151
|
Jiang X, Shao M, Liu X, Liu X, Zhang X, Wang Y, Yin K, Wang S, Hu Y, Jose PA, Zhou Z, Xu F, Yang Z. Reversible Treatment of Pressure Overload-Induced Left Ventricular Hypertrophy through Drd5 Nucleic Acid Delivery Mediated by Functional Polyaminoglycoside. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003706. [PMID: 33717857 PMCID: PMC7927605 DOI: 10.1002/advs.202003706] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/23/2020] [Indexed: 05/12/2023]
Abstract
Left ventricular hypertrophy and fibrosis are major risk factors for heart failure, which require timely and effective treatment. Genetic therapy has been shown to ameliorate hypertrophic cardiac damage. In this study, it is found that in mice, the dopamine D5 receptor (D5R) expression in the left ventricle (LV) progressively decreases with worsening of transverse aortic constriction-induced left ventricular hypertrophy. Then, a reversible treatment of left ventricular hypertrophy with Drd5 nucleic acids delivered by tobramycin-based hyperbranched polyaminoglycoside (SS-HPT) is studied. The heart-specific increase in D5R expression by SS-HPT/Drd5 plasmid in the early stage of left ventricular hypertrophy attenuates cardiac hypertrophy and fibrosis by preventing oxidative and endoplasmic reticulum (ER) stress and ameliorating autophagic dysregulation. By contrast, SS-HPT/Drd5 siRNA promotes the progression of left ventricular hypertrophy and accelerates the deterioration of myocardial function into heart failure. The reduction in cardiac D5R expression and dysregulated autophagy are observed in patients with hypertrophic cardiomyopathy and heart failure. The data show a cardiac-specific beneficial effect of SS-HPT/Drd5 plasmid on myocardial remodeling and dysfunction, which may provide an effective therapy of patients with left ventricular hypertrophy and heart failure.
Collapse
Affiliation(s)
- Xiaoliang Jiang
- NHC Key Laboratory of Human Disease Comparative Medicine (The Institute of Laboratory Animal Sciences, CAMS & PUMC), and Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases5 Pan Jia Yuan Nan Li, Chaoyang DistrictBeijing100021P. R. China
| | - Meiyu Shao
- Key Lab of Biomedical Materials of Natural MacromoleculesMinistry of EducationBeijing Laboratory of Biomedical MaterialsBeijing Advanced Innovation Center for Soft Matter Science and EngineeringBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Xue Liu
- NHC Key Laboratory of Human Disease Comparative Medicine (The Institute of Laboratory Animal Sciences, CAMS & PUMC), and Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases5 Pan Jia Yuan Nan Li, Chaoyang DistrictBeijing100021P. R. China
| | - Xing Liu
- NHC Key Laboratory of Human Disease Comparative Medicine (The Institute of Laboratory Animal Sciences, CAMS & PUMC), and Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases5 Pan Jia Yuan Nan Li, Chaoyang DistrictBeijing100021P. R. China
| | - Xu Zhang
- Department of Hepato‐Biliary‐Pancreatic SurgeryHenan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversityZhengzhouHenan450003P. R. China
| | - Yuming Wang
- Department of Hepato‐Biliary‐Pancreatic SurgeryHenan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversityZhengzhouHenan450003P. R. China
| | - Kunlun Yin
- State Key Laboratory of Cardiovascular DiseaseBeijing Key Laboratory for Molecular Diagnostics of Cardiovascular DiseasesDiagnostic Laboratory ServiceFuwai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037P. R. China
| | - Shuiyun Wang
- Department of Cardiovascular SurgeryState Key Laboratory of Cardiovascular DiseaseFuwai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037P. R. China
| | - Yang Hu
- Key Lab of Biomedical Materials of Natural MacromoleculesMinistry of EducationBeijing Laboratory of Biomedical MaterialsBeijing Advanced Innovation Center for Soft Matter Science and EngineeringBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Pedro A Jose
- Department of Pharmacology and PhysiologyThe George Washington University School of Medicine & Health SciencesWashingtonDC20052USA
- Department of MedicineDivision of Kidney Diseases & HypertensionThe George Washington University School of Medicine & Health SciencesWashingtonDC20052USA
| | - Zhou Zhou
- State Key Laboratory of Cardiovascular DiseaseBeijing Key Laboratory for Molecular Diagnostics of Cardiovascular DiseasesDiagnostic Laboratory ServiceFuwai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037P. R. China
| | - Fu‐Jian Xu
- Key Lab of Biomedical Materials of Natural MacromoleculesMinistry of EducationBeijing Laboratory of Biomedical MaterialsBeijing Advanced Innovation Center for Soft Matter Science and EngineeringBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Zhiwei Yang
- NHC Key Laboratory of Human Disease Comparative Medicine (The Institute of Laboratory Animal Sciences, CAMS & PUMC), and Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases5 Pan Jia Yuan Nan Li, Chaoyang DistrictBeijing100021P. R. China
| |
Collapse
|
152
|
Chowdhury EA, Meno-Tetang G, Chang HY, Wu S, Huang HW, Jamier T, Chandran J, Shah DK. Current progress and limitations of AAV mediated delivery of protein therapeutic genes and the importance of developing quantitative pharmacokinetic/pharmacodynamic (PK/PD) models. Adv Drug Deliv Rev 2021; 170:214-237. [PMID: 33486008 DOI: 10.1016/j.addr.2021.01.017] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 12/17/2022]
Abstract
While protein therapeutics are one of the most successful class of drug molecules, they are expensive and not suited for treating chronic disorders that require long-term dosing. Adeno-associated virus (AAV) mediated in vivo gene therapy represents a viable alternative, which can deliver the genes of protein therapeutics to produce long-term expression of proteins in target tissues. Ongoing clinical trials and recent regulatory approvals demonstrate great interest in these therapeutics, however, there is a lack of understanding regarding their cellular disposition, whole-body disposition, dose-exposure relationship, exposure-response relationship, and how product quality and immunogenicity affects these important properties. In addition, there is a lack of quantitative studies to support the development of pharmacokinetic-pharmacodynamic models, which can support the discovery, development, and clinical translation of this delivery system. In this review, we have provided a state-of-the-art overview of current progress and limitations related to AAV mediated delivery of protein therapeutic genes, along with our perspective on the steps that need to be taken to improve clinical translation of this therapeutic modality.
Collapse
|
153
|
Uddin MS, Khan ZA, Sumsuzzman DM, Perveen A, Ashraf GM. Challenges of Gene Therapy for Neurodegenerative Disorders. Curr Gene Ther 2021; 21:3-10. [PMID: 33153421 DOI: 10.2174/1566523220999201105150442] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/12/2020] [Accepted: 09/29/2020] [Indexed: 11/22/2022]
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
| | | | | | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Saharanpur, India
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
154
|
Singh M, Singh SP, Yadav D, Agarwal M, Agarwal S, Agarwal V, Swargiary G, Srivastava S, Tyagi S, Kaur R, Mani S. Targeted Delivery for Neurodegenerative Disorders Using Gene Therapy Vectors: Gene Next Therapeutic Goals. Curr Gene Ther 2021; 21:23-42. [PMID: 32811395 DOI: 10.2174/1566523220999200817164907] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/18/2020] [Accepted: 07/21/2020] [Indexed: 11/22/2022]
Abstract
The technique of gene therapy, ever since its advent nearly fifty years ago, has been utilized by scientists as a potential treatment option for various disorders. This review discusses some of the major neurodegenerative diseases (NDDs) like Alzheimer's disease (AD), Parkinson's Disease (PD), Motor neuron diseases (MND), Spinal Muscular Atrophy (SMA), Huntington's Disease (HD), Multiple Sclerosis (MS), etc. and their underlying genetic mechanisms along with the role that gene therapy can play in combating them. The pathogenesis and the molecular mechanisms specifying the altered gene expression of each of these NDDs have also been discussed in elaboration. The use of gene therapy vectors can prove to be an effective tool in the field of curative modern medicine for the generations to come. Therefore, consistent efforts and progressive research towards its implementation can provide us with powerful treatment options for disease conditions that have so far been considered as incurable.
Collapse
Affiliation(s)
- Manisha Singh
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT) Noida, U.P, India
| | - Surinder P Singh
- Bhartiya Nirdeshak Dravya Division, CSIR-National Physical Laboratory, New Delhi, India
| | - Deepshikha Yadav
- Bhartiya Nirdeshak Dravya Division, CSIR-National Physical Laboratory, New Delhi, India
| | - Mugdha Agarwal
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT) Noida, U.P., India
| | - Shriya Agarwal
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT) Noida, U.P., India
| | - Vinayak Agarwal
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT) Noida, U.P., India
| | - Geeta Swargiary
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT) Noida, U.P., India
| | - Sahil Srivastava
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT) Noida, U.P., India
| | - Sakshi Tyagi
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT) Noida, U.P., India
| | - Ramneek Kaur
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Shalini Mani
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT) Noida, U.P., India
| |
Collapse
|
155
|
Weber-Adrian D, Kofoed RH, Silburt J, Noroozian Z, Shah K, Burgess A, Rideout S, Kügler S, Hynynen K, Aubert I. Systemic AAV6-synapsin-GFP administration results in lower liver biodistribution, compared to AAV1&2 and AAV9, with neuronal expression following ultrasound-mediated brain delivery. Sci Rep 2021; 11:1934. [PMID: 33479314 PMCID: PMC7820310 DOI: 10.1038/s41598-021-81046-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 12/20/2020] [Indexed: 02/06/2023] Open
Abstract
Non-surgical gene delivery to the brain can be achieved following intravenous injection of viral vectors coupled with transcranial MRI-guided focused ultrasound (MRIgFUS) to temporarily and locally permeabilize the blood-brain barrier. Vector and promoter selection can provide neuronal expression in the brain, while limiting biodistribution and expression in peripheral organs. To date, the biodistribution of adeno-associated viruses (AAVs) within peripheral organs had not been quantified following intravenous injection and MRIgFUS delivery to the brain. We evaluated the quantity of viral DNA from the serotypes AAV9, AAV6, and a mosaic AAV1&2, expressing green fluorescent protein (GFP) under the neuron-specific synapsin promoter (syn). AAVs were administered intravenously during MRIgFUS targeting to the striatum and hippocampus in mice. The syn promoter led to undetectable levels of GFP expression in peripheral organs. In the liver, the biodistribution of AAV9 and AAV1&2 was 12.9- and 4.4-fold higher, respectively, compared to AAV6. The percentage of GFP-positive neurons in the FUS-targeted areas of the brain was comparable for AAV6-syn-GFP and AAV1&2-syn-GFP. In summary, MRIgFUS-mediated gene delivery with AAV6-syn-GFP had lower off-target biodistribution in the liver compared to AAV9 and AAV1&2, while providing neuronal GFP expression in the striatum and hippocampus.
Collapse
Affiliation(s)
- Danielle Weber-Adrian
- grid.410356.50000 0004 1936 8331Present Address: Faculty of Health Sciences, School of Medicine, Queen′s University, Kingston, ON Canada ,grid.17063.330000 0001 2157 2938Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Rikke Hahn Kofoed
- grid.17063.330000 0001 2157 2938Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Joseph Silburt
- grid.17063.330000 0001 2157 2938Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Zeinab Noroozian
- grid.17063.330000 0001 2157 2938Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Kairavi Shah
- grid.17063.330000 0001 2157 2938Institute of Medical Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Alison Burgess
- grid.17063.330000 0001 2157 2938Physical Sciences, Sunnybrook Research Institute, Toronto, ON Canada
| | - Shawna Rideout
- grid.17063.330000 0001 2157 2938Physical Sciences, Sunnybrook Research Institute, Toronto, ON Canada
| | - Sebastian Kügler
- grid.411984.10000 0001 0482 5331Department of Neurology, Center Nanoscale Microscopy and Physiology of the Brain (CNMPB) at University Medical Center Göttingen, Göttingen, Germany
| | - Kullervo Hynynen
- grid.17063.330000 0001 2157 2938Physical Sciences, Sunnybrook Research Institute, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Isabelle Aubert
- grid.17063.330000 0001 2157 2938Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON Canada
| |
Collapse
|
156
|
Bashiri FA, Temsah MH, Hundallah K, Alsohime F, AlRuthia Y. 2020 Update to Spinal Muscular Atrophy Management in Saudi Arabia. Front Pediatr 2021; 9:684134. [PMID: 34136444 PMCID: PMC8200403 DOI: 10.3389/fped.2021.684134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 04/29/2021] [Indexed: 11/17/2022] Open
Abstract
Novel therapeutic strategies have shown some promise in treating spinal muscular atrophy (SMA). However, the outcomes and acceptance of these new strategies are yet to be explored. We aimed to investigate physicians' opinions and perceptions toward management strategies of SMA across Saudi Arabia. This is a cross-sectional survey using a self-administered, structured questionnaire sent to physicians who care for SMA patients during the Saudi Pediatric Neurology Society annual conference. A total of 72 clinicians of different neurological subspecialties were included. 48.6% prescribed nusinersen to their patients, with 39% of them having patients started on nusinersen. Though, 8.3% prescribed onasemnogene abeparvovec for 1-3 patients, while none of their patients started on the treatment. 64.3% stated that the only treatment available for SMA in their settings is supportive care. Around 69.4% described having a moderate to high knowledge on SMA gene therapy, and 79.2% would recommend it. 48.6% confirmed they would prescribe gene therapy at the age of 6 months, and 78.3% would prescribe it for type-I SMA. Pediatric neurologists are receptive to novel and innovative therapies for SMA in Saudi Arabia. However, the high treatment acquisition cost, strict regulations, logistical issues, and budget constraints delay their adoption and implementation.
