1
|
Mercado NM, Szarowicz C, Stancati JA, Sortwell CE, Boezwinkle SA, Collier TJ, Caulfield ME, Steece-Collier K. Advancing age and the rs6265 BDNF SNP are permissive to graft-induced dyskinesias in parkinsonian rats. NPJ Parkinsons Dis 2024; 10:163. [PMID: 39179609 PMCID: PMC11344059 DOI: 10.1038/s41531-024-00771-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 08/05/2024] [Indexed: 08/26/2024] Open
Abstract
The rs6265 single nucleotide polymorphism (SNP) in the gene for brain-derived neurotrophic factor is a common variant that alters therapeutic outcomes for individuals with Parkinson's disease (PD). We previously investigated the effects of this SNP on the experimental therapeutic approach of neural grafting, demonstrating that young adult parkinsonian rats carrying the variant Met allele exhibited enhanced graft function compared to wild-type rats and also exclusively developed aberrant graft-induced dyskinesias (GID). Aging is the primary risk factor for PD and reduces graft efficacy. Here we investigated whether aging interacts with this SNP to further alter cell transplantation outcomes. We hypothesized that aging would reduce enhancement of graft function associated with this genetic variant and exacerbate GID in all grafted subjects. Unexpectedly, beneficial graft function was maintained in aged rs6265 subjects. However, aging was permissive to GID induction, regardless of genotype, with the greatest incidence and severity found in rs6265-expressing animals.
Collapse
Affiliation(s)
- Natosha M Mercado
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA
| | - Carlye Szarowicz
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Jennifer A Stancati
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA
| | - Caryl E Sortwell
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA
- Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, MI, 49503, USA
| | - Samuel A Boezwinkle
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Timothy J Collier
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA
- Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, MI, 49503, USA
| | - Margaret E Caulfield
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA
| | - Kathy Steece-Collier
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA.
- Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, MI, 49503, USA.
| |
Collapse
|
2
|
Kitaeva KV, Solovyeva VV, Blatt NL, Rizvanov AA. Eternal Youth: A Comprehensive Exploration of Gene, Cellular, and Pharmacological Anti-Aging Strategies. Int J Mol Sci 2024; 25:643. [PMID: 38203812 PMCID: PMC10778954 DOI: 10.3390/ijms25010643] [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: 11/08/2023] [Revised: 12/21/2023] [Accepted: 12/30/2023] [Indexed: 01/12/2024] Open
Abstract
The improvement of human living conditions has led to an increase in average life expectancy, creating a new social and medical problem-aging, which diminishes the overall quality of human life. The aging process of the body begins with the activation of effector signaling pathways of aging in cells, resulting in the loss of their normal functions and deleterious effects on the microenvironment. This, in turn, leads to chronic inflammation and similar transformations in neighboring cells. The cumulative retention of these senescent cells over a prolonged period results in the deterioration of tissues and organs, ultimately leading to a reduced quality of life and an elevated risk of mortality. Among the most promising methods for addressing aging and age-related illnesses are pharmacological, genetic, and cellular therapies. Elevating the activity of aging-suppressing genes, employing specific groups of native and genetically modified cells, and utilizing senolytic medications may offer the potential to delay aging and age-related ailments over the long term. This review explores strategies and advancements in the field of anti-aging therapies currently under investigation, with a particular emphasis on gene therapy involving adeno-associated vectors and cell-based therapeutic approaches.
Collapse
Affiliation(s)
- Kristina V. Kitaeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (K.V.K.); (V.V.S.); (N.L.B.)
| | - Valeriya V. Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (K.V.K.); (V.V.S.); (N.L.B.)
| | - Nataliya L. Blatt
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (K.V.K.); (V.V.S.); (N.L.B.)
| | - Albert A. Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (K.V.K.); (V.V.S.); (N.L.B.)
- Division of Medical and Biological Sciences, Tatarstan Academy of Sciences, 420111 Kazan, Russia
| |
Collapse
|
3
|
Ling Q, Herstine JA, Bradbury A, Gray SJ. AAV-based in vivo gene therapy for neurological disorders. Nat Rev Drug Discov 2023; 22:789-806. [PMID: 37658167 DOI: 10.1038/s41573-023-00766-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2023] [Indexed: 09/03/2023]
Abstract
Recent advancements in gene supplementation therapy are expanding the options for the treatment of neurological disorders. Among the available delivery vehicles, adeno-associated virus (AAV) is often the favoured vector. However, the results have been variable, with some trials dramatically altering the course of disease whereas others have shown negligible efficacy or even unforeseen toxicity. Unlike traditional drug development with small molecules, therapeutic profiles of AAV gene therapies are dependent on both the AAV capsid and the therapeutic transgene. In this rapidly evolving field, numerous clinical trials of gene supplementation for neurological disorders are ongoing. Knowledge is growing about factors that impact the translation of preclinical studies to humans, including the administration route, timing of treatment, immune responses and limitations of available model systems. The field is also developing potential solutions to mitigate adverse effects, including AAV capsid engineering and designs to regulate transgene expression. At the same time, preclinical research is addressing new frontiers of gene supplementation for neurological disorders, with a focus on mitochondrial and neurodevelopmental disorders. In this Review, we describe the current state of AAV-mediated neurological gene supplementation therapy, including critical factors for optimizing the safety and efficacy of treatments, as well as unmet needs in this field.
Collapse
Affiliation(s)
- Qinglan Ling
- Department of Paediatrics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jessica A Herstine
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Paediatrics, The Ohio State University, Columbus, OH, USA
| | - Allison Bradbury
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Paediatrics, The Ohio State University, Columbus, OH, USA
| | - Steven J Gray
- Department of Paediatrics, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
4
|
Lejman J, Panuciak K, Nowicka E, Mastalerczyk A, Wojciechowska K, Lejman M. Gene Therapy in ALS and SMA: Advances, Challenges and Perspectives. Int J Mol Sci 2023; 24:ijms24021130. [PMID: 36674643 PMCID: PMC9860634 DOI: 10.3390/ijms24021130] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/31/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Gene therapy is defined as the administration of genetic material to modify, manipulate gene expression or alter the properties of living cells for therapeutic purposes. Recent advances and improvements in this field have led to many breakthroughs in the treatment of various diseases. As a result, there has been an increasing interest in the use of these therapies to treat motor neuron diseases (MNDs), for which many potential molecular targets have been discovered. MNDs are neurodegenerative disorders that, in their most severe forms, can lead to respiratory failure and death, for instance, spinal muscular atrophy (SMA) or amyotrophic lateral sclerosis (ALS). Despite the fact that SMA has been known for many years, it is still one of the most common genetic diseases causing infant mortality. The introduction of drugs based on ASOs-nusinersen; small molecules-risdiplam; and replacement therapy (GRT)-Zolgensma has shown a significant improvement in both event-free survival and the quality of life of patients after using these therapies in the available trial results. Although there is still no drug that would effectively alleviate the course of the disease in ALS, the experience gained from SMA gene therapy gives hope for a positive outcome of the efforts to produce an effective and safe drug. The aim of this review is to present current progress and prospects for the use of gene therapy in the treatment of both SMA and ALS.
Collapse
Affiliation(s)
- Jan Lejman
- Student Scientific Society, Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
- Correspondence:
| | - Kinga Panuciak
- Student Scientific Society, Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
| | - Emilia Nowicka
- Student Scientific Society, Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
| | - Angelika Mastalerczyk
- Student Scientific Society, Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
| | - Katarzyna Wojciechowska
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
| | - Monika Lejman
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
5
|
Motaghinejad M, Gholami M, Emanuele E. Constant romantic feelings and experiences can protect against neurodegeneration: Potential role of oxytocin-induced nerve growth factor/protein kinase B/Cyclic response element-binding protein and nerve growth factor/protein kinase B/Phospholipase C-Gamma signaling pathways. BIOMEDICAL AND BIOTECHNOLOGY RESEARCH JOURNAL (BBRJ) 2023. [DOI: 10.4103/bbrj.bbrj_28_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
|
6
|
Caulfield ME, Manfredsson FP, Steece-Collier K. The Role of Striatal Cav1.3 Calcium Channels in Therapeutics for Parkinson's Disease. Handb Exp Pharmacol 2023; 279:107-137. [PMID: 36592226 DOI: 10.1007/164_2022_629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Parkinson's disease (PD) is a relentlessly progressive neurodegenerative disorder with typical motor symptoms that include rigidity, tremor, and akinesia/bradykinesia, in addition to a host of non-motor symptoms. Motor symptoms are caused by progressive and selective degeneration of dopamine (DA) neurons in the SN pars compacta (SNpc) and the accompanying loss of striatal DA innervation from these neurons. With the exception of monogenic forms of PD, the etiology of idiopathic PD remains unknown. While there are a number of symptomatic treatment options available to individuals with PD, these therapies do not work uniformly well in all patients, and eventually most are plagued with waning efficacy and significant side-effect liability with disease progression. The incidence of PD increases with aging, and as such the expected burden of this disease will continue to escalate as our aging population increases (Dorsey et al. Neurology 68:384-386, 2007). The daunting personal and socioeconomic burden has pressed scientists and clinicians to find improved symptomatic treatment options devoid side-effect liability and meaningful disease-modifying therapies. Federal and private sources have supported clinical investigations over the past two-plus decades; however, no trial has yet been successful in finding an effective therapy to slow progression of PD, and there is currently just one FDA approved drug to treat the antiparkinsonian side-effect known as levodopa-induced dyskinesia (LID) that impacts approximately 90% of all individuals with PD. In this review, we present biological rationale and experimental evidence on the potential therapeutic role of the L-type voltage-gated Cav1.3 calcium (Ca2+) channels in two distinct brain regions, with two distinct mechanisms of action, in impacting the lives of individuals with PD. Our primary emphasis will be on the role of Cav1.3 channels in the striatum and the compelling evidence of their involvement in LID side-effect liability. We also briefly discuss the role of these same Ca2+ channels in the SNpc and the longstanding interest in Cav1.3 in this brain region in halting or delaying progression of PD.
Collapse
Affiliation(s)
- Margaret E Caulfield
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Fredric P Manfredsson
- Parkinson's Disease Research Unit, Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Kathy Steece-Collier
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA.
- Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, MI, USA.
| |
Collapse
|
7
|
Quintero JE, Slevin JT, Gurwell JA, McLouth CJ, El Khouli R, Chau MJ, Guduru Z, Gerhardt GA, van Horne CG. Direct delivery of an investigational cell therapy in patients with Parkinson's disease: an interim analysis of feasibility and safety of an open-label study using DBS-Plus clinical trial design. BMJ Neurol Open 2022; 4:e000301. [PMID: 35949912 PMCID: PMC9295654 DOI: 10.1136/bmjno-2022-000301] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/13/2022] [Indexed: 12/29/2022] Open
Abstract
Objective To evaluate the interim feasibility, safety and clinical measures data of direct delivery of regenerating peripheral nerve tissue (PNT) to the substantia nigra (SN) in participants with Parkinson’s disease (PD). Methods Eighteen (13 men/5 women) participants were unilaterally implanted with PNT to the SN, contralateral to the most affected side during the same surgery they were receiving deep brain stimulation (DBS) surgery. Autologous PNT was collected from the sural nerve. Participants were followed for safety and clinical outcomes for 2 years (including off-state Unified Parkinson’s Disease Rating Scale (UPDRS) Part III assessments) with study visits every 6 months. Results All 18 participants scheduled to receive PNT implantation received targeted delivery to the SN in addition to their DBS. All subjects were discharged the following day except for two: post-op day 2; post-op day 3. The most common study-related adverse events were hypoaesthesia and hyperaesthesias to the lateral aspect of the foot and ankle of the biopsied nerve (6 of 18 participants experienced). Clinical measures did not identify any hastening of PD measures providing evidence of safety and tolerability. Off-state UPDRS Part III mean difference scores were reduced at 12 months compared with baseline (difference=−8.1, 95% CI −2.4 to −13.9 points, p=0.005). No complications involving dyskinesias were observed. Conclusions Targeting the SN for direct delivery of PNT was feasible with no serious adverse events related to the study intervention. Interim clinical outcomes show promising results meriting continued examination of this investigational approach. Trial registration number NCT02369003.
