1
|
Narasimhan K, Hakami A, Comini G, Patton T, Newland B, Dowd E. Cryogel microcarriers loaded with glial cell line-derived neurotrophic factor enhance the engraftment of primary dopaminergic neurons in a rat model of Parkinson's disease. J Neural Eng 2024; 21:056011. [PMID: 39231475 DOI: 10.1088/1741-2552/ad7761] [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: 06/13/2024] [Accepted: 09/04/2024] [Indexed: 09/06/2024]
Abstract
Objective.Cryogel microcarriers made of poly(ethylene glycol) diacrylate and 3-sulfopropyl acrylate have the potential to act as delivery vehicles for long-term retention of neurotrophic factors (NTFs) in the brain. In addition, they can potentially enhance stem cell-derived dopaminergic (DAergic) cell replacement strategies for Parkinson's disease (PD), by addressing the limitations of variable survival and poor differentiation of the transplanted precursors due to neurotrophic deprivation post-transplantation in the brain. In this context, to develop a proof-of-concept, the aim of this study was to determine the efficacy of glial cell line-derived NTF (GDNF)-loaded cryogel microcarriers by assessing their impact on the survival of, and reinnervation by, primary DAergic grafts after intra-striatal delivery in Parkinsonian rat brains.Approach.Rat embryonic day 14 ventral midbrain cells were transplanted into the 6-hydroxydopamine-lesioned striatum either alone, or with GDNF, or with unloaded cryogel microcarriers, or with GDNF-loaded cryogel microcarriers.Post-mortem, GDNF and tyrosine hydroxylase immunostaining were used to identify retention of the delivered GDNF within the implanted cryogel microcarriers, and to identify the transplanted DAergic neuronal cell bodies and fibres in the brains, respectively.Main results.We found an intact presence of GDNF-stained cryogel microcarriers in graft sites, indicating their ability for long-term retention of the delivered GDNF up to 4 weeks in the brain. This resulted in an enhanced survival (1.9-fold) of, and striatal reinnervation (density & volume) by, the grafted DAergic neurons, in addition to an enhanced sprouting of fibres within graft sites.Significance.This data provides an important proof-of-principle for the beneficial effects of neurotrophin-loaded cryogel microcarriers on engraftment of cells in the context of cell replacement therapy in PD. For clinical translation, further studies will be needed to assess the impact of cryogel microcarriers on the survival and differentiation of stem cell-derived DAergic precursors in Parkinsonian rat brains.
Collapse
Affiliation(s)
- Kaushik Narasimhan
- Pharmacology & Therapeutics and Galway Neuroscience Centre, University of Galway, Galway, Ireland
| | - Abrar Hakami
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, United Kingdom
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Giulia Comini
- Pharmacology & Therapeutics and Galway Neuroscience Centre, University of Galway, Galway, Ireland
| | - Tommy Patton
- Pharmacology & Therapeutics and Galway Neuroscience Centre, University of Galway, Galway, Ireland
| | - Ben Newland
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, United Kingdom
- Leibniz-Institut für Polymerforschung Dresden e.V., 01069 Dresden, Germany
| | - Eilís Dowd
- Pharmacology & Therapeutics and Galway Neuroscience Centre, University of Galway, Galway, Ireland
| |
Collapse
|
2
|
The promise of the TGF-β superfamily as a therapeutic target for Parkinson's disease. Neurobiol Dis 2022; 171:105805. [PMID: 35764291 DOI: 10.1016/j.nbd.2022.105805] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 11/20/2022] Open
Abstract
A large body of evidence underscore the regulatory role of TGF-β superfamily in the central nervous system. Components of the TGF-β superfamily modulate key events during embryonic brain development and adult brain tissue injury repair. With respect to Parkinson's disease (PD), TGF-ß signaling pathways are implicated in the differentiation, maintenance and synaptic function of the dopaminergic neurons, as well as in processes related to the activation state of astrocytes and microglia. In vitro and in vivo studies using toxin models, have interrogated on the dopaminotrophic and protective role of the TGF-β superfamily members. The evolution of genetic and animal models of PD that more closely recapitulate the disease condition has made possible the dissection of intracellular pathways in response to TGF-ß treatment. Although the first clinical trials using GDNF did not meet their primary endpoints, substantial work has been carried out to reappraise the TGF-β superfamily's clinical benefit.
Collapse
|
3
|
Anantha J, Goulding SR, Tuboly E, O'Mahony AG, Moloney GM, Lomansey G, McCarthy CM, Collins LM, Sullivan AM, O'Keeffe GW. NME1 Protects Against Neurotoxin-, α-Synuclein- and LRRK2-Induced Neurite Degeneration in Cell Models of Parkinson's Disease. Mol Neurobiol 2022; 59:61-76. [PMID: 34623600 PMCID: PMC8786793 DOI: 10.1007/s12035-021-02569-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 09/14/2021] [Indexed: 11/26/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterised by the progressive degeneration of midbrain dopaminergic neurons, coupled with the intracellular accumulation of α-synuclein. Axonal degeneration is a central part of the pathology of PD. While the majority of PD cases are sporadic, some are genetic; the G2019S mutation in leucine-rich repeat kinase 2 (LRRK2) is the most common genetic form. The application of neurotrophic factors to protect dopaminergic neurons is a proposed experimental therapy. One such neurotrophic factor is growth differentiation factor (GDF)5. GDF5 is a dopaminergic neurotrophic factor that has been shown to upregulate the expression of a protein called nucleoside diphosphate kinase A (NME1). However, whether NME1 is neuroprotective in cell models of axonal degeneration of relevance to PD is unknown. Here we show that treatment with NME1 can promote neurite growth in SH-SY5Y cells, and in cultured dopaminergic neurons treated with the neurotoxin 6-hydroxydopamine (6-OHDA). Similar effects of NME1 were found in SH-SY5Y cells and dopaminergic neurons overexpressing human wild-type α-synuclein, and in stable SH-SY5Y cell lines carrying the G2019S LRRK2 mutation. We found that the effects of NME1 require the RORα/ROR2 receptors. Furthermore, increased NF-κB-dependent transcription was partially required for the neurite growth-promoting effects of NME1. Finally, a combined bioinformatics and biochemical analysis of the mitochondrial oxygen consumption rate revealed that NME1 enhanced mitochondrial function, which is known to be impaired in PD. These data show that recombinant NME1 is worthy of further study as a potential therapeutic agent for axonal protection in PD.
Collapse
Affiliation(s)
- Jayanth Anantha
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Susan R Goulding
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Eszter Tuboly
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Adam G O'Mahony
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Gerard M Moloney
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Gareth Lomansey
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Cathal M McCarthy
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Louise M Collins
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- Department of Physiology, University College Cork, Cork, Ireland
- Parkinson's Disease Research Cluster (PDRC), University College Cork, Cork, Ireland
| | - Aideen M Sullivan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
- Parkinson's Disease Research Cluster (PDRC), University College Cork, Cork, Ireland.
| | - Gerard W O'Keeffe
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
- Parkinson's Disease Research Cluster (PDRC), University College Cork, Cork, Ireland.
| |
Collapse
|
4
|
Goulding SR, Anantha J, Collins LM, Sullivan AM, O'Keeffe GW. Growth differentiation factor 5: a neurotrophic factor with neuroprotective potential in Parkinson's disease. Neural Regen Res 2022; 17:38-44. [PMID: 34100424 PMCID: PMC8451580 DOI: 10.4103/1673-5374.314290] [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: 11/25/2020] [Revised: 01/08/2021] [Accepted: 01/24/2021] [Indexed: 11/04/2022] Open
Abstract
Parkinson's disease is the most common movement disorder worldwide, affecting over 6 million people. It is an age-related disease, occurring in 1% of people over the age of 60, and 3% of the population over 80 years. The disease is characterized by the progressive loss of midbrain dopaminergic neurons from the substantia nigra, and their axons, which innervate the striatum, resulting in the characteristic motor and non-motor symptoms of Parkinson's disease. This is paralleled by the intracellular accumulation of α-synuclein in several regions of the nervous system. Current therapies are solely symptomatic and do not stop or slow disease progression. One promising disease-modifying strategy to arrest the loss of dopaminergic neurons is the targeted delivery of neurotrophic factors to the substantia nigra or striatum, to protect the remaining dopaminergic neurons of the nigrostriatal pathway. However, clinical trials of two well-established neurotrophic factors, glial cell line-derived neurotrophic factor and neurturin, have failed to meet their primary end-points. This failure is thought to be at least partly due to the downregulation by α-synuclein of Ret, the common co-receptor of glial cell line-derived neurorophic factor and neurturin. Growth/differentiation factor 5 is a member of the bone morphogenetic protein family of neurotrophic factors, that signals through the Ret-independent canonical Smad signaling pathway. Here, we review the evidence for the neurotrophic potential of growth/differentiation factor 5 in in vitro and in vivo models of Parkinson's disease. We discuss new work on growth/differentiation factor 5's mechanisms of action, as well as data showing that viral delivery of growth/differentiation factor 5 to the substantia nigra is neuroprotective in the α-synuclein rat model of Parkinson's disease. These data highlight the potential for growth/differentiation factor 5 as a disease-modifying therapy for Parkinson's disease.
