1
|
Kalyn M, Garvey R, Lee H, Mbesha HA, Curry J, Saxena V, Mennigen JA, Ekker M. Differential roles of NR4A2 (NURR1) paralogs in the brain and behavior of zebrafish. J Neurochem 2025; 169:e16234. [PMID: 39388214 DOI: 10.1111/jnc.16234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 10/15/2024]
Abstract
Dopaminergic (DAnergic) dysfunction and imbalanced dopamine (DA) levels are known contributors to the pathogenesis of numerous psychiatric and neurodegenerative disorders. Of the many identified risk factors for DA-associated disorders, nuclear receptor subfamily 4 group A2 (NR4A2; or nuclear receptor related-1 protein (NURR1)), a transcription factor involved in DAnergic differentiation, has been associated with Parkinson's disease and attention deficit hyperactive disorder (ADHD). In zebrafish, transient loss of nr4a2 was previously shown to decrease tyrosine hydroxylase (TH) expression and impair locomotion. To further characterize the roles of the two zebrafish nr4a2 paralogs, nr4a2a, and nr4a2b, we produced targeted loss-of-function mutants and examined DAnergic neuron regeneration, oxidative respiration, and behavioral traits. The loss of nr4a2a function more closely recapitulated Parkinsonian phenotypes and affected neurotrophic factor gene expression. Conversely, nr4a2b mutants displayed behavioral symptoms reminiscent of mice deficient in Nr4a2 with increased neurotrophic output. In contrast, nr4a2b mutants also displayed increased metabolic input from non-mitochondrial sources indicative of high cytosolic reactive oxygen species and perturbed mitochondrial function. The nr4a2a mutants also showed increased maximal respiration, which may suggest a compensatory mechanism to meet the metabolic requirements of DAnergic neuron health. Overall, the zebrafish mutants generated in this study helped uncover molecular mechanisms involved in DA-related disease pathologies, and in the regeneration of DAnergic neurons.
Collapse
Affiliation(s)
- Michael Kalyn
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Rose Garvey
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Hyojin Lee
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | | | - Jory Curry
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Vishal Saxena
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Jan A Mennigen
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Marc Ekker
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
2
|
Palasz E, Gasiorowska-Bien A, Drapich P, Niewiadomski W, Niewiadomska G. Steady Moderate Exercise Confers Resilience Against Neurodegeneration and Neuroinflammation in a Mouse Model of Parkinson's Disease. Int J Mol Sci 2025; 26:1146. [PMID: 39940916 PMCID: PMC11818830 DOI: 10.3390/ijms26031146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Intensive aerobic exercise slows the progression of movement disorders in Parkinson's disease (PD) and is therefore recommended as an important component of treatment for PD patients. Studies in animal models of PD have shown that vigorous exercise has neuroprotective effects, and emerging evidence suggests that it may be a disease-modifying treatment in humans. However, many people with PD may not be able to participate in vigorous exercise because of multiple medical conditions that severely limit their physical activity. In this study, we have shown that chronic MPTP treatment in sedentary mice resulted in loss of dopaminergic neurons in the SNpc, decreased levels of neurotrophins, BDNF and GDNF, and increased levels of inflammatory markers and pro-inflammatory changes in immunocompetent cells. Moderate exercise, initiated both before and after chronic MPTP treatment, significantly attenuated the loss of dopaminergic neurons and increased BDNF and GDNF levels even above those in sedentary control mice. No signs of inflammation were observed in MPTP-treated mice, either when training began before or after MPTP treatment. Training induced beneficial changes in the dopaminergic system, increased levels of neurotrophins and suppression of inflammation were similar for both steady moderate (present data) and intense training (our previously published data). This suggests that there is a kind of saturation when the percentage of rescued dopaminergic neurons reaches the highest possible value, and therefore further increases in exercise intensity do not enhance neuroprotection. In conclusion, our present results compared with the previous data show that increasing exercise intensity beyond the level used in this study does not increase the neuroprotective effect of aerobic training in a mouse model of Parkinson's disease.
Collapse
Affiliation(s)
- Ewelina Palasz
- Department of Cellular Signalling, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Anna Gasiorowska-Bien
- Clinical and Research Department of Applied Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (A.G.-B.); (P.D.); (W.N.)
| | - Patrycja Drapich
- Clinical and Research Department of Applied Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (A.G.-B.); (P.D.); (W.N.)
| | - Wiktor Niewiadomski
- Clinical and Research Department of Applied Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (A.G.-B.); (P.D.); (W.N.)
| | - Grazyna Niewiadomska
- Clinical and Research Department of Applied Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (A.G.-B.); (P.D.); (W.N.)
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
| |
Collapse
|
3
|
Ozgür-Gunes Y, Le Stunff C, Bougnères P. Oligodendrocytes, the Forgotten Target of Gene Therapy. Cells 2024; 13:1973. [PMID: 39682723 PMCID: PMC11640421 DOI: 10.3390/cells13231973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
If the billions of oligodendrocytes (OLs) populating the central nervous system (CNS) of patients could express their feelings, they would undoubtedly tell gene therapists about their frustration with the other neural cell populations, neurons, microglia, or astrocytes, which have been the favorite targets of gene transfer experiments. This review questions why OLs have been left out of most gene therapy attempts. The first explanation is that the pathogenic role of OLs is still discussed in most CNS diseases. Another reason is that the so-called ubiquitous CAG, CBA, CBh, or CMV promoters-widely used in gene therapy studies-are unable or poorly able to activate the transcription of episomal transgene copies brought by adeno-associated virus (AAV) vectors in OLs. Accordingly, transgene expression in OLs has either not been found or not been evaluated in most gene therapy studies in rodents or non-human primates. The aims of the current review are to give OLs their rightful place among the neural cells that future gene therapy could target and to encourage researchers to test the effect of OL transduction in various CNS diseases.
Collapse
Affiliation(s)
- Yasemin Ozgür-Gunes
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA;
| | - Catherine Le Stunff
- MIRCen Institute, Laboratoire des Maladies Neurodégénératives, Commissariat à l’Energie Atomique, 92260 Fontenay-aux-Roses, France;
- NEURATRIS at MIRCen, 92260 Fontenay-aux-Roses, France
- UMR1195 Inserm and University Paris Saclay, 94270 Le Kremlin-Bicêtre, France
| | - Pierre Bougnères
- MIRCen Institute, Laboratoire des Maladies Neurodégénératives, Commissariat à l’Energie Atomique, 92260 Fontenay-aux-Roses, France;
- NEURATRIS at MIRCen, 92260 Fontenay-aux-Roses, France
- Therapy Design Consulting, 94300 Vincennes, France
| |
Collapse
|
4
|
Kumagai S, Nakajima T, Muramatsu SI. Intraparenchymal delivery of adeno-associated virus vectors for the gene therapy of neurological diseases. Expert Opin Biol Ther 2024; 24:773-785. [PMID: 39066718 DOI: 10.1080/14712598.2024.2386339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 07/07/2024] [Accepted: 07/25/2024] [Indexed: 07/30/2024]
Abstract
INTRODUCTION In gene therapy with adeno-associated virus (AAV) vectors for diseases of the central nervous system, the vectors can be administered into blood vessels, cerebrospinal fluid space, or the brain parenchyma. When gene transfer to a large area of the brain is required, the first two methods are used, but for diseases in which local gene transfer is expected to be effective, vectors are administered directly into the brain parenchyma. AREAS COVERED Strategies for intraparenchymal vector delivery in gene therapy for Parkinson's disease, aromatic l-amino acid decarboxylase (AADC) deficiency, and epilepsy are reviewed. EXPERT OPINION Stereotactic intraparenchymal injection of AAV vectors allows precise gene delivery to the target site. Although more surgically invasive than intravascular or intrathecal administration, intraparenchymal vector delivery has the advantage of a lower vector dose, and preexisting neutralizing antibodies have little effect on the transduction efficacy. This approach improves motor function in AADC deficiency and led to regulatory approval of an AAV vector for the disease in the EU. Although further validation through clinical studies is needed, direct infusion of viral vectors into the brain parenchyma is expected to be a novel treatment for Parkinson's disease and drug-resistant epilepsy.
Collapse
Affiliation(s)
- Shinichi Kumagai
- Department of Neurosurgery, Jichi Medical University, Tochigi, Japan
| | - Takeshi Nakajima
- Department of Neurosurgery, Jichi Medical University, Tochigi, Japan
| | - Shin-Ichi Muramatsu
- Division of Neurological Gene Therapy, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
5
|
Ge G, Sivasubramanian BP, Geng BD, Zhao S, Zhou Q, Huang G, O'Connor JC, Clark RA, Li S. Long-term benefits of hematopoietic stem cell-based macrophage/microglia delivery of GDNF to the CNS in a mouse model of Parkinson's disease. Gene Ther 2024; 31:324-334. [PMID: 38627469 PMCID: PMC11245959 DOI: 10.1038/s41434-024-00451-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 03/21/2024] [Accepted: 03/27/2024] [Indexed: 05/03/2024]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) protects dopaminergic neurons in various models of Parkinson's disease (PD). Cell-based GDNF gene delivery mitigates neurodegeneration and improves both motor and non-motor functions in PD mice. As PD is a chronic condition, this study aims to investigate the long-lasting benefits of hematopoietic stem cell (HSC)-based macrophage/microglia-mediated CNS GDNF (MMC-GDNF) delivery in an MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) mouse model. The results indicate that GDNF treatment effectively ameliorated MPTP-induced motor deficits for up to 12 months, which coincided with the protection of nigral dopaminergic neurons and their striatal terminals. Also, the HSC-derived macrophages/microglia were recruited selectively to the neurodegenerative areas of the substantia nigra. The therapeutic benefits appear to involve two mechanisms: (1) macrophage/microglia release of GDNF-containing exosomes, which are transferred to target neurons, and (2) direct release of GDNF by macrophage/microglia, which diffuses to target neurons. Furthermore, the study found that plasma GDNF levels were significantly increased from baseline and remained stable over time, potentially serving as a convenient biomarker for future clinical trials. Notably, no weight loss, altered food intake, cerebellar pathology, or other adverse effects were observed. Overall, this study provides compelling evidence for the long-term therapeutic efficacy and safety of HSC-based MMC-GDNF delivery in the treatment of PD.
Collapse
Affiliation(s)
- Guo Ge
- Audie L. Murphy VA Medical Center, 7400 Merton Minter Boulevard, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX, 78229, USA
- Department of Human Anatomy, School of Basic Medicine, Guizhou Medical University, Guian New Area, Guizhou, 550025, China
| | | | - Bill D Geng
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | - Shujie Zhao
- Audie L. Murphy VA Medical Center, 7400 Merton Minter Boulevard, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | - Qing Zhou
- Audie L. Murphy VA Medical Center, 7400 Merton Minter Boulevard, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | - Gang Huang
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Jason C O'Connor
- Audie L. Murphy VA Medical Center, 7400 Merton Minter Boulevard, San Antonio, TX, 78229, USA
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Robert A Clark
- Audie L. Murphy VA Medical Center, 7400 Merton Minter Boulevard, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | - Senlin Li
- Audie L. Murphy VA Medical Center, 7400 Merton Minter Boulevard, San Antonio, TX, 78229, USA.
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX, 78229, USA.
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
6
|
Lutfi Ismaeel G, Makki AlHassani OJ, S Alazragi R, Hussein Ahmed A, H Mohamed A, Yasir Jasim N, Hassan Shari F, Almashhadani HA. Genetically engineered neural stem cells (NSCs) therapy for neurological diseases; state-of-the-art. Biotechnol Prog 2023; 39:e3363. [PMID: 37221947 DOI: 10.1002/btpr.3363] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/28/2023] [Accepted: 05/01/2023] [Indexed: 05/25/2023]
Abstract
Neural stem cells (NSCs) are multipotent stem cells with remarkable self-renewal potential and also unique competencies to differentiate into neurons, astrocytes, and oligodendrocytes (ODCs) and improve the cellular microenvironment. In addition, NSCs secret diversity of mediators, including neurotrophic factors (e.g., BDNF, NGF, GDNF, CNTF, and NT-3), pro-angiogenic mediators (e.g., FGF-2 and VEGF), and anti-inflammatory biomolecules. Thereby, NSCs transplantation has become a reasonable and effective treatment for various neurodegenerative disorders by their capacity to induce neurogenesis and vasculogenesis and dampen neuroinflammation and oxidative stress. Nonetheless, various drawbacks such as lower migration and survival and less differential capacity to a particular cell lineage concerning the disease pathogenesis hinder their application. Thus, genetic engineering of NSCs before transplantation is recently regarded as an innovative strategy to bypass these hurdles. Indeed, genetically modified NSCs could bring about more favored therapeutic influences post-transplantation in vivo, making them an excellent option for neurological disease therapy. This review for the first time offers a comprehensive review of the therapeutic capability of genetically modified NSCs rather than naïve NSCs in neurological disease beyond brain tumors and sheds light on the recent progress and prospect in this context.
Collapse
Affiliation(s)
- Ghufran Lutfi Ismaeel
- Department of Pharmacology, College of Pharmacy, University of Al-Ameed, Karbala, Iraq
| | | | - Reem S Alazragi
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Ammar Hussein Ahmed
- Department of Radiology and Sonar, College of Medical Techniques, Al-Farahidi University, Baghdad, Iraq
| | - Asma'a H Mohamed
- Intelligent Medical Systems Department, Al-Mustaqbal University College, Babylon, Iraq
| | - Nisreen Yasir Jasim
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Falah Hassan Shari
- Department of Clinical Laboratory Sciences, College of Pharmacy, University of Basrah, Basrah, Iraq
| | | |
Collapse
|
7
|
Zhang CT, Qin DL, Cao XY, Kan JS, Huang XX, Gao DS, Gao J. Dephosphorylation of Six2Y129 protects tyrosine hydroxylase-positive cells in SNpc by regulating TEA domain 1 expression. iScience 2023; 26:107049. [PMID: 37534182 PMCID: PMC10391717 DOI: 10.1016/j.isci.2023.107049] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 04/03/2023] [Accepted: 06/01/2023] [Indexed: 08/04/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by selective loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc). We recently reported that Six2 could reverse the degeneration of DA neurons in a dephosphorylation state. Here we further identified that Eya1 was the phosphatase of Six2 that could dephosphorylate the tyrosine 129 (Y129) site by forming a complex with Six2 in damaged DA cells. Dephosphorylated Six2 then translocates from the cytoplasm to the nucleus. Using ChIP-qPCR and dual luciferase assay, we found that dephosphorylated Six2 down-regulates TEA domain1 (Tead1) expression, thus inhibiting 6-hydroxydopamine (6-OHDA)-induced apoptosis in DA cells. Furthermore, we showed Six2Y129F/Tead1 signaling could protect against the loss of SNpc tyrosine hydroxylase-positive (TH+) cells and improve motor function in PD model rats. Our results demonstrate a dephosphorylation-dependent mechanism of Six2 that restores the degeneration of DA neurons, which could represent a potential therapeutic target for PD.