Collapse
Affiliation(s)
- Fahad A Bashiri
- College of Medicine, King Saud University, Riyadh, Saudi Arabia.,Division of Pediatric Neurology, Department of Pediatrics, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Mohamad-Hani Temsah
- College of Medicine, King Saud University, Riyadh, Saudi Arabia.,Pediatric Intensive Care Unit, Pediatric Department, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Khalid Hundallah
- Division of Neurology, Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Fahad Alsohime
- College of Medicine, King Saud University, Riyadh, Saudi Arabia.,Pediatric Intensive Care Unit, Pediatric Department, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Yazed AlRuthia
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.,Pharmacoeconomics Research Unit, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
157
|
Liu D, Zhu M, Zhang Y, Diao Y. Crossing the blood-brain barrier with AAV vectors. Metab Brain Dis 2021; 36:45-52. [PMID: 33201426 DOI: 10.1007/s11011-020-00630-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/12/2020] [Indexed: 12/26/2022]
Abstract
Central nervous system (CNS) diseases are some of the most difficult to treat because the blood-brain barrier (BBB) almost entirely limits the passage of many therapeutic drugs into the CNS. Gene therapy based on the adeno-associated virus (AAV) vector has the potential to overcome this problem. For example, an AAV serotype AAV9 has been widely studied for its ability to cross the BBB to transduce astrocytes, but its efficiency is limited. The emergence of AAV directed evolution technology provides a solution, and the variants derived from AAV9 directed evolution have been shown to have significantly higher crossing efficiency than AAV9. However, the mechanisms by which AAV crosses the BBB are still unclear. In this review, we focus on recent advances in crossing the blood-brain barrier with AAV vectors. We first review the AAV serotypes that can be applied to treating CNS diseases. Recent progress in possible AAV crossing the BBB and transduction mechanisms are then summarized. Finally, the methods to improve the AAV transduction efficiency are discussed.
Collapse
Affiliation(s)
- Dan Liu
- School of Biomedical Sciences, College of Chemical Engineering, Huaqiao University, Xiamen, Fujian, China.
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China.
| | - Mingyang Zhu
- School of Biomedical Sciences, College of Chemical Engineering, Huaqiao University, Xiamen, Fujian, China
| | - Yuqian Zhang
- School of Biomedical Sciences, College of Chemical Engineering, Huaqiao University, Xiamen, Fujian, China
| | - Yong Diao
- School of Biomedical Sciences, College of Chemical Engineering, Huaqiao University, Xiamen, Fujian, China
| |
Collapse
|
158
|
Nonnenmacher M, Wang W, Child MA, Ren XQ, Huang C, Ren AZ, Tocci J, Chen Q, Bittner K, Tyson K, Pande N, Chung CHY, Paul SM, Hou J. Rapid evolution of blood-brain-barrier-penetrating AAV capsids by RNA-driven biopanning. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 20:366-378. [PMID: 33553485 PMCID: PMC7841218 DOI: 10.1016/j.omtm.2020.12.006] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 12/16/2020] [Indexed: 12/26/2022]
Abstract
Therapeutic payload delivery to the central nervous system (CNS) remains a major challenge in gene therapy. Recent studies using function-driven evolution of adeno-associated virus (AAV) vectors have successfully identified engineered capsids with improved blood-brain barrier (BBB) penetration and CNS tropism in mouse. However, these strategies require transgenic animals and thus are limited to rodents. To address this issue, we developed a directed evolution approach based on recovery of capsid library RNA transcribed from CNS-restricted promoters. This RNA-driven screen platform, termed TRACER (Tropism Redirection of AAV by Cell-type-specific Expression of RNA), was tested in the mouse with AAV9 peptide display libraries and showed rapid emergence of dominant sequences. Ten individual variants were characterized and showed up to 400-fold higher brain transduction over AAV9 following systemic administration. Our results demonstrate that the TRACER platform allows rapid selection of AAV capsids with robust BBB penetration and CNS tropism in non-transgenic animals.
Collapse
Affiliation(s)
| | - Wei Wang
- Voyager Therapeutics, Cambridge, MA 02139, USA
| | | | | | - Carol Huang
- Voyager Therapeutics, Cambridge, MA 02139, USA
| | | | - Jenna Tocci
- Voyager Therapeutics, Cambridge, MA 02139, USA
| | | | | | | | | | | | | | - Jay Hou
- Voyager Therapeutics, Cambridge, MA 02139, USA
| |
Collapse
|
159
|
Current Status and Challenges Associated with CNS-Targeted Gene Delivery across the BBB. Pharmaceutics 2020; 12:pharmaceutics12121216. [PMID: 33334049 PMCID: PMC7765480 DOI: 10.3390/pharmaceutics12121216] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/19/2020] [Accepted: 12/11/2020] [Indexed: 12/21/2022] Open
Abstract
The era of the aging society has arrived, and this is accompanied by an increase in the absolute numbers of patients with neurological disorders, such as Alzheimer’s disease (AD) and Parkinson’s disease (PD). Such neurological disorders are serious costly diseases that have a significant impact on society, both globally and socially. Gene therapy has great promise for the treatment of neurological disorders, but only a few gene therapy drugs are currently available. Delivery to the brain is the biggest hurdle in developing new drugs for the central nervous system (CNS) diseases and this is especially true in the case of gene delivery. Nanotechnologies such as viral and non-viral vectors allow efficient brain-targeted gene delivery systems to be created. The purpose of this review is to provide a comprehensive review of the current status of the development of successful drug delivery to the CNS for the treatment of CNS-related disorders especially by gene therapy. We mainly address three aspects of this situation: (1) blood-brain barrier (BBB) functions; (2) adeno-associated viral (AAV) vectors, currently the most advanced gene delivery vector; (3) non-viral brain targeting by non-invasive methods.
Collapse
|
160
|
Conniot J, Talebian S, Simões S, Ferreira L, Conde J. Revisiting gene delivery to the brain: silencing and editing. Biomater Sci 2020; 9:1065-1087. [PMID: 33315025 DOI: 10.1039/d0bm01278e] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurodegenerative disorders, ischemic brain diseases, and brain tumors are debilitating diseases that severely impact a person's life and could possibly lead to their demise if left untreated. Many of these diseases do not respond to small molecule therapeutics and have no effective long-term therapy. Gene therapy offers the promise of treatment or even a cure for both genetic and acquired brain diseases, mediated by either silencing or editing disease-specific genes. Indeed, in the last 5 years, significant progress has been made in the delivery of non-coding RNAs as well as gene-editing formulations to the brain. Unfortunately, the delivery is a major limiting factor for the success of gene therapies. Both viral and non-viral vectors have been used to deliver genetic information into a target cell, but they have limitations. Viral vectors provide excellent transduction efficiency but are associated with toxic effects and have limited packaging capacity; however, non-viral vectors are less toxic and show a high packaging capacity at the price of low transfection efficiency. Herein, we review the progress made in the field of brain gene therapy, particularly in the design of non-toxic and trackable non-viral vectors, capable of controlled release of genes in response to internal/external triggers, and in the delivery of formulations for gene editing. The application of these systems in the context of various brain diseases in pre-clinical and clinical tests will be discussed. Such promising approaches could potentially pave the way for clinical realization of brain gene therapies.
Collapse
Affiliation(s)
- João Conniot
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal.
| | | | | | | | | |
Collapse
|
161
|
Zhu G, Harischandra DS, Ghaisas S, Zhang P, Prall W, Huang L, Maghames C, Guo L, Luna E, Mack KL, Torrente MP, Luk KC, Shorter J, Yang X. TRIM11 Prevents and Reverses Protein Aggregation and Rescues a Mouse Model of Parkinson's Disease. Cell Rep 2020; 33:108418. [PMID: 33264628 PMCID: PMC7906527 DOI: 10.1016/j.celrep.2020.108418] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 07/15/2020] [Accepted: 10/30/2020] [Indexed: 12/25/2022] Open
Abstract
Neurodegenerative diseases are characterized by the formation and propagation of protein aggregates, especially amyloid fibrils. However, what normally suppresses protein misfolding and aggregation in metazoan cells remains incompletely understood. Here, we show that TRIM11, a member of the metazoan tripartite motif (TRIM) family, both prevents the formation of protein aggregates and dissolves pre-existing protein deposits, including amyloid fibrils. These molecular chaperone and disaggregase activities are ATP independent. They enhance folding and solubility of normal proteins and cooperate with TRIM11 SUMO ligase activity to degrade aberrant proteins. TRIM11 abrogates α-synuclein fibrillization and restores viability in cell models of Parkinson's disease (PD). Intracranial adeno-associated viral delivery of TRIM11 mitigates α-synuclein-mediated pathology, neurodegeneration, and motor impairments in a PD mouse model. Other TRIMs can also function as ATP-independent molecular chaperones and disaggregases. Thus, we define TRIMs as a potent and multifunctional protein quality-control system in metazoa, which might be applied to treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Guixin Zhu
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dilshan S Harischandra
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shivani Ghaisas
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Pengfei Zhang
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wil Prall
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Liangqian Huang
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Chantal Maghames
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lili Guo
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Esteban Luna
- Department of Pathology and Laboratory Medicine and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Korrie L Mack
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mariana P Torrente
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kelvin C Luk
- Department of Pathology and Laboratory Medicine and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - James Shorter
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiaolu Yang
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
162
|
Boucher P, Cui X, Curiel DT. Adenoviral vectors for in vivo delivery of CRISPR-Cas gene editors. J Control Release 2020; 327:788-800. [PMID: 32891680 PMCID: PMC8091654 DOI: 10.1016/j.jconrel.2020.09.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 12/11/2022]
Abstract
Harnessing the bacterial clustered regularly interspaced short palindromic repeats (CRISPR) system for genome editing in eukaryotes has revolutionized basic biomedical research and translational sciences. The ability to create targeted alterations of the genome through this easy to design system has presented unprecedented opportunities to treat inherited disorders and other diseases such as cancer through gene therapy. A major hurdle is the lack of an efficient and safe in vivo delivery system, limiting most of the current gene therapy efforts to ex vivo editing of extracted cells. Here we discuss the unique features of adenoviral vectors that enable tissue specific and efficient delivery of the CRISPR-Cas machinery for in vivo genome editing.
Collapse
Affiliation(s)
- Paul Boucher
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in Saint Louis, St. Louis, MO 63130, USA; Division of Cancer Biology, Department of Radiation Oncology, School of Medicine, Washington University in Saint Louis, St. Louis, MO 63110, USA
| | - Xiaoxia Cui
- Genome Engineering & iPSC Center, Department of Genetics, School of Medicine, Washington University in Saint Louis, St. Louis, MO 63110, USA
| | - David T Curiel
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in Saint Louis, St. Louis, MO 63130, USA; Division of Cancer Biology, Department of Radiation Oncology, School of Medicine, Washington University in Saint Louis, St. Louis, MO 63110, USA; Biologic Therapeutics Center, Department of Radiation Oncology, School of Medicine, Washington University in Saint Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
163
|
Li S, Shi X, Wang H, Xiao L. A multifunctional dual-shell magnetic nanocomposite with near-infrared light response for synergistic chemo-thermal tumor therapy. J Biomed Mater Res B Appl Biomater 2020; 109:841-852. [PMID: 33135302 DOI: 10.1002/jbm.b.34749] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/26/2020] [Accepted: 10/06/2020] [Indexed: 01/29/2023]
Abstract
The synergistic tumor therapy in single nanoplatform has always been the goal for high efficacy tumor treatment while still remains great challenge. This paper reports a versatile nanotheranostic platform enlisting magnetic iron oxide nanoparticles, polydopamine (PDA), gold nanocages (Au nanocage) and metal organic framework (MOF, MIL101-NH2 ) in order to achieve synergistic chemothermal tumor therapy both in vitro and in vivo. The prepared magnetic photothermal nanoparticles (MPNPs) exhibit high drug loading capacity (31.34 mg/g), superior photo-thermal capacity (11.5°C enhancement in 180 s), low bio-toxicity, good magnetic resonance with a low dosage of 22 μg/g, as well as high antitumor efficacy in vivo. Such a novel and multifunctional nanoplatform is expected to find promising applications in target tumor synergistic therapy.
