higher dose animals eventually becoming apathetic. Time to onset of symptoms was dose dependent, with the highest-dose cohort producing symptoms within a month of infusion. One monkey in the lowest-dose cohort was behaviorally asymptomatic but had magnetic resonance imaging abnormalities in the thalami. Histopathology was similar in all monkeys injected with AAVrh8-cmHexα/β, showing severe white and gray matter necrosis along the injection track, reactive vasculature, and the presence of neurons with granular eosinophilic material. Lesions were minimal to absent in both control cohorts. Despite cellular loss, a dramatic increase in Hex activity was measured in the thalamus, and none of the animals presented with antibody titers against Hex. The high overexpression of Hex protein is likely to blame for this negative outcome, and this study demonstrates the variations in safety profiles of AAVrh8-Hexα/β intracranial injection among different species, despite encoding for self-proteins.
Collapse
Affiliation(s)
- Diane Golebiowski
- 1 Department of Neurology, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Horae Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Imramsjah M J van der Bom
- 3 Department of Radiology, University of Massachusetts Medical School , Worcester, Massachusetts.,4 New England Center for Stroke Research, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Churl-Su Kwon
- 5 Department of Neurosurgery, Massachusetts General Hospital , Boston, Massachusetts
| | - Andrew D Miller
- 6 New England Primate Research Center, Harvard Medical School , Southborough, Massachusetts
| | - Keiko Petrosky
- 6 New England Primate Research Center, Harvard Medical School , Southborough, Massachusetts
| | - Allison M Bradbury
- 7 Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University , Alabama.,8 Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University , Alabama
| | - Stacy Maitland
- 1 Department of Neurology, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Horae Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Anna Luisa Kühn
- 3 Department of Radiology, University of Massachusetts Medical School , Worcester, Massachusetts.,4 New England Center for Stroke Research, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Nina Bishop
- 9 Department of Animal Medicine, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Elizabeth Curran
- 6 New England Primate Research Center, Harvard Medical School , Southborough, Massachusetts
| | - Nilsa Silva
- 6 New England Primate Research Center, Harvard Medical School , Southborough, Massachusetts
| | - Dwijit GuhaSarkar
- 1 Department of Neurology, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Horae Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Susan V Westmoreland
- 6 New England Primate Research Center, Harvard Medical School , Southborough, Massachusetts
| | - Douglas R Martin
- 7 Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University , Alabama.,8 Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University , Alabama
| | - Matthew J Gounis
- 3 Department of Radiology, University of Massachusetts Medical School , Worcester, Massachusetts.,4 New England Center for Stroke Research, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Wael F Asaad
- 10 Department of Neurosurgery, Alpert Medical School, Brown University , Providence, Rhode Island.,11 Brown Institute for Brain Science, Brown University , Providence, Rhode Island.,12 Rhode Island Hospital , Providence, Rhode Island
| | - Miguel Sena-Esteves
- 1 Department of Neurology, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Horae Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| |
Collapse
|
8
|
Kirik D, Cederfjäll E, Halliday G, Petersén Å. Gene therapy for Parkinson's disease: Disease modification by GDNF family of ligands. Neurobiol Dis 2017; 97:179-188. [DOI: 10.1016/j.nbd.2016.09.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 08/24/2016] [Accepted: 09/06/2016] [Indexed: 10/21/2022] Open
|
9
|
Meng Q, Ying Z, Noble E, Zhao Y, Agrawal R, Mikhail A, Zhuang Y, Tyagi E, Zhang Q, Lee JH, Morselli M, Orozco L, Guo W, Kilts TM, Zhu J, Zhang B, Pellegrini M, Xiao X, Young MF, Gomez-Pinilla F, Yang X. Systems Nutrigenomics Reveals Brain Gene Networks Linking Metabolic and Brain Disorders. EBioMedicine 2016; 7:157-66. [PMID: 27322469 PMCID: PMC4909610 DOI: 10.1016/j.ebiom.2016.04.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/05/2016] [Accepted: 04/07/2016] [Indexed: 12/30/2022] Open
Abstract
Nutrition plays a significant role in the increasing prevalence of metabolic and brain disorders. Here we employ systems nutrigenomics to scrutinize the genomic bases of nutrient–host interaction underlying disease predisposition or therapeutic potential. We conducted transcriptome and epigenome sequencing of hypothalamus (metabolic control) and hippocampus (cognitive processing) from a rodent model of fructose consumption, and identified significant reprogramming of DNA methylation, transcript abundance, alternative splicing, and gene networks governing cell metabolism, cell communication, inflammation, and neuronal signaling. These signals converged with genetic causal risks of metabolic, neurological, and psychiatric disorders revealed in humans. Gene network modeling uncovered the extracellular matrix genes Bgn and Fmod as main orchestrators of the effects of fructose, as validated using two knockout mouse models. We further demonstrate that an omega-3 fatty acid, DHA, reverses the genomic and network perturbations elicited by fructose, providing molecular support for nutritional interventions to counteract diet-induced metabolic and brain disorders. Our integrative approach complementing rodent and human studies supports the applicability of nutrigenomics principles to predict disease susceptibility and to guide personalized medicine. Fructose promotes transcriptomic and epigenomic reprogramming to perturb brain networks linking metabolism and brain function. The extracellular matrix genes Bgn and Fmod emerge as key regulators of gene networks responsive to fructose. The omega-3 fatty acid DHA reverses fructose-induced genomic and network reprogramming.