Collapse
Affiliation(s)
- Jorge E Quintero
- Neurosurgery, University of Kentucky Medical Center, Lexington, Kentucky, USA.,Neuroscience, University of Kentucky Medical Center, Lexington, Kentucky, USA
| | - John T Slevin
- Neurology, University of Kentucky Medical Center, Lexington, Kentucky, USA.,Neurology, VA Medical Center, Lexington, Kentucky, USA
| | - Julie A Gurwell
- Neurology, University of Kentucky Medical Center, Lexington, Kentucky, USA
| | | | - Riham El Khouli
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, University of Kentucky Medical Center, Lexington, Kentucky, USA
| | - Monica J Chau
- Neurosurgery, University of Kentucky Medical Center, Lexington, Kentucky, USA
| | - Zain Guduru
- Neurology, University of Kentucky Medical Center, Lexington, Kentucky, USA
| | - Greg A Gerhardt
- Neurosurgery, University of Kentucky Medical Center, Lexington, Kentucky, USA.,Neuroscience, University of Kentucky Medical Center, Lexington, Kentucky, USA.,Neurology, University of Kentucky Medical Center, Lexington, Kentucky, USA
| | - Craig G van Horne
- Neurosurgery, University of Kentucky Medical Center, Lexington, Kentucky, USA.,Neuroscience, University of Kentucky Medical Center, Lexington, Kentucky, USA
| |
Collapse
|
8
|
Therapeutic potential of neurotrophic factors in Alzheimer's Disease. Mol Biol Rep 2021; 49:2345-2357. [PMID: 34826049 DOI: 10.1007/s11033-021-06968-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/17/2021] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the most common cause of dementia among the elderly population. AD is accompanied with the dysregulation of specific neurotrophic factors (NTFs) and their receptors, which plays a critical role in neuronal degeneration. NTFs are small proteins with therapeutic potential for human neurodegenerative diseases. These growth factors are categorized into four families: neurotrophins, neurokines, the glial cell line-derived NTF family of ligands, and the newly discovered cerebral dopamine NTF/mesencephalic astrocyte-derived NTF family. NTFs are capable of preventing cell death in degenerative conditions and can increase the neuronal growth and function in these disorders. Nevertheless, the adverse side effects of NTFs delivery and poor diffusion of these factors in the brain restrict the efficacy of NTFs therapy in clinical situations. MATERIALS AND METHODS In this review, we focus on the current advances in the use of NTFs to treat AD and summarize previous experimental and clinical studies for supporting the protective and therapeutic effects of these factors. CONCLUSION Based on reports, NTFs are considered as new and promising candidates for treating AD and AD-associated cognitive impairment.
Collapse
|
9
|
Kambey PA, Kanwore K, Ayanlaja AA, Nadeem I, Du Y, Buberwa W, Liu W, Gao D. Failure of Glial Cell-Line Derived Neurotrophic Factor (GDNF) in Clinical Trials Orchestrated By Reduced NR4A2 (NURR1) Transcription Factor in Parkinson's Disease. A Systematic Review. Front Aging Neurosci 2021; 13:645583. [PMID: 33716718 PMCID: PMC7943926 DOI: 10.3389/fnagi.2021.645583] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 01/29/2021] [Indexed: 12/23/2022] Open
Abstract
Parkinson’s disease (PD) is one of the most common neurodegenerative maladies with unforeseen complex pathologies. While this neurodegenerative disorder’s neuropathology is reasonably well known, its etiology remains a mystery, making it challenging to aim therapy. Glial cell-line derived neurotrophic factor (GDNF) remains an auspicious therapeutic molecule for treating PD. Neurotrophic factor derived from glial cell lines is effective in rodents and nonhuman primates, but clinical findings have been equivocal. Laborious exertions have been made over the past few decades to improve and assess GDNF in treating PD (clinical studies). Definitive clinical trials have, however, failed to demonstrate a survival advantage. Consequently, there seemed to be a doubt as to whether GDNF has merit in the potential treatment of PD. The purpose of this cutting edge review is to speculate as to why the clinical trials have failed to meet the primary endpoint. We introduce a hypothesis, “Failure of GDNF in clinical trials succumbed by nuclear receptor-related factor 1 (Nurr1) shortfall.” We demonstrate how Nurr1 binds to GDNF to induce dopaminergic neuron synthesis. Due to its undisputable neuro-protection aptitude, we display Nurr1 (also called Nr4a2) as a promising therapeutic target for PD.
Collapse
Affiliation(s)
- Piniel Alphayo Kambey
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Kouminin Kanwore
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Abiola Abdulrahman Ayanlaja
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Iqra Nadeem
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| | - YinZhen Du
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| | | | - WenYa Liu
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Dianshuai Gao
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
10
|
Gross SK, Shim BS, Bartus RT, Deaver D, McEachin Z, Bétourné A, Boulis NM, Maragakis NJ. Focal and dose-dependent neuroprotection in ALS mice following AAV2-neurturin delivery. Exp Neurol 2020; 323:113091. [DOI: 10.1016/j.expneurol.2019.113091] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 10/16/2019] [Accepted: 10/23/2019] [Indexed: 12/13/2022]
|
11
|
FGF2 and dual agonist of NCAM and FGF receptor 1, Enreptin, rescue neurite outgrowth loss in hippocampal neurons expressing mutated huntingtin proteins. J Neural Transm (Vienna) 2019; 126:1493-1500. [PMID: 31501979 DOI: 10.1007/s00702-019-02073-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 08/30/2019] [Indexed: 10/26/2022]
Abstract
In the present study, we developed an in vitro model of Huntington disease (HD) by transfecting primary rat hippocampal neurons with plasmids coding for m-htt exon 1 with different number of CAG repeats (18, 50 and 115) and demonstrated the influence of the length of polyQ sequence on neurite elongation. We found that exogenously applied FGF2 significantly rescued the m-htt-induced loss of neurite outgrowth. Moreover, the Enreptin peptide, an FGFR1 and NCAM dual agonist, had a similar neuritogenic effect to FGF2 in clinically relevant m-htt 50Q-expressing neurons. This study has developed an in vitro model of primary hippocampal neurons transfected with m-htt-coding vectors that is a powerful tool to study m-htt-related effects on neuronal placticity.
Collapse
|
12
|
González-Casacuberta I, Juárez-Flores DL, Morén C, Garrabou G. Bioenergetics and Autophagic Imbalance in Patients-Derived Cell Models of Parkinson Disease Supports Systemic Dysfunction in Neurodegeneration. Front Neurosci 2019; 13:894. [PMID: 31551675 PMCID: PMC6748355 DOI: 10.3389/fnins.2019.00894] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 08/09/2019] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder worldwide affecting 2-3% of the population over 65 years. This prevalence is expected to rise as life expectancy increases and diagnostic and therapeutic protocols improve. PD encompasses a multitude of clinical, genetic, and molecular forms of the disease. Even though the mechanistic of the events leading to neurodegeneration remain largely unknown, some molecular hallmarks have been repeatedly reported in most patients and models of the disease. Neuroinflammation, protein misfolding, disrupted endoplasmic reticulum-mitochondria crosstalk, mitochondrial dysfunction and consequent bioenergetic failure, oxidative stress and autophagy deregulation, are amongst the most commonly described. Supporting these findings, numerous familial forms of PD are caused by mutations in genes that are crucial for mitochondrial and autophagy proper functioning. For instance, late and early onset PD associated to mutations in Leucine-rich repeat kinase 2 (LRRK2) and Parkin (PRKN) genes, responsible for the most frequent dominant and recessive inherited forms of PD, respectively, have emerged as promising examples of disease due to their established role in commanding bioenergetic and autophagic balance. Concomitantly, the development of animal and cell models to investigate the etiology of the disease, potential biomarkers and therapeutic approaches are being explored. One of the emerging approaches in this context is the use of patient's derived cells models, such as skin-derived fibroblasts that preserve the genetic background and some environmental cues of the patients. An increasing number of reports in these PD cell models postulate that deficient mitochondrial function and impaired autophagic flux may be determinant in PD accelerated nigral cell death in terms of limitation of cell energy supply and accumulation of obsolete and/or unfolded proteins or dysfunctional organelles. The reliance of neurons on mitochondrial oxidative metabolism and their post-mitotic nature, may explain their increased vulnerability to undergo degeneration upon mitochondrial challenges or autophagic insults. In this scenario, proper mitochondrial function and turnover through mitophagy, are gaining in strength as protective targets to prevent neurodegeneration, together with the use of patient-derived fibroblasts to further explore these events. These findings point out the presence of molecular damage beyond the central nervous system (CNS) and proffer patient-derived cell platforms to the clinical and scientific community, which enable the study of disease etiopathogenesis and therapeutic approaches focused on modifying the natural history of PD through, among others, the enhancement of mitochondrial function and autophagy.
Collapse
Affiliation(s)
- Ingrid González-Casacuberta
- Muscle Research and Mitochondrial Function Laboratory, Cellex-IDIBAPS, Faculty of Medicine and Health Sciences-University of Barcelona, Internal Medicine Service-Hospital Clínic of Barcelona, Barcelona, Spain.,CIBERER-U722, Madrid, Spain
| | - Diana Luz Juárez-Flores
- Muscle Research and Mitochondrial Function Laboratory, Cellex-IDIBAPS, Faculty of Medicine and Health Sciences-University of Barcelona, Internal Medicine Service-Hospital Clínic of Barcelona, Barcelona, Spain.,CIBERER-U722, Madrid, Spain
| | - Constanza Morén
- Muscle Research and Mitochondrial Function Laboratory, Cellex-IDIBAPS, Faculty of Medicine and Health Sciences-University of Barcelona, Internal Medicine Service-Hospital Clínic of Barcelona, Barcelona, Spain.,CIBERER-U722, Madrid, Spain
| | - Gloria Garrabou
- Muscle Research and Mitochondrial Function Laboratory, Cellex-IDIBAPS, Faculty of Medicine and Health Sciences-University of Barcelona, Internal Medicine Service-Hospital Clínic of Barcelona, Barcelona, Spain.,CIBERER-U722, Madrid, Spain
| |
Collapse
|
13
|
Abstract
Neurotrophic factors (NTF) are a subgroup of growth factors that promote survival and
differentiation of neurons. Due to their neuroprotective and neurorestorative properties,
their therapeutic potential has been tested in various neurodegenerative diseases.