Collapse
Affiliation(s)
- Can-tang Zhang
- Department of Respiratory and Critical Care, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Deng-li Qin
- Department of Neurobiology and Cell Biology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xia-yin Cao
- Department of Neurobiology and Cell Biology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Jia-shuo Kan
- Department of Neurobiology and Cell Biology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xin-xing Huang
- Department of Neurobiology and Cell Biology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Dian-shuai Gao
- Department of Neurobiology and Cell Biology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Jin Gao
- Department of Neurobiology and Cell Biology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| |
Collapse
|
8
|
Alarcón TA, Presti-Silva SM, Simões APT, Ribeiro FM, Pires RGW. Molecular mechanisms underlying the neuroprotection of environmental enrichment in Parkinson's disease. Neural Regen Res 2023; 18:1450-1456. [PMID: 36571341 PMCID: PMC10075132 DOI: 10.4103/1673-5374.360264] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Parkinson's disease is the most common movement disorder, affecting about 1% of the population over the age of 60 years. Parkinson's disease is characterized clinically by resting tremor, bradykinesia, rigidity and postural instability, as a result of the progressive loss of nigrostriatal dopaminergic neurons. In addition to this neuronal cell loss, Parkinson's disease is characterized by the accumulation of intracellular protein aggregates, Lewy bodies and Lewy neurites, composed primarily of the protein α-synuclein. Although it was first described almost 200 years ago, there are no disease-modifying drugs to treat patients with Parkinson's disease. In addition to conventional therapies, non-pharmacological treatment strategies are under investigation in patients and animal models of neurodegenerative disorders. Among such strategies, environmental enrichment, comprising physical exercise, cognitive stimulus, and social interactions, has been assessed in preclinical models of Parkinson's disease. Environmental enrichment can cause structural and functional changes in the brain and promote neurogenesis and dendritic growth by modifying gene expression, enhancing the expression of neurotrophic factors and modulating neurotransmission. In this review article, we focus on the current knowledge about the molecular mechanisms underlying environmental enrichment neuroprotection in Parkinson's disease, highlighting its influence on the dopaminergic, cholinergic, glutamatergic and GABAergic systems, as well as the involvement of neurotrophic factors. We describe experimental pre-clinical data showing how environmental enrichment can act as a modulator in a neurochemical and behavioral context in different animal models of Parkinson's disease, highlighting the potential of environmental enrichment as an additional strategy in the management and prevention of this complex disease.
Collapse
Affiliation(s)
- Tamara Andrea Alarcón
- Department of Physiological Sciences; Laboratory of Molecular and Behavioral Neurobiology, Health Science Center, Universidade Federal do Espirito Santo, Vitoria, Brazil
| | - Sarah Martins Presti-Silva
- Laboratory of Molecular and Behavioral Neurobiology, Health Science Center, Universidade Federal do Espirito Santo, Vitoria; Department of Biochemistry and Immunology, Institute o Biological Sciences, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, Belo Horizonte, Brazil
| | - Ana Paula Toniato Simões
- Department of Physiological Sciences; Laboratory of Molecular and Behavioral Neurobiology, Health Science Center, Universidade Federal do Espirito Santo, Vitoria, Brazil
| | - Fabiola Mara Ribeiro
- Department of Biochemistry and Immunology, Institute o Biological Sciences, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, Belo Horizonte, Brazil
| | - Rita Gomes Wanderley Pires
- Department of Physiological Sciences; Laboratory of Molecular and Behavioral Neurobiology, Health Science Center, Universidade Federal do Espirito Santo, Vitoria, Brazil
| |
Collapse
|
9
|
Kong C, Chang WS. Preclinical Research on Focused Ultrasound-Mediated Blood-Brain Barrier Opening for Neurological Disorders: A Review. Neurol Int 2023; 15:285-300. [PMID: 36810473 PMCID: PMC9944161 DOI: 10.3390/neurolint15010018] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/02/2023] [Accepted: 02/10/2023] [Indexed: 02/17/2023] Open
Abstract
Several therapeutic agents for neurological disorders are usually not delivered to the brain owing to the presence of the blood-brain barrier (BBB), a special structure present in the central nervous system (CNS). Focused ultrasound (FUS) combined with microbubbles can reversibly and temporarily open the BBB, enabling the application of various therapeutic agents in patients with neurological disorders. In the past 20 years, many preclinical studies on drug delivery through FUS-mediated BBB opening have been conducted, and the use of this method in clinical applications has recently gained popularity. As the clinical application of FUS-mediated BBB opening expands, it is crucial to understand the molecular and cellular effects of FUS-induced microenvironmental changes in the brain so that the efficacy of treatment can be ensured, and new treatment strategies established. This review describes the latest research trends in FUS-mediated BBB opening, including the biological effects and applications in representative neurological disorders, and suggests future directions.
Collapse
Affiliation(s)
| | - Won Seok Chang
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| |
Collapse
|
10
|
Alfonsetti M, d’Angelo M, Castelli V. Neurotrophic factor-based pharmacological approaches in neurological disorders. Neural Regen Res 2022; 18:1220-1228. [PMID: 36453397 PMCID: PMC9838155 DOI: 10.4103/1673-5374.358619] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Aging is a physiological event dependent on multiple pathways that are linked to lifespan and processes leading to cognitive decline. This process represents the major risk factor for aging-related diseases such as Alzheimer's disease, Parkinson's disease, and ischemic stroke. The incidence of all these pathologies increases exponentially with age. Research on aging biology has currently focused on elucidating molecular mechanisms leading to the development of those pathologies. Cognitive deficit and neurodegeneration, common features of aging-related pathologies, are related to the alteration of the activity and levels of neurotrophic factors, such as brain-derived neurotrophic factor, nerve growth factor, and glial cell-derived neurotrophic factor. For this reason, treatments that modulate neurotrophin levels have acquired a great deal of interest in preventing neurodegeneration and promoting neural regeneration in several neurological diseases. Those treatments include both the direct administration of neurotrophic factors and the induced expression with viral vectors, neurotrophins' binding with biomaterials or other molecules to increase their bioavailability but also cell-based therapies. Considering neurotrophins' crucial role in aging pathologies, here we discuss the involvement of several neurotrophic factors in the most common brain aging-related diseases and the most recent therapeutic approaches that provide direct and sustained neurotrophic support.
Collapse
Affiliation(s)
- Margherita Alfonsetti
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | - Michele d’Angelo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy,Correspondence to: Vanessa Castelli, .
| |
Collapse
|
11
|
Stefani A, Pierantozzi M, Cardarelli S, Stefani L, Cerroni R, Conti M, Garasto E, Mercuri NB, Marini C, Sucapane P. Neurotrophins as Therapeutic Agents for Parkinson’s Disease; New Chances From Focused Ultrasound? Front Neurosci 2022; 16:846681. [PMID: 35401084 PMCID: PMC8990810 DOI: 10.3389/fnins.2022.846681] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 01/31/2022] [Indexed: 01/02/2023] Open
Abstract
Magnetic Resonance–guided Focused Ultrasound (MRgFUS) represents an effective micro-lesioning approach to target pharmaco-resistant tremor, mostly in patients afflicted by essential tremor (ET) and/or Parkinson’s disease (PD). So far, experimental protocols are verifying the clinical extension to other facets of the movement disorder galaxy (i.e., internal pallidus for disabling dyskinesias). Aside from those neurosurgical options, one of the most intriguing opportunities of this technique relies on its capability to remedy the impermeability of blood–brain barrier (BBB). Temporary BBB opening through low-intensity focused ultrasound turned out to be safe and feasible in patients with PD, Alzheimer’s disease, and amyotrophic lateral sclerosis. As a mere consequence of the procedures, some groups described even reversible but significant mild cognitive amelioration, up to hippocampal neurogenesis partially associated to the increased of endogenous brain-derived neurotrophic factor (BDNF). A further development elevates MRgFUS to the status of therapeutic tool for drug delivery of putative neurorestorative therapies. Since 2012, FUS-assisted intravenous administration of BDNF or neurturin allowed hippocampal or striatal delivery. Experimental studies emphasized synergistic modalities. In a rodent model for Huntington’s disease, engineered liposomes can carry glial cell line–derived neurotrophic factor (GDNF) plasmid DNA (GDNFp) to form a GDNFp-liposome (GDNFp-LPs) complex through pulsed FUS exposures with microbubbles; in a subacute MPTP-PD model, the combination of intravenous administration of neurotrophic factors (either through protein or gene delivery) plus FUS did curb nigrostriatal degeneration. Here, we explore these arguments, focusing on the current, translational application of neurotrophins in neurodegenerative diseases.
Collapse
Affiliation(s)
- Alessandro Stefani
- Department of System Medicine, Parkinson Center, University Tor Vergata, Rome, Italy
- *Correspondence: Alessandro Stefani,
| | | | - Silvia Cardarelli
- Department of System Medicine, Parkinson Center, University Tor Vergata, Rome, Italy
| | - Lucrezia Stefani
- Department of System Medicine, Parkinson Center, University Tor Vergata, Rome, Italy
| | - Rocco Cerroni
- Department of System Medicine, Parkinson Center, University Tor Vergata, Rome, Italy
| | - Matteo Conti
- Department of System Medicine, UOC Neurology, University Tor Vergata, Rome, Italy
| | - Elena Garasto
- Department of System Medicine, UOC Neurology, University Tor Vergata, Rome, Italy
| | - Nicola B. Mercuri
- Department of System Medicine, UOC Neurology, University Tor Vergata, Rome, Italy
| | - Carmine Marini
- UOC Neurology and Stroke Unit, University of L’Aquila, L’Aquila, Italy
| | | |
Collapse
|
12
|
Lin MS, Chen SM, Hua KF, Chen WJ, Hsieh CC, Lin CC. Freshwater Clam Extract Mitigates Neuroinflammation and Amplifies Neurotrophic Activity of Glia: Insights from In Vitro Model of Neurodegenerative Pathomechanism. J Clin Med 2022; 11:jcm11030553. [PMID: 35160004 PMCID: PMC8836940 DOI: 10.3390/jcm11030553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/16/2022] [Accepted: 01/20/2022] [Indexed: 11/28/2022] Open
Abstract
Background. An extensive body of research suggests that brain inflammation and oxidative stress are the underlying causes of Parkinson’s disease (PD), for which no potent therapeutic approach exists to mitigate the degradation of dopamine neurons. Freshwater clams, an ancient health food of Chinese origin, have been documented to exhibit anti-inflammatory and antioxidant effects. We previously reported that freshwater clam extract (FCE) can attenuate astrocytic activation and subsequent proinflammatory cytokine production from substantia nigra in an MPTP-induced PD mouse model. This article provides insight into the potential mechanisms through which FCE regulates neuroinflammation in a glia model of injury. Materials and methods. In total, 1 μg/mL lipopolysaccharide (LPS) and 200 μM rotenone were conducted in primary glial cell cultures to mimic the respective neuroinflammation and oxidative stress during injury-induced glial cell reactivation, which is relevant to the pathological process of PD. Results. FCE markedly reduced LPS-induced neuroinflammation by suppressing NO and TNF-α production and the expression of pro-inflammatory cytokines. In addition, FCE was effective at reducing rotenone-induced toxicity by diminishing ROS production, promoting antioxidant enzymes (SOD, catalase, and GPx) and minimizing the decline in glial-cell-secreted neurotrophic factors (GDNF, BDNF). These impacts ultimately led to a decrease in glial apoptosis. Conclusions. Evidence reveals that FCE is capable of stabilizing reactive glia, as demonstrated by reduced neuroinflammation, oxidative stress, the increased release of neurotrophic factors and the inhibition of apoptosis, which provides therapeutic insight into neurodegenerative diseases, including PD.
Collapse
Affiliation(s)
- Muh-Shi Lin
- Division of Neurosurgery, Department of Surgery, Kuang Tien General Hospital, Taichung 43303, Taiwan;
- Department of Biotechnology and Animal Science, College of Bioresources, National Ilan University, Yilan 26047, Taiwan; (K.-F.H.); (W.-J.C.); (C.-C.H.)
- Department of Biotechnology, College of Medical and Health Care, Hung Kuang University, Taichung 43302, Taiwan
- Department of Health Business Administration, College of Medical and Health Care, Hung Kuang University, Taichung 43302, Taiwan
| | - Shu-Mei Chen
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei Medical University, Taipei 110, Taiwan;
- Department of Surgery, School of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Kuo-Feng Hua
- Department of Biotechnology and Animal Science, College of Bioresources, National Ilan University, Yilan 26047, Taiwan; (K.-F.H.); (W.-J.C.); (C.-C.H.)
| | - Wei-Jung Chen
- Department of Biotechnology and Animal Science, College of Bioresources, National Ilan University, Yilan 26047, Taiwan; (K.-F.H.); (W.-J.C.); (C.-C.H.)
| | - Cho-Chen Hsieh
- Department of Biotechnology and Animal Science, College of Bioresources, National Ilan University, Yilan 26047, Taiwan; (K.-F.H.); (W.-J.C.); (C.-C.H.)
| | - Chai-Ching Lin
- Department of Biotechnology and Animal Science, College of Bioresources, National Ilan University, Yilan 26047, Taiwan; (K.-F.H.); (W.-J.C.); (C.-C.H.)