Meng et al. report fructose as a powerful inducer of genomic and epigenomic variability with the capacity to reorganize gene networks critical for central metabolic regulation and neuronal processes in the brain; conversely, an omega-3 fatty acid, DHA, has the potential to normalize the genomic impact of fructose. Our findings help explain the pathogenic actions of fructose on prevalent metabolic and brain disorders and provide proof-of-concept for nutritional remedies supported by nutrigenomics evidence. Our integrative approach complementing rodent and human studies supports the applicability of nutrigenomics principles to predict disease susceptibility and to guide personalized medicine.
Collapse
Affiliation(s)
- Qingying Meng
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zhe Ying
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Emily Noble
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yuqi Zhao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Rahul Agrawal
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Andrew Mikhail
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yumei Zhuang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ethika Tyagi
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Qing Zhang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jae-Hyung Lee
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Maxillofacial Biomedical Engineering, School of Dentistry, Kyung Hee University, Seoul 130-701, Korea
| | - Marco Morselli
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Luz Orozco
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Weilong Guo
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Center for Synthetic & Systems Biology, TNLIST, Tsinghua University, Beijing 100084, China
| | - Tina M Kilts
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jun Zhu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York 10029, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York 10029, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Xinshu Xiao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Marian F Young
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Fernando Gomez-Pinilla
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
10
|
Gene Therapy of CNS Disorders Using Recombinant AAV Vectors. Transl Neurosci 2016. [DOI: 10.1007/978-1-4899-7654-3_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
11
|
O'Connor DM, Boulis NM. Gene therapy for neurodegenerative diseases. Trends Mol Med 2015; 21:504-12. [PMID: 26122838 DOI: 10.1016/j.molmed.2015.06.001] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 06/02/2015] [Accepted: 06/03/2015] [Indexed: 12/18/2022]
Abstract
Gene therapy is, potentially, a powerful tool for treating neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS), spinal muscular atrophy, Parkinson's disease (PD) and Alzheimer's disease (AD). To date, clinical trials have failed to show any improvement in outcome beyond the placebo effect. Efforts to improve outcomes are focusing on three main areas: vector design and the identification of new vector serotypes, mode of delivery of gene therapies, and identification of new therapeutic targets. These advances are being tested both individually and together to improve efficacy. These improvements may finally make gene therapy successful for these disorders.
Collapse
Affiliation(s)
- Deirdre M O'Connor
- Department of Neurosurgery, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Nicholas M Boulis
- Department of Neurosurgery, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA.
| |
Collapse
|
12
|
Mallory GW, Grahn PJ, Hachmann JT, Lujan JL, Lee KH. Optical stimulation for restoration of motor function after spinal cord injury. Mayo Clin Proc 2015; 90:300-7. [PMID: 25659246 PMCID: PMC4339262 DOI: 10.1016/j.mayocp.2014.12.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 12/03/2014] [Accepted: 12/05/2014] [Indexed: 12/31/2022]
Abstract
Spinal cord injury can be defined as a loss of communication between the brain and the body due to disrupted pathways within the spinal cord. Although many promising molecular strategies have emerged to reduce secondary injury and promote axonal regrowth, there is still no effective cure, and recovery of function remains limited. Functional electrical stimulation (FES) represents a strategy developed to restore motor function without the need for regenerating severed spinal pathways. Despite its technological success, however, FES has not been widely integrated into the lives of spinal cord injury survivors. In this review, we briefly discuss the limitations of existing FES technologies. Additionally, we discuss how optogenetics, a rapidly evolving technique used primarily to investigate select neuronal populations within the brain, may eventually be used to replace FES as a form of therapy for functional restoration after spinal cord injury.
