1
|
Emborg ME, Gambardella JC, Zhang A, Federoff HJ. Autologous vs heterologous cell replacement strategies for Parkinson disease and other neurologic diseases. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:41-56. [PMID: 39341662 DOI: 10.1016/b978-0-323-90120-8.00010-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Successful cell replacement strategies for brain repair depend on graft integration into the neural network, which is affected by the immune response to the grafted cells. Using Parkinson disease as an example, in this chapter, we consider the immune system interaction and its role in autologous vs heterologous graft survival and integration, as well as past and emerging strategies to overcome the immunologic response. We also reflect on the role of nonhuman primate research to assess novel approaches and consider the role of different stakeholders on advancing the most promising new approaches into the clinic.
Collapse
Affiliation(s)
- Marina E Emborg
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States; Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI, United States; Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, United States.
| | - Julia C Gambardella
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States; Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI, United States
| | - Ai Zhang
- Aspen Neuroscience, San Diego, CA, United States
| | - Howard J Federoff
- Kenai Therapeutics, San Diego, CA, United States; Georgetown University Medical Center, Georgetown, Washington, DC, United States
| |
Collapse
|
2
|
Melatonin-A Potent Therapeutic for Stroke and Stroke-Related Dementia. Antioxidants (Basel) 2020; 9:antiox9080672. [PMID: 32731545 PMCID: PMC7463751 DOI: 10.3390/antiox9080672] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 07/24/2020] [Accepted: 07/24/2020] [Indexed: 12/20/2022] Open
Abstract
Secreted by the pineal gland to regulate the circadian rhythm, melatonin is a powerful antioxidant that has been used to combat oxidative stress in the central nervous system. Melatonin-based therapies have been shown to provide neuroprotective effects in the setting of ischemic stroke by mitigating neuroinflammation and accelerating brain tissue restoration. Melatonin treatment includes injection of exogenous melatonin, pineal gland grafting and melatonin-mediated stem cell therapy. This review will discuss the current preclinical and clinical studies investigating melatonin-based therapeutics to treat stroke.
Collapse
|
3
|
MacPherson A, Kimmelman J. Ethical development of stem-cell-based interventions. Nat Med 2019; 25:1037-1044. [PMID: 31270501 DOI: 10.1038/s41591-019-0511-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 06/04/2019] [Indexed: 02/07/2023]
Abstract
The process of developing new and complex stem-cell-based therapeutics is incremental and requires decades of sustained collaboration among different stakeholders. In this Perspective, we address key ethical and policy challenges confronting the clinical translation of stem-cell-based interventions (SCBIs), including premature diffusion of SCBIs to clinical practice, assessment of risk in trials, obtaining valid informed consent for research participants, balanced and complete scientific reporting and public communications, regulation, and equitable access to treatment. We propose a way forward for translating these therapies with the above challenges in mind.
Collapse
Affiliation(s)
- Amanda MacPherson
- Biomedical Ethics Unit, STREAM Research Group, McGill University, Montreal, Canada
| | - Jonathan Kimmelman
- Biomedical Ethics Unit, STREAM Research Group, McGill University, Montreal, Canada.
| |
Collapse
|
4
|
Sekiya T, Holley MC. 'Surface Transplantation' for Nerve Injury and Repair: The Quest for Minimally Invasive Cell Delivery. Trends Neurosci 2018; 41:429-441. [PMID: 29625774 DOI: 10.1016/j.tins.2018.03.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 02/22/2018] [Accepted: 03/07/2018] [Indexed: 12/15/2022]
Abstract
Cell transplantation is an ambitious, but arguably realistic, therapy for repair of the nervous system. Cell delivery is a major challenge for clinical translation, especially given the apparently inhibitory astrogliotic environment in degenerated tissue. However, astrogliotic tissue also contains endogenous structural and biochemical cues that can be harnessed for functional repair. Minimizing damage to these cues during cell delivery could enhance cell integration. This theory is supported by studies with an auditory astrocyte scar model, in which cells delivered onto the surface of the damaged nerve were more successfully integrated in the host than those injected into the tissue. We consider the application of this less invasive approach for nerve injury and its potential application to some neurodegenerative disorders.
