1
|
Ye D, Chukwu C, Yang Y, Hu Z, Chen H. Adeno-associated virus vector delivery to the brain: Technology advancements and clinical applications. Adv Drug Deliv Rev 2024; 211:115363. [PMID: 38906479 DOI: 10.1016/j.addr.2024.115363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 05/13/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024]
Abstract
Adeno-associated virus (AAV) vectors have emerged as a promising tool in the development of gene therapies for various neurological diseases, including Alzheimer's disease and Parkinson's disease. However, the blood-brain barrier (BBB) poses a significant challenge to successfully delivering AAV vectors to the brain. Strategies that can overcome the BBB to improve the AAV delivery efficiency to the brain are essential to successful brain-targeted gene therapy. This review provides an overview of existing strategies employed for AAV delivery to the brain, including direct intraparenchymal injection, intra-cerebral spinal fluid injection, intranasal delivery, and intravenous injection of BBB-permeable AAVs. Focused ultrasound has emerged as a promising technology for the noninvasive and spatially targeted delivery of AAV administered by intravenous injection. This review also summarizes each strategy's current preclinical and clinical applications in treating neurological diseases. Moreover, this review includes a detailed discussion of the recent advances in the emerging focused ultrasound-mediated AAV delivery. Understanding the state-of-the-art of these gene delivery approaches is critical for future technology development to fulfill the great promise of AAV in neurological disease treatment.
Collapse
Affiliation(s)
- Dezhuang Ye
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Chinwendu Chukwu
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Yaoheng Yang
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Zhongtao Hu
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Hong Chen
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA; Department of Neurosurgery, Washington University School of Medicine, Saint Louis, MO 63110 USA; Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| |
Collapse
|
2
|
Heiss JD, Ray-Chaudhury A, Kleiner DE, Ehrlich D, Scott G, Edwards NA, Goldstein DS, Hammoud DA, Hadaczek P, Van Laar VS, Graff S, Herscovitch P, Lungu C, Hallett M, Lonser RR, Zaghloul K, Bankiewicz KS. Persistent GDNF Expression 45 Months after Putaminal Infusion of AAV2-GDNF in a Patient with Parkinson's Disease. Mov Disord 2024; 39:1412-1417. [PMID: 38718138 PMCID: PMC11341257 DOI: 10.1002/mds.29820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 04/08/2024] [Indexed: 08/23/2024] Open
Abstract
OBJECTIVE Gene therapy by convection-enhanced delivery of type 2 adeno-associated virus-glial cell derived neurotrophic factor (AAV2-GDNF) to the bilateral putamina seeks to increase GDNF gene expression and treat Parkinson's disease (PD). METHODS A 63-year-old man with advanced PD received AAV2-GDNF in a clinical trial. He died from pneumonia after anterior cervical discectomy and fusion 45 months later. An autopsy included brain examination for GDNF transgene expression. Putaminal catecholamine concentrations were compared to in vivo 18F-Fluorodopa (18F-FDOPA) positron emission tomography (PET) scanning results before and 18 months after AAV2-GDNF infusion. RESULTS Parkinsonian progression stabilized clinically. Postmortem neuropathology confirmed PD. Bilateral putaminal regions previously infused with AAV2-GDNF expressed the GDNF gene. Total putaminal dopamine was 1% of control, confirming the striatal dopaminergic deficiency suggested by baseline 18F-DOPA-PET scanning. Putaminal regions responded as expected to AAV2-GDNF. CONCLUSION After AAV2-GDNF infusion, infused putaminal regions showed increased GDNF gene expression, tyrosine hydroxylase immunoreactive sprouting, catechol levels, and 18F-FDOPA-PET signal, suggesting the regenerative potential of AAV2-GDNF in PD.
Collapse
Affiliation(s)
- John D. Heiss
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Abhik Ray-Chaudhury
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - David E. Kleiner
- Laboratory of Pathology, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD 20892
| | - Debra Ehrlich
- Parkinson’s Disease Clinic, Office of the Clinical Director, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Gretchen Scott
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Nancy A. Edwards
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - David S. Goldstein
- Autonomic Medicine Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Dima A. Hammoud
- Center for Infectious Disease Imaging, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Piotr Hadaczek
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Victor S. Van Laar
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Shantelle Graff
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Peter Herscovitch
- Positron Emission Tomography Department, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Codrin Lungu
- Division of Clinical Research, and Office of the Clinical Director, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Mark Hallett
- Human Motor Control Section, Medical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Russell R. Lonser
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Kareem Zaghloul
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Krystof S. Bankiewicz
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
3
|
Patel RV, Nanda P, Richardson RM. Neurosurgical gene therapy for central nervous system diseases. Neurotherapeutics 2024; 21:e00434. [PMID: 39191071 PMCID: PMC11445594 DOI: 10.1016/j.neurot.2024.e00434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 07/16/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
Viral vector mediated gene therapies for neurodegenerative and neurodevelopmental conditions that require neurosurgical administration continue to expand. We systematically reviewed the National Institutes of Health (NIH) ClinicalTrials.gov database to identify all clinical trials studying in-vivo viral vector mediated gene therapies targeted to the CNS for neurodegenerative and neurodevelopmental diseases. We isolated studies which delivered therapies using neurosurgical approaches: intracisternal, intraventricular, and/or intraparenchymal. Clinical trials primarily registered in international countries were included if they were referenced by an NIH registered clinical trial. We performed a scoping review to identify the preclinical studies that supported each human clinical trial. Key preclinical and clinical data were aggregated to characterize vector capsid design, delivery methods, gene expression profile, and clinical benefit. A total of 64 clinical trials were identified in active, completed, terminated, and long-term follow-up stages. A range of CNS conditions across pediatric and adult populations are being studied with CNS targeted viral vector gene therapy, including Alzheimer's disease, Parkinson's disease, AADC deficiency, sphingolipidoses, mucopolysaccharidoses, neuronal ceroid lipofuscinoses, spinal muscular atrophy, adrenoleukodystrophy, Canavan disease, frontotemporal dementia, Huntington's disease, Rett syndrome, Dravet syndrome, mesial temporal lobe epilepsy, and glutaric acidemia. Adeno-associated viral vectors (AAVs) were utilized by the majority of tested therapies, with vector serotypes, regulatory elements, delivery methods, and vector monitoring varying based on the disease being studied. Intraparenchymal delivery has evolved significantly, with MRI-guided convection-enhanced delivery established as a gold standard method for pioneering novel gene targets.
Collapse
Affiliation(s)
- Ruchit V Patel
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Pranav Nanda
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - R Mark Richardson
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
Lee MH, Um KH, Lee SW, Sun YJ, Gu DH, Jo YO, Kim SH, Seol W, Hwang H, Baek K, Choi JW. Bi-directional regulation of AIMP2 and its splice variant on PARP-1-dependent neuronal cell death; Therapeutic implication for Parkinson's disease. Acta Neuropathol Commun 2024; 12:5. [PMID: 38172953 PMCID: PMC10765824 DOI: 10.1186/s40478-023-01697-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/28/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Parthanatos represents a critical molecular aspect of Parkinson's disease, wherein AIMP2 aberrantly activates PARP-1 through direct physical interaction. Although AIMP2 ought to be a therapeutic target for the disease, regrettably, it is deemed undruggable due to its non-enzymatic nature and predominant localization within the tRNA synthetase multi-complex. Instead, AIMP2 possesses an antagonistic splice variant, designated DX2, which counteracts AIMP2-induced apoptosis in the p53 or inflammatory pathway. Consequently, we examined whether DX2 competes with AIMP2 for PARP-1 activation and is therapeutically effective in Parkinson's disease. METHODS The binding affinity of AIMP2 and DX2 to PARP-1 was contrasted through immunoprecipitation. The efficacy of DX2 in neuronal cell death was assessed under 6-OHDA and H2O2 in vitro conditions. Additionally, endosomal and exosomal activity of synaptic vesicles was gauged in AIMP2 or DX2 overexpressed hippocampal primary neurons utilizing optical live imaging with VAMP-vGlut1 probes. To ascertain the role of DX2 in vivo, rotenone-induced behavioral alterations were compared between wild-type and DX2 transgenic animals. A DX2-encoding self-complementary adeno-associated virus (scAAV) was intracranially injected into 6-OHDA induced in vivo animal models, and their mobility was examined. Subsequently, the isolated brain tissues were analyzed. RESULTS DX2 translocates into the nucleus upon ROS stress more rapidly than AIMP2. The binding affinity of DX2 to PARP-1 appeared to be more robust compared to that of AIMP2, resulting in the inhibition of PARP-1 induced neuronal cell death. DX2 transgenic animals exhibited neuroprotective behavior in rotenone-induced neuronal damage conditions. Following a single intracranial injection of AAV-DX2, both behavior and mobility were consistently ameliorated in neurodegenerative animal models induced by 6-OHDA. CONCLUSION AIMP2 and DX2 are proposed to engage in bidirectional regulation of parthanatos. They physically interact with PARP-1. Notably, DX2's cell survival properties manifest exclusively in the context of abnormal AIMP2 accumulation, devoid of any tumorigenic effects. This suggests that DX2 could represent a distinctive therapeutic target for addressing Parkinson's disease in patients.
Collapse
Affiliation(s)
- Min Hak Lee
- Department of Pharmacology, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Biomedical and Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Regulatory Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Ki-Hwan Um
- Department of Biomedical and Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Regulatory Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Seok Won Lee
- Department of Pharmacology, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Biomedical and Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Ye Ji Sun
- Department of Pharmacology, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Biomedical and Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Regulatory Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Da-Hye Gu
- Department of Pharmacology, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Biomedical and Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Young Ok Jo
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Sung Hyun Kim
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Physiology, School of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Wongi Seol
- InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Sanbonro 321, Gunposi, Gyeonggido, 15865, Republic of Korea
| | - Hyorin Hwang
- Generoath Ltd., Seoul, 04168, Republic of Korea
- Department of Pharmacology, College of Dentistry and Research Institute of Oral Science, Gangneung-Wonju National University, Gangneung, Gangwon-Do, 25457, Republic of Korea
| | - Kyunghwa Baek
- Department of Pharmacology, College of Dentistry and Research Institute of Oral Science, Gangneung-Wonju National University, Gangneung, Gangwon-Do, 25457, Republic of Korea
| | - Jin Woo Choi
- Department of Pharmacology, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea.
- Department of Biomedical and Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.
- Department of Regulatory Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
5
|
Ford MM, George BE, Van Laar VS, Holleran KM, Naidoo J, Hadaczek P, Vanderhooft LE, Peck EG, Dawes MH, Ohno K, Bringas J, McBride JL, Samaranch L, Forsayeth JR, Jones SR, Grant KA, Bankiewicz KS. GDNF gene therapy for alcohol use disorder in male non-human primates. Nat Med 2023; 29:2030-2040. [PMID: 37580533 PMCID: PMC10602124 DOI: 10.1038/s41591-023-02463-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 06/15/2023] [Indexed: 08/16/2023]
Abstract
Alcohol use disorder (AUD) exacts enormous personal, social and economic costs globally. Return to alcohol use in treatment-seeking patients with AUD is common, engendered by a cycle of repeated abstinence-relapse episodes even with use of currently available pharmacotherapies. Repeated ethanol use induces dopaminergic signaling neuroadaptations in ventral tegmental area (VTA) neurons of the mesolimbic reward pathway, and sustained dysfunction of reward circuitry is associated with return to drinking behavior. We tested this hypothesis by infusing adeno-associated virus serotype 2 vector encoding human glial-derived neurotrophic factor (AAV2-hGDNF), a growth factor that enhances dopaminergic neuron function, into the VTA of four male rhesus monkeys, with another four receiving vehicle, following induction of chronic alcohol drinking. GDNF expression ablated the return to alcohol drinking behavior over a 12-month period of repeated abstinence-alcohol reintroduction challenges. This behavioral change was accompanied by neurophysiological modulations to dopamine signaling in the nucleus accumbens that countered the hypodopaminergic signaling state associated with chronic alcohol use, indicative of a therapeutic modulation of limbic circuits countering the effects of alcohol. These preclinical findings suggest gene therapy targeting relapse prevention may be a potential therapeutic strategy for AUD.