Collapse
Affiliation(s)
- Susan R. Goulding
- Department of Anatomy and Neuroscience, and Cork Neuroscience Centre, University College Cork, Cork, Ireland
| | - Jayanth Anantha
- Department of Anatomy and Neuroscience, and Cork Neuroscience Centre, 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
| | - Aideen M. Sullivan
- Department of Anatomy and Neuroscience, and Cork Neuroscience Centre, University College Cork, Cork, Ireland
| | - Gerard W. O'Keeffe
- Department of Anatomy and Neuroscience, and Cork Neuroscience Centre, University College Cork, Cork, Ireland
| |
Collapse
|
5
|
Goulding SR, Concannon RM, Morales-Prieto N, Villalobos-Manriquez F, Clarke G, Collins LM, Lévesque M, Wyatt SL, Sullivan AM, O'Keeffe GW. Growth differentiation factor 5 exerts neuroprotection in an α-synuclein rat model of Parkinson's disease. Brain 2021; 144:e14. [PMID: 33253375 DOI: 10.1093/brain/awaa367] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Affiliation(s)
- Susan R Goulding
- Department of Anatomy and Neuroscience, University College Cork (UCC), Cork, Ireland.,Department of Biological Sciences, Cork Institute of Technology (CIT), Cork, Ireland
| | - Ruth M Concannon
- Department of Anatomy and Neuroscience, University College Cork (UCC), Cork, Ireland
| | - Noelia Morales-Prieto
- Department of Anatomy and Neuroscience, University College Cork (UCC), Cork, Ireland
| | | | - Gerard Clarke
- Department of Psychiatry and Neurobehavioural Sciences, UCC, Cork, Ireland.,APC Microbiome Ireland, UCC, Cork, Ireland.,Cork Neuroscience Centre, UCC, Cork, Ireland
| | - Louise M Collins
- Department of Anatomy and Neuroscience, University College Cork (UCC), Cork, Ireland
| | - Martin Lévesque
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Sean L Wyatt
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, UK
| | - Aideen M Sullivan
- Department of Anatomy and Neuroscience, University College Cork (UCC), Cork, Ireland.,APC Microbiome Ireland, UCC, Cork, Ireland.,Cork Neuroscience Centre, UCC, Cork, Ireland
| | - Gerard W O'Keeffe
- Department of Anatomy and Neuroscience, University College Cork (UCC), Cork, Ireland.,APC Microbiome Ireland, UCC, Cork, Ireland.,Cork Neuroscience Centre, UCC, Cork, Ireland
| |
Collapse
|
6
|
Anantha J, Goulding SR, Wyatt SL, Concannon RM, Collins LM, Sullivan AM, O'Keeffe GW. STRAP and NME1 Mediate the Neurite Growth-Promoting Effects of the Neurotrophic Factor GDF5. iScience 2020; 23:101457. [PMID: 32853992 PMCID: PMC7452236 DOI: 10.1016/j.isci.2020.101457] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/17/2020] [Accepted: 08/10/2020] [Indexed: 12/23/2022] Open
Abstract
Loss of midbrain dopaminergic (mDA) neurons and their axons is central to Parkinson's disease (PD). Growth differentiation factor (GDF)5 is a potential neurotrophic factor for PD therapy. However, the molecular mediators of its neurotrophic action are unknown. Our proteomics analysis shows that GDF5 increases the expression of serine threonine receptor-associated protein kinase (STRAP) and nucleoside diphosphate kinase (NME)1 in the SH-SY5Y neuronal cell line. GDF5 overexpression increased NME1 expression in adult rat brain in vivo. NME and STRAP mRNAs are expressed in developing and adult rodent midbrain. Expression of both STRAP and NME1 is necessary and sufficient for the promotion of neurite growth in SH-SY5Y cells by GDF5. NME1 treatment increased neurite growth in both SH-SY5Y cells and cultured mDA neurons. Expression patterns of NME and STRAP are altered in PD midbrain. NME1 and STRAP are thus key mediators of GDF5's neurotrophic effects, rationalizing their future study as therapeutic targets for PD.
Collapse
Affiliation(s)
- Jayanth Anantha
- Department of Anatomy & Neuroscience, University College Cork (UCC), Cork, Ireland
| | - Susan R. Goulding
- Department of Anatomy & Neuroscience, University College Cork (UCC), Cork, Ireland
- Department of Biological Sciences, Cork Institute of Technology, Cork, Ireland
| | - Sean L. Wyatt
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, UK
| | - Ruth M. Concannon
- Department of Anatomy & Neuroscience, University College Cork (UCC), Cork, Ireland
| | - Louise M. Collins
- Department of Anatomy & Neuroscience, University College Cork (UCC), Cork, Ireland
- Department of Physiology, UCC, Cork, Ireland
| | - Aideen M. Sullivan
- Department of Anatomy & Neuroscience, University College Cork (UCC), Cork, Ireland
- APC Microbiome Ireland, UCC, Cork, Ireland
- Cork Neuroscience Centre, UCC, Cork, Ireland
| | - Gerard W. O'Keeffe
- Department of Anatomy & Neuroscience, University College Cork (UCC), Cork, Ireland
- APC Microbiome Ireland, UCC, Cork, Ireland
- Cork Neuroscience Centre, UCC, Cork, Ireland
| |
Collapse
|
7
|
Iarkov A, Barreto GE, Grizzell JA, Echeverria V. Strategies for the Treatment of Parkinson's Disease: Beyond Dopamine. Front Aging Neurosci 2020; 12:4. [PMID: 32076403 PMCID: PMC7006457 DOI: 10.3389/fnagi.2020.00004] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/09/2020] [Indexed: 12/11/2022] Open
Abstract
Parkinson’s disease (PD) is the second-leading cause of dementia and is characterized by a progressive loss of dopaminergic neurons in the substantia nigra alongside the presence of intraneuronal α-synuclein-positive inclusions. Therapies to date have been directed to the restoration of the dopaminergic system, and the prevention of dopaminergic neuronal cell death in the midbrain. This review discusses the physiological mechanisms involved in PD as well as new and prospective therapies for the disease. The current data suggest that prevention or early treatment of PD may be the most effective therapeutic strategy. New advances in the understanding of the underlying mechanisms of PD predict the development of more personalized and integral therapies in the years to come. Thus, the development of more reliable biomarkers at asymptomatic stages of the disease, and the use of genetic profiling of patients will surely permit a more effective treatment of PD.
Collapse
Affiliation(s)
- Alexandre Iarkov
- Laboratorio de Neurobiología, Facultad de Ciencias de la Salud, Universidad San Sebastián, Concepción, Chile
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.,Health Research Institute, University of Limerick, Limerick, Ireland
| | - J Alex Grizzell
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado, Boulder, CO, United States
| | - Valentina Echeverria
- Laboratorio de Neurobiología, Facultad de Ciencias de la Salud, Universidad San Sebastián, Concepción, Chile.,Research & Development Service, Bay Pines VA Healthcare System, Bay Pines, FL, United States
| |
Collapse
|
8
|
Wang T, Nimkingratana P, Smith CA, Cheng A, Hardingham TE, Kimber SJ. Enhanced chondrogenesis from human embryonic stem cells. Stem Cell Res 2019; 39:101497. [PMID: 31326745 PMCID: PMC6745516 DOI: 10.1016/j.scr.2019.101497] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 05/17/2019] [Accepted: 07/08/2019] [Indexed: 02/06/2023] Open
Abstract
Human embryonic stem cells (hESCs) have great potential for the repair of damaged articular cartilage. We developed a serum-free 14-day protocol for hESC differentiation into chondrocyte progenitors, which surprisingly lacked strong cartilage matrix production in in vitro tests. In order to direct these progenitors to a more mature phenotype, we investigated substituting different members of the TGFβ family in the protocol. Initially, we supplemented, or substituted GDF5 (day 11-14), with combinations of BMP7 and TGFβ-1, or -3, but these modifications yielded no improvement in matrix gene expression. However, replacing BMP4 with BMP2 (days 3-10 of the protocol) resulted in a more rapid increase in SOX9 gene expression and increased expression of chondrogenic genes SOX5, ACAN and COL2A1. The replacement of BMP4 with BMP2 also enhanced the formation of chondrogenic cell aggregates, with greater deposition of type II collagen. This change was not accompanied by hypertrophic chondrocyte marker COL10A1 expression. The results demonstrate that BMP2 has greater specificity for the generation of chondrogenic cells from hESCs than BMP4 and this was consistent in two hESC lines (HUES1 and MAN7). hESC-chondrogenic cells derived with either BMP2 or BMP4 were tested in vivo by implanting them in fibrin into osteochondral defects in the femur of RNU rats. Repaired cartilage tissue, positive for Safranin O and type II collagen was detected at 6 and 12 weeks with both cell sources, but the BMP2 cells scored higher for tissue quality (Pineda score). Therefore, BMP2 is more effective at driving chondrogenic differentiation from human pluripotent stem cells than BMP4 and the effect on the resulting chondroprogenitors is sustained in an in vivo setting.
Collapse
Affiliation(s)
- Tao Wang
- Faculty of Biology, Medicine and Health, University of Manchester, UK
| | | | | | - Aixin Cheng
- Faculty of Biology, Medicine and Health, University of Manchester, UK
| | - Timothy E Hardingham
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Susan J Kimber
- Faculty of Biology, Medicine and Health, University of Manchester, UK.
| |
Collapse
|
9
|
de Natale ER, Wilson H, Pagano G, Politis M. Imaging Transplantation in Movement Disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2018; 143:213-263. [PMID: 30473196 DOI: 10.1016/bs.irn.2018.10.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cell replacement therapy with graft transplantation has been tested as a disease-modifying treatment in neurodegenerative diseases characterized by the damage of a predominant cell type, such as substantia nigra dopaminergic neurons in Parkinson's disease (PD) or striatal medium spiny projection neurons in Huntington's disease (HD). The results of these trials are mixed with success in preclinical and pilot open-label trials, which were not consistently reproduced in randomized controlled trials. Positron emission tomography (PET) and single photon emission computed tomography (SPECT) molecular imaging and functional magnetic resonance imaging allow the graft survival, and its relationship with the host tissues to be studied in vivo. In PD, PET with [18F]DOPA showed that graft survival does not necessarily correlate with the clinical improvement and PD patients with worse outcome had lower binding in the ventral striatum and a high serotonin ([11C]DASB PET) to dopamine ([18F]DOPA PET) ratio in the grafted neurons. In HD, PET with [11C]PK11195 showed the graft survival and the clinical responses may be related to the reactive activation of the host inflammatory/immune system. Findings from these studies have been used to refine study protocols and patient selection in current clinical trials, which includes identifying suitable candidates for transplantation using imaging markers and employing multiple and/or novel PET tracers to better assess graft functions and inflammatory responses to grafts.