- Correspondence: ; Tel.: +886-3-9310592; Fax: +886-3-9280609
| |
Collapse
|
13
|
Bentea E, De Pauw L, Verbruggen L, Winfrey LC, Deneyer L, Moore C, Albertini G, Sato H, Van Eeckhaut A, Meshul CK, Massie A. Aged xCT-Deficient Mice Are Less Susceptible for Lactacystin-, but Not 1-Methyl-4-Phenyl-1,2,3,6- Tetrahydropyridine-, Induced Degeneration of the Nigrostriatal Pathway. Front Cell Neurosci 2022; 15:796635. [PMID: 34975413 PMCID: PMC8718610 DOI: 10.3389/fncel.2021.796635] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 11/24/2021] [Indexed: 12/23/2022] Open
Abstract
The astrocytic cystine/glutamate antiporter system x c - (with xCT as the specific subunit) imports cystine in exchange for glutamate and has been shown to interact with multiple pathways in the brain that are dysregulated in age-related neurological disorders, including glutamate homeostasis, redox balance, and neuroinflammation. In the current study, we investigated the effect of genetic xCT deletion on lactacystin (LAC)- and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced degeneration of the nigrostriatal pathway, as models for Parkinson's disease (PD). Dopaminergic neurons of adult xCT knock-out mice (xCT-/-) demonstrated an equal susceptibility to intranigral injection of the proteasome inhibitor LAC, as their wild-type (xCT+/+) littermates. Contrary to adult mice, aged xCT-/- mice showed a significant decrease in LAC-induced degeneration of nigral dopaminergic neurons, depletion of striatal dopamine (DA) and neuroinflammatory reaction, compared to age-matched xCT+/+ littermates. Given this age-related protection, we further investigated the sensitivity of aged xCT-/- mice to chronic and progressive MPTP treatment. However, in accordance with our previous observations in adult mice (Bentea et al., 2015a), xCT deletion did not confer protection against MPTP-induced nigrostriatal degeneration in aged mice. We observed an increased loss of nigral dopaminergic neurons, but equal striatal DA denervation, in MPTP-treated aged xCT-/- mice when compared to age-matched xCT+/+ littermates. To conclude, we reveal age-related protection against proteasome inhibition-induced nigrostriatal degeneration in xCT-/- mice, while xCT deletion failed to protect nigral dopaminergic neurons of aged mice against MPTP-induced toxicity. Our findings thereby provide new insights into the role of system x c - in mechanisms of dopaminergic cell loss and its interaction with aging.
Collapse
Affiliation(s)
- Eduard Bentea
- Laboratory of Neuro-Aging and Viro-Immunotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Laura De Pauw
- Laboratory of Neuro-Aging and Viro-Immunotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lise Verbruggen
- Laboratory of Neuro-Aging and Viro-Immunotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lila C Winfrey
- Neurocytology Laboratory, Veterans Affairs Medical Center, Research Services, Portland, OR, United States
| | - Lauren Deneyer
- Laboratory of Neuro-Aging and Viro-Immunotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Cynthia Moore
- Neurocytology Laboratory, Veterans Affairs Medical Center, Research Services, Portland, OR, United States
| | - Giulia Albertini
- Laboratory of Neuro-Aging and Viro-Immunotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hideyo Sato
- Department of Medical Technology, Niigata University, Niigata, Japan
| | - Ann Van Eeckhaut
- Research Group Experimental Pharmacology, Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Vrije Universiteit Brussel, Brussels, Belgium
| | - Charles K Meshul
- Neurocytology Laboratory, Veterans Affairs Medical Center, Research Services, Portland, OR, United States.,Department of Behavioral Neuroscience and Pathology, Oregon Health and Science University, Portland, OR, United States
| | - Ann Massie
- Laboratory of Neuro-Aging and Viro-Immunotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
14
|
Negrini M, Tomasello G, Davidsson M, Fenyi A, Adant C, Hauser S, Espa E, Gubinelli F, Manfredsson FP, Melki R, Heuer A. Sequential or Simultaneous Injection of Preformed Fibrils and AAV Overexpression of Alpha-Synuclein Are Equipotent in Producing Relevant Pathology and Behavioral Deficits. JOURNAL OF PARKINSON'S DISEASE 2022; 12:1133-1153. [PMID: 35213388 PMCID: PMC9198765 DOI: 10.3233/jpd-212555] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 02/04/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND Preclinical rodent models for Parkinson's disease (PD) based on viral human alpha-synuclein (h-αSyn) overexpression recapitulate some of the pathological hallmarks as it presents in humans, such as progressive cell loss and additional synucleinopathy in cortical and subcortical structures. Recent studies have combined viral vector-based overexpression of human wild-type αSyn with the sequential or simultaneous inoculation of preformed fibrils (PFFs) derived from human αSyn. OBJECTIVE The goal of the study was to investigate whether sequential or combined delivery of the AAV vector and the PFFs are equipotent in inducing stable neurodegeneration and behavioral deficits. METHODS Here we compare between four experimental paradigms (PFFs only, AAV-h-αSyn only, AAV-h-αSyn with simultaneous PFFs, and AAV-h-αSyn with sequential PFFs) and their respective GFP control groups. RESULTS We observed reduction of TH expression and loss of neurons in the midbrain in all AAV (h-αSyn or GFP) injected groups, with or without additional PFFs inoculation. The overexpression of either h-αSyn or GFP alone induced motor deficits and dysfunctional dopamine release/reuptake in electrochemical recordings in the ipsilateral striatum. However, we observed a substantial formation of insoluble h-αSyn aggregates and inflammatory response only when h-αSyn and PFFs were combined. Moreover, the presence of h-αSyn induced higher axonal pathology compared to control groups. CONCLUSION Simultaneous AAV and PFFs injections are equipotent in the presented experimental setup in inducing histopathological and behavioral changes. This model provides new and interesting possibilities for characterizing PD pathology in preclinical models and means to assess future therapeutic interventions.
Collapse
Affiliation(s)
- Matilde Negrini
- Behavioural Neuroscience Laboratory, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Giuseppe Tomasello
- Behavioural Neuroscience Laboratory, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Marcus Davidsson
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, USA
- Molecular Neuromodulation, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Alexis Fenyi
- Institut Francois Jacob (MIRCen), CEA and Laboratory of Neurodegenerative Diseases, CNRS, Fontenay-aux-Roses, France
| | - Cécile Adant
- Behavioural Neuroscience Laboratory, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Swantje Hauser
- Behavioural Neuroscience Laboratory, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Elena Espa
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Francesco Gubinelli
- Behavioural Neuroscience Laboratory, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Fredric P. Manfredsson
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Ronald Melki
- Institut Francois Jacob (MIRCen), CEA and Laboratory of Neurodegenerative Diseases, CNRS, Fontenay-aux-Roses, France
| | - Andreas Heuer
- Behavioural Neuroscience Laboratory, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
15
|
Mesa-Infante V, Afonso-Oramas D, Salas-Hernández J, Rodríguez-Núñez J, Barroso-Chinea P. Long-term exposure to GDNF induces dephosphorylation of Ret, AKT, and ERK1/2, and is ineffective at protecting midbrain dopaminergic neurons in cellular models of Parkinson's disease. Mol Cell Neurosci 2021; 118:103684. [PMID: 34826608 DOI: 10.1016/j.mcn.2021.103684] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 11/12/2021] [Accepted: 11/13/2021] [Indexed: 12/01/2022] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) promotes differentiation, proliferation, and survival in different cell types, including dopaminergic neurons. Thus, GDNF has been proposed as a promising neuroprotective therapy in Parkinson's disease. Although findings from cellular and animal models of Parkinson's disease were encouraging, results emerging from clinical trials were not as good as expected, probably due to the inappropriate administration protocols. Despite the growing information on GDNF action mechanisms, many aspects of its pharmacological effects are still unclear and data from different studies are still contradictory. Considering that GDNF action mechanisms are mediated by its receptor tyrosine kinase Ret, which activates PI3K/AKT and MAPK/ERK signaling pathways, we aimed to investigate Ret activation and its effect over both signaling pathways in midbrain cell cultures treated with GDNF at different doses (0.3, 1, and 10 ng/ml) and times (15 min, 24 h, 24 h (7 days), and 7 continuous days). The results showed that short-term or acute (15 min, 24 h, and 24 h (7 days)) GDNF treatment in rat midbrain neurons increases Tyrosine hydroxylase (TH) expression and the phosphorylation levels of Ret (Tyr 1062), AKT (Ser 473), ERK1/2 (Thr202/Tyr204), S6 (Ser 235/236), and GSK3-β (Ser 9). However, the phosphorylation level of these kinases, TH expression, and dopamine uptake, decreased below basal levels after long-term or prolonged treatment with 1 and 10 ng/ml GDNF (7 continuous days). Our data suggest that long-term GDNF treatment inactivates the receptor by an unknown mechanism, affecting its neuroprotective capacity against degeneration caused by 6-OHDA or rotenone, while short-term exposure to GDNF promoted dopaminergic cell survival. These findings highlight the need to find new and more effective long-acting therapeutic approaches for disorders in which GDNF plays a beneficial role, including Parkinson's disease. In this regard, it is necessary to propose new GDNF treatment guidelines to regulate and control its long-term expression levels and optimize the clinical use of this trophic factor in patients with Parkinson's disease.
Collapse
Affiliation(s)
- V Mesa-Infante
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain
| | - D Afonso-Oramas
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain; Instituto de Tecnologías Biomédicas de Canarias (ITB), Universidad de La Laguna, Tenerife, Spain.
| | - J Salas-Hernández
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain
| | - J Rodríguez-Núñez
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain
| | - P Barroso-Chinea
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain; Instituto de Tecnologías Biomédicas de Canarias (ITB), Universidad de La Laguna, Tenerife, Spain.
| |
Collapse
|
16
|
Renko JM, Mahato AK, Visnapuu T, Valkonen K, Karelson M, Voutilainen MH, Saarma M, Tuominen RK, Sidorova YA. Neuroprotective Potential of a Small Molecule RET Agonist in Cultured Dopamine Neurons and Hemiparkinsonian Rats. JOURNAL OF PARKINSONS DISEASE 2021; 11:1023-1046. [PMID: 34024778 PMCID: PMC8461720 DOI: 10.3233/jpd-202400] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Parkinson's disease (PD) is a progressive neurological disorder where loss of dopamine neurons in the substantia nigra and dopamine depletion in the striatum cause characteristic motor symptoms. Currently, no treatment is able to halt the progression of PD. Glial cell line-derived neurotrophic factor (GDNF) rescues degenerating dopamine neurons both in vitro and in animal models of PD. When tested in PD patients, however, the outcomes from intracranial GDNF infusion paradigms have been inconclusive, mainly due to poor pharmacokinetic properties. OBJECTIVE We have developed drug-like small molecules, named BT compounds that activate signaling through GDNF's receptor, the transmembrane receptor tyrosine kinase RET, both in vitro and in vivo and are able to penetrate through the blood-brain barrier. Here we evaluated the properties of BT44, a second generation RET agonist, in immortalized cells, dopamine neurons and rat 6-hydroxydopamine model of PD. METHODS We used biochemical, immunohistochemical and behavioral methods to evaluate the effects of BT44 on dopamine system in vitro and in vivo. RESULTS BT44 selectively activated RET and intracellular pro-survival AKT and MAPK signaling pathways in immortalized cells. In primary midbrain dopamine neurons cultured in serum-deprived conditions, BT44 promoted the survival of the neurons derived from wild-type, but not from RET knockout mice. BT44 also protected cultured wild-type dopamine neurons from MPP+-induced toxicity. In a rat 6-hydroxydopamine model of PD, BT44 reduced motor imbalance and seemed to protect dopaminergic fibers in the striatum. CONCLUSION BT44 holds potential for further development into a novel, possibly disease-modifying, therapy for PD.
Collapse
Affiliation(s)
- Juho-Matti Renko
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Arun Kumar Mahato
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Tanel Visnapuu
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Konsta Valkonen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Mati Karelson
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Merja H Voutilainen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Raimo K Tuominen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Yulia A Sidorova
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
17
|
Amalric M, Pattij T, Sotiropoulos I, Silva JM, Sousa N, Ztaou S, Chiamulera C, Wahlberg LU, Emerich DF, Paolone G. Where Dopaminergic and Cholinergic Systems Interact: A Gateway for Tuning Neurodegenerative Disorders. Front Behav Neurosci 2021; 15:661973. [PMID: 34366802 PMCID: PMC8340002 DOI: 10.3389/fnbeh.2021.661973] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 06/14/2021] [Indexed: 12/18/2022] Open
Abstract
Historically, many investigations into neurodegenerative diseases have focused on alterations in specific neuronal populations such as, for example, the loss of midbrain dopaminergic neurons in Parkinson's disease (PD) and loss of cholinergic transmission in Alzheimer's disease (AD). However, it has become increasingly clear that mammalian brain activities, from executive and motor functioning to memory and emotional responses, are strictly regulated by the integrity of multiple interdependent neuronal circuits. Among subcortical structures, the dopaminergic nigrostriatal and mesolimbic pathways as well as cholinergic innervation from basal forebrain and brainstem, play pivotal roles in orchestrating cognitive and non-cognitive symptoms in PD and AD. Understanding the functional interactions of these circuits and the consequent neurological changes that occur during degeneration provides new opportunities to understand the fundamental inter-workings of the human brain as well as develop new potential treatments for patients with dysfunctional neuronal circuits. Here, excerpted from a session of the European Behavioral Pharmacology Society meeting (Braga, Portugal, August 2019), we provide an update on our recent work in behavioral and cellular neuroscience that primarily focuses on interactions between cholinergic and dopaminergic systems in PD models, as well as stress in AD. These brief discussions include descriptions of (1) striatal cholinergic interneurons (CINs) and PD, (2) dopaminergic and cholinergic modulation of impulse control, and (3) the use of an implantable cell-based system for drug delivery directly the into brain and (4) the mechanisms through which day life stress, a risk factor for AD, damage protein and RNA homeostasis leading to AD neuronal malfunction.
Collapse
Affiliation(s)
- Marianne Amalric
- Centre National de la Recherche Scientifique (CNRS), UMR 7291, Laboratoire de Neurosciences Cognitives, Aix-Marseille University (AMU), Marseille, France
| | - Tommy Pattij
- Amsterdam Neuroscience, Department of Anatomy and Neurosciences, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Ioannis Sotiropoulos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Braga, Portugal
| | - Joana M. Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Braga, Portugal
| | - Nuno Sousa
- ICVS/3B’s – PT Government Associate Laboratory, Braga, Portugal
| | - Samira Ztaou
- Centre National de la Recherche Scientifique (CNRS), UMR 7291, Laboratoire de Neurosciences Cognitives, Aix-Marseille University (AMU), Marseille, France
- Department of Molecular Therapeutics, New York State Psychiatric Institute, Department of Psychiatry, Columbia University, New York, NY, United States
| | - Cristiano Chiamulera
- Department of Diagnostic and Public Health, Section of Pharmacology, University of Verona, Verona, Italy
| | | | | | - Giovanna Paolone
- Department of Diagnostic and Public Health, Section of Pharmacology, University of Verona, Verona, Italy
| |
Collapse
|
18
|
Goulding SR, Lévesque M, Sullivan AM, Collins LM, O'Keeffe GW. Quinacrine and Niclosamide Promote Neurite Growth in Midbrain Dopaminergic Neurons Through the Canonical BMP-Smad Pathway and Protect Against Neurotoxin and α-Synuclein-Induced Neurodegeneration. Mol Neurobiol 2021; 58:3405-3416. [PMID: 33713017 DOI: 10.1007/s12035-021-02351-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/05/2021] [Indexed: 11/25/2022]
Abstract
Parkinson's disease is a neurodegenerative disorder characterised by nigrostriatal dopaminergic degeneration, and intracellular α-synuclein aggregation. Current pharmacological treatments are solely symptomatic so there is a need to identify agents that can slow or stop dopaminergic degeneration. One proposed class of therapeutics are neurotrophic factors which promote the survival of nigrostriatal dopaminergic neurons. However, neurotrophic factors need to be delivered directly to the brain. An alternative approach may be to identify pharmacological agents which can reach the brain to stimulate neurotrophic factor expression and/or their signalling pathways in dopaminergic neurons. BMP2 is a neurotrophic factor that is expressed in the human substantia nigra; exogenous BMP2 administration protects against dopaminergic degeneration in in vitro models of PD. In this study, we investigated the neurotrophic potential of two FDA-approved drugs, quinacrine and niclosamide, that are modulators of BMP2 signalling. We report that quinacrine and niclosamide, like BMP2, significantly increased neurite length, as a readout of neurotrophic action, in SH-SY5Y cells and dopaminergic neurons in primary cultures of rat ventral mesencephalon. We also show that these effects of quinacrine and niclosamide require the activation of BMP-Smad signalling. Finally, we demonstrate that quinacrine and niclosamide are neuroprotective against degeneration induced by the neurotoxins, MPP+ and 6-OHDA, and by viral-mediated overexpression of α-synuclein in vitro. Collectively, this study identifies two drugs, that are safe for use in patients' to 'are approved for human use, that exert neurotrophic effects on dopaminergic neurons through modulation of BMP-Smad signalling. This rationalises the further study of drugs that target the BMP-Smad pathway as potential neuroprotective pharmacotherapy for Parkinson's disease.
