1
|
An M, Davis JR, Levy JM, Serack FE, Harvey JW, Brauer PP, Pirtle CP, Berríos KN, Newby GA, Yeh WH, Kamath N, Mortberg M, Lian Y, Howard M, DeSouza-Lenz K, Guzman K, Thai A, Graffam S, Laversenne V, Coffey AA, Frei J, Pierce SE, Safar JG, Deverman BE, Minikel EV, Vallabh SM, Liu DR. In vivo base editing extends lifespan of a humanized mouse model of prion disease. Nat Med 2025:10.1038/s41591-024-03466-w. [PMID: 39810005 DOI: 10.1038/s41591-024-03466-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 12/12/2024] [Indexed: 01/16/2025]
Abstract
Prion disease is a fatal neurodegenerative disease caused by the misfolding of prion protein (PrP) encoded by the PRNP gene. While there is currently no cure for the disease, depleting PrP in the brain is an established strategy to prevent or stall templated misfolding of PrP. Here we developed in vivo cytosine and adenine base strategies delivered by adeno-associated viruses to permanently modify the PRNP locus to achieve PrP knockdown in the mouse brain. Systemic injection of dual-adeno-associated virus PHP.eB encoding BE3.9max and single guide RNA installing PRNP R37X resulted in 37% average installation of the desired edit, 50% reduction of PrP in the mouse brain and 52% extension of lifespan in transgenic human PRNP mice inoculated with pathogenic human prion isolates representing the most common sporadic and genetic subtypes of prion disease. We further engineered base editing systems to achieve improved in vivo potency and reduced base editor expression in nontargeting tissues, resulting in 63% average PrP reduction in the mouse brain from a 6.7-fold lower viral dose, with no detected off-target editing of anticipated clinical significance observed in either human cells or mouse tissues. These findings support the potential of in vivo base editing as one-time treatment for prion disease.
Collapse
Affiliation(s)
- Meirui An
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Jessie R Davis
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Jonathan M Levy
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Fiona E Serack
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - John W Harvey
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Pamela P Brauer
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Catherine P Pirtle
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kiara N Berríos
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Gregory A Newby
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Wei-Hsi Yeh
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Nikita Kamath
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Meredith Mortberg
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yuan Lian
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michael Howard
- Comparative Medicine, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Kenia Guzman
- Comparative Medicine, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Aaron Thai
- Comparative Medicine, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Samantha Graffam
- Comparative Medicine, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Vanessa Laversenne
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alissa A Coffey
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jeannine Frei
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sarah E Pierce
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Jiri G Safar
- Case Western Reserve University, Cleveland, OH, USA
| | - Benjamin E Deverman
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Eric Vallabh Minikel
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- McCance Center for Brain Health and Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Department of Neurology, Harvard Medical School, Boston, MA, USA.
- Prion Alliance, Cambridge, MA, USA.
| | - Sonia M Vallabh
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- McCance Center for Brain Health and Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Department of Neurology, Harvard Medical School, Boston, MA, USA.
- Prion Alliance, Cambridge, MA, USA.
| | - David R Liu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
2
|
Bolon B, Buza E, Galbreath E, Wicks J, Cargnin F, Hordeaux J. Neuropathological Findings in Nonclinical Species Following Administration of Adeno-Associated Virus (AAV)-Based Gene Therapy Vectors. Toxicol Pathol 2024; 52:489-505. [PMID: 39668663 DOI: 10.1177/01926233241300314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
Adeno-associated virus (AAV) gene therapy vectors are an accepted platform for treating severe neurological diseases. Test article (TA)-related and procedure-related neuropathological effects following administration of AAV-based vectors are observed in the central nervous system (CNS) and peripheral nervous system (PNS). Leukocyte accumulation (mononuclear cell infiltration > inflammation) may occur in brain, spinal cord, spinal nerve roots (SNRs), sensory and autonomic ganglia, and rarely nerves. Leukocyte accumulation may be associated with neuron necrosis (sensory ganglia > CNS) and/or glial changes (microgliosis and/or astrocytosis in the CNS, increased satellite glial cellularity in ganglia and/or Schwann cellularity in nerves). Axonal degeneration secondary to neuronal injury may occur in the SNR (dorsal > ventral), spinal cord (dorsal and occasionally lateral funiculi), and brainstem centrally and in nerves peripherally. Patterns of AAV-associated microscopic findings in the CNS and PNS differ for TAs administered into brain parenchyma (where tissue at the injection site is affected most) versus TAs delivered into cerebrospinal fluid (CSF) or systemically (which primarily impacts sensory ganglion neurons and their processes in SNR and spinal cord). Changes related to the TA and procedure may overlap. While often interpreted as adverse, AAV-associated neuronal necrosis and axonal degeneration of limited severity generally do not preclude clinical testing.
Collapse
Affiliation(s)
| | - Elizabeth Buza
- University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Joan Wicks
- Spark Therapeutics, Philadelphia, Pennsylvania, USA
| | | | | |
Collapse
|
3
|
Artemyev V, Gubaeva A, Paremskaia AI, Dzhioeva AA, Deviatkin A, Feoktistova SG, Mityaeva O, Volchkov PY. Synthetic Promoters in Gene Therapy: Design Approaches, Features and Applications. Cells 2024; 13:1963. [PMID: 39682712 DOI: 10.3390/cells13231963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/22/2024] [Accepted: 11/24/2024] [Indexed: 12/18/2024] Open
Abstract
Gene therapy is a promising approach to the treatment of various inherited diseases, but its development is complicated by a number of limitations of the natural promoters used. The currently used strong ubiquitous natural promoters do not allow for the specificity of expression, while natural tissue-specific promoters have lowactivity. These limitations of natural promoters can be addressed by creating new synthetic promoters that achieve high levels of tissue-specific target gene expression. This review discusses recent advances in the development of synthetic promoters that provide a more precise regulation of gene expression. Approaches to the design of synthetic promoters are reviewed, including manual design and bioinformatic methods using machine learning. Examples of successful applications of synthetic promoters in the therapy of hereditary diseases and cancer are presented, as well as prospects for their clinical use.
Collapse
Affiliation(s)
- Valentin Artemyev
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592 Moscow, Russia
| | - Anna Gubaeva
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia
| | - Anastasiia Iu Paremskaia
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia
| | - Amina A Dzhioeva
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592 Moscow, Russia
| | - Andrei Deviatkin
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia
| | - Sofya G Feoktistova
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia
| | - Olga Mityaeva
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592 Moscow, Russia
- Faculty of Fundamental Medicine, Moscow State University, Lomonosovsky Pr., 27, 119991 Moscow, Russia
| | - Pavel Yu Volchkov
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia
- Faculty of Fundamental Medicine, Moscow State University, Lomonosovsky Pr., 27, 119991 Moscow, Russia
- Moscow Clinical Scientific Center N.A. A.S. Loginov, 111123 Moscow, Russia
| |
Collapse
|
4
|
Dooley J, Hughes JG, Needham EJ, Palios KA, Liston A. The potential of gene delivery for the treatment of traumatic brain injury. J Neuroinflammation 2024; 21:183. [PMID: 39069631 DOI: 10.1186/s12974-024-03156-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 06/17/2024] [Indexed: 07/30/2024] Open
Abstract
Therapeutics for traumatic brains injuries constitute a global unmet medical need. Despite the advances in neurocritical care, which have dramatically improved the survival rate for the ~ 70 million patients annually, few treatments have been developed to counter the long-term neuroinflammatory processes and accompanying cognitive impairments, frequent among patients. This review looks at gene delivery as a potential therapeutic development avenue for traumatic brain injury. We discuss the capacity of gene delivery to function in traumatic brain injury, by producing beneficial biologics within the brain. Gene delivery modalities, promising vectors and key delivery routes are discussed, along with the pathways that biological cargos could target to improve long-term outcomes for patients. Coupling blood-brain barrier crossing with sustained local production, gene delivery has the potential to convert proteins with useful biological properties, but poor pharmacodynamics, into effective therapeutics. Finally, we review the limitations and health economics of traumatic brain injury, and whether future gene delivery approaches will be viable for patients and health care systems.
Collapse
Affiliation(s)
- James Dooley
- Department of Pathology, University of Cambridge, Cambridge, UK.
| | - Jasmine G Hughes
- Department of Pathology, University of Cambridge, Cambridge, UK
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, UK
| | - Edward J Needham
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, UK
| | | | - Adrian Liston
- Department of Pathology, University of Cambridge, Cambridge, UK
| |
Collapse
|
5
|
Griffin H, Hanson J, Phelan KD, Baldini G. MC4R Localizes at Excitatory Postsynaptic and Peri-Postsynaptic Sites of Hypothalamic Neurons in Primary Culture. Cells 2024; 13:1235. [PMID: 39120267 PMCID: PMC11311852 DOI: 10.3390/cells13151235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 07/13/2024] [Accepted: 07/18/2024] [Indexed: 08/10/2024] Open
Abstract
The melanocortin-4 receptor (MC4R) is a G protein-coupled receptor (GPCR) that is expressed in several brain locations encompassing the hypothalamus and the brainstem, where the receptor controls several body functions, including metabolism. In a well-defined pathway to decrease appetite, hypothalamic proopiomelanocortin (POMC) neurons localized in the arcuate nucleus (Arc) project to MC4R neurons in the paraventricular nuclei (PVN) to release the natural MC4R agonist α-melanocyte-stimulating hormone (α-MSH). Arc neurons also project excitatory glutamatergic fibers to the MC4R neurons in the PVN for a fast synaptic transmission to regulate a satiety pathway potentiated by α-MSH. By using super-resolution microscopy, we found that in hypothalamic neurons in a primary culture, postsynaptic density protein 95 (PSD95) colocalizes with GluN1, a subunit of the ionotropic N-methyl-D-aspartate receptor (NMDAR). Thus, hypothalamic neurons form excitatory postsynaptic specializations. To study the MC4R distribution at these sites, tagged HA-MC4R under the synapsin promoter was expressed in neurons by adeno-associated virus (AAV) gene transduction. HA-MC4R immunofluorescence peaked at the center and in proximity to the PSD95- and NMDAR-expressing sites. These data provide morphological evidence that MC4R localizes together with glutamate receptors at postsynaptic and peri-postsynaptic sites.
Collapse
Affiliation(s)
- Haven Griffin
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (H.G.); (J.H.)
| | - Jude Hanson
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (H.G.); (J.H.)
| | - Kevin D. Phelan
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Giulia Baldini
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (H.G.); (J.H.)
| |
Collapse
|
6
|
Trofimova AM, Amakhin DV, Postnikova TY, Tiselko VS, Alekseev A, Podoliak E, Gordeliy VI, Chizhov AV, Zaitsev AV. Light-Driven Sodium Pump as a Potential Tool for the Control of Seizures in Epilepsy. Mol Neurobiol 2024; 61:4691-4704. [PMID: 38114761 DOI: 10.1007/s12035-023-03865-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 12/06/2023] [Indexed: 12/21/2023]
Abstract
The marine flavobacterium Krokinobactereikastus light-driven sodium pump (KR2) generates an outward sodium ion current under 530 nm light stimulation, representing a promising optogenetic tool for seizure control. However, the specifics of KR2 application to suppress epileptic activity have not yet been addressed. In the present study, we investigated the possibility of KR2 photostimulation to suppress epileptiform activity in mouse brain slices using the 4-aminopyrindine (4-AP) model. We injected the adeno-associated viral vector (AAV-PHP.eB-hSyn-KR2-YFP) containing the KR2 sodium pump gene enhanced with appropriate trafficking tags. KR2 expression was observed in the lateral entorhinal cortex and CA1 hippocampus. Using whole-cell patch clamp in mouse brain slices, we show that KR2, when stimulated with LED light, induces a substantial hyperpolarization of entorhinal neurons. However, continuous photostimulation of KR2 does not interrupt ictal discharges in mouse entorhinal cortex slices induced by a solution containing 4-AP. KR2-induced hyperpolarization strongly activates neuronal HCN channels. Consequently, turning off photostimulation resulted in HCN channel-mediated rebound depolarization accompanied by a transient increase in spontaneous network activity. Using low-frequency pulsed photostimulation, we induced the generation of short HCN channel-mediated discharges that occurred in response to the light stimulus being turned off; these discharges reliably interrupt ictal activity. Thus, low-frequency pulsed photostimulation of KR2 can be considered as a potential tool for controlling epileptic seizures.