Collapse
Affiliation(s)
- Edoardo Rosario de Natale
- Neurodegeneration Imaging Group, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, United Kingdom
| | - Heather Wilson
- Neurodegeneration Imaging Group, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, United Kingdom
| | - Gennaro Pagano
- Neurodegeneration Imaging Group, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, United Kingdom
| | - Marios Politis
- Neurodegeneration Imaging Group, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, United Kingdom.
| |
Collapse
|
10
|
Meyers EA, Kessler JA. TGF-β Family Signaling in Neural and Neuronal Differentiation, Development, and Function. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022244. [PMID: 28130363 DOI: 10.1101/cshperspect.a022244] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Signaling by the transforming growth factor β (TGF-β) family is necessary for proper neural development and function throughout life. Sequential waves of activation, inhibition, and reactivation of TGF-β family members regulate numerous elements of the nervous system from the earliest stages of embryogenesis through adulthood. This review discusses the expression, regulation, and function of TGF-β family members in the central nervous system at various developmental stages, beginning with induction and patterning of the nervous system to their importance in the adult as modulators of inflammatory response and involvement in degenerative diseases.
Collapse
Affiliation(s)
- Emily A Meyers
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - John A Kessler
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| |
Collapse
|
11
|
Baker KA, Purdy MB, Sadi D, Mukhida K, Mendez I. A Sequential Intrastriatal Dopaminergic Graft Strategy in the Rodent Model for Parkinson's Disease: Implications for Graft Survival and Targeting. Cell Transplant 2017. [DOI: 10.3727/096020198389951] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Optimal placement of intrastriatal dopaminergic grafts is likely crucial to optimize clinical recovery in Parkinson's disease (PD). The target sites of dopaminergic grafts vary among clinical trials and may partially explain the variable results in clinical efficacy reported thus far. In this study we hypothesized that a subsequent dopaminergic graft may promote functional recovery following a suboptimal initial graft. To test this hypothesis, rats with unilateral 6-hydroxydopamine lesions of the right nigrostriatal pathway were randomly divided into three groups. The first group received 900,000 fetal nigral cells in the medial striatum only (n = 6). The second group received 900,000 cells in both the medial and lateral striatum simultaneously (1.8 million total; n = 8). The final group received a second graft of 900,000 cells in the lateral striatum 6 weeks following initial transplantation of a medial graft (n = 6). Amphetamine-induced circling behavior was significantly reduced in both simultaneous and sequential graft groups at 9 and 12 weeks following transplantation of the initial graft. However, no recovery was noted in the single medial graft group at those time points. Furthermore, increased survival of dopaminergic cells was observed in the lateral graft of sequentially grafted animals compared with the medial graft. We conclude that a well-positioned subsequent graft can restore function in animals with a suboptimal initial graft and that the initial graft may improve survival of the second graft. These results are further discussed in relation to their important clinical implication for neural transplantation in PD.
Collapse
Affiliation(s)
- K. A. Baker
- Neural Transplantation Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada, B3H 4H7
- Department of Anatomy and Neurobiology, Dalhousie University, Halifax, Nova Scotia, Canada, B3H 4H7
| | - M. B. Purdy
- Neural Transplantation Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada, B3H 4H7
| | - D. Sadi
- Neural Transplantation Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada, B3H 4H7
| | - K. Mukhida
- Neural Transplantation Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada, B3H 4H7
| | - I. Mendez
- Neural Transplantation Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada, B3H 4H7
- Department of Anatomy and Neurobiology, Dalhousie University, Halifax, Nova Scotia, Canada, B3H 4H7
- Department of Surgery (Division of Neurosurgery), Dalhousie University, Halifax, Nova Scotia, Canada, B3H 4H7
| |
Collapse
|
12
|
Macauley SL, Horsch AD, Oterdoom M, Zheng MH, Stewart GR. The Effects of Transforming Growth Factor-β2 on Dopaminergic Graft Survival. Cell Transplant 2017; 13:245-52. [PMID: 15191162 DOI: 10.3727/000000004783984043] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Dopaminergic cell transplantation is a promising therapeutic approach for the treatment of Parkinson's disease, the potential of which is limited due to poor survival and low dopamine content within engrafted tissue. In this study, the ability of transforming growth factor-β2 (TGF-β2) to influence transplant survival was evaluated. Cell suspensions containing fetal rat ventral mesencephalon (VM) cells were incubated prior to surgery with vehicle (DPBS), varying concentrations of TGF-β2 (5–1000 ng/ml), or a pan-specific antibody against TGF-β (1D11, 100 ng/ml). VM cell suspensions (200,000 cells) were unilaterally implanted into the striatum of adult Sprague-Dawley rats (n = 5–11 animals/group). Following a 3-week survival period, small but viable VM grafts containing tyrosine hydroxylase-positive (TH+) neurons and fibers were present in all animals. Addition of TGF-β2 resulted in a steep, bell-shaped dose-response curve with a significant effect on TH+/dopamine cell survival. At 50 ng/ml TGF-β2, the number of surviving dopamine neurons was increased twofold compared with controls. Addition of TGF-β2 or 1D11 did not significantly influence graft volume. Further studies, possibly in combination with other neurotrophic factors, need to be performed to obtain a greater understanding of the effects of TGF-β on dopamine neurons and fetal VM cell engraftment.
Collapse
|
13
|
Targeting bone morphogenetic protein signalling in midbrain dopaminergic neurons as a therapeutic approach in Parkinson's disease. Neuronal Signal 2017; 1:NS20170027. [PMID: 32714578 PMCID: PMC7373244 DOI: 10.1042/ns20170027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 02/20/2017] [Accepted: 02/23/2017] [Indexed: 11/17/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, characterized by the degeneration of midbrain dopaminergic (mDA) neurons and their axons, and aggregation of α-synuclein, which leads to motor and late-stage cognitive impairments. As the motor symptoms of PD are caused by the degeneration of a specific population of mDA neurons, PD lends itself to neurotrophic factor therapy. The goal of this therapy is to apply a neurotrophic factor that can slow down, halt or even reverse the progressive degeneration of mDA neurons. While the best known neurotrophic factors are members of the glial cell line-derived neurotrophic factor (GDNF) family, their lack of clinical efficacy to date means that it is important to continue to study other neurotrophic factors. Bone morphogenetic proteins (BMPs) are naturally secreted proteins that play critical roles during nervous system development and in the adult brain. In this review, we provide an overview of the BMP ligands, BMP receptors (BMPRs) and their intracellular signalling effectors, the Smad proteins. We review the available evidence that BMP-Smad signalling pathways play an endogenous role in mDA neuronal survival in vivo, before outlining how exogenous application of BMPs exerts potent effects on mDA neuron survival and axon growth in vitro and in vivo. We discuss the molecular mechanisms that mediate these effects, before highlighting the potential of targeting the downstream effectors of BMP-Smad signalling as a novel neuroprotective approach to slow or stop the degeneration of mDA neurons in PD.
Collapse
|
14
|
Viral vector delivery of neurotrophic factors for Parkinson's disease therapy. Expert Rev Mol Med 2015; 17:e8. [DOI: 10.1017/erm.2015.6] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterised by the progressive loss of midbrain dopaminergic neurons, which causes motor impairments. Current treatments involve dopamine replacement to address the disease symptoms rather than its cause. Factors that promote the survival of dopaminergic neurons have been proposed as novel therapies for PD. Several dopaminergic neurotrophic factors (NTFs) have been examined for their ability to protect and/or restore degenerating dopaminergic neurons, both in animal models and in clinical trials. These include glial cell line-derived neurotrophic factor, neurturin, cerebral dopamine neurotrophic factor and growth/differentiation factor 5. Delivery of these NTFs via injection or infusion to the brain raises several practical problems. A new delivery approach for NTFs involves the use of recombinant viral vectors to enable long-term expression of these factors in brain cells. Vectors used include those based on adenoviruses, adeno-associated viruses and lentiviruses. Here we review progress to date on the potential of each of these four NTFs as novel therapeutic strategies for PD, as well as the challenges that have arisen, from pre-clinical analysis to clinical trials. We conclude by discussing recently-developed approaches to optimise the delivery of NTF-carrying viral vectors to the brain.
Collapse
|
15
|
Hegarty SV, O'Keeffe GW, Sullivan AM. Neurotrophic factors: from neurodevelopmental regulators to novel therapies for Parkinson's disease. Neural Regen Res 2014; 9:1708-11. [PMID: 25422631 PMCID: PMC4238158 DOI: 10.4103/1673-5374.143410] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2014] [Indexed: 11/23/2022] Open
Abstract
Neuroprotection and neuroregeneration are two of the most promising disease-modifying therapies for the incurable and widespread Parkinson's disease. In Parkinson's disease, progressive degeneration of nigrostriatal dopaminergic neurons causes debilitating motor symptoms. Neurotrophic factors play important regulatory roles in the development, survival and maintenance of specific neuronal populations. These factors have the potential to slow down, halt or reverse the loss of nigrostriatal dopaminergic neurons in Parkinson's disease. Several neurotrophic factors have been investigated in this regard. This review article discusses the neurodevelopmental roles and therapeutic potential of three dopaminergic neurotrophic factors: glial cell line-derived neurotrophic factor, neurturin and growth/differentiation factor 5.
Collapse
Affiliation(s)
- Shane V Hegarty
- Department of Anatomy and Neuroscience, Biosciences Institute, University College Cork, Cork, Ireland
| | - Gerard W O'Keeffe
- Department of Anatomy and Neuroscience, Biosciences Institute, University College Cork, Cork, Ireland
| | - Aideen M Sullivan
- Department of Anatomy and Neuroscience, Biosciences Institute, University College Cork, Cork, Ireland
| |
Collapse
|
16
|
Hegarty SV, Collins LM, Gavin AM, Roche SL, Wyatt SL, Sullivan AM, O'Keeffe GW. Canonical BMP-Smad signalling promotes neurite growth in rat midbrain dopaminergic neurons. Neuromolecular Med 2014; 16:473-89. [PMID: 24682653 DOI: 10.1007/s12017-014-8299-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 03/07/2014] [Indexed: 01/01/2023]
Abstract
Ventral midbrain (VM) dopaminergic (DA) neurons project to the dorsal striatum via the nigrostriatal pathway to regulate voluntary movements, and their loss leads to the motor dysfunction seen in Parkinson's disease (PD). Despite recent progress in the understanding of VM DA neurogenesis, the factors regulating nigrostriatal pathway development remain largely unknown. The bone morphogenetic protein (BMP) family regulates neurite growth in the developing nervous system and may contribute to nigrostriatal pathway development. Two related members of this family, BMP2 and growth differentiation factor (GDF)5, have neurotrophic effects, including promotion of neurite growth, on cultured VM DA neurons. However, the molecular mechanisms regulating their effects on DA neurons are unknown. By characterising the temporal expression profiles of endogenous BMP receptors (BMPRs) in the developing and adult rat VM and striatum, this study identified BMP2 and GDF5 as potential regulators of nigrostriatal pathway development. Furthermore, through the use of noggin, dorsomorphin and BMPR/Smad plasmids, this study demonstrated that GDF5- and BMP2-induced neurite outgrowth from cultured VM DA neurons is dependent on BMP type I receptor activation of the Smad 1/5/8 signalling pathway.
