1
|
Sandoval IM, Kelley CM, Bernal-Conde LD, Steece-Collier K, Marmion DJ, Davidsson M, Crosson SM, Boye SL, Boye SE, Manfredsson FP. Engineered AAV capsid transport mutants overcome transduction deficiencies in the aged CNS. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102332. [PMID: 39445231 PMCID: PMC11497394 DOI: 10.1016/j.omtn.2024.102332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 09/05/2024] [Indexed: 10/25/2024]
Abstract
Adeno-associated virus (AAV)-based gene therapy has enjoyed great successes over the past decade, with Food and Drug Administration-approved therapeutics and a robust clinical pipeline. Nonetheless, barriers to successful translation remain. For example, advanced age is associated with impaired brain transduction, with the diminution of infectivity depending on anatomical region and capsid. Given that CNS gene transfer is often associated with neurodegenerative diseases where age is the chief risk factor, we sought to better understand the causes of this impediment. We assessed two AAV variants hypothesized to overcome factors negatively impacting transduction in the aged brain; specifically, changes in extracellular and cell-surface glycans, and intracellular transport. We evaluated a heparin sulfate proteoglycan null variant with or without mutations enhancing intracellular transport. Vectors were injected into the striatum of young adult or aged rats to address whether improving extracellular diffusion, removing glycan receptor dependence, or improving intracellular transport are important factors in transducing the aged brain. We found that, regardless of the viral capsid, there was a reduction in many of our metrics of transduction in the aged brain. However, the transport mutant was less sensitive to age, suggesting that changes in the cellular transport of AAV capsids are a key factor in age-related transduction deficiency.
Collapse
Affiliation(s)
- Ivette M. Sandoval
- Parkinson’s Disease Research Unit, Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Christy M. Kelley
- Parkinson’s Disease Research Unit, Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Luis Daniel Bernal-Conde
- Parkinson’s Disease Research Unit, Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Kathy Steece-Collier
- Department of Translational Neuroscience, Michigan State University College of Human Medicine, Grand Rapids, MI 49506, USA
| | - David J. Marmion
- Parkinson’s Disease Research Unit, Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Marcus Davidsson
- Parkinson’s Disease Research Unit, Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Sean M. Crosson
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL 32610, USA
| | - Sanford L. Boye
- Powell Gene Therapy Center, Department of Pediatrics, University of Florida, Gainesville, FL 32610, USA
| | - Shannon E. Boye
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL 32610, USA
| | - Fredric P. Manfredsson
- Parkinson’s Disease Research Unit, Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| |
Collapse
|
2
|
Li S, Xu X, Li C, Xu Z, Wu K, Ye Q, Zhang Y, Jiang X, Cang C, Tian C, Wen J. In vivo labeling and quantitative imaging of neuronal populations using MRI. Neuroimage 2023; 281:120374. [PMID: 37729795 DOI: 10.1016/j.neuroimage.2023.120374] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 08/30/2023] [Accepted: 09/10/2023] [Indexed: 09/22/2023] Open
Abstract
The study of neural circuits, which underlies perception, cognition, emotion, and behavior, is essential for understanding the mammalian brain, a complex organ consisting of billions of neurons. To study the structure and function of the brain, in vivo neuronal labeling and imaging techniques are crucial as they provide true physiological information that ex vivo methods cannot offer. In this paper, we present a new strategy for in vivo neuronal labeling and quantification using MRI. We demonstrate the efficacy of this method by delivering the oatp1a1 gene to the target neurons using rAAV2-retro virus. OATP1A1 protein expression on the neuronal membrane increased the uptake of a specific MRI contrast agent (Gd-EOB-DTPA), leading to hyperintense signals on T1W images of labeled neuronal populations. We also used dynamic contrast enhancement-based methods to obtain quantitative information on labeled neuronal populations in vivo.