Collapse
Affiliation(s)
- Matthew M Ford
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
- Department of Psychology, Lewis & Clark College, Portland, OR, USA
| | - Brianna E George
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Victor S Van Laar
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | - Katherine M Holleran
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Jerusha Naidoo
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Piotr Hadaczek
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Lauren E Vanderhooft
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Emily G Peck
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Monica H Dawes
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Kousaku Ohno
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - John Bringas
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Jodi L McBride
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Lluis Samaranch
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - John R Forsayeth
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Sara R Jones
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Kathleen A Grant
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA.
| | - Krystof S Bankiewicz
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA.
- Department of Neurological Surgery, University of California, San Francisco, CA, USA.
| |
Collapse
|
6
|
Meyer AH, Feldsien TM, Mezler M, Untucht C, Venugopalan R, Lefebvre DR. Novel Developments to Enable Treatment of CNS Diseases with Targeted Drug Delivery. Pharmaceutics 2023; 15:pharmaceutics15041100. [PMID: 37111587 PMCID: PMC10145602 DOI: 10.3390/pharmaceutics15041100] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/07/2023] [Accepted: 03/17/2023] [Indexed: 04/29/2023] Open
Abstract
The blood-brain barrier (BBB) is a major hurdle for the development of systemically delivered drugs against diseases of the central nervous system (CNS). Because of this barrier there is still a huge unmet need for the treatment of these diseases, despite years of research efforts across the pharmaceutical industry. Novel therapeutic entities, such as gene therapy and degradomers, have become increasingly popular in recent years, but have not been the focus for CNS indications so far. To unfold their full potential for the treatment of CNS diseases, these therapeutic entities will most likely have to rely on innovative delivery technologies. Here we will describe and assess approaches, both invasive and non-invasive, that can enable, or at least increase, the probability of a successful drug development of such novel therapeutics for CNS indications.
Collapse
Affiliation(s)
- Axel H Meyer
- Quantitative, Translational & ADME Sciences, AbbVie Deutschland GmbH & Co. KG, Knollstraße, 67061 Ludwigshafen, Germany
| | - Thomas M Feldsien
- Drug Delivery and Combination Products, Development Sciences, AbbVie Inc., 1 N Waukegan Road, North Chicago, IL 60064, USA
| | - Mario Mezler
- Quantitative, Translational & ADME Sciences, AbbVie Deutschland GmbH & Co. KG, Knollstraße, 67061 Ludwigshafen, Germany
| | - Christopher Untucht
- Neuroscience Discovery, AbbVie Deutschland GmbH & Co. KG, Knollstraße, 67061 Ludwigshafen, Germany
| | - Ramakrishna Venugopalan
- Drug Delivery and Combination Products, Development Sciences, AbbVie Inc., 1 N Waukegan Road, North Chicago, IL 60064, USA
| | - Didier R Lefebvre
- Drug Delivery and Combination Products, Development Sciences, AbbVie Inc., 1 N Waukegan Road, North Chicago, IL 60064, USA
| |
Collapse
|
7
|
Nance E, Pun SH, Saigal R, Sellers DL. Drug delivery to the central nervous system. NATURE REVIEWS. MATERIALS 2022; 7:314-331. [PMID: 38464996 PMCID: PMC10923597 DOI: 10.1038/s41578-021-00394-w] [Citation(s) in RCA: 96] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/05/2021] [Indexed: 03/12/2024]
Abstract
Despite the rising global incidence of central nervous system (CNS) disorders, CNS drug development remains challenging, with high costs, long pathways to clinical use and high failure rates. The CNS is highly protected by physiological barriers, in particular, the blood-brain barrier and the blood-cerebrospinal fluid barrier, which limit access of most drugs. Biomaterials can be designed to bypass or traverse these barriers, enabling the controlled delivery of drugs into the CNS. In this Review, we first examine the effects of normal and diseased CNS physiology on drug delivery to the brain and spinal cord. We then discuss CNS drug delivery designs and materials that are administered systemically, directly to the CNS, intranasally or peripherally through intramuscular injections. Finally, we highlight important challenges and opportunities for materials design for drug delivery to the CNS and the anticipated clinical impact of CNS drug delivery.
Collapse
Affiliation(s)
- Elizabeth Nance
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
- These authors contributed equally: Elizabeth Nance, Suzie H. Pun, Rajiv Saigal, Drew L. Sellers
| | - Suzie H. Pun
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- These authors contributed equally: Elizabeth Nance, Suzie H. Pun, Rajiv Saigal, Drew L. Sellers
| | - Rajiv Saigal
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
- These authors contributed equally: Elizabeth Nance, Suzie H. Pun, Rajiv Saigal, Drew L. Sellers
| | - Drew L. Sellers
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- These authors contributed equally: Elizabeth Nance, Suzie H. Pun, Rajiv Saigal, Drew L. Sellers
| |
Collapse
|
8
|
Lonser RR, Akhter AS, Zabek M, Elder JB, Bankiewicz KS. Direct convective delivery of adeno-associated virus gene therapy for treatment of neurological disorders. J Neurosurg 2021; 134:1751-1763. [PMID: 32915526 DOI: 10.3171/2020.4.jns20701] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/16/2020] [Indexed: 11/06/2022]
Abstract
Molecular biological insights have led to a fundamental understanding of the underlying genomic mechanisms of nervous system disease. These findings have resulted in the identification of therapeutic genes that can be packaged in viral capsids for the treatment of a variety of neurological conditions, including neurodegenerative, metabolic, and enzyme deficiency disorders. Recent data have demonstrated that gene-carrying viral vectors (most often adeno-associated viruses) can be effectively distributed by convection-enhanced delivery (CED) in a safe, reliable, targeted, and homogeneous manner across the blood-brain barrier. Critically, these vectors can be monitored using real-time MRI of a co-infused surrogate tracer to accurately predict vector distribution and transgene expression at the perfused site. The unique properties of CED of adeno-associated virus vectors allow for cell-specific transgene manipulation of the infused anatomical site and/or widespread interconnected sites via antero- and/or retrograde transport. The authors review the convective properties of viral vectors, associated technology, and clinical applications.
Collapse
Affiliation(s)
- Russell R Lonser
- 1Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio; and
| | - Asad S Akhter
- 1Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio; and
| | - Mirosław Zabek
- 2Department of Neurological Surgery, Bródno Hospital, Warsaw, Poland
| | - J Bradley Elder
- 1Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio; and
| | - Krystof S Bankiewicz
- 1Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio; and
| |
Collapse
|
9
|
Wang J, Zhang L. Retrograde Axonal Transport Property of Adeno-Associated Virus and Its Possible Application in Future. Microbes Infect 2021; 23:104829. [PMID: 33878458 DOI: 10.1016/j.micinf.2021.104829] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/30/2021] [Accepted: 04/05/2021] [Indexed: 12/19/2022]
Abstract
Gene therapy has become a treatment method for many diseases. Adeno-associated virus (AAV) is one of the most common virus vectors, is also widely used in the gene therapy field. During the past 2 decades, the retrograde axonal transportability of AAV has been discovered and utilized. Many studies have worked on the retrograde axonal transportability of AAV, and more and more people are interested in this field. This review described the current application, influence factors, and mechanism of retrograde axonal transportability of AAV and predicted its potential use in disease treatment in near future.
Collapse
Affiliation(s)
- Jingjing Wang
- Department of Gastroenterology, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin, 300170, China
| | - Liqin Zhang
- Department of Otolaryngology, Peking Union Medical College Hospital, Dongcheng Qu, Beijing, 100730, China.
| |
Collapse
|
10
|
Terse PS, Kells AP, Noker P, Wright JF, Bankiewicz KS. Safety Assessment of AAV2-hGDNF Administered Via Intracerebral Injection in Rats for Treatment of Parkinson's Disease. Int J Toxicol 2021; 40:4-14. [PMID: 33131343 PMCID: PMC8171122 DOI: 10.1177/1091581820966315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is a potent neuroprotective biologic in Parkinson's disease models. Adeno-associated viral vector serotype 2 (AAV2)-human GDNF safety was assessed in rats treated with a single intracerebral dose of vehicle, 6.8 × 108, 6.8 × 109, or 5.2 × 1010 vector genomes (vg)/dose followed by interim sacrifices on day 7, 31, 90, and 376. There were no treatment-related effects observed on food consumption, body weight, hematology, clinical chemistry, coagulation parameters, neurobehavioral parameters, organ weights, or serum GDNF and anti-GDNF antibody levels. Increased serum anti-AAV2 neutralizing antibody titers were observed in the 5.2 × 1010 vg/dose group. Histopathological lesions were observed at the injection site in the 6.8 × 109 vg/dose (day 7) and 5.2 × 1010 vg/dose groups (days 7 and 31) and consisted of gliosis, mononuclear perivascular cuffing, intranuclear inclusion bodies, and/or apoptosis on day 7 and mononuclear perivascular cuffing on day 31. GDNF immunostaining was observed in the injection site in all dose groups through day 376 indicating no detectable impacts of anti-AAV2 neutralizing antibody. There was no evidence of increased expression of calcitonin gene-related peptide or Swann cell hyperplasia in the cervical and lumbar spinal cord or medulla oblongata at the 5.2 × 1010 vg/dose level indicating lack of hyperplastic effects. In conclusion, no systemic toxicity was observed, and the local toxicity observed at the injection site appeared to be reversible demonstrating a promising safety profile of intracerebral AAV2-GDNF delivery. Furthermore, an intracerebral dose of 6.8 × 108 AAV2-GDNF vg/dose was considered to be a no observed adverse effect level in rats.
Collapse
Affiliation(s)
- Pramod S. Terse
- National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | | | | | - J Fraser Wright
- Center for Definitive and Curative Medicine, Stanford University School of Medicine, CA, USA
| | | |
Collapse
|
11
|
Abstract
Multiple system atrophy (MSA) is a progressive neurodegenerative disease variably associated with motor, nonmotor, and autonomic symptoms, resulting from putaminal and cerebellar degeneration and associated with glial cytoplasmic inclusions enriched with α-synuclein in oligodendrocytes and neurons. Although symptomatic treatment of MSA can provide significant improvements in quality of life, the benefit is often partial, limited by adverse effects, and fails to treat the underlying cause. Consistent with the multisystem nature of the disease and evidence that motor symptoms, autonomic failure, and depression drive patient assessments of quality of life, treatment is best achieved through a coordinated multidisciplinary approach driven by the patient's priorities and goals of care. Research into disease-modifying therapies is ongoing with a particular focus on synuclein-targeted therapies among others. This review focuses on both current management and emerging therapies for this devastating disease.