Bioavailability of NTFs in the target tissue remains a major challenge for NTF-based
therapies. Various intracerebral delivery approaches, both protein and gene
transfer-based, have been tested with varying outcomes. Three growth factors, glial
cell-line derived neurotrophic factor (GDNF), neurturin (NRTN) and platelet-derived growth
factor (PDGF-BB) have been tested in clinical trials in Parkinson’s disease (PD) during
the past 20 years. A new protein can now be added to this list, as cerebral dopamine
neurotrophic factor (CDNF) has recently entered clinical trials. Despite their misleading
names, CDNF, together with its closest relative mesencephalic astrocyte-derived
neurotrophic factor (MANF), form a novel family of unconventional NTF that are both
structurally and mechanistically distinct from other growth factors. CDNF and MANF are
localized mainly to the lumen of endoplasmic reticulum (ER) and their primary function
appears to be modulation of the unfolded protein response (UPR) pathway. Prolonged ER
stress, via the UPR signaling pathways, contributes to the pathogenesis in a number of
chronic degenerative diseases, and is an important target for therapeutic modulation.
Intraputamenally administered recombinant human CDNF has shown robust neurorestorative
effects in a number of small and large animal models of PD, and had a good safety profile
in preclinical toxicology studies. Intermittent monthly bilateral intraputamenal infusions
of CDNF are currently being tested in a randomized placebo-controlled phase I–II clinical
study in moderately advanced PD patients. Here, we review the history of growth
factor-based clinical trials in PD, and discuss how CDNF differs from the previously
tested growth factors.
Collapse
Affiliation(s)
- Henri J Huttunen
- 1 Herantis Pharma Plc, Espoo, Finland.,2 Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- 3 Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
14
|
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
|
15
|
Pandemrix-induced narcolepsy is associated with genes related to immunity and neuronal survival. EBioMedicine 2019; 40:595-604. [PMID: 30711515 PMCID: PMC6413474 DOI: 10.1016/j.ebiom.2019.01.041] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 01/14/2019] [Accepted: 01/22/2019] [Indexed: 12/24/2022] Open
Abstract
Background The incidence of narcolepsy rose sharply after the swine influenza A (H1N1) vaccination campaign with Pandemrix. Narcolepsy is an immune-related disorder with excessive daytime sleepiness. The most frequent form is strongly associated with HLA-DQB1*06:02, but only a minority of carriers develop narcolepsy. We aimed to identify genetic markers that predispose to Pandemrix-induced narcolepsy. Methods We tested for genome-wide and candidate gene associations in 42 narcolepsy cases and 4981 controls. Genotyping was performed on Illumina arrays, HLA alleles were imputed using SNP2HLA, and single nucleotide polymorphisms were imputed using the haplotype reference consortium panel. The genome-wide significance threshold was p < 5 × 10−8, and the nominal threshold was p < 0.05. Results were replicated in 32 cases and 7125 controls. Chromatin data was used for functional annotation. Findings Carrying HLA-DQB1*06:02 was significantly associated with narcolepsy, odds ratio (OR) 39.4 [95% confidence interval (CI) 11.3, 137], p = 7.9 × 10−9. After adjustment for HLA, GDNF-AS1 (rs62360233) was significantly associated, OR = 8.7 [95% CI 4.2, 17.5], p = 2.6 × 10−9, and this was replicated, OR = 3.4 [95% CI 1.2–9.6], p = 0.022. Functional analysis revealed variants in high LD with rs62360233 that might explain the detected association. The candidate immune-gene locus TRAJ (rs1154155) was nominally associated in both the discovery and replication cohorts, meta-analysis OR = 2.0 [95% CI 1.4, 2.8], p = 0.0002. Interpretation We found a novel association between Pandemrix-induced narcolepsy and the non-coding RNA gene GDNF-AS1, which has been shown to regulate expression of the essential neurotrophic factor GDNF. Changes in regulation of GDNF have been associated with neurodegenerative diseases. This finding may increase the understanding of disease mechanisms underlying narcolepsy. Associations between Pandemrix-induced narcolepsy and immune-related genes were replicated.
Collapse
|
16
|
van Horne CG, Quintero JE, Slevin JT, Anderson-Mooney A, Gurwell JA, Welleford AS, Lamm JR, Wagner RP, Gerhardt GA. Peripheral nerve grafts implanted into the substantia nigra in patients with Parkinson's disease during deep brain stimulation surgery: 1-year follow-up study of safety, feasibility, and clinical outcome. J Neurosurg 2018; 129:1550-1561. [PMID: 29451447 DOI: 10.3171/2017.8.jns163222] [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: 01/03/2017] [Accepted: 08/08/2017] [Indexed: 11/06/2022]
Abstract
OBJECTIVECurrently, there is no treatment that slows or halts the progression of Parkinson's disease. Delivery of various neurotrophic factors to restore dopaminergic function has become a focus of study in an effort to fill this unmet need for patients with Parkinson's disease. Schwann cells provide a readily available source of such factors. This study presents a 12-month evaluation of safety and feasibility, as well as the clinical response, of implanting autologous peripheral nerve grafts into the substantia nigra of patients with Parkinson's disease at the time of deep brain stimulation (DBS) surgery.METHODSStandard DBS surgery targeting the subthalamic nucleus was performed in 8 study participants. After DBS lead implantation, a section of the sural nerve containing Schwann cells was harvested and unilaterally grafted to the substantia nigra. Adverse events were continually monitored. Baseline clinical data were obtained during standard preoperative evaluations. Clinical outcome data were obtained with postoperative clinical evaluations, neuropsychological testing, and MRI at 1 year after surgery.RESULTSAll 8 participants were implanted with DBS systems and grafts. Adverse event profiles were comparable to those of standard DBS surgery with the exception of 1 superficial infection at the sural nerve harvest site. Three participants also reported numbness in the distribution of the sural nerve distal to the harvest site. Motor scores on Unified Parkinson's Disease Rating Scale (UPDRS) part III while the participant was off therapy at 12 months improved from baseline (mean ± SD 25.1 ± 15.9 points at 12 months vs 32.5 ± 9.7 points at baseline). An analysis of the lateralized UPDRS scores also showed a greater overall reduction in scores on the side contralateral to the graft.CONCLUSIONSPeripheral nerve graft delivery to the substantia nigra at the time of DBS surgery is feasible and safe based on the results of this initial pilot study. Clinical outcome data from this phase I trial suggests that grafting may have some clinical benefit and certainly warrants further study to determine if this is an efficacious and neurorestorative therapy.Clinical trial registration no.: NCT01833364 (clinicaltrials.gov).
Collapse
Affiliation(s)
- Craig G van Horne
- 1Brain Restoration Center and
- Departments of2Neurosurgery
- 1Brain Restoration Center and
| | | | - John T Slevin
- 1Brain Restoration Center and
- 4Neurology, University of Kentucky, Lexington, Kentucky
| | - Amelia Anderson-Mooney
- 1Brain Restoration Center and
- Departments of2Neurosurgery
- 4Neurology, University of Kentucky, Lexington, Kentucky
| | - Julie A Gurwell
- 1Brain Restoration Center and
- 4Neurology, University of Kentucky, Lexington, Kentucky
| | | | - John R Lamm
- 1Brain Restoration Center and
- Departments of2Neurosurgery
| | | | - Greg A Gerhardt
- 1Brain Restoration Center and
- Departments of2Neurosurgery
- 3Neuroscience, and
- 4Neurology, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
17
|
Maximising coverage of brain structures using controlled reflux, convection-enhanced delivery and the recessed step catheter. J Neurosci Methods 2018; 308:337-345. [DOI: 10.1016/j.jneumeth.2018.08.029] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 08/14/2018] [Accepted: 08/31/2018] [Indexed: 11/18/2022]
|
18
|
Sanchez-Ramos J, Song S, Kong X, Foroutan P, Martinez G, Dominguez-Viqueria W, Mohapatra S, Mohapatra S, Haraszti RA, Khvorova A, Aronin N, Sava V. Chitosan-Mangafodipir nanoparticles designed for intranasal delivery of siRNA and DNA to brain. J Drug Deliv Sci Technol 2017; 43:453-460. [PMID: 29805475 DOI: 10.1016/j.jddst.2017.11.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The overall objective of the present research was to develop a nanocarrier system for non-invasive delivery to brain of molecules useful for gene therapy. Manganese-containing nanoparticles (mNPs) carrying anti-eGFP siRNA were tested in cell cultures of eGFP-expressing cell line of mouse fibroblasts (NIH3T3). The optimal mNPs were then tested in vivo in mice. Following intranasal instillation, mNPs were visualized by 7T MRI throughout brain at 24 and 48 hrs. mNPs were effective in significantly reducing GFP mRNA expression in Tg GFP+ mice in olfactory bulb, striatum, hippocampus and cortex. Intranasal instillation of mNPS loaded with dsDNA encoding RFP also resulted in expression of the RFP in multiple brain regions. In conclusion, mNPs carrying siRNA, or dsDNA were capable of delivering the payload from nose to brain. This approach for delivery of gene therapies to humans, if successful, will have a significant impact on disease-modifying therapeutics of neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Shijie Song
- Department of Neurology, University of South Florida, Tampa, FL, USA
| | - Xiaoyuan Kong
- Department of Neurology, University of South Florida, Tampa, FL, USA
| | | | - Gary Martinez
- Mofftt Cancer Center and Research Institute, Tampa, FL, USA
| | | | | | | | - Reka A Haraszti
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Anastasia Khvorova
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Neil Aronin
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Vasyl Sava
- Department of Neurology, University of South Florida, Tampa, FL, USA
| |
Collapse
|
19
|
Chansel-Debordeaux L, Bourdenx M, Dovero S, Grouthier V, Dutheil N, Espana A, Groc L, Jimenez C, Bezard E, Dehay B. In utero delivery of rAAV2/9 induces neuronal expression of the transgene in the brain: towards new models of Parkinson’s disease. Gene Ther 2017; 24:801-809. [DOI: 10.1038/gt.2017.84] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 06/26/2017] [Accepted: 08/23/2017] [Indexed: 12/21/2022]
|
20
|