Collapse
Affiliation(s)
- Shane V Hegarty
- Department of Anatomy and Neuroscience, Biosciences Institute, University College Cork, Cork, Ireland
| | | | | | | | | | | | | |
Collapse
|
17
|
Roles for the TGFβ superfamily in the development and survival of midbrain dopaminergic neurons. Mol Neurobiol 2014; 50:559-73. [PMID: 24504901 DOI: 10.1007/s12035-014-8639-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 01/02/2014] [Indexed: 12/29/2022]
Abstract
The adult midbrain contains 75% of all dopaminergic neurons in the CNS. Within the midbrain, these neurons are divided into three anatomically and functionally distinct clusters termed A8, A9 and A10. The A9 group plays a functionally non-redundant role in the control of voluntary movement, which is highlighted by the motor syndrome that results from their progressive degeneration in the neurodegenerative disorder, Parkinson's disease. Despite 50 years of investigation, treatment for Parkinson's disease remains symptomatic, but an intensive research effort has proposed delivering neurotrophic factors to the brain to protect the remaining dopaminergic neurons, or using these neurotrophic factors to differentiate dopaminergic neurons from stem cell sources for cell transplantation. Most neurotrophic factors studied in this context have been members of the transforming growth factor β (TGFβ) superfamily. In recent years, an intensive research effort has focused on understanding the function of these proteins in midbrain dopaminergic neuron development and their role in the molecular architecture that regulates the development of this brain region, with the goal of applying this knowledge to develop novel therapies for Parkinson's disease. In this review, the current evidence showing that TGFβ superfamily members play critical roles in the regulation of midbrain dopaminergic neuron induction, differentiation, target innervation and survival during embryonic and postnatal development is analysed, and the implications of these findings are discussed.
Collapse
|
18
|
6-Hydroxydopamine induces distinct alterations in GDF5 and GDNF mRNA expression in the rat nigrostriatal system in vivo. Neurosci Lett 2013; 561:176-81. [PMID: 24373993 DOI: 10.1016/j.neulet.2013.12.046] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 12/05/2013] [Accepted: 12/20/2013] [Indexed: 01/07/2023]
Abstract
Growth/differentiation factor (GDF)5 and glial cell line-derived neurotrophic factor (GDNF) are neurotrophic factors that promote the survival of midbrain dopaminergic neurons in vitro and in vivo. Both factors have potent neurotrophic and neuroprotective effects in rat models of Parkinson's disease (PD) and represent promising new therapies for PD. The aim of this study was to investigate the expression of GDF5, GDNF and their receptors in the nigrostriatal dopaminergic system in rat models of PD. It found that endogenous GDF5, GDNF and their receptors are differentially expressed in two 6-hydroxydopamine lesion models of PD. In both striatal and medial forebrain bundle (MFB) lesion models, striatal levels of GDF5 mRNA increased at 10 days post-lesion, while GDNF mRNA levels in the nigrostriatal system decreased after 10 and 28 days. Midbrain mRNA levels for both GDF5 receptors transiently increased after striatal lesion, whereas those of two GDNF receptors decreased at later time-points in both models. Despite the fact that exogenous GDF5 and GDNF have comparable effects on dopaminergic neurons in vitro and in vivo, their endogenous responses to neurotoxic injury are different. This highlights the importance of studying neurotrophic factor expression at distinct disease stages and in various animal models of PD.
Collapse
|
19
|
BMP2 and GDF5 induce neuronal differentiation through a Smad dependant pathway in a model of human midbrain dopaminergic neurons. Mol Cell Neurosci 2013; 56:263-71. [PMID: 23831389 DOI: 10.1016/j.mcn.2013.06.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 05/24/2013] [Accepted: 06/25/2013] [Indexed: 01/12/2023] Open
Abstract
Parkinson's disease is the second most common neurodegenerative disease, and is characterised by the progressive degeneration of the nigrostriatal dopaminergic (DA) system. Current treatments are symptomatic, and do not protect against the DA neuronal loss. One of the most promising treatment approaches is the application of neurotrophic factors to rescue the remaining population of nigrostriatal DA neurons. Therefore, the identification of new neurotrophic factors for midbrain DA neurons, and the subsequent elucidation of the molecular bases of their effects, are important. Two related members of the bone morphogenetic protein (BMP) family, BMP2 and growth differentiation factor 5 (GDF5), have been shown to have neurotrophic effects on midbrain DA neurons both in vitro and in vivo. However, the molecular (signalling pathway(s)) and cellular (direct neuronal or indirect via glial cells) mechanisms of their effects remain to be elucidated. Using the SH-SH5Y human neuronal cell line, as a model of human midbrain DA neurons, we have shown that GDF5 and BMP2 induce neurite outgrowth via a direct mechanism. Furthermore, we demonstrate that these effects are dependent on BMP type I receptor activation of canonical Smad 1/5/8 signalling.
Collapse
|
20
|
Midbrain dopaminergic neurons: a review of the molecular circuitry that regulates their development. Dev Biol 2013; 379:123-38. [PMID: 23603197 DOI: 10.1016/j.ydbio.2013.04.014] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 03/27/2013] [Accepted: 04/12/2013] [Indexed: 12/21/2022]
Abstract
Dopaminergic (DA) neurons of the ventral midbrain (VM) play vital roles in the regulation of voluntary movement, emotion and reward. They are divided into the A8, A9 and A10 subgroups. The development of the A9 group of DA neurons is an area of intense investigation to aid the generation of these neurons from stem cell sources for cell transplantation approaches to Parkinson's disease (PD). This review discusses the molecular processes that are involved in the identity, specification, maturation, target innervation and survival of VM DA neurons during development. The complex molecular interactions of a number of genetic pathways are outlined, as well as recent advances in the mechanisms that regulate subset identity within the VM DA neuronal pool. A thorough understanding of the cellular and molecular mechanisms involved in the development of VM DA neurons will greatly facilitate the use of cell replacement therapy for the treatment of PD.
Collapse
|
21
|
Costello DJ, O'Keeffe GW, Hurley FM, Sullivan AM. Transplantation of novel human GDF5-expressing CHO cells is neuroprotective in models of Parkinson's disease. J Cell Mol Med 2013; 16:2451-60. [PMID: 22436046 PMCID: PMC3823439 DOI: 10.1111/j.1582-4934.2012.01562.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Growth/differentiation factor 5 (GDF5) is a neurotrophic factor that promotes the survival of midbrain dopaminergic neurons in vitro and in vivo and as such is potentially useful in the treatment of Parkinson's disease (PD). This study shows that a continuous supply of GDF5, produced by transplanted GDF5-overexpressing CHO cells in vivo, has neuroprotective and neurorestorative effects on midbrain dopaminergic neurons following 6-hydroxydopamine (6-OHDA)-induced lesions of the adult rat nigrostriatal pathway. It also increases the survival and improves the function of transplanted embryonic dopaminergic neurons in the 6-OHDA-lesioned rat model of PD. This study provides the first proof-of-principle that sustained delivery of GDF5 in vivo may be useful in the treatment of PD.
Collapse
Affiliation(s)
- Daniel J Costello
- Department of Anatomy and Neuroscience, Biosciences Institute, University College Cork, Cork, Ireland
| | | | | | | |
Collapse
|
22
|
Survival, differentiation, and connectivity of ventral mesencephalic dopamine neurons following transplantation. PROGRESS IN BRAIN RESEARCH 2012. [DOI: 10.1016/b978-0-444-59575-1.00004-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
23
|
Politis M, Piccini P. In vivo imaging of the integration and function of nigral grafts in clinical trials. PROGRESS IN BRAIN RESEARCH 2012. [PMID: 23195420 DOI: 10.1016/b978-0-444-59575-1.00009-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In vivo functional imaging has provided objective evidence for the integration and function of nigral grafts in the brains of patients with Parkinson's disease. Clinical trials with the use of positron emission tomography have shown that transplants of human dopamine-rich fetal ventral mesencephalic tissue can survive, grow, and release dopamine providing motor symptom relief, and also that they can restore brain activation related to movement. Positron emission tomography has aided in the elucidation of the pathophysiology of serious adverse effects, so-called graft-induced dyskinesias. With the use of newly established radioligands, positron emission tomography and single-photon emission computed tomography could help to improve Parkinson's patient selection in future clinical trials by selecting those with better predicted outcomes. Moreover, positron emission tomography could help monitoring postoperational inflammatory processes around the grafted tissue and the effect of immunosuppression. Recent evidence from positron emission tomography has provided insight of how ongoing extrastriatal serotonergic denervation may have relevance to nonmotor symptoms in transplanted Parkinson's disease patients indicating new cell therapy targets for a more complete relief of symptoms. Functional and structural magnetic resonance imaging techniques could help to better assess the integration of nigral graft with the host brain by assessing the restoration of brain activation during movement and of functional and structural connectivity. This knowledge should lead to the development of new, optimized in vivo imaging protocols that could help to better schedule, monitor, and modify the clinical outcomes of future human trials assessing the efficacy of fetal or stem cell therapy in Parkinson's disease.