Collapse
Affiliation(s)
- Susan R Goulding
- Department of Biological Sciences, Munster Technological University, Cork, Ireland
- Department of Anatomy and Neuroscience and Cork Neuroscience Centre, University College Cork, Cork, Ireland
| | - Martin Lévesque
- Department of Psychiatry and Neurosciences, Cervo Brain Research Centre, Université Laval, Quebec, QC, Canada
| | - Aideen M Sullivan
- Department of Anatomy and Neuroscience and Cork Neuroscience Centre, University College Cork, Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Louise M Collins
- Department of Anatomy and Neuroscience and Cork Neuroscience Centre, University College Cork, Cork, Ireland.
- Department of Physiology, University College Cork, Cork, Ireland.
| | - Gerard W O'Keeffe
- Department of Anatomy and Neuroscience and Cork Neuroscience Centre, University College Cork, Cork, Ireland.
- APC Microbiome Institute, University College Cork, Cork, Ireland.
| |
Collapse
|
19
|
Yang R, Boldrey J, Jiles D, Schneider I, Que L. On chip detection of glial cell-derived neurotrophic factor secreted from dopaminergic cells under magnetic stimulation. Biosens Bioelectron 2021; 182:113179. [PMID: 33774433 DOI: 10.1016/j.bios.2021.113179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/13/2021] [Accepted: 03/17/2021] [Indexed: 11/24/2022]
Abstract
Glial cell-derived neurotrophic factor (GDNF) is a small protein potently promoting the survival of dopaminergic and motor neurons. GDNF can be secreted from different types of cells including the dopaminergic neural cell line, N27. N27 cells, a rat dopaminergic neural cell line, is regarded as a suitable in vitro model for Parkinson's disease (PD) research. For PD treatment, transcranial magnetic stimulation (TMS), a noninvasive therapeutic method, showed beneficial clinical effects, but the mechanism for its benefit is not understood. Because GDNF is a potent neurotrophic factor, it is of great value to evaluate if GDNF secretion from N27 cells can be affected by magnetic stimulation (MS). However, the current methods for detecting GDNF are time-consuming and expensive. In this paper we outline the detection of GDNF secretion from N27 cells by ultrasensitive nanopore thin film sensors (nanosensor) for the first time. As low as 2 pg/mL GDNF can be readily detected by the nanosensor. Furthermore, we show that MS can promote GDNF secretion from N27 cells. Specifically, the GDNF concentration in N27 cell-conditioned media under MS treatment shows statistically significant increase up to 2-fold after 5 days in vitro in comparison with the control. This nanosensor along with the in vitro PD model N27 cells provides a low-cost, easy-to-use, sensitive approach for studying potential cell biological mechanisms of the clinical benefits of MS on PD.
Collapse
Affiliation(s)
- Renyuan Yang
- Department of Electrical and Computer Engineering, Iowa State University, United States
| | - Joseph Boldrey
- Department of Electrical and Computer Engineering, Iowa State University, United States
| | - David Jiles
- Department of Electrical and Computer Engineering, Iowa State University, United States
| | - Ian Schneider
- Department of Chemical and Biological Engineering, Iowa State University, United States; Department of Genetics, Development and Cell Biology, Iowa State University, United States
| | - Long Que
- Department of Electrical and Computer Engineering, Iowa State University, United States.
| |
Collapse
|
20
|
Dolatshahi M, Ranjbar Hameghavandi MH, Sabahi M, Rostamkhani S. Nuclear factor-kappa B (NF-κB) in pathophysiology of Parkinson disease: Diverse patterns and mechanisms contributing to neurodegeneration. Eur J Neurosci 2021; 54:4101-4123. [PMID: 33884689 DOI: 10.1111/ejn.15242] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/25/2021] [Accepted: 04/07/2021] [Indexed: 01/02/2023]
Abstract
Parkinson's disease (PD), the most common movement disorder, comprises several pathophysiologic mechanisms including misfolded alpha-synuclein aggregation, inflammation, mitochondrial dysfunction, and synaptic loss. Nuclear Factor-Kappa B (NF-κB), as a key regulator of a myriad of cellular reactions, is shown to be involved in such mechanisms associated with PD, and the changes in NF-κB expression is implicated in PD. Alpha-synuclein accumulation, the characteristic feature of PD pathology, is known to trigger NF-κB activation in neurons, thereby propagating apoptosis through several mechanisms. Furthermore, misfolded alpha-synuclein released from degenerated neurons, activates several signaling pathways in glial cells which culminate in activation of NF-κB and production of pro-inflammatory cytokines, thereby aggravating neurodegenerative processes. On the other hand, NF-κB activation, acting as a double-edged sword, can be necessary for survival of neurons. For instance, NF-κB activation is necessary for competent mitochondrial function and deficiency in c-Rel, one of the NF-κB proteins, is known to propagate DA neuron loss via several mechanisms. Despite the dual role of NF-κB in PD, several agents by selectively modifying the mechanisms and pathways associated with NF-κB, can be effective in attenuating DA neuron loss and PD, as reviewed in this paper.
Collapse
Affiliation(s)
- Mahsa Dolatshahi
- Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | | | - Mohammadmahdi Sabahi
- NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Neurosurgery Research Group (NRG), Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sabra Rostamkhani
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Bressan C, Saghatelyan A. Intrinsic Mechanisms Regulating Neuronal Migration in the Postnatal Brain. Front Cell Neurosci 2021; 14:620379. [PMID: 33519385 PMCID: PMC7838331 DOI: 10.3389/fncel.2020.620379] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/08/2020] [Indexed: 01/19/2023] Open
Abstract
Neuronal migration is a fundamental brain development process that allows cells to move from their birthplaces to their sites of integration. Although neuronal migration largely ceases during embryonic and early postnatal development, neuroblasts continue to be produced and to migrate to a few regions of the adult brain such as the dentate gyrus and the subventricular zone (SVZ). In the SVZ, a large number of neuroblasts migrate into the olfactory bulb (OB) along the rostral migratory stream (RMS). Neuroblasts migrate in chains in a tightly organized micro-environment composed of astrocytes that ensheath the chains of neuroblasts and regulate their migration; the blood vessels that are used by neuroblasts as a physical scaffold and a source of molecular factors; and axons that modulate neuronal migration. In addition to diverse sets of extrinsic micro-environmental cues, long-distance neuronal migration involves a number of intrinsic mechanisms, including membrane and cytoskeleton remodeling, Ca2+ signaling, mitochondria dynamics, energy consumption, and autophagy. All these mechanisms are required to cope with the different micro-environment signals and maintain cellular homeostasis in order to sustain the proper dynamics of migrating neuroblasts and their faithful arrival in the target regions. Neuroblasts in the postnatal brain not only migrate into the OB but may also deviate from their normal path to migrate to a site of injury induced by a stroke or by certain neurodegenerative disorders. In this review, we will focus on the intrinsic mechanisms that regulate long-distance neuroblast migration in the adult brain and on how these pathways may be modulated to control the recruitment of neuroblasts to damaged/diseased brain areas.
Collapse
Affiliation(s)
- Cedric Bressan
- CERVO Brain Research Center, Quebec City, QC, Canada.,Department of Psychiatry and Neuroscience, Université Laval, Quebec City, QC, Canada
| | - Armen Saghatelyan
- CERVO Brain Research Center, Quebec City, QC, Canada.,Department of Psychiatry and Neuroscience, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
22
|
Ferrini F, Salio C, Boggio EM, Merighi A. Interplay of BDNF and GDNF in the Mature Spinal Somatosensory System and Its Potential Therapeutic Relevance. Curr Neuropharmacol 2021; 19:1225-1245. [PMID: 33200712 PMCID: PMC8719296 DOI: 10.2174/1570159x18666201116143422] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/17/2020] [Accepted: 10/05/2020] [Indexed: 11/22/2022] Open
Abstract
The growth factors BDNF and GDNF are gaining more and more attention as modulators of synaptic transmission in the mature central nervous system (CNS). The two molecules undergo a regulated secretion in neurons and may be anterogradely transported to terminals where they can positively or negatively modulate fast synaptic transmission. There is today a wide consensus on the role of BDNF as a pro-nociceptive modulator, as the neurotrophin has an important part in the initiation and maintenance of inflammatory, chronic, and/or neuropathic pain at the peripheral and central level. At the spinal level, BDNF intervenes in the regulation of chloride equilibrium potential, decreases the excitatory synaptic drive to inhibitory neurons, with complex changes in GABAergic/glycinergic synaptic transmission, and increases excitatory transmission in the superficial dorsal horn. Differently from BDNF, the role of GDNF still remains to be unraveled in full. This review resumes the current literature on the interplay between BDNF and GDNF in the regulation of nociceptive neurotransmission in the superficial dorsal horn of the spinal cord. We will first discuss the circuitries involved in such a regulation, as well as the reciprocal interactions between the two factors in nociceptive pathways. The development of small molecules specifically targeting BDNF, GDNF and/or downstream effectors is opening new perspectives for investigating these neurotrophic factors as modulators of nociceptive transmission and chronic pain. Therefore, we will finally consider the molecules of (potential) pharmacological relevance for tackling normal and pathological pain.
Collapse
Affiliation(s)
- Francesco Ferrini
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
- Department of Psychiatry & Neuroscience, Université Laval, Québec, Canada
| | - Chiara Salio
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| | - Elena M. Boggio
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| | - Adalberto Merighi
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
- National Institute of Neuroscience, Grugliasco, Italy
| |
Collapse
|
23
|
Efimova EV, Kozlova AA, Razenkova V, Katolikova NV, Antonova KA, Sotnikova TD, Merkulyeva NS, Veshchitskii AS, Kalinina DS, Korzhevskii DE, Musienko PE, Kanov EV, Gainetdinov RR. Increased dopamine transmission and adult neurogenesis in trace amine-associated receptor 5 (TAAR5) knockout mice. Neuropharmacology 2020; 182:108373. [PMID: 33132188 DOI: 10.1016/j.neuropharm.2020.108373] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/24/2022]
Abstract
Trace amine-associated receptors (TAARs) are a class of sensory G protein-coupled receptors that detect biogenic amines, products of decarboxylation of amino acids. The majority of TAARs (TAAR2-TAAR9) have been described mainly in the olfactory epithelium and considered to be olfactory receptors sensing innate odors. However, there is recent evidence that one of the members of this family, TAAR5, is expressed also in the limbic brain areas receiving projection from the olfactory system and involved in the regulation of emotions. In this study, we further characterized a mouse line lacking TAAR5 (TAAR5 knockout, TAAR5-KO mice) that express beta-galactosidase mapping TAAR5 expression. We found that in TAAR5-KO mice the number of dopamine neurons, the striatal levels of dopamine and its metabolites, as well as striatal levels of GDNF mRNA, are elevated indicating a potential increase in dopamine neuron proliferation. Furthermore, an analysis of TAAR5 beta-galactosidase expression revealed that TAAR5 is present in the major neurogenic areas of the brain such as the subventricular zone (SVZ), the subgranular zone (SGZ) and the less characterized potentially neurogenic zone surrounding the 3rd ventricle. Direct analysis of neurogenesis by using specific markers doublecortin (DCX) and proliferating cell nuclear antigen (PCNA) revealed at least 2-fold increase in the number of proliferating neurons in the SVZ and SGZ of TAAR5-KO mice, but no such markers were detected in mutant or control mice in the areas surrounding the 3rd ventricle. These observations indicate that TAAR5 involved not only in regulation of emotional status but also adult neurogenesis and dopamine transmission. Thus, future TAAR5 antagonists may exert not only antidepressant and/or anxiolytic action but may also provide new treatment opportunity for neurodegenerative disorders such as Parkinson's disease.
Collapse
Affiliation(s)
- Evgeniya V Efimova
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, 199034, Russia
| | - Alena A Kozlova
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, 199034, Russia
| | | | - Nataliia V Katolikova
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, 199034, Russia
| | - Kristina A Antonova
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, 199034, Russia
| | - Tatyana D Sotnikova
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, 199034, Russia
| | - Natalia S Merkulyeva
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, 199034, Russia; Pavlov Institute of Physiology RAS, St. Petersburg, Russia
| | | | - Daria S Kalinina
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, 199034, Russia
| | | | - Pavel E Musienko
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, 199034, Russia; St. Petersburg State Research Institute of Phthisiopulmonology, Ministry of Healthcare of the RF, St. Petersburg, 191036, Russia
| | - Evgeny V Kanov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, 199034, Russia
| | - Raul R Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, 199034, Russia; St. Petersburg University Hospital, St. Petersburg State University, St. Petersburg, 199034, Russia.
| |
Collapse
|
24
|
Paolone G. From the Gut to the Brain and Back: Therapeutic Approaches for the Treatment of Network Dysfunction in Parkinson's Disease. Front Neurol 2020; 11:557928. [PMID: 33117258 PMCID: PMC7575743 DOI: 10.3389/fneur.2020.557928] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 09/04/2020] [Indexed: 12/16/2022] Open
Abstract
Parkinson's disease (PD) is a complex, multisystem, progressive, degenerative disorder characterized by severe, debilitating motor dysfunction, cognitive impairments, and mood disorders. Although preclinical research has traditionally focused on the motor deficits resulting from the loss of nigrostriatal dopaminergic neurons, up to two thirds of PD patients present separate and distinct behavioral changes. Loss of basal forebrain cholinergic neurons occurs as early as the loss of dopaminergic cells and contributes to the cognitive decline in PD. In addition, attentional deficits can limit posture control and movement efficacy caused by dopaminergic cell loss. Complicating the picture further is intracellular α-synuclein accumulation beginning in the enteric nervous system and diffusing to the substantia nigra through the dorsal motor neurons of the vagus nerve. It seems that α-synuclein's role is that of mediating dopamine synthesis, storage, and release, and its function has not been completely understood. Treating a complex, multistage network disorder, such as PD, likely requires a multipronged approach. Here, we describe a few approaches that could be used alone or perhaps in combination to achieve a greater mosaic of behavioral benefit. These include (1) using encapsulated, genetically modified cells as delivery vehicles for administering neuroprotective trophic factors, such as GDNF, in a direct and sustained means to the brain; (2) immunotherapeutic interventions, such as vaccination or the use of monoclonal antibodies against aggregated, pathological α-synuclein; (3) the continuous infusion of levodopa-carbidopa through an intestinal gel pad to attenuate the loss of dopaminergic function and manage the motor and non-motor complications in PD patients; and (4) specific rehabilitation treatment programs for drug-refractory motor complications.