Collapse
Affiliation(s)
- Sheng Li
- School of Resource and Environmental Science, Hubei International Scientific and Technological Cooperation Base of Sustainable Resource and Energy, Hubei Biomass-Resource Chemistry and Environmental Biotechnology Key Laboratory, Wuhan University, Wuhan, PR China.,School of Chemistry and Environmental Engineering, Wuhan Polytechnic University, Wuhan, PR China
| | - Xiaowen Shi
- School of Resource and Environmental Science, Hubei International Scientific and Technological Cooperation Base of Sustainable Resource and Energy, Hubei Biomass-Resource Chemistry and Environmental Biotechnology Key Laboratory, Wuhan University, Wuhan, PR China
| | - Haibo Wang
- School of Chemistry and Environmental Engineering, Wuhan Polytechnic University, Wuhan, PR China
| | - Ling Xiao
- School of Resource and Environmental Science, Hubei International Scientific and Technological Cooperation Base of Sustainable Resource and Energy, Hubei Biomass-Resource Chemistry and Environmental Biotechnology Key Laboratory, Wuhan University, Wuhan, PR China
| |
Collapse
|
164
|
|
165
|
Iqubal A, Iqubal MK, Khan A, Ali J, Baboota S, Haque SE. Gene Therapy, A Novel Therapeutic Tool for Neurological Disorders: Current Progress, Challenges and Future Prospective. Curr Gene Ther 2020; 20:184-194. [DOI: 10.2174/1566523220999200716111502] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/02/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023]
Abstract
:
Neurological disorders are one of the major threat for health care system as they put enormous
socioeconomic burden. All aged populations are susceptible to one or other neurological problems
with symptoms of neuroinflammation, neurodegeneration and cognitive dysfunction. At present,
available pharmacotherapeutics are insufficient to treat these diseased conditions and in most cases,
they provide only palliative effect. It was also found that the molecular etiology of neurological disorders
is directly linked with the alteration in genetic makeup, which can be inherited or triggered by the
injury, environmental toxins and by some existing disease. Therefore, to take care of this situation,
gene therapy has emerged as an advanced modality that claims to permanently cure the disease by deletion,
silencing or edition of faulty genes and by insertion of healthier genes. In this modality, vectors
(viral and non-viral) are used to deliver targeted gene into a specific region of the brain via various
routes. At present, gene therapy has shown positive outcomes in complex neurological disorders, such
as Parkinson's disease, Alzheimer's disease, Huntington disease, Multiple sclerosis, Amyotrophic lateral
sclerosis and in lysosomal storage disease. However, there are some limitations such as immunogenic
reactions non-specificity of viral vectors and a lack of effective biomarkers to understand the efficacy
of therapy. Considerable progress has been made to improve vector design, gene selection and
targeted delivery. This review article deals with the current status of gene therapy in neurological disorders
along with its clinical relevance, challenges and future prospective.
Collapse
Affiliation(s)
- Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi- 110062, India
| | - Mohammad Kashif Iqubal
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi- 110062, India
| | - Aamir Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi- 110062, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi- 110062, India
| | - Sanjula Baboota
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi- 110062, India
| | - Syed Ehtaishamul Haque
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi- 110062, India
| |
Collapse
|
166
|
Feldman AG, Parsons JA, Dutmer CM, Veerapandiyan A, Hafberg E, Maloney N, Mack CL. Subacute Liver Failure Following Gene Replacement Therapy for Spinal Muscular Atrophy Type 1. J Pediatr 2020; 225:252-258.e1. [PMID: 32473148 PMCID: PMC10152980 DOI: 10.1016/j.jpeds.2020.05.044] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 02/06/2023]
Abstract
Spinal muscular atrophy is a neurodegenerative disease resulting from irreversible loss of anterior horn cells owing to biallelic deletions/mutations in the survival motor neuron (SMN) 1 gene. Gene replacement therapy using an adeno-associated virus vector containing the SMN gene was approved by the US Food and Drug Administration in May 2019. We report 2 cases of transient, drug-induced liver failure after this therapy.
Collapse
Affiliation(s)
- Amy G Feldman
- Children's Hospital Colorado, University of Colorado School of Medicine, Denver; Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO.
| | - Julie A Parsons
- Children's Hospital Colorado, University of Colorado School of Medicine, Denver; Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO
| | - Cullen M Dutmer
- Children's Hospital Colorado, University of Colorado School of Medicine, Denver; Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO
| | | | - Einar Hafberg
- Monroe Carell Jr. Children's Hospital at Vanderbilt, Nashville, TN
| | - Nolan Maloney
- University of Colorado School of Medicine, Aurora, CO
| | - Cara L Mack
- Children's Hospital Colorado, University of Colorado School of Medicine, Denver; Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO
| |
Collapse
|
167
|
Fatima N, Gromnicova R, Loughlin J, Sharrack B, Male D. Gold nanocarriers for transport of oligonucleotides across brain endothelial cells. PLoS One 2020; 15:e0236611. [PMID: 32941446 PMCID: PMC7498062 DOI: 10.1371/journal.pone.0236611] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 08/31/2020] [Indexed: 12/04/2022] Open
Abstract
Treatment of diseases that affect the CNS by gene therapy requires delivery of oligonucleotides to target cells within the brain. As the blood brain barrier prevents movement of large biomolecules, current approaches involve direct injection of the oligonucleotides, which is invasive and may have only a localised effect. The aim of this study was to investigate the potential of 2 nm galactose-coated gold nanoparticles (NP-Gal) as a delivery system of oligonucleotides across brain endothelium. DNA oligonucleotides of different types were attached to NP-Gal by the place exchange reaction and were characterised by EMSA (electrophoretic mobility shift assay). Several nanoparticle formulations were created, with single- or double-stranded (20nt or 40nt) DNA oligonucleotides, or with different amounts of DNA attached to the carriers. These nanocarriers were applied to transwell cultures of human brain endothelium in vitro (hCMEC/D3 cell-line) or to a 3D-hydrogel model of the blood-brain barrier including astrocytes. Transfer rates were measured by quantitative electron microscopy for the nanoparticles and qPCR for DNA. Despite the increase in nanoparticle size caused by attachment of oligonucleotides to the NP-Gal carrier, the rates of endocytosis and transcytosis of nanoparticles were both considerably increased when they carried an oligonucleotide cargo. Carriers with 40nt dsDNA were most efficient, accumulating in vesicles, in the cytosol and beneath the basal membrane of the endothelium. The oligonucleotide cargo remained attached to the nanocarriers during transcytosis and the transport rate across the endothelial cells was increased at least 50fold compared with free DNA. The nanoparticles entered the extracellular matrix and were taken up by the astrocytes in biologically functional amounts. Attachment of DNA confers a strong negative charge to the nanoparticles which may explain the enhanced binding to the endothelium and transcytosis by both vesicular transport and the transmembrane/cytosol pathway. These gold nanoparticles have the potential to transport therapeutic amounts of nucleic acids into the CNS.
Collapse
Affiliation(s)
- Nayab Fatima
- Department of Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| | - Radka Gromnicova
- Department of Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| | - Jane Loughlin
- Department of Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| | - Basil Sharrack
- Academic Department of Neuroscience and Sheffield, NIHR Translational Neuroscience BRC, Sheffield Teaching Hospitals, NHS Foundation Trust, University of Sheffield, Sheffield, United Kingdom
| | - David Male
- Department of Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| |
Collapse
|
168
|
Brenner D, Ludolph AC, Weishaupt JH. Gene specific therapies - the next therapeutic milestone in neurology. Neurol Res Pract 2020; 2:25. [PMID: 33324928 PMCID: PMC7650126 DOI: 10.1186/s42466-020-00075-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 06/22/2020] [Indexed: 02/07/2023] Open
Abstract
Gene selective approaches that either correct a disease mutation or a pathogenic mechanism will fundamentally change the treatment of neurological disorders. Basically, gene specific therapies are designed to manipulate RNA expression or reconstitute gene expression and function depending on the disease mechanism. Considerable methodological advances in the last years have made successful clinical translation of gene selective approaches possible, based on RNA interference or viral gene reconstitution in spinal muscular atrophy (SMA), Duchenne muscular dystrophy (DMD), and familial amyloid polyneuropathy (FAP). In this review, we provide an overview of the existing and coming gene specific therapies in neurology and discuss benefits, risks and challenges.
Collapse
Affiliation(s)
- David Brenner
- Department of Neurology, University of Ulm, Ulm, Germany
- Division of Neurodegenerative Diseases, Neurology Department, University Medicine Mannheim, Mannheim, Germany
| | - Albert C. Ludolph
- Department of Neurology, University of Ulm, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | - Jochen H. Weishaupt
- Department of Neurology, University of Ulm, Ulm, Germany
- Division of Neurodegenerative Diseases, Neurology Department, University Medicine Mannheim, Mannheim, Germany
| |
Collapse
|
169
|
Systemic Delivery of AAV-Fdxr Mitigates the Phenotypes of Mitochondrial Disorders in Fdxr Mutant Mice. Mol Ther Methods Clin Dev 2020; 18:84-97. [PMID: 32995353 PMCID: PMC7488755 DOI: 10.1016/j.omtm.2020.05.021] [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: 02/06/2020] [Accepted: 05/19/2020] [Indexed: 11/23/2022]
Abstract
Gene therapy now provides a novel approach for treating inherited monogenetic disorders, including nuclear gene mutations associated with mitochondrial diseases. In this study, we have utilized a mouse model carrying a p.Arg389Gln mutation of the mitochondrial Ferredoxin Reductase gene (Fdxr) and treated them with neurotropic AAV-PHP.B vector loaded with the mouse Fdxr cDNA sequence. We then used immunofluorescence staining and western blot to test the transduction efficiency of this vector. Toluidine blue staining and electronic microscopy were also utilized to assess the morphology of optic and sciatic nerves, and the mitochondrial respiratory chain activity was determined as well. The AAV vector effectively transduced in the central nervous system and peripheral organs, and AAV-Fdxr treatment reversed almost all the symptoms of the mutants (FdxrR389Q/R389Q). This therapy also improved the electronic conductivity of the sciatic nerves, prevented optic atrophy, improved mobility, and restored mitochondrial complex function. Most notably, the sensory neuropathy, neurodegeneration, and chronic neuroinflammation in the brain were alleviated. Overall, we present the first demonstration of a potential definitive treatment that significantly improves optic and sciatic nerve atrophy, sensory neuropathy, and mitochondrial dysfunction in FDXR-related mitochondriopathy. Our study provides substantial support for the translation of AAV-based Fdxr gene therapy into clinical applications.
Collapse
|
170
|
Uchitel J, Kantor B, Smith EC, Mikati MA. Viral-Mediated Gene Replacement Therapy in the Developing Central Nervous System: Current Status and Future Directions. Pediatr Neurol 2020; 110:5-19. [PMID: 32684374 DOI: 10.1016/j.pediatrneurol.2020.04.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/17/2020] [Accepted: 04/14/2020] [Indexed: 12/13/2022]
Abstract
The past few years have witnessed rapid developments in viral-mediated gene replacement therapy for pediatric central nervous system neurogenetic disorders. Here, we provide pediatric neurologists with an up-to-date, comprehensive overview of these developments and note emerging trends for future research. This review presents the different types of viral vectors used in viral-mediated gene replacement therapy; the fundamental properties of viral-mediated gene replacement therapy; the challenges associated with the use of this therapy in the central nervous system; the pathway for therapy development, from translational basic science studies to clinical trials; and an overview of the therapies that have reached clinical trials in patients. Current viral platforms under investigation include adenovirus vectors, adeno-associated viral vectors, lentiviral/retroviral vectors, and herpes simplex virus type 1 vectors. This review also presents an in-depth analysis of numerous studies that investigated these viral platforms in cultured cells and in transgenic animal models for pediatric neurogenetic disorders. Viral vectors have been applied to clinical trials for many different pediatric neurogenetic disorders, including Canavan disease, metachromatic leukodystrophy, neuronal ceroid lipofuscinosis, mucopolysaccharidosis III, spinal muscular atrophy, and aromatic l-amino acid decarboxylase deficiency. Of these diseases, only spinal muscular atrophy has a viral-mediated gene replacement therapy approved for marketing. Despite significant progress in therapy development, many challenges remain. Surmounting these challenges is critical to advancing the current status of viral-mediated gene replacement therapy for pediatric central nervous system neurogenetic disorders.