Obeso J, Stamelou M, Goetz C, Poewe W, Lang A, Weintraub D, Burn D, Halliday G, Bezard E, Przedborski S, Lehericy S, Brooks D, Rothwell J, Hallett M, DeLong M, Marras C, Tanner C, Ross G, Langston J, Klein C, Bonifati V, Jankovic J, Lozano A, Deuschl G, Bergman H, Tolosa E, Rodriguez-Violante M, Fahn S, Postuma R, Berg D, Marek K, Standaert D, Surmeier D, Olanow C, Kordower J, Calabresi P, Schapira A, Stoessl A. Past, present, and future of Parkinson's disease: A special essay on the 200th Anniversary of the Shaking Palsy. Mov Disord 2017; 32:1264-1310. [PMID: 28887905 PMCID: PMC5685546 DOI: 10.1002/mds.27115] [Citation(s) in RCA: 501] [Impact Index Per Article: 71.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 06/27/2017] [Indexed: 12/12/2022] Open
Abstract
This article reviews and summarizes 200 years of Parkinson's disease. It comprises a relevant history of Dr. James Parkinson's himself and what he described accurately and what he missed from today's perspective. Parkinson's disease today is understood as a multietiological condition with uncertain etiopathogenesis. Many advances have occurred regarding pathophysiology and symptomatic treatments, but critically important issues are still pending resolution. Among the latter, the need to modify disease progression is undoubtedly a priority. In sum, this multiple-author article, prepared to commemorate the bicentenary of the shaking palsy, provides a historical state-of-the-art account of what has been achieved, the current situation, and how to progress toward resolving Parkinson's disease. © 2017 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- J.A. Obeso
- HM CINAC, Hospital Universitario HM Puerta del Sur, Mostoles, Madrid, Spain
- Universidad CEU San Pablo, Madrid, Spain
- CIBERNED, Madrid, Spain
| | - M. Stamelou
- Department of Neurology, Philipps University, Marburg, Germany
- Parkinson’s Disease and Movement Disorders Department, HYGEIA Hospital and Attikon Hospital, University of Athens, Athens, Greece
| | - C.G. Goetz
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - W. Poewe
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - A.E. Lang
- Morton and Gloria Shulman Movement Disorders Clinic and the Edmond J Safra Program in Parkinson’s Disease, Toronto Western Hospital, Toronto, Canada
- Department of Medicine, University of Toronto, Toronto, Canada
| | - D. Weintraub
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Parkinson’s Disease and Mental Illness Research, Education and Clinical Centers (PADRECC and MIRECC), Corporal Michael J. Crescenz Veteran’s Affairs Medical Center, Philadelphia, Pennsylvania, USA
| | - D. Burn
- Medical Sciences, Newcastle University, Newcastle, UK
| | - G.M. Halliday
- Brain and Mind Centre, Sydney Medical School, The University of Sydney, Sydney, Australia
- School of Medical Sciences, University of New South Wales and Neuroscience Research Australia, Sydney, Australia
| | - E. Bezard
- Université de Bordeaux, Institut des Maladies Neurodégénératives, Centre National de la Recherche Scientifique Unité Mixte de Recherche 5293, Institut des Maladies Neurodégénératives, Bordeaux, France
- China Academy of Medical Sciences, Institute of Lab Animal Sciences, Beijing, China
| | - S. Przedborski
- Departments of Neurology, Pathology, and Cell Biology, the Center for Motor Neuron Biology and Disease, Columbia University, New York, New York, USA
- Columbia Translational Neuroscience Initiative, Columbia University, New York, New York, USA
| | - S. Lehericy
- Institut du Cerveau et de la Moelle épinière – ICM, Centre de NeuroImagerie de Recherche – CENIR, Sorbonne Universités, UPMC Univ Paris 06, Inserm U1127, CNRS UMR 7225, Paris, France
- Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - D.J. Brooks
- Clinical Sciences Department, Newcastle University, Newcastle, UK
- Department of Nuclear Medicine, Aarhus University, Aarhus, Denmark
| | - J.C. Rothwell
- Human Neurophysiology, Sobell Department, UCL Institute of Neurology, London, UK
| | - M. Hallett
- Human Motor Control Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - M.R. DeLong
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - C. Marras
- Morton and Gloria Shulman Movement Disorders Centre and the Edmond J Safra Program in Parkinson’s disease, Toronto Western Hospital, University of Toronto, Toronto, Canada
| | - C.M. Tanner
- Movement Disorders and Neuromodulation Center, Department of Neurology, University of California–San Francisco, San Francisco, California, USA
- Parkinson’s Disease Research, Education and Clinical Center, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA
| | - G.W. Ross
- Veterans Affairs Pacific Islands Health Care System, Honolulu, Hawaii, USA
| | | | - C. Klein
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
| | - V. Bonifati
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - J. Jankovic
- Parkinson’s Disease Center and Movement Disorders Clinic, Department of Neurology, Baylor College of Medicine, Houston, Texas, USA
| | - A.M. Lozano
- Department of Neurosurgery, Toronto Western Hospital, University of Toronto, Toronto, Canada
| | - G. Deuschl
- Department of Neurology, Universitätsklinikum Schleswig-Holstein, Christian Albrechts University Kiel, Kiel, Germany
| | - H. Bergman
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada, Jerusalem, Israel
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University, Jerusalem, Israel
- Department of Neurosurgery, Hadassah University Hospital, Jerusalem, Israel
| | - E. Tolosa
- Parkinson’s Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, Barcelona, Spain
- Department of Medicine, Universitat de Barcelona, IDIBAPS, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - M. Rodriguez-Violante
- Movement Disorders Clinic, Clinical Neurodegenerative Research Unit, Mexico City, Mexico
- Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | - S. Fahn
- Department of Neurology, Columbia University Medical Center, New York, New York, USA
| | - R.B. Postuma
- Department of Neurology, McGill University, Montreal General Hospital, Montreal, Quebec, Canada
| | - D. Berg
- Klinikfür Neurologie, UKSH, Campus Kiel, Christian-Albrechts-Universität, Kiel, Germany
| | - K. Marek
- Institute for Neurodegenerative Disorders, New Haven, Connecticut, USA
| | - D.G. Standaert
- Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - D.J. Surmeier
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - C.W. Olanow
- Departments of Neurology and Neuroscience, Mount Sinai School of Medicine, New York, New York, USA
| | - J.H. Kordower
- Research Center for Brain Repair, Rush University Medical Center, Chicago, Illinois, USA
- Neuroscience Graduate Program, Rush University Medical Center, Chicago, Illinois, USA
| | - P. Calabresi
- Neurological Clinic, Department of Medicine, Hospital Santa Maria della Misericordia, University of Perugia, Perugia, Italy
- Laboratory of Neurophysiology, Santa Lucia Foundation, IRCCS, Rome, Italy
| | - A.H.V. Schapira
- University Department of Clinical Neurosciences, UCL Institute of Neurology, University College London, London, UK
| | - A.J. Stoessl
- Pacific Parkinson’s Research Centre, Division of Neurology & Djavadf Mowafaghian Centre for Brain Health, University of British Columbia, British Columbia, Canada
- Vancouver Coastal Health, Vancouver, British Columbia, Canada
| |
Collapse
|
21
|
Tang T, Li Y, Jiao Q, Du X, Jiang H. Cerebral Dopamine Neurotrophic Factor: A Potential Therapeutic Agent for Parkinson's Disease. Neurosci Bull 2017; 33:568-575. [PMID: 28337696 DOI: 10.1007/s12264-017-0123-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 12/18/2016] [Indexed: 11/27/2022] Open
Abstract
The application of neurotrophic factors (NTFs) is a promising therapeutic strategy for neurodegenerative disorders such as Parkinson's disease (PD). Many NTFs have been reported to enhance the survival, regeneration, and differentiation of neurons and to induce synaptic plasticity. However, because of their potential side-effects and low efficacy after clinical administration, more potent treatments for neurodegenerative disorders are being sought. Cerebral dopamine neurotrophic factor (CDNF), a newly-identified NTF homologous to mesencephalic astrocyte-derived NTF, is structurally and functionally different from other NTFs, providing new hope especially for PD patients. In various animal models of PD, CDNF is efficient in protecting and repairing dopaminergic neurons, and it inhibits endoplasmic reticulum stress, neuroinflammation, and apoptosis. Recent progress in all facets of CDNF research has enabled researchers to better understand its beneficial effects in the treatment of PD.
Collapse
Affiliation(s)
- Tingting Tang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, 266071, China
| | - Yong Li
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, 266071, China
| | - Qian Jiao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, 266071, China
| | - Xixun Du
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, 266071, China
| | - Hong Jiang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
22
|
Pignataro D, Sucunza D, Vanrell L, Lopez-Franco E, Dopeso-Reyes IG, Vales A, Hommel M, Rico AJ, Lanciego JL, Gonzalez-Aseguinolaza G. Adeno-Associated Viral Vectors Serotype 8 for Cell-Specific Delivery of Therapeutic Genes in the Central Nervous System. Front Neuroanat 2017; 11:2. [PMID: 28239341 PMCID: PMC5301009 DOI: 10.3389/fnana.2017.00002] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 01/13/2017] [Indexed: 12/19/2022] Open
Abstract
Adeno-associated viruses (AAVs) have become highly promising tools for research and clinical applications in the central nervous system (CNS). However, specific delivery of genes to the cell type of interest is essential for the success of gene therapy and therefore a correct selection of the promoter plays a very important role. Here, AAV8 vectors carrying enhanced green fluorescent protein (eGFP) as reporter gene under the transcriptional control of different CNS-specific promoters were used and compared with a strong ubiquitous promoter. Since one of the main limitations of AAV-mediated gene delivery lies in its restricted cloning capacity, we focused our work on small-sized promoters. We tested the transduction efficacy and specificity of each vector after stereotactic injection into the mouse striatum. Three glia-specific AAV vectors were generated using two truncated forms of the human promoter for glial fibrillar acidic protein (GFAP) as well as a truncated form of the murine GFAP promoter. All three vectors resulted in predominantly glial expression; however we also observed eGFP expression in other cell-types such as oligodendrocytes, but never in neurons. In addition, robust and neuron-specific eGFP expression was observed using the minimal promoters for the neural protein BM88 and the neuronal nicotinic receptor β2 (CHRNB2). In summary, we developed a set of AAV vectors designed for specific expression in cells of the CNS using minimal promoters to drive gene expression when the size of the therapeutic gene matters.