Collapse
Affiliation(s)
- Alina M Trofimova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Saint Petersburg, Russia
| | - Dmitry V Amakhin
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Saint Petersburg, Russia
| | - Tatyana Y Postnikova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Saint Petersburg, Russia
| | - Vasilii S Tiselko
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Saint Petersburg, Russia
| | - Alexey Alekseev
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology (National Research University), Dolgoprudny, Russia
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
| | - Elizaveta Podoliak
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology (National Research University), Dolgoprudny, Russia
- Department of Ophthalmology, Universitäts-Augenklinik Bonn, University of Bonn, Bonn, Germany
| | - Valentin I Gordeliy
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology (National Research University), Dolgoprudny, Russia
- Institut de Biologie Structurale (IBS), Université Grenoble Alpes, CEA, CNRS, Grenoble, France
| | - Anton V Chizhov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Saint Petersburg, Russia
- MathNeuro Team, Inria Centre at Université Côte d'Azur, Sophia Antipolis, France
| | - Aleksey V Zaitsev
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Saint Petersburg, Russia.
| |
Collapse
|
7
|
Gao Z. Strategies for enhanced gene delivery to the central nervous system. NANOSCALE ADVANCES 2024; 6:3009-3028. [PMID: 38868835 PMCID: PMC11166101 DOI: 10.1039/d3na01125a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 04/12/2024] [Indexed: 06/14/2024]
Abstract
The delivery of genes to the central nervous system (CNS) has been a persistent challenge due to various biological barriers. The blood-brain barrier (BBB), in particular, hampers the access of systemically injected drugs to parenchymal cells, allowing only a minimal percentage (<1%) to pass through. Recent scientific insights highlight the crucial role of the extracellular space (ECS) in governing drug diffusion. Taking into account advancements in vectors, techniques, and knowledge, the discussion will center on the most notable vectors utilized for gene delivery to the CNS. This review will explore the influence of the ECS - a dynamically regulated barrier-on drug diffusion. Furthermore, we will underscore the significance of employing remote-control technologies to facilitate BBB traversal and modulate the ECS. Given the rapid progress in gene editing, our discussion will also encompass the latest advances focused on delivering therapeutic editing in vivo to the CNS tissue. In the end, a brief summary on the impact of Artificial Intelligence (AI)/Machine Learning (ML), ultrasmall, soft endovascular robots, and high-resolution endovascular cameras on improving the gene delivery to the CNS will be provided.
Collapse
Affiliation(s)
- Zhenghong Gao
- Mechanical Engineering, The University of Texas at Dallas USA
| |
Collapse
|
8
|
Maturana CJ. Engineered compact pan-neuronal promoter from Alphaherpesvirus LAP2 enhances target gene expression in the mouse brain and reduces tropism in the liver. Gene Ther 2024; 31:335-344. [PMID: 38012300 PMCID: PMC11090813 DOI: 10.1038/s41434-023-00430-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 10/29/2023] [Accepted: 11/09/2023] [Indexed: 11/29/2023]
Abstract
Small promoters capable of driving potent neuron-restricted gene expression are required to support successful brain circuitry and clinical gene therapy studies. However, converting large promoters into functional MiniPromoters, which can be used in vectors with limited capacity, remains challenging. In this study, we describe the generation of a novel version of alphaherpesvirus latency-associated promoter 2 (LAP2), which facilitates precise transgene expression exclusively in the neurons of the mouse brain while minimizing undesired targeting in peripheral tissues. Additionally, we aimed to create a compact neural promoter to facilitate packaging of larger transgenes. Our results revealed that MiniLAP2 (278 bp) drives potent transgene expression in all neurons in the mouse brain, with little to no expression in glial cells. In contrast to the native promoter, MiniLAP2 reduced tropism in the spinal cord and liver. No expression was detected in the kidney or skeletal muscle. In summary, we developed a minimal pan-neuronal promoter that drives specific and robust transgene expression in the mouse brain when delivered intravenously via AAV-PHP.eB vector. The use of this novel MiniPromoter may broaden the range of deliverable therapeutics and improve their safety and efficacy by minimizing the potential for off-target effects.
Collapse
Affiliation(s)
- Carola J Maturana
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
9
|
Vacca O, Zarrouki F, Izabelle C, Belmaati Cherkaoui M, Rendon A, Dalkara D, Vaillend C. AAV-Mediated Restoration of Dystrophin-Dp71 in the Brain of Dp71-Null Mice: Molecular, Cellular and Behavioral Outcomes. Cells 2024; 13:718. [PMID: 38667332 PMCID: PMC11049308 DOI: 10.3390/cells13080718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/05/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
A deficiency in the shortest dystrophin-gene product, Dp71, is a pivotal aggravating factor for intellectual disabilities in Duchenne muscular dystrophy (DMD). Recent advances in preclinical research have achieved some success in compensating both muscle and brain dysfunctions associated with DMD, notably using exon skipping strategies. However, this has not been studied for distal mutations in the DMD gene leading to Dp71 loss. In this study, we aimed to restore brain Dp71 expression in the Dp71-null transgenic mouse using an adeno-associated virus (AAV) administrated either by intracardiac injections at P4 (ICP4) or by bilateral intracerebroventricular (ICV) injections in adults. ICP4 delivery of the AAV9-Dp71 vector enabled the expression of 2 to 14% of brain Dp71, while ICV delivery enabled the overexpression of Dp71 in the hippocampus and cortex of adult mice, with anecdotal expression in the cerebellum. The restoration of Dp71 was mostly located in the glial endfeet that surround capillaries, and it was associated with partial localization of Dp71-associated proteins, α1-syntrophin and AQP4 water channels, suggesting proper restoration of a scaffold of proteins involved in blood-brain barrier function and water homeostasis. However, this did not result in significant improvements in behavioral disturbances displayed by Dp71-null mice. The potential and limitations of this AAV-mediated strategy are discussed. This proof-of-concept study identifies key molecular markers to estimate the efficiencies of Dp71 rescue strategies and opens new avenues for enhancing gene therapy targeting cognitive disorders associated with a subgroup of severely affected DMD patients.
Collapse
Affiliation(s)
- Ophélie Vacca
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400 Saclay, France (M.B.C.)
| | - Faouzi Zarrouki
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400 Saclay, France (M.B.C.)
| | - Charlotte Izabelle
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400 Saclay, France (M.B.C.)
| | - Mehdi Belmaati Cherkaoui
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400 Saclay, France (M.B.C.)
| | - Alvaro Rendon
- Department of Therapeutics, Sorbonne University, Institut de la Vision, 75012 Paris, France; (A.R.)
| | - Deniz Dalkara
- Department of Therapeutics, Sorbonne University, Institut de la Vision, 75012 Paris, France; (A.R.)
| | - Cyrille Vaillend
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400 Saclay, France (M.B.C.)
| |
Collapse
|
10
|
Bacman SR, Barrera-Paez JD, Pinto M, Van Booven D, Stewart JB, Griswold AJ, Moraes CT. mitoTALEN reduces the mutant mtDNA load in neurons. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102132. [PMID: 38404505 PMCID: PMC10883830 DOI: 10.1016/j.omtn.2024.102132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 01/29/2024] [Indexed: 02/27/2024]
Abstract
Mutations within mtDNA frequently give rise to severe encephalopathies. Given that a majority of these mtDNA defects exist in a heteroplasmic state, we harnessed the precision of mitochondrial-targeted TALEN (mitoTALEN) to selectively eliminate mutant mtDNA within the CNS of a murine model harboring a heteroplasmic mutation in the mitochondrial tRNA alanine gene (m.5024C>T). This targeted approach was accomplished by the use of AAV-PHP.eB and a neuron-specific synapsin promoter for effective neuronal delivery and expression of mitoTALEN. We found that most CNS regions were effectively transduced and showed a significant reduction in mutant mtDNA. This reduction was accompanied by an increase in mitochondrial tRNA alanine levels, which are drastically reduced by the m.5024C>T mutation. These results showed that mitochondrial-targeted gene editing can be effective in reducing CNS-mutant mtDNA in vivo, paving the way for clinical trials in patients with mitochondrial encephalopathies.
Collapse
Affiliation(s)
- Sandra R. Bacman
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jose Domingo Barrera-Paez
- Graduate Program in Human Genetics and Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Milena Pinto
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Derek Van Booven
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - James B. Stewart
- Biosciences Institute, Faculty of Medical Sciences, Wellcome Centre for Mitochondrial Research, Newcastle University, Newcastle upon Tyne, UK
| | - Anthony J. Griswold
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Carlos T. Moraes
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
11
|
Kolesnik VV, Nurtdinov RF, Oloruntimehin ES, Karabelsky AV, Malogolovkin AS. Optimization strategies and advances in the research and development of AAV-based gene therapy to deliver large transgenes. Clin Transl Med 2024; 14:e1607. [PMID: 38488469 PMCID: PMC10941601 DOI: 10.1002/ctm2.1607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 02/07/2024] [Accepted: 02/15/2024] [Indexed: 03/18/2024] Open
Abstract
Adeno-associated virus (AAV)-based therapies are recognized as one of the most potent next-generation treatments for inherited and genetic diseases. However, several biological and technological aspects of AAV vectors remain a critical issue for their widespread clinical application. Among them, the limited capacity of the AAV genome significantly hinders the development of AAV-based gene therapy. In this context, genetically modified transgenes compatible with AAV are opening up new opportunities for unlimited gene therapies for many genetic disorders. Recent advances in de novo protein design and remodelling are paving the way for new, more efficient and targeted gene therapeutics. Using computational and genetic tools, AAV expression cassette and transgenic DNA can be split, miniaturized, shuffled or created from scratch to mediate efficient gene transfer into targeted cells. In this review, we highlight recent advances in AAV-based gene therapy with a focus on its use in translational research. We summarize recent research and development in gene therapy, with an emphasis on large transgenes (>4.8 kb) and optimizing strategies applied by biomedical companies in the research pipeline. We critically discuss the prospects for AAV-based treatment and some emerging challenges. We anticipate that the continued development of novel computational tools will lead to rapid advances in basic gene therapy research and translational studies.
Collapse
Affiliation(s)
- Valeria V. Kolesnik
- Martsinovsky Institute of Medical ParasitologyTropical and Vector‐Borne Diseases, Sechenov UniversityMoscowRussia
| | - Ruslan F. Nurtdinov
- Martsinovsky Institute of Medical ParasitologyTropical and Vector‐Borne Diseases, Sechenov UniversityMoscowRussia
| | - Ezekiel Sola Oloruntimehin
- Martsinovsky Institute of Medical ParasitologyTropical and Vector‐Borne Diseases, Sechenov UniversityMoscowRussia
| | | | - Alexander S. Malogolovkin
- Martsinovsky Institute of Medical ParasitologyTropical and Vector‐Borne Diseases, Sechenov UniversityMoscowRussia
- Center for Translational MedicineSirius University of Science and TechnologySochiRussia
| |
Collapse
|
12
|
van der Veen I, Heredero Berzal A, Koster C, ten Asbroek ALMA, Bergen AA, Boon CJF. The Road towards Gene Therapy for X-Linked Juvenile Retinoschisis: A Systematic Review of Preclinical Gene Therapy in Cell-Based and Rodent Models of XLRS. Int J Mol Sci 2024; 25:1267. [PMID: 38279267 PMCID: PMC10816913 DOI: 10.3390/ijms25021267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 01/28/2024] Open
Abstract
X-linked juvenile retinoschisis (XLRS) is an early-onset progressive inherited retinopathy affecting males. It is characterized by abnormalities in the macula, with formation of cystoid retinal cavities, frequently accompanied by splitting of the retinal layers, impaired synaptic transmission of visual signals, and associated loss of visual acuity. XLRS is caused by loss-of-function mutations in the retinoschisin gene located on the X chromosome (RS1, MIM 30083). While proof-of-concept studies for gene augmentation therapy have been promising in in vitro and rodent models, clinical trials in XLRS patients have not been successful thus far. We performed a systematic literature investigation using search strings related to XLRS and gene therapy in in vivo and in vitro models. Three rounds of screening (title/abstract, full text and qualitative) were performed by two independent reviewers until consensus was reached. Characteristics related to study design and intervention were extracted from all studies. Results were divided into studies using (1) viral and (2) non-viral therapies. All in vivo rodent studies that used viral vectors were assessed for quality and risk of bias using the SYRCLE's risk-of-bias tool. Studies using alternative and non-viral delivery techniques, either in vivo or in vitro, were extracted and reviewed qualitatively, given the diverse and dispersed nature of the information. For in-depth analysis of in vivo studies using viral vectors, outcome data for optical coherence tomography (OCT), immunohistopathology and electroretinography (ERG) were extracted. Meta-analyses were performed on the effect of recombinant adeno-associated viral vector (AAV)-mediated gene augmentation therapies on a- and b-wave amplitude as well as the ratio between b- and a-wave amplitudes (b/a-ratio) extracted from ERG data. Subgroup analyses and meta-regression were performed for model, dose, age at injection, follow-up time point and delivery method. Between-study heterogeneity was assessed with a Chi-square test of homogeneity (I2). We identified 25 studies that target RS1 and met our search string. A total of 19 of these studies reported rodent viral methods in vivo. Six of the 25 studies used non-viral or alternative delivery methods, either in vitro or in vivo. Of these, five studies described non-viral methods and one study described an alternative delivery method. The 19 aforementioned in vivo studies were assessed for risk of bias and quality assessments and showed inconsistency in reporting. This resulted in an unclear risk of bias in most included studies. All 19 studies used AAVs to deliver intact human or murine RS1 in rodent models for XLRS. Meta-analyses of a-wave amplitude, b-wave amplitude, and b/a-ratio showed that, overall, AAV-mediated gene augmentation therapy significantly ameliorated the disease phenotype on these parameters. Subgroup analyses and meta-regression showed significant correlations between b-wave amplitude effect size and dose, although between-study heterogeneity was high. This systematic review reiterates the high potential for gene therapy in XLRS, while highlighting the importance of careful preclinical study design and reporting. The establishment of a systematic approach in these studies is essential to effectively translate this knowledge into novel and improved treatment alternatives.