Collapse
Affiliation(s)
- Julie Uchitel
- Division of Pediatric Neurology and Developmental Medicine, Duke University Medical Center, Durham, North Carolina
| | - Boris Kantor
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina
| | - Edward C Smith
- Division of Pediatric Neurology and Developmental Medicine, Duke University Medical Center, Durham, North Carolina
| | - Mohamad A Mikati
- Division of Pediatric Neurology and Developmental Medicine, Duke University Medical Center, Durham, North Carolina; Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina.
| |
Collapse
|
171
|
Cunnane SC, Trushina E, Morland C, Prigione A, Casadesus G, Andrews ZB, Beal MF, Bergersen LH, Brinton RD, de la Monte S, Eckert A, Harvey J, Jeggo R, Jhamandas JH, Kann O, la Cour CM, Martin WF, Mithieux G, Moreira PI, Murphy MP, Nave KA, Nuriel T, Oliet SHR, Saudou F, Mattson MP, Swerdlow RH, Millan MJ. Brain energy rescue: an emerging therapeutic concept for neurodegenerative disorders of ageing. Nat Rev Drug Discov 2020; 19:609-633. [PMID: 32709961 PMCID: PMC7948516 DOI: 10.1038/s41573-020-0072-x] [Citation(s) in RCA: 548] [Impact Index Per Article: 109.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2020] [Indexed: 12/11/2022]
Abstract
The brain requires a continuous supply of energy in the form of ATP, most of which is produced from glucose by oxidative phosphorylation in mitochondria, complemented by aerobic glycolysis in the cytoplasm. When glucose levels are limited, ketone bodies generated in the liver and lactate derived from exercising skeletal muscle can also become important energy substrates for the brain. In neurodegenerative disorders of ageing, brain glucose metabolism deteriorates in a progressive, region-specific and disease-specific manner - a problem that is best characterized in Alzheimer disease, where it begins presymptomatically. This Review discusses the status and prospects of therapeutic strategies for countering neurodegenerative disorders of ageing by improving, preserving or rescuing brain energetics. The approaches described include restoring oxidative phosphorylation and glycolysis, increasing insulin sensitivity, correcting mitochondrial dysfunction, ketone-based interventions, acting via hormones that modulate cerebral energetics, RNA therapeutics and complementary multimodal lifestyle changes.
Collapse
Affiliation(s)
- Stephen C Cunnane
- Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada.
- Research Center on Aging, Sherbrooke, QC, Canada.
| | | | - Cecilie Morland
- Department of Pharmaceutical Biosciences, Institute of Pharmacy, University of Oslo, Oslo, Norway
| | - Alessandro Prigione
- Department of General Pediatrics, Neonatology, and Pediatric Cardiology, University of Dusseldorf, Dusseldorf, Germany
| | - Gemma Casadesus
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Zane B Andrews
- Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Department of Physiology, Monash University, Clayton, VIC, Australia
| | - M Flint Beal
- Department of Neurology, Weill Cornell Medicine, New York, NY, USA
| | - Linda H Bergersen
- Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | | | | | | | - Jenni Harvey
- Ninewells Hospital, University of Dundee, Dundee, UK
- Medical School, University of Dundee, Dundee, UK
| | - Ross Jeggo
- Centre for Therapeutic Innovation in Neuropsychiatry, Institut de Recherche Servier, Croissy sur Seine, France
| | - Jack H Jhamandas
- Department of Medicine, University of Albeta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Albeta, Edmonton, AB, Canada
| | - Oliver Kann
- Institute of Physiology and Pathophysiology, University of Heidelberg, Heidelberg, Germany
| | - Clothide Mannoury la Cour
- Centre for Therapeutic Innovation in Neuropsychiatry, Institut de Recherche Servier, Croissy sur Seine, France
| | - William F Martin
- Institute of Molecular Evolution, University of Dusseldorf, Dusseldorf, Germany
| | | | - Paula I Moreira
- CNC Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Klaus-Armin Nave
- Department of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Tal Nuriel
- Columbia University Medical Center, New York, NY, USA
| | - Stéphane H R Oliet
- Neurocentre Magendie, INSERM U1215, Bordeaux, France
- Université de Bordeaux, Bordeaux, France
| | - Frédéric Saudou
- University of Grenoble Alpes, Grenoble, France
- INSERM U1216, CHU Grenoble Alpes, Grenoble Institute Neurosciences, Grenoble, France
| | - Mark P Mattson
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Mark J Millan
- Centre for Therapeutic Innovation in Neuropsychiatry, Institut de Recherche Servier, Croissy sur Seine, France.
| |
Collapse
|
172
|
Vormstein-Schneider D, Lin JD, Pelkey KA, Chittajallu R, Guo B, Arias-Garcia MA, Allaway K, Sakopoulos S, Schneider G, Stevenson O, Vergara J, Sharma J, Zhang Q, Franken TP, Smith J, Ibrahim LA, M Astro KJ, Sabri E, Huang S, Favuzzi E, Burbridge T, Xu Q, Guo L, Vogel I, Sanchez V, Saldi GA, Gorissen BL, Yuan X, Zaghloul KA, Devinsky O, Sabatini BL, Batista-Brito R, Reynolds J, Feng G, Fu Z, McBain CJ, Fishell G, Dimidschstein J. Viral manipulation of functionally distinct interneurons in mice, non-human primates and humans. Nat Neurosci 2020; 23:1629-1636. [PMID: 32807948 DOI: 10.1038/s41593-020-0692-9] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 07/10/2020] [Indexed: 12/21/2022]
Abstract
Recent success in identifying gene-regulatory elements in the context of recombinant adeno-associated virus vectors has enabled cell-type-restricted gene expression. However, within the cerebral cortex these tools are largely limited to broad classes of neurons. To overcome this limitation, we developed a strategy that led to the identification of multiple new enhancers to target functionally distinct neuronal subtypes. By investigating the regulatory landscape of the disease gene Scn1a, we discovered enhancers selective for parvalbumin (PV) and vasoactive intestinal peptide-expressing interneurons. Demonstrating the functional utility of these elements, we show that the PV-specific enhancer allowed for the selective targeting and manipulation of these neurons across vertebrate species, including humans. Finally, we demonstrate that our selection method is generalizable and characterizes additional PV-specific enhancers with exquisite specificity within distinct brain regions. Altogether, these viral tools can be used for cell-type-specific circuit manipulation and hold considerable promise for use in therapeutic interventions.
Collapse
Affiliation(s)
| | - Jessica D Lin
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Kenneth A Pelkey
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Ramesh Chittajallu
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Baolin Guo
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Mario A Arias-Garcia
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Kathryn Allaway
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA.,Department of Neurobiology, Harvard Medical School, Boston, MA, USA.,NYU Langone Medical Center, New York University, New York, NY, USA
| | - Sofia Sakopoulos
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Gates Schneider
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Olivia Stevenson
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Josselyn Vergara
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Jitendra Sharma
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Qiangge Zhang
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tom P Franken
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jared Smith
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Leena A Ibrahim
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA.,Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Kevin J M Astro
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA.,Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Ehsan Sabri
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Shuhan Huang
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA.,Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Emilia Favuzzi
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA.,Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Timothy Burbridge
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA.,Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Qing Xu
- Center for Genomics & Systems Biology, New York University, Abu Dhabi, UAE
| | - Lihua Guo
- Center for Genomics & Systems Biology, New York University, Abu Dhabi, UAE
| | - Ian Vogel
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Vanessa Sanchez
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Giuseppe A Saldi
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA.,Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Bram L Gorissen
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Xiaoqing Yuan
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Kareem A Zaghloul
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Orrin Devinsky
- Comprehensive Epilepsy Center, New York University School of Medicine, New York, NY, USA
| | - Bernardo L Sabatini
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA.,Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | | | - John Reynolds
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Guoping Feng
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA.,McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Zhanyan Fu
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Chris J McBain
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Gord Fishell
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA.,Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Jordane Dimidschstein
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
173
|
Cell-Type-Specific Gene Inactivation and In Situ Restoration via Recombinase-Based Flipping of Targeted Genomic Region. J Neurosci 2020; 40:7169-7186. [PMID: 32801153 DOI: 10.1523/jneurosci.1044-20.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/22/2020] [Accepted: 07/30/2020] [Indexed: 11/21/2022] Open
Abstract
Conditional gene inactivation and restoration are powerful tools for studying gene functions in the nervous system and for modeling neuropsychiatric diseases. The combination of the two is necessary to interrogate specific cell types within defined developmental stages. However, very few methods and animal models have been developed for such purpose. Here we present a versatile method for conditional gene inactivation and in situ restoration through reversibly inverting a critical part of its endogenous genomic sequence by Cre- and Flp-mediated recombinations. Using this method, we generated a mouse model to manipulate Mecp2, an X-linked dosage-sensitive gene whose mutations cause Rett syndrome. Combined with multiple Cre- and Flp-expressing drivers and viral tools, we achieved efficient and reliable Mecp2 inactivation and restoration in the germline and several neuronal cell types, and demonstrated phenotypic reversal and prevention on cellular and behavioral levels in male mice. This study not only provides valuable tools and critical insights for Mecp2 and Rett syndrome, but also offers a generally applicable strategy to decipher other neurologic disorders.SIGNIFICANCE STATEMENT Studying neurodevelopment and modeling neurologic disorders rely on genetic tools, such as conditional gene regulation. We developed a new method to combine conditional gene inactivation and restoration on a single allele without disturbing endogenous expression pattern or dosage. We applied it to manipulate Mecp2, a gene residing on X chromosome whose malfunction leads to neurologic disease, including Rett syndrome. Our results demonstrated the efficiency, specificity, and versatility of this new method, provided valuable tools and critical insights for Mecp2 function and Rett syndrome research, and offered a generally applicable strategy to investigate other genes and genetic disorders.
Collapse
|
174
|
Chen W, Hu Y, Ju D. Gene therapy for neurodegenerative disorders: advances, insights and prospects. Acta Pharm Sin B 2020; 10:1347-1359. [PMID: 32963936 PMCID: PMC7488363 DOI: 10.1016/j.apsb.2020.01.015] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/09/2019] [Accepted: 12/06/2019] [Indexed: 02/07/2023] Open
Abstract
Gene therapy is rapidly emerging as a powerful therapeutic strategy for a wide range of neurodegenerative disorders, including Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington's disease (HD). Some early clinical trials have failed to achieve satisfactory therapeutic effects. Efforts to enhance effectiveness are now concentrating on three major fields: identification of new vectors, novel therapeutic targets, and reliable of delivery routes for transgenes. These approaches are being assessed closely in preclinical and clinical trials, which may ultimately provide powerful treatments for patients. Here, we discuss advances and challenges of gene therapy for neurodegenerative disorders, highlighting promising technologies, targets, and future prospects.
Collapse
Key Words
- AADC, aromatic-l-amino-acid
- AAVs, adeno-associated viruses
- AD, Alzheimer's disease
- ARSA, arylsulfatase A
- ASOs, antisense oligonucleotides
- ASPA, aspartoacylase
- Adeno-associated viruses
- Adv, adenovirus
- BBB, blood–brain barrier
- BCSFB, blood–cerebrospinal fluid barrier
- BRB, blood–retina barrier
- Bip, glucose regulated protein 78
- CHOP, CCAAT/enhancer binding homologous protein
- CLN6, ceroidlipofuscinosis neuronal protein 6
- CNS, central nervous system
- CSF, cerebrospinal fluid
- Central nervous system
- Delivery routes
- ER, endoplasmic reticulum
- FDA, U.S. Food and Drug Administration
- GAA, lysosomal acid α-glucosidase
- GAD, glutamic acid decarboxylase
- GDNF, glial derived neurotrophic factor
- Gene therapy
- HD, Huntington's disease
- HSPGs, heparin sulfate proteoglycans
- HTT, mutant huntingtin
- IDS, iduronate 2-sulfatase
- LVs, retrovirus/lentivirus
- Lamp2a, lysosomal-associated membrane protein 2a
- NGF, nerve growth factor
- Neurodegenerative disorders
- PD, Parkinson's disease
- PGRN, Progranulin
- PINK1, putative kinase 1
- PTEN, phosphatase and tensin homolog
- RGCs, retinal ganglion cells
- RNAi, RNA interference
- RPE, retinal pigmented epithelial
- SGSH, lysosomal heparan-N-sulfamidase gene
- SMN, survival motor neuron
- SOD, superoxide dismutase
- SUMF, sulfatase-modifying factor
- TFEB, transcription factor EB
- TPP1, tripeptidyl peptidase 1
- TREM2, triggering receptor expressed on myeloid cells 2
- UPR, unfolded protein response
- ZFPs, zinc finger proteins
- mTOR, mammalian target of rapamycin
- siRNA, small interfering RNA
Collapse
Affiliation(s)
- Wei Chen
- Department of Biological Medicines, Fudan University School of Pharmacy, Shanghai 201203, China
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Yang Hu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Dianwen Ju
- Department of Biological Medicines, Fudan University School of Pharmacy, Shanghai 201203, China
| |
Collapse
|
175
|
Sudhakar V, Naidoo J, Samaranch L, Bringas JR, Lonser RR, Fiandaca MS, Bankiewicz KS. Infuse-as-you-go convective delivery to enhance coverage of elongated brain targets: technical note. J Neurosurg 2020; 133:530-537. [PMID: 31299656 DOI: 10.3171/2019.4.jns19826] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 04/29/2019] [Indexed: 01/07/2023]
Abstract
OBJECTIVE To develop and assess a convective delivery technique that enhances the effectiveness of drug delivery to nonspherical brain nuclei, the authors developed an occipital "infuse-as-you-go" approach to the putamen and compared it to the currently used transfrontal approach. METHODS Eleven nonhuman primates received a bilateral putamen injection of adeno-associated virus with 2 mM gadolinium-DTPA by real-time MR-guided convective perfusion via either a transfrontal (n = 5) or occipital infuse-as-you-go (n = 6) approach. RESULTS MRI provided contemporaneous assessment and monitoring of putaminal infusions for transfrontal (2 to 3 infusion deposits) and occipital infuse-as-you-go (stepwise infusions) putaminal approaches. The infuse-as-you-go technique was more efficient than the transfrontal approach (mean 35 ± 1.1 vs 88 ± 8.3 minutes [SEM; p < 0.001]). More effective perfusion of the postcommissural and total putamen was achieved with the infuse-as-you-go versus transfronatal approaches (100-µl infusion volumes; mean posterior commissural coverage 76.2% ± 5.0% vs 32.8% ± 2.9% [p < 0.001]; and mean total coverage 53.5% ± 3.0% vs 38.9% ± 2.3% [p < 0.01]). CONCLUSIONS The infuse-as-you-go approach, paralleling the longitudinal axis of the target structure, provides a more effective and efficient method for convective infusate coverage of elongated, irregularly shaped subcortical brain nuclei.