Collapse
Affiliation(s)
- Diego Pignataro
- Department of Gene Therapy and Regulation of Gene Expression, Center for Applied Medical ResearchPamplona, Spain; Department of Neurosciences, Center for Applied Medical ResearchPamplona, Spain
| | - Diego Sucunza
- Department of Gene Therapy and Regulation of Gene Expression, Center for Applied Medical ResearchPamplona, Spain; Department of Neurosciences, Center for Applied Medical ResearchPamplona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades NeurodegenerativasSpain
| | - Lucia Vanrell
- Department of Gene Therapy and Regulation of Gene Expression, Center for Applied Medical Research Pamplona, Spain
| | | | - Iria G Dopeso-Reyes
- Department of Neurosciences, Center for Applied Medical ResearchPamplona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades NeurodegenerativasSpain; Instituto de Investigación Sanitaria de NavarraPamplona, Spain
| | - Africa Vales
- Department of Gene Therapy and Regulation of Gene Expression, Center for Applied Medical Research Pamplona, Spain
| | - Mirja Hommel
- Department of Gene Therapy and Regulation of Gene Expression, Center for Applied Medical ResearchPamplona, Spain; Instituto de Investigación Sanitaria de NavarraPamplona, Spain
| | - Alberto J Rico
- Department of Neurosciences, Center for Applied Medical ResearchPamplona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades NeurodegenerativasSpain; Instituto de Investigación Sanitaria de NavarraPamplona, Spain
| | - Jose L Lanciego
- Department of Neurosciences, Center for Applied Medical ResearchPamplona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades NeurodegenerativasSpain; Instituto de Investigación Sanitaria de NavarraPamplona, Spain
| | - Gloria Gonzalez-Aseguinolaza
- Department of Gene Therapy and Regulation of Gene Expression, Center for Applied Medical ResearchPamplona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades NeurodegenerativasSpain; Instituto de Investigación Sanitaria de NavarraPamplona, Spain
| |
Collapse
|
23
|
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
|
24
|
Clinical tests of neurotrophic factors for human neurodegenerative diseases, part 2: Where do we stand and where must we go next? Neurobiol Dis 2017; 97:169-178. [DOI: 10.1016/j.nbd.2016.03.026] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 03/30/2016] [Indexed: 12/13/2022] Open
|
25
|
Intranasal Cerebrolysin Attenuates Learning and Memory Impairments in D-galactose-Induced Senescence in Mice. Exp Gerontol 2016; 87:16-22. [PMID: 27894939 DOI: 10.1016/j.exger.2016.11.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 11/11/2016] [Accepted: 11/21/2016] [Indexed: 11/20/2022]
Abstract
Neurotrophic factors are currently being considered as pro-cognitive therapeutic approaches for management of cognitive deficits. This study aims to evaluate the effects of intranasal (i.n.) or intraperitoneal (i.p.) administration of Cerebrolysin (CBL) (as a mixture of neurotrophic factors) on the d-galactose-induced oxidative stress, apoptosis and memory as well as learning impairment in mice. For this purpose, CBL (1, 2.5, 5 ml/kg/i.p.) or (1 ml/kg/i.n.), were administrated daily in d-galactose-received (100 mg/kg/subcutaneous (s.c.)) mice model of aging for eight weeks. Spatial and recognition memories were assessed by the Morris water maze and novel object recognition tasks. Brain and blood of animals were analysed for oxidative stress biomarkers including malondialdehyde, total antioxidant capacity, glutathione peroxidase and superoxide dismutase. Apoptosis rate in the hippocampus was evaluated by TUNEL staining of brain tissue. 5 ml/kg/i.p. dose of CBL increased the locomotor activity but, 1 ml/kg/i.p. dose didn't show detectable behavioural or molecular effects on aged mice. Treatment with 2.5 ml/kg/i.p. and 1 ml/kg/i.n. doses attenuated d-galactose-impaired spatial and recognition memories. Results showed an obvious increase in the antioxidant biomarkers and decrease in the malondialdehyde levels both in the blood and brain of aged mice in 2.5 ml/kg/i.p. dose, and only in the brain in 1 ml/kg/i.n. dose of CBL. Anti-apoptotic effects also were seen in the same dose/rout of CBL administration in aged animals. This study proves the usefulness of i.n. CBL administration as a non-invasive and efficient method of drug delivery to the brain to improve aging-induced oxidative stress, apoptosis and learning as well as memory impairment.
Collapse
|
26
|
Olanow CW, Bartus RT, Volpicelli-Daley LA, Kordower JH. Trophic factors for Parkinson's disease: To live or let die. Mov Disord 2016; 30:1715-24. [PMID: 26769457 DOI: 10.1002/mds.26426] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 08/06/2015] [Accepted: 08/19/2015] [Indexed: 12/23/2022] Open
Abstract
Trophic factors show great promise in laboratory studies as potential therapies for PD. However, multiple double-blind, clinical trials have failed to show benefits in comparison to a placebo control. This article will review the scientific rationale for testing trophic factors in PD, the results of the different clinical trials that have been performed to date, and the possible explanations for these failed outcomes. We will also consider future directions and the likelihood that trophic factors will become a viable therapy for patients with PD.
Collapse
Affiliation(s)
- C Warren Olanow
- Department of Neurology, Mount Sinai School of Medicine, New York, New York, USA
| | | | | | | |
Collapse
|
27
|
Abstract
The dream that trophic factors could be effectively delivered and potently forestall and reverse the symptoms of Parkinson's disease (PD) has yet to be realized. Research in this area has been active for 20 years, but after much work, the prospects for utilizing trophic factors in the treatment of PD are currently dim. Millions of dollars have been spent, numerous academic, foundation, and government resources have been invested, and hundreds of patient research volunteers have contributed their time and hope to this effort without a therapeutic breakthrough. As a scientist who has journeyed these events from the beginning and participated in many of the decisions that navigated this field, I consider it important for the movement disorder scientific community to reflect on the evolution of thought and to participate in the dialog over whether the investments were worthwhile.The most studied group of trophic factor for PD is the glial cell derived family of ligands, of which glial cell derived neurotrophic factor (GDNF) and neurturin are members, and are the best studied. I trace the development of these factors chronologically with commentary on the key decision-making points. Before we collectively invest further, I offer this scientific reflection on the past and offer my own view on the next steps of research in the field of neurotrophins as potential therapeutic agents in PD.
Collapse
|
28
|
Runeberg-Roos P, Piccinini E, Penttinen AM, Mätlik K, Heikkinen H, Kuure S, Bespalov MM, Peränen J, Garea-Rodríguez E, Fuchs E, Airavaara M, Kalkkinen N, Penn R, Saarma M. Developing therapeutically more efficient Neurturin variants for treatment of Parkinson's disease. Neurobiol Dis 2016; 96:335-345. [PMID: 27425888 DOI: 10.1016/j.nbd.2016.07.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/04/2016] [Accepted: 07/13/2016] [Indexed: 10/21/2022] Open
Abstract
In Parkinson's disease midbrain dopaminergic neurons degenerate and die. Oral medications and deep brain stimulation can relieve the initial symptoms, but the disease continues to progress. Growth factors that might support the survival, enhance the activity, or even regenerate degenerating dopamine neurons have been tried with mixed results in patients. As growth factors do not pass the blood-brain barrier, they have to be delivered intracranially. Therefore their efficient diffusion in brain tissue is of crucial importance. To improve the diffusion of the growth factor neurturin (NRTN), we modified its capacity to attach to heparan sulfates in the extracellular matrix. We present four new, biologically fully active variants with reduced heparin binding. Two of these variants are more stable than WT NRTN in vitro and diffuse better in rat brains. We also show that one of the NRTN variants diffuses better than its close homolog GDNF in monkey brains. The variant with the highest stability and widest diffusion regenerates dopamine fibers and improves the conditions of rats in a 6-hydroxydopamine model of Parkinson's disease more potently than GDNF, which previously showed modest efficacy in clinical trials. The new NRTN variants may help solve the major problem of inadequate distribution of NRTN in human brain tissue.
Collapse
Affiliation(s)
- Pia Runeberg-Roos
- Institute of Biotechnology, University of Helsinki, PB 56 (Viikinkaari 5D), FIN-00014, Finland.
| | - Elisa Piccinini
- Institute of Biotechnology, University of Helsinki, PB 56 (Viikinkaari 5D), FIN-00014, Finland
| | - Anna-Maija Penttinen
- Institute of Biotechnology, University of Helsinki, PB 56 (Viikinkaari 5D), FIN-00014, Finland
| | - Kert Mätlik
- Institute of Biotechnology, University of Helsinki, PB 56 (Viikinkaari 5D), FIN-00014, Finland
| | - Hanna Heikkinen
- Institute of Biotechnology, University of Helsinki, PB 56 (Viikinkaari 5D), FIN-00014, Finland
| | - Satu Kuure
- Institute of Biotechnology, University of Helsinki, PB 56 (Viikinkaari 5D), FIN-00014, Finland
| | - Maxim M Bespalov
- Institute of Biotechnology, University of Helsinki, PB 56 (Viikinkaari 5D), FIN-00014, Finland
| | - Johan Peränen
- Institute of Biotechnology, University of Helsinki, PB 56 (Viikinkaari 5D), FIN-00014, Finland
| | - Enrique Garea-Rodríguez
- Department of Neuroanatomy, Institute for Anatomy and Cell Biology, University of Freiburg, Freiburg, Germany
| | | | - Mikko Airavaara
- Institute of Biotechnology, University of Helsinki, PB 56 (Viikinkaari 5D), FIN-00014, Finland
| | - Nisse Kalkkinen
- Institute of Biotechnology, University of Helsinki, PB 56 (Viikinkaari 5D), FIN-00014, Finland
| | - Richard Penn
- CNS Therapeutics Inc., 332 Minnesota Street, Ste W1750, St. Paul, MN 55101, USA
| | - Mart Saarma
- Institute of Biotechnology, University of Helsinki, PB 56 (Viikinkaari 5D), FIN-00014, Finland
| |
Collapse
|
29
|
Peng S, Ma Y, Flores J, Cornfeldt M, Mitrovic B, Eidelberg D, Doudet DJ. Modulation of Abnormal Metabolic Brain Networks by Experimental Therapies in a Nonhuman Primate Model of Parkinson Disease: An Application to Human Retinal Pigment Epithelial Cell Implantation. J Nucl Med 2016; 57:1591-1598. [PMID: 27056614 DOI: 10.2967/jnumed.115.161513] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 03/07/2016] [Indexed: 01/30/2023] Open
Abstract
Abnormal covariance pattern of regional metabolism associated with Parkinson disease (PD) is modulated by dopaminergic pharmacotherapy. Using high-resolution 18F-FDG PET and network analysis, we previously derived and validated a parkinsonism-related metabolic pattern (PRP) in nonhuman primate models of PD. It is currently not known whether this network is modulated by experimental therapeutics. In this study, we examined changes in network activity by striatal implantation of human levodopa-producing retinal pigment epithelial (hRPE) cells in parkinsonian macaques and evaluated the reproducibility of network activity in a small test-retest study. METHODS 18F-FDG PET scans were acquired in 8 healthy macaques and 8 macaques with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced bilateral nigrostriatal dopaminergic lesions after unilateral putaminal implantation of hRPE cells or sham surgery. PRP activity was measured prospectively in all animals and in a subset of test-retest animals using a network quantification approach. Network activity and regional metabolic values were compared on a hemispheric basis between animal groups and treatment conditions. RESULTS All individual macaques showed clinical improvement after hRPE cell implantation compared with the sham surgery. PRP activity was elevated in the untreated MPTP hemispheres relative to those of the normal controls (P < 0.00005) but was reduced (P < 0.05) in the hRPE-implanted hemispheres. The modulation observed in network activity was supported by concurrent local and remote changes in regional glucose metabolism. PRP activity remained unchanged in the untreated MPTP hemispheres versus the sham-operated hemispheres. PRP activity was also stable (P ≥ 0.29) and correlated (R2 ≥ 0.926; P < 0.00005) in the test-retest hemispheres. These findings were highly reproducible across several PRP topographies generated in multiple cohorts of parkinsonian and healthy macaques. CONCLUSION We have demonstrated long-term therapeutic effects of hRPE cell implantation in nonhuman primate models of PD. The implantation of such levodopa-producing cells can concurrently decrease the elevated metabolic network activity in parkinsonian brains on an individual basis. These results parallel the analogous findings reported in patients with PD undergoing levodopa therapy and other symptomatic interventions. With further validation in large samples, 18F-FDG PET imaging with network analysis may provide a viable biomarker for assessing treatment response in animal models of PD after experimental therapies.