Collapse
Affiliation(s)
- Marios Politis
- Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK.
| | | |
Collapse
|
24
|
Toulouse A, Collins GC, Sullivan AM. Neurotrophic effects of growth/differentiation factor 5 in a neuronal cell line. Neurotox Res 2011; 21:256-65. [PMID: 21858606 DOI: 10.1007/s12640-011-9266-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Revised: 08/02/2011] [Accepted: 08/04/2011] [Indexed: 10/17/2022]
Abstract
The neurotrophin growth/differentiation factor 5 (GDF5) is studied as a potential therapeutic agent for Parkinson's disease as it is believed to play a role in the development and maintenance of the nigrostriatal system. Progress in understanding the effects of GDF5 on dopaminergic neurones has been hindered by the use of mixed cell populations derived from primary cultures or in vivo experiments, making it difficult to differentiate between direct and indirect effects of GDF5 treatment on neurones. In an attempt to establish an useful model to study the direct neuronal influence of GDF5, we have characterised the effects of GDF5 on a human neuronal cell line, SH-SY5Y. Our results show that GDF5 has the capability to promote neuronal but not dopaminergic differentiation. We also show that it promotes neuronal survival in vitro following a 6-hydroxydopamine insult. Our results show that application of GDF5 to SH-SY5Y cultures induces the SMAD pathway which could potentially be implicated in the intracellular transmission of GDF5's neurotrophic effects. Overall, our study shows that the SH-SY5Y neuroblastoma cell line provides an excellent neuronal model to study the neurotrophic effects of GDF5.
Collapse
Affiliation(s)
- André Toulouse
- Department of Anatomy and Neuroscience, University College Cork, College Road, Cork, Ireland.
| | | | | |
Collapse
|
25
|
Neurotrophic factors for the treatment of Parkinson's disease. Cytokine Growth Factor Rev 2011; 22:157-65. [DOI: 10.1016/j.cytogfr.2011.05.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Accepted: 05/20/2011] [Indexed: 11/20/2022]
|
26
|
O’Sullivan DB, Harrison PT, Sullivan AM. Effects of GDF5 overexpression on embryonic rat dopaminergic neurones in vitro and in vivo. J Neural Transm (Vienna) 2010; 117:559-72. [DOI: 10.1007/s00702-010-0392-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Accepted: 03/07/2010] [Indexed: 12/26/2022]
|
27
|
|
28
|
Yurek DM, Flectcher AM, Kowalczyk TH, Padegimas L, Cooper MJ. Compacted DNA nanoparticle gene transfer of GDNF to the rat striatum enhances the survival of grafted fetal dopamine neurons. Cell Transplant 2009; 18:1183-96. [PMID: 19650971 DOI: 10.3727/096368909x12483162196881] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Previously it was established that infusion of glial cell line-derived neurotrophic factor (GDNF) protein into grafts of embryonic dopamine cells has a neurotrophic effect on the grafted cells. In this study we used a nonviral technique to transfer the gene encoding for GDNF to striatal cells. Plasmid DNA encoding for GDNF was compacted into DNA nanoparticles (DNPs) by 10 kDa polyethylene glycol (PEG)-substituted lysine 30-mers (CK(30)PEG10k) and then injected into the denervated striatum of rats with unilateral 6-hydroxydopamine lesions. Sham controls were injected with saline. One week later, experimental animals received either a ventral mesencephalic (VM) tissue chunk graft or a cell suspension VM graft implanted into the denervated striatum. Grafts were allowed to integrate for 4-6 weeks and during this period we monitored spontaneous and drug-induced motor activity. Using stereological cell counting we observed a 16-fold increase in the number of surviving TH(+) cells within tissue chunk grafts placed into the striatum pretreated with pGDNF DNPs (14,923 +/- 4,326) when compared to grafts placed into striatum pretreated with saline (955 +/- 343). Similarly, we observed a sevenfold increase in the number of TH(+) cells within cell suspension grafts placed into the striatum treated with pGDNF DNPs when compared to cell suspension grafts placed into the saline dosed striatum. Behaviorally, we observed significant improvement in rotational scores and in spontaneous forepaw usage of the affected forelimb in grafted animals receiving prior treatment with compacted pGDNF DNPs when compared to grafted animals receiving saline control pretreatment. Data analysis for protein, morphological, and behavioral measures suggests that compacted pGDNF DNPs injected into the striatum can result in transfected cells overexpressing GDNF protein at levels that provide neurotrophic support for grafted embryonic dopamine neurons.
Collapse
Affiliation(s)
- David M Yurek
- Department of Neurosurgery, University of Kentucky College of Medicine, Lexington, KY 40536-0305, USA.
| | | | | | | | | |
Collapse
|
29
|
Progress in Parkinson's disease—Where do we stand? Prog Neurobiol 2008; 85:376-92. [DOI: 10.1016/j.pneurobio.2008.05.003] [Citation(s) in RCA: 137] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2007] [Revised: 02/26/2008] [Accepted: 05/30/2008] [Indexed: 12/21/2022]
|
30
|
Correia AS, Anisimov SV, Li JY, Brundin P. Growth factors and feeder cells promote differentiation of human embryonic stem cells into dopaminergic neurons: a novel role for fibroblast growth factor-20. Front Neurosci 2008; 2:26-34. [PMID: 18982104 PMCID: PMC2570076 DOI: 10.3389/neuro.01.011.2008] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2008] [Accepted: 05/21/2008] [Indexed: 11/25/2022] Open
Abstract
Human embryonic stem cells (hESCs) are a potential source of dopaminergic neurons for treatment of patients with Parkinson's disease (PD). Dopaminergic neurons can be derived from hESCs and display a characteristic midbrain phenotype. Once transplanted, they can induce partial behavioral recovery in animal models of PD. However, the potential research field faces several challenges that need to be overcome before clinical application of hESCs in a transplantation therapy in PD can be considered. These include low survival of the hESC-derived, grafted dopaminergic neurons after transplantation; unclear functional integration of the grafted neurons in the host brain; and, the risk of teratoma/tumor formation from the transplanted cells. This review is focused on our recent efforts to improve the survival of hESC-dervied dopaminergic neurons. In a recent study, we examined the effect of fibroblast growth factor (FGF)-20 in the differentiation of hESCs into dopaminergic neurons. We supplemented cultures of hESCs with FGF-20 during differentiation on PA6 mouse stromal cells for 3 weeks. When we added FGF-20 the yield of neurons expressing tyrosine hydroxylase increased. We demonstrated that at least part of the effect is contributed by enhanced cell differentiation towards the dopaminergic phenotype as well as reduced cell death. We compare our results with those obtained in other published protocols using different sets of growth factors. Taken together, our data indicate that FGF-20 has potent effects to generate large number of dopaminergic neurons derived from hESCs, which may be useful for hESC-based therapy in PD.
Collapse
Affiliation(s)
- Ana Sofia Correia
- Neuronal Survival Unit, Department of Experimental Medical Science, Lund University, Wallenberg Neuroscience Center Lund, Sweden. Sofi
| | | | | | | |
Collapse
|
31
|
Torres EM, Dowd E, Dunnett SB. Recovery of functional deficits following early donor age ventral mesencephalic grafts in a rat model of Parkinson's disease. Neuroscience 2008; 154:631-40. [PMID: 18468807 DOI: 10.1016/j.neuroscience.2008.03.048] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2007] [Revised: 03/10/2008] [Accepted: 03/20/2008] [Indexed: 12/12/2022]
Abstract
It has previously been reported that dopaminergic grafts derived from early donor age, embryonic age 12-day-old (E12) rat embryos produced a fivefold greater yield of dopamine neurons than those derived from conventional E14 donors. The present study addresses whether E12 grafts are able to ameliorate lesion-induced behavioral deficits to the same extent as E14 grafts. In a unilateral rat model of Parkinson's disease, animals received grafts derived from either E12 or E14 donor embryos, dispersed at four sites in the lesioned striatum. Both E12 and E14 grafts were able to induce recovery on both amphetamine and apomorphine rotation tests, and to ameliorate deficits in the cylinder, stepping test, and corridor tests, but were unable to restore function in the paw reaching task. E12 grafts were equivalent to E14 grafts in their effects on lesion-induced deficits. However, E12 grafts resulted in cell yields greater than previously reported for untreated primary tissue, with mean TH-positive cell counts in excess of 25,000 neurons, compared with E14 TH cell counts of 4000-5000 cells, representing survival rates of 75% and 12.5%, respectively, based on the expected adult complement. The equivalence of graft induced behavioral recovery between the two graft groups is attributed to a threshold number of cells, above which no further improvement is seen. Such high dopamine cell survival rates should mean that multiple, functioning grafts can be derived from a single embryonic donor, and if similar yields could be obtained from human tissues then the goal of one embryo per patient would be achieved.
Collapse
Affiliation(s)
- E M Torres
- Department of Biosciences, Biomedical Sciences Building, Museum Avenue, Cardiff University, PO Box 911 Cardiff South Glamorgan, UK.
| | | | | |
Collapse
|
32
|
AAV2-mediated gene transfer of GDNF to the striatum of MPTP monkeys enhances the survival and outgrowth of co-implanted fetal dopamine neurons. Exp Neurol 2008; 211:252-8. [PMID: 18346734 DOI: 10.1016/j.expneurol.2008.01.026] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2007] [Revised: 01/25/2008] [Accepted: 01/31/2008] [Indexed: 11/20/2022]
Abstract
Neural transplantation offers the potential of treating Parkinson's disease by grafting fetal dopamine neurons to depleted regions of the brain. However, clinical studies of neural grafting in Parkinson's disease have produced only modest improvements. One of the main reasons for this is the low survival rate of transplanted neurons. The inadequate supply of critical neurotrophic factors in the adult brain is likely to be a major cause of early cell death and restricted outgrowth of fetal grafts placed into the mature striatum. Glial derived neurotrophic factor (GDNF) is a potent neurotrophic factor that is crucial to the survival, outgrowth and maintenance of dopamine neurons, and so is a candidate for protecting grafted fetal dopamine neurons in the adult brain. We found that implantation of adeno-associated virus type 2 encoding GDNF (AAV2-GDNF) in the normal monkey caudate nucleus induced overexpression of GDNF that persisted for at least 6 months after injection. In a 6-month within-animal controlled study, AAV2-GDNF enhanced the survival of fetal dopamine neurons by 4-fold, and increased the outgrowth of grafted fetal dopamine neurons by almost 3-fold in the caudate nucleus of MPTP-treated monkeys, compared with control grafts in the other caudate nucleus. Thus, the addition of GDNF gene therapy to neural transplantation may be a useful strategy to improve treatment for Parkinson's disease.