Collapse
Affiliation(s)
- Isa van der Veen
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (I.v.d.V.); (A.H.B.); (C.K.); (A.A.B.)
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
| | - Andrea Heredero Berzal
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (I.v.d.V.); (A.H.B.); (C.K.); (A.A.B.)
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
| | - Céline Koster
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (I.v.d.V.); (A.H.B.); (C.K.); (A.A.B.)
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
| | - Anneloor L. M. A. ten Asbroek
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
| | - Arthur A. Bergen
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (I.v.d.V.); (A.H.B.); (C.K.); (A.A.B.)
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
| | - Camiel J. F. Boon
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (I.v.d.V.); (A.H.B.); (C.K.); (A.A.B.)
- Department of Ophthalmology, Leiden University Medical Center, Leiden University, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
13
|
Brunet de Courssou JB, Deiva K. Les thérapies géniques en neurologie. PRATIQUE NEUROLOGIQUE - FMC 2023; 14:208-224. [DOI: 10.1016/j.praneu.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
14
|
Li L, Vasan L, Kartono B, Clifford K, Attarpour A, Sharma R, Mandrozos M, Kim A, Zhao W, Belotserkovsky A, Verkuyl C, Schmitt-Ulms G. Advances in Recombinant Adeno-Associated Virus Vectors for Neurodegenerative Diseases. Biomedicines 2023; 11:2725. [PMID: 37893099 PMCID: PMC10603849 DOI: 10.3390/biomedicines11102725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 09/29/2023] [Accepted: 10/03/2023] [Indexed: 10/29/2023] Open
Abstract
Recombinant adeno-associated virus (rAAV) vectors are gene therapy delivery tools that offer a promising platform for the treatment of neurodegenerative diseases. Keeping up with developments in this fast-moving area of research is a challenge. This review was thus written with the intention to introduce this field of study to those who are new to it and direct others who are struggling to stay abreast of the literature towards notable recent studies. In ten sections, we briefly highlight early milestones within this field and its first clinical success stories. We showcase current clinical trials, which focus on gene replacement, gene augmentation, or gene suppression strategies. Next, we discuss ongoing efforts to improve the tropism of rAAV vectors for brain applications and introduce pre-clinical research directed toward harnessing rAAV vectors for gene editing applications. Subsequently, we present common genetic elements coded by the single-stranded DNA of rAAV vectors, their so-called payloads. Our focus is on recent advances that are bound to increase treatment efficacies. As needed, we included studies outside the neurodegenerative disease field that showcased improved pre-clinical designs of all-in-one rAAV vectors for gene editing applications. Finally, we discuss risks associated with off-target effects and inadvertent immunogenicity that these technologies harbor as well as the mitigation strategies available to date to make their application safer.
Collapse
Affiliation(s)
- Leyao Li
- Department of Biochemistry, University of Toronto, Medical Sciences Building, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, 6th Floor, 60 Leonard Avenue, Toronto, ON M5T 0S8, Canada
| | - Lakshmy Vasan
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| | - Bryan Kartono
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, 6th Floor, 60 Leonard Avenue, Toronto, ON M5T 0S8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| | - Kevan Clifford
- Institute of Medical Science, University of Toronto, Medical Sciences Building, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
- Centre for Addiction and Mental Health (CAMH), 250 College St., Toronto, ON M5T 1R8, Canada
| | - Ahmadreza Attarpour
- Department of Medical Biophysics, University of Toronto, 101 College St., Toronto, ON M5G 1L7, Canada
| | - Raghav Sharma
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, 6th Floor, 60 Leonard Avenue, Toronto, ON M5T 0S8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| | - Matthew Mandrozos
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| | - Ain Kim
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, 6th Floor, 60 Leonard Avenue, Toronto, ON M5T 0S8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| | - Wenda Zhao
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, 6th Floor, 60 Leonard Avenue, Toronto, ON M5T 0S8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| | - Ari Belotserkovsky
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, 6th Floor, 60 Leonard Avenue, Toronto, ON M5T 0S8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| | - Claire Verkuyl
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, 6th Floor, 60 Leonard Avenue, Toronto, ON M5T 0S8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| | - Gerold Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, 6th Floor, 60 Leonard Avenue, Toronto, ON M5T 0S8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
15
|
Tosolini AP, Smith GM. Editorial: Gene therapy for the central and peripheral nervous system, volume II. Front Mol Neurosci 2023; 16:1258458. [PMID: 37593465 PMCID: PMC10431961 DOI: 10.3389/fnmol.2023.1258458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 07/14/2023] [Indexed: 08/19/2023] Open
Affiliation(s)
- Andrew P. Tosolini
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, United Kingdom
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD, Australia
| | - George M. Smith
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
16
|
Timalsina B, Choi HJ, Moon IS. N-Acetylglucosamine Kinase-Small Nuclear Ribonucleoprotein Polypeptide N Interaction Promotes Axodendritic Branching in Neurons via Dynein-Mediated Microtubule Transport. Int J Mol Sci 2023; 24:11672. [PMID: 37511433 PMCID: PMC10380243 DOI: 10.3390/ijms241411672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
N-acetylglucosamine kinase (NAGK) has been identified as an anchor protein that facilitates neurodevelopment with its non-canonical structural role. Similarly, small nuclear ribonucleoprotein polypeptide N (SNRPN) regulates neurodevelopment and cognitive ability. In our previous study, we revealed the interaction between NAGK and SNRPN in the neuron. However, the precise role in neurodevelopment is elusive. In this study, we investigate the role of NAGK and SNRPN in the axodendritic development of neurons. NAGK and SNRPN interaction is significantly increased in neurons at the crucial stages of neurodevelopment. Furthermore, overexpression of the NAGK and SNRPN proteins increases axodendritic branching and neuronal complexity, whereas the knockdown inhibits neurodevelopment. We also observe the interaction of NAGK and SNRPN with the dynein light-chain roadblock type 1 (DYNLRB1) protein variably during neurodevelopment, revealing the microtubule-associated delivery of the complex. Interestingly, NAGK and SNRPN proteins rescued impaired axodendritic development in an SNRPN depletion model of Prader-Willi syndrome (PWS) patient-derived induced pluripotent stem cell neurons. Taken together, these findings are crucial in developing therapeutic approaches for neurodegenerative diseases.
Collapse
Affiliation(s)
- Binod Timalsina
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea
| | - Ho Jin Choi
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea
| | - Il Soo Moon
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea
| |
Collapse
|
17
|
Hughes MP, Nelvagal HR, Coombe-Tennant O, Smith D, Smith C, Massaro G, Poupon-Bejuit L, Platt FM, Rahim AA. A Novel Small NPC1 Promoter Enhances AAV-Mediated Gene Therapy in Mouse Models of Niemann-Pick Type C1 Disease. Cells 2023; 12:1619. [PMID: 37371089 PMCID: PMC10296851 DOI: 10.3390/cells12121619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/22/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Niemann-Pick disease type C1 (NP-C) is a prematurely lethal genetic lysosomal storage disorder with neurological and visceral pathology resulting from mutations in the NPC1 gene encoding the lysosomal transmembrane protein NPC1. There is currently no cure for NP-C, and the only disease modifying treatment, miglustat, slows disease progression but does not significantly attenuate neurological symptoms. AAV-mediated gene therapy is an attractive option for NP-C, but due to the large size of the human NPC1 gene, there may be packaging and truncation issues during vector manufacturing. One option is to reduce the size of DNA regulatory elements that are essential for gene expression, such as the promoter sequence. Here, we describe a novel small truncated endogenous NPC1 promoter that leads to high gene expression both in vitro and in vivo and compare its efficacy to other commonly used promoters. Following neonatal intracerebroventricular (ICV) injection into the CNS, this novel promoter provided optimal therapeutic efficacy compared to all other promoters including increased survival, improved behavioural phenotypes, and attenuated neuropathology in mouse models of NP-C. Taken together, we propose that this novel promoter can be extremely efficient in designing an optimised AAV9 vector for gene therapy for NP-C.
Collapse
Affiliation(s)
- Michael Paul Hughes
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK (H.R.N.); (O.C.-T.); (G.M.)
| | - Hemanth Ramesh Nelvagal
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK (H.R.N.); (O.C.-T.); (G.M.)
| | - Oliver Coombe-Tennant
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK (H.R.N.); (O.C.-T.); (G.M.)
| | - Dave Smith
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK; (D.S.); (C.S.); (F.M.P.)
| | - Claire Smith
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK; (D.S.); (C.S.); (F.M.P.)
| | - Giulia Massaro
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK (H.R.N.); (O.C.-T.); (G.M.)
| | - Laura Poupon-Bejuit
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK (H.R.N.); (O.C.-T.); (G.M.)
| | - Frances Mary Platt
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK; (D.S.); (C.S.); (F.M.P.)
| | - Ahad Abdul Rahim
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK (H.R.N.); (O.C.-T.); (G.M.)
| |
Collapse
|
18
|
Chai S, Wakefield L, Norgard M, Li B, Enicks D, Marks DL, Grompe M. Strong ubiquitous micro-promoters for recombinant adeno-associated viral vectors. Mol Ther Methods Clin Dev 2023; 29:504-512. [PMID: 37287749 PMCID: PMC10241652 DOI: 10.1016/j.omtm.2023.05.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 05/12/2023] [Indexed: 06/09/2023]
Abstract
Significant progress has been made in developing recombinant adeno-associated virus (rAAV) for clinical gene therapy. While rAAV is a versatile gene delivery platform, its packaging limit of 4.7 kb limits the diseases it can target. Here, we report two unusually small promoters that enable the expression of larger transgenes than standard promoters. These micro-promoters are only 84 (MP-84) and 135 bp (MP-135) in size but have activity in most cells and tissues comparable to the CAG promoter, the strongest ubiquitous promoter to date. MP-84- and MP-135-based rAAV constructs displayed robust activity in cultured cells from the three different germ-layer lineages. In addition, reporter gene expression was documented in human primary hepatocytes and pancreatic islets and in multiple mouse tissues in vivo, including brain and skeletal muscle. MP-84 and MP-135 will enable the therapeutic expression of transgenes currently too large for rAAV vectors.