Collapse
Affiliation(s)
- Tetsuji Sekiya
- Department of Otolaryngology, Head and Neck Surgery, Kyoto University Graduate School of Medicine, Sakyou-ku, Kyoto, 606-8507, Japan; Hikone Chuo Hospital, Department of Neurological Surgery, Hikone Chuo Hospital, 421 Nishiima-cho, Hikone, 522-0054, Japan.
| | - Matthew C Holley
- Department of Biomedical Science, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, United Kingdom
| |
Collapse
|
5
|
Vermilyea SC, Emborg ME. The role of nonhuman primate models in the development of cell-based therapies for Parkinson's disease. J Neural Transm (Vienna) 2017; 125:365-384. [PMID: 28326445 PMCID: PMC5847191 DOI: 10.1007/s00702-017-1708-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 03/12/2017] [Indexed: 12/23/2022]
Abstract
Through the course of over three decades, nonhuman primate (NHP) studies on cell-based therapies (CBTs) for Parkinson’s disease (PD) have provided insight into the feasibility, safety and efficacy of the approach, methods of cell collection and preparation, cell viability, as well as potential brain targets. Today, NHP research continues to be a vital source of information for improving cell grafts and analyzing how the host affects graft survival, integration and function. Overall, this article aims to discuss the role that NHP models of PD have played in CBT development and highlights specific issues that need to be considered to maximize the value of NHP studies for the successful clinical translation of CBTs.
Collapse
Affiliation(s)
- Scott C Vermilyea
- Neuroscience Training Program, University of Wisconsin, Madison, 1220 Capitol Court, Madison, WI, 53715, USA.,Wisconsin National Primate Research Center, University of Wisconsin, Madison, USA
| | - Marina E Emborg
- Neuroscience Training Program, University of Wisconsin, Madison, 1220 Capitol Court, Madison, WI, 53715, USA. .,Wisconsin National Primate Research Center, University of Wisconsin, Madison, USA. .,Department of Medical Physics, University of Wisconsin, Madison, USA.
| |
Collapse
|
6
|
Peng SP, Copray S. Comparison of Human Primary with Human iPS Cell-Derived Dopaminergic Neuron Grafts in the Rat Model for Parkinson's Disease. Stem Cell Rev Rep 2016; 12:105-20. [PMID: 26438376 PMCID: PMC4720696 DOI: 10.1007/s12015-015-9623-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Neuronal degeneration within the substantia nigra and the loss of the dopaminergic nigro-striatal pathway are the major hallmarks of Parkinson's disease (PD). Grafts of foetal ventral mesencephalic (VM) dopaminergic (DA) neurons into the striatum have been shown to be able to restore striatal dopamine levels and to improve overall PD symptoms. However, human foetus-derived cell grafts are not feasible for clinical application. Autologous induced pluripotent stem cell (iPS cell)-derived DA neurons are emerging as an unprecedented alternative. In this review, we summarize and compare the efficacy of human iPS cell-derived DA neuron grafts to restore normal behaviour in a rat model for PD with that of human foetal primary DA neurons. The differences we observed in the efficacy to restore normal function between the 2 types of DA neuron grafts could be ascribed to intrinsic properties of the iPS cell-derived DA neurons that critically affected survival and proper neurite extension in the striatum after implantation.
Collapse
Affiliation(s)
- Su-Ping Peng
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
- Department of Neuroscience, Medical Physiology, University Medical Center Groningen, University of Groningen, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Sjef Copray
- Department of Neuroscience, Medical Physiology, University Medical Center Groningen, University of Groningen, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands.