Collapse
|
33
|
Plöger F, Seemann P, Schmidt-von Kegler M, Lehmann K, Seidel J, Kjaer KW, Pohl J, Mundlos S. Brachydactyly type A2 associated with a defect in proGDF5 processing. Hum Mol Genet 2008; 17:1222-33. [PMID: 18203755 DOI: 10.1093/hmg/ddn012] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
34
|
Crotty S, Fitzgerald P, Tuohy E, Harris DM, Fisher A, Mandel A, Bolton AE, Sullivan AM, Nolan Y. Neuroprotective effects of novel phosphatidylglycerol-based phospholipids in the 6-hydroxydopamine model of Parkinson's disease. Eur J Neurosci 2008; 27:294-300. [DOI: 10.1111/j.1460-9568.2007.06018.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
35
|
López de Maturana R, Martin B, Millar RP, Brown P, Davidson L, Pawson AJ, Nicol MR, Mason JI, Barran P, Naor Z, Maudsley S. GnRH-mediated DAN production regulates the transcription of the GnRH receptor in gonadotrope cells. Neuromolecular Med 2007; 9:230-48. [PMID: 17914181 DOI: 10.1007/s12017-007-8004-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2006] [Revised: 11/30/1999] [Accepted: 02/02/2007] [Indexed: 10/23/2022]
Abstract
The primary function of gonadotropin-releasing hormone (GnRH) is the regulation of pituitary gonadotropin hormone gene transcription, biosynthesis and release. These effects are mediated through intracellular mobilization of Ca2+ and activation of PKC isoforms and MAP kinases. We show here that DAN (differential screening-selected gene aberrative in neuroblastoma) which is a secreted bone morphogenic protein (BMP) antagonist belonging to the TGFbeta protein superfamily, is controlled by GnRH in murine gonadotrope cells. Acute GnRH stimulation induced a rapid, 27-fold, elevation of DAN mRNA, accompanied by an approximate 3-fold increase in the amount of mature DAN glycoprotein in the cell cytoplasm and in DAN secretion into the culture medium. Incubation of L beta T2 cells in DAN-containing medium altered the levels of a number of cellular proteins. Two of these were identified as the steroidogenic acute regulatory protein (StAR) and the actin-related protein 2/3 complex subunits 2 (p34-ARC) which are primarily involved in steroidogenesis and cytoskeleton remodelling, respectively. DAN caused an approximate 2-fold specific elevation in the cytoplasmic levels of both these proteins in L beta T2 cells. We further tested the effects of DAN on classical GnRH effects viz. gonadotropin and GnRH receptor gene expression. Co-transfection of L beta T2 cells with DAN and gonadotropin subunit promoter luciferase reporter genes had no effect on GnRH stimulation of alpha GSU and LH beta or on the additive GnRH and activin induction of FSH beta subunit transcription. However, co-transfection of DAN markedly inhibited the synergistic activation of GnRH and activin on GnRH receptor gene expression thus implicating DAN as a novel autocrine/paracrine factor that modulates GnRH function in pituitary gonadotropes.
Collapse
Affiliation(s)
- Rakel López de Maturana
- MRC Human Reproductive Sciences Unit, Centre for Human Reproductive Biology, Edinburgh, EH16 4SB, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Clayton KB, Sullivan AM. Differential effects of GDF5 on the medial and lateral rat ventral mesencephalon. Neurosci Lett 2007; 427:132-7. [DOI: 10.1016/j.neulet.2007.09.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2007] [Revised: 08/29/2007] [Accepted: 09/13/2007] [Indexed: 11/25/2022]
|
37
|
Terpstra BT, Collier TJ, Marchionini DM, Levine ND, Paumier KL, Sortwell CE. Increased cell suspension concentration augments the survival rate of grafted tyrosine hydroxylase immunoreactive neurons. J Neurosci Methods 2007; 166:13-9. [PMID: 17706789 PMCID: PMC2067253 DOI: 10.1016/j.jneumeth.2007.06.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2007] [Revised: 06/21/2007] [Accepted: 06/21/2007] [Indexed: 11/23/2022]
Abstract
The poor survival rate (5-20%) of grafted embryonic dopamine (DA) neurons is one of the primary factors preventing cell replacement from becoming a viable treatment for Parkinson's disease. Previous studies have demonstrated that graft volume impacts grafted DA neuron survival, indicating that transplant parameters influence survival rates. However, the effects of mesencephalic cell concentration on grafted DA neuron survival have not been investigated. The current study compares the survival rates of DA neurons in grafts of varying concentrations. Mesencephalic cell suspensions derived from E14 Fisher 344 rat pups were concentrated to 25,000, 50,000, 100,000 and 200,000 cells/microl and transplanted into two 0.5 microl sites in the 6-OHDA-denervated rat striatum. Animals were sacrificed 10 days and 6 weeks post-transplantation for histochemical analysis of striatal grafts. The absolute number of DA neurons per graft increased proportionally to the total number of cells transplanted. However, our results show that the 200,000 cells/microl group exhibited significantly higher survival rates (5.48+/-0.83%) compared to the 25,000 cells/microl (2.81+/-0.39%) and 50,000 cells/microl (3.36+/-0.51%) groups (p=0.02 and 0.03, respectively). Soma size of grafted DA neurons in the 200,000 cells/microl group was significantly larger than that of the 25,000 cells/microl (p<0.0001) and 50,000 cells/microl groups (p=0.004). In conclusion, increasing the concentration of mesencephalic cells prior to transplantation, augments the survival and functionality of grafted DA neurons. These data have the potential to identify optimal transplantation parameters that can be applied to procedures utilizing stem cells, neural progenitors, and primary mesencephalic cells.
Collapse
Affiliation(s)
- Brian T Terpstra
- Department of Neurology, University of Cincinnati, PO Box 670525, Cincinnati, OH 45267-0525, United States.
| | | | | | | | | | | |
Collapse
|
38
|
Torres EM, Monville C, Gates MA, Bagga V, Dunnett SB. Improved survival of young donor age dopamine grafts in a rat model of Parkinson's disease. Neuroscience 2007; 146:1606-17. [PMID: 17478050 DOI: 10.1016/j.neuroscience.2007.03.037] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2007] [Revised: 03/08/2007] [Accepted: 03/24/2007] [Indexed: 12/12/2022]
Abstract
In an attempt to improve the survival of implanted dopamine cells, we have readdressed the optimal embryonic donor age for dopamine grafts. In a rat model of Parkinson's disease, animals with unilateral 6-hydroxydopamine lesions of the median forebrain bundle received dopamine-rich ventral mesencephalic grafts derived from embryos of crown to rump length 4, 6, 9, or 10.5 mm (estimated embryonic age (E) 11, E12, E13 and E14 days post-coitus, respectively). Grafts derived from 4 mm embryos survived poorly, with less than 1% of the implanted dopamine cells surviving. Grafts derived from 9 mm and 10.5 mm embryos were similar to those seen in previous experiments with survival rates of 8% and 7% respectively. The best survival was seen in the group that received 6 mm grafts, which were significantly larger than all other graft groups. Mean dopamine cell survival in the 6 mm group (E12) was 36%, an extremely high survival rate for primary, untreated ventral mesencephalic grafts applied as a single placement, and more than fivefold larger than the survival rate observed in the 10.5 mm (E14) group. As E12 ventral mesencephalic tissues contain few, if any, differentiated dopamine cells we conclude that the large numbers of dopamine cells seen in the 6 mm grafts must have differentiated post-implantation. We consider the in vivo conditions which allow this differentiation to occur, and the implications for the future of clinical trials based on dopamine cell replacement therapy.
Collapse
Affiliation(s)
- E M Torres
- Department of Biosciences, Cardiff University, Museum Avenue, PO Box 911, Cardiff CF10 3US, UK.
| | | | | | | | | |
Collapse
|
39
|
Bjerkén SA, Boger HA, Nelson M, Hoffer BJ, Granholm AC, Strömberg I. Effects of glial cell line-derived neurotrophic factor deletion on ventral mesencephalic organotypic tissue cultures. Brain Res 2007; 1133:10-9. [PMID: 17184739 PMCID: PMC2670563 DOI: 10.1016/j.brainres.2006.11.052] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2006] [Revised: 11/01/2006] [Accepted: 11/14/2006] [Indexed: 01/03/2023]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is potent for survival and promotion of nerve fibers from midbrain dopamine neurons. It is also known to exert different effects on specific subpopulations of dopamine neurons. In organotypic tissue cultures, dopamine neurons form two diverse nerve fiber growth patterns, targeting the striatum differently. The aim of this study was to investigate the effect of GDNF on the formation of dopamine nerve fibers. Organotypic tissue cultures of ventral mesencephalon of gdnf gene-deleted mice were studied. The results revealed that dopamine neurons survive in the absence of GDNF. Tyrosine hydroxylase immunoreactivity demonstrated, in gdnf knockout and wildtype cultures, nerve fiber formation with two separate morphologies occurring either in the absence or the presence of astrocytes. The outgrowth that occurred in the absence of astrocytes was unaffected by gdnf deletion, whereas nerve fibers guided by the presence of astrocytes were affected in that they reached significantly shorter distances from the gdnf gene-deleted tissue slice, compared to those measured in wildtype cultures. Treatment with GDNF reversed this effect and increased nerve fiber density independent of genotype. Furthermore, migration of astrocytes reached significantly shorter distances from the tissue slice in GDNF knockout compared to wildtype cultures. Exogenous GDNF increased astrocytic migration in gdnf gene-deleted tissue cultures, comparable to lengths observed in wildtype tissue cultures. In conclusion, cultured midbrain dopamine neurons survive in the absence of GDNF, and the addition of GDNF improved dopamine nerve fiber formation - possibly as an indirect effect of astrocytic stimulation.