Collapse
Affiliation(s)
- Giovanna Paolone
- Department of Diagnostic and Public Health - Section of Pharmacology, University of Verona, Verona, Italy
| |
Collapse
|
25
|
Conway JA, Ince S, Black S, Kramer ER. GDNF/RET signaling in dopamine neurons in vivo. Cell Tissue Res 2020; 382:135-146. [PMID: 32870383 DOI: 10.1007/s00441-020-03268-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 07/24/2020] [Indexed: 12/15/2022]
Abstract
The glial cell line-derived neurotrophic factor (GDNF) and its canonical receptor Ret can signal both in tandem and separately to exert many vital functions in the midbrain dopamine system. It is known that Ret has effects on maintenance, physiology, protection and regeneration in the midbrain dopamine system, with the physiological functions of GDNF still somewhat unclear. Notwithstanding, Ret ligands, such as GDNF, are considered as promising candidates for neuroprotection and/or regeneration in Parkinson's disease, although data from clinical trials are so far inconclusive. In this review, we discuss the current knowledge of GDNF/Ret signaling in the dopamine system in vivo as well as crosstalk with pathology-associated proteins and their signaling in mammals.
Collapse
Affiliation(s)
- James A Conway
- Peninsula Medical School, Institute of Translational and Stratified Medicine, Faculty of Health, University of Plymouth, Plymouth, UK
| | - Selvi Ince
- Peninsula Medical School, Institute of Translational and Stratified Medicine, Faculty of Health, University of Plymouth, Plymouth, UK
| | | | - Edgar R Kramer
- Peninsula Medical School, Institute of Translational and Stratified Medicine, Faculty of Health, University of Plymouth, Plymouth, UK.
| |
Collapse
|
26
|
Chen C, Guderyon MJ, Li Y, Ge G, Bhattacharjee A, Ballard C, He Z, Masliah E, Clark RA, O’Connor JC, Li S. Non-toxic HSC Transplantation-Based Macrophage/Microglia-Mediated GDNF Delivery for Parkinson's Disease. Mol Ther Methods Clin Dev 2020; 17:83-98. [PMID: 31890743 PMCID: PMC6931095 DOI: 10.1016/j.omtm.2019.11.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 11/15/2019] [Indexed: 02/08/2023]
Abstract
Glial cell-line-derived neurotrophic factor (GDNF) is a potent neuroprotective agent in cellular and animal models of Parkinson's disease (PD). However, CNS delivery of GDNF in clinical trials has proven challenging due to blood-brain barrier (BBB) impermeability, poor diffusion within brain tissue, and large brain size. We report that using non-toxic mobilization-enabled preconditioning, hematopoietic stem cell (HSC) transplantation-based macrophage-mediated gene delivery may provide a solution to overcome these obstacles. Syngeneic bone marrow HSCs were transduced ex vivo with a lentiviral vector expressing macrophage promoter-driven GDNF and transplanted into 14-week-old MitoPark mice exhibiting PD-like impairments. Transplant preconditioning with granulocyte colony-stimulating factor (G-CSF) and AMD3100 was used to vacate bone marrow stem cell niches. Chimerism reached ∼80% after seven transplantation cycles. Transgene-expressing macrophages infiltrated degenerating CNS regions of MitoPark mice (not wild-type littermate controls), resulting in increased GDNF levels in the midbrain. Macrophage GDNF delivery not only markedly improved motor and non-motor dysfunction, but also dramatically mitigated the loss of dopaminergic neurons in both substantia nigra and the ventral tegmental area and preserved axonal terminals in the striatum. Striatal dopamine levels were almost completely restored. Our data support further development of mobilization-enabled HSC transplantation (HSCT)-based macrophage-mediated GDNF gene delivery as a disease-modifying therapy for PD.
Collapse
Affiliation(s)
- Cang Chen
- Department of Medicine, The University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Michael J. Guderyon
- Department of Medicine, The University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Yang Li
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Guo Ge
- Department of Medicine, The University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Anindita Bhattacharjee
- Department of Medicine, The University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Cori Ballard
- Department of Medicine, The University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Zhixu He
- Department of Pediatrics, Zunyi Medical University Affiliated Hospital and Key Laboratory of Adult Stem Cell Transformation Research, Chinese Academy of Medical Science, Guiyang, Guizhou 550025, China
| | | | - Robert A. Clark
- Department of Medicine, The University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
- Audie L. Murphy VA Hospital, 7400 Merton Minter Boulevard, San Antonio, TX 78229, USA
| | - Jason C. O’Connor
- Department of Pharmacology, The University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
- Audie L. Murphy VA Hospital, 7400 Merton Minter Boulevard, San Antonio, TX 78229, USA
| | - Senlin Li
- Department of Medicine, The University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
- Department of Pharmacology, The University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
- Audie L. Murphy VA Hospital, 7400 Merton Minter Boulevard, San Antonio, TX 78229, USA
| |
Collapse
|
27
|
Elsworth JD. Parkinson's disease treatment: past, present, and future. J Neural Transm (Vienna) 2020; 127:785-791. [PMID: 32172471 PMCID: PMC8330829 DOI: 10.1007/s00702-020-02167-1] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 03/03/2020] [Indexed: 12/15/2022]
Abstract
The substantial contributions of Dr. Gerald Stern to past and current treatments for Parkinson's disease patients are reviewed, which form the foundation for an evaluation of future options to control symptoms and halt progression of the disease. These opportunities will depend on a greater understanding of the relative contributions of the environment, genetic and epigenetic influences to disease onset, and promise to emerge as strategies for improving mitochondrial function, halting accumulation of synuclein and neuromelanin, in addition to refinement of stem cell and gene therapies. Such advances will be achieved through deployment of improved models for the disease.
Collapse
Affiliation(s)
- John D Elsworth
- Yale University School of Medicine, New Haven, CT, 06511, USA.
| |
Collapse
|
28
|
Schneider A, Sari AT, Alhaddad H, Sari Y. Overview of Therapeutic Drugs and Methods for the Treatment of Parkinson's Disease. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2020; 19:195-206. [PMID: 32448109 DOI: 10.2174/1871527319666200525011110] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/21/2020] [Accepted: 04/23/2020] [Indexed: 12/28/2022]
Abstract
Parkinson's Disease (PD) is a neurodegenerative disease involving degeneration of dopaminergic neurons of the nigrostriatal pathways. Over the past decades, most of the medications for the treatment of PD patients have been used to modulate dopamine concentrations in the basal ganglia. This includes levodopa and its inhibitory metabolizing enzymes. In addition to modulating dopamine concentrations in the brain, there are D2-like dopamine receptor agonists that mimic the action of dopamine to compensate for the deficit in dopamine found in PD patients. Muscarinic antagonists' drugs are used rarely due to some side effects. Monoamine oxidase inhibitors are among the first in line, and are considered popular drugs that reduce the metabolism of dopamine in PD patients. Furthermore, we discussed in this review the existence of certain glutamate receptor antagonists for the treatment of PD. Alternatively, we further discussed the potential therapeutic role of adenosine (2A) receptor antagonists, such as tozadenant and istradefylline in the treatment of PD. We also discussed the important role of serotonin1A receptor agonist, adrenergic autoreceptors (α2) antagonists and calcium channel blockers in the treatment of PD. Finally, neurotrophic factors, such as glial cell line-derived neurotrophic growth factor and brain-derived neurotrophic factor are considered the primary factors for neuroprotection in PD.
Collapse
Affiliation(s)
- Andrew Schneider
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, United States
| | - Adam T Sari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, United States
| | - Hasan Alhaddad
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, United States
| | - Youssef Sari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, United States
| |
Collapse
|
29
|
Tao L, Ma W, Wu L, Xu M, Yang Y, Zhang W, Sha W, Li H, Xu J, Feng R, Xue D, Zhang J, Dooley S, Seki E, Liu P, Liu C. Glial cell line-derived neurotrophic factor (GDNF) mediates hepatic stellate cell activation via ALK5/Smad signalling. Gut 2019; 68:2214-2227. [PMID: 31171625 PMCID: PMC6842044 DOI: 10.1136/gutjnl-2018-317872] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 04/18/2019] [Accepted: 05/03/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Although glial cell line-derived neurotrophic factor (GDNF) is a member of the transforming growth factor-β superfamily, its function in liver fibrosis has rarely been studied. Here, we investigated the role of GDNF in hepatic stellate cell (HSC) activation and liver fibrosis in humans and mice. DESIGN GDNF expression was examined in liver biopsies and sera from patients with liver fibrosis. The functional role of GDNF in liver fibrosis was examined in mice with adenoviral delivery of the GDNF gene, GDNF sgRNA CRISPR/Cas9 and the administration of GDNF-blocking antibodies. GDNF was examined on HSC activation using human and mouse primary HSCs. The binding of activin receptor-like kinase 5 (ALK5) to GDNF was determined using surface plasmon resonance (SPR), molecular docking, mutagenesis and co-immunoprecipitation. RESULTS GDNF mRNA and protein levels are significantly upregulated in patients with stage F4 fibrosis. Serum GDNF content correlates positively with α-smooth muscle actin (α-SMA) and Col1A1 mRNA in human fibrotic livers. Mice with overexpressed GDNF display aggravated liver fibrosis, while mice with silenced GDNF expression or signalling inhibition by GDNF-blocking antibodies have reduced fibrosis and HSC activation. GDNF is confined mainly to HSCs and contributes to HSC activation through ALK5 at His39 and Asp76 and through downstream signalling via Smad2/3, but not through GDNF family receptor alpha-1 (GFRα1). GDNF, ALK5 and α-SMA colocalise in human and mouse HSCs, as demonstrated by confocal microscopy. CONCLUSIONS GDNF promotes HSC activation and liver fibrosis through ALK5/Smad signalling. Inhibition of GDNF could be a novel therapeutic strategy to combat liver fibrosis.
Collapse
Affiliation(s)
- Le Tao
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Laboratory of Liver Disease, Department of Infectious Disease, PutuoHospital , Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenting Ma
- Laboratory of Liver Disease, Department of Infectious Disease, PutuoHospital , Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liu Wu
- Laboratory of Liver Disease, Department of Infectious Disease, PutuoHospital , Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mingyi Xu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanqin Yang
- Department of Pathology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Zhang
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenjun Sha
- Departmentof Endocrinology and Metabolism, PutuoHospital , Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hongshan Li
- Department of Hepatology, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Jianrong Xu
- Department of Pharmacology and Chemical Biology, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rilu Feng
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Dongying Xue
- Laboratory of Liver Disease, Department of Infectious Disease, PutuoHospital , Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jie Zhang
- Laboratory of Liver Disease, Department of Infectious Disease, PutuoHospital , Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Steven Dooley
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ekihiro Seki
- Division of Digestive and Liver Diseases, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Ping Liu
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cheng Liu
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Laboratory of Liver Disease, Department of Infectious Disease, PutuoHospital , Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Shanghai, China
| |
Collapse
|
30
|
The Potential Influence of the Bacterial Microbiome on the Development and Progression of ADHD. Nutrients 2019; 11:nu11112805. [PMID: 31744191 PMCID: PMC6893446 DOI: 10.3390/nu11112805] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/11/2019] [Accepted: 11/13/2019] [Indexed: 02/06/2023] Open
Abstract
The latest research cumulates staggering information about the correlation between the microbiota-gut-brain axis and neurodevelopmental disorders. This review aims to shed light on the potential influence of the microbiome on the development of the most prevalent neurodevelopmental disease, attention-deficit-hyperactive disorder (ADHD). As the etiology and pathophysiology of ADHD are still unclear, finding viable biomarkers and effective treatment still represent a challenge. Therefore, we focused on factors that have been associated with a higher risk of developing ADHD, while simultaneously influencing the microbial composition. We reviewed the effect of a differing microbial makeup on neurotransmitter concentrations important in the pathophysiology of ADHD. Additionally, we deduced factors that correlate with a high prevalence of ADHD, while simultaneously affecting the gut microbiome, such as emergency c-sections, and premature birth as the former leads to a decrease of the gut microbial diversity and the latter causes neuroprotective Lactobacillus levels to be reduced. Also, we assessed nutritional influences, such as breastfeeding, ingestion of short-chain fatty acids (SCFAs) and polyunsaturated fatty acids (PUFAs) on the host′s microbiome and development of ADHD. Finally, we discussed the potential significance of Bifidobacterium as a biomarker for ADHD, the importance of preventing premature birth as prophylaxis and nutrition as a prospective therapeutic measurement against ADHD.
Collapse
|
31
|
Abstract
In vivo gene therapy for neurodegenerative disorders has turned out to be a formidable challenge. It is a field not much older than twenty years, but we were many who would have predicted a much easier path towards the clinic using this treatment modality. For Parkinson’s disease patients, this has meant a frustrating wait, seeing many promising therapies being forgotten after a few pre-clinical proof-of-concept studies. The reasons for this are both scientific and economical. However, this is slowly but surely changing and over the next two decades we will see a very exciting development in this field. In a foreseeable future, gene therapy will be a very natural component of many clinical therapies, not least in Parkinson’s disease.