Collapse
Affiliation(s)
- Vivek Sudhakar
- 1Department of Neurological Surgery, University of California, San Francisco, California; and
| | - Jerusha Naidoo
- 1Department of Neurological Surgery, University of California, San Francisco, California; and
| | - Lluis Samaranch
- 1Department of Neurological Surgery, University of California, San Francisco, California; and
| | - John R Bringas
- 1Department of Neurological Surgery, University of California, San Francisco, California; and
| | - Russell R Lonser
- 2Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Massimo S Fiandaca
- 1Department of Neurological Surgery, University of California, San Francisco, California; and
| | - Krystof S Bankiewicz
- 1Department of Neurological Surgery, University of California, San Francisco, California; and
- 2Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio
| |
Collapse
|
176
|
Orefice NS. Development of New Strategies Using Extracellular Vesicles Loaded with Exogenous Nucleic Acid. Pharmaceutics 2020; 12:E705. [PMID: 32722622 PMCID: PMC7464422 DOI: 10.3390/pharmaceutics12080705] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/19/2020] [Accepted: 07/24/2020] [Indexed: 12/13/2022] Open
Abstract
Gene therapy is a therapeutic strategy of delivering foreign genetic material (encoding for an important protein) into a patient's target cell to replace a defective gene. Nucleic acids are embedded within the adeno-associated virus (AAVs) vectors; however, preexisting immunity to AAVs remains a significant concern that impairs their clinical application. Extracellular vesicles (EVs) hold great potential for therapeutic applications as vectors of nucleic acids due to their endogenous intercellular communication functions through their cargo delivery, including lipids and proteins. So far, small RNAs (siRNA and micro (mi)RNA) have been mainly loaded into EVs to treat several diseases, but the potential use of EVs to load and deliver exogenous plasmid DNA has not been thoroughly described. This review provides a comprehensive overview of the principal methodologies currently employed to load foreign genetic material into EVs, highlighting the need to find the most effective strategies for their successful clinical translations.
Collapse
Affiliation(s)
- Nicola Salvatore Orefice
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; or ; Tel.: +1-608-262-21-89
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
177
|
Tosolini AP, Sleigh JN. Intramuscular Delivery of Gene Therapy for Targeting the Nervous System. Front Mol Neurosci 2020; 13:129. [PMID: 32765219 PMCID: PMC7379875 DOI: 10.3389/fnmol.2020.00129] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 06/26/2020] [Indexed: 12/11/2022] Open
Abstract
Virus-mediated gene therapy has the potential to deliver exogenous genetic material into specific cell types to promote survival and counteract disease. This is particularly enticing for neuronal conditions, as the nervous system is renowned for its intransigence to therapeutic targeting. Administration of gene therapy viruses into skeletal muscle, where distal terminals of motor and sensory neurons reside, has been shown to result in extensive transduction of cells within the spinal cord, brainstem, and sensory ganglia. This route is minimally invasive and therefore clinically relevant for gene therapy targeting to peripheral nerve soma. For successful transgene expression, viruses administered into muscle must undergo a series of processes, including host cell interaction and internalization, intracellular sorting, long-range retrograde axonal transport, endosomal liberation, and nuclear import. In this review article, we outline key characteristics of major gene therapy viruses—adenovirus, adeno-associated virus (AAV), and lentivirus—and summarize the mechanisms regulating important steps in the virus journey from binding at peripheral nerve terminals to nuclear delivery. Additionally, we describe how neuropathology can negatively influence these pathways, and conclude by discussing opportunities to optimize the intramuscular administration route to maximize gene delivery and thus therapeutic potential.
Collapse
Affiliation(s)
- Andrew P Tosolini
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - James N Sleigh
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.,UK Dementia Research Institute, University College London, London, United Kingdom
| |
Collapse
|
178
|
Transport rate of EAAT2 is regulated by amino acid located at the interface between the scaffolding and substrate transport domains. Neurochem Int 2020; 139:104792. [PMID: 32668264 DOI: 10.1016/j.neuint.2020.104792] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/19/2020] [Accepted: 06/21/2020] [Indexed: 01/24/2023]
Abstract
Excitatory Amino Acid Transporters (EAATs) are plasma membrane proteins responsible for maintenance of low extracellular concentrations of glutamate in the CNS. Dysfunction in their activity is implicated in various neurological disorders. Glutamate transport by EAATs occurs through the movement of the central transport domain relative to the scaffold domain in the EAAT membrane protein. Previous studies suggested that residues located within the interface of these two domains in EAAT2, the main subtype of glutamate transporter in the brain, are involved in regulating transport rates. We used mutagenesis, structure-function relationship, surface protein expression and electrophysiology studies, in transfected COS-7 cells and oocytes, to examine residue glycine at position 298, which is located within this interface. Mutation G298A results in increased transport rate without changes in surface expression, suggesting a more hydrophobic and larger alanine results in facilitated transport movement. The increased transport rate does not involve changes in sodium affinity. Electrophysiological currents show that G298A increase both transport and anion currents, suggesting faster transitions through the transport cycle. This work identifies a region critically involved in setting the glutamate transport rate.
Collapse
|
179
|
Hacker UT, Bentler M, Kaniowska D, Morgan M, Büning H. Towards Clinical Implementation of Adeno-Associated Virus (AAV) Vectors for Cancer Gene Therapy: Current Status and Future Perspectives. Cancers (Basel) 2020; 12:E1889. [PMID: 32674264 PMCID: PMC7409174 DOI: 10.3390/cancers12071889] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 02/06/2023] Open
Abstract
Adeno-associated virus (AAV) vectors have gained tremendous attention as in vivo delivery systems in gene therapy for inherited monogenetic diseases. First market approvals, excellent safety data, availability of large-scale production protocols, and the possibility to tailor the vector towards optimized and cell-type specific gene transfer offers to move from (ultra) rare to common diseases. Cancer, a major health burden for which novel therapeutic options are urgently needed, represents such a target. We here provide an up-to-date overview of the strategies which are currently developed for the use of AAV vectors in cancer gene therapy and discuss the perspectives for the future translation of these pre-clinical approaches into the clinic.
Collapse
Affiliation(s)
- Ulrich T. Hacker
- Department of Oncology, Gastroenterology, Hepatology, Pulmonology, and Infectious Diseases, University Cancer Center Leipzig (UCCL), Leipzig University Medical Center, 04103 Leipzig, Germany;
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (M.B.); (M.M.)
| | - Martin Bentler
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (M.B.); (M.M.)
| | - Dorota Kaniowska
- Department of Oncology, Gastroenterology, Hepatology, Pulmonology, and Infectious Diseases, University Cancer Center Leipzig (UCCL), Leipzig University Medical Center, 04103 Leipzig, Germany;
| | - Michael Morgan
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (M.B.); (M.M.)
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Hildegard Büning
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (M.B.); (M.M.)
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Inhoffenstraße 7, 38124 Braunschweig, Germany
| |
Collapse
|
180
|
French LS, Mellough CB, Chen FK, Carvalho LS. A Review of Gene, Drug and Cell-Based Therapies for Usher Syndrome. Front Cell Neurosci 2020; 14:183. [PMID: 32733204 PMCID: PMC7363968 DOI: 10.3389/fncel.2020.00183] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 05/28/2020] [Indexed: 12/12/2022] Open
Abstract
Usher syndrome is a genetic disorder causing neurosensory hearing loss and blindness from retinitis pigmentosa (RP). Adaptive techniques such as braille, digital and optical magnifiers, mobility training, cochlear implants, or other assistive listening devices are indispensable for reducing disability. However, there is currently no treatment to reduce or arrest sensory cell degeneration. There are several classes of treatments for Usher syndrome being investigated. The present article reviews the progress this research has made towards delivering commercial options for patients with Usher syndrome.
Collapse
Affiliation(s)
- Lucy S French
- Centre for Ophthalmology and Visual Sciences (incorporating Lions Eye Institute), The University of Western Australia, Nedlands, WA, Australia
| | - Carla B Mellough
- Centre for Ophthalmology and Visual Sciences (incorporating Lions Eye Institute), The University of Western Australia, Nedlands, WA, Australia
| | - Fred K Chen
- Centre for Ophthalmology and Visual Sciences (incorporating Lions Eye Institute), The University of Western Australia, Nedlands, WA, Australia.,Department of Ophthalmology, Royal Perth Hospital, Perth, WA, Australia.,Department of Ophthalmology, Perth Children's Hospital, Nedlands, WA, Australia
| | - Livia S Carvalho
- Centre for Ophthalmology and Visual Sciences (incorporating Lions Eye Institute), The University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
181
|
Zakharova M. Modern approaches in gene therapy of motor neuron diseases. Med Res Rev 2020; 41:2634-2655. [PMID: 32638429 DOI: 10.1002/med.21705] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 12/12/2022]
Abstract
Motor neuron disorders are a group of neurodegenerative diseases characterized by muscle weakness, loss of ambulation, respiratory insufficiency, leading to an early death. Spinal muscular atrophy (SMA) and amyotrophic lateral sclerosis are the most common and fatal motor neuron diseases. The last 3 years became very successful for novel gene therapy approaches in SMA in infants. Two innovative drugs-nusinersen (Spinraza) and onasemnogene abeparvovec (Zolgensma) have been approved by health authorities. The numerous molecular and genetic overlaps between different neurodegenerative diseases are of great importance in the development of innovative therapeutic strategies, including viral vector therapy and RNA modulating approaches.
Collapse
Affiliation(s)
- Maria Zakharova
- Sixth Neurology Department (Department of Neuroinfectious Diseases), Research Center of Neurology, Moscow, Russia
| |
Collapse
|
182
|
Maximizing lentiviral vector gene transfer in the CNS. Gene Ther 2020; 28:75-88. [PMID: 32632267 PMCID: PMC7902268 DOI: 10.1038/s41434-020-0172-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/20/2020] [Accepted: 06/25/2020] [Indexed: 12/19/2022]
Abstract
Gene transfer is a widely developed technique for studying and treating genetic diseases. However, the development of therapeutic strategies is challenging, due to the cellular and functional complexity of the central nervous system (CNS), its large size and restricted access. We explored two parameters for improving gene transfer efficacy and capacity for the selective targeting of subpopulations of cells with lentiviral vectors (LVs). We first developed a second-generation LV specifically targeting astrocytes for the efficient expression or silencing of genes of interest, and to better study the importance of cell subpopulations in neurological disorders. We then made use of the retrograde transport properties of a chimeric envelope to target brain circuits affected in CNS diseases and achieve a broad distribution. The combination of retrograde transport and specific tropism displayed by this LV provides opportunities for delivering therapeutic genes to specific cell populations and ensuring high levels of transduction in interconnected brain areas following local administration. This new LV and delivery strategy should be of greater therapeutic benefit and opens up new possibilities for the preclinical development of gene therapy for neurodegenerative diseases.