| |
Collapse
|
7
|
Watson N, Diamandis T, Gonzales-Portillo C, Reyes S, Borlongan CV. Melatonin as an Antioxidant for Stroke Neuroprotection. Cell Transplant 2015; 25:883-91. [PMID: 26497887 DOI: 10.3727/096368915x689749] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Melatonin (N-acetyl-5-methoxytryptamine) is a hormone derived from the pineal gland that has a wide range of clinical applications. While melatonin was originally assessed as a hormone specializing in regulation of the normal circadian rhythm in mammals, it now has been shown to be an effective free radical scavenger and antioxidant. Current research has focused on central nervous system (CNS) disorders, stroke in particular, for potential melatonin-based therapeutics. As of now, the realm of potential therapy regimens is focused on three main treatments: exogenously delivered melatonin, pineal gland grafting, and melatonin-mediated stem cell therapy. All therapies contain both costs and benefits, and current research is still focused on finding the best treatment plan. While comprehensive research has been conducted, more research regarding the safety of such therapies is needed in order to transition into the clinical level of testing. Antioxidants such as traditional Chinese medicine, (-)-epigallocatechin-3-gallate (EGCG), and lavender oil, which have been used for thousands of years as treatment, are now gaining recognition as effective melatonin treatment alternatives. This review will further discuss relevant studies assessing melatonin-based therapeutics and provide evidence of other natural melatonin treatment alternatives for the treatment of stroke.
Collapse
Affiliation(s)
- Nate Watson
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
| | | | | | | | | |
Collapse
|
8
|
Cell based therapies in Parkinson's Disease. Ann Neurosci 2014; 18:76-83. [PMID: 25205926 PMCID: PMC4117039 DOI: 10.5214/ans.0972.7531.1118209] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2011] [Revised: 04/09/2011] [Accepted: 04/30/2011] [Indexed: 12/27/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disorder after Alzheimer’s disease. It is characterized by bradykinesia, hypokinesia/ akinesia, rigidity, tremor, and postural instability, caused by dopaminergic (DA) striatal denervation. The prevalence of PD increases from 50 years of age with steep rise after age 60 years. Current treatment of PD relies heavily on replacing lost dopamine either with its precursor L-dopa or dopamine agonists (ropinirole, pramipexole, bromocriptine, lisuride etc). Other pharmacological measures like catechol-O-methyltrasferase (COMT) inhibitors like entacopone, telcapone and monoamine oxidase B (MAO-B) inhibitors like selegiline and rasagiline are also useful, while L-dopa remains the gold standard in the treatment of PD. Emerging therapies are focusing on cell based therapeutics derived from various sources.
Collapse
|
9
|
Heumann R, Moratalla R, Herrero MT, Chakrabarty K, Drucker-Colín R, Garcia-Montes JR, Simola N, Morelli M. Dyskinesia in Parkinson's disease: mechanisms and current non-pharmacological interventions. J Neurochem 2014; 130:472-89. [DOI: 10.1111/jnc.12751] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 04/23/2014] [Accepted: 04/27/2014] [Indexed: 01/24/2023]
Affiliation(s)
- Rolf Heumann
- Molecular Neurobiochemistry; Ruhr-University Bochum; Bochum Germany
| | | | - Maria Trinidad Herrero
- Clinical & Experimental Neuroscience (NiCE-CIBERNED); School of Health Sciences; University Jaume I; Castelló, and School of Medicine; University of Murcia; Murcia Spain
| | | | - René Drucker-Colín
- Instituto de Fisiología Celular; Universidad Nacional Autónoma de México; Mexico City México
| | | | - Nicola Simola
- Department of Biomedical Sciences; Section of Neuropsychopharmacology; University of Cagliari; Cagliari Italy
| | - Micaela Morelli
- Department of Biomedical Sciences; Section of Neuropsychopharmacology; University of Cagliari; Cagliari Italy
- National Institute of Neuroscience (INN); University of Cagliari; Cagliari Italy
- National Research Council (CNR); Neuroscience Institute; Cagliari Italy
- Center of Excellence on Neurobiology of Dependence; University of Cagliari; Cagliari Italy
| |
Collapse
|
10
|
Bloch J, Brunet JF, McEntire CRS, Redmond DE. Primate adult brain cell autotransplantation produces behavioral and biological recovery in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced parkinsonian St. Kitts monkeys. J Comp Neurol 2014; 522:2729-40. [PMID: 24610674 DOI: 10.1002/cne.23579] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 10/25/2013] [Accepted: 11/04/2013] [Indexed: 02/03/2023]