Collapse
Affiliation(s)
- Shichun Peng
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, New York, New York
| | - Yilong Ma
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, New York, New York
| | - Joseph Flores
- Department of Neurology, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | - David Eidelberg
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, New York, New York
| | - Doris J Doudet
- Department of Neurology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
30
|
Bartus RT, Johnson EM. Clinical tests of neurotrophic factors for human neurodegenerative diseases, part 1: Where have we been and what have we learned? Neurobiol Dis 2016; 97:156-168. [PMID: 27063798 DOI: 10.1016/j.nbd.2016.03.027] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 03/24/2016] [Accepted: 03/30/2016] [Indexed: 02/08/2023] Open
Abstract
Over the past 25years, about 3 dozen clinical reports have been published regarding the safety and possible efficacy of neurotrophic factors in patients with various neurodegenerative diseases. This effort involved a half dozen different neurotrophic factors, using at least 5 different general delivery approaches for ALS (amyolateral sclerosis), peripheral neuropathies, PD (Parkinson's disease) and AD (Alzheimer's disease). While none of these efforts have yet produced efficacy data sufficiently robust or reliable to establish neurotrophic factors as treatments for any human disease, the obstacles encountered and novel information reported, when viewed collectively, provide important insight to help future efforts. Three consistent themes emerge from these publications: (1) unexpected and undesirable side effects, at times serious, have plagued many efforts to deliver neurotrophic factors to humans; (2) the magnitude and consistency of clinical benefit has been disappointing; (3) by far that most consistently proposed reason for the side effects and poor efficacy has been inadequate dosing and delivery. This paper reviews and attempts to synthesize the available data derived from clinical tests of neurotrophic factors for neurodegenerative diseases. The obstacles encountered, the solutions attempted, and the lessons learned are discussed. The vast majority of solutions have involved changes in dosing paradigms and dose levels, which has primarily led to improved safety outcomes. However, lack of adequate efficacy remains a significant issue. While current efforts continue to focus exclusively on still-further changes in dosing parameters, a review of available data argues that it may now be the time to ask whether other, non-dose-related variables should be given more serious consideration as being responsible for the great divide that exists between the robust effects seen in animal models and the relatively weak effects seen in human neurodegenerative patients. Foremost among these appears to be the severe degeneration seen in the majority of patients enrolled in past and current trials testing neurotrophic factors in humans. A companion paper (Bartus and Johnson, 2016), reviews the contemporary data and concludes that compelling empirical evidence already exists for enrolling earlier-stage subjects as likely essential to achieving more robust and reliable benefit.
Collapse
Affiliation(s)
| | - Eugene M Johnson
- Departments of Neurology and Developmental Biology, Washington University Medical School, St. Louis, MO, USA
| |
Collapse
|
31
|
A regulatable AAV vector mediating GDNF biological effects at clinically-approved sub-antimicrobial doxycycline doses. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 5:16027. [PMID: 27069954 PMCID: PMC4813607 DOI: 10.1038/mtm.2016.27] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 02/26/2016] [Accepted: 02/26/2016] [Indexed: 12/19/2022]
Abstract
Preclinical and clinical data stress the importance of pharmacologically-controlling glial cell line-derived neurotrophic factor (GDNF) intracerebral administration to treat PD. The main challenge is finding a combination of a genetic switch and a drug which, when administered at a clinically-approved dose, reaches the brain in sufficient amounts to induce a therapeutic effect. We describe a highly-sensitive doxycycline-inducible adeno-associated virus (AAV) vector. This vector allowed for the first time a longitudinal analysis of inducible transgene expression in the brain using bioluminescence imaging. To evaluate the dose range of GDNF biological activity, the inducible AAV vector (8.0 × 10(9) viral genomes) was injected in the rat striatum at four delivery sites and increasing doxycycline doses administered orally. ERK/Akt signaling activation as well as tyrosine hydroxylase downregulation, a consequence of long-term GDNF treatment, were induced at plasmatic doxycycline concentrations of 140 and 320 ng/ml respectively, which are known not to increase antibiotic-resistant microorganisms in patients. In these conditions, GDNF covered the majority of the striatum. No behavioral abnormalities or weight loss were observed. Motor asymmetry resulting from unilateral GDNF treatment only appeared with a 2.5-fold higher vector and a 13-fold higher inducer doses. Our data suggest that using the herein-described inducible AAV vector, biological effects of GDNF can be obtained in response to sub-antimicrobial doxycycline doses.
Collapse
|
32
|
Volta M, Milnerwood AJ, Farrer MJ. Insights from late-onset familial parkinsonism on the pathogenesis of idiopathic Parkinson's disease. Lancet Neurol 2015; 14:1054-64. [PMID: 26376970 DOI: 10.1016/s1474-4422(15)00186-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 07/17/2015] [Accepted: 07/20/2015] [Indexed: 01/24/2023]
Abstract
Disease-modifying therapies that slow or halt the progression of Parkinson's disease are an unmet clinical need. Many hypotheses have been put forward to explain the pathogenesis of the disease, but none has led to the development of disease-modifying drugs. Here we focus on familial forms of late-onset parkinsonism that most closely resemble idiopathic Parkinson's disease and present a synthesis of emerging molecular advances. Genetic discoveries and mechanistic investigations have highlighted early alterations to synaptic function, endosomal maturation, and protein sorting that might lead to an intracellular proteinopathy. We propose that these cellular processes constitute one pathway to pathogenesis and suggest that neuroprotection, as an adjunct to current symptomatic treatments, need not remain an elusive goal.
Collapse
Affiliation(s)
- Mattia Volta
- Department of Medical Genetics, Centre for Applied Neurogenetics, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Austen J Milnerwood
- Division of Neurology, Centre for Applied Neurogenetics, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Matthew J Farrer
- Department of Medical Genetics, Centre for Applied Neurogenetics, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
33
|
Bartus RT. Gene therapy for Parkinson's disease: a decade of progress supported by posthumous contributions from volunteer subjects. Neural Regen Res 2015; 10:1586-8. [PMID: 26692850 PMCID: PMC4660746 DOI: 10.4103/1673-5374.167783] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Raymond T Bartus
- RTBioconsultants, Inc., San Diego, CA, USA; formerly, EVP and CSO, Ceregene, Inc., San Diego, CA, USA
| |
Collapse
|
34
|
Chen H. Adeno-associated virus vectors for human gene therapy. World J Med Genet 2015; 5:28-45. [DOI: 10.5496/wjmg.v5.i3.28] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 04/08/2015] [Accepted: 05/18/2015] [Indexed: 02/06/2023] Open
Abstract
Adeno-associated virus (AAV) is a small, non-enveloped virus that contains a single-stranded DNA genome. It was the first gene therapy drug approved in the Western world in November 2012 to treat patients with lipoprotein lipase deficiency. AAV made history and put human gene therapy in the forefront again. More than four decades of research on AAV vector biology and human gene therapy has generated a huge amount of valuable information. Over 100 AAV serotypes and variants have been isolated and at least partially characterized. A number of them have been used for preclinical studies in a variety of animal models. Several AAV vector production platforms, especially the baculovirus-based system have been established for commercial-scale AAV vector production. AAV purification technologies such as density gradient centrifugation, column chromatography, or a combination, have been well developed. More than 117 clinical trials have been conducted with AAV vectors. Although there are still challenges down the road, such as cross-species variation in vector tissue tropism and gene transfer efficiency, pre-existing humoral immunity to AAV capsids and vector dose-dependent toxicity in patients, the gene therapy community is forging ahead with cautious optimism. In this review I will focus on the properties and applications of commonly used AAV serotypes and variants, and the technologies for AAV vector production and purification. I will also discuss the advancement of several promising gene therapy clinical trials.
Collapse
|
35
|
Domanskyi A, Saarma M, Airavaara M. Prospects of Neurotrophic Factors for Parkinson's Disease: Comparison of Protein and Gene Therapy. Hum Gene Ther 2015; 26:550-9. [DOI: 10.1089/hum.2015.065] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Andrii Domanskyi
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Mikko Airavaara
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
36
|
Janelidze S, Nordström U, Kügler S, Brundin P. Pre-existing immunity to adeno-associated virus (AAV)2 limits transgene expression following intracerebral AAV2-based gene delivery in a 6-hydroxydopamine model of Parkinson's disease. J Gene Med 2015; 16:300-8. [PMID: 25303717 DOI: 10.1002/jgm.2779] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 07/31/2014] [Accepted: 08/12/2014] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Adeno-associated virus (AAV) vectors are used to deliver potentially therapeutic genes in clinical trials in Parkinson's disease (PD). Pre-existing immunity to AAV and a local neuroinflammatory response might negatively affect the efficacy of such AAV-mediated gene delivery. METHODS We pre-immunized rats with wild-type AAV-2. Three months later, we created PD-like lesions by intrastriatal injections of 6-hydroxydopamine (6-OHDA) in 50% of the animals. One month later, we injected AAV2 vector expressing enhanced green fluorescent protein (eGFP) in the striatum. Using immunohistochemistry, we assessed eGFP expression, microglia activation and CD8 T cell infiltration. We also measured AAV-2 specific neutralizing antibody titers in the serum. RESULTS The number of striatal cells transduced with AAV2 vector expressing eGFP was reduced by 71% in rats pre-immunized with wild-type AAV2 compared to non-immunized animals. We detected elevated numbers of OX6(+) activated microglia in the striatum and circulating AAV2-specific neutralizing antibodies in pre-immunized rats. We also observed that the intrastriatal 6-OHDA injection promoted CD8(+) T cell infiltration and enhanced microglia activation. Nevertheless, the 6-OHDA lesion did not alter AAV2-mediated expression of eGFP in either pre-immunized or non-immunized rats. CONCLUSIONS Our findings indicate that intracerebral AAV2-based gene therapy is compromised in rats with pre-existing immunity to AAV2. By contrast, a local neuroinflammatory response, caused by intrastriatal a 6-OHDA injection, does not affect viral vector-mediated transgene expression. Our results emphasize the importance of monitoring circulating AAV-specific neutralizing antibodies in patients undergoing intracerebral gene therapy using AAV vectors.
Collapse
Affiliation(s)
- Shorena Janelidze
- Neuronal Survival Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | | | | |
Collapse
|
37
|
O'Connor DM, Boulis NM. Gene therapy for neurodegenerative diseases. Trends Mol Med 2015; 21:504-12. [PMID: 26122838 DOI: 10.1016/j.molmed.2015.06.001] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 06/02/2015] [Accepted: 06/03/2015] [Indexed: 12/18/2022]
Abstract
Gene therapy is, potentially, a powerful tool for treating neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS), spinal muscular atrophy, Parkinson's disease (PD) and Alzheimer's disease (AD). To date, clinical trials have failed to show any improvement in outcome beyond the placebo effect. Efforts to improve outcomes are focusing on three main areas: vector design and the identification of new vector serotypes, mode of delivery of gene therapies, and identification of new therapeutic targets. These advances are being tested both individually and together to improve efficacy. These improvements may finally make gene therapy successful for these disorders.
Collapse
Affiliation(s)
- Deirdre M O'Connor
- Department of Neurosurgery, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Nicholas M Boulis
- Department of Neurosurgery, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA.
| |
Collapse
|
38
|
Gurevich EV, Gurevich VV. Beyond traditional pharmacology: new tools and approaches. Br J Pharmacol 2015; 172:3229-41. [PMID: 25572005 DOI: 10.1111/bph.13066] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 11/24/2014] [Accepted: 01/02/2015] [Indexed: 12/14/2022] Open
Abstract
Traditional pharmacology is defined as the science that deals with drugs and their actions. While small molecule drugs have clear advantages, there are many cases where they have proved to be ineffective, prone to unacceptable side effects, or where due to a particular disease aetiology they cannot possibly be effective. A dominant feature of the small molecule drugs is their single mindedness: they provide either continuous inhibition or continuous activation of the target. Because of that, these drugs tend to engage compensatory mechanisms leading to drug tolerance, drug resistance or, in some cases, sensitization and consequent loss of therapeutic efficacy over time and/or unwanted side effects. Here we discuss new and emerging therapeutic tools and approaches that have potential for treating the majority of disorders for which small molecules are either failing or cannot be developed. These new tools include biologics, such as recombinant hormones and antibodies, as well as approaches involving gene transfer (gene therapy and genome editing) and the introduction of specially designed self-replicating cells. It is clear that no single method is going to be a 'silver bullet', but collectively, these novel approaches hold promise for curing practically every disorder.