Collapse
Affiliation(s)
- Shana Li
- The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
| | - Xiang Xu
- The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
| | - Canjun Li
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
| | - Ziyan Xu
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
| | - Ke Wu
- The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
| | - Qiong Ye
- Key Laboratory of High Field Magnetic Resonance Image of Anhui Province, High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, PR China
| | - Yan Zhang
- The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
| | - Xiaohua Jiang
- The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
| | - Chunlei Cang
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
| | - Changlin Tian
- The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China; School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China; Key Laboratory of High Field Magnetic Resonance Image of Anhui Province, High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, PR China.
| | - Jie Wen
- The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China.
| |
Collapse
|
3
|
Brown D, Altermatt M, Dobreva T, Chen S, Wang A, Thomson M, Gradinaru V. Deep Parallel Characterization of AAV Tropism and AAV-Mediated Transcriptional Changes via Single-Cell RNA Sequencing. Front Immunol 2021; 12:730825. [PMID: 34759919 PMCID: PMC8574206 DOI: 10.3389/fimmu.2021.730825] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/17/2021] [Indexed: 12/21/2022] Open
Abstract
Engineered variants of recombinant adeno-associated viruses (rAAVs) are being developed rapidly to meet the need for gene-therapy delivery vehicles with particular cell-type and tissue tropisms. While high-throughput AAV engineering and selection methods have generated numerous variants, subsequent tropism and response characterization have remained low throughput and lack resolution across the many relevant cell and tissue types. To fully leverage the output of these large screening paradigms across multiple targets, we have developed an experimental and computational single-cell RNA sequencing (scRNA-seq) pipeline for in vivo characterization of barcoded rAAV pools at high resolution. Using this platform, we have both corroborated previously reported viral tropisms and discovered unidentified AAV capsid targeting biases. As expected, we observed that the tropism profile of AAV.CAP-B10 in mice was shifted toward neurons and away from astrocytes when compared with AAV-PHP.eB. Transcriptomic analysis revealed that this neuronal bias is due mainly to increased targeting efficiency for glutamatergic neurons, which we confirmed by RNA fluorescence in situ hybridization. We further uncovered cell subtype tropisms of AAV variants in vascular and glial cells, such as low transduction of pericytes and Myoc+ astrocytes. Additionally, we have observed cell-type-specific transitory responses to systemic AAV-PHP.eB administration, such as upregulation of genes involved in p53 signaling in endothelial cells three days post-injection, which return to control levels by day twenty-five. The presented experimental and computational approaches for parallel characterization of AAV tropism will facilitate the advancement of safe and precise gene delivery vehicles, and showcase the power of understanding responses to gene therapies at the single-cell level.
Collapse
Affiliation(s)
- David Brown
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| | - Michael Altermatt
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| | - Tatyana Dobreva
- Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, United States
| | - Sisi Chen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| | - Alexander Wang
- Division of Engineering and Applied Science, California Institute of Technology, Pasadena, CA, United States
| | - Matt Thomson
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| |
Collapse
|
4
|
Self-complementarity in adeno-associated virus enhances transduction and gene expression in mouse cochlear tissues. PLoS One 2020; 15:e0242599. [PMID: 33227033 PMCID: PMC7682903 DOI: 10.1371/journal.pone.0242599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 11/06/2020] [Indexed: 12/03/2022] Open
Abstract
Sensorineural hearing loss is one of the most common disabilities worldwide. Such prevalence necessitates effective tools for studying the molecular workings of cochlear cells. One prominent and effective vector for expressing genes of interest in research models is adeno-associated virus (AAV). However, AAV efficacy in transducing cochlear cells can vary for a number of reasons including serotype, species, and methodology, and oftentimes requires high multiplicity of infection which can damage the sensory cells. Reports in other systems suggest multiple approaches can be used to enhance AAV transduction including self-complementary vector design and pharmacological inhibition of degradation. Here we produced AAV to drive green fluorescent protein (GFP) expression in explanted neonatal mouse cochleae. Treatment with eeyarestatin I, tyrphostin 23, or lipofectamine 2000 did not result in increased transduction, however, self-complementary vector design resulted in significantly more GFP positive cells when compared to single-stranded controls. Similarly, self-complementary AAV2 vectors demonstrated enhanced transduction efficiency compared to single stranded AAV2 when injected via the posterior semicircular canal, in vivo. Self-complementary vectors for AAV1, 8, and 9 serotypes also demonstrated robust GFP transduction in cochlear cells in vivo, though these were not directly compared to single stranded vectors. These findings suggest that second-strand synthesis may be a rate limiting step in AAV transduction of cochlear tissues and that self-complementary AAV can be used to effectively target large numbers of cochlear cells in vitro and in vivo.