Collapse
Affiliation(s)
- Matthew R. Burns
- Norman Fixel Institute for Neurological Diseases at UFHealth, Movement Disorders Division, Department of Neurology, University of Florida, 3009 SW Williston Rd, Gainesville, FL 32608 USA
| | - Nikolaus R. McFarland
- Norman Fixel Institute for Neurological Diseases at UFHealth, Movement Disorders Division, Department of Neurology, University of Florida, 3009 SW Williston Rd, Gainesville, FL 32608 USA
| |
Collapse
|
12
|
Cushnie AK, El-Nahal HG, Bohlen MO, May PJ, Basso MA, Grimaldi P, Wang MZ, de Velasco Ezequiel MF, Sommer MA, Heilbronner SR. Using rAAV2-retro in rhesus macaques: Promise and caveats for circuit manipulation. J Neurosci Methods 2020; 345:108859. [PMID: 32668316 DOI: 10.1016/j.jneumeth.2020.108859] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/01/2020] [Accepted: 07/10/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Recent genetic technologies such as opto- and chemogenetics allow for the manipulation of brain circuits with unprecedented precision. Most studies employing these techniques have been undertaken in rodents, but a more human-homologous model for studying the brain is the nonhuman primate (NHP). Optimizing viral delivery of transgenes encoding actuator proteins could revolutionize the way we study neuronal circuits in NHPs. NEW METHOD: rAAV2-retro, a popular new capsid variant, produces robust retrograde labeling in rodents. Whether rAAV2-retro's highly efficient retrograde transport would translate to NHPs was unknown. Here, we characterized the anatomical distribution of labeling following injections of rAAV2-retro encoding opsins or DREADDs in the cortico-basal ganglia and oculomotor circuits of rhesus macaques. RESULTS rAAV2-retro injections in striatum, frontal eye field, and superior colliculus produced local labeling at injection sites and robust retrograde labeling in many afferent regions. In every case, however, a few brain regions with well-established projections to the injected structure lacked retrogradely labeled cells. We also observed robust terminal field labeling in downstream structures. COMPARISON WITH EXISTING METHOD(S) Patterns of labeling were similar to those obtained with traditional tract-tracers, except for some afferent labeling that was noticeably absent. CONCLUSIONS rAAV2-retro promises to be useful for circuit manipulation via retrograde transduction in NHPs, but caveats were revealed by our findings. Some afferently connected regions lacked retrogradely labeled cells, showed robust axon terminal labeling, or both. This highlights the importance of anatomically characterizing rAAV2-retro's expression in target circuits in NHPs before moving to manipulation studies.
Collapse
Affiliation(s)
- Adriana K Cushnie
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, United States
| | - Hala G El-Nahal
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, United States
| | - Martin O Bohlen
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, United States
| | - Paul J May
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, 39216, United States
| | - Michele A Basso
- Fuster Laboratory of Cognitive Neuroscience, Department of Psychiatry and Biobehavioral Sciences and Neurobiology, Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, Univ. of California Los Angeles, Los Angeles, CA 90095, United States
| | - Piercesare Grimaldi
- Fuster Laboratory of Cognitive Neuroscience, Department of Psychiatry and Biobehavioral Sciences and Neurobiology, Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, Univ. of California Los Angeles, Los Angeles, CA 90095, United States
| | - Maya Zhe Wang
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, United States
| | | | - Marc A Sommer
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, United States; Department of Neurobiology, Duke University School of Medicine, Durham, NC 27708, United States; Center for Cognitive Neuroscience, Duke University, Durham, NC 27708, United States
| | - Sarah R Heilbronner
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, United States.
| |
Collapse
|
13
|
Heiss JD, Lungu C, Hammoud DA, Herscovitch P, Ehrlich DJ, Argersinger DP, Sinharay S, Scott G, Wu T, Federoff HJ, Zaghloul KA, Hallett M, Lonser RR, Bankiewicz KS. Trial of magnetic resonance-guided putaminal gene therapy for advanced Parkinson's disease. Mov Disord 2019; 34:1073-1078. [PMID: 31145831 DOI: 10.1002/mds.27724] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/26/2019] [Accepted: 05/06/2019] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE To investigate the safety and tolerability of convection-enhanced delivery of an adeno-associated virus, serotype-2 vector carrying glial cell line-derived neurotrophic factor into the bilateral putamina of PD patients. METHODS Thirteen adult patients with advanced PD underwent adeno-associated virus, serotype-2 vector carrying glial cell line-derived neurotrophic factor and gadoteridol (surrogate MRI tracer) coinfusion (450 μL/hemisphere) at escalating doses: 9 × 1010 vg (n = 6); 3 × 1011 vg (n = 6); and 9 × 1011 vg (n = 1). Intraoperative MRI monitored infusion distribution. Patients underwent UPDRS assessment and [18 F]FDOPA-PET scanning preoperatively and 6 and 18 months postoperatively. RESULTS Adeno-associated virus, serotype-2 vector carrying glial cell line-derived neurotrophic factor was tolerated without clinical or radiographic toxicity. Average putaminal coverage was 26%. UPDRS scores remained stable. Ten of thirteen and 12 of 13 patients had increased [18 F]FDOPA Kis at 6 and 18 months postinfusion (increase range: 5-274% and 8-130%; median, 36% and 54%), respectively. Ki differences between baseline and 6- and 18-month follow-up were statistically significant (P < 0.0002). CONCLUSION Adeno-associated virus, serotype-2 vector carrying glial cell line-derived neurotrophic factor infusion was safe and well tolerated. Increased [18 F]FDOPA uptake suggests a neurotrophic effect on dopaminergic neurons. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- John D Heiss
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Codrin Lungu
- Division of Clinical Research, and Office of the Clinical Director, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Dima A Hammoud
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Peter Herscovitch
- Positron Emission Tomography Department, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Debra J Ehrlich
- Parkinson's Disease Clinic, Office of the Clinical Director, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Davis P Argersinger
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Sanhita Sinharay
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Gretchen Scott
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Tianxia Wu
- Clinical Trials Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Howard J Federoff
- Department of Neurology, University of California-Irvine, Irvine, California, USA
| | - Kareem A Zaghloul
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Mark Hallett
- Human Motor Control Section, Medical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Russell R Lonser
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Krystof S Bankiewicz
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA.,Department of Neurological Surgery, University of California-San Francisco, San Francisco, California, USA
| |
Collapse
|
14
|
Hudry E, Vandenberghe LH. Therapeutic AAV Gene Transfer to the Nervous System: A Clinical Reality. Neuron 2019; 101:839-862. [DOI: 10.1016/j.neuron.2019.02.017] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/07/2019] [Accepted: 02/11/2019] [Indexed: 02/07/2023]
|
15
|
Paul G, Sullivan AM. Trophic factors for Parkinson's disease: Where are we and where do we go from here? Eur J Neurosci 2019; 49:440-452. [DOI: 10.1111/ejn.14102] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 06/25/2018] [Accepted: 07/22/2018] [Indexed: 01/15/2023]
Affiliation(s)
- Gesine Paul
- Translational Neurology GroupDepartment of Clinical ScienceLund University Lund Sweden
- Wallenberg Center for Molecular MedicineLund University Lund Sweden
- Department of NeurologyScania University Hospital Lund Sweden
| | - Aideen M. Sullivan
- Department of Anatomy and NeuroscienceUniversity College Cork Cork Ireland
| |
Collapse
|
16
|
Abstract
Gene therapy has the potential to provide therapeutic benefit to millions of people with neurodegenerative diseases through several means, including direct correction of pathogenic mechanisms, neuroprotection, neurorestoration, and symptom control. Therapeutic efficacy is therefore dependent on knowledge of the disease pathogenesis and the required temporal and spatial specificity of gene expression. An additional critical challenge is achieving the most complete transduction of the target structure while avoiding leakage into neighboring regions or perivascular spaces. The gene therapy field has recently entered a new technological era, in which interventional MRI-guided convection-enhanced delivery (iMRI-CED) is the gold standard for verifying accurate vector delivery in real time. The availability of this advanced neurosurgical technique may accelerate the translation of the promising preclinical therapeutics under development for neurodegenerative disorders, including Parkinson's, Huntington's, and Alzheimer's diseases.
Collapse
Affiliation(s)
- Vivek Sudhakar
- Brain Modulation Laboratory, Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 15213, USA
| | - R Mark Richardson
- Brain Modulation Laboratory, Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 15213, USA.
| |
Collapse
|
17
|
Raghunathan R, Polinski NK, Klein JA, Hogan JD, Shao C, Khatri K, Leon D, McComb ME, Manfredsson FP, Sortwell CE, Zaia J. Glycomic and Proteomic Changes in Aging Brain Nigrostriatal Pathway. Mol Cell Proteomics 2018; 17:1778-1787. [PMID: 29915149 PMCID: PMC6126385 DOI: 10.1074/mcp.ra118.000680] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 06/12/2018] [Indexed: 12/19/2022] Open
Abstract
Parkinson's disease (PD) is a neurological disorder characterized by the progressive loss of functional dopaminergic neurons in the nigrostriatal pathway in the brain. Although current treatments provide only symptomatic relief, gene therapy has the potential to slow or halt the degeneration of nigrostriatal dopamine neurons in PD patients. Adeno-associated viruses (AAV) are vectors of choice in gene therapy because of their well-characterized safety and efficacy profiles; however, although gene therapy has been successful in preclinical models of the disease, clinical trials in humans have failed to demonstrate efficacy. Significantly, all primary AAV receptors of the virus are glycans. We thus hypothesize that age related changes in glycan receptors of heparan sulfate (HS) proteoglycans (receptor for rAAV2), and/or N-glycans with terminal galactose (receptor for rAAV9) results in poor adeno-associated virus binding in either the striatum or substantia nigra, or both, affecting transduction and gene delivery. To test our hypothesis we analyzed the striatum and substantia nigra for changes in HS, N-glycans and proteomic signatures in young versus aged rat brain striatum and substantia nigra. We observed different brain region-specific HS disaccharide profiles in aged compared with young adult rats for brain region-specific profiles in striatum versus substantia nigra. We observed brain region- and age-specific N-glycan compositional profiles with respect to the terminal galactose units that serve as receptors for AAV9. We also observed brain region-specific changes in protein expression in the aging nigrostriatal pathway. These studies provide insight into age- and brain region-specific changes in glycan receptors and proteome that will inform design of improved viral vectors for Parkinson Disease (PD) gene therapy.