Collapse
Affiliation(s)
- Grant W Mallory
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN
| | - Peter J Grahn
- Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN
| | - Jan T Hachmann
- School of Medicine, Heidelberg University, Neuenheimer Feld, Bergheim, Germany
| | - J Luis Lujan
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN
| | - Kendall H Lee
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN.
| |
Collapse
|
13
|
Controlled Striatal DOPA Production From a Gene Delivery System in a Rodent Model of Parkinson's Disease. Mol Ther 2015; 23:896-906. [PMID: 25592335 DOI: 10.1038/mt.2015.8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 12/29/2014] [Indexed: 12/25/2022] Open
Abstract
Conventional symptomatic treatment for Parkinson's disease (PD) with long-term L-3,4-dihydroxyphenylalanine (DOPA) is complicated with development of drug-induced side effects. In vivo viral vector-mediated gene expression encoding tyrosine hydroxylase (TH) and GTP cyclohydrolase 1 (GCH1) provides a drug delivery strategy of DOPA with distinct advantages over pharmacotherapy. Since the brain alterations made with current gene transfer techniques are irreversible, the therapeutic approaches taken to the clinic should preferably be controllable to match the needs of each individual during the course of their disease. We used a recently described tunable gene expression system based on the use of destabilized dihydrofolate reductase (DD) and generated a N-terminally coupled GCH1 enzyme (DD-GCH1) while the TH enzyme was constitutively expressed, packaged in adeno-associated viral (AAV) vectors. Expression of DD-GCH1 was regulated by the activating ligand trimethoprim (TMP) that crosses the blood-brain barrier. We show that the resulting intervention provides a TMP-dose-dependent regulation of DOPA synthesis that is closely linked to the magnitude of functional effects. Our data constitutes the first proof of principle for controlled reconstitution of dopamine capacity in the brain and suggests that such next-generation gene therapy strategies are now mature for preclinical development toward use in patients with PD.
Collapse
|
14
|
Ayers JI, Fromholt S, Sinyavskaya O, Siemienski Z, Rosario AM, Li A, Crosby KW, Cruz PE, DiNunno NM, Janus C, Ceballos-Diaz C, Borchelt DR, Golde TE, Chakrabarty P, Levites Y. Widespread and efficient transduction of spinal cord and brain following neonatal AAV injection and potential disease modifying effect in ALS mice. Mol Ther 2014; 23:53-62. [PMID: 25228069 DOI: 10.1038/mt.2014.180] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 09/09/2014] [Indexed: 01/01/2023] Open
Abstract
The architecture of the spinal cord makes efficient delivery of recombinant adeno-associated virus (rAAV) vectors throughout the neuraxis challenging. We describe a paradigm in which small amounts of virus delivered intraspinally to newborn mice result in robust rAAV-mediated transgene expression in the spinal cord. We compared the efficacy of rAAV2/1, 2/5, 2/8, and 2/9 encoding EGFP delivered to the hindlimb muscle (IM), cisterna magna (ICM), or lumbar spinal cord (IS) of neonatal pups. IS injection of all four capsids resulted in robust transduction of the spinal cord with rAAV2/5, 2/8, and 2/9 vectors appearing to be transported to brain. ICM injection resulted in widespread expression of EGFP in the brain, and upper spinal cord. IM injection resulted in robust muscle expression, with only rAAV2/8 and 2/9 transducing spinal motor and sensory neurons. As proof of concept, we use the IS paradigm to express murine Interleukin (IL)-10 in the spinal cord of the SOD1-G93A transgenic mouse model of amyotrophic lateral sclerosis. We show that expression of IL-10 in the spinal axis of SOD1-G93A mice altered the immune milieu and significantly prolonged survival. These data establish an efficient paradigm for somatic transgene delivery of therapeutic biologics to the spinal cord of mice.