Collapse
|
5
|
Zhao J, Yue Y, Patel A, Wasala L, Karp JF, Zhang K, Duan D, Lai Y. High-Resolution Histological Landscape of AAV DNA Distribution in Cellular Compartments and Tissues following Local and Systemic Injection. Mol Ther Methods Clin Dev 2020; 18:856-868. [PMID: 32953935 PMCID: PMC7479330 DOI: 10.1016/j.omtm.2020.08.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 08/06/2020] [Indexed: 12/21/2022]
Abstract
Adeno-associated virus (AAV) is one of the most important gene delivery vehicles for in vivo gene therapy. Intramuscular (i.m.) and intravascular (i.v.) injection are commonly used for AAV gene transfer. Unfortunately, the fate of AAV vectors following administration remains unclear at the histological level. Taking advantage of RNAscope, a recently developed in situ hybridization technique that can reveal high-resolution viral DNA localization information, in this study, we evaluated body-wide distribution of an AAV9 vector in the context of the cell and tissue microenvironments. We observed distinctive kinetics of cell and nuclear entry of the AAV DNA in striated muscle and liver following i.m. and i.v. injection. We also found characteristic distribution patterns of the AAV DNA in various histological structures in internal organs, including gonads and lymph nodes, following i.v. injection. Finally, we showed significantly body-wide spreading of the AAV DNA following i.m. injection. These results add a new dimension to our understanding of AAV transduction biology and provide a basis for assessing the full impact of AAV gene therapy.
Collapse
Affiliation(s)
- Junling Zhao
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Aman Patel
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
- School of Medicine, Saint Louis University, St. Louis, MO 63104, USA
| | - Lakmini Wasala
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Jacob F. Karp
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Keqing Zhang
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO 65212, USA
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
- Department of Bioengineering, University of Missouri, Columbia, MO 65212, USA
| | - Yi Lai
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
6
|
Raghunathan R, Polinski NK, Klein JA, Hogan JD, Shao C, Khatri K, Leon D, McComb ME, Manfredsson FP, Sortwell CE, Zaia J. Glycomic and Proteomic Changes in Aging Brain Nigrostriatal Pathway. Mol Cell Proteomics 2018; 17:1778-1787. [PMID: 29915149 PMCID: PMC6126385 DOI: 10.1074/mcp.ra118.000680] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 06/12/2018] [Indexed: 12/19/2022] Open
Abstract
Parkinson's disease (PD) is a neurological disorder characterized by the progressive loss of functional dopaminergic neurons in the nigrostriatal pathway in the brain. Although current treatments provide only symptomatic relief, gene therapy has the potential to slow or halt the degeneration of nigrostriatal dopamine neurons in PD patients. Adeno-associated viruses (AAV) are vectors of choice in gene therapy because of their well-characterized safety and efficacy profiles; however, although gene therapy has been successful in preclinical models of the disease, clinical trials in humans have failed to demonstrate efficacy. Significantly, all primary AAV receptors of the virus are glycans. We thus hypothesize that age related changes in glycan receptors of heparan sulfate (HS) proteoglycans (receptor for rAAV2), and/or N-glycans with terminal galactose (receptor for rAAV9) results in poor adeno-associated virus binding in either the striatum or substantia nigra, or both, affecting transduction and gene delivery. To test our hypothesis we analyzed the striatum and substantia nigra for changes in HS, N-glycans and proteomic signatures in young versus aged rat brain striatum and substantia nigra. We observed different brain region-specific HS disaccharide profiles in aged compared with young adult rats for brain region-specific profiles in striatum versus substantia nigra. We observed brain region- and age-specific N-glycan compositional profiles with respect to the terminal galactose units that serve as receptors for AAV9. We also observed brain region-specific changes in protein expression in the aging nigrostriatal pathway. These studies provide insight into age- and brain region-specific changes in glycan receptors and proteome that will inform design of improved viral vectors for Parkinson Disease (PD) gene therapy.