Collapse
Affiliation(s)
- Rekha Raghunathan
- From the ‡Department of Molecular and Translational Medicine, Center for Biomedical Mass Spectrometry, Boston University Medical Campus, Boston, Massachusetts
| | - Nicole K Polinski
- ‖Department of Translational Science and Molecular Medicine, Michigan State University
| | - Joshua A Klein
- ¶Bioinformatics Program, Boston University, Boston, Massachusetts
| | - John D Hogan
- ¶Bioinformatics Program, Boston University, Boston, Massachusetts
| | - Chun Shao
- §Department of Biochemistry, Center for Biomedical Mass Spectrometry, Boston University Medical Campus, Boston, Massachusetts
| | - Kshitij Khatri
- §Department of Biochemistry, Center for Biomedical Mass Spectrometry, Boston University Medical Campus, Boston, Massachusetts
| | - Deborah Leon
- §Department of Biochemistry, Center for Biomedical Mass Spectrometry, Boston University Medical Campus, Boston, Massachusetts
| | - Mark E McComb
- §Department of Biochemistry, Center for Biomedical Mass Spectrometry, Boston University Medical Campus, Boston, Massachusetts
| | - Fredric P Manfredsson
- ‖Department of Translational Science and Molecular Medicine, Michigan State University
| | - Caryl E Sortwell
- ‖Department of Translational Science and Molecular Medicine, Michigan State University
| | - Joseph Zaia
- From the ‡Department of Molecular and Translational Medicine, Center for Biomedical Mass Spectrometry, Boston University Medical Campus, Boston, Massachusetts;
- §Department of Biochemistry, Center for Biomedical Mass Spectrometry, Boston University Medical Campus, Boston, Massachusetts
- ¶Bioinformatics Program, Boston University, Boston, Massachusetts
| |
Collapse
|
18
|
Penttinen AM, Parkkinen I, Voutilainen MH, Koskela M, Bäck S, Their A, Richie CT, Domanskyi A, Harvey BK, Tuominen RK, Nevalaita L, Saarma M, Airavaara M. Pre-α-pro-GDNF and Pre-β-pro-GDNF Isoforms Are Neuroprotective in the 6-hydroxydopamine Rat Model of Parkinson's Disease. Front Neurol 2018; 9:457. [PMID: 29973907 PMCID: PMC6019446 DOI: 10.3389/fneur.2018.00457] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 05/29/2018] [Indexed: 11/13/2022] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is one of the most studied neurotrophic factors. GDNF has two splice isoforms, full-length pre-α-pro-GDNF (α-GDNF) and pre-β-pro-GDNF (β-GDNF), which has a 26 amino acid deletion in the pro-region. Thus far, studies have focused solely on the α-GDNF isoform, and nothing is known about the in vivo effects of the shorter β-GDNF variant. Here we compare for the first time the effects of overexpressed α-GDNF and β-GDNF in non-lesioned rat striatum and the partial 6-hydroxydopamine lesion model of Parkinson's disease. GDNF isoforms were overexpressed with their native pre-pro-sequences in the striatum using an adeno-associated virus (AAV) vector, and the effects on motor performance and dopaminergic phenotype of the nigrostriatal pathway were assessed. In the non-lesioned striatum, both isoforms increased the density of dopamine transporter-positive fibers at 3 weeks after viral vector delivery. Although both isoforms increased the activity of the animals in cylinder assay, only α-GDNF enhanced the use of contralateral paw. Four weeks later, the striatal tyrosine hydroxylase (TH)-immunoreactivity was decreased in both α-GDNF and β-GDNF treated animals. In the neuroprotection assay, both GDNF splice isoforms increased the number of TH-immunoreactive cells in the substantia nigra but did not promote behavioral recovery based on amphetamine-induced rotation or cylinder assays. Thus, the shorter GDNF isoform, β-GDNF, and the full-length α-isoform have comparable neuroprotective efficacy on dopamine neurons of the nigrostriatal circuitry.
Collapse
Affiliation(s)
- Anna-Maija Penttinen
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Ilmari Parkkinen
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Merja H Voutilainen
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Maryna Koskela
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Susanne Bäck
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Anna Their
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Christopher T Richie
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, United States
| | - Andrii Domanskyi
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Brandon K Harvey
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, United States
| | - Raimo K Tuominen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Liina Nevalaita
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Mikko Airavaara
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
19
|
Forsayeth J, Hadaczek P. Ganglioside Metabolism and Parkinson's Disease. Front Neurosci 2018; 12:45. [PMID: 29459819 PMCID: PMC5807391 DOI: 10.3389/fnins.2018.00045] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 01/18/2018] [Indexed: 12/21/2022] Open
Abstract
Here we advance the hypothesis that Parkinson's disease (PD) is fundamentally a failure of trophic support for specific classes of neurons, primarily catecholaminergic. Evidence from our laboratory provides a framework into which a broad array of findings from many quarters can be integrated into a general theory that offers testable hypotheses to new and established investigators. Mice deficient in the ability to synthesize series-a gangliosides, specifically GM1 ganglioside, develop parkinsonism. We found that this seems to be due to a failure in signaling efficiency by the important catecholaminergic growth factor, GDNF. Interestingly, these mice accumulate alpha-synuclein in nigral neurons. Striatal over-expression of GDNF eliminates these aggregates and also restores normal motor function. These findings bring into question common beliefs about alpha-synuclein pathology and may help us to reinterpret other experimental findings in a new light. The purpose of this article is to provoke new thinking about PD and hopefully encourage younger scientists to explore some of the ideas presented below.
Collapse
Affiliation(s)
- John Forsayeth
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Piotr Hadaczek
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
20
|
Axelsen TM, Woldbye DP. Gene Therapy for Parkinson's Disease, An Update. JOURNAL OF PARKINSON'S DISEASE 2018; 8:195-215. [PMID: 29710735 PMCID: PMC6027861 DOI: 10.3233/jpd-181331] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 03/25/2018] [Indexed: 12/19/2022]
Abstract
The current mainstay treatment of Parkinson's disease (PD) consists of dopamine replacement therapy which, in addition to causing several side effects, does not delay disease progression. The field of gene therapy offers a potential means to improve current therapy. The present review gives an update of the present status of gene therapy for PD. Both non-disease and disease modifying transgenes have been tested for PD gene therapy in animal and human studies. Non-disease modifying treatments targeting dopamine or GABA synthesis have been successful and promising at improving PD symptomatology in randomized clinical studies, but substantial testing remains before these can be implemented in the standard clinical treatment repertoire. As for disease modifying targets that theoretically offer the possibility of slowing the progression of disease, several neurotrophic factors show encouraging results in preclinical models (e.g., neurturin, GDNF, BDNF, CDNF, VEGF-A). However, so far, clinical trials have only tested neurturin, and, unfortunately, no trial has been able to meet its primary endpoint. Future clinical trials with neurotrophic factors clearly deserve to be conducted, considering the still enticing goal of actually slowing the disease process of PD. As alternative types of gene therapy, opto- and chemogenetics might also find future use in PD treatment and novel genome-editing technology could also potentially be applied as individualized gene therapy for genetic types of PD.
Collapse
Affiliation(s)
- Tobias M. Axelsen
- Department of Neurology, Herlev University Hospital, Herlev, Denmark
| | - David P.D. Woldbye
- Department of Neuroscience, Panum Institute, Mærsk Tower, University of Copenhagen, Copenhagen N, Denmark
| |
Collapse
|
21
|
Fan CH, Lin CY, Liu HL, Yeh CK. Ultrasound targeted CNS gene delivery for Parkinson's disease treatment. J Control Release 2017; 261:246-262. [DOI: 10.1016/j.jconrel.2017.07.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 07/05/2017] [Accepted: 07/05/2017] [Indexed: 10/19/2022]
|
22
|
Lin JY, Xie CL, Zhang SF, Yuan W, Liu ZG. Current Experimental Studies of Gene Therapy in Parkinson's Disease. Front Aging Neurosci 2017; 9:126. [PMID: 28515689 PMCID: PMC5413509 DOI: 10.3389/fnagi.2017.00126] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 04/13/2017] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease (PD) was characterized by late-onset, progressive dopamine neuron loss and movement disorders. The progresses of PD affected the neural function and integrity. To date, most researches had largely addressed the dopamine replacement therapies, but the appearance of L-dopa-induced dyskinesia hampered the use of the drug. And the mechanism of PD is so complicated that it's hard to solve the problem by just add drugs. Researchers began to focus on the genetic underpinnings of Parkinson's disease, searching for new method that may affect the neurodegeneration processes in it. In this paper, we reviewed current delivery methods used in gene therapies for PD, we also summarized the primary target of the gene therapy in the treatment of PD, such like neurotrophic factor (for regeneration), the synthesis of neurotransmitter (for prolong the duration of L-dopa), and the potential proteins that might be a target to modulate via gene therapy. Finally, we discussed RNA interference therapies used in Parkinson's disease, it might act as a new class of drug. We mainly focus on the efficiency and tooling features of different gene therapies in the treatment of PD.
Collapse
Affiliation(s)
- Jing-Ya Lin
- Department of Neurology, Xinhua Hospital Affiliated to the Medical School of Shanghai JiaoTong UniversityShanghai, China
| | - Cheng-Long Xie
- Department of Neurology, The first Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical UniversityWenzhou, China
| | - Su-Fang Zhang
- Department of Neurology, Xinhua Hospital Affiliated to the Medical School of Shanghai JiaoTong UniversityShanghai, China
| | - Weien Yuan
- School of Pharmacy, Shanghai JiaoTong UniversityShanghai, China
| | - Zhen-Guo Liu
- Department of Neurology, Xinhua Hospital Affiliated to the Medical School of Shanghai JiaoTong UniversityShanghai, China
| |
Collapse
|
23
|
Tenenbaum L, Humbert-Claude M. Glial Cell Line-Derived Neurotrophic Factor Gene Delivery in Parkinson's Disease: A Delicate Balance between Neuroprotection, Trophic Effects, and Unwanted Compensatory Mechanisms. Front Neuroanat 2017; 11:29. [PMID: 28442998 PMCID: PMC5385337 DOI: 10.3389/fnana.2017.00029] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 03/23/2017] [Indexed: 12/14/2022] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) and Neurturin (NRTN) bind to a receptor complex consisting of a member of the GDNF family receptor (GFR)-α and the Ret tyrosine kinase. Both factors were shown to protect nigro-striatal dopaminergic neurons and reduce motor symptoms when applied terminally in toxin-induced Parkinson's disease (PD) models. However, clinical trials based on intraputaminal GDNF protein administration or recombinant adeno-associated virus (rAAV)-mediated NRTN gene delivery have been disappointing. In this review, several factors that could have limited the clinical benefits are discussed. Retrograde transport of GDNF/NRTN to the dopaminergic neurons soma is thought to be necessary for NRTN/GFR-α/Ret signaling mediating the pro-survival effect. Therefore, the feasibility of treating advanced patients with neurotrophic factors is questioned by recent data showing that: (i) tyrosine hydroxylase-positive putaminal innervation has almost completely disappeared at 5 years post-diagnosis and (ii) in patients enrolled in the rAAV-NRTN trial more than 5 years post-diagnosis, NRTN was almost not transported to the substantia nigra pars compacta. In addition to its anti-apoptotic and neurotrophic properties, GDNF also interferes with dopamine homeostasis via time and dose-dependent effects such as: stimulation of dopamine neuron excitability, inhibition of dopamine transporter activity, tyrosine hydroxylase phosphorylation, and inhibition of tyrosine hydroxylase transcription. Depending on the delivery parameters, the net result of this intricate network of regulations could be either beneficial or deleterious. In conclusion, further unraveling of the mechanism of action of GDNF gene delivery in relevant animal models is still needed to optimize the clinical benefits of this new therapeutic approach. Recent developments in the design of regulated viral vectors will allow to finely adjust the GDNF dose and period of administration. Finally, new clinical studies in less advanced patients are warranted to evaluate the potential of AAV-mediated neurotrophic factors gene delivery in PD. These will be facilitated by the demonstration of the safety of rAAV administration into the human brain.