Collapse
Affiliation(s)
- Sara af Bjerkén
- Department of Integrative Medical Biology, Umeå University, S 901 87 Umeå, Sweden
| | - Heather A. Boger
- Department of Integrative Medical Biology, Umeå University, S 901 87 Umeå, Sweden
| | - Matthew Nelson
- Department of Physiology and Neuroscience and the Center on Aging, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Barry J. Hoffer
- Cellular Neurophysiology, National Institute on Drug Abuse/NIH, Baltimore, MD 21224, USA
| | - Ann-Charlotte Granholm
- Department of Physiology and Neuroscience and the Center on Aging, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ingrid Strömberg
- Department of Integrative Medical Biology, Umeå University, S 901 87 Umeå, Sweden
| |
Collapse
|
40
|
Gash DM, Chen Y, Gerhardt G. Neurotrophic factors and Parkinson's disease. HANDBOOK OF CLINICAL NEUROLOGY 2007; 83:521-533. [PMID: 18808932 DOI: 10.1016/s0072-9752(07)83025-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
|
41
|
Sullivan AM, O'Keeffe GW. The role of growth/differentiation factor 5 (GDF5) in the induction and survival of midbrain dopaminergic neurones: relevance to Parkinson's disease treatment. J Anat 2006; 207:219-26. [PMID: 16185246 PMCID: PMC1571542 DOI: 10.1111/j.1469-7580.2005.00447.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Growth/differentiation factor-5 (GDF5) is a member of the transforming growth factor-beta superfamily which has potent effects on dopaminergic neurones in vitro and in vivo. GDF5 is under investigation as a potential therapeutic agent for Parkinson's disease (PD), which is caused by the progressive degeneration of dopaminergic neurones projecting from the substantia nigra (SN) to the striatum. In the rat ventral mesencephalon (VM; the developing SN), GDF5 expression peaks at embryonic day 14, the time at which dopaminergic neurones undergo terminal differentiation. Addition of GDF5 protein to cultures of embryonic rat VM increases the survival and improves the morphology of dopaminergic neurones in these cultures. GDF5 treatment also increases the number of cells which adopt a dopaminergic phenotype in cultures of VM progenitor cells. Intracerebral administration of GDF5 has potent neuroprotective and restorative effects on the nigrostriatal pathway in animal models of PD. Furthermore, addition of GDF5 protein to embryonic rat dopaminergic neuronal transplants improves their survival and function in a rat model of PD. Thus, GDF5 has potential applications to PD therapy as a dopaminergic neuroprotective agent and as a factor that may induce a dopaminergic neuronal fate in unrestricted progenitor cells.
Collapse
Affiliation(s)
- Aideen M Sullivan
- Department of Neuroscience/Anatomy, Biosciences Research Institute, National University of Ireland Cork (NUIC), College Road, Cork, Ireland.
| | | |
Collapse
|
42
|
Ahn YH, Bensadoun JC, Aebischer P, Zurn AD, Seiger A, Björklund A, Lindvall O, Wahlberg L, Brundin P, Kaminski Schierle GS. Increased fiber outgrowth from xeno-transplanted human embryonic dopaminergic neurons with co-implants of polymer-encapsulated genetically modified cells releasing glial cell line-derived neurotrophic factor. Brain Res Bull 2005; 66:135-42. [PMID: 15982530 DOI: 10.1016/j.brainresbull.2005.04.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2004] [Revised: 03/29/2005] [Accepted: 04/18/2005] [Indexed: 01/31/2023]
Abstract
We investigated whether a continuous supply of glial cell line-derived neurotrophic factor (GDNF) via encapsulated genetically modified cells can promote survival and fiber outgrowth from xenotransplanted human dopaminergic neurons. Cells genetically engineered to continuously secrete GDNF were confined in hollow fiber-based macrocapsules. Each hemiparkinsonian rat received either a single C2C12-hGDNF capsule (n=8) or a C2C12-control capsule (n=8) concomitantly with human embryonic ventral mesencephalic cell suspension transplants. Our results show that fiber outgrowth in the area between the capsule and the graft is more extensive in rats with GDNF-releasing capsules than in rats with control capsules. We suggest that continuous and safe delivery of GDNF to the brain could be a potential way to optimize neural transplantation as a therapy for Parkinson's disease.
Collapse
Affiliation(s)
- Young-Hwan Ahn
- Wallenberg Neuroscience Center, Section for Neuronal Survival, BMC A10, SE-221 84 Lund, Sweden.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Kim DH, Jahng TA, Fu TS, Zhang HY, Novak SA. Evaluation of HealosMP52 osteoinductive bone graft for instrumented lumbar intertransverse process fusion in sheep. Spine (Phila Pa 1976) 2004; 29:2800-8. [PMID: 15599282 DOI: 10.1097/01.brs.0000148155.79353.ac] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN HealosMP52 was evaluated in a sheep model of instrumented lumbar intertransverse process spine fusion and compared to autogenous bone graft. OBJECTIVES To determine the long-term efficacy and safety of HealosMP52 as a bone graft substitute in posterolateral instrumented spinal fusion. SUMMARY OF BACKGROUND DATA Although the standard intertransverse fusion method employs autogenous iliac crest bone, autograft has certain limitations. HealosMP52, an osteoinductive bone graft material, can facilitate noninstrumented posterolateral spine fusion in rabbits and nonhuman primates, but the long-term outcome of such fusions has not been evaluated. METHODS Eleven skeletally mature, female sheep were instrumented with pedicle screws and rods at L2-L3 and L5-L6. Each animal was treated with autograft bone at one fusion level and HealosMP52 at the other. At 6 and 12 months after surgery, bone formation was measured on contact microradiographs and by backscattered electron imaging. Bone core biopsies taken from 6-month and 12-month specimens were evaluated histologically for pathology indicative of osteosarcoma. RESULTS Grossly, all autograft- and HealosMP52-treated levels showed stable fusions at 6 and 12 months. HealosMP52 and autograft treatments resulted in equivalent mean percent bone volumes within fusion bodies; similar values were observed at 6 and 12 months. Fusion bodies contained cortical and trabecular bone with osteoid seams and fatty marrow, and fusion masses showed maturation from 6 to 12 months. HealosMP52 treatment was not associated with implant migration, ectopic bone formation, or pathologic abnormality. No histologic evidence of osteosarcoma was seen on bone core biopsies. CONCLUSIONS This long-term assessment of the use of HealosMP52 in posterolateral instrumented spine fusion indicates that HealosMP52 possesses safe and efficacious bone grafting properties and can potentially serve as anosteoinductive alternative to autograft bone.
Collapse
Affiliation(s)
- Daniel H Kim
- Department of Neurosurgery, Stanford University Medical Center, Stanford, California 94305-5327, USA.
| | | | | | | | | |
Collapse
|
44
|
Georgievska B, Carlsson T, Lacar B, Winkler C, Kirik D. Dissociation between short-term increased graft survival and long-term functional improvements in Parkinsonian rats overexpressing glial cell line-derived neurotrophic factor. Eur J Neurosci 2004; 20:3121-30. [PMID: 15579166 DOI: 10.1111/j.1460-9568.2004.03770.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The present study was designed to analyse whether continuous overexpression of glial cell line-derived neurotrophic factor (GDNF) in the striatum by a recombinant lentiviral vector can provide improved cell survival and additional long-term functional benefits after transplantation of fetal ventral mesencephalic cells in Parkinsonian rats. A four-site intrastriatal 6-hydroxydopamine lesion resulted in an 80-90% depletion of nigral dopamine cells and striatal fiber innervation, leading to stable motor impairments. Histological analysis performed at 4 weeks after grafting into the GDNF-overexpressing striatum revealed a twofold increase in the number of surviving tyrosine hydroxylase (TH)-positive cells, as compared with grafts placed in control (green fluorescent protein-overexpressing) animals. However, in animals that were allowed to survive for 6 months, the numbers of surviving TH-positive cells in the grafts were equal in both groups, suggesting that the cells initially protected at 4 weeks failed to survive despite the continued presence of GDNF. Although cell survival was similar in both grafted groups, the TH-positive fiber innervation density was lower in the GDNF-treated grafted animals (30% of normal) compared with animals with control grafts (55% of normal). The vesicular monoamine transporter-2-positive fiber density in the striatum, by contrast, was equal in both groups, suggesting that long-term GDNF overexpression induced a selective down-regulation of TH in the grafted dopamine neurons. Behavioral analysis in the long-term grafted animals showed that the control grafted animals improved their performance in spontaneous motor behaviors to approximately 50% of normal, whereas the GDNF treatment did not provide any additional recovery.
Collapse
Affiliation(s)
- Biljana Georgievska
- Wallenberg Neuroscience Center, Division of Neurobiology, Lund University, BMC A11, 22184, Lund, Sweden.