Collapse
Affiliation(s)
- Rekha Raghunathan
- From the ‡Department of Molecular and Translational Medicine, Center for Biomedical Mass Spectrometry, Boston University Medical Campus, Boston, Massachusetts
| | - Nicole K Polinski
- ‖Department of Translational Science and Molecular Medicine, Michigan State University
| | - Joshua A Klein
- ¶Bioinformatics Program, Boston University, Boston, Massachusetts
| | - John D Hogan
- ¶Bioinformatics Program, Boston University, Boston, Massachusetts
| | - Chun Shao
- §Department of Biochemistry, Center for Biomedical Mass Spectrometry, Boston University Medical Campus, Boston, Massachusetts
| | - Kshitij Khatri
- §Department of Biochemistry, Center for Biomedical Mass Spectrometry, Boston University Medical Campus, Boston, Massachusetts
| | - Deborah Leon
- §Department of Biochemistry, Center for Biomedical Mass Spectrometry, Boston University Medical Campus, Boston, Massachusetts
| | - Mark E McComb
- §Department of Biochemistry, Center for Biomedical Mass Spectrometry, Boston University Medical Campus, Boston, Massachusetts
| | - Fredric P Manfredsson
- ‖Department of Translational Science and Molecular Medicine, Michigan State University
| | - Caryl E Sortwell
- ‖Department of Translational Science and Molecular Medicine, Michigan State University
| | - Joseph Zaia
- From the ‡Department of Molecular and Translational Medicine, Center for Biomedical Mass Spectrometry, Boston University Medical Campus, Boston, Massachusetts;
- §Department of Biochemistry, Center for Biomedical Mass Spectrometry, Boston University Medical Campus, Boston, Massachusetts
- ¶Bioinformatics Program, Boston University, Boston, Massachusetts
| |
Collapse
|
7
|
Kritzinger A, Ferger B, Gillardon F, Stierstorfer B, Birk G, Kochanek S, Ciossek T. Age-related pathology after adenoviral overexpression of the leucine-rich repeat kinase 2 in the mouse striatum. Neurobiol Aging 2018; 66:97-111. [DOI: 10.1016/j.neurobiolaging.2018.02.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 01/04/2018] [Accepted: 02/10/2018] [Indexed: 02/07/2023]
|
8
|
Subthalamic Nucleus Deep Brain Stimulation Does Not Modify the Functional Deficits or Axonopathy Induced by Nigrostriatal α-Synuclein Overexpression. Sci Rep 2017; 7:16356. [PMID: 29180681 PMCID: PMC5703955 DOI: 10.1038/s41598-017-16690-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 11/16/2017] [Indexed: 12/21/2022] Open
Abstract
Subthalamic nucleus deep brain stimulation (STN DBS) protects dopaminergic neurons of the substantia nigra pars compacta (SNpc) against 6-OHDA and MPTP. We evaluated STN DBS in a parkinsonian model that displays α-synuclein pathology using unilateral, intranigral injections of recombinant adeno-associated virus pseudotype 2/5 to overexpress wildtype human α-synuclein (rAAV2/5 α-syn). A low titer of rAAV2/5 α-syn results in progressive forelimb asymmetry, loss of striatal dopaminergic terminal density and modest loss of SNpc dopamine neurons after eight weeks, corresponding to robust human-Snca expression and no effect on rat-Snca, Th, Bdnf or Trk2. α-syn overexpression increased phosphorylation of ribosomal protein S6 (p-rpS6) in SNpc neurons, a readout of trkB activation. Rats received intranigral injections of rAAV2/5 α-syn and three weeks later received four weeks of STN DBS or electrode implantation that remained inactive. STN DBS did not protect against α-syn-mediated deficits in forelimb akinesia, striatal denervation or loss of SNpc neuron, nor did STN DBS elevate p-rpS6 levels further. ON stimulation, forelimb asymmetry was exacerbated, indicating α-syn overexpression-mediated neurotransmission deficits. These results demonstrate that STN DBS does not protect the nigrostriatal system against α-syn overexpression-mediated toxicity. Whether STN DBS can be protective in other models of synucleinopathy is unknown.