Collapse
|
183
|
Hsu HL, Brown A, Loveland AB, Lotun A, Xu M, Luo L, Xu G, Li J, Ren L, Su Q, Gessler DJ, Wei Y, Tai PWL, Korostelev AA, Gao G. Structural characterization of a novel human adeno-associated virus capsid with neurotropic properties. Nat Commun 2020; 11:3279. [PMID: 32606306 PMCID: PMC7327033 DOI: 10.1038/s41467-020-17047-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 05/27/2020] [Indexed: 02/05/2023] Open
Abstract
Recombinant adeno-associated viruses (rAAVs) are currently considered the safest and most reliable gene delivery vehicles for human gene therapy. Three serotype capsids, AAV1, AAV2, and AAV9, have been approved for commercial use in patients, but they may not be suitable for all therapeutic contexts. Here, we describe a novel capsid identified in a human clinical sample by high-throughput, long-read sequencing. The capsid, which we have named AAVv66, shares high sequence similarity with AAV2. We demonstrate that compared to AAV2, AAVv66 exhibits enhanced production yields, virion stability, and CNS transduction. Unique structural properties of AAVv66 visualized by cryo-EM at 2.5-Å resolution, suggest that critical residues at the three-fold protrusion and at the interface of the five-fold axis of symmetry likely contribute to the beneficial characteristics of AAVv66. Our findings underscore the potential of AAVv66 as a gene therapy vector.
Collapse
Affiliation(s)
- Hung-Lun Hsu
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Alexander Brown
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Anna B Loveland
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Anoushka Lotun
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Meiyu Xu
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Li Luo
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, P. R., China
| | - Guangchao Xu
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, P. R., China
| | - Jia Li
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Lingzhi Ren
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Qin Su
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
- Viral Vector Core, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Dominic J Gessler
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Yuquan Wei
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, P. R., China
| | - Phillip W L Tai
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA.
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA.
| | - Andrei A Korostelev
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA.
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA.
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA.
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
184
|
Taghian T, Horn E, Shazeeb MS, Bierfeldt LJ, Tuominen SM, Koehler J, Fernau D, Bertrand S, Frey S, Cataltepe OI, Gounis MJ, Abayazeed AH, Flotte TR, Sena-Esteves M, Gray-Edwards HL. Volume and Infusion Rate Dynamics of Intraparenchymal Central Nervous System Infusion in a Large Animal Model. Hum Gene Ther 2020; 31:617-625. [PMID: 32363942 DOI: 10.1089/hum.2019.288] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Thalamic infusion of adeno-associated viral (AAV) vectors has been shown to have therapeutic effects in neuronopathic lysosomal storage diseases. Preclinical studies in sheep model of Tay-Sachs disease demonstrated that bilateral thalamic injections of AAV gene therapy are required for maximal benefit. Translation of thalamic injection to patients carries risks in that (1) it has never been done in humans, and (2) dosing scale-up based on brain weight from animals to humans requires injection of larger volumes. To increase the safety margin of this infusion, a flexible cannula was selected to enable simultaneous bilateral thalamic infusion in infants while monitoring by imaging and/or to enable awake infusions for injection of large volumes at low infusion rates. In this study, we tested various infusion volumes (200-800 μL) and rates (0.5-5 μL/min) to determine the maximum tolerated combination of injection parameters. Animals were followed for ∼1 month postinjection with magnetic resonance imaging (MRI) performed at 14 and 28 days. T1-weighted MRI was used to quantify thalamic damage followed by histopathological assessment of the brain. Trends in data show that infusion volumes of 800 μL (2 × the volume required in sheep based on thalamic size) resulted in larger lesions than lower volumes, where the long infusion times (between 13 and 26 h) could have contributed to the generation of larger lesions. The target volume (400 μL, projected to be sufficient to cover most of the sheep thalamus) created the smallest lesion size. Cannula placement alone did result in damage, but this is likely associated with an inherent limitation of its use in a small brain due to the length of the distal rigid portion and lack of stable fixation. An injection rate of 5 μL/min at a volume ∼1/3 of the thalamus (400-600 μL) appears to be well tolerated in sheep both clinically and histopathologically.
Collapse
Affiliation(s)
- Toloo Taghian
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Erin Horn
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Mohammed Salman Shazeeb
- Department of Radiology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Lindsey J Bierfeldt
- Department of Animal Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Susan M Tuominen
- Department of Animal Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Jennifer Koehler
- Department of Pathology, Auburn University, Auburn, Alabama, USA
| | - Deborah Fernau
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Stephanie Bertrand
- Department of Environmental Population Health, Cummings Veterinary School at Tufts University, Grafton, Massachusetts, USA
| | | | - Oguz I Cataltepe
- Department of Neurological Surgery, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Matthew J Gounis
- Department of Radiology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Aly H Abayazeed
- Department of Radiology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Terence R Flotte
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA.,Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Miguel Sena-Esteves
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA.,Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Heather L Gray-Edwards
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA.,Department of Radiology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
185
|
Liu JT, Corbett JL, Heslop JA, Duncan SA. Enhanced genome editing in human iPSCs with CRISPR-CAS9 by co-targeting ATP1a1. PeerJ 2020; 8:e9060. [PMID: 32391204 PMCID: PMC7197401 DOI: 10.7717/peerj.9060] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 04/04/2020] [Indexed: 12/11/2022] Open
Abstract
Genome editing in human induced pluripotent stem cells (iPSCs) provides the potential for disease modeling and cell therapy. By generating iPSCs with specific mutations, researchers can differentiate the modified cells to their lineage of interest for further investigation. However, the low efficiency of targeting in iPSCs has hampered the application of genome editing. In this study we used a CRISPR-Cas9 system that introduces a specific point substitution into the sequence of the Na+/K+-ATPase subunit ATP1A1. The introduced mutation confers resistance to cardiac glycosides, which can then be used to select successfully targeted cells. Using this system, we introduced different formats of donor DNA for homology-directed repair (HDR), including single-strand DNAs, double-strand DNAs, and plasmid donors. We achieved a 35-fold increase in HDR when using plasmid donor with a 400 bp repair template. We further co-targeted ATP1A1 and a second locus of interest to determine the enrichment of mutagenesis after cardiac glycoside selection. Through this approach, INDEL rate was increased after cardiac glycoside treatment, while HDR enrichment was only observed at certain loci. Collectively, these results suggest that a plasmid donor with a 400 bp repair template is an optimal donor DNA for targeted substitution and co-targeting ATP1A1 with the second locus enriches for mutagenesis events through cardiac glycoside selection in human iPSCs.
Collapse
Affiliation(s)
- Jui-Tung Liu
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States of America
| | - James L Corbett
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States of America
| | - James A Heslop
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States of America
| | - Stephen A Duncan
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States of America
| |
Collapse
|
186
|
Engineering adeno-associated virus vectors for gene therapy. Nat Rev Genet 2020; 21:255-272. [DOI: 10.1038/s41576-019-0205-4] [Citation(s) in RCA: 342] [Impact Index Per Article: 68.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2019] [Indexed: 02/06/2023]
|
187
|
Iarkov A, Barreto GE, Grizzell JA, Echeverria V. Strategies for the Treatment of Parkinson's Disease: Beyond Dopamine. Front Aging Neurosci 2020; 12:4. [PMID: 32076403 PMCID: PMC7006457 DOI: 10.3389/fnagi.2020.00004] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/09/2020] [Indexed: 12/11/2022] Open
Abstract
Parkinson’s disease (PD) is the second-leading cause of dementia and is characterized by a progressive loss of dopaminergic neurons in the substantia nigra alongside the presence of intraneuronal α-synuclein-positive inclusions. Therapies to date have been directed to the restoration of the dopaminergic system, and the prevention of dopaminergic neuronal cell death in the midbrain. This review discusses the physiological mechanisms involved in PD as well as new and prospective therapies for the disease. The current data suggest that prevention or early treatment of PD may be the most effective therapeutic strategy. New advances in the understanding of the underlying mechanisms of PD predict the development of more personalized and integral therapies in the years to come. Thus, the development of more reliable biomarkers at asymptomatic stages of the disease, and the use of genetic profiling of patients will surely permit a more effective treatment of PD.
Collapse
Affiliation(s)
- Alexandre Iarkov
- Laboratorio de Neurobiología, Facultad de Ciencias de la Salud, Universidad San Sebastián, Concepción, Chile
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.,Health Research Institute, University of Limerick, Limerick, Ireland
| | - J Alex Grizzell
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado, Boulder, CO, United States
| | - Valentina Echeverria
- Laboratorio de Neurobiología, Facultad de Ciencias de la Salud, Universidad San Sebastián, Concepción, Chile.,Research & Development Service, Bay Pines VA Healthcare System, Bay Pines, FL, United States
| |
Collapse
|
188
|
Nabirotchkin S, Peluffo AE, Rinaudo P, Yu J, Hajj R, Cohen D. Next-generation drug repurposing using human genetics and network biology. Curr Opin Pharmacol 2020; 51:78-92. [PMID: 31982325 DOI: 10.1016/j.coph.2019.12.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/16/2019] [Accepted: 12/19/2019] [Indexed: 12/26/2022]
Abstract
Drug repurposing has attracted increased attention, especially in the context of drug discovery rates that remain too low despite a recent wave of approvals for biological therapeutics (e.g. gene therapy). These new biological entities-based treatments have high costs that are difficult to justify for small markets that include rare diseases. Drug repurposing, involving the identification of single or combinations of existing drugs based on human genetics data and network biology approaches represents a next-generation approach that has the potential to increase the speed of drug discovery at a lower cost. This Pharmacological Perspective reviews progress and perspectives in combining human genetics, especially genome-wide association studies, with network biology to drive drug repurposing for rare and common diseases with monogenic or polygenic etiologies. Also, highlighted here are important features of this next generation approach to drug repurposing, which can be combined with machine learning methods to meet the challenges of personalized medicine.
Collapse
Affiliation(s)
- Serguei Nabirotchkin
- Network Biology & Drug Discovery Department, Pharnext, 11 rue René Jacques, 92130 Issy-les-Moulineaux, France
| | - Alex E Peluffo
- Data Science Department, Pharnext, 11 rue René Jacques, 92130 Issy-les-Moulineaux, France.
| | - Philippe Rinaudo
- Data Science Department, Pharnext, 11 rue René Jacques, 92130 Issy-les-Moulineaux, France
| | - Jinchao Yu
- Data Science Department, Pharnext, 11 rue René Jacques, 92130 Issy-les-Moulineaux, France
| | - Rodolphe Hajj
- Preclinical Research and Pharmacology Department, Pharnext, 11 rue René Jacques, 92130 Issy-les-Moulineaux, France
| | - Daniel Cohen
- Chief Executive Officer, Pharnext, 11 rue René Jacques, 92130 Issy-les-Moulineaux, France
| |
Collapse
|
189
|
Hirbec H, Déglon N, Foo LC, Goshen I, Grutzendler J, Hangen E, Kreisel T, Linck N, Muffat J, Regio S, Rion S, Escartin C. Emerging technologies to study glial cells. Glia 2020; 68:1692-1728. [PMID: 31958188 DOI: 10.1002/glia.23780] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 12/20/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022]
Abstract
Development, physiological functions, and pathologies of the brain depend on tight interactions between neurons and different types of glial cells, such as astrocytes, microglia, oligodendrocytes, and oligodendrocyte precursor cells. Assessing the relative contribution of different glial cell types is required for the full understanding of brain function and dysfunction. Over the recent years, several technological breakthroughs were achieved, allowing "glio-scientists" to address new challenging biological questions. These technical developments make it possible to study the roles of specific cell types with medium or high-content workflows and perform fine analysis of their mutual interactions in a preserved environment. This review illustrates the potency of several cutting-edge experimental approaches (advanced cell cultures, induced pluripotent stem cell (iPSC)-derived human glial cells, viral vectors, in situ glia imaging, opto- and chemogenetic approaches, and high-content molecular analysis) to unravel the role of glial cells in specific brain functions or diseases. It also illustrates the translation of some techniques to the clinics, to monitor glial cells in patients, through specific brain imaging methods. The advantages, pitfalls, and future developments are discussed for each technique, and selected examples are provided to illustrate how specific "gliobiological" questions can now be tackled.