Collapse
Affiliation(s)
- Jacob I Ayers
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Susan Fromholt
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Olga Sinyavskaya
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Zoe Siemienski
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Awilda M Rosario
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Andrew Li
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Keith W Crosby
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Pedro E Cruz
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Nadia M DiNunno
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Christopher Janus
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Carolina Ceballos-Diaz
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - David R Borchelt
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Todd E Golde
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Paramita Chakrabarty
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Yona Levites
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
15
|
Golpich M, Rahmani B, Mohamed Ibrahim N, Dargahi L, Mohamed Z, Raymond AA, Ahmadiani A. Preconditioning as a potential strategy for the prevention of Parkinson's disease. Mol Neurobiol 2014; 51:313-30. [PMID: 24696268 DOI: 10.1007/s12035-014-8689-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Accepted: 03/23/2014] [Indexed: 12/16/2022]
Abstract
Parkinson's disease (PD) is a chronic neurodegenerative movement disorder characterized by the progressive and massive loss of dopaminergic neurons by neuronal apoptosis in the substantia nigra pars compacta and depletion of dopamine in the striatum, which lead to pathological and clinical abnormalities. A numerous of cellular processes including oxidative stress, mitochondrial dysfunction, and accumulation of α-synuclein aggregates are considered to contribute to the pathogenesis of Parkinson's disease. A further understanding of the cellular and molecular mechanisms involved in the pathophysiology of PD is crucial for developing effective diagnostic, preventative, and therapeutic strategies to cure this devastating disorder. Preconditioning (PC) is assumed as a natural adaptive process whereby a subthreshold stimulus can promote protection against a subsequent lethal stimulus in the brain as well as in other tissues that affords robust brain tolerance facing neurodegenerative insults. Multiple lines of evidence have demonstrated that preconditioning as a possible neuroprotective technique may reduce the neural deficits associated with neurodegenerative diseases such as PD. Throughout the last few decades, a lot of efforts have been made to discover the molecular determinants involved in preconditioning-induced protective responses; although, the accurate mechanisms underlying this "tolerance" phenomenon are not fully understood in PD. In this review, we will summarize pathophysiology and current therapeutic approaches in PD and discuss about preconditioning in PD as a potential neuroprotective strategy. Also the role of gene reprogramming and mitochondrial biogenesis involved in the preconditioning-mediated neuroprotective events will be highlighted. Preconditioning may represent a promising therapeutic weapon to combat neurodegeneration.
Collapse
Affiliation(s)
- Mojtaba Golpich
- Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Cheras, Kuala Lumpur, Malaysia
| | | | | | | | | | | | | |
Collapse
|
16
|
Ojala DS, Amara DP, Schaffer DV. Adeno-associated virus vectors and neurological gene therapy. Neuroscientist 2014; 21:84-98. [PMID: 24557878 DOI: 10.1177/1073858414521870] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Gene therapy has strong potential for treating a variety of genetic disorders, as demonstrated in recent clinical trials. There is unfortunately no scarcity of disease targets, and the grand challenge in this field has instead been the development of safe and efficient gene delivery platforms. To date, approximately two thirds of the 1800 gene therapy clinical trials completed worldwide have used viral vectors. Among these, adeno-associated virus (AAV) has emerged as particularly promising because of its impressive safety profile and efficiency in transducing a wide range of cell types. Gene delivery to the CNS involves both considerable promise and unique challenges, and better AAV vectors are thus needed to translate CNS gene therapy approaches to the clinic. This review discusses strategies for vector design, potential routes of administration, immune responses, and clinical applications of AAV in the CNS.
Collapse
Affiliation(s)
- David S Ojala
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA
| | - Dominic P Amara
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - David V Schaffer
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA Department of Bioengineering, University of California, Berkeley, CA, USA The Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| |
Collapse
|
17
|
Abstract
Parkinson's disease (PD) is a progressive neurological disorder characterized primarily by the degeneration of nigrostriatal dopaminergic neurons and diminution of the neurotransmitter dopamine. Though dopamine replacement therapies such as levodopa can improve the symptoms of PD, the benefits may be overshadowed by side effects and the onset of symptoms not responsive to dopaminergic treatments (e.g., autonomic symptoms, gait and balance problems, and cognitive impairment). Furthermore, no therapies have proven to slow the neurodegenerative process. Novel approaches to address these difficult problems, and others, are being sought. Over the last decade, several innovative gene therapies for PD have entered human clinical trials in an effort to address both symptomatic and potential disease-modifying effects. Though the results of these trials have been mixed, the therapies have generally been safe and well-tolerated, suggesting gene therapy may be a viable treatment for PD in the future. This article will review past and current clinical trials of gene therapies for PD. In addition, novel preclinical approaches to gene therapy for PD will be described.
Collapse
Affiliation(s)
- Patricia J. Allen
- Center for Neurosciences, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY 11030 USA
| | - Andrew Feigin
- Center for Neurosciences, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY 11030 USA
| |
Collapse
|