Collapse
|
9
|
Kanaan NM, Sellnow RC, Boye SL, Coberly B, Bennett A, Agbandje-McKenna M, Sortwell CE, Hauswirth WW, Boye SE, Manfredsson FP. Rationally Engineered AAV Capsids Improve Transduction and Volumetric Spread in the CNS. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 8:184-197. [PMID: 28918020 PMCID: PMC5503098 DOI: 10.1016/j.omtn.2017.06.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 06/14/2017] [Accepted: 06/15/2017] [Indexed: 12/13/2022]
Abstract
Adeno-associated virus (AAV) is the most common vector for clinical gene therapy of the CNS. This popularity originates from a high safety record and the longevity of transgene expression in neurons. Nevertheless, clinical efficacy for CNS indications is lacking, and one reason for this is the relatively limited spread and transduction efficacy in large regions of the human brain. Using rationally designed modifications of the capsid, novel AAV capsids have been generated that improve intracellular processing and result in increased transgene expression. Here, we sought to improve AAV-mediated neuronal transduction to minimize the existing limitations of CNS gene therapy. We investigated the efficacy of CNS transduction using a variety of tyrosine and threonine capsid mutants based on AAV2, AAV5, and AAV8 capsids, as well as AAV2 mutants incapable of binding heparan sulfate (HS). We found that mutating several tyrosine residues on the AAV2 capsid significantly enhanced neuronal transduction in the striatum and hippocampus, and the ablation of HS binding also increased the volumetric spread of the vector. Interestingly, the analogous tyrosine substitutions on AAV5 and AAV8 capsids did not improve the efficacy of these serotypes. Our results demonstrate that the efficacy of CNS gene transfer can be significantly improved with minor changes to the AAV capsid and that the effect is serotype specific.
Collapse
Affiliation(s)
- Nicholas M Kanaan
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI 49503, USA; Mercy Health Saint Mary's, Grand Rapids, MI 49503, USA
| | - Rhyomi C Sellnow
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Sanford L Boye
- Department of Ophthalmology, University of Florida, Gainesville, FL 32610, USA
| | - Ben Coberly
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI 49503, USA; Neuroscience Program, Michigan State University, East Lansing, MI 48825, USA
| | - Antonette Bennett
- Department of Biochemistry and Molecular Biology, The McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Mavis Agbandje-McKenna
- Department of Biochemistry and Molecular Biology, The McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Caryl E Sortwell
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI 49503, USA; Mercy Health Saint Mary's, Grand Rapids, MI 49503, USA
| | - William W Hauswirth
- Department of Ophthalmology, University of Florida, Gainesville, FL 32610, USA
| | - Shannon E Boye
- Department of Ophthalmology, University of Florida, Gainesville, FL 32610, USA
| | - Fredric P Manfredsson
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI 49503, USA; Mercy Health Saint Mary's, Grand Rapids, MI 49503, USA.
| |
Collapse
|
10
|
Alpha-Synuclein mRNA Is Not Increased in Sporadic PD and Alpha-Synuclein Accumulation Does Not Block GDNF Signaling in Parkinson's Disease and Disease Models. Mol Ther 2017; 25:2231-2235. [PMID: 28522034 DOI: 10.1016/j.ymthe.2017.04.018] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 04/16/2017] [Accepted: 04/17/2017] [Indexed: 12/20/2022] Open
|
11
|
Polinski NK, Manfredsson FP, Benskey MJ, Fischer DL, Kemp CJ, Steece-Collier K, Sandoval IM, Paumier KL, Sortwell CE. Impact of age and vector construct on striatal and nigral transgene expression. Mol Ther Methods Clin Dev 2016; 3:16082. [PMID: 27933309 PMCID: PMC5142515 DOI: 10.1038/mtm.2016.82] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 10/25/2016] [Indexed: 02/06/2023]
Abstract
Therapeutic protein delivery using viral vectors has shown promise in preclinical models of Parkinson's disease (PD) but clinical trial success remains elusive. This may partially be due to a failure to include advanced age as a covariate despite aging being the primary risk factor for PD. We investigated transgene expression following intracerebral injections of recombinant adeno-associated virus pseudotypes 2/2 (rAAV2/2), 2/5 (rAAV2/5), 2/9 (rAAV2/9), and lentivirus (LV) expressing green fluorescent protein (GFP) in aged versus young adult rats. Both rAAV2/2 and rAAV2/5 yielded lower GFP expression following injection to either the aged substantia nigra or striatum. rAAV2/9-mediated GFP expression was deficient in the aged striatonigral system but displayed identical transgene expression between ages in the nigrostriatal system. Young and aged rats displayed equivalent GFP levels following LV injection to the striatonigral system but LV-delivered GFP was deficient in delivering GFP to the aged nigrostriatal system. Notably, age-related transgene expression deficiencies revealed by protein quantitation were poorly predicted by GFP-immunoreactive cell counts. Further, in situ hybridization for the viral CβA promoter revealed surprisingly limited tropism for astrocytes compared to neurons. Our results demonstrate that aging is a critical covariate to consider when designing gene therapy approaches for PD.