Collapse
Affiliation(s)
- Tomas Björklund
- Molecular Neuromodulation, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| |
Collapse
|
32
|
Pena SA, Iyengar R, Eshraghi RS, Bencie N, Mittal J, Aljohani A, Mittal R, Eshraghi AA. Gene therapy for neurological disorders: challenges and recent advancements. J Drug Target 2019; 28:111-128. [DOI: 10.1080/1061186x.2019.1630415] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Stefanie A. Pena
- Department of Otolaryngology, Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Rahul Iyengar
- Department of Otolaryngology, Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Rebecca S. Eshraghi
- Department of Otolaryngology, Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Nicole Bencie
- Department of Otolaryngology, Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jeenu Mittal
- Department of Otolaryngology, Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Abdulrahman Aljohani
- Department of Otolaryngology, Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Rahul Mittal
- Department of Otolaryngology, Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Adrien A. Eshraghi
- Department of Otolaryngology, Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Biomedical Engineering, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
33
|
Barsuglia JP, Polanco M, Palmer R, Malcolm BJ, Kelmendi B, Calvey T. A case report SPECT study and theoretical rationale for the sequential administration of ibogaine and 5-MeO-DMT in the treatment of alcohol use disorder. PROGRESS IN BRAIN RESEARCH 2018; 242:121-158. [PMID: 30471678 DOI: 10.1016/bs.pbr.2018.08.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Ibogaine is a plant-derived alkaloid and dissociative psychedelic that demonstrates anti-addictive properties with several substances of abuse, including alcohol. 5-methoxy-N,N-dimethyltryptamine (5-MeO-DMT) is a naturally occurring psychedelic known to occasion potent mystical-type experiences and also demonstrates anti-addictive properties. The potential therapeutic effects of both compounds in treating alcohol use disorder require further investigation and there are no published human neuroimaging findings of either treatment to date. We present the case of a 31-year-old male military veteran with moderate alcohol use disorder who sought treatment at an inpatient clinic in Mexico that utilized a sequential protocol with ibogaine hydrochloride (1550mg, 17.9mg/kg) on day 1, followed by vaporized 5-MeO-DMT (bufotoxin source 50mg, estimated 5-MeO-DMT content, 5-7mg) on day 3. The patient received SPECT neuroimaging that included a resting-state protocol before, and 3 days after completion of the program. During the patient's ibogaine treatment, he experienced dream-like visions that included content pertaining to his alcohol use and resolution of past developmental traumas. He described his treatment with 5-MeO-DMT as a peak transformational and spiritual breakthrough. On post-treatment SPECT neuroimaging, increases in brain perfusion were noted in bilateral caudate nuclei, left putamen, right insula, as well as temporal, occipital, and cerebellar regions compared to the patient's baseline scan. The patient reported improvement in mood, cessation of alcohol use, and reduced cravings at 5 days post-treatment, effects which were sustained at 1 month, with a partial return to mild alcohol use at 2 months. In this case, serial administration of ibogaine and 5-MeO-DMT resulted in increased perfusion in multiple brain regions broadly associated with alcohol use disorders and known pharmacology of both compounds, which coincided with a short-term therapeutic outcome. We present theoretical considerations regarding the potential of both psychedelic medicines in treating alcohol use disorders in the context of these isolated findings, and areas for future investigation.
Collapse
Affiliation(s)
- Joseph P Barsuglia
- Crossroads Treatment Center, Tijuana, Mexico; Mission Within, Oakland, CA, United States; New School Research, LLC, North Hollywood, CA, United States; Terra Incognita Project, NGO, Ben Lomond, CA, United States.
| | - Martin Polanco
- Crossroads Treatment Center, Tijuana, Mexico; Mission Within, Oakland, CA, United States
| | - Robert Palmer
- Yale School of Medicine, New Haven, CT, United States
| | - Benjamin J Malcolm
- College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States
| | - Benjamin Kelmendi
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Tanya Calvey
- Faculty of Health Sciences, University of the Witwatersrand Medical School, Johannesburg, South Africa
| |
Collapse
|
34
|
Ge G, Chen C, Guderyon MJ, Liu J, He Z, Yu Y, Clark RA, Li S. Regulatable Lentiviral Hematopoietic Stem Cell Gene Therapy in a Mouse Model of Parkinson's Disease. Stem Cells Dev 2018; 27:995-1005. [PMID: 29562865 DOI: 10.1089/scd.2018.0030] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) exhibits potent neuroprotective properties in preclinical models of Parkinson's disease (PD), but challenges in GDNF delivery have been reported from clinical trials. To address this barrier, we developed a hematopoietic stem cell transplantation-based macrophage-mediated GDNF therapy platform. Here, we introduced a regulatable lentiviral vector (LV-MSP-Tet-Off-hGDNF) to allow the expression of human GDNF (hGDNF) to be adjusted or stopped by oral administration of doxycycline (Dox). C57BL/6J mice were lethally irradiated with head protection and then transplanted with syngeneic bone marrow cells transduced with either the hGDNF-expressing vector or a corresponding GFP-expressing vector, LV-MSP-Tet-Off-GFP. Suppression of vector gene expression was achieved through administration of Dox in drinking water. To create a toxin-induced Parkinsonian model, mice were injected in two cycles with MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) to yield nigral cell/striatal dopamine loss and behavioral deficits. During the presence of Dox in the drinking water, plasma GDNF was at a basal level, whereas during the absence of Dox, plasma GDNF was significantly elevated, indicating reliable regulation of therapeutic gene expression. Midbrain GDNF levels were altered in parallel, although these did not return completely to basal levels during the periods of Dox withdrawal. Motor activities of the MPTP-Tet-off-hGDNF group were comparable to those of the Tet-off-GFP (subject to no MPTP treatment) group, but substantially better than those of the MPTP-Tet-off-GFP group. Interestingly, the improvement in motor activities was sustained during the Dox-withdrawn periods in MPTP-Tet-off-hGDNF animals. Neuroprotection by therapeutic GDNF expression was further evidenced by significant amelioration of nigral tyrosine hydroxylase loss after both the first and second MPTP treatment cycles. These data suggest that neurotrophic factor expression can be upregulated to achieve efficacy or downregulated in case of off-target effects or adverse events, a feature that may eventually increase the acceptance of this potentially neuroprotective/disease-modifying PD therapy.
Collapse
Affiliation(s)
- Guo Ge
- 1 Department of Medicine, University of Texas Health San Antonio , San Antonio, Texas
- 2 Stem Cells Research Center and Department of Pathology, Guizhou Medical University , Guiyang, Guizhou, China
| | - Cang Chen
- 1 Department of Medicine, University of Texas Health San Antonio , San Antonio, Texas
| | - Michael J Guderyon
- 1 Department of Medicine, University of Texas Health San Antonio , San Antonio, Texas
| | - Jingwei Liu
- 1 Department of Medicine, University of Texas Health San Antonio , San Antonio, Texas
| | - Zhixu He
- 2 Stem Cells Research Center and Department of Pathology, Guizhou Medical University , Guiyang, Guizhou, China
| | - Yanni Yu
- 2 Stem Cells Research Center and Department of Pathology, Guizhou Medical University , Guiyang, Guizhou, China
| | - Robert A Clark
- 1 Department of Medicine, University of Texas Health San Antonio , San Antonio, Texas
- 3 Research & Development Service, Audie L. Murphy VA Hospital , San Antonio, Texas
| | - Senlin Li
- 1 Department of Medicine, University of Texas Health San Antonio , San Antonio, Texas
- 3 Research & Development Service, Audie L. Murphy VA Hospital , San Antonio, Texas
- 4 Department of Pharmacology, University of Texas Health San Antonio , San Antonio, Texas
| |
Collapse
|
35
|
Chen C, Li X, Ge G, Liu J, Biju KC, Laing SD, Qian Y, Ballard C, He Z, Masliah E, Clark RA, O'Connor JC, Li S. GDNF-expressing macrophages mitigate loss of dopamine neurons and improve Parkinsonian symptoms in MitoPark mice. Sci Rep 2018; 8:5460. [PMID: 29615705 PMCID: PMC5882968 DOI: 10.1038/s41598-018-23795-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 03/14/2018] [Indexed: 01/12/2023] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is the most potent neuroprotective agent tested in cellular and animal models of Parkinson's disease (PD). However, CNS delivery of GDNF is restricted by the blood-brain barrier (BBB). Using total body irradiation as transplant preconditioning, we previously reported that hematopoietic stem cell (HSC) transplantation (HSCT)-based macrophage-mediated gene therapy could deliver GDNF to the brain to prevent degeneration of nigrostriatal dopamine (DA) neurons in an acute murine neurotoxicity model. Here, we validate this therapeutic approach in a chronic progressive PD model - the MitoPark mouse, with head shielding to avoid inducing neuroinflammation and compromising BBB integrity. Bone marrow HSCs were transduced ex vivo with a lentiviral vector expressing macrophage promoter-driven GDNF and transplanted into MitoPark mice exhibiting well developed PD-like impairments. Transgene-expressing macrophages infiltrated the midbrains of MitoPark mice, but not normal littermates, and delivered GDNF locally. Macrophage GDNF delivery markedly improved both motor and non-motor symptoms, and dramatically mitigated the loss of both DA neurons in the substantia nigra and tyrosine hydroxylase-positive axonal terminals in the striatum. Our data support further development of this HSCT-based macrophage-mediated GDNF delivery approach in order to address the unmet need for a disease-modifying therapy for PD.
Collapse
Affiliation(s)
- Cang Chen
- Department of Medicine, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, Texas, 78229, USA
| | - Xiuhua Li
- Department of Medicine, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, Texas, 78229, USA
| | - Guo Ge
- Department of Medicine, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, Texas, 78229, USA
| | - Jingwei Liu
- Department of Medicine, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, Texas, 78229, USA
| | - K C Biju
- Department of Medicine, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, Texas, 78229, USA
| | - Suzette D Laing
- Department of Medicine, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, Texas, 78229, USA
| | - Yusheng Qian
- Department of Medicine, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, Texas, 78229, USA
| | - Cori Ballard
- Department of Medicine, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, Texas, 78229, USA
| | - Zhixu He
- Stem Cells Research Center of Guizhou Medical University and Key Laboratory of Adult Stem cell Transformation Research, Chinese Academy of Medical Science, Guiyang, Guizhou, 550025, China
| | | | - Robert A Clark
- Department of Medicine, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, Texas, 78229, USA
- Audie L. Murphy VA Hospital, 7400 Merton Minter Boulevard, San Antonio, Texas, 78229, USA
| | - Jason C O'Connor
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, Texas, 78229, USA.
- Audie L. Murphy VA Hospital, 7400 Merton Minter Boulevard, San Antonio, Texas, 78229, USA.
| | - Senlin Li
- Department of Medicine, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, Texas, 78229, USA.
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, Texas, 78229, USA.
- Audie L. Murphy VA Hospital, 7400 Merton Minter Boulevard, San Antonio, Texas, 78229, USA.
| |
Collapse
|
36
|
Treatment with the noradrenaline re-uptake inhibitor atomoxetine alone and in combination with the α2-adrenoceptor antagonist idazoxan attenuates loss of dopamine and associated motor deficits in the LPS inflammatory rat model of Parkinson's disease. Brain Behav Immun 2018; 69:456-469. [PMID: 29339319 DOI: 10.1016/j.bbi.2018.01.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/04/2018] [Accepted: 01/11/2018] [Indexed: 01/14/2023] Open
Abstract
The impact of treatment with the noradrenaline (NA) re-uptake inhibitor atomoxetine and the α2-adrenoceptor (AR) antagonist idazoxan in an animal model of Parkinson's disease (PD) was assessed. Concurrent systemic treatment with atomoxetine and idazoxan, a combination which serves to enhance the extra-synaptic availability of NA, exerts anti-inflammatory and neuroprotective effects following delivery of an inflammatory stimulus, the bacterial endotoxin, lipopolysaccharide (LPS) into the substantia nigra. Lesion-induced deficits in motor function (akinesia, forelimb-use asymmetry) and striatal dopamine (DA) loss were rescued to varying degrees depending on the treatment. Treatment with atomoxetine following LPS-induced lesion to the substantia nigra, yielded a robust anti-inflammatory effect, suppressing microglial activation and expression of the pro-inflammatory cytokine TNF-α whilst increasing the expression of neurotrophic factors. Furthermore atomoxetine treatment prevented loss of tyrosine hydroxylase (TH) positive nigral dopaminergic neurons and resulted in functional improvements in motor behaviours. Atomoxetine alone was sufficient to achieve most of the observed effects. In combination with idazoxan, an additional improvement in the impairment of contralateral limb use 7 days post lesion and a reduction in amphetamine-mediated rotational asymmetry 14 days post-lesion was observed, compared to atomoxetine or idazoxan treatments alone. The results indicate that increases in central NA tone has the propensity to regulate the neuroinflammatory phenotype in vivo and may act as an endogenous neuroprotective mechanism where inflammation contributes to the progression of DA loss. In accordance with this, the clinical use of agents such as NA re-uptake inhibitors and α2-AR antagonists may prove useful in enhancing the endogenous neuroimmunomodulatory potential of NA in conditions associated with brain inflammation.
Collapse
|
37
|
Krivoy A, Hochman E, Sendt KV, Hollander S, Vilner Y, Selakovic M, Weizman A, Taler M. Association between serum levels of glutamate and neurotrophic factors and response to clozapine treatment. Schizophr Res 2018; 192:226-231. [PMID: 28599751 DOI: 10.1016/j.schres.2017.05.040] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 05/07/2017] [Accepted: 05/30/2017] [Indexed: 12/17/2022]
Abstract
Clozapine is the only available therapy for about 30% of schizophrenia patients otherwise refractory to antipsychotics. Unfortunately, the mechanism of action of the drug is still unknown and there are no biomarkers that can predict a positive response to clozapine. We aimed to examine serum neurotrophins and glutamate levels as putative biomarkers for clozapine response based on the hypothesized mode-of-action of the compound. Blood samples of 89 chronic schizophrenia patients maintained on clozapine were analyzed in a cross-sectional design. Serum brain derived neurotrophic factor (BDNF), vascular endothelial growth factor (VEGF), neurotrophic growth factor (NGF), glial derived neurotrophic factor (GDNF) and glutamate were determined. Differences between responders and non-responders to clozapine and correlation between clinical and biological measures were analyzed. Our sample consisted of 54 (61%) responders and 35 (39%) non-responders. Responders had higher mean BDNF levels than non-responders (2066±814 vs. 1668±820pg/ml, p<0.05. respectively) and higher serum glutamate levels (1.61±2.2 vs. 0.66±0.9pg/ml, respectively, p<0.05). Furthermore, there was a significant correlation between serum glutamate levels and positive symptoms among the clozapine-responder group (rho=0.47, p<0.005). High serum levels of BDNF and glutamate were associated with response to clozapine, while glutamate levels correlated with the psychosis severity in clozapine responders only. Large-scale, prospective longitudinal studies are needed to support these findings and the assumption that serum glutamate and BDNF can discriminate between clozapine responders and non-responders.