Collapse
Affiliation(s)
- E V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - V V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
39
|
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
|
40
|
|
41
|
Bartus RT, Kordower JH, Johnson EM, Brown L, Kruegel BR, Chu Y, Baumann TL, Lang AE, Olanow CW, Herzog CD. Post-mortem assessment of the short and long-term effects of the trophic factor neurturin in patients with α-synucleinopathies. Neurobiol Dis 2015; 78:162-71. [PMID: 25841760 DOI: 10.1016/j.nbd.2015.03.023] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 03/13/2015] [Accepted: 03/23/2015] [Indexed: 11/29/2022] Open
Abstract
Substantial interest persists for developing neurotrophic factors to treat neurodegenerative diseases. At the same time, significant progress has been made in implementing gene therapy as a means to provide long-term expression of bioactive neurotrophic factors to targeted sites in the brain. Nonetheless, to date, no double-blind clinical trial has achieved positive results on its primary endpoint despite robust benefits achieved in animal models. A major issue with advancing the field is the paucity of information regarding the expression and effects of neurotrophic factors in human neurodegenerative brain, relative to the well-characterized responses in animal models. To help fill this information void, we examined post-mortem brain tissue from four patients with nigrostriatal degeneration who had participated in clinical trials testing gene delivery of neurturin to the putamen of patients. Each had died of unrelated causes ranging from 1.5-to-3-months (2 Parkinson's disease patients), to 4+-years (1 Parkinson's disease and 1 multiple-system atrophy-parkinsonian type patient) following gene therapy. Quantitative and immunohistochemical evaluation of neurturin, alpha-synuclein, tyrosine hydroxylase (TH) and an oligodendroglia marker (Olig 2) were performed in each brain. Comparable volumes-of-expression of neurturin were seen in the putamen in all cases (~15-22%; mean=18.5%). TH-signal in the putamen was extremely sparse in the shorter-term cases. A 6-fold increase was seen in longer-term cases, but was far less than achieved in animal models of nigrostriatal degeneration with similar or even far less NRTN exposure. Less than 1% of substantia nigra (SN) neurons stained for neurturin in the shorter-term cases. A 15-fold increase was seen in the longer-term cases, but neurturin was still only detected in ~5% of nigral cells. These data provide unique insight into the functional status of advanced, chronic nigrostriatal degeneration in human brain and the response of these neurons to neurotrophic factor stimulation. They demonstrate mild but persistent expression of gene-mediated neurturin over 4-years, with an apparent, time-related amplification of its transport and biological effects, albeit quite weak, and provide unique information to help plan and design future trials.
Collapse
Affiliation(s)
- R T Bartus
- RTBioconsultants, Inc., San Diego, CA, USA; Ceregene, Inc, USA.
| | - J H Kordower
- Rush Presbyterian Medical Center, Chicago, IL, USA
| | - E M Johnson
- Washington University Medical School, St. Louis, MO, USA
| | | | | | - Y Chu
- Rush Presbyterian Medical Center, Chicago, IL, USA
| | - T L Baumann
- Isis Pharmaceuticals, Carlsbad, CA, USA; Ceregene, Inc, USA
| | - A E Lang
- Toronto Western Hospital, Toronto, Canada
| | - C W Olanow
- Mount Sinai School of Medicine, NYC, USA
| | | |
Collapse
|
42
|
Abstract
Adeno-associated virus (AAV) is a small, nonenveloped virus that was adapted 30 years ago for use as a gene transfer vehicle. It is capable of transducing a wide range of species and tissues in vivo with no evidence of toxicity, and it generates relatively mild innate and adaptive immune responses. We review the basic biology of AAV, the history of progress in AAV vector technology, and some of the clinical and research applications where AAV has shown success.
Collapse
Affiliation(s)
- R. Jude Samulski
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Nicholas Muzyczka
- Powell Gene Therapy Center, College of Medicine, University of Florida, Gainesville, Florida 32610
| |
Collapse
|
43
|
Dong Y, Eltoukhy AA, Alabi CA, Khan OF, Veiseh O, Dorkin JR, Sirirungruang S, Yin H, Tang BC, Pelet JM, Chen D, Gu Z, Xue Y, Langer R, Anderson DG. Lipid-like nanomaterials for simultaneous gene expression and silencing in vivo. Adv Healthc Mater 2014; 3:1392-7. [PMID: 24623658 DOI: 10.1002/adhm.201400054] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Indexed: 01/20/2023]
Abstract
New lipid-like nanomaterials are developed to simultaneously regulate expression of multiple genes. Self-assembled nanoparticles are capable of efficiently encapsulating pDNA and siRNA. These nanoparticles are shown to induce simultaneous gene expression and silencing both in vitro and in vivo.
Collapse
Affiliation(s)
- Yizhou Dong
- David H. Koch Institute for Integrative Cancer Research; Massachusetts Institute of Technology; Cambridge MA 02139 USA
- Department of Chemical Engineering; Massachusetts Institute of Technology; Cambridge MA 02139 USA
- Department of Anesthesiology; Children's Hospital Boston; 300 Longwood Avenue Boston MA 02115 USA
| | - Ahmed A. Eltoukhy
- David H. Koch Institute for Integrative Cancer Research; Massachusetts Institute of Technology; Cambridge MA 02139 USA
| | - Christopher A. Alabi
- David H. Koch Institute for Integrative Cancer Research; Massachusetts Institute of Technology; Cambridge MA 02139 USA
- Department of Chemical Engineering; Massachusetts Institute of Technology; Cambridge MA 02139 USA
| | - Omar F. Khan
- David H. Koch Institute for Integrative Cancer Research; Massachusetts Institute of Technology; Cambridge MA 02139 USA
- Department of Chemical Engineering; Massachusetts Institute of Technology; Cambridge MA 02139 USA
| | - Omid Veiseh
- David H. Koch Institute for Integrative Cancer Research; Massachusetts Institute of Technology; Cambridge MA 02139 USA
- Department of Chemical Engineering; Massachusetts Institute of Technology; Cambridge MA 02139 USA
- Department of Anesthesiology; Children's Hospital Boston; 300 Longwood Avenue Boston MA 02115 USA
| | - J. Robert Dorkin
- Department of Biology; Massachusetts Institute of Technology; Cambridge MA 02139 USA
| | | | - Hao Yin
- David H. Koch Institute for Integrative Cancer Research; Massachusetts Institute of Technology; Cambridge MA 02139 USA
| | - Benjamin C. Tang
- David H. Koch Institute for Integrative Cancer Research; Massachusetts Institute of Technology; Cambridge MA 02139 USA
- Department of Anesthesiology; Children's Hospital Boston; 300 Longwood Avenue Boston MA 02115 USA
| | - Jeisa M. Pelet
- David H. Koch Institute for Integrative Cancer Research; Massachusetts Institute of Technology; Cambridge MA 02139 USA
| | - Delai Chen
- David H. Koch Institute for Integrative Cancer Research; Massachusetts Institute of Technology; Cambridge MA 02139 USA
- Department of Anesthesiology; Children's Hospital Boston; 300 Longwood Avenue Boston MA 02115 USA
| | - Zhen Gu
- David H. Koch Institute for Integrative Cancer Research; Massachusetts Institute of Technology; Cambridge MA 02139 USA
- Department of Chemical Engineering; Massachusetts Institute of Technology; Cambridge MA 02139 USA
- Department of Anesthesiology; Children's Hospital Boston; 300 Longwood Avenue Boston MA 02115 USA
| | - Yuan Xue
- David H. Koch Institute for Integrative Cancer Research; Massachusetts Institute of Technology; Cambridge MA 02139 USA
| | - Robert Langer
- Department of Chemical Engineering; Massachusetts Institute of Technology; Cambridge MA 02139 USA
- Harvard-MIT Division of Health Science Technology and Institute for Medical Engineering and Science; Massachusetts Institute of Technology; Cambridge MA 02139 USA
| | - Daniel G. Anderson
- David H. Koch Institute for Integrative Cancer Research; Massachusetts Institute of Technology; Cambridge MA 02139 USA
- Department of Chemical Engineering; Massachusetts Institute of Technology; Cambridge MA 02139 USA
- Harvard-MIT Division of Health Science Technology and Institute for Medical Engineering and Science; Massachusetts Institute of Technology; Cambridge MA 02139 USA
| |
Collapse
|
44
|
Bioactivity-guided fractionation identifies amygdalin as a potent neurotrophic agent from herbal medicine Semen Persicae extract. BIOMED RESEARCH INTERNATIONAL 2014; 2014:306857. [PMID: 25050339 PMCID: PMC4094722 DOI: 10.1155/2014/306857] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Revised: 06/03/2014] [Accepted: 06/03/2014] [Indexed: 01/21/2023]
Abstract
Herbal medicine Semen Persicae is widely used to treat blood stasis in Chinese medicine and other oriental folk medicines. Although little is known about the effects of Semen Persicae and its active compounds on neuron differentiation, our pilot study showed that Semen Persicae extract promoted neurite outgrowth in rat dopaminergic PC12 cells. In the present study, we developed a bioactivity-guided fractionation procedure for the characterization of the neurotrophic activity of Semen Persicae extract. The resultant fractions were assayed for neurite outgrowth in PC12 cells based on microscopic assessment. Through liquid-liquid extraction and reverse phase HPLC separation, a botanical glycoside amygdalin was isolated as the active compound responsible for the neurotrophic activity of Semen Persicae extract. Moreover, we found that amygdalin rapidly induced the activation of extracellular-signal-regulated kinase 1/2 (ERK1/2). A specific ERK1/2 inhibitor PD98059 attenuated the stimulatory effect of amygdalin on neurite outgrowth. Taken together, amygdalin was identified as a potent neurotrophic agent from Semen Persicae extract through a bioactivity-guided fractional procedure. The neurotrophic activity of amygdalin may be mediated by the activation of ERK1/2 pathway.
Collapse
|
45
|
Bourdenx M, Dutheil N, Bezard E, Dehay B. Systemic gene delivery to the central nervous system using Adeno-associated virus. Front Mol Neurosci 2014; 7:50. [PMID: 24917785 PMCID: PMC4040820 DOI: 10.3389/fnmol.2014.00050] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 05/14/2014] [Indexed: 12/12/2022] Open
Abstract
Adeno-associated virus (AAV)-mediated gene delivery has emerged as an effective and safe tool for both preclinical and clinical studies of neurological disorders. The recent discovery that several serotypes are able to cross the blood–brain barrier when administered systemically has been a real breakthrough in the field of neurodegenerative diseases. Widespread transgene expression after systemic injection could spark interest as a therapeutic approach. Such strategy will avoid invasive brain surgery and allow non-focal gene therapy promising for CNS diseases affecting large portion of the brain. Here, we will review the recent results achieved through different systemic routes of injection generated in the last decade using systemic AAV-mediated delivery and propose a brief assessment of their values. In particular, we emphasize how the methods used for virus engineering could improve brain transduction after peripheral delivery.