Collapse
Affiliation(s)
- Liliane Tenenbaum
- Laboratory of Cellular and Molecular Neurotherapies, Clinical Neuroscience Department, Center for Neuroscience Research, Lausanne University HospitalLausanne, Switzerland
| | - Marie Humbert-Claude
- Laboratory of Cellular and Molecular Neurotherapies, Clinical Neuroscience Department, Center for Neuroscience Research, Lausanne University HospitalLausanne, Switzerland
| |
Collapse
|
24
|
Non-human primate models of PD to test novel therapies. J Neural Transm (Vienna) 2017; 125:291-324. [PMID: 28391443 DOI: 10.1007/s00702-017-1722-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 04/04/2017] [Indexed: 12/13/2022]
Abstract
Non-human primate (NHP) models of Parkinson disease show many similarities with the human disease. They are very useful to test novel pharmacotherapies as reviewed here. The various NHP models of this disease are described with their characteristics including the macaque, the marmoset, and the squirrel monkey models. Lesion-induced and genetic models are described. There is no drug to slow, delay, stop, or cure Parkinson disease; available treatments are symptomatic. The dopamine precursor, L-3,4-dihydroxyphenylalanine (L-Dopa) still remains the gold standard symptomatic treatment of Parkinson. However, involuntary movements termed L-Dopa-induced dyskinesias appear in most patients after chronic treatment and may become disabling. Dyskinesias are very difficult to manage and there is only amantadine approved providing only a modest benefit. In this respect, NHP models have been useful to seek new drug targets, since they reproduce motor complications observed in parkinsonian patients. Therapies to treat motor symptoms in NHP models are reviewed with a discussion of their translational value to humans. Disease-modifying treatments tested in NHP are reviewed as well as surgical treatments. Many biochemical changes in the brain of post-mortem Parkinson disease patients with dyskinesias are reviewed and compare well with those observed in NHP models. Non-motor symptoms can be categorized into psychiatric, autonomic, and sensory symptoms. These symptoms are present in most parkinsonian patients and are already installed many years before the pre-motor phase of the disease. The translational usefulness of NHP models of Parkinson is discussed for non-motor symptoms.
Collapse
|
25
|
Blesa J, Trigo-Damas I, del Rey NLG, Obeso JA. The use of nonhuman primate models to understand processes in Parkinson’s disease. J Neural Transm (Vienna) 2017; 125:325-335. [DOI: 10.1007/s00702-017-1715-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Accepted: 03/16/2017] [Indexed: 02/07/2023]
|
26
|
Albert K, Voutilainen MH, Domanskyi A, Airavaara M. AAV Vector-Mediated Gene Delivery to Substantia Nigra Dopamine Neurons: Implications for Gene Therapy and Disease Models. Genes (Basel) 2017; 8:genes8020063. [PMID: 28208742 PMCID: PMC5333052 DOI: 10.3390/genes8020063] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 02/03/2017] [Indexed: 12/21/2022] Open
Abstract
Gene delivery using adeno-associated virus (AAV) vectors is a widely used method to transduce neurons in the brain, especially due to its safety, efficacy, and long-lasting expression. In addition, by varying AAV serotype, promotor, and titer, it is possible to affect the cell specificity of expression or the expression levels of the protein of interest. Dopamine neurons in the substantia nigra projecting to the striatum, comprising the nigrostriatal pathway, are involved in movement control and degenerate in Parkinson’s disease. AAV-based gene targeting to the projection area of these neurons in the striatum has been studied extensively to induce the production of neurotrophic factors for disease-modifying therapies for Parkinson’s disease. Much less emphasis has been put on AAV-based gene therapy targeting dopamine neurons in substantia nigra. We will review the literature related to targeting striatum and/or substantia nigra dopamine neurons using AAVs in order to express neuroprotective and neurorestorative molecules, as well as produce animal disease models of Parkinson’s disease. We discuss difficulties in targeting substantia nigra dopamine neurons and their vulnerability to stress in general. Therefore, choosing a proper control for experimental work is not trivial. Since the axons along the nigrostriatal tract are the first to degenerate in Parkinson’s disease, the location to deliver the therapy must be carefully considered. We also review studies using AAV-α-synuclein (α-syn) to target substantia nigra dopamine neurons to produce an α-syn overexpression disease model in rats. Though these studies are able to produce mild dopamine system degeneration in the striatum and substantia nigra and some behavioural effects, there are studies pointing to the toxicity of AAV-carrying green fluorescent protein (GFP), which is often used as a control. Therefore, we discuss the potential difficulties in overexpressing proteins in general in the substantia nigra.
Collapse
Affiliation(s)
- Katrina Albert
- Institute of Biotechnology, University of Helsinki, Helsinki 00014, Finland.
| | - Merja H Voutilainen
- Institute of Biotechnology, University of Helsinki, Helsinki 00014, Finland.
| | - Andrii Domanskyi
- Institute of Biotechnology, University of Helsinki, Helsinki 00014, Finland.
| | - Mikko Airavaara
- Institute of Biotechnology, University of Helsinki, Helsinki 00014, Finland.
| |
Collapse
|
27
|
Pignataro D, Sucunza D, Rico AJ, Dopeso-Reyes IG, Roda E, Rodríguez-Perez AI, Labandeira-Garcia JL, Broccoli V, Kato S, Kobayashi K, Lanciego JL. Gene therapy approaches in the non-human primate model of Parkinson's disease. J Neural Transm (Vienna) 2017; 125:575-589. [PMID: 28130586 DOI: 10.1007/s00702-017-1681-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 01/16/2017] [Indexed: 12/23/2022]
Abstract
The field of gene therapy has recently witnessed a number of major conceptual changes. Besides the traditional thinking that comprises the use of viral vectors for the delivery of a given therapeutic gene, a number of original approaches have been recently envisaged, focused on using vectors carrying genes to further modify basal ganglia circuits of interest. It is expected that these approaches will ultimately induce a therapeutic potential being sustained by gene-induced changes in brain circuits. Among others, at present, it is technically feasible to use viral vectors to (1) achieve a controlled release of neurotrophic factors, (2) conduct either a transient or permanent silencing of any given basal ganglia circuit of interest, (3) perform an in vivo cellular reprogramming by promoting the conversion of resident cells into dopaminergic-like neurons, and (4) improving levodopa efficacy over time by targeting aromatic L-amino acid decarboxylase. Furthermore, extensive research efforts based on viral vectors are currently ongoing in an attempt to better replicate the dopaminergic neurodegeneration phenomena inherent to the progressive intraneuronal aggregation of alpha-synuclein. Finally, a number of incoming strategies will soon emerge over the horizon, these being sustained by the underlying goal of promoting alpha-synuclein clearance, such as, for instance, gene therapy initiatives based on increasing the activity of glucocerebrosidase. To provide adequate proof-of-concept on safety and efficacy and to push forward true translational initiatives based on these different types of gene therapies before entering into clinical trials, the use of non-human primate models undoubtedly plays an instrumental role.
Collapse
Affiliation(s)
- D Pignataro
- Department of Neurosciences, Center for Applied Medical Research (CIMA), University of Navarra, Pio XII Ave 55, Edificio CIMA, 31008, Pamplona, Navarra, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - D Sucunza
- Department of Neurosciences, Center for Applied Medical Research (CIMA), University of Navarra, Pio XII Ave 55, Edificio CIMA, 31008, Pamplona, Navarra, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - A J Rico
- Department of Neurosciences, Center for Applied Medical Research (CIMA), University of Navarra, Pio XII Ave 55, Edificio CIMA, 31008, Pamplona, Navarra, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - I G Dopeso-Reyes
- Department of Neurosciences, Center for Applied Medical Research (CIMA), University of Navarra, Pio XII Ave 55, Edificio CIMA, 31008, Pamplona, Navarra, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - E Roda
- Department of Neurosciences, Center for Applied Medical Research (CIMA), University of Navarra, Pio XII Ave 55, Edificio CIMA, 31008, Pamplona, Navarra, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - A I Rodríguez-Perez
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - J L Labandeira-Garcia
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - V Broccoli
- Division of Neuroscience, Ospedale San Raffaele, 20132, Milan, Italy
- CNR Institute of Neuroscience, 20129, Milan, Italy
| | - S Kato
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - K Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - José L Lanciego
- Department of Neurosciences, Center for Applied Medical Research (CIMA), University of Navarra, Pio XII Ave 55, Edificio CIMA, 31008, Pamplona, Navarra, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.
| |
Collapse
|
28
|
Stayte S, Rentsch P, Tröscher AR, Bamberger M, Li KM, Vissel B. Activin A Inhibits MPTP and LPS-Induced Increases in Inflammatory Cell Populations and Loss of Dopamine Neurons in the Mouse Midbrain In Vivo. PLoS One 2017; 12:e0167211. [PMID: 28121982 PMCID: PMC5266209 DOI: 10.1371/journal.pone.0167211] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 11/10/2016] [Indexed: 01/11/2023] Open
Abstract
Parkinson’s disease is a chronic neurodegenerative disease characterized by a significant loss of dopaminergic neurons within the substantia nigra pars compacta region and a subsequent loss of dopamine within the striatum. A promising avenue of research has been the administration of growth factors to promote the survival of remaining midbrain neurons, although the mechanism by which they provide neuroprotection is not understood. Activin A, a member of the transforming growth factor β superfamily, has been shown to be a potent anti-inflammatory following acute brain injury and has been demonstrated to play a role in the neuroprotection of midbrain neurons against MPP+-induced degeneration in vitro. We hypothesized that activin A may offer similar anti-inflammatory and neuroprotective effects in in vivo mouse models of Parkinson’s disease. We found that activin A significantly attenuated the inflammatory response induced by both MPTP and intranigral administration of lipopolysaccharide in C57BL/6 mice. We found that administration of activin A promoted survival of dopaminergic and total neuron populations in the pars compacta region both 8 days and 8 weeks after MPTP-induced degeneration. Surprisingly, no corresponding protection of striatal dopamine levels was found. Furthermore, activin A failed to protect against loss of striatal dopamine transporter expression in the striatum, suggesting the neuroprotective action of activin A may be localized to the substantia nigra. Together, these results provide the first evidence that activin A exerts potent neuroprotection and anti-inflammatory effects in the MPTP and lipopolysaccharide mouse models of Parkinson’s disease.