Collapse
Affiliation(s)
- Sunghee Chai
- Papé Family Pediatric Research Institute, Oregon Stem Cell Center, Portland, OR, USA
| | - Leslie Wakefield
- Papé Family Pediatric Research Institute, Oregon Stem Cell Center, Portland, OR, USA
| | - Mason Norgard
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - Bin Li
- Papé Family Pediatric Research Institute, Oregon Stem Cell Center, Portland, OR, USA
| | - David Enicks
- Papé Family Pediatric Research Institute, Oregon Stem Cell Center, Portland, OR, USA
| | - Daniel L. Marks
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - Markus Grompe
- Papé Family Pediatric Research Institute, Oregon Stem Cell Center, Portland, OR, USA
| |
Collapse
|
19
|
Tian G, Cao C, Li S, Wang W, Zhang Y, Lv Y. rAAV2-Mediated Restoration of GALC in Neural Stem Cells from Krabbe Patient-Derived iPSCs. Pharmaceuticals (Basel) 2023; 16:ph16040624. [PMID: 37111381 PMCID: PMC10143348 DOI: 10.3390/ph16040624] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/28/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Krabbe disease is a rare neurodegenerative fatal disease. It is caused by deficiency of the lysosomal enzyme galactocerebrosidase (GALC), which results in progressive accumulation of galactolipid substrates in myelin-forming cells. However, there is still a lack of appropriate neural models and effective approaches for Krabbe disease. We generated induced pluripotent stem cells (iPSCs) from a Krabbe patient previously. Here, Krabbe patient-derived neural stem cells (K-NSCs) were induced from these iPSCs. By using nine kinds of recombinant adeno-associated virus (rAAV) vectors to infect K-NSCs, we found that the rAAV2 vector has high transduction efficiency for K-NSCs. Most importantly, rAAV2-GALC rescued GALC enzymatic activity in K-NSCs. Our findings not only establish a novel patient NSC model for Krabbe disease, but also firstly indicate the potential of rAAV2-mediated gene therapy for this devastating disease.
Collapse
Affiliation(s)
- Guoshuai Tian
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Chunyu Cao
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443000, China
| | - Shuyue Li
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443000, China
| | - Wei Wang
- Department of Neurology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Ye Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Yafeng Lv
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443000, China
| |
Collapse
|
20
|
Zoabi S, Andreyanov M, Heinrich R, Ron S, Carmi I, Gutfreund Y, Berlin S. A custom-made AAV1 variant (AAV1-T593K) enables efficient transduction of Japanese quail neurons in vitro and in vivo. Commun Biol 2023; 6:337. [PMID: 36977781 PMCID: PMC10050006 DOI: 10.1038/s42003-023-04712-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
The widespread use of rodents in neuroscience has prompted the development of optimized viral variants for transduction of brain cells, in vivo. However, many of the viruses developed are less efficient in other model organisms, with birds being among the most resistant to transduction by current viral tools. Resultantly, the use of genetically-encoded tools and methods in avian species is markedly lower than in rodents; likely holding the field back. We sought to bridge this gap by developing custom viruses towards the transduction of brain cells of the Japanese quail. We first develop a protocol for culturing primary neurons and glia from quail embryos, followed by characterization of cultures via immunostaining, single cell mRNA sequencing, patch clamp electrophysiology and calcium imaging. We then leveraged the cultures for the rapid screening of various viruses, only to find that all yielded poor to no infection of cells in vitro. However, few infected neurons were obtained by AAV1 and AAV2. Scrutiny of the sequence of the AAV receptor found in quails led us to rationally design a custom-made AAV variant (AAV1-T593K; AAV1*) that exhibits improved transduction efficiencies in vitro and in vivo (14- and five-fold, respectively). Together, we present unique culturing method, transcriptomic profiles of quail's brain cells and a custom-tailored AAV1 for transduction of quail neurons in vitro and in vivo.
Collapse
Affiliation(s)
- Shaden Zoabi
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion- Israel Institute of Technology, Haifa, Israel
| | - Michael Andreyanov
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion- Israel Institute of Technology, Haifa, Israel
| | - Ronit Heinrich
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion- Israel Institute of Technology, Haifa, Israel
| | - Shaked Ron
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion- Israel Institute of Technology, Haifa, Israel
| | - Ido Carmi
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion- Israel Institute of Technology, Haifa, Israel
| | - Yoram Gutfreund
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion- Israel Institute of Technology, Haifa, Israel
| | - Shai Berlin
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion- Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
21
|
Tsuchimochi R, Yamagami K, Kubo N, Amimoto N, Raudzus F, Samata B, Kikuchi T, Doi D, Yoshimoto K, Mihara A, Takahashi J. Viral delivery of L1CAM promotes axonal extensions by embryonic cerebral grafts in mouse brain. Stem Cell Reports 2023; 18:899-914. [PMID: 36963389 PMCID: PMC10147836 DOI: 10.1016/j.stemcr.2023.02.012] [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: 06/21/2022] [Revised: 02/24/2023] [Accepted: 02/25/2023] [Indexed: 03/26/2023] Open
Abstract
Cell replacement therapy is expected as a new and more radical treatment against brain damage. We previously reported that transplanted human cerebral organoids extend their axons along the corticospinal tract in rodent brains. The axons reached the spinal cord but were still sparse. Therefore, this study optimized the host brain environment by the adeno-associated virus (AAV)-mediated expression of axon guidance proteins in mouse brain. Among netrin-1, SEMA3, and L1CAM, only L1CAM significantly promoted the axonal extension of mouse embryonic brain tissue-derived grafts. L1CAM was also expressed by donor neurons, and this promotion was exerted in a haptotactic manner by their homophilic binding. Primary cortical neurons cocultured on L1CAM-expressing HEK-293 cells supported this mechanism. These results suggest that optimizing the host environment by the AAV-mediated expression of axon guidance molecules enhances the effect of cell replacement therapy.
Collapse
Affiliation(s)
- Ryosuke Tsuchimochi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan; Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Keitaro Yamagami
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan; Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Naoko Kubo
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Naoya Amimoto
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Fabian Raudzus
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Bumpei Samata
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Tetsuhiro Kikuchi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Daisuke Doi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Koji Yoshimoto
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Aya Mihara
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Jun Takahashi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan; Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.
| |
Collapse
|
22
|
Menon V, Cerri D, Lee B, Yuan R, Lee SH, Shih YYI. Optogenetic stimulation of anterior insular cortex neurons in male rats reveals causal mechanisms underlying suppression of the default mode network by the salience network. Nat Commun 2023; 14:866. [PMID: 36797303 PMCID: PMC9935890 DOI: 10.1038/s41467-023-36616-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 02/09/2023] [Indexed: 02/18/2023] Open
Abstract
The salience network (SN) and default mode network (DMN) play a crucial role in cognitive function. The SN, anchored in the anterior insular cortex (AI), has been hypothesized to modulate DMN activity during stimulus-driven cognition. However, the causal neural mechanisms underlying changes in DMN activity and its functional connectivity with the SN are poorly understood. Here we combine feedforward optogenetic stimulation with fMRI and computational modeling to dissect the causal role of AI neurons in dynamic functional interactions between SN and DMN nodes in the male rat brain. Optogenetic stimulation of Chronos-expressing AI neurons suppressed DMN activity, and decreased AI-DMN and intra-DMN functional connectivity. Our findings demonstrate that feedforward optogenetic stimulation of AI neurons induces dynamic suppression and decoupling of the DMN and elucidates previously unknown features of rodent brain network organization. Our study advances foundational knowledge of causal mechanisms underlying dynamic cross-network interactions and brain network switching.
Collapse
Grants
- R01 MH121069 NIMH NIH HHS
- P50 HD103573 NICHD NIH HHS
- T32 AA007573 NIAAA NIH HHS
- R01 NS091236 NINDS NIH HHS
- R01 MH126518 NIMH NIH HHS
- S10 MH124745 NIMH NIH HHS
- U01 AA020023 NIAAA NIH HHS
- R01 MH111429 NIMH NIH HHS
- S10 OD026796 NIH HHS
- R01 NS086085 NINDS NIH HHS
- R01 EB022907 NIBIB NIH HHS
- P60 AA011605 NIAAA NIH HHS
- RF1 NS086085 NINDS NIH HHS
- RF1 MH117053 NIMH NIH HHS
- This work was supported in part by the National Institute of Mental Health (R01MH121069 to V.M., and R01MH126518, RF1MH117053, R01MH111429, S10MH124745 to Y.-Y.I.S.), National Institute on Alcohol Abuse and Alcoholism (P60AA011605 and U01AA020023 to Y.-Y.I.S., T32AA007573 to D.C.), National Institute of Neurological Disorders and Stroke (R01NS086085 to V.M., R01NS091236 to Y.-Y.I.S.), National Institute of Child Health and Human Development (P50HD103573 to Y.-Y.I.S.), National Institute of Biomedical Imaging and Bioengineering (R01EB022907 to V.M.), and National Institute of Health Office of the Director (S10OD026796 to Y.-Y.I.S.).
Collapse
Affiliation(s)
- Vinod Menon
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Department of Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Wu Tsai Neuroscience Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Domenic Cerri
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Byeongwook Lee
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Rui Yuan
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Sung-Ho Lee
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yen-Yu Ian Shih
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
23
|
Day-Cooney J, Dalangin R, Zhong H, Mao T. Genetically encoded fluorescent sensors for imaging neuronal dynamics in vivo. J Neurochem 2023; 164:284-308. [PMID: 35285522 PMCID: PMC11322610 DOI: 10.1111/jnc.15608] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/14/2022] [Accepted: 02/25/2022] [Indexed: 11/29/2022]
Abstract
The brain relies on many forms of dynamic activities in individual neurons, from synaptic transmission to electrical activity and intracellular signaling events. Monitoring these neuronal activities with high spatiotemporal resolution in the context of animal behavior is a necessary step to achieve a mechanistic understanding of brain function. With the rapid development and dissemination of highly optimized genetically encoded fluorescent sensors, a growing number of brain activities can now be visualized in vivo. To date, cellular calcium imaging, which has been largely used as a proxy for electrical activity, has become a mainstay in systems neuroscience. While challenges remain, voltage imaging of neural populations is now possible. In addition, it is becoming increasingly practical to image over half a dozen neurotransmitters, as well as certain intracellular signaling and metabolic activities. These new capabilities enable neuroscientists to test previously unattainable hypotheses and questions. This review summarizes recent progress in the development and delivery of genetically encoded fluorescent sensors, and highlights example applications in the context of in vivo imaging.
Collapse
Affiliation(s)
- Julian Day-Cooney
- Vollum Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Rochelin Dalangin
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, California, USA
| | - Haining Zhong
- Vollum Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Tianyi Mao
- Vollum Institute, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
24
|
Nieuwenhuis B, Laperrousaz E, Tribble JR, Verhaagen J, Fawcett JW, Martin KR, Williams PA, Osborne A. Improving adeno-associated viral (AAV) vector-mediated transgene expression in retinal ganglion cells: comparison of five promoters. Gene Ther 2023:10.1038/s41434-022-00380-z. [PMID: 36635457 DOI: 10.1038/s41434-022-00380-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 12/02/2022] [Accepted: 12/21/2022] [Indexed: 01/14/2023]
Abstract
Recombinant adeno-associated viral vectors (AAVs) are an effective system for gene transfer. AAV serotype 2 (AAV2) is commonly used to deliver transgenes to retinal ganglion cells (RGCs) via intravitreal injection. The AAV serotype however is not the only factor contributing to the effectiveness of gene therapies. Promoters influence the strength and cell-selectivity of transgene expression. This study compares five promoters designed to maximise AAV2 cargo space for gene delivery: chicken β-actin (CBA), cytomegalovirus (CMV), short CMV early enhancer/chicken β-actin/short β-globulin intron (sCAG), mouse phosphoglycerate kinase (PGK), and human synapsin (SYN). The promoters driving enhanced green fluorescent protein (eGFP) were examined in adult C57BL/6J mice eyes and tissues of the visual system. eGFP expression was strongest in the retina, optic nerves and brain when driven by the sCAG and SYN promoters. CBA, CMV, and PGK had moderate expression by comparison. The SYN promoter had almost exclusive transgene expression in RGCs. The PGK promoter had predominant expression in both RGCs and AII amacrine cells. The ubiquitous CBA, CMV, and sCAG promoters expressed eGFP in a variety of cell types across multiple retinal layers including Müller glia and astrocytes. We also found that these promoters could transduce human retina ex vivo, although expression was predominantly in glial cells due to low RGC viability. Taken together, this promoter comparison study contributes to optimising AAV-mediated transduction in the retina, and could be valuable for research in ocular disorders, particularly those with large or complex genetic cargos.