Collapse
Affiliation(s)
- Hélène Hirbec
- Institute for Functional Genomics (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Nicole Déglon
- Laboratory of Neurotherapies and Neuromodulation, Department of Clinical Neuroscience, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland.,Laboratory of Neurotherapies and Neuromodulation, Neuroscience Research Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Lynette C Foo
- Neuroimmunology and Neurodegeneration Section, The Neuroscience and Rare Diseases Discovery and Translational Area, F. Hoffman-La Roche, Basel, Switzerland
| | - Inbal Goshen
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jaime Grutzendler
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Emilie Hangen
- Commissariat à l'Energie Atomique et aux Energies Alternatives, Département de la Recherche Fondamentale, Institut de Biologie François Jacob, MIRCen, Fontenay-aux-Roses, France.,Centre National de la Recherche Scientifique, Neurodegenerative Diseases Laboratory, Université Paris-Sud, Université Paris-Saclay, UMR 9199, Fontenay-aux-Roses, France
| | - Tirzah Kreisel
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nathalie Linck
- Institute for Functional Genomics (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Julien Muffat
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, and Department of Molecular Genetics, The University of Toronto, Toronto, Canada
| | - Sara Regio
- Laboratory of Neurotherapies and Neuromodulation, Department of Clinical Neuroscience, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland.,Laboratory of Neurotherapies and Neuromodulation, Neuroscience Research Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Sybille Rion
- Neuroimmunology and Neurodegeneration Section, The Neuroscience and Rare Diseases Discovery and Translational Area, F. Hoffman-La Roche, Basel, Switzerland
| | - Carole Escartin
- Commissariat à l'Energie Atomique et aux Energies Alternatives, Département de la Recherche Fondamentale, Institut de Biologie François Jacob, MIRCen, Fontenay-aux-Roses, France.,Centre National de la Recherche Scientifique, Neurodegenerative Diseases Laboratory, Université Paris-Sud, Université Paris-Saclay, UMR 9199, Fontenay-aux-Roses, France
| |
Collapse
|
190
|
Chen W, Chen H, Zheng D, Zhang H, Deng L, Cui W, Zhang Y, Santos HA, Shen H. Gene-Hydrogel Microenvironment Regulates Extracellular Matrix Metabolism Balance in Nucleus Pulposus. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1902099. [PMID: 31921568 PMCID: PMC6947697 DOI: 10.1002/advs.201902099] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/18/2019] [Indexed: 05/17/2023]
Abstract
Gene therapy provides an ideal potential treatment for intervertebral disk degeneration by delivering synthetic microRNAs (miRNAs) to regulate the gene expression levels. However, it is very challenging to deliver miRNAs directly, which leads to inactivation, low transfection efficiency, and short half-life. Here, Agomir is loaded in hydrogel to construct a gene-hydrogel microenvironment for regulating the synthesis/catabolism balance of the tissue extracellular matrix (ECM) to treat degenerative diseases. Agomir is a cholesterol-, methylation-, and phosphorothioate-modified miRNA, which can mimic the function of miRNA to regulate the expression of the target gene. Agomir874 that mimics miRNA874 is synthesized to down regulate the expression of matrix metalloproteinases (MMPs) in nucleus pulposus (NP). At the same time, a polyethylene glycol (PEG) hydrogel is synthesized through Ag-S coordination of 4-arm PEG-SH and silver ion solution, which has injectable, self-healing, antimicrobial, degradable, and superabsorbent properties and matches perfectly with the mechanism of intervertebral disk. By delivering Agomir-loaded PEG-hydrogel to a degenerative intervertebral disk, a gene-hydrogel microenvironment is constructed in situ, which reduces the expression of MMPs, regulates the synthesis/catabolism balance of ECM in the NP of the intervertebral disk, and improves the tissue microenvironment regeneration.
Collapse
Affiliation(s)
- Wei Chen
- Department of Spine SurgeryRenji HospitalShanghai JiaoTong University School of Medicine160 Pujian RoadShanghai200127P. R. China
| | - Hao Chen
- Department of Spine SurgeryRenji HospitalShanghai JiaoTong University School of Medicine160 Pujian RoadShanghai200127P. R. China
| | - Dandan Zheng
- Department of Spine SurgeryRenji HospitalShanghai JiaoTong University School of Medicine160 Pujian RoadShanghai200127P. R. China
| | - Hongbo Zhang
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Pharmaceutical Sciences Laboratory and Turku Bioscience CenterÅbo Akademi UniversityTurkuFI‐20520Finland
| | - Lianfu Deng
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Wenguo Cui
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Yuhui Zhang
- Department of Spine SurgeryRenji HospitalShanghai JiaoTong University School of Medicine160 Pujian RoadShanghai200127P. R. China
| | - Hélder A. Santos
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
- Helsinki Institute of Life Science (HiLIFE)University of HelsinkiHelsinkiFI‐00014Finland
| | - Hongxing Shen
- Department of Spine SurgeryRenji HospitalShanghai JiaoTong University School of Medicine160 Pujian RoadShanghai200127P. R. China
| |
Collapse
|
191
|
Davidsson M, Wang G, Aldrin-Kirk P, Cardoso T, Nolbrant S, Hartnor M, Mudannayake J, Parmar M, Björklund T. A systematic capsid evolution approach performed in vivo for the design of AAV vectors with tailored properties and tropism. Proc Natl Acad Sci U S A 2019; 116:27053-27062. [PMID: 31818949 PMCID: PMC6936499 DOI: 10.1073/pnas.1910061116] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Adeno-associated virus (AAV) capsid modification enables the generation of recombinant vectors with tailored properties and tropism. Most approaches to date depend on random screening, enrichment, and serendipity. The approach explored here, called BRAVE (barcoded rational AAV vector evolution), enables efficient selection of engineered capsid structures on a large scale using only a single screening round in vivo. The approach stands in contrast to previous methods that require multiple generations of enrichment. With the BRAVE approach, each virus particle displays a peptide, derived from a protein, of known function on the AAV capsid surface, and a unique molecular barcode in the packaged genome. The sequencing of RNA-expressed barcodes from a single-generation in vivo screen allows the mapping of putative binding sequences from hundreds of proteins simultaneously. Using the BRAVE approach and hidden Markov model-based clustering, we present 25 synthetic capsid variants with refined properties, such as retrograde axonal transport in specific subtypes of neurons, as shown for both rodent and human dopaminergic neurons.
Collapse
Affiliation(s)
- Marcus Davidsson
- Molecular Neuromodulation, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| | - Gang Wang
- Molecular Neuromodulation, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| | - Patrick Aldrin-Kirk
- Molecular Neuromodulation, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| | - Tiago Cardoso
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Lund Stem Cell Center, Lund University, 221 84 Lund, Sweden
| | - Sara Nolbrant
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Lund Stem Cell Center, Lund University, 221 84 Lund, Sweden
| | - Morgan Hartnor
- Molecular Neuromodulation, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| | - Janitha Mudannayake
- Molecular Neuromodulation, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| | - Malin Parmar
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Lund Stem Cell Center, Lund University, 221 84 Lund, Sweden
| | - Tomas Björklund
- Molecular Neuromodulation, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| |
Collapse
|
192
|
Tambuyzer E, Vandendriessche B, Austin CP, Brooks PJ, Larsson K, Miller Needleman KI, Valentine J, Davies K, Groft SC, Preti R, Oprea TI, Prunotto M. Therapies for rare diseases: therapeutic modalities, progress and challenges ahead. Nat Rev Drug Discov 2019; 19:93-111. [PMID: 31836861 DOI: 10.1038/s41573-019-0049-9] [Citation(s) in RCA: 203] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2019] [Indexed: 12/26/2022]
Abstract
Most rare diseases still lack approved treatments despite major advances in research providing the tools to understand their molecular basis, as well as legislation providing regulatory and economic incentives to catalyse the development of specific therapies. Addressing this translational gap is a multifaceted challenge, for which a key aspect is the selection of the optimal therapeutic modality for translating advances in rare disease knowledge into potential medicines, known as orphan drugs. With this in mind, we discuss here the technological basis and rare disease applicability of the main therapeutic modalities, including small molecules, monoclonal antibodies, protein replacement therapies, oligonucleotides and gene and cell therapies, as well as drug repurposing. For each modality, we consider its strengths and limitations as a platform for rare disease therapy development and describe clinical progress so far in developing drugs based on it. We also discuss selected overarching topics in the development of therapies for rare diseases, such as approval statistics, engagement of patients in the process, regulatory pathways and digital tools.
Collapse
Affiliation(s)
- Erik Tambuyzer
- BioPontis Alliance for Rare Diseases Foundation fup/son, Brussels, Belgium. .,BioPontis Alliance Rare Disease Foundation, Inc, Raleigh, NC, USA.
| | - Benjamin Vandendriessche
- Byteflies, Antwerp, Belgium.,Department of Electrical, Computer, and Systems Engineering (ECSE), Case Western Reserve University, Cleveland, OH, USA
| | - Christopher P Austin
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Philip J Brooks
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Kristina Larsson
- Orphan Medicines Office, European Medicines Agency, Amsterdam, Netherlands
| | | | | | - Kay Davies
- MDUK Oxford Neuromuscular Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Stephen C Groft
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Robert Preti
- Hitachi Chemical Regenerative Medicine Business Sector, Allendale, NJ, USA
| | - Tudor I Oprea
- Translational Informatics Division, Department of Internal Medicine, University of New Mexico Albuquerque, Albuquerque, NM, USA.,UNM Comprehensive Cancer Center, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Marco Prunotto
- School of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
193
|
Wu G, Jiang Q, Cui T, Liu X, Gong D, Yin Y, Wang C, Wang T, Lu Y, Zhu D, Han F. The glymphatic system delivery enhances the transduction efficiency of AAV1 to brain endothelial cells in adult mice. J Neurosci Methods 2019; 328:108441. [PMID: 31574288 DOI: 10.1016/j.jneumeth.2019.108441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/23/2019] [Accepted: 09/23/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND Recombinant adeno-associated virus (rAAV) is increasingly applied in neuroscience research or gene therapy. However, there is no simple and efficient tool for specific transfection of rAAV into cerebrovascular tissues. It has been reported that fluorescent tracers or beta-amyloid protein can enter the brain through perivascular spaces, named as "glymphatic system". The purpose of this study was to explore whether rAAV could transduce the cerebral vasculature through the glymphatic pathway. NEW METHOD An AAV1-GFP vector suspension (15 μL) was injected into the intracisternal space of anesthetized mice (n = 2) and 5 μl was injected into the bulbus medullae (n = 2). As controls, 15 μl of artificial cerebrospinal fluid (aCSF) was injected into the cisterna magna. The endothelial specific transduction was verified by Glut1 or PDGFRβ immunofluorescent staining. Immunofluorescence images for all groups were captured with a laser microscope. RESULTS It was observed that infection with rAAV1 vectors encoding green fluorescence protein resulted in a successful cerebrovascular transduction when injected into cisterna magna, compared to aCSF or intra-parenchymal injection at 30 days post-transduction in adult mice. In addition, GFP was co-localized with Glut1 based on immuno-fluorescence. These results indicate that glymphatic system delivery enhances the transduction efficiency of AAV1 to brain endothelial cells. COMPARISON WITH EXISTING METHODS The AAV1 vector can simply and efficiently transduce the cerebral endothelial cells through the glymphatic pathway. CONCLUSION The findings of this study reveal that rAAV1-based vectors have high application potential for endothelial-targeted neurologic disease research or gene-based therapies.
Collapse
Affiliation(s)
- Gang Wu
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China; Department of Pharmacy, Taizhou Hospital of Zhejiang Province, Linhai, Zhejiang, China
| | - Quan Jiang
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Tiantian Cui
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiuxiu Liu
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Dongmei Gong
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China; School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, China
| | - Yixuan Yin
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China; School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, China
| | - Chengkun Wang
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Tiantian Wang
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China; Department of Pharmacy, Run Run Shaw Hospital affiliated to School of Medicine of Zhejiang University, Hangzhou, Zhejiang, China
| | - YingMei Lu
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China; School of Pharmacy, Nanjing Medical University, Nanjing, Jiang Su, China; School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, China
| | - Danyan Zhu
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Feng Han
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China; School of Pharmacy, Nanjing Medical University, Nanjing, Jiang Su, China.
| |
Collapse
|
194
|
Rosenberg JB, Chen A, Kaminsky SM, Crystal RG, Sondhi D. Advances in the Treatment of Neuronal Ceroid Lipofuscinosis. Expert Opin Orphan Drugs 2019; 7:473-500. [PMID: 33365208 PMCID: PMC7755158 DOI: 10.1080/21678707.2019.1684258] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 10/21/2019] [Indexed: 12/27/2022]
Abstract
Neuronal ceroid lipofuscinoses (NCL) represent a class of neurodegenerative disorders involving defective lysosomal processing enzymes or receptors, leading to lysosomal storage disorders, typically characterized by observation of cognitive and visual impairments, epileptic seizures, ataxia, and deterioration of motor skills. Recent success of a biologic (Brineura®) for the treatment of neurologic manifestations of the central nervous system (CNS) has led to renewed interest in therapeutics for NCL, with the goal of ablating or reversing the impact of these devastating disorders. Despite complex challenges associated with CNS therapy, many treatment modalities have been evaluated, including enzyme replacement therapy, gene therapy, stem cell therapy, and small molecule pharmacotherapy. Because the clinical endpoints for the evaluation of candidate therapies are complex and often reliant on subjective clinical scales, the development of quantitative biomarkers for NCLs has become an apparent necessity for the validation of potential treatments. We will discuss the latest findings in the search for relevant biomarkers for assessing disease progression. For this review, we will focus primarily on recent pre-clinical and clinical developments for treatments to halt or cure these NCL diseases. Continued development of current therapies and discovery of newer modalities will be essential for successful therapeutics for NCL. AREAS COVERED The reader will be introduced to the NCL subtypes, natural histories, experimental animal models, and biomarkers for NCL progression; challenges and different therapeutic approaches, and the latest pre-clinical and clinical research for therapeutic development for the various NCLs. This review corresponds to the literatures covering the years from 1968 to mid-2019, but primarily addresses pre-clinical and clinical developments for the treatment of NCL disease in the last decade and as a follow-up to our 2013 review of the same topic in this journal. EXPERT OPINION Much progress has been made in the treatment of neurologic diseases, such as the NCLs, including better animal models and improved therapeutics with better survival outcomes. Encouraging results are being reported at symposiums and in the literature, with multiple therapeutics reaching the clinical trial stage for the NCLs. The potential for a cure could be at hand after many years of trial and error in the preclinical studies. The clinical development of enzyme replacement therapy (Brineura® for CLN2), immunosuppression (CellCept® for CLN3), and gene therapy vectors (for CLN1, CLN2, CLN3, and CLN6) are providing encouragement to families that have a child afflicted with NCL. We believe that successful therapies in the future may involve the combination of two or more therapeutic modalities to provide therapeutic benefit especially as the patients grow older.