Collapse
Affiliation(s)
- Nicole K Polinski
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, Michigan, USA
- Neuroscience Graduate Program, Michigan State University, East Lansing, Michigan, USA
| | - Fredric P Manfredsson
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, Michigan, USA
- Mercy Health Saint Mary’s, Grand Rapids, Michigan, USA
| | - Matthew J Benskey
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, Michigan, USA
| | - D Luke Fischer
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, Michigan, USA
| | - Christopher J Kemp
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, Michigan, USA
| | - Kathy Steece-Collier
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, Michigan, USA
- Mercy Health Saint Mary’s, Grand Rapids, Michigan, USA
| | - Ivette M Sandoval
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, Michigan, USA
| | - Katrina L Paumier
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, Michigan, USA
| | - Caryl E Sortwell
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, Michigan, USA
- Mercy Health Saint Mary’s, Grand Rapids, Michigan, USA
| |
Collapse
|
12
|
Fischer DL, Gombash SE, Kemp CJ, Manfredsson FP, Polinski NK, Duffy MF, Sortwell CE. Viral Vector-Based Modeling of Neurodegenerative Disorders: Parkinson's Disease. Methods Mol Biol 2016; 1382:367-82. [PMID: 26611600 DOI: 10.1007/978-1-4939-3271-9_26] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Gene therapy methods are increasingly used to model Parkinson's disease (PD) in animals in an effort to test experimental therapeutics within a more relevant context to disease pathophysiology and neuropathology. We have detailed several criteria that are critical or advantageous to accurately modeling PD in a murine model or in a nonhuman primate. Using these criteria, we then evaluate approaches made to model PD using viral vectors to date, including both adeno-associated viruses and lentiviruses. Lastly, we comment on the consideration of aging as a critical factor for modeling PD.
Collapse
Affiliation(s)
- D Luke Fischer
- Department of Translational Science & Molecular Medicine, Michigan State University, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503-2532, USA
- MD/PhD Program, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Graduate Program, Michigan State University, Grand Rapids, MI, USA
| | - Sara E Gombash
- Department of Translational Science & Molecular Medicine, Michigan State University, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503-2532, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, USA
| | - Christopher J Kemp
- Department of Translational Science & Molecular Medicine, Michigan State University, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503-2532, USA
| | - Fredric P Manfredsson
- Translational Science and Molecular Medicine, Michigan State University, College of Human Science, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503-2532, USA
| | - Nicole K Polinski
- Department of Translational Science & Molecular Medicine, Michigan State University, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503-2532, USA
- Neuroscience Graduate Program, Michigan State University, Grand Rapids, MI, USA
| | - Megan F Duffy
- Department of Translational Science & Molecular Medicine, Michigan State University, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503-2532, USA
- Neuroscience Graduate Program, Michigan State University, Grand Rapids, MI, USA
| | - Caryl E Sortwell
- Department of Translational Science & Molecular Medicine, Michigan State University, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503-2532, USA.
| |
Collapse
|
13
|
Grimmig B, Morganti J, Nash K, Bickford PC. Immunomodulators as Therapeutic Agents in Mitigating the Progression of Parkinson's Disease. Brain Sci 2016; 6:brainsci6040041. [PMID: 27669315 PMCID: PMC5187555 DOI: 10.3390/brainsci6040041] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 09/06/2016] [Accepted: 09/20/2016] [Indexed: 12/21/2022] Open
Abstract
Parkinson’s disease (PD) is a common neurodegenerative disorder that primarily afflicts the elderly. It is characterized by motor dysfunction due to extensive neuron loss in the substantia nigra pars compacta. There are multiple biological processes that are negatively impacted during the pathogenesis of PD, and are implicated in the cell death in this region. Neuroinflammation is evidently involved in PD pathology and mitigating the inflammatory cascade has been a therapeutic strategy. Age is the number one risk factor for PD and thus needs to be considered in the context of disease pathology. Here, we discuss the role of neuroinflammation within the context of aging as it applies to the development of PD, and the potential for two representative compounds, fractalkine and astaxanthin, to attenuate the pathophysiology that modulates neurodegeneration that occurs in Parkinson’s disease.