Collapse
Affiliation(s)
- Bart Nieuwenhuis
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK. .,Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK.
| | - Elise Laperrousaz
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - James R Tribble
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Joost Verhaagen
- Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, The Netherlands.,Centre for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - James W Fawcett
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Centre of Reconstructive Neuroscience, Institute of Experimental Medicine, Prague, Czech Republic
| | - Keith R Martin
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, VIC, Australia.,Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, VIC, Australia
| | - Pete A Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Andrew Osborne
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK. .,Ikarovec Ltd, The Norwich Research Park Innovation Centre, Norwich, UK.
| |
Collapse
|
25
|
Giovannelli I, Higginbottom A, Kirby J, Azzouz M, Shaw PJ. Prospects for gene replacement therapies in amyotrophic lateral sclerosis. Nat Rev Neurol 2023; 19:39-52. [PMID: 36481799 DOI: 10.1038/s41582-022-00751-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/10/2022] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating and incurable neurodegenerative disease characterized by the progressive loss of upper and lower motor neurons. ALS causes death, usually within 2-5 years of diagnosis. Riluzole, the only drug currently approved in Europe for the treatment of this condition, offers only a modest benefit, increasing survival by 3 months on average. Recent advances in our understanding of causative or disease-modifying genetic variants and in the development of genetic therapy strategies present exciting new therapeutic opportunities for ALS. In addition, the approval of adeno-associated virus-mediated delivery of functional copies of the SMN1 gene to treat spinal muscular atrophy represents an important therapeutic milestone and demonstrates the potential of gene replacement therapies for motor neuron disorders. In this Review, we describe the current landscape of genetic therapies in ALS, highlighting achievements and critical challenges. In particular, we discuss opportunities for gene replacement therapy in subgroups of people with ALS, and we describe loss-of-function mutations that are known to contribute to the pathophysiology of ALS and could represent novel targets for gene replacement therapies.
Collapse
Affiliation(s)
- Ilaria Giovannelli
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Adrian Higginbottom
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Janine Kirby
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Mimoun Azzouz
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK.
| |
Collapse
|
26
|
Kuzmin AA, Tomilin AN. Building Blocks of Artificial CRISPR-Based Systems beyond Nucleases. Int J Mol Sci 2022; 24:ijms24010397. [PMID: 36613839 PMCID: PMC9820447 DOI: 10.3390/ijms24010397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/19/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
Tools developed in the fields of genome engineering, precise gene regulation, and synthetic gene networks have an increasing number of applications. When shared with the scientific community, these tools can be used to further unlock the potential of precision medicine and tissue engineering. A large number of different genetic elements, as well as modifications, have been used to create many different systems and to validate some technical concepts. New studies have tended to optimize or improve existing elements or approaches to create complex synthetic systems, especially those based on the relatively new CRISPR technology. In order to maximize the output of newly developed approaches and to move from proof-of-principle experiments to applications in regenerative medicine, it is important to navigate efficiently through the vast number of genetic elements to choose those most suitable for specific needs. In this review, we have collected information regarding the main genetic elements and their modifications, which can be useful in different synthetic systems with an emphasis of those based on CRISPR technology. We have indicated the most suitable elements and approaches to choose or combine in planning experiments, while providing their deeper understanding, and have also stated some pitfalls that should be avoided.
Collapse
|
27
|
Lim CKW, McCallister TX, Saporito-Magriña C, McPheron GD, Krishnan R, Zeballos C MA, Powell JE, Clark LV, Perez-Pinera P, Gaj T. CRISPR base editing of cis-regulatory elements enables the perturbation of neurodegeneration-linked genes. Mol Ther 2022; 30:3619-3631. [PMID: 35965414 PMCID: PMC9734028 DOI: 10.1016/j.ymthe.2022.08.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/25/2022] [Accepted: 08/09/2022] [Indexed: 12/15/2022] Open
Abstract
CRISPR technology has demonstrated broad utility for controlling target gene expression; however, there remains a need for strategies capable of modulating expression via the precise editing of non-coding regulatory elements. Here, we demonstrate that CRISPR base editors, a class of gene-modifying proteins capable of creating single-base substitutions in DNA, can be used to perturb gene expression via their targeted mutagenesis of cis-acting sequences. Using the promoter region of the human huntingtin (HTT) gene as an initial target, we show that editing of the binding site for the transcription factor NF-κB led to a marked reduction in HTT gene expression in base-edited cell populations. We found that these gene perturbations were persistent and specific, as a transcriptome-wide RNA analysis revealed minimal off-target effects resulting from the action of the base editor protein. We further demonstrate that this base-editing platform could influence gene expression in vivo as its delivery to a mouse model of Huntington's disease led to a potent decrease in HTT mRNA in striatal neurons. Finally, to illustrate the applicability of this concept, we target the amyloid precursor protein, showing that multiplex editing of its promoter region significantly perturbed its expression. These findings demonstrate the potential for base editors to regulate target gene expression.
Collapse
Affiliation(s)
- Colin K W Lim
- Department of Bioengineering, University of Illinois, Urbana, IL 61801, USA
| | | | | | - Garrett D McPheron
- Department of Bioengineering, University of Illinois, Urbana, IL 61801, USA
| | - Ramya Krishnan
- Department of Bioengineering, University of Illinois, Urbana, IL 61801, USA
| | | | - Jackson E Powell
- Department of Bioengineering, University of Illinois, Urbana, IL 61801, USA
| | - Lindsay V Clark
- Roy J. Carver Biotechnology Center, University of Illinois, Urbana, IL 61801, USA
| | - Pablo Perez-Pinera
- Department of Bioengineering, University of Illinois, Urbana, IL 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana, IL 61801, USA; Department of Biomedical and Translational Sciences, Carle-Illinois College of Medicine, University of Illinois, Urbana, IL 61801, USA; Cancer Center at Illinois, University of Illinois, Urbana, IL 61801, USA.
| | - Thomas Gaj
- Department of Bioengineering, University of Illinois, Urbana, IL 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana, IL 61801, USA.
| |
Collapse
|
28
|
Fluorochromized tyramide-glucose oxidase as a multiplex fluorescent tyramide signal amplification system for histochemical analysis. Sci Rep 2022; 12:14807. [PMID: 36097273 PMCID: PMC9468149 DOI: 10.1038/s41598-022-19085-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/24/2022] [Indexed: 11/08/2022] Open
Abstract
Tyramide signal amplification (TSA) is a highly sensitive method for histochemical analysis. Previously, we reported a TSA system, biotinyl tyramine-glucose oxidase (BT-GO), for bright-filed imaging. Here, we develop fluorochromized tyramide-glucose oxidase (FT-GO) as a multiplex fluorescent TSA system. FT-GO involves peroxidase-catalyzed deposition of fluorochromized tyramide (FT) with hydrogen peroxide produced by enzymatic reaction between glucose and glucose oxidase. We showed that FT-GO enhanced immunofluorescence signals while maintaining low background signals. Compared with indirect immunofluorescence detections, FT-GO demonstrated a more widespread distribution of monoaminergic projection systems in mouse and marmoset brains. For multiplex labeling with FT-GO, we quenched antibody-conjugated peroxidase using sodium azide. We applied FT-GO to multiplex fluorescent in situ hybridization, and succeeded in labeling neocortical interneuron subtypes by coupling with immunofluorescence. FT-GO immunofluorescence further increased the detectability of an adeno-associated virus tracer. Given its simplicity and a staining with a high signal-to-noise ratio, FT-GO would provide a versatile platform for histochemical analysis.
Collapse
|
29
|
Rybarikova M, Almacellas Barbanoj A, Schorge S, Déglon N. CNS gene therapy: present developments and emerging trends accelerating industry-academia pathways. Hum Gene Ther 2022; 33:913-922. [PMID: 36070435 DOI: 10.1089/hum.2022.177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The recent success of first central nervous system gene therapies has reinvigorated the growing community of gene therapy researchers and strengthened the field's market position. We are witnessing an increase of clinical trials with long-term efficiency mainly for neurometabolic, neurodegenerative and neurodevelopmental diseases caused by loss-of-function mutations. The ever-expanding knowledge and accessibility to the most advanced tools allow enrichment of applications to more complex diseases. This gradually contributes towards sealing the gap between top diseases impacting current global health and those towards which gene therapy development is currently aimed. Here, we highlight innovative therapeutic approaches that have reached the clinics and outline the latest improvements of vector design and targeting. Finally, we address the pressing challenges faced by clinical trials and the direction they are heading.
Collapse
Affiliation(s)
- Margareta Rybarikova
- Lausanne University Hospital, Department of Clinical Neurosciences, Lausanne, Vaud, Switzerland.,Lausanne University Hospital, Neuroscience Research Center , Lausanne, Vaud, Switzerland;
| | - Amanda Almacellas Barbanoj
- University College London, Institute of Neurology (IoN), Department of Clinical and Experimental Epilepsy (DCEE), London, London, United Kingdom of Great Britain and Northern Ireland;
| | - Stephanie Schorge
- University College London, Institute of Neurology (IoN), Department of Clinical and Experimental Epilepsy (DCEE), London, London, United Kingdom of Great Britain and Northern Ireland;
| | - Nicole Déglon
- Lausanne University Hospital, Department of Clinical Neurosciences, Lausanne, Vaud, Switzerland.,Lausanne University Hospital, Neuroscience Research Center, Lausanne, Vaud, Switzerland;
| |
Collapse
|
30
|
Jung KM, Park KJ, Kim YM, Han JY. Efficient gene delivery into the embryonic chicken brain using neuron-specific promoters and in ovo electroporation. BMC Biotechnol 2022; 22:25. [PMID: 36056347 PMCID: PMC9440574 DOI: 10.1186/s12896-022-00756-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/29/2022] [Indexed: 11/30/2022] Open
Abstract
Background The chicken in ovo model is an attractive system to explore underlying mechanisms of neural and brain development, and it is important to develop effective genetic modification techniques that permit analyses of gene functions in vivo. Although electroporation and viral vector-mediated gene delivery techniques have been used to introduce exogenous DNA into chicken embryonic cells, transducing neurons efficiently and specifically remains challenging.
Methods In the present study, we performed a comparative study of the ubiquitous CMV promoter and three neuron-specific promoters, chicken Ca2+/calmodulin-dependent kinase (cCaMKII), chicken Nestin (cNestin), and human synapsin I. We explored the possibility of manipulating gene expression in chicken embryonic brain cells using in ovo electroporation with the selected promoters.
Results Transgene expression by two neuron-specific promoters (cCaMKII and cNestin) was preliminarily verified in vitro in cultured brain cells, and in vivo, expression levels of an EGFP transgene in brain cells by neuron-specific promoters were comparable to or higher than those of the ubiquitous CMV promoter. Overexpression of the FOXP2 gene driven by the cNestin promoter in brain cells significantly affected expression levels of target genes, CNTNAP2 and ELAVL4. Conclusion We demonstrated that exogenous DNA can be effectively introduced into neuronal cells in living embryos by in ovo electroporation with constructs containing neuron-specific promoters. In ovo electroporation offers an easier and more efficient way to manipulate gene expression during embryonic development, and this technique will be useful for neuron-targeted transgene expression. Supplementary Information The online version contains supplementary material available at 10.1186/s12896-022-00756-4.
Collapse
Affiliation(s)
- Kyung Min Jung
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Kyung Je Park
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Young Min Kim
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Jae Yong Han
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea.
| |
Collapse
|
31
|
Marrone L, Marchi PM, Azzouz M. Circumventing the packaging limit of AAV-mediated gene replacement therapy for neurological disorders. Expert Opin Biol Ther 2022; 22:1163-1176. [PMID: 34904932 DOI: 10.1080/14712598.2022.2012148] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 11/25/2021] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Gene therapy provides the exciting opportunity of a curative single treatment for devastating diseases, eradicating the need for chronic medication. Adeno-associated viruses (AAVs) are among the most attractive vector carriers for gene replacement in vivo. Yet, despite the success of recent AAV-based clinical trials, the clinical use of these vectors has been limited. For instance, the AAV packaging capacity is restricted to ~4.7 kb, making it a substantial challenge to deliver large gene products. AREAS COVERED In this review, we explore established and emerging strategies that circumvent the packaging limit of AAVs to make them effective vehicles for gene replacement therapy of monogenic disorders, with a particular focus on diseases affecting the nervous system. We report historical references, design remarks, as well as strengths and weaknesses of these approaches. We additionally discuss examples of neurological disorders for which such strategies have been attempted. EXPERT OPINION The field of AAV-gene therapy has experienced enormous advancements in the last decade. However, there is still ample space for improvement aimed at overcoming existing challenges that are slowing down the progressive trajectory of this field.