Collapse
Affiliation(s)
- Amir Krivoy
- Geha Mental Health Center, Petach-Tikva, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, Petach-Tikva, Israel; Department of Psychosis Studies, Institute Of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Eldar Hochman
- Geha Mental Health Center, Petach-Tikva, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, Petach-Tikva, Israel
| | - Kyra-Verena Sendt
- Department of Psychosis Studies, Institute Of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Sarah Hollander
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, Petach-Tikva, Israel
| | - Yael Vilner
- Geha Mental Health Center, Petach-Tikva, Israel
| | - Mirjana Selakovic
- Department of Psychiatry, Sismanoglio General Hospital, Athens, Greece
| | - Abraham Weizman
- Geha Mental Health Center, Petach-Tikva, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, Petach-Tikva, Israel
| | - Michal Taler
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, Petach-Tikva, Israel
| |
Collapse
|
38
|
Abstract
The neurotrophins are a family of closely related proteins that were first identified as survival factors for sympathetic and sensory neurons and have since been shown to control a number of aspects of survival, development, and function of neurons in both the central and peripheral nervous systems. Limiting quantities of neurotrophins during development control the numbers of surviving neurons to ensure a match between neurons and the requirement for a suitable density of target innervation. Biological effects of each of the four mammalian neurotrophins are mediated through activation of one or more of the three members of the tropomyosin-related kinase (Trk) family of receptor tyrosine kinases (TrkA, TrkB, and TrkC). In addition, all neurotrophins activate the p75 neurotrophin receptor (p75NTR), a member of the tumor necrosis factor receptor superfamily. Neurotrophin engagement of Trk receptors leads to activation of Ras, phosphatidylinositol 3-kinase, phospholipase C-γ1, and signaling pathways controlled through these proteins, including the mitogen-activated protein kinases. Neurotrophin availability is required into adulthood, where they control synaptic function and plasticity and sustain neuronal cell survival, morphology, and differentiation. This article will provide an overview of neurotrophin biology, their receptors, and signaling pathways.
Collapse
Affiliation(s)
- Stephen D Skaper
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy.
| |
Collapse
|
39
|
Hou L, Chen W, Liu X, Qiao D, Zhou FM. Exercise-Induced Neuroprotection of the Nigrostriatal Dopamine System in Parkinson's Disease. Front Aging Neurosci 2017; 9:358. [PMID: 29163139 PMCID: PMC5675869 DOI: 10.3389/fnagi.2017.00358] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 10/19/2017] [Indexed: 12/11/2022] Open
Abstract
Epidemiological studies indicate that physical activity and exercise may reduce the risk of developing Parkinson's disease (PD), and clinical observations suggest that physical exercise can reduce the motor symptoms in PD patients. In experimental animals, a profound observation is that exercise of appropriate timing, duration, and intensity can reduce toxin-induced lesion of the nigrostriatal dopamine (DA) system in animal PD models, although negative results have also been reported, potentially due to inappropriate timing and intensity of the exercise regimen. Exercise may also minimize DA denervation-induced medium spiny neuron (MSN) dendritic atrophy and other abnormalities such as enlarged corticostriatal synapse and abnormal MSN excitability and spiking activity. Taken together, epidemiological studies, clinical observations, and animal research indicate that appropriately dosed physical activity and exercise may not only reduce the risk of developing PD in vulnerable populations but also benefit PD patients by potentially protecting the residual DA neurons or directly restoring the dysfunctional cortico-basal ganglia motor control circuit, and these benefits may be mediated by exercise-triggered production of endogenous neuroprotective molecules such as neurotrophic factors. Thus, exercise is a universally available, side effect-free medicine that should be prescribed to vulnerable populations as a preventive measure and to PD patients as a component of treatment. Future research needs to establish standardized exercise protocols that can reliably induce DA neuron protection, enabling the delineation of the underlying cellular and molecular mechanisms that in turn can maximize exercise-induced neuroprotection and neurorestoration in animal PD models and eventually in PD patients.
Collapse
Affiliation(s)
- Lijuan Hou
- Exercise Physiology Laboratory, College of Physical Education and Sports, Beijing Normal University, Beijing, China
| | - Wei Chen
- Exercise Physiology Laboratory, College of Physical Education and Sports, Beijing Normal University, Beijing, China.,Department of Exercise and Rehabilitation, Physical Education College, Hebei Normal University, Shijiazhuang, China
| | - Xiaoli Liu
- Exercise Physiology Laboratory, College of Physical Education and Sports, Beijing Normal University, Beijing, China
| | - Decai Qiao
- Exercise Physiology Laboratory, College of Physical Education and Sports, Beijing Normal University, Beijing, China
| | - Fu-Ming Zhou
- Department of Pharmacology, University of Tennessee College of Medicine, Memphis, TN, United States
| |
Collapse
|
40
|
Contrasting effects of selective MAGL and FAAH inhibition on dopamine depletion and GDNF expression in a chronic MPTP mouse model of Parkinson's disease. Neurochem Int 2017; 110:14-24. [PMID: 28826718 DOI: 10.1016/j.neuint.2017.08.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 06/08/2017] [Accepted: 08/03/2017] [Indexed: 01/07/2023]
Abstract
The modulation of the brain endocannabinoid system has been identified as an option to treat neurodegenerative diseases including Parkinson's disease (PD). Especially the elevation of endocannabinoid levels by inhibition of hydrolytic degradation represents a valuable approach. To evaluate whether monoacylglycerol lipase (MAGL) or fatty acid amide hydrolase (FAAH) inhibition could be beneficial for PD, we examined in parallel the therapeutic potential of the highly selective MAGL inhibitor KML29 elevating 2-arachidonoylglyerol (2-AG) levels and the highly selective FAAH inhibitor PF-3845 elevating anandamide (AEA) levels in a chronic methyl-4-phenyl-1,2,3,6-tetrahydropyridine/probenecid (MPTP/probenecid) mouse model of PD. Chronic administration of KML29 (10 mg/kg) but not PF-3845 (10 mg/kg) attenuated striatal MPTP/probenecid-induced dopamine depletion. Furthermore, KML29 induced an increase in Gdnf but not Bdnf expression, whereas PF-3845 decreased the MPTP/probenecid-induced Cnr2 expression without any effects on neurotrophin expression. Investigation of treatment-naïve striatal mRNA levels revealed a high presence of Gdnf and Mgll in contrast to Bdnf and Faah. Treatment of primary mouse microglia with 2-AG increased Gdnf but not Bdnf expression, suggesting that microglia might mediate the observed KML29-induced increase in Gdnf. In summary, pharmacological MAGL but not FAAH inhibition in the chronic MPTP/probenecid model attenuated the MPTP/probenecid-induced effects on striatal dopamine levels which were accompanied by an increase in 2-AG levels.
Collapse
|
41
|
Mcgrath J, Lintz E, Hoffer BJ, Gerhardt GA, Quintero EM, Granholm AC. Adeno-Associated Viral Delivery of GDNF Promotes Recovery of Dopaminergic Phenotype following a Unilateral 6-Hydroxydopamine Lesion. Cell Transplant 2017. [DOI: 10.3727/096020198389988] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is a potent neurotrophic factor for dopamine neurons that has been proposed for use in the treatment of Parkinson's disease (PD). Previous studies using viral vectors to deliver GDNF in rodent models of PD have entailed administering the virus either prior to or immediately after neurotoxin-induced lesions, when the nigrostriatal pathway is largely intact, a paradigm that does not accurately reflect the clinical situation encountered with Parkinson's patients. In this study, recombinant adeno-associated virus carrying the gene encoding GDNF (rAAV-GDNF) was administered to animals bearing a maximal lesion in the nigrostriatal system, more closely resembling fully developed PD. Rats were treated with 6-hydroxydopamine into the medial forebrain bundle and assessed by apomorphine-induced rotational behavior for 5 weeks prior to virus administration. Within 4 weeks of a single intrastriatal injection of rAAV-GDNF, unilaterally lesioned animals exhibited significant behavioral recovery, which correlated with increased expression of dopaminergic markers in the substantia nigra, the medial forebrain bundle, and the striatum. Our findings demonstrate that rAAV-GDNF is capable of rescuing adult dopaminergic neurons from near complete phenotypic loss following a neurotoxic lesion, effectively restoring a functional dopaminergic pathway and diminishing motoric deficits. These data provide further support for the therapeutic potential of rAAV-GDNF-based gene therapy in the treatment of PD.
Collapse
Affiliation(s)
| | - Elishia Lintz
- University of Colorado Health Science Center, Denver, CO
| | - Barry J. Hoffer
- Intramural Research Program National Institute on Drug Abuse, Baltimore, MD
| | - Greg A. Gerhardt
- Departments of Anatomy & Neurobiology, and Neurology, and the Morris K. Udall Parkinson's Disease Research Center of Excellence, and the Center for Sensor Technology, University of Kentucky, Chandler Medical Center, Lexington, KY
| | - E. Matthew Quintero
- ¶Department of Physiology and Neuroscience, Medical University of South Carolina, Charleston, SC
| | - Ann-Charlotte Granholm
- ¶Department of Physiology and Neuroscience, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
42
|
Dall AM, Danielsen EH, Sørensen JC, Andersen F, Møller A, Zimmer J, Gjedde AH, Cumming P, Zimmer J, Brevig T, Dall AM, Meyer M, Pedersen EB, Gjedde A, Danielsen EH, Cumming P, Andersen F, Bender D, Falborg L, Gee A, Gillings NM, Hansen SB, Hermansen F, Jørgensen HA, Munk O, Poulsen PH, Rodell AB, Sakoh M, Simonsen CZ, Smith DF, Sørensen JC, Østergård L, Moller A, Johansen TE. Quantitative [18F]Fluorodopa/PET and Histology of Fetal Mesencephalic Dopaminergic Grafts to the Striatum of MPTP-Poisoned Minipigs. Cell Transplant 2017. [DOI: 10.3727/000000002783985314] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The functional restoration of the dopamine innervation of striatum in MPTP-poisoned Göttingen minipigs was assessed for 6 months following grafting of fetal pig mesencephalic neurons. Pigs were assigned to a normal control group and a MPTP-poisoned group, members of which received no further treatment, or which received bilateral grafts to the striatum of tissue blocks harvested from E28 fetal pig mesencephalon with and without immunosuppressive treatment after grafting, or with additional co-grafting with immortalized rat neural cells transfected to produce GDNF. In the baseline condition, and again at 3 and 6 months postsurgery, all animals were subjected to quantitative [18F]fluorodopa PET scans and testing for motor impairment. At the end of 6 months, tyrosine hydroxylase (TH)-containing neurons were counted in the grafts by stereological methods. The MPTP poisoning persistently reduced the magnitude of k3D, the relative activity of DOPA decarboxylase in striatum, by 60%. Grafting restored the rate of [18F]fluorodopa decarboxylation to the normal range, and normalized the scores in motor function. The biochemical and functional recovery was associated with survival of approximately 100,000 TH-positive graft neurons in each hemisphere. Immunosuppression did not impart a greater recovery of [18F]fluorodopa uptake, nor were the number of TH-positive graft neurons or the volumes of the grafts increased in the immunosuppressed group. Contrary to expectation, co-grafting of transfected GDNF-expressing HiB5 cells, a rat-derived neural cell line, tended to impair the survival of the grafts with the lowest values for graft volumes, TH-positive cell numbers, behavioral scores, and relative DOPA decarboxylase activity. From the results we conclude that pig ventral mesencephalic allografts can restore functional dopamine innervation in adult MPTP-lesioned minipigs.