Collapse
Affiliation(s)
- Jonathan B Rosenberg
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Alvin Chen
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Stephen M Kaminsky
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Ronald G Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Dolan Sondhi
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
195
|
Haggerty DL, Grecco GG, Reeves KC, Atwood B. Adeno-Associated Viral Vectors in Neuroscience Research. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 17:69-82. [PMID: 31890742 PMCID: PMC6931098 DOI: 10.1016/j.omtm.2019.11.012] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Adeno-associated viral vectors (AAVs) are increasingly useful preclinical tools in neuroscience research studies for interrogating cellular and neurocircuit functions and mapping brain connectivity. Clinically, AAVs are showing increasing promise as viable candidates for treating multiple neurological diseases. Here, we briefly review the utility of AAVs in mapping neurocircuits, manipulating neuronal function and gene expression, and activity labeling in preclinical research studies as well as AAV-based gene therapies for diseases of the nervous system. This review highlights the vast potential that AAVs have for transformative research and therapeutics in the neurosciences.
Collapse
Affiliation(s)
- David L. Haggerty
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Gregory G. Grecco
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Medical Scientist Training Program, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kaitlin C. Reeves
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Brady Atwood
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Stark Neurosciences Research Institute, Indianapolis, IN 46202, USA
- Corresponding author: Brady Atwood, PhD, Department of Pharmacology & Toxicology, Indiana University School of Medicine, 320 West 15th Street, NB-400C, Indianapolis, IN 46202, USA.
| |
Collapse
|
196
|
Ferreira LMR, Muller YD, Bluestone JA, Tang Q. Next-generation regulatory T cell therapy. Nat Rev Drug Discov 2019; 18:749-769. [PMID: 31541224 PMCID: PMC7773144 DOI: 10.1038/s41573-019-0041-4] [Citation(s) in RCA: 323] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2019] [Indexed: 02/08/2023]
Abstract
Regulatory T cells (Treg cells) are a small subset of immune cells that are dedicated to curbing excessive immune activation and maintaining immune homeostasis. Accordingly, deficiencies in Treg cell development or function result in uncontrolled immune responses and tissue destruction and can lead to inflammatory disorders such as graft-versus-host disease, transplant rejection and autoimmune diseases. As Treg cells deploy more than a dozen molecular mechanisms to suppress immune responses, they have potential as multifaceted adaptable smart therapeutics for treating inflammatory disorders. Indeed, early-phase clinical trials of Treg cell therapy have shown feasibility, tolerability and potential efficacy in these disease settings. In the meantime, progress in the development of chimeric antigen receptors and in genome editing (including the application of CRISPR-Cas9) over the past two decades has facilitated the genetic optimization of primary T cell therapy for cancer. These technologies are now being used to enhance the specificity and functionality of Treg cells. In this Review, we describe the key advances and prospects in designing and implementing Treg cell-based therapy in autoimmunity and transplantation.
Collapse
Affiliation(s)
- Leonardo M R Ferreira
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
- Sean N. Parker Autoimmune Research Laboratory, University of California, San Francisco, San Francisco, CA, USA
| | - Yannick D Muller
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Jeffrey A Bluestone
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA.
- Sean N. Parker Autoimmune Research Laboratory, University of California, San Francisco, San Francisco, CA, USA.
| | - Qizhi Tang
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA.
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
197
|
Cappella M, Ciotti C, Cohen-Tannoudji M, Biferi MG. Gene Therapy for ALS-A Perspective. Int J Mol Sci 2019; 20:E4388. [PMID: 31500113 PMCID: PMC6771059 DOI: 10.3390/ijms20184388] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/30/2019] [Accepted: 09/03/2019] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal motor neuron disease (MND) with no cure. Recent advances in gene therapy open a new perspective to treat this disorder-particularly for the characterized genetic forms. Gene therapy approaches, involving the delivery of antisense oligonucleotides into the central nervous system (CNS) are being tested in clinical trials for patients with mutations in SOD1 or C9orf72 genes. Viral vectors can be used to deliver therapeutic sequences to stably transduce motor neurons in the CNS. Vectors derived from adeno-associated virus (AAV), can efficiently target genes and have been tested in several pre-clinical settings with promising outcomes. Recently, the Food and Drug Administration (FDA) approved Zolgensma, an AAV-mediated treatment for another MND-the infant form of spinal muscular atrophy. Given the accelerated progress in gene therapy, it is potentially a promising avenue to develop an efficient and safe cure for ALS.
Collapse
Affiliation(s)
- Marisa Cappella
- Sorbonne Université, Inserm UMRS 974, Centre of Research in Myology (CRM), Institut de Myologie, GH Pitié Salpêtrière, 75013 Paris, France
| | - Chiara Ciotti
- Sorbonne Université, Inserm UMRS 974, Centre of Research in Myology (CRM), Institut de Myologie, GH Pitié Salpêtrière, 75013 Paris, France
| | - Mathilde Cohen-Tannoudji
- Sorbonne Université, Inserm UMRS 974, Centre of Research in Myology (CRM), Institut de Myologie, GH Pitié Salpêtrière, 75013 Paris, France
| | - Maria Grazia Biferi
- Sorbonne Université, Inserm UMRS 974, Centre of Research in Myology (CRM), Institut de Myologie, GH Pitié Salpêtrière, 75013 Paris, France.
| |
Collapse
|
198
|
Shen X, Beasley S, Putman JN, Li Y, Prakash TP, Rigo F, Napierala M, Corey DR. Efficient electroporation of neuronal cells using synthetic oligonucleotides: identifying duplex RNA and antisense oligonucleotide activators of human frataxin expression. RNA (NEW YORK, N.Y.) 2019; 25:1118-1129. [PMID: 31151992 PMCID: PMC6800520 DOI: 10.1261/rna.071290.119] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 05/23/2019] [Indexed: 05/08/2023]
Abstract
Oligonucleotide drugs are experiencing greater success in the clinic, encouraging the initiation of new projects. Resources are insufficient to develop every potentially important project, and persuasive experimental data using cell lines close to disease target tissue is needed to prioritize candidates. Friedreich's ataxia (FRDA) is a devastating and currently incurable disease caused by insufficient expression of the enzyme frataxin (FXN). We have previously shown that synthetic nucleic acids can activate FXN expression in human patient-derived fibroblast cells. We chose to further test these compounds in induced pluripotent stem cell-derived neuronal progenitor cells (iPSC-NPCs). Here we describe methods to deliver oligonucleotides and duplex RNAs into iPSC-NPCs using electroporation. Activation of FXN expression is potent, easily reproducible, and potencies parallel those determined using patient-derived fibroblast cells. A duplex RNA and several antisense oligonucleotides (ASOs) with different combinations of 2'-methoxyethyl (2'-MOE), 2'-fluoro (2'-F), and constrained ethyl (cEt) were active, providing multiple starting points for further development and highlighting improved potency as an important goal for preclinical development. Our data support the conclusion that ASO-mediated activation of FXN is a feasible approach for treating FRDA and that electroporation is a robust method for introducing ASOs to modulate gene expressions in neuronal cells.
Collapse
Affiliation(s)
- Xiulong Shen
- Departments of Pharmacology and Biochemistry, UT Southwestern Medical Center at Dallas, Dallas, Texas 75390, USA
| | | | - Jennifer N Putman
- Departments of Pharmacology and Biochemistry, UT Southwestern Medical Center at Dallas, Dallas, Texas 75390, USA
| | - Yanjie Li
- Department of Biochemistry and Molecular Genetics, UAB Stem Cell Institute, University of Alabama, Birmingham, Alabama 35294, USA
| | | | - Frank Rigo
- Ionis Pharmaceuticals, Carlsbad, California 92010, USA
| | - Marek Napierala
- Department of Biochemistry and Molecular Genetics, UAB Stem Cell Institute, University of Alabama, Birmingham, Alabama 35294, USA
| | - David R Corey
- Departments of Pharmacology and Biochemistry, UT Southwestern Medical Center at Dallas, Dallas, Texas 75390, USA
| |
Collapse
|
199
|
Divergent engagements between adeno-associated viruses with their cellular receptor AAVR. Nat Commun 2019; 10:3760. [PMID: 31434885 PMCID: PMC6704107 DOI: 10.1038/s41467-019-11668-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 07/27/2019] [Indexed: 12/12/2022] Open
Abstract
Adeno-associated virus (AAV) receptor (AAVR) is an essential receptor for the entry of multiple AAV serotypes with divergent rules; however, the mechanism remains unclear. Here, we determine the structures of the AAV1-AAVR and AAV5-AAVR complexes, revealing the molecular details by which PKD1 recognizes AAV5 and PKD2 is solely engaged with AAV1. PKD2 lies on the plateau region of the AAV1 capsid. However, the AAV5-AAVR interface is strikingly different, in which PKD1 is bound at the opposite side of the spike of the AAV5 capsid than the PKD2-interacting region of AAV1. Residues in strands F/G and the CD loop of PKD1 interact directly with AAV5, whereas residues in strands B/C/E and the BC loop of PKD2 make contact with AAV1. These findings further the understanding of the distinct mechanisms by which AAVR recognizes various AAV serotypes and provide an example of a single receptor engaging multiple viral serotypes with divergent rules. Multiple adeno-associated viruses (AAV) use the same receptor (AAVR), but the binding mode is not clear. Here, the authors determine the structures of the AAV1-AAVR and AAV5-AAVR complexes, identify residues necessary for virus entry and compare the receptor interfaces of different AAV capsids.
Collapse
|
200
|
Foffani G, Trigo‐Damas I, Pineda‐Pardo JA, Blesa J, Rodríguez‐Rojas R, Martínez‐Fernández R, Obeso JA. Focused ultrasound in Parkinson's disease: A twofold path toward disease modification. Mov Disord 2019; 34:1262-1273. [DOI: 10.1002/mds.27805] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/14/2019] [Accepted: 06/27/2019] [Indexed: 12/18/2022] Open
Affiliation(s)
- Guglielmo Foffani
- CINACHospital Universitario HM Puerta del Sur, Móstoles, Universidad CEU‐San Pablo Madrid Spain
- Hospital Nacional de Parapléjicos Toledo Spain
| | - Inés Trigo‐Damas
- CINACHospital Universitario HM Puerta del Sur, Móstoles, Universidad CEU‐San Pablo Madrid Spain
- CIBERNEDInstituto de Salud Carlos III Madrid Spain
| | - José A. Pineda‐Pardo
- CINACHospital Universitario HM Puerta del Sur, Móstoles, Universidad CEU‐San Pablo Madrid Spain
- CIBERNEDInstituto de Salud Carlos III Madrid Spain
| | - Javier Blesa
- CINACHospital Universitario HM Puerta del Sur, Móstoles, Universidad CEU‐San Pablo Madrid Spain
- CIBERNEDInstituto de Salud Carlos III Madrid Spain
| | - Rafael Rodríguez‐Rojas
- CINACHospital Universitario HM Puerta del Sur, Móstoles, Universidad CEU‐San Pablo Madrid Spain
- CIBERNEDInstituto de Salud Carlos III Madrid Spain
| | - Raul Martínez‐Fernández
- CINACHospital Universitario HM Puerta del Sur, Móstoles, Universidad CEU‐San Pablo Madrid Spain
- CIBERNEDInstituto de Salud Carlos III Madrid Spain
| | - José A. Obeso
- CINACHospital Universitario HM Puerta del Sur, Móstoles, Universidad CEU‐San Pablo Madrid Spain
- CIBERNEDInstituto de Salud Carlos III Madrid Spain
| |
Collapse
|