Collapse
Affiliation(s)
- Lara Marrone
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Paolo M Marchi
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Mimoun Azzouz
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| |
Collapse
|
32
|
Overexpression of Brain- and Glial Cell Line-Derived Neurotrophic Factors Is Neuroprotective in an Animal Model of Acute Hypobaric Hypoxia. Int J Mol Sci 2022; 23:ijms23179733. [PMID: 36077134 PMCID: PMC9456324 DOI: 10.3390/ijms23179733] [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: 07/10/2022] [Revised: 08/21/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022] Open
Abstract
Currently, the role of the neurotrophic factors BDNF and GDNF in maintaining the brain’s resistance to the damaging effects of hypoxia and functional recovery of neural networks after exposure to damaging factors are actively studied. The assessment of the effect of an increase in the level of these neurotrophic factors in brain tissues using genetic engineering methods on the resistance of laboratory animals to hypoxia may pave the way for the future clinical use of neurotrophic factors BDNF and GDNF in the treatment of hypoxic damage. This study aimed to evaluate the antihypoxic and neuroprotective properties of BDNF and GDNF expression level increase using adeno-associated viral vectors in modeling hypoxia in vivo. To achieve overexpression of neurotrophic factors in the central nervous system’s cells, viral constructs were injected into the brain ventricles of newborn male C57Bl6 (P0) mice. Acute hypobaric hypoxia was modeled on the 30th day after the injection of viral vectors. Survival, cognitive, and mnestic functions in the late post-hypoxic period were tested. Evaluation of growth and weight characteristics and the neurological status of animals showed that the overexpression of neurotrophic factors does not affect the development of mice. It was found that the use of adeno-associated viral vectors increased the survival rate of male mice under hypoxic conditions. The present study indicates that the neurotrophic factors’ overexpression, induced by the specially developed viral constructs carrying the BDNF and GDNF genes, is a prospective neuroprotection method, increasing the survival rate of animals after hypoxic injury.
Collapse
|
33
|
Dong C, Zheng Y, Long-Iyer K, Wright EC, Li Y, Tian L. Fluorescence Imaging of Neural Activity, Neurochemical Dynamics, and Drug-Specific Receptor Conformation with Genetically Encoded Sensors. Annu Rev Neurosci 2022; 45:273-294. [PMID: 35316611 PMCID: PMC9940643 DOI: 10.1146/annurev-neuro-110520-031137] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Recent advances in fluorescence imaging permit large-scale recording of neural activity and dynamics of neurochemical release with unprecedented resolution in behaving animals. Calcium imaging with highly optimized genetically encoded indicators provides a mesoscopic view of neural activity from genetically defined populations at cellular and subcellular resolutions. Rigorously improved voltage sensors and microscopy allow for robust spike imaging of populational neurons in various brain regions. In addition, recent protein engineering efforts in the past few years have led to the development of sensors for neurotransmitters and neuromodulators. Here, we discuss the development and applications of these genetically encoded fluorescent indicators in reporting neural activity in response to various behaviors in different biological systems as well as in drug discovery. We also report a simple model to guide sensor selection and optimization.
Collapse
Affiliation(s)
- Chunyang Dong
- Graduate Program in Biochemistry, Molecular, Cellular, and Developmental Biology, University of California, Davis, California, USA
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, California, USA;
| | - Yu Zheng
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences; PKU-IDG/McGovern Institute for Brain Research; and Peking-Tsinghua Center for Life Sciences, Beijing, China;
| | - Kiran Long-Iyer
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, California, USA;
- Neuroscience Graduate Program, University of California, Davis, California, USA
| | - Emily C Wright
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, California, USA;
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences; PKU-IDG/McGovern Institute for Brain Research; and Peking-Tsinghua Center for Life Sciences, Beijing, China;
| | - Lin Tian
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, California, USA;
| |
Collapse
|
34
|
Pouchelon G, Vergara J, McMahon J, Gorissen BL, Lin JD, Vormstein-Schneider D, Niehaus JL, Burbridge TJ, Wester JC, Sherer M, Fernandez-Otero M, Allaway KC, Pelkey K, Chittajallu R, McBain CJ, Fan M, Nasse JS, Wildenberg GA, Fishell G, Dimidschstein J. A versatile viral toolkit for functional discovery in the nervous system. CELL REPORTS METHODS 2022; 2:100225. [PMID: 35784651 PMCID: PMC9243523 DOI: 10.1016/j.crmeth.2022.100225] [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] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/21/2022] [Accepted: 05/04/2022] [Indexed: 12/24/2022]
Abstract
The ability to precisely control transgene expression is essential for basic research and clinical applications. Adeno-associated viruses (AAVs) are non-pathogenic and can be used to drive stable expression in virtually any tissue, cell type, or species, but their limited genomic payload results in a trade-off between the transgenes that can be incorporated and the complexity of the regulatory elements controlling their expression. Resolving these competing imperatives in complex experiments inevitably results in compromises. Here, we assemble an optimized viral toolkit (VTK) that addresses these limitations and allows for efficient combinatorial targeting of cell types. Moreover, their modular design explicitly enables further refinements. We achieve this in compact vectors by integrating structural improvements of AAV vectors with innovative molecular tools. We illustrate the potential of this approach through a systematic demonstration of their utility for targeting cell types and querying their biology using a wide array of genetically encoded tools.
Collapse
Affiliation(s)
- Gabrielle Pouchelon
- Broad Institute, Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA
- Harvard Medical School, Blavatnik Institute, Neurobiology, Boston, MA 02115, USA
| | - Josselyn Vergara
- Broad Institute, Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA
| | - Justin McMahon
- Broad Institute, Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA
| | - Bram L. Gorissen
- Broad Institute, Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA
- Harvard Medical School, Blavatnik Institute, Neurobiology, Boston, MA 02115, USA
| | - Jessica D. Lin
- Broad Institute, Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA
| | | | | | - Timothy J. Burbridge
- Broad Institute, Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA
- Harvard Medical School, Blavatnik Institute, Neurobiology, Boston, MA 02115, USA
| | - Jason C. Wester
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Mia Sherer
- Broad Institute, Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA
- Harvard Medical School, Blavatnik Institute, Neurobiology, Boston, MA 02115, USA
| | - Marian Fernandez-Otero
- Broad Institute, Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA
- Harvard Medical School, Blavatnik Institute, Neurobiology, Boston, MA 02115, USA
| | - Kathryn C. Allaway
- Broad Institute, Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA
- Harvard Medical School, Blavatnik Institute, Neurobiology, Boston, MA 02115, USA
| | - Kenneth Pelkey
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Ramesh Chittajallu
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Chris J. McBain
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | | | | | - Gregg A. Wildenberg
- Department of Neurobiology, University of Chicago, Chicago, IL 60637, USA
- Biosciences Division, Argonne National Laboratory, Lemont, IL 60439, USA
| | - Gordon Fishell
- Broad Institute, Stanley Center for Psychiatric Research, Cambridge, MA 02142, USA
- Harvard Medical School, Blavatnik Institute, Neurobiology, Boston, MA 02115, USA
| | | |
Collapse
|
35
|
Peng Z, Lu H, Yang Q, Xie Q. Astrocyte Reprogramming in Stroke: Opportunities and Challenges. Front Aging Neurosci 2022; 14:885707. [PMID: 35663583 PMCID: PMC9160982 DOI: 10.3389/fnagi.2022.885707] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/11/2022] [Indexed: 11/21/2022] Open
Abstract
Stroke is a major cause of morbidity and mortality worldwide. In the early stages of stroke, irreversible damage to neurons leads to high mortality and disability rates in patients. However, there are still no effective prevention and treatment measures for the resulting massive neuronal death in clinical practice. Astrocyte reprogramming has recently attracted much attention as an avenue for increasing neurons in mice after cerebral ischemia. However, the field of astrocyte reprogramming has recently been mired in controversy due to reports questioning whether newborn neurons are derived from astrocyte transformation. To better understand the process and controversies of astrocyte reprogramming, this review introduces the method of astrocyte reprogramming and its application in stroke. By targeting key transcription factors or microRNAs, astrocytes in the mouse brain could be reprogrammed into functional neurons. Additionally, we summarize some of the current controversies over the lack of cell lineage tracing and single-cell sequencing experiments to provide evidence of gene expression profile changes throughout the process of astrocyte reprogramming. Finally, we present recent advances in cell lineage tracing and single-cell sequencing, suggesting that it is possible to characterize the entire process of astrocyte reprogramming by combining these techniques.
Collapse
Affiliation(s)
- Zhouzhou Peng
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, China
| | - Hui Lu
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, China
| | - Qingwu Yang
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, China
| | - Qi Xie
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, China
- *Correspondence: Qi Xie,
| |
Collapse
|
36
|
Lunev E, Karan A, Egorova T, Bardina M. Adeno-Associated Viruses for Modeling Neurological Diseases in Animals: Achievements and Prospects. Biomedicines 2022; 10:biomedicines10051140. [PMID: 35625877 PMCID: PMC9139062 DOI: 10.3390/biomedicines10051140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/13/2022] [Accepted: 05/13/2022] [Indexed: 02/04/2023] Open
Abstract
Adeno-associated virus (AAV) vectors have become an attractive tool for efficient gene transfer into animal tissues. Extensively studied as the vehicles for therapeutic constructs in gene therapy, AAVs are also applied for creating animal models of human genetic disorders. Neurological disorders are challenging to model in laboratory animals by transgenesis or genome editing, at least partially due to the embryonic lethality and the timing of the disease onset. Therefore, gene transfer with AAV vectors provides a more flexible option for simulating genetic neurological disorders. Indeed, the design of the AAV expression construct allows the reproduction of various disease-causing mutations, and also drives neuron-specific expression. The natural and newly created AAV serotypes combined with various delivery routes enable differentially targeting neuronal cell types and brain areas in vivo. Moreover, the same viral vector can be used to reproduce the main features of the disorder in mice, rats, and large laboratory animals such as non-human primates. The current review demonstrates the general principles for the development and use of AAVs in modeling neurological diseases. The latest achievements in AAV-mediated modeling of the common (e.g., Alzheimer’s disease, Parkinson’s disease, ataxias, etc.) and ultra-rare disorders affecting the central nervous system are described. The use of AAVs to create multiple animal models of neurological disorders opens opportunities for studying their mechanisms, understanding the main pathological features, and testing therapeutic approaches.
Collapse
Affiliation(s)
- Evgenii Lunev
- Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
- Marlin Biotech LLC, 354340 Sochi, Russia; (A.K.); (T.E.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
- Correspondence: (E.L.); (M.B.)
| | - Anna Karan
- Marlin Biotech LLC, 354340 Sochi, Russia; (A.K.); (T.E.)
| | - Tatiana Egorova
- Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
- Marlin Biotech LLC, 354340 Sochi, Russia; (A.K.); (T.E.)
| | - Maryana Bardina
- Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
- Marlin Biotech LLC, 354340 Sochi, Russia; (A.K.); (T.E.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
- Correspondence: (E.L.); (M.B.)
| |
Collapse
|
37
|
Morris G, Schorge S. Gene Therapy for Neurological Disease: State of the Art and Opportunities for Next-generation Approaches. Neuroscience 2022; 490:309-314. [PMID: 35304290 DOI: 10.1016/j.neuroscience.2022.03.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/22/2022] [Accepted: 03/09/2022] [Indexed: 12/11/2022]
Abstract
Gene therapy for rare monogenetic neurological disorders is reaching clinics and offering hope to families affected by these diseases. There is also potential for gene therapy to offer new and effective treatments for common, non-genetic disorders. Treatments for Parkinson's Disease are in clinical trials, and treatments for refractory epilepsies are due to enter first-in-human clinical trials in 2022. Gene therapies for these disorders are based on delivering genes that address the mechanism of the disease, not repairing a mutated gene. Similar 'mechanistic' gene therapies could offer treatments to a wide range of neurological and neuropsychiatric diseases where there is a known mechanism that could be restored using gene therapy. However, the permanent nature of most gene therapies is a serious drawback for translation of gene therapies to a wide-range of diseases because it could present risk of irreversible adverse effects. Several lines of research are aimed at developing gene therapy approaches that allow for the treatment to be turned on and off, including: using proteins activated by exogenous ligands, and promoters turned on by activators. We review these approaches and propose an overall de-risking strategy for gene therapy for common neurological and psychiatric diseases. This approach is based on using a temporary mRNA-based treatment to initially assess efficacy and safety of the planned manipulation, and only following with permanent, virally-delivered treatment if the approach appears safe and effective.