| | | | | | | | | |
Collapse
|
45
|
O'Keeffe GW, Dockery P, Sullivan AM. Effects of growth/differentiation factor 5 on the survival and morphology of embryonic rat midbrain dopaminergic neurones in vitro. ACTA ACUST UNITED AC 2004; 33:479-88. [PMID: 15906156 DOI: 10.1007/s11068-004-0511-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2004] [Revised: 08/11/2004] [Accepted: 08/18/2004] [Indexed: 12/24/2022]
Abstract
Growth/differentiation factor 5 (GDF5) is a member of the transforming growth factor-beta superfamily that is expressed in the developing CNS, including the ventral mesencephalon (VM). GDF5 has been shown to increase the survival of dopaminergic neurones in animal models of Parkinson's disease. This study was aimed at characterising the effects of GDF5 on dopaminergic neurones in vitro. Treatment with GDF5 induced a three-fold increase in the number of dopaminergic neurones in embryonic day 14 rat VM cultures after six days in vitro. A significant increase was also observed in the numbers of astrocytes in GDF5-treated cultures. GDF5 treatment also had significant effects on the morphology of dopaminergic neurones in these cultures; total neurite length, number of branch points and somal area were all significantly increased after six days in vitro. Analysis of neurite length and numbers of branch points at each level of the neuritic field revealed that the most pronounced effects of GDF5 were on the secondary and tertiary levels of the neuritic field. The specific type I receptor for GDF5, bone morphogenetic protein receptor (BMPR)-Ib, was found to be strongly expressed in freshly-dissected E14 VM tissue, but its expression was lost with increasing time in culture. Accordingly, treatment with GDF5 for 24 h from the time of plating induced increases in the numbers of dopaminergic neurones, while treatment with GDF5 for 24 h after six days in vitro did not. This study shows that GDF5 can promote both the survival and morphological differentiation of VM dopaminergic neurones in vitro, lending support to its potential as a candidate dopaminergic neurotrophin for use in the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Gerard W O'Keeffe
- Department of Neuroscience/Anatomy, Biosciences Research Institute, National University of Ireland Cork (NUIC), Cork, Ireland
| | | | | |
Collapse
|
46
|
Agrawal AK, Shukla S, Chaturvedi RK, Seth K, Srivastava N, Ahmad A, Seth PK. Olfactory ensheathing cell transplantation restores functional deficits in rat model of Parkinson's disease: a cotransplantation approach with fetal ventral mesencephalic cells. Neurobiol Dis 2004; 16:516-26. [PMID: 15262263 DOI: 10.1016/j.nbd.2004.04.014] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2003] [Revised: 04/22/2004] [Accepted: 04/27/2004] [Indexed: 11/22/2022] Open
Abstract
Different strategies have been worked out to promote survival of transplanted fetal ventral mesencephalic cells (VMCs) using trophic and nontrophic support. Olfactory ensheathing cells (OECs) express high level of growth factors including NGF, bFGF, GDNF, and NT3, which are known to play important role in functional restoration or neurodegeneration. In the present investigation, an attempt has been made to study functional restoration in 6-hydroxydopamine (6-OHDA)-lesioned rat model of Parkinson's disease (PD) following cotransplantation of VMC and OECs (cultured from olfactory bulb, OB) in striatal region. The functional restoration was assessed using neurobehavioral, neurochemical, and immunohistochemical approach. At 12 weeks, post-transplantation, a significant recovery (P < 0.001) in D-amphetamine induced circling behavior (73%), and spontaneous locomotor activity (SLA, 81%) was evident in cotransplanted animals when compared with 6-OHDA-lesioned animals. A significant restoration (P < 0.001) in [3H]-spiperone binding (77%), dopamine (DA) (82%) and 3,4-dihydroxy phenyl acetic acid (DOPAC) level (75%) was observed in animals cotransplanted with OECs and VMC in comparison to lesioned animals. A significantly high expression and quantification of tyrosine hydroxylase (TH)-positive cells in cotransplanted animals further confirmed the supportive role of OECs in viability of transplanted dopaminergic cells, which in turn may be helping in functional restoration. This was further substantiated by our observation of enhanced TH immunoreactivity and differentiation in VMC cocultured with OECs under in vitro conditions as compared to VMC alone cultures. The results suggest that cotransplantation of OECs and VMC may be a better approach for functional restoration in 6-OHDA-induced rat model of Parkinson's disease.
Collapse
Affiliation(s)
- A K Agrawal
- Developmental Toxicology Division, Industrial Toxicology Research Centre, M.G. Marg, Lucknow 226 001, India.
| | | | | | | | | | | | | |
Collapse
|
47
|
O'Keeffe GW, Hanke M, Pohl J, Sullivan AM. Expression of growth differentiation factor-5 in the developing and adult rat brain. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2004; 151:199-202. [PMID: 15246706 DOI: 10.1016/j.devbrainres.2004.04.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/10/2004] [Indexed: 10/26/2022]
Abstract
Expression of the dopaminergic neurotrophin GDF-5 in developing rat ventral mesencephalon (VM) was found to begin at embryonic day (E) 12 and peak on E14, when dopaminergic neurones undergo terminal differentiation. In the adult rat, GDF-5 was found to be restricted to heart and brain, being expressed in many areas of the brain, including striatum and midbrain. This indicates a role for GDF-5 in the development and maintenance of dopaminergic neurones.
Collapse
Affiliation(s)
- Gerard W O'Keeffe
- Department of Neuroscience/Anatomy, Biosciences Research Institute, National University of Ireland Cork, College Road, Cork, Ireland
| | | | | | | |
Collapse
|
48
|
Zuch CL, David D, Ujhelyi L, Hudson JL, Gerhardt GA, Kaplan PL, Bickford PC. Beneficial effects of intraventricularly administered BMP-7 following a striatal 6-hydroxydopamine lesion. Brain Res 2004; 1010:10-6. [PMID: 15126112 DOI: 10.1016/j.brainres.2003.12.058] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2003] [Indexed: 11/25/2022]
Abstract
The present study was undertaken to investigate the effects of bone morphogenetic protein-7 (BMP-7), also named osteogenic protein-1 (OP-1), on the progression of a striatal 6-hydroxydopamine (6-OHDA) lesion. BMP-7, a member of the transforming growth factor-beta (TGF-beta) superfamily of proteins, has been shown to have protective effects in other animal models of neuronal damage. In this study, male Fischer 344 rats received striatal 6-OHDA lesions followed 1 week later by an intraventricular dose of BMP-7. No significant effect of BMP-7 treatment on spontaneous locomotor activity was observed, however BMP-7 significantly increased the density of tyrosine hydroxylase (TH) immunoreactivity (TH-ir) in the substantia nigra (SN) pars compacta, in the lesioned hemisphere [31.7+/-5.2 (optical density (O.D.) arbitrary units) control vs. 50.2+/-4.3 O.D. BMP-7-treated; p<0.05]. Interestingly, BMP-7 significantly increased TH-ir in the SN of the non-lesioned hemisphere (pars reticulata: 14.8+/-1.19 O.D. control vs. 36+/-2.6 O.D. BMP-7-treated, p<0.05; pars compacta: 29.0+/-4.9 O.D. control vs. 64.4+/-6.9 O.D. BMP-7-treated, p<0.001). A significant increase in DA concentration in the contralateral, non-lesioned hemisphere was also noted (113.2 ng/g control vs. 198.2 ng/g BMP-7-treated, p<0.01). In contrast to other intraventricularly administered neurotrophic factors, BMP-7 was not associated with an increase in the sensitivity to pain. These results suggest that BMP-7 is able to act as a dopaminotrophic agent without unwanted side effects and as such may be a useful pharmacological tool in the treatment of Parkinson's disease in humans.
Collapse
Affiliation(s)
- Christina L Zuch
- Department of Pharmacology, University of Colorado Health Sciences Center, Denver, CO, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Sortwell CE, Collier TJ, Camargo MD, Pitzer MR. An in vitro interval before transplantation of mesencephalic reaggregates does not compromise survival or functionality. Exp Neurol 2004; 187:58-64. [PMID: 15081588 DOI: 10.1016/j.expneurol.2003.12.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2003] [Revised: 09/09/2003] [Accepted: 12/02/2003] [Indexed: 11/28/2022]
Abstract
Grafts of primary ventral mesencephalic tissue and cell suspensions to the denervated striatum are currently utilized as a treatment strategy for Parkinson's disease. Survival rates of grafted dopamine (DA) neurons are extremely poor (5-20%) and is even poorer in grafts to the aged striatum. Short pretreatment of grafted cells with various survival-promoting agents has elicited 2- to 3-fold improvements in these survival rates. However, the duration of pretreatment is limited by the necessity of implanting the embryonic cells within a critical period after tissue harvest, potentially limiting the beneficial effects of these interventions. This study details the use of a modified mesencephalic reaggregate culture system combined with striatal-derived trophic factor support to provide an extended ex vivo cell culture interval before grafting. Mesencephalic cell suspension grafts implanted immediately following dissociation were compared to grafts of an equivalent number of cells reaggregated in the presence of striatal oligodendrocyte-type-2 astrocyte (SO2A) conditioned medium for 3 or 7 days. All grafts were placed in the denervated striatum of young adult male Fischer 344 rats. Rotational assessment of amphetamine-induced rotations indicates that aggregates maintained for 3 days in culture present statistically similar functional recovery profiles as compared to cell suspension grafts. Grafts of mesencephalic reaggregates maintained in vitro for 7 days did not display significant improvements in functional recovery. Immunohistochemical analysis for tyrosine hydroxylase immunoreactive (THir) neurons conducted at 10 weeks post-grafting revealed equivalent survival rates of THir neurons in grafts of fresh cell suspensions and aggregates held in culture for 3 days. Grafts of reaggregates held in culture for 7 days possessed significantly fewer THir neurons, about 25% of the cell suspension or 3-day aggregate grafts. This ex vivo reaggregate system allows for extended pretreatment (3 days) of mesencephalic cells with survival-promoting agents and immunological screening of tissue before transplantation.
Collapse
Affiliation(s)
- Caryl E Sortwell
- Department of Neurological Sciences, Research Center for Brain Repair, Rush-Presbyterian-St. Luke's Medical Center, Chicago, IL 60612, USA.
| | | | | | | |
Collapse
|
50
|
Hurley FM, Costello DJ, Sullivan AM. Neuroprotective effects of delayed administration of growth/differentiation factor-5 in the partial lesion model of Parkinson's disease. Exp Neurol 2004; 185:281-9. [PMID: 14736509 DOI: 10.1016/j.expneurol.2003.10.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Neurotrophic factors have the potential for therapeutic use in Parkinson's disease (PD) to support the remaining dopaminergic neurons and protect them against the ongoing disease process. We have examined the effects of the neurotrophin growth and differentiation factor-5 (GDF-5) in a rat model of Parkinson's disease, the intrastriatal 6-hydroxydopamine (6-OHDA) lesion. GDF-5 (25 microg) was injected into either the striatum or substantia nigra (SN) of adult rats at 1 or 2 weeks after 6-hydroxydopamine administration. The behavioral effects of GDF-5 treatment were examined in vivo by amphetamine-induced rotational testing. Injection of GDF-5 into the nigra at either 1 or 2 weeks, or into the striatum at 1 week, after the lesion induced significant decreases in rotations. Post-mortem immunocytochemistry after 6 weeks showed that GDF-5 administration into either site protected dopaminergic cell bodies of the nigra when injected at 1 but not 2 weeks after 6-hydroxydopamine. However, no significant protection of striatal dopaminergic fiber density was observed after GDF-5 treatment. This study shows that the delayed administration of a single dose of GDF-5 has significant protective effects on the damaged adult rat nigrostriatal pathway, reinforcing its therapeutic potential for Parkinson's disease.
Collapse
Affiliation(s)
- Fiona M Hurley
- Department of Anatomy and Biosciences Research Institute, University College Cork, Cork, Ireland
| | | | | |
Collapse
|