Collapse
Affiliation(s)
- Sandy Stayte
- Neuroscience Department, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Peggy Rentsch
- Neuroscience Department, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | | | | | - Kong M. Li
- Pharmacology Department, Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Bryce Vissel
- Neuroscience Department, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- School of Life Sciences, University of Technology Sydney, Sydney, Australia
- * E-mail:
| |
Collapse
|
29
|
Blits B, Petry H. Perspective on the Road toward Gene Therapy for Parkinson's Disease. Front Neuroanat 2017; 10:128. [PMID: 28119578 PMCID: PMC5220060 DOI: 10.3389/fnana.2016.00128] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 12/20/2016] [Indexed: 11/19/2022] Open
Abstract
Many therapeutic strategies aimed at relieving symptoms of Parkinson’s disease (PD) are currently used for treatment of this disease. With a hallmark of progressive degeneration of dopaminergic neurons, the absence of properly operational dopaminergic circuitry becomes a therapeutic target. Following diagnosis, dopamine replacement can be given in the form of L-DOPA (L-3,4-dihydroxyphenylalanine). Even though it is recognized as standard of care, this treatment strategy does not prevent the affected neurons from degenerating. Therefore, studies have been performed using gene therapy (GT) to make dopamine (DA) available from within the brain using an artificial DA circuitry. One approach is to administer a GT aimed at delivering the key enzymes for DA synthesis using a lentiviral vector system (Palfi et al., 2014). A similar approach has been investigated with adeno-associated virus (AAV) expressing aromatic L-amino acid decarboxylase, tyrosine hydroxylase, and GTP-cyclohydrolase I (Bankiewicz et al., 2000), which are downregulated in PD. Another GT approach to mitigate symptoms of PD used AAV-mediated delivery of GAD-67 (glutamate decarboxylase) (Kaplitt et al., 2007). This approach mimics the inhibitory effect of DA neurons on their targets, in reducing motor abnormalities. Finally, disease modifying strategies have been undertaken using neurotrophic factors such as neurturin (NTN) (Marks et al., 2008; Bartus et al., 2013a) or are ongoing with the closely related Glial cell line-derived neurotrophic factor. Those approaches are aiming at rescuing the degenerating neurons. All of the above mentioned strategies have their own merits, but also some disadvantages. So far, none of clinical applied GT studies has resulted in significant clinical benefit, although some clinical studies are ongoing and results are expected over the next few years.
Collapse
Affiliation(s)
- Bas Blits
- Neurobiology Research, uniQure BV Amsterdam, Netherlands
| | - Harald Petry
- Neurobiology Research, uniQure BV Amsterdam, Netherlands
| |
Collapse
|
30
|
Kirik D, Cederfjäll E, Halliday G, Petersén Å. Gene therapy for Parkinson's disease: Disease modification by GDNF family of ligands. Neurobiol Dis 2017; 97:179-188. [DOI: 10.1016/j.nbd.2016.09.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 08/24/2016] [Accepted: 09/06/2016] [Indexed: 10/21/2022] Open
|
31
|
Hocquemiller M, Giersch L, Audrain M, Parker S, Cartier N. Adeno-Associated Virus-Based Gene Therapy for CNS Diseases. Hum Gene Ther 2016; 27:478-96. [PMID: 27267688 PMCID: PMC4960479 DOI: 10.1089/hum.2016.087] [Citation(s) in RCA: 201] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 06/07/2016] [Indexed: 12/11/2022] Open
Abstract
Gene therapy is at the cusp of a revolution for treating a large spectrum of CNS disorders by providing a durable therapeutic protein via a single administration. Adeno-associated virus (AAV)-mediated gene transfer is of particular interest as a therapeutic tool because of its safety profile and efficiency in transducing a wide range of cell types. The purpose of this review is to describe the most notable advancements in preclinical and clinical research on AAV-based CNS gene therapy and to discuss prospects for future development based on a new generation of vectors and delivery.
Collapse
Affiliation(s)
| | | | - Mickael Audrain
- Université Paris Descartes, Paris, France
- INSERM UMR1169, Université Paris-Sud,Université Paris-Saclay, Orsay, France
- CEA, DSV, IBM, MIRCen, Fontenay-aux-Roses, France
| | | | - Nathalie Cartier
- INSERM UMR1169, Université Paris-Sud,Université Paris-Saclay, Orsay, France
- CEA, DSV, IBM, MIRCen, Fontenay-aux-Roses, France
| |
Collapse
|
32
|
Shen Y, Huang J, Liu L, Xu X, Han C, Zhang G, Jiang H, Li J, Lin Z, Xiong N, Wang T. A Compendium of Preparation and Application of Stem Cells in Parkinson's Disease: Current Status and Future Prospects. Front Aging Neurosci 2016; 8:117. [PMID: 27303288 PMCID: PMC4885841 DOI: 10.3389/fnagi.2016.00117] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 05/09/2016] [Indexed: 12/22/2022] Open
Abstract
Parkinson's Disease (PD) is a progressively neurodegenerative disorder, implicitly characterized by a stepwise loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc) and explicitly marked by bradykinesia, rigidity, resting tremor and postural instability. Currently, therapeutic approaches available are mainly palliative strategies, including L-3,4-dihydroxy-phenylalanine (L-DOPA) replacement therapy, DA receptor agonist and deep brain stimulation (DBS) procedures. As the disease proceeds, however, the pharmacotherapeutic efficacy is inevitably worn off, worse still, implicated by side effects of motor response oscillations as well as L-DOPA induced dyskinesia (LID). Therefore, the frustrating status above has propeled the shift to cell replacement therapy (CRT), a promising restorative therapy intending to secure a long-lasting relief of patients' symptoms. By far, stem cell lines of multifarious origins have been established, which can be further categorized into embryonic stem cells (ESCs), neural stem cells (NSCs), induced neural stem cells (iNSCs), mesenchymal stem cells (MSCs), and induced pluripotent stem cells (iPSCs). In this review, we intend to present a compendium of preparation and application of multifarious stem cells, especially in relation to PD research and therapy. In addition, the current status, potential challenges and future prospects for practical CRT in PD patients will be elaborated as well.
Collapse
Affiliation(s)
- Yan Shen
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Jinsha Huang
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Ling Liu
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Xiaoyun Xu
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Chao Han
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Guoxin Zhang
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Haiyang Jiang
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Jie Li
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Zhicheng Lin
- Department of Psychiatry, Harvard Medical School, Division of Alcohol and Drug Abuse, and Mailman Neuroscience Research Center, McLean Hospital Belmont, MA, USA
| | - Nian Xiong
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Tao Wang
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| |
Collapse
|
33
|
Bankiewicz KS, Sudhakar V, Samaranch L, San Sebastian W, Bringas J, Forsayeth J. AAV viral vector delivery to the brain by shape-conforming MR-guided infusions. J Control Release 2016; 240:434-442. [PMID: 26924352 DOI: 10.1016/j.jconrel.2016.02.034] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 02/17/2016] [Accepted: 02/22/2016] [Indexed: 11/18/2022]
Abstract
Gene transfer technology offers great promise as a potential therapeutic approach to the brain but has to be viewed as a very complex technology. Success of ongoing clinical gene therapy trials depends on many factors such as selection of the correct genetic and anatomical target in the brain. In addition, selection of the viral vector capable of transfer of therapeutic gene into target cells, along with long-term expression that avoids immunotoxicity has to be established. As with any drug development strategy, delivery of gene therapy has to be consistent and predictable in each study subject. Failed drug and vector delivery will lead to failed clinical trials. In this article, we describe our experience with AAV viral vector delivery system, that allows us to optimize and monitor in real time viral vector administration into affected regions of the brain. In addition to discussing MRI-guided technology for administration of AAV vectors we have developed and now employ in current clinical trials, we also describe ways in which infusion cannula design and stereotactic trajectory may be used to maximize the anatomical coverage by using fluid backflow. This innovative approach enables more precise coverage by fitting the shape of the infusion to the shape of the anatomical target.
Collapse
Affiliation(s)
- Krystof S Bankiewicz
- Interventional Neuro Center, Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94110, USA.
| | - Vivek Sudhakar
- Interventional Neuro Center, Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94110, USA
| | - Lluis Samaranch
- Interventional Neuro Center, Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94110, USA
| | - Waldy San Sebastian
- Interventional Neuro Center, Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94110, USA
| | - John Bringas
- Interventional Neuro Center, Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94110, USA
| | - John Forsayeth
- Interventional Neuro Center, Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94110, USA
| |
Collapse
|
34
|
GDNF and AADC Gene Therapy for Parkinson’s Disease. Transl Neurosci 2016. [DOI: 10.1007/978-1-4899-7654-3_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
35
|
Aron Badin R, Vadori M, Cozzi E, Hantraye P. Translational research for Parkinson׳s disease: The value of pre-clinical primate models. Eur J Pharmacol 2015; 759:118-26. [DOI: 10.1016/j.ejphar.2015.03.038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 03/10/2015] [Accepted: 03/12/2015] [Indexed: 12/15/2022]
|
36
|
Barak S, Wang J, Ahmadiantehrani S, Ben Hamida S, Kells AP, Forsayeth J, Bankiewicz KS, Ron D. Glial cell line-derived neurotrophic factor (GDNF) is an endogenous protector in the mesolimbic system against excessive alcohol consumption and relapse. Addict Biol 2015; 20:629-42. [PMID: 24801661 DOI: 10.1111/adb.12152] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Moderate social consumption of alcohol is common; however, only a small percentage of individuals transit from social to excessive, uncontrolled alcohol drinking. This suggests the existence of protective mechanisms that prevent the development of alcohol addiction. Here, we tested the hypothesis that the glial cell line-derived neurotrophic factor (GDNF) in the mesolimbic system [e.g. the nucleus accumbens (Acb) and ventral tegmental area (VTA)] is part of such a mechanism. We found that GDNF knockdown, by infecting rat Acb neurons with a small hairpin RNA (shRNA) targeting the GDNF gene, produced a rapid escalation to excessive alcohol consumption and enhanced relapse to alcohol drinking. Conversely, viral-mediated overexpression of the growth factor in the mesolimbic system blocked the escalation from moderate to excessive alcohol drinking. To access the mechanism underlying GDNF's actions, we measured the firing rate of dopaminergic (DAergic) neurons in the VTA after a history of excessive alcohol intake with or without elevating GDNF levels. We found that the spontaneous firing rate of DAergic neurons in the VTA was reduced during alcohol withdrawal and that GDNF reversed this alcohol-induced DA deficiency. Together, our results suggest that endogenous GDNF in the mesolimbic system controls the transition from moderate to excessive alcohol drinking and relapse via reversal of alcohol-dependent neuro-adaptations in DAergic VTA neurons.
Collapse
Affiliation(s)
- Segev Barak
- The Gallo Research Center; University of California; San Francisco CA USA
- Department of Neurology; University of California; San Francisco CA USA
| | - Jun Wang
- The Gallo Research Center; University of California; San Francisco CA USA
- Department of Neurology; University of California; San Francisco CA USA
| | - Somayeh Ahmadiantehrani
- The Gallo Research Center; University of California; San Francisco CA USA
- Department of Neurology; University of California; San Francisco CA USA
| | - Sami Ben Hamida
- The Gallo Research Center; University of California; San Francisco CA USA
- Department of Neurology; University of California; San Francisco CA USA
| | - Adrian P. Kells
- Department of Neurological Surgery; University of California; San Francisco CA USA
| | - John Forsayeth
- Department of Neurological Surgery; University of California; San Francisco CA USA
| | | | - Dorit Ron
- The Gallo Research Center; University of California; San Francisco CA USA
- Department of Neurology; University of California; San Francisco CA USA
| |
Collapse
|
37
|
Viral vector delivery of neurotrophic factors for Parkinson's disease therapy. Expert Rev Mol Med 2015; 17:e8. [DOI: 10.1017/erm.2015.6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterised by the progressive loss of midbrain dopaminergic neurons, which causes motor impairments. Current treatments involve dopamine replacement to address the disease symptoms rather than its cause. Factors that promote the survival of dopaminergic neurons have been proposed as novel therapies for PD. Several dopaminergic neurotrophic factors (NTFs) have been examined for their ability to protect and/or restore degenerating dopaminergic neurons, both in animal models and in clinical trials. These include glial cell line-derived neurotrophic factor, neurturin, cerebral dopamine neurotrophic factor and growth/differentiation factor 5. Delivery of these NTFs via injection or infusion to the brain raises several practical problems. A new delivery approach for NTFs involves the use of recombinant viral vectors to enable long-term expression of these factors in brain cells. Vectors used include those based on adenoviruses, adeno-associated viruses and lentiviruses. Here we review progress to date on the potential of each of these four NTFs as novel therapeutic strategies for PD, as well as the challenges that have arisen, from pre-clinical analysis to clinical trials. We conclude by discussing recently-developed approaches to optimise the delivery of NTF-carrying viral vectors to the brain.