Collapse
Affiliation(s)
- Gareth Morris
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Stephanie Schorge
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom.
| |
Collapse
|
38
|
Kim H, Gao EB, Draper A, Berens NC, Vihma H, Zhang X, Higashi-Howard A, Ritola KD, Simon JM, Kennedy AJ, Philpot BD. Rescue of behavioral and electrophysiological phenotypes in a Pitt-Hopkins syndrome mouse model by genetic restoration of Tcf4 expression. eLife 2022; 11:e72290. [PMID: 35535852 PMCID: PMC9090324 DOI: 10.7554/elife.72290] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 04/19/2022] [Indexed: 12/14/2022] Open
Abstract
Pitt-Hopkins syndrome (PTHS) is a neurodevelopmental disorder caused by monoallelic mutation or deletion in the transcription factor 4 (TCF4) gene. Individuals with PTHS typically present in the first year of life with developmental delay and exhibit intellectual disability, lack of speech, and motor incoordination. There are no effective treatments available for PTHS, but the root cause of the disorder, TCF4 haploinsufficiency, suggests that it could be treated by normalizing TCF4 gene expression. Here, we performed proof-of-concept viral gene therapy experiments using a conditional Tcf4 mouse model of PTHS and found that postnatally reinstating Tcf4 expression in neurons improved anxiety-like behavior, activity levels, innate behaviors, and memory. Postnatal reinstatement also partially corrected EEG abnormalities, which we characterized here for the first time, and the expression of key TCF4-regulated genes. Our results support a genetic normalization approach as a treatment strategy for PTHS, and possibly other TCF4-linked disorders.
Collapse
Affiliation(s)
- Hyojin Kim
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, United States
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Eric B Gao
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, United States
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Adam Draper
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, United States
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Noah C Berens
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Hanna Vihma
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, United States
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Xinyuan Zhang
- Department of Chemistry and Biochemistry, Bates College, Lewiston, United States
| | | | | | - Jeremy M Simon
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, United States
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, United States
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hil, Chapel Hill, United States
| | - Andrew J Kennedy
- Department of Chemistry and Biochemistry, Bates College, Lewiston, United States
| | - Benjamin D Philpot
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, United States
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, United States
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hil, Chapel Hill, United States
| |
Collapse
|
39
|
Islam A, Tom VJ. The use of viral vectors to promote repair after spinal cord injury. Exp Neurol 2022; 354:114102. [PMID: 35513025 DOI: 10.1016/j.expneurol.2022.114102] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 04/21/2022] [Accepted: 04/27/2022] [Indexed: 11/16/2022]
Abstract
Spinal cord injury (SCI) is a devastating event that can permanently disrupt multiple modalities. Unfortunately, the combination of the inhibitory environment at a central nervous system (CNS) injury site and the diminished intrinsic capacity of adult axons for growth results in the failure for robust axonal regeneration, limiting the ability for repair. Delivering genetic material that can either positively or negatively modulate gene expression has the potential to counter the obstacles that hinder axon growth within the spinal cord after injury. A popular gene therapy method is to deliver the genetic material using viral vectors. There are considerations when deciding on a viral vector approach for a particular application, including the type of vector, as well as serotypes, and promoters. In this review, we will discuss some of the aspects to consider when utilizing a viral vector approach to as a therapy for SCI. Additionally, we will discuss some recent applications of gene therapy to target extrinsic and/or intrinsic barriers to promote axon regeneration after SCI in preclinical models. While still in early stages, this approach has potential to treat those living with SCI.
Collapse
Affiliation(s)
- Ashraful Islam
- Drexel University College of Medicine, Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Philadelphia, PA, USA
| | - Veronica J Tom
- Drexel University College of Medicine, Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Philadelphia, PA, USA.
| |
Collapse
|
40
|
Characterization of an immune-evading doxycycline-inducible lentiviral vector for gene therapy in the spinal cord. Exp Neurol 2022; 355:114120. [DOI: 10.1016/j.expneurol.2022.114120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 05/12/2022] [Accepted: 05/17/2022] [Indexed: 11/18/2022]
|
41
|
Oyarzabal EA, Hsu LM, Das M, Chao THH, Zhou J, Song S, Zhang W, Smith KG, Sciolino NR, Evsyukova IY, Yuan H, Lee SH, Cui G, Jensen P, Shih YYI. Chemogenetic stimulation of tonic locus coeruleus activity strengthens the default mode network. SCIENCE ADVANCES 2022; 8:eabm9898. [PMID: 35486721 PMCID: PMC9054017 DOI: 10.1126/sciadv.abm9898] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 03/15/2022] [Indexed: 05/31/2023]
Abstract
The default mode network (DMN) of the brain is functionally associated with a wide range of behaviors. In this study, we used functional magnetic resonance imaging (fMRI), positron emission tomography (PET), and spectral fiber photometry to investigate the selective neuromodulatory effect of norepinephrine (NE)-releasing noradrenergic neurons in the locus coeruleus (LC) on the mouse DMN. Chemogenetic-induced tonic LC activity decreased cerebral blood volume (CBV) and glucose uptake and increased synchronous low-frequency fMRI activity within the frontal cortices of the DMN. Fiber photometry results corroborated these findings, showing that LC-NE activation induced NE release, enhanced calcium-weighted neuronal spiking, and reduced CBV in the anterior cingulate cortex. These data suggest that LC-NE alters conventional coupling between neuronal activity and CBV in the frontal DMN. We also demonstrated that chemogenetic activation of LC-NE neurons strengthened functional connectivity within the frontal DMN, and this effect was causally mediated by reduced modulatory inputs from retrosplenial and hippocampal regions to the association cortices of the DMN.
Collapse
Affiliation(s)
- Esteban A. Oyarzabal
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
- Curriculum in Neurobiology, University of North Carolina, Chapel Hill, NC, USA
| | - Li-Ming Hsu
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Manasmita Das
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Tzu-Hao Harry Chao
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Jingheng Zhou
- In Vivo Neurobiology Group, Neurobiology Laboratory, NIEHS/NIH, Research Triangle Park, NC, USA
| | - Sheng Song
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Weiting Zhang
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Kathleen G. Smith
- Developmental Neurobiology Group, Neurobiology Laboratory, NIEHS/NIH, Research Triangle Park, NC, USA
| | - Natale R. Sciolino
- Developmental Neurobiology Group, Neurobiology Laboratory, NIEHS/NIH, Research Triangle Park, NC, USA
| | - Irina Y. Evsyukova
- Developmental Neurobiology Group, Neurobiology Laboratory, NIEHS/NIH, Research Triangle Park, NC, USA
| | - Hong Yuan
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
| | - Sung-Ho Lee
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Guohong Cui
- In Vivo Neurobiology Group, Neurobiology Laboratory, NIEHS/NIH, Research Triangle Park, NC, USA
| | - Patricia Jensen
- Developmental Neurobiology Group, Neurobiology Laboratory, NIEHS/NIH, Research Triangle Park, NC, USA
| | - Yen-Yu Ian Shih
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
42
|
Chen HH, Lu HY, Chang CH, Lin SH, Huang CW, Wei PH, Chen YW, Lin YR, Huang HS, Wang PY, Tsao YP, Chen SL. Breast carcinoma-amplified sequence 2 regulates adult neurogenesis via β-catenin. Stem Cell Res Ther 2022; 13:160. [PMID: 35410459 PMCID: PMC8996563 DOI: 10.1186/s13287-022-02837-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 03/31/2022] [Indexed: 11/10/2022] Open
Abstract
Background Breast carcinoma-amplified sequence 2 (BCAS2) regulates β-catenin gene splicing. The conditional knockout of BCAS2 expression in the forebrain (BCAS2 cKO) of mice confers impaired learning and memory along with decreased β-catenin expression. Because β-catenin reportedly regulates adult neurogenesis, we wondered whether BCAS2 could regulate adult neurogenesis via β-catenin. Methods BCAS2-regulating neurogenesis was investigated by characterizing BCAS2 cKO mice. Also, lentivirus-shBCAS2 was intracranially injected into the hippocampus of wild-type mice to knock down BCAS2 expression. We evaluated the rescue effects of BCAS2 cKO by intracranial injection of adeno-associated virus encoding BCAS2 (AAV-DJ8-BCAS2) and AAV-β-catenin gene therapy. Results To show that BCAS2-regulating adult neurogenesis via β-catenin, first, BCAS2 cKO mice showed low SRY-box 2-positive (Sox2+) neural stem cell proliferation and doublecortin-positive (DCX+) immature neurons. Second, stereotaxic intracranial injection of lentivirus-shBCAS2 knocked down BCAS2 in the hippocampus of wild-type mice, and we confirmed the BCAS2 regulation of adult neurogenesis via β-catenin. Third, AAV-DJ8-BCAS2 gene therapy in BCAS2 cKO mice reversed the low proliferation of Sox2+ neural stem cells and the decreased number of DCX+ immature neurons with increased β-catenin expression. Moreover, AAV-β-catenin gene therapy restored neuron stem cell proliferation and immature neuron differentiation, which further supports BCAS2-regulating adult neurogenesis via β-catenin. In addition, cells targeted by AAV-DJ8 injection into the hippocampus included Sox2 and DCX immature neurons, interneurons, and astrocytes. BCAS2 may regulate adult neurogenesis by targeting Sox2+ and DCX+ immature neurons for autocrine effects and interneurons or astrocytes for paracrine effects. Conclusions BCAS2 can regulate adult neurogenesis in mice via β-catenin. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02837-9.
Collapse
Affiliation(s)
- Hsin-Hsiung Chen
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, 7F, No1, Sec. 1, Jen-Ai Rd., Taipei 100, Taiwan
| | - Hao-Yu Lu
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, 7F, No1, Sec. 1, Jen-Ai Rd., Taipei 100, Taiwan
| | - Chao-Hsin Chang
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, 7F, No1, Sec. 1, Jen-Ai Rd., Taipei 100, Taiwan
| | - Shih-Hao Lin
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, 7F, No1, Sec. 1, Jen-Ai Rd., Taipei 100, Taiwan
| | - Chu-Wei Huang
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, 7F, No1, Sec. 1, Jen-Ai Rd., Taipei 100, Taiwan
| | - Po-Han Wei
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, 7F, No1, Sec. 1, Jen-Ai Rd., Taipei 100, Taiwan
| | - Yi-Wen Chen
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, 7F, No1, Sec. 1, Jen-Ai Rd., Taipei 100, Taiwan
| | - Yi-Rou Lin
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, 7F, No1, Sec. 1, Jen-Ai Rd., Taipei 100, Taiwan
| | - Hsien-Sung Huang
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, No. 1, Section 1, Jen Ai Road, Taipei 100, Taiwan
| | - Pei-Yu Wang
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, No. 1, Section 1, Jen Ai Road, Taipei 100, Taiwan
| | - Yeou-Ping Tsao
- Department of Ophthalmology, Mackay Memorial Hospital, No. 92, Sec. 2, Chung Shan North Road, Taipei 104, Taiwan
| | - Show-Li Chen
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, 7F, No1, Sec. 1, Jen-Ai Rd., Taipei 100, Taiwan.
| |
Collapse
|
43
|
Lateralized deficits after unilateral AAV-vector based overexpression of alpha-synuclein in the midbrain of rats on drug-free behavioural tests. Behav Brain Res 2022; 429:113887. [DOI: 10.1016/j.bbr.2022.113887] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/03/2022] [Accepted: 03/28/2022] [Indexed: 02/08/2023]
|
44
|
Dopamine D3 receptor in the nucleus accumbens alleviates neuroinflammation in a mouse model of depressive-like behavior. Brain Behav Immun 2022; 101:165-179. [PMID: 34971757 DOI: 10.1016/j.bbi.2021.12.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 12/20/2021] [Accepted: 12/23/2021] [Indexed: 12/13/2022] Open
Abstract
We recently reported that dopamine D3 receptor (D3R) was involved in inflammation-related depression. Nucleus accumbens (NAc) inflammation is implicated in the development and progression of depression, but its regulatory mechanism remains largely unknown. In a mouse model of NAc neuroinflammation induced by bilateral NAc injection of lipopolysaccharide (LPS), we observed that NAc neuroinflammation triggered depressive-like behaviors, and D3R expression decline and microglial activation in the NAc. A selective knockdown of D3R in the NAc elicited depressive-like behaviors, while re-expression of D3R in the NAc of global D3RKO mice alleviated depressive-like behaviors induced by D3R deficiency. D3R downregulation in the NAc shifted microglia toward a proinflammatory state, which was validated with cultured mouse microglial cultures. Further in vitro results demonstrated that D3R inhibition induced microglia to enter a proinflammatory state primarily through the Akt signaling pathway. In conclusion, our results suggest that D3R expression in the NAc may inhibit microglial proinflammatory responses in the NAc, thus alleviating NAc neuroinflammation and subsequent depressive-like behaviors through the Akt signaling pathway.