Collapse
Affiliation(s)
- Mathieu Bourdenx
- Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux Bordeaux, France ; CNRS, Institut des Maladies Neurodégénératives, UMR 5293 Bordeaux, France
| | - Nathalie Dutheil
- Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux Bordeaux, France ; CNRS, Institut des Maladies Neurodégénératives, UMR 5293 Bordeaux, France
| | - Erwan Bezard
- Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux Bordeaux, France ; CNRS, Institut des Maladies Neurodégénératives, UMR 5293 Bordeaux, France
| | - Benjamin Dehay
- Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux Bordeaux, France ; CNRS, Institut des Maladies Neurodégénératives, UMR 5293 Bordeaux, France
| |
Collapse
|
46
|
Herrán E, Requejo C, Ruiz-Ortega JA, Aristieta A, Igartua M, Bengoetxea H, Ugedo L, Pedraz JL, Lafuente JV, Hernández RM. Increased antiparkinson efficacy of the combined administration of VEGF- and GDNF-loaded nanospheres in a partial lesion model of Parkinson's disease. Int J Nanomedicine 2014; 9:2677-87. [PMID: 24920904 PMCID: PMC4043720 DOI: 10.2147/ijn.s61940] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Current research efforts are focused on the application of growth factors, such as glial cell line-derived neurotrophic factor (GDNF) and vascular endothelial growth factor (VEGF), as neuroregenerative approaches that will prevent the neurodegenerative process in Parkinson’s disease. Continuing a previous work published by our research group, and with the aim to overcome different limitations related to growth factor administration, VEGF and GDNF were encapsulated in poly(lactic-co-glycolic acid) nanospheres (NS). This strategy facilitates the combined administration of the VEGF and GDNF into the brain of 6-hydroxydopamine (6-OHDA) partially lesioned rats, resulting in a continuous and simultaneous drug release. The NS particle size was about 200 nm and the simultaneous addition of VEGF NS and GDNF NS resulted in significant protection of the PC-12 cell line against 6-OHDA in vitro. Once the poly(lactic-co-glycolic acid) NS were implanted into the striatum of 6-OHDA partially lesioned rats, the amphetamine rotation behavior test was carried out over 10 weeks, in order to check for in vivo efficacy. The results showed that VEGF NS and GDNF NS significantly decreased the number of amphetamine-induced rotations at the end of the study. In addition, tyrosine hydroxylase immunohistochemical analysis in the striatum and the external substantia nigra confirmed a significant enhancement of neurons in the VEGF NS and GDNF NS treatment group. The synergistic effect of VEGF NS and GDNF NS allows for a reduction of the dose by half, and may be a valuable neurogenerative/neuroreparative approach for treating Parkinson’s disease.
Collapse
Affiliation(s)
- Enara Herrán
- NanoBioCel Group, Laboratory of Pharmaceutics, University of the Basque Country (UPV/EHU), School of Pharmacy, Vitoria, Spain ; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria, Spain
| | - Catalina Requejo
- LaNCE, Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
| | | | - Asier Aristieta
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Manoli Igartua
- NanoBioCel Group, Laboratory of Pharmaceutics, University of the Basque Country (UPV/EHU), School of Pharmacy, Vitoria, Spain ; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria, Spain
| | - Harkaitz Bengoetxea
- LaNCE, Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Luisa Ugedo
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Jose Luis Pedraz
- NanoBioCel Group, Laboratory of Pharmaceutics, University of the Basque Country (UPV/EHU), School of Pharmacy, Vitoria, Spain ; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria, Spain
| | - Jose Vicente Lafuente
- LaNCE, Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Rosa Maria Hernández
- NanoBioCel Group, Laboratory of Pharmaceutics, University of the Basque Country (UPV/EHU), School of Pharmacy, Vitoria, Spain ; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria, Spain
| |
Collapse
|
47
|
Tarazi FI, Sahli ZT, Wolny M, Mousa SA. Emerging therapies for Parkinson's disease: from bench to bedside. Pharmacol Ther 2014; 144:123-33. [PMID: 24854598 DOI: 10.1016/j.pharmthera.2014.05.010] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 05/01/2014] [Indexed: 02/08/2023]
Abstract
The prevalence of Parkinson's disease (PD) increases with age and is projected to increase in parallel to the rising average age of the population. The disease can have significant health-related, social, and financial implications not only for the patient and the caregiver, but for the health care system as well. While the neuropathology of this neurodegenerative disorder is fairly well understood, its etiology remains a mystery, making it difficult to target therapy. The currently available drugs for treatment provide only symptomatic relief and do not control or prevent disease progression, and as a result patient compliance and satisfaction are low. Several emerging pharmacotherapies for PD are in different stages of clinical development. These therapies include adenosine A2A receptor antagonists, glutamate receptor antagonists, monoamine oxidase inhibitors, anti-apoptotic agents, and antioxidants such as coenzyme Q10, N-acetyl cysteine, and edaravone. Other emerging non-pharmacotherapies include viral vector gene therapy, microRNAs, transglutaminases, RTP801, stem cells and glial derived neurotrophic factor (GDNF). In addition, surgical procedures including deep brain stimulation, pallidotomy, thalamotomy and gamma knife surgery have emerged as alternative interventions for advanced PD patients who have completely utilized standard treatments and still suffer from persistent motor fluctuations. While several of these therapies hold much promise in delaying the onset of the disease and slowing its progression, more pharmacotherapies and surgical interventions need to be investigated in different stages of PD. It is hoped that these emerging therapies and surgical procedures will strengthen our clinical armamentarium for improved treatment of PD.
Collapse
Affiliation(s)
- F I Tarazi
- Department of Psychiatry and Neuroscience Program, Harvard Medical School, McLean Hospital, Belmont, MA 02478, USA.
| | - Z T Sahli
- Department of Psychiatry and Neuroscience Program, Harvard Medical School, McLean Hospital, Belmont, MA 02478, USA; School of Medicine, American University of Beirut, Beirut, Lebanon
| | - M Wolny
- The Pharmaceutical Research Institute at Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA
| | - S A Mousa
- The Pharmaceutical Research Institute at Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA
| |
Collapse
|
48
|
Buttery PC, Barker RA. Treating Parkinson's disease in the 21st century: can stem cell transplantation compete? J Comp Neurol 2014; 522:2802-16. [PMID: 24610597 PMCID: PMC4233918 DOI: 10.1002/cne.23577] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Revised: 08/07/2013] [Accepted: 10/08/2013] [Indexed: 12/25/2022]
Abstract
The characteristic and selective degeneration of a unique population of cells—the nigrostriatal dopamine (DA) neurons—that occurs in Parkinson’s disease (PD) has made the condition an iconic target for cell replacement therapies. Indeed, transplantation of fetal ventral mesencephalic cells into the DA-deficient striatum was first trialled nearly 30 years ago, at a time when other treatments for the disease were less well developed. Over recent decades standard treatments for PD have advanced, and newer biological therapies are now emerging. In the 21st century, stem cell technology will have to compete alongside other sophisticated treatments, including deep brain stimulation and gene therapies. In this review we examine how stem cell–based transplantation therapies compare with these novel and emerging treatments in the management of this common condition. J. Comp. Neurol. 522:2802–2816, 2014.
Collapse
Affiliation(s)
- Philip C Buttery
- John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
| | | |
Collapse
|
49
|
Ojala DS, Amara DP, Schaffer DV. Adeno-associated virus vectors and neurological gene therapy. Neuroscientist 2014; 21:84-98. [PMID: 24557878 DOI: 10.1177/1073858414521870] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Gene therapy has strong potential for treating a variety of genetic disorders, as demonstrated in recent clinical trials. There is unfortunately no scarcity of disease targets, and the grand challenge in this field has instead been the development of safe and efficient gene delivery platforms. To date, approximately two thirds of the 1800 gene therapy clinical trials completed worldwide have used viral vectors. Among these, adeno-associated virus (AAV) has emerged as particularly promising because of its impressive safety profile and efficiency in transducing a wide range of cell types. Gene delivery to the CNS involves both considerable promise and unique challenges, and better AAV vectors are thus needed to translate CNS gene therapy approaches to the clinic. This review discusses strategies for vector design, potential routes of administration, immune responses, and clinical applications of AAV in the CNS.
Collapse
Affiliation(s)
- David S Ojala
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA
| | - Dominic P Amara
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - David V Schaffer
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA Department of Bioengineering, University of California, Berkeley, CA, USA The Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| |
Collapse
|
50
|
Tereshchenko J, Maddalena A, Bähr M, Kügler S. Pharmacologically controlled, discontinuous GDNF gene therapy restores motor function in a rat model of Parkinson's disease. Neurobiol Dis 2014; 65:35-42. [PMID: 24440408 DOI: 10.1016/j.nbd.2014.01.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 12/13/2013] [Accepted: 01/08/2014] [Indexed: 11/18/2022] Open
Abstract
Neurotrophic factors have raised hopes to be able to cure symptoms and to prevent progressive neurodegeneration in devastating neurological diseases. Gene therapy by means of viral vectors can overcome the hurdle of targeted delivery, but its current configuration is irreversible and thus much less controllable than that of classical pharmacotherapies. We thus aimed at developing a strategy allowing for both curative and controllable neurotrophic factor expression. Therefore, the short-term, intermittent and reversible expression of a neutrophic factor was evaluated for therapeutic efficacy in a slowly progressive animal model of Parkinson's disease (PD). We demonstrate that short-term induced expression of glial cell line derived neurotrophic factor (GDNF) is sufficient to provide i) substantial protection of nigral dopaminergic neurons from degeneration and ii) restoration of dopamine supply and motor behaviour in the partial striatal 6-OHDA model PD. These neurorestorative effects of GDNF lasted several weeks beyond the time of its expression. Later on, therapeutic efficacy ceased, but was restored by a second short induction of GDNF expression, demonstrating that monthly application of the inducing drug mifepristone was sufficient to maintain neuroprotective and neurorestorative GDNF levels. These findings suggest that forthcoming gene therapies for PD or other neurodegenerative disorders can be designed in a way that low frequency application of an approved drug can provide controllable and therapeutically efficient levels of GDNF or other neurotrophic factors. Neurotrophic factor expression can be withdrawn in case of off-target effects or sufficient clinical benefit, a feature that may eventually increase the acceptance of gene therapy for less advanced patients, which may profit better from such approaches.
Collapse
Affiliation(s)
- Julia Tereshchenko
- University Medicine Göttingen, Dept. of Neurology, Center for Molecular Physiology of the Brain, Waldweg 33, 37073 Göttingen, Germany
| | - Andrea Maddalena
- University Medicine Göttingen, Dept. of Neurology, Center for Molecular Physiology of the Brain, Waldweg 33, 37073 Göttingen, Germany
| | - Mathias Bähr
- University Medicine Göttingen, Dept. of Neurology, Center for Molecular Physiology of the Brain, Waldweg 33, 37073 Göttingen, Germany
| | - Sebastian Kügler
- University Medicine Göttingen, Dept. of Neurology, Center for Molecular Physiology of the Brain, Waldweg 33, 37073 Göttingen, Germany.
| |
Collapse
|