Collapse
|
38
|
Stayte S, Rentsch P, Li KM, Vissel B. Activin A protects midbrain neurons in the 6-hydroxydopamine mouse model of Parkinson's disease. PLoS One 2015; 10:e0124325. [PMID: 25902062 PMCID: PMC4406584 DOI: 10.1371/journal.pone.0124325] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Accepted: 03/02/2015] [Indexed: 01/11/2023] Open
Abstract
Parkinson’s disease (PD) is a chronic neurodegenerative disease characterized by a significant loss of dopaminergic neurons within the substantia nigra pars compacta (SNpc) and a subsequent loss of dopamine (DA) within the striatum. Despite advances in the development of pharmacological therapies that are effective at alleviating the symptoms of PD, the search for therapeutic treatments that halt or slow the underlying nigral degeneration remains a particular challenge. Activin A, a member of the transforming growth factor β superfamily, has been shown to play a role in the neuroprotection of midbrain neurons against 6-hydroxydopamine (6-OHDA) in vitro, suggesting that activin A may offer similar neuroprotective effects in in vivo models of PD. Using robust stereological methods, we found that intrastriatal injections of 6-OHDA results in a significant loss of both TH positive and NeuN positive populations in the SNpc at 1, 2, and 3 weeks post-lesioning in drug naïve mice. Exogenous application of activin A for 7 days, beginning the day prior to 6-OHDA administration, resulted in a significant survival of both dopaminergic and total neuron numbers in the SNpc against 6-OHDA-induced toxicity. However, we found no corresponding protection of striatal DA or dopamine transporter (DAT) expression levels in animals receiving activin A compared to vehicle controls. These results provide the first evidence that activin A exerts potent neuroprotection in a mouse model of PD, however this neuroprotection may be localized to the midbrain.
Collapse
Affiliation(s)
- Sandy Stayte
- Neuroscience Department, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- Faculty of Medicine, UNSW Australia, Sydney, Australia
| | - Peggy Rentsch
- Neuroscience Department, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- School of Biotechnology and Biomolecular Sciences, UNSW Australia, Sydney, Australia
| | - Kong M. Li
- Pharmacology Department, Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Bryce Vissel
- Neuroscience Department, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- * E-mail:
| |
Collapse
|
39
|
Hadaczek P, Wu G, Sharma N, Ciesielska A, Bankiewicz K, Davidow AL, Lu ZH, Forsayeth J, Ledeen RW. GDNF signaling implemented by GM1 ganglioside; failure in Parkinson's disease and GM1-deficient murine model. Exp Neurol 2014; 263:177-89. [PMID: 25448159 DOI: 10.1016/j.expneurol.2014.10.010] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 09/16/2014] [Accepted: 10/16/2014] [Indexed: 11/26/2022]
Abstract
GDNF is indispensible for adult catecholaminergic neuron survival, and failure of GDNF signaling has been linked to loss of dopaminergic neurons in Parkinson's disease (PD). This study demonstrates attenuated GDNF signaling in neurons deficient in ganglio-series gangliosides, and restoration of such signaling with LIGA20, a membrane permeable analog of GM1. GM1 is shown to associate in situ with GFRα1 and RET, the protein components of the GDNF receptor, this being necessary for assembly of the tripartite receptor complex. Mice wholly or partially deficient in GM1 due to disruption of the B4galnt1 gene developed PD symptoms based on behavioral and neuropathological criteria which were largely ameliorated by gene therapy with AAV2-GDNF and also with LIGA20 treatment. The nigral neurons of PD subjects that were severely deficient in GM1 showed subnormal levels of tyrosine phosphorylated RET. Also in PD brain, GM1 levels in the occipital cortex, a region of limited PD pathology, were significantly below age-matched controls, suggesting the possibility of systemic GM1 deficiency as a risk factor in PD. This would accord with our finding that mice with partial GM1 deficiency represent a faithful recapitulation of the human disease. Together with the previously demonstrated age-related decline of GM1 in human brain, this points to gradual development of subthreshold levels of GM1 in the brain of PD subjects below that required for effective GDNF signaling. This hypothesis offers a dramatically different explanation for the etiology of sporadic PD as a manifestation of acquired resistance to GDNF.
Collapse
Affiliation(s)
- Piotr Hadaczek
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94103-0555, USA
| | - Gusheng Wu
- Department of Neurology and Neurosciences MSB-H506, The State University of New Jersey, 185 South Orange Ave., Newark, NJ 07103, USA
| | - Nitasha Sharma
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94103-0555, USA
| | - Agnieszka Ciesielska
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94103-0555, USA
| | - Krystof Bankiewicz
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94103-0555, USA
| | - Amy L Davidow
- Department of Biostatistics/Epidemiology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 South Orange Ave., Newark, NJ 07103, USA
| | - Zi-Hua Lu
- Department of Neurology and Neurosciences MSB-H506, The State University of New Jersey, 185 South Orange Ave., Newark, NJ 07103, USA
| | - John Forsayeth
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94103-0555, USA.
| | - Robert W Ledeen
- Department of Neurology and Neurosciences MSB-H506, The State University of New Jersey, 185 South Orange Ave., Newark, NJ 07103, USA.
| |
Collapse
|
40
|
Ghosh B, Zhang C, Smith GM. Bridging between transplantation therapy and neurotrophic factors in Parkinson's disease. Front Biosci (Elite Ed) 2014; 6:225-35. [PMID: 24896204 PMCID: PMC11375561 DOI: 10.2741/e704] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Parkinson's disease (PD) represents a challenging condition where different therapeutic options have evolved over the course of the last 50 years. The potential for therapeutic use of cell transplantation for cell replacement or for gene delivery of neurotrophic factors has received a great deal of attention. Currently, all available treatment options are directed towards the amelioration of symptoms. A greater understanding of the distinctive pathology underlying PD might offer some novel therapeutic approaches. Transplantation of embryonic ventral mesencephalon (VM) dopaminergic neurons has shown promise in animal studies, but similar transplant procedures have shown limited success in clinical trials. One important issue may be the site of transplantation. Previous studies have transplanted VM into the striatum, which is the target of these neurons. With increased understanding of growth and guidance molecule effecting dopaminergic neurons, it may be feasible to place transplants in the damaged substantia nigra and direct the growth of axons into target regions to reconstruction of midbrain dopamine (DA) circuitry. Our established and on-going understanding of the molecular cues which support directed growth of DA neurons form an important basis for the refinement and optimization of VM grafting procedures, and also the development of new procedures based on the use of stem cells. In this review, we discuss transplantation therapy and how selective guidance molecules could be used to reconstruction of nigrostriatal circuit.
Collapse
Affiliation(s)
- Biswarup Ghosh
- Center for Neural Repair and Rehabilitation, Department of Neuroscience, and Shriners Hospitals for Pediatric Research, Temple University, School of Medicine, Philadelphia, PA 19140-4106
| | - Chen Zhang
- Center for Neural Repair and Rehabilitation, Department of Neuroscience, and Shriners Hospitals for Pediatric Research, Temple University, School of Medicine, Philadelphia, PA 19140-4106
| | - George M Smith
- Center for Neural Repair and Rehabilitation, Department of Neuroscience, and Shriners Hospitals for Pediatric Research, Temple University, School of Medicine, Philadelphia, PA 19140-4106
| |
Collapse
|
41
|
Stayte S, Vissel B. Advances in non-dopaminergic treatments for Parkinson's disease. Front Neurosci 2014; 8:113. [PMID: 24904259 PMCID: PMC4033125 DOI: 10.3389/fnins.2014.00113] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 04/30/2014] [Indexed: 01/05/2023] Open
Abstract
Since the 1960's treatments for Parkinson's disease (PD) have traditionally been directed to restore or replace dopamine, with L-Dopa being the gold standard. However, chronic L-Dopa use is associated with debilitating dyskinesias, limiting its effectiveness. This has resulted in extensive efforts to develop new therapies that work in ways other than restoring or replacing dopamine. Here we describe newly emerging non-dopaminergic therapeutic strategies for PD, including drugs targeting adenosine, glutamate, adrenergic, and serotonin receptors, as well as GLP-1 agonists, calcium channel blockers, iron chelators, anti-inflammatories, neurotrophic factors, and gene therapies. We provide a detailed account of their success in animal models and their translation to human clinical trials. We then consider how advances in understanding the mechanisms of PD, genetics, the possibility that PD may consist of multiple disease states, understanding of the etiology of PD in non-dopaminergic regions as well as advances in clinical trial design will be essential for ongoing advances. We conclude that despite the challenges ahead, patients have much cause for optimism that novel therapeutics that offer better disease management and/or which slow disease progression are inevitable.
Collapse
Affiliation(s)
- Sandy Stayte
- Neuroscience Department, Neurodegenerative Disorders Laboratory, Garvan Institute of Medical Research, Sydney NSW, Australia ; Faculty of Medicine, University of New South Wales, Sydney NSW, Australia
| | - Bryce Vissel
- Neuroscience Department, Neurodegenerative Disorders Laboratory, Garvan Institute of Medical Research, Sydney NSW, Australia ; Faculty of Medicine, University of New South Wales, Sydney NSW, Australia
| |
Collapse
|
42
|
San Sebastian W, Kells AP, Bringas J, Samaranch L, Hadaczek P, Ciesielska A, Macayan M, Pivirotto PJ, Forsayeth J, Osborne S, Wright JF, Green F, Heller G, Bankiewicz KS. SAFETY AND TOLERABILITY OF MRI-GUIDED INFUSION OF AAV2-hAADC INTO THE MID-BRAIN OF NON-HUMAN PRIMATE. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2014; 3:S2329-0501(16)30117-6. [PMID: 25541617 PMCID: PMC4274790 DOI: 10.1038/mtm.2014.49] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Aromatic L-amino acid decarboxylase (AADC) deficiency is a rare, autosomal-recessive neurological disorder caused by mutations in the DDC gene that leads to an inability to synthesize catecholamines and serotonin. As a result, patients suffer compromised development, particularly in motor function. A recent gene replacement clinical trial explored putaminal delivery of recombinant adeno-associated virus serotype 2 vector encoding human AADC (AAV2-hAADC) in AADC-deficient children. Unfortunately, patients presented only modest amelioration of motor symptoms, which authors acknowledged could be due to insufficient transduction of putamen. We hypothesize that, with the development of a highly accurate MRI-guided cannula placement technology, a more effective approach might be to target the affected mid-brain neurons directly. Transduction of AADC-deficient dopaminergic neurons in the substantia nigra and ventral tegmental area with locally infused AAV2-hAADC would be expected to lead to restoration of normal dopamine levels in affected children. The objective of this study was to assess the long-term safety and tolerability of bilateral AAV2-hAADC MRI-guided pressurized infusion into the mid-brain of nonhuman primates. Animals received either vehicle, low or high AAV2-hAADC vector dose and were euthanized 1, 3, or 9 months after surgery. Our data indicate that effective mid-brain transduction was achieved without untoward effects.