Collapse
|
45
|
Li Z, Zhang L, Jiang K, Zhang Y, Liu Y, Hu G, Song J. Biosafety assessment of delivery systems for clinical nucleic acid therapeutics. BIOSAFETY AND HEALTH 2022. [DOI: 10.1016/j.bsheal.2022.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
46
|
Au HKE, Isalan M, Mielcarek M. Gene Therapy Advances: A Meta-Analysis of AAV Usage in Clinical Settings. Front Med (Lausanne) 2022; 8:809118. [PMID: 35223884 PMCID: PMC8864161 DOI: 10.3389/fmed.2021.809118] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/06/2021] [Indexed: 01/04/2023] Open
Abstract
Adeno-associated viruses (AAVs) are the safest and most effective gene delivery vehicles to drive long-term transgene expression in gene therapy. While animal studies have shown promising results, the translatability of AAVs into clinical settings has been partly limited due to their restricted gene packaging capacities, off-target transduction, and immunogenicity. In this study, we analysed over two decades of AAV applications, in 136 clinical trials. This meta-analysis aims to provide an up-to-date overview of the use and successes of AAVs in clinical trials, while evaluating the approaches used to address the above challenges. First, this study reveals that the speed of novel AAV development has varied between therapeutic areas, with particular room for improvement in Central Nervous System disorders, where development has been slow. Second, the lack of dose-dependent toxicity and efficacy data indicates that optimal dosing regimes remain elusive. Third, more clinical data on the effectiveness of various immune-modulation strategies and gene editing approaches are required to direct future research and to accelerate the translation of AAV-mediated gene therapy into human applications.
Collapse
Affiliation(s)
- Hau Kiu Edna Au
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Mark Isalan
- Department of Life Sciences, Imperial College London, London, United Kingdom
- Imperial College Centre for Synthetic Biology, Imperial College London, London, United Kingdom
| | - Michal Mielcarek
- Department of Life Sciences, Imperial College London, London, United Kingdom
- Imperial College Centre for Synthetic Biology, Imperial College London, London, United Kingdom
| |
Collapse
|
47
|
Powell JE, Lim CKW, Krishnan R, McCallister TX, Saporito-Magriña C, Zeballos MA, McPheron GD, Gaj T. Targeted gene silencing in the nervous system with CRISPR-Cas13. SCIENCE ADVANCES 2022; 8:eabk2485. [PMID: 35044815 PMCID: PMC8769545 DOI: 10.1126/sciadv.abk2485] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 11/24/2021] [Indexed: 05/14/2023]
Abstract
Cas13 nucleases are a class of programmable RNA-targeting CRISPR effector proteins that are capable of silencing target gene expression in mammalian cells. Here, we demonstrate that RfxCas13d, a Cas13 ortholog with favorable characteristics to other family members, can be delivered to the mouse spinal cord and brain to silence neurodegeneration-associated genes. Intrathecally delivering an adeno-associated virus vector encoding an RfxCas13d variant programmed to target superoxide dismutase 1 (SOD1), a protein whose mutation can cause amyotrophic lateral sclerosis, reduced SOD1 mRNA and protein in the spinal cord by >50% and improved outcomes in a mouse model of the disorder. We further show that intrastriatally delivering an RfxCas13d variant programmed to target huntingtin (HTT), a protein whose mutation is causative for Huntington’s disease, led to a ~50% reduction in HTT protein in the mouse brain. Our results establish RfxCas13d as a versatile platform for knocking down gene expression in the nervous system.
Collapse
Affiliation(s)
- Jackson E. Powell
- Department of Bioengineering, University of Illinois, Urbana, IL 61801, USA
| | - Colin K. W. Lim
- Department of Bioengineering, University of Illinois, Urbana, IL 61801, USA
| | - Ramya Krishnan
- Department of Bioengineering, University of Illinois, Urbana, IL 61801, USA
| | | | | | - Maria A. Zeballos
- Department of Bioengineering, University of Illinois, Urbana, IL 61801, USA
| | | | - Thomas Gaj
- Department of Bioengineering, University of Illinois, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana, IL 61801, USA
| |
Collapse
|
48
|
Peptide-Functionalized Dendrimer Nanocarriers for Targeted Microdystrophin Gene Delivery. Pharmaceutics 2021; 13:pharmaceutics13122159. [PMID: 34959441 PMCID: PMC8708248 DOI: 10.3390/pharmaceutics13122159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/07/2021] [Accepted: 12/12/2021] [Indexed: 12/13/2022] Open
Abstract
Gene therapy is a good alternative for determined congenital disorders; however, there are numerous limitations for gene delivery in vivo including targeted cellular uptake, intracellular trafficking, and transport through the nuclear membrane. Here, a modified G5 polyamidoamine (G5 PAMAM) dendrimer-DNA complex was developed, which will allow cell-specific targeting to skeletal muscle cells and transport the DNA through the intracellular machinery and the nuclear membrane. The G5 PAMAM nanocarrier was modified with a skeletal muscle-targeting peptide (SMTP), a DLC8-binding peptide (DBP) for intracellular transport, and a nuclear localization signaling peptide (NLS) for nuclear uptake, and polyplexed with plasmid DNA containing the GFP-tagged microdystrophin (µDys) gene. The delivery of µDys has been considered as a therapeutic modality for patients suffering from a debilitating Duchenne muscular dystrophy (DMD) disorder. The nanocarrier-peptide-DNA polyplexes were prepared with different charge ratios and characterized for stability, size, surface charge, and cytotoxicity. Using the optimized nanocarrier polyplexes, the transfection efficiency in vitro was determined by demonstrating the expression of the GFP and the µDys protein using fluorescence and Western blotting studies, respectively. Protein expression in vivo was determined by injecting an optimal nanocarrier polyplex formulation to Duchenne model mice, mdx4Cv. Ultimately, these nanocarrier polyplexes will allow targeted delivery of the microdystrophin gene to skeletal muscle cells and result in improved muscle function in Duchenne muscular dystrophy patients.
Collapse
|
49
|
Ito D. Promise of Nucleic Acid Therapeutics for Amyotrophic Lateral Sclerosis. Ann Neurol 2021; 91:13-20. [PMID: 34704267 DOI: 10.1002/ana.26259] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/25/2021] [Accepted: 10/25/2021] [Indexed: 12/14/2022]
Abstract
Nucleic acid therapeutics have been attracting attention as novel drug discovery modalities for intractable diseases, including amyotrophic lateral sclerosis. This review provides an overview of the current status and prospects of antisense oligonucleotide treatment for amyotrophic lateral sclerosis. Recently, the results of a phase I/II study using the antisense oligonucleotides Tofersen to treat familial amyotrophic lateral sclerosis with superoxide dismutase 1 mutation have been reported. Intrathecal Tofersen administration resulted in a 36% reduction in superoxide dismutase 1 level in the cerebrospinal fluid. Another report described 2 patients with mutant superoxide dismutase 1 treated with an adeno-associated virus encoding a microRNA targeting superoxide dismutase 1. The first patient, who possessed the fast progressive mutant A5V, received a single intrathecal infusion. Although the patient died of respiratory arrest 16 months after treatment, autopsy findings showed a reduction of >90% in superoxide dismutase 1 level in the spinal cord. Clinical trials on antisense oligonucleotide therapies targeting other major amyotrophic lateral sclerosis-causative genes, fused in sarcoma and chromosome 9 open reading frame 72, are ongoing. To attenuate the pathology of TDP-43, strategies targeting regulators of TDP-43 (ataxin 2) and proteins downstream of TDP-43 (stathmin 2) by antisense oligonucleotides are being developed. The advent of nucleic acid therapeutics has enabled to specifically attack the molecules in the amyotrophic lateral sclerosis pathological cascade, expanding the options for therapeutic targets. ANN NEUROL 2021.
Collapse
Affiliation(s)
- Daisuke Ito
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
50
|
Guzman RA, Maruyama M, Moeinzadeh S, Lui E, Zhang N, Storaci HW, Tam K, Huang EE, Utsunomiya T, Rhee C, Gao Q, Yao Z, Yang YP, Goodman SB. The effect of genetically modified platelet-derived growth factor-BB over-expressing mesenchymal stromal cells during core decompression for steroid-associated osteonecrosis of the femoral head in rabbits. Stem Cell Res Ther 2021; 12:503. [PMID: 34526115 PMCID: PMC8444495 DOI: 10.1186/s13287-021-02572-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/27/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Approximately one third of patients undergoing core decompression (CD) for early-stage osteonecrosis of the femoral head (ONFH) experience progression of the disease, and subsequently require total hip arthroplasty (THA). Thus, identifying adjunctive treatments to optimize bone regeneration during CD is an unmet clinical need. Platelet-derived growth factor (PDGF)-BB plays a central role in cell growth and differentiation. The aim of this study was to characterize mesenchymal stromal cells (MSCs) that were genetically modified to overexpress PDGF-BB (PDGF-BB-MSCs) in vitro and evaluate their therapeutic effect when injected into the bone tunnel at the time of CD in an in vivo rabbit model of steroid-associated ONFH. METHODS In vitro studies: Rabbit MSCs were transduced with a lentivirus vector carrying the human PDGF-BB gene under the control of either the cytomegalovirus (CMV) or phosphoglycerate (PGK) promoter. The proliferative rate, PDGF-BB expression level, and osteogenic differentiation capacity of unmodified MSCs, CMV-PDGF-BB-MSCs, and PGK-PDGF-BB-MSCs were assessed. In vivo studies: Twenty-four male New Zealand white rabbits received an intramuscular (IM) injection of methylprednisolone 20 mg/kg. Four weeks later, the rabbits were divided into four groups: the CD group, the hydrogel [HG, (a collagen-alginate mixture)] group, the MSC group, and the PGK-PDGF-BB-MSC group. Eight weeks later, the rabbits were sacrificed, their femurs were harvested, and microCT, mechanical testing, and histological analyses were performed. RESULTS In vitro studies: PGK-PDGF-BB-MSCs proliferated more rapidly than unmodified MSCs (P < 0.001) and CMV-PDGF-BB-MSCs (P < 0.05) at days 3 and 7. CMV-PDGF-BB-MSCs demonstrated greater PDGF-BB expression than PGK-PDGF-BB-MSCs (P < 0.01). However, PGK-PDGF-BB-MSCs exhibited greater alkaline phosphatase staining at 14 days (P < 0.01), and osteogenic differentiation at 28 days (P = 0.07) than CMV-PDGF-BB-MSCs. In vivo: The PGK-PDGF-BB-MSC group had a trend towards greater bone mineral density (BMD) than the CD group (P = 0.074). The PGK-PDGF-BB-MSC group demonstrated significantly lower numbers of empty lacunae (P < 0.001), greater osteoclast density (P < 0.01), and greater angiogenesis (P < 0.01) than the other treatment groups. CONCLUSION The use of PGK-PDGF-BB-MSCs as an adjunctive treatment with CD may reduce progression of osteonecrosis and enhance bone regeneration and angiogenesis in the treatment of early-stage ONFH.
Collapse
Affiliation(s)
- Roberto Alfonso Guzman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA
| | - Masahiro Maruyama
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA
| | - Seyedsina Moeinzadeh
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA
| | - Elaine Lui
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA.,Department of Mechanical Engineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Ning Zhang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA
| | - Hunter W Storaci
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA
| | - Kaysie Tam
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA
| | - Elijah Ejun Huang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA
| | - Takeshi Utsunomiya
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA
| | - Claire Rhee
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA
| | - Yunzhi Peter Yang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA. .,Department of Material Science and Engineering, Stanford University School of Medicine, Stanford, CA, USA. .,Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA.
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards R155, Stanford, CA, 94305, USA. .,Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA. .,Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Redwood City, CA, 94063, USA.
| |
Collapse
|