Collapse
Affiliation(s)
- Bethany Grimmig
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| | - Josh Morganti
- Sanders-Brown Center on Aging, Department of Anatomy and Neurobiology, University of Kentucky, Lexington, KY 40508, USA.
| | - Kevin Nash
- Byrd Alzheimer's Institute, Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33613, USA.
| | - Paula C Bickford
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
- James A Haley VA Hospital, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA.
| |
Collapse
|
14
|
Abstract
Dementias are among the most common neurological disorders, and Alzheimer's disease (AD) is the most common cause of dementia worldwide. AD remains a looming health crisis despite great efforts to learn the mechanisms surrounding the neuron dysfunction and neurodegeneration that accompanies AD primarily in the medial temporal lobe. In addition to AD, a group of diseases known as frontotemporal dementias (FTDs) are degenerative diseases involving atrophy and degeneration in the frontal and temporal lobe regions. Importantly, AD and a number of FTDs are collectively known as tauopathies due to the abundant accumulation of pathological tau inclusions in the brain. The precise role tau plays in disease pathogenesis remains an area of strong research focus. A critical component to effectively study any human disease is the availability of models that recapitulate key features of the disease. Accordingly, a number of animal models are currently being pursued to fill the current gaps in our knowledge of the causes of dementias and to develop effective therapeutics. Recent developments in gene therapy-based approaches, particularly in recombinant adeno-associated viruses (rAAVs), have provided new tools to study AD and other related neurodegenerative disorders. Additionally, gene therapy approaches have emerged as an intriguing possibility for treating these diseases in humans. This chapter explores the current state of rAAV models of AD and other dementias, discuss recent efforts to improve these models, and describe current and future possibilities in the use of rAAVs and other viruses in treatments of disease.
Collapse
Affiliation(s)
- Benjamin Combs
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, 333 Bostwick Avenue NE, Grand Rapids, MI, 49503, USA
| | - Andrew Kneynsberg
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, 333 Bostwick Avenue NE, Grand Rapids, MI, 49503, USA
- Neuroscience Program, Michigan State University, Grand Rapids, MI, USA
| | - Nicholas M Kanaan
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, 333 Bostwick Avenue NE, Grand Rapids, MI, 49503, USA.
- Neuroscience Program, Michigan State University, Grand Rapids, MI, USA.
| |
Collapse
|
15
|
The transfection of BDNF to dopamine neurons potentiates the effect of dopamine D3 receptor agonist recovering the striatal innervation, dendritic spines and motor behavior in an aged rat model of Parkinson's disease. PLoS One 2015; 10:e0117391. [PMID: 25693197 PMCID: PMC4332861 DOI: 10.1371/journal.pone.0117391] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 12/21/2014] [Indexed: 01/28/2023] Open
Abstract
The progressive degeneration of the dopamine neurons of the pars compacta of substantia nigra and the consequent loss of the dopamine innervation of the striatum leads to the impairment of motor behavior in Parkinson's disease. Accordingly, an efficient therapy of the disease should protect and regenerate the dopamine neurons of the substantia nigra and the dopamine innervation of the striatum. Nigral neurons express Brain Derived Neurotropic Factor (BDNF) and dopamine D3 receptors, both of which protect the dopamine neurons. The chronic activation of dopamine D3 receptors by their agonists, in addition, restores, in part, the dopamine innervation of the striatum. Here we explored whether the over-expression of BDNF by dopamine neurons potentiates the effect of the activation of D3 receptors restoring nigrostriatal innervation. Twelve-month old Wistar rats were unilaterally injected with 6-hydroxydopamine into the striatum. Five months later, rats were treated with the D3 agonist 7-hydroxy-N,N-di-n-propy1-2-aminotetralin (7-OH-DPAT) administered i.p. during 4½ months via osmotic pumps and the BDNF gene transfection into nigral cells using the neurotensin-polyplex nanovector (a non-viral transfection) that selectively transfect the dopamine neurons via the high-affinity neurotensin receptor expressed by these neurons. Two months after the withdrawal of 7-OH-DPAT when rats were aged (24 months old), immunohistochemistry assays were made. The over-expression of BDNF in rats receiving the D3 agonist normalized gait and motor coordination; in addition, it eliminated the muscle rigidity produced by the loss of dopamine. The recovery of motor behavior was associated with the recovery of the nigral neurons, the dopamine innervation of the striatum and of the number of dendritic spines of the striatal neurons. Thus, the over-expression of BDNF in dopamine neurons associated with the chronic activation of the D3 receptors appears to be a promising strategy for restoring dopamine neurons in Parkinson's disease.
Collapse
|