Collapse
Affiliation(s)
- Annette Møller Dall
- Department of Anatomy and Neurobiology, University of Southern Denmark, 5000 Odense C, Denmark
| | | | | | | | | | - Jens Zimmer
- Department of Anatomy and Neurobiology, University of Southern Denmark, 5000 Odense C, Denmark
| | - Albert H. Gjedde
- Department of Anatomy and Neurobiology, University of Southern Denmark, 5000 Odense C, Denmark
- McGill University, Montreal, Quebec, Canada
| | - Paul Cumming
- PET Centre, Aarhus General Hospital, 8000 Aarhus C, Denmark
| | - J. Zimmer
- Department of Anatomy and Neurobiology, SDU Odense University
| | - T. Brevig
- Department of Anatomy and Neurobiology, SDU Odense University
| | - A. M. Dall
- Department of Anatomy and Neurobiology, SDU Odense University
| | - M. Meyer
- Department of Anatomy and Neurobiology, SDU Odense University
| | - E. B. Pedersen
- Department of Anatomy and Neurobiology, SDU Odense University
| | - A. Gjedde
- PET-Center and Departments of Neuroradiology, Neurosurgery, Neuroanaesthesia, and Biological Psychiatry, Aarhus University Hospital
| | - E. H. Danielsen
- PET-Center and Departments of Neuroradiology, Neurosurgery, Neuroanaesthesia, and Biological Psychiatry, Aarhus University Hospital
| | - P. Cumming
- PET-Center and Departments of Neuroradiology, Neurosurgery, Neuroanaesthesia, and Biological Psychiatry, Aarhus University Hospital
| | - F. Andersen
- PET-Center and Departments of Neuroradiology, Neurosurgery, Neuroanaesthesia, and Biological Psychiatry, Aarhus University Hospital
| | - D. Bender
- PET-Center and Departments of Neuroradiology, Neurosurgery, Neuroanaesthesia, and Biological Psychiatry, Aarhus University Hospital
| | - L. Falborg
- PET-Center and Departments of Neuroradiology, Neurosurgery, Neuroanaesthesia, and Biological Psychiatry, Aarhus University Hospital
| | - A. Gee
- PET-Center and Departments of Neuroradiology, Neurosurgery, Neuroanaesthesia, and Biological Psychiatry, Aarhus University Hospital
| | - N. M. Gillings
- PET-Center and Departments of Neuroradiology, Neurosurgery, Neuroanaesthesia, and Biological Psychiatry, Aarhus University Hospital
| | - S. B. Hansen
- PET-Center and Departments of Neuroradiology, Neurosurgery, Neuroanaesthesia, and Biological Psychiatry, Aarhus University Hospital
| | - F. Hermansen
- PET-Center and Departments of Neuroradiology, Neurosurgery, Neuroanaesthesia, and Biological Psychiatry, Aarhus University Hospital
| | - H. A. Jørgensen
- PET-Center and Departments of Neuroradiology, Neurosurgery, Neuroanaesthesia, and Biological Psychiatry, Aarhus University Hospital
| | - O. Munk
- PET-Center and Departments of Neuroradiology, Neurosurgery, Neuroanaesthesia, and Biological Psychiatry, Aarhus University Hospital
| | - P. H. Poulsen
- PET-Center and Departments of Neuroradiology, Neurosurgery, Neuroanaesthesia, and Biological Psychiatry, Aarhus University Hospital
| | - A. B. Rodell
- PET-Center and Departments of Neuroradiology, Neurosurgery, Neuroanaesthesia, and Biological Psychiatry, Aarhus University Hospital
| | - M. Sakoh
- PET-Center and Departments of Neuroradiology, Neurosurgery, Neuroanaesthesia, and Biological Psychiatry, Aarhus University Hospital
| | - C. Z. Simonsen
- PET-Center and Departments of Neuroradiology, Neurosurgery, Neuroanaesthesia, and Biological Psychiatry, Aarhus University Hospital
| | - D. F. Smith
- PET-Center and Departments of Neuroradiology, Neurosurgery, Neuroanaesthesia, and Biological Psychiatry, Aarhus University Hospital
| | - J. C. Sørensen
- PET-Center and Departments of Neuroradiology, Neurosurgery, Neuroanaesthesia, and Biological Psychiatry, Aarhus University Hospital
| | - L. Østergård
- PET-Center and Departments of Neuroradiology, Neurosurgery, Neuroanaesthesia, and Biological Psychiatry, Aarhus University Hospital
| | | | | | | |
Collapse
|
43
|
Freed WJ. Article Commentary: A Perspective on Transplantation Therapy and Stem Cells for Parkinson's Disease. Cell Transplant 2017; 13:319-327. [DOI: 10.3727/000000004783984025] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- William J. Freed
- Cellular Neurobiology Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, MD, 21224
| |
Collapse
|
44
|
Zigova T, Pencea V, Betarbet R, Wiegand SJ, Alexander C, Bakay RA, Luskin MB. Neuronal Progenitor Cells of the Neonatal Subventricular Zone Differentiate and Disperse following Transplantation into the Adult Rat Striatum. Cell Transplant 2017; 7:137-56. [PMID: 9588596 DOI: 10.1177/096368979800700209] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
We have investigated the suitability of a recently identified and characterized population of neuronal progenitor cells for their potential use in the replacement of degenerating or damaged neurons in the mammalian brain. The unique population of neuronal progenitor cells is situated in a well-delineated region of the anterior part of the neonatal subventricular zone (referred to as SVZa). This region can be separated from the remaining proliferative, gliogenic, subventricular zone encircling the lateral ventricles of the forebrain. Because the neurons arising from the highly enriched neurogenic progenitor cell population of the SVZa ordinarily migrate considerable distances and ultimately express the neurotransmitters GABA and dopamine, we have examined whether they could serve as an alternative source of tissue for neural transplantation. SVZa cells from postnatal day 0-2 rats, prelabeled by intraperitoneal injections of the cell proliferation marker BrdU, were implanted into the striatum of adult rats approximately 1 mo after unilateral denervation by 6-OHDA. To examine the spatio-temporal distribution and phenotype of the transplanted SVZa cells, the experimental recipients were perfused at short (less than 1 wk), intermediate (2-3 wk) and long (5 mo) postimplantation times. The host brains were sectioned and stained with an antibody to BrdU and one of several cell-type specific markers to determine the phenotypic characteristics of the transplanted SVZa cells. To identify neurons we used the neuron-specific antibody TuJ1, or antimembrane-associated protein 2 (MAP-2), and anti-GFAP was used to identify astrocytic glia. At all studied intervals the majority of the surviving SVZa cells exhibited a neuronal phenotype. Moreover, morphologically they could be distinguished from the cells of the host striatum because they resembled the intrinsic granule cells of the olfactory bulb, their usual fate. At longer times, a greater number of the transplanted SVZa cells had migrated from their site of implantation, often towards an outlying blood vessel, and the density of cells within the core of the transplant was reduced. Furthermore, there were rarely signs of transplant rejection or a glial scar surrounding the transplant. In the core of the transplant there were low numbers of GFAP-positive cells, indicating that the transplanted SVZa cells, predominantly TuJ1-positive/MAP2-positive, express a neuronal phenotype. Collectively, the propensity of the SVZa cells to express a neuronal phenotype and to survive and integrate in the striatal environment suggest that they may be useful in the reconstruction of the brain following CNS injury or disease.
Collapse
Affiliation(s)
- T Zigova
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Weis C, Kaufmann WA, Humpel C. Lipid-Mediated In Vivo Gene Transfer Replaces the Loss of Choline Acetyltransferase Activity after Unilateral Fimbria-Fornix Aspiration. Cell Transplant 2017. [DOI: 10.3727/000000001783986233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Carla Weis
- Laboratory of Psychiatry, Clinic of Psychiatry, University Hospital Innsbruck, Austria
| | - Walter A. Kaufmann
- Laboratory of Psychiatry, Clinic of Psychiatry, University Hospital Innsbruck, Austria
| | - Christian Humpel
- Laboratory of Psychiatry, Clinic of Psychiatry, University Hospital Innsbruck, Austria
| |
Collapse
|
46
|
Tenenbaum L, Humbert-Claude M. Glial Cell Line-Derived Neurotrophic Factor Gene Delivery in Parkinson's Disease: A Delicate Balance between Neuroprotection, Trophic Effects, and Unwanted Compensatory Mechanisms. Front Neuroanat 2017; 11:29. [PMID: 28442998 PMCID: PMC5385337 DOI: 10.3389/fnana.2017.00029] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 03/23/2017] [Indexed: 12/14/2022] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) and Neurturin (NRTN) bind to a receptor complex consisting of a member of the GDNF family receptor (GFR)-α and the Ret tyrosine kinase. Both factors were shown to protect nigro-striatal dopaminergic neurons and reduce motor symptoms when applied terminally in toxin-induced Parkinson's disease (PD) models. However, clinical trials based on intraputaminal GDNF protein administration or recombinant adeno-associated virus (rAAV)-mediated NRTN gene delivery have been disappointing. In this review, several factors that could have limited the clinical benefits are discussed. Retrograde transport of GDNF/NRTN to the dopaminergic neurons soma is thought to be necessary for NRTN/GFR-α/Ret signaling mediating the pro-survival effect. Therefore, the feasibility of treating advanced patients with neurotrophic factors is questioned by recent data showing that: (i) tyrosine hydroxylase-positive putaminal innervation has almost completely disappeared at 5 years post-diagnosis and (ii) in patients enrolled in the rAAV-NRTN trial more than 5 years post-diagnosis, NRTN was almost not transported to the substantia nigra pars compacta. In addition to its anti-apoptotic and neurotrophic properties, GDNF also interferes with dopamine homeostasis via time and dose-dependent effects such as: stimulation of dopamine neuron excitability, inhibition of dopamine transporter activity, tyrosine hydroxylase phosphorylation, and inhibition of tyrosine hydroxylase transcription. Depending on the delivery parameters, the net result of this intricate network of regulations could be either beneficial or deleterious. In conclusion, further unraveling of the mechanism of action of GDNF gene delivery in relevant animal models is still needed to optimize the clinical benefits of this new therapeutic approach. Recent developments in the design of regulated viral vectors will allow to finely adjust the GDNF dose and period of administration. Finally, new clinical studies in less advanced patients are warranted to evaluate the potential of AAV-mediated neurotrophic factors gene delivery in PD. These will be facilitated by the demonstration of the safety of rAAV administration into the human brain.
Collapse
Affiliation(s)
- Liliane Tenenbaum
- Laboratory of Cellular and Molecular Neurotherapies, Clinical Neuroscience Department, Center for Neuroscience Research, Lausanne University HospitalLausanne, Switzerland
| | - Marie Humbert-Claude
- Laboratory of Cellular and Molecular Neurotherapies, Clinical Neuroscience Department, Center for Neuroscience Research, Lausanne University HospitalLausanne, Switzerland
| |
Collapse
|
47
|
Zuo F, Xiong F, Wang X, Li X, Wang R, Ge W, Bao X. Intrastriatal Transplantation of Human Neural Stem Cells Restores the Impaired Subventricular Zone in Parkinsonian Mice. Stem Cells 2017; 35:1519-1531. [PMID: 28328168 DOI: 10.1002/stem.2616] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 02/17/2017] [Accepted: 02/26/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Fuxing Zuo
- Department of Neurosurgery; Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing China
| | - Feng Xiong
- State Key Laboratory of Medical Molecular Biology & Department of Immunology; Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; Beijing China
| | - Xia Wang
- State Key Laboratory of Medical Molecular Biology & Department of Immunology; Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; Beijing China
| | - Xueyuan Li
- Department of Neurosurgery; Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing China
| | - Renzhi Wang
- Department of Neurosurgery; Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing China
| | - Wei Ge
- State Key Laboratory of Medical Molecular Biology & Department of Immunology; Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; Beijing China
| | - Xinjie Bao
- Department of Neurosurgery; Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; Beijing China
| |
Collapse
|
48
|
Quantitative analysis of the therapeutic effect of magnolol on MPTP-induced mouse model of Parkinson's disease using in vivo 18F-9-fluoropropyl-(+)-dihydrotetrabenazine PET imaging. PLoS One 2017; 12:e0173503. [PMID: 28257461 PMCID: PMC5336287 DOI: 10.1371/journal.pone.0173503] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 02/21/2017] [Indexed: 02/04/2023] Open
Abstract
18F-9-Fluoropropyl-(+)-dihydrotetrabenazine [18F-FP-(+)-DTBZ] positron emission tomography (PET) has been shown to detect dopaminergic neuron loss associated with Parkinson’s disease (PD) in human and neurotoxin-induced animal models. A polyphenol compound, magnolol, was recently proposed as having a potentially restorative effect in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)- or 6-hydroxydopamine-treated animal models. In this study, 18F-FP-(+)-DTBZ PET was used to determine the therapeutic efficacy of magnolol in an MPTP–PD mouse model that was prepared by giving an intraperitoneally (i.p.) daily dose of 25 mg/kg MPTP to male C57BL/6 mice for 5 consecutive days. Twenty-minute static 18F-FP-(+)-DTBZ PET scans were performed before MPTP treatment and 5 days after the termination of MPTP treatment to set up the baseline control. Half of the MPTP-treated mice then received a daily dose of magnolol (10 mg/kg dissolved in corn oil, i.p.) for 6 days. 18F-FP-(+)-DTBZ PET imaging was performed the day after the final treatment. All 18F-FP-(+)-DTBZ PET images were analysed and the specific uptake ratio (SUr) was calculated. Ex vivo autoradiography (ARG) and corresponding immunohistochemistry (IHC) studies were conducted to confirm the distribution of dopaminergic terminals in the striatum. The striatal SUr ratios of 18F-FP-(+)-DTBZ PET images for the Sham, the MPTP, and the MPTP + Magnolol-treated groups were 1.25 ± 0.05, 0.75 ± 0.06, and 1.00 ± 0.11, respectively (n = 4 for each group). The ex vivo 18F-FP-(+)-DTBZ ARG and IHC results correlated favourably with the PET imaging results. 18F-FP-(+)-DTBZ PET imaging suggested that magnolol post-treatment may reverse the neuronal damage in the MPTP-lesioned PD mice. In vivo imaging of the striatal vesicular monoamine transporter type 2 (VMAT2) distribution using 18F-FP-(+)-DTBZ animal PET is a useful method to evaluate the efficacy of therapeutic drugs i.e., magnolol, for the management of PD.
Collapse
|
49
|
Differential behavioral outcomes following neonatal versus fetal human retinal pigment epithelial cell striatal implants in parkinsonian rats. J Neural Transm (Vienna) 2017; 124:455-462. [PMID: 28160153 DOI: 10.1007/s00702-017-1683-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 01/16/2017] [Indexed: 02/04/2023]
Abstract
Following the failure of a Phase II clinical study evaluating human retinal pigment epithelial (hRPE) cell implants as a potential treatment option for Parkinson's disease, speculation has centered on implant function and survival as possible contributors to the therapeutic outcomes. We recently reported that neonatal hRPE cells, similar to hRPE cells used in the Phase II clinical study, produced short-lived in vitro and limited in vivo trophic factors, which supports that assumption. We hypothesize that the switch from fetal to neonatal hRPE cells, between the Phase I and the Phase II clinical trial may be partly responsible for the later negative outcomes. To investigate this hypothesis, we used two neonatal hRPE cell lots, prepared in a similar manner to neonatal hRPE cells used in the Phase II clinical study, and compared them to previously evaluated fetal hRPE cells for behavioral changes following unilateral striatal implantation in 6-hydroxydopamine-lesioned rats. The results showed that only fetal, not neonatal, hRPE cell implants, were able to improve behavioral outcomes following striatal implantation in the lesioned rats. These data suggest that fetal hRPE cells may be preferential to neonatal hRPE cells in restoring behavioral deficits.
Collapse
|
50
|
Destination Brain: the Past, Present, and Future of Therapeutic Gene Delivery. J Neuroimmune Pharmacol 2017; 12:51-83. [PMID: 28160121 DOI: 10.1007/s11481-016-9724-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 12/12/2016] [Indexed: 12/20/2022]
Abstract
Neurological diseases and disorders (NDDs) present a significant societal burden and currently available drug- and biological-based therapeutic strategies have proven inadequate to alleviate it. Gene therapy is a suitable alternative to treat NDDs compared to conventional systems since it can be tailored to specifically alter select gene expression, reverse disease phenotype and restore normal function. The scope of gene therapy has broadened over the years with the advent of RNA interference and genome editing technologies. Consequently, encouraging results from central nervous system (CNS)-targeted gene delivery studies have led to their transition from preclinical to clinical trials. As we shift to an exciting gene therapy era, a retrospective of available literature on CNS-associated gene delivery is in order. This review is timely in this regard, since it analyzes key challenges and major findings from the last two decades and evaluates future prospects of brain gene delivery. We emphasize major areas consisting of physiological and pharmacological challenges in gene therapy, function-based selection of a ideal cellular target(s), available therapy modalities, and diversity of viral vectors and nanoparticles as vehicle systems. Further, we present plausible answers to key questions such as strategies to circumvent low blood-brain barrier permeability and most suitable CNS cell types for targeting. We compare and contrast pros and cons of the tested viral vectors in the context of delivery systems used in past and current clinical trials. Gene vector design challenges are also evaluated in the context of cell-specific promoters. Key challenges and findings reported for recent gene therapy clinical trials, assessing viral vectors and nanoparticles are discussed from the perspective of bench to bedside gene therapy translation. We conclude this review by tying together gene delivery challenges, available vehicle systems and comprehensive analyses of neuropathogenesis to outline future prospects of CNS-targeted gene therapies.
Collapse
|