Collapse
Affiliation(s)
- Waldy San Sebastian
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Adrian P Kells
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - John Bringas
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Lluis Samaranch
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Piotr Hadaczek
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Agnieszka Ciesielska
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Michael Macayan
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Phillip J Pivirotto
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - John Forsayeth
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | | | - J Fraser Wright
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA ; Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Foad Green
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Gregory Heller
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Krystof S Bankiewicz
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
43
|
Glavaski-Joksimovic A, Bohn MC. Mesenchymal stem cells and neuroregeneration in Parkinson's disease. Exp Neurol 2013; 247:25-38. [DOI: 10.1016/j.expneurol.2013.03.016] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 03/14/2013] [Indexed: 02/06/2023]
|
44
|
Kordower JH, Bjorklund A. Trophic factor gene therapy for Parkinson's disease. Mov Disord 2013; 28:96-109. [PMID: 23390096 DOI: 10.1002/mds.25344] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 12/05/2012] [Accepted: 12/13/2012] [Indexed: 11/06/2022] Open
Abstract
Parkinson's disease (PD) is a chronic and progressive neurodegenerative movement disorder for which there is presently no cure. Pharmacological remedies targeting the dopaminergic network are relatively effective at ameliorating motor deficits, especially in the early stages of the disease, but none of these therapies are curative and many generate their own problems. Recent advances in PD research have demonstrated that gene delivery of trophic factors, glial cell line-derived neurotrophic factor (GDNF) and neurturin, in particular, can provide structural and functional recovery in rodent and nonhuman primate models of PD. Similar success has been gleaned in open-label clinical trials, although this has yet to be realized in double-blinded analyses. This work reviews the field of trophic factor gene delivery for PD.
Collapse
Affiliation(s)
- Jeffrey H Kordower
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois 60612, USA.
| | | |
Collapse
|
45
|
Redmond DE, McEntire CRS, Kingsbery JP, Leranth C, Elsworth JD, Bjugstad KB, Roth RH, Samulski RJ, Sladek JR. Comparison of fetal mesencephalic grafts, AAV-delivered GDNF, and both combined in an MPTP-induced nonhuman primate Parkinson's model. Mol Ther 2013; 21:2160-8. [PMID: 23913185 DOI: 10.1038/mt.2013.180] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Accepted: 07/20/2013] [Indexed: 02/08/2023] Open
Abstract
We combined viral vector delivery of human glial-derived neurotrophic factor (GDNF) with the grafting of dopamine (DA) precursor cells from fetal ventral mesencephalon (VM) to determine whether these strategies would improve the anti-Parkinson's effects in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated monkeys, an animal model for Parkinson's disease (PD). Both strategies have been reported as individually beneficial in animal models of PD, leading to clinical studies. GDNF delivery has also been reported to augment VM tissue implants, but no combined studies have been done in monkeys. Monkeys were treated with MPTP and placed into four balanced treatment groups receiving only recombinant adeno-associated virus serotype 5 (rAAV5)/hu-GDNF, only fetal DA precursor cells, both together, or a buffered saline solution (control). The combination of fetal precursors with rAAV5/hu-GDNF showed significantly higher striatal DA concentrations compared with the other treatments, but did not lead to greater functional improvement in this study. For the first time under identical conditions in primates, we show that all three treatments lead to improvement compared with control animals.
Collapse
Affiliation(s)
- D Eugene Redmond
- 1] Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA [2] Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Intrastriatal gene delivery of GDNF persistently attenuates methamphetamine self-administration and relapse in mice. Int J Neuropsychopharmacol 2013; 16:1559-67. [PMID: 23432945 DOI: 10.1017/s1461145712001575] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Relapse of drug abuse after abstinence is a major challenge to the treatment of addicts. In our well-established mouse models of methamphetamine (Meth) self-administration and reinstatement, bilateral microinjection of adeno-associated virus vectors expressing GDNF (AAV-Gdnf) into the striatum significantly reduced Meth self-administration, without affecting locomotor activity. Moreover, the intrastriatal AAV-Gdnf attenuated cue-induced reinstatement of Meth-seeking behaviour in a sustainable manner. In addition, this manipulation showed that Meth-primed reinstatement of Meth-seeking behaviour was reduced. These findings suggest that the AAV vector-mediated Gdnf gene transfer into the striatum is an effective and sustainable approach to attenuate Meth self-administration and Meth-associated cue-induced relapsing behaviour and that the AAV-mediated Gdnf gene transfer in the brain may be a valuable gene therapy against drug dependence and protracted relapse in clinical settings.
Collapse
|
47
|
Allen SJ, Watson JJ, Shoemark DK, Barua NU, Patel NK. GDNF, NGF and BDNF as therapeutic options for neurodegeneration. Pharmacol Ther 2013; 138:155-75. [PMID: 23348013 DOI: 10.1016/j.pharmthera.2013.01.004] [Citation(s) in RCA: 568] [Impact Index Per Article: 51.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 01/07/2013] [Indexed: 12/16/2022]
Abstract
Glial cell-derived neurotrophic factor (GDNF), and the neurotrophin nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) are important for the survival, maintenance and regeneration of specific neuronal populations in the adult brain. Depletion of these neurotrophic factors has been linked with disease pathology and symptoms, and replacement strategies are considered as potential therapeutics for neurodegenerative diseases such as Parkinson's, Alzheimer's and Huntington's diseases. GDNF administration has recently been shown to be an effective treatment for Parkinson's disease, with clinical trials currently in progress. Trials with NGF for Alzheimer's disease are ongoing, with some degree of success. Preclinical results using BDNF also show much promise, although there are accompanying difficulties. Ultimately, the administration of a therapy involving proteins in the brain has inherent problems. Because of the blood-brain-barrier, the protein must be infused directly, produced by viral constructs, secreted from implanted protein-secreting cells or actively transported across the brain. An alternative to this is the use of a small molecule agonist, a modulator or enhancer targeting the associated receptors. We evaluate these neurotrophic factors as potential short or long-term treatments, weighing up preclinical and clinical results with the possible effects on the underlying neurodegenerative process.
Collapse
|
48
|
Coune PG, Schneider BL, Aebischer P. Parkinson's disease: gene therapies. Cold Spring Harb Perspect Med 2013; 2:a009431. [PMID: 22474617 DOI: 10.1101/cshperspect.a009431] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
With the recent development of effective gene delivery systems, gene therapy for the central nervous system is finding novel applications. Here, we review existing viral vectors and discuss gene therapy strategies that have been proposed for Parkinson's disease. To date, most of the clinical trials were based on viral vectors to deliver therapeutic transgenes to neurons within the basal ganglia. Initial trials used genes to relieve the major motor symptoms caused by nigrostriatal degeneration. Although these new genetic approaches still need to prove more effective than existing symptomatic treatments, there is a need for disease-modifying strategies. The investigation of the genetic factors implicated in Parkinson's disease is providing precious insights in disease pathology that, combined with innovative gene delivery systems, will hopefully offer novel opportunities for gene therapy interventions to slow down, or even halt disease progression.
Collapse
Affiliation(s)
- Philippe G Coune
- Neurodegenerative Studies Laboratory, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | | | | |
Collapse
|
49
|
Bäck S, Peränen J, Galli E, Pulkkila P, Lonka-Nevalaita L, Tamminen T, Voutilainen MH, Raasmaja A, Saarma M, Männistö PT, Tuominen RK. Gene therapy with AAV2-CDNF provides functional benefits in a rat model of Parkinson's disease. Brain Behav 2013; 3:75-88. [PMID: 23532969 PMCID: PMC3607149 DOI: 10.1002/brb3.117] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 11/29/2012] [Accepted: 12/16/2012] [Indexed: 11/10/2022] Open
Abstract
Cerebral dopamine neurotrophic factor (CDNF) protein has been shown to protect the nigrostriatal dopaminergic pathway when given as intrastriatal infusions in rat and mouse models of Parkinson's disease (PD). In this study, we assessed the neuroprotective effect of CDNF delivered with a recombinant adeno-associated viral (AAV) serotype 2 vector in a rat 6-hydroxydopamine (6-OHDA) model of PD. AAV2 vectors encoding CDNF, glial cell line-derived neurotrophic factor (GDNF), or green fluorescent protein were injected into the rat striatum. Protein expression analysis showed that our AAV2 vector efficiently delivered the neurotrophic factor genes into the brain and gave rise to a long-lasting expression of the proteins. Two weeks after AAV2 vector injection, 6-OHDA was injected into the rat striatum, creating a progressive degeneration of the nigrostriatal dopaminergic system. Treatment with AAV2-CDNF resulted in a marked decrease in amphetamine-induced ipsilateral rotations while it provided only partial protection of tyrosine hydroxylase (TH)-immunoreactive cells in the rat substantia nigra pars compacta and TH-reactive fibers in the striatum. Results from this study provide additional evidence that CDNF can be considered a potential treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Susanne Bäck
- Division of Pharmacology and Toxicology Faculty of Pharmacy, University of Helsinki Helsinki, Finland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Miranpuri GS, Kumbier L, Hinchman A, Schomberg D, Wang A, Marshall H, Kubota K, Ross C, Sillay K. Gene-based therapy of Parkinson's Disease: Translation from animal model to human clinical trial employing convection enhanced delivery. Ann Neurosci 2012; 19:133-46. [PMID: 25205986 PMCID: PMC4117084 DOI: 10.5214/ans.0972.7531.190310] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Revised: 07/02/2012] [Accepted: 07/02/2012] [Indexed: 11/17/2022] Open
Abstract
The existing treatment of Parkinson's disease (PD) is directed towards substituting dopamine loss with either dopamine replacement therapy or pharmacological therapies aimed at increasing dopamine at the synapse level. Emerging viable alternatives include the use of cell-based and gene-based therapeutics. In this review, we discuss efforts in developing in vitro and in vivo models and their translation to human clinical trials for gene-based therapy of this distressing and prevalent neurodegenerative disorder. Given the mismatch between expectations from preclinical data and results of human pivotal trials, drug delivery has been identified as the key emerging area for translational research due to limitation of limited efficacy. The chief highlights of the current topic include use of improved delivery methods of gene-based therapeutic agents. Convection-enhanced delivery (CED), an advanced infusion technique with demonstrated utility in ex vivo and in vivo animal models has recently been adopted for PD gene-based therapy trials. Several preclinical studies suggest that magnetic resonance imaging (MRI)-guided navigation for accurately targeting and real time monitoring viral vector delivery (rCED) in future clinical trials involving detection of gene expression and restoration of dopaminergic function loss using pro-drug approach will greatly enhance these PD treatments.
Collapse
Affiliation(s)
- Gurwattan S. Miranpuri
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, USA
| | - Lauren Kumbier
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, USA
| | - Angelica Hinchman
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, USA
| | - Dominic Schomberg
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, USA
| | - Anyi Wang
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, USA
| | - Hope Marshall
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, USA
| | - Ken Kubota
- Kinetic Foundation, Los Altos, CA, 94023, USA
| | - Chris Ross
- Engineering Resources Group Inc, Pembroke Pines, FL, 33029, USA
| | - Karl Sillay
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, USA
| |
Collapse
|