Abstract
The potential for "replacement cells" to restore function in Parkinson's disease has been widely reported over the past 3 decades, rejuvenating the central nervous system rather than just relieving symptoms. Most such experiments have used fetal or embryonic sources that may induce immunological rejection and generate ethical concerns. Autologous sources, in which the cells to be implanted are derived from recipients' own cells after reprogramming to stem cells, direct genetic modifications, or epigenetic modifications in culture, could eliminate many of these problems. In a previous study on autologous brain cell transplantation, we demonstrated that adult monkey brain cells, obtained from cortical biopsies and kept in culture for 7 weeks, exhibited potential as a method of brain repair after low doses of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) caused dopaminergic cell death. The present study exposed monkeys to higher MPTP doses to produce significant parkinsonism and behavioral impairments. Cerebral cortical cells were biopsied from the animals, held in culture for 7 weeks to create an autologous neural cell "ecosystem" and reimplanted bilaterally into the striatum of the same six donor monkeys. These cells expressed neuroectodermal and progenitor markers such as nestin, doublecortin, GFAP, neurofilament, and vimentin. Five to six months after reimplantation, histological analysis with the dye PKH67 and unbiased stereology showed that reimplanted cells survived, migrated bilaterally throughout the striatum, and seemed to exert a neurorestorative effect. More tyrosine hydroxylase-immunoreactive neurons and significant behavioral improvement followed reimplantation of cultured autologous neural cells as a result of unknown trophic factors released by the grafts.
Collapse
Affiliation(s)
- Jocelyne Bloch
- Department of Clinical Neurosciences, Lausanne University Hospital, 1011, Lausanne, Switzerland
| | | | | | | |
Collapse
|
11
|
de la Pena I, Pabon M, Acosta S, Sanberg PR, Tajiri N, Kaneko Y, Borlongan CV. Oligodendrocytes engineered with migratory proteins as effective graft source for cell transplantation in multiple sclerosis. CELL MEDICINE 2014; 6:123-127. [PMID: 24999443 DOI: 10.3727/215517913x674144] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Multiple sclerosis (MS) is characterized by widespread immunomodulatory demyelination of the CNS resulting in nerve cell dysfunction. Accordingly, treatment strategies have been centered on immunodulation and remyelination, with the former primarily focused on reducing the pathology rather than enhancing myelin repair which the latter targets. While conceding to the emerging view of heterogeneity in the pathology of MS, which precludes variations in degree of immune response (i.e., inflammation) and demyelination, the concept of enhancing myelin repair is appealing since it is likely to provide both disease-reducing and disease-inhibiting therapeutic approach to MS. In this regard, we and several others, have proposed that cell replacement therapy is an effective strategy to repair the myelin in MS. Here, we hypothesize that transplantation of mouse bone marrow-derived oligodendrocytes (BMDOs) and BMDOs transfected with Ephrin proteins (BMDO+Ephrin), which are known to enhance cell and axonal migratory capacity, may produce therapeutic benefits in animal models of MS.
Collapse
Affiliation(s)
- Ike de la Pena
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair. University of South Florida, Morsani College of Medicine 12901 Bruce B. Downs Blvd., Tampa, FL 33612
| | - Mibel Pabon
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair. University of South Florida, Morsani College of Medicine 12901 Bruce B. Downs Blvd., Tampa, FL 33612
| | - Sandra Acosta
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair. University of South Florida, Morsani College of Medicine 12901 Bruce B. Downs Blvd., Tampa, FL 33612
| | - Paul R Sanberg
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair. University of South Florida, Morsani College of Medicine 12901 Bruce B. Downs Blvd., Tampa, FL 33612
| | - Naoki Tajiri
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair. University of South Florida, Morsani College of Medicine 12901 Bruce B. Downs Blvd., Tampa, FL 33612
| | - Yuji Kaneko
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair. University of South Florida, Morsani College of Medicine 12901 Bruce B. Downs Blvd., Tampa, FL 33612
| | - Cesar V Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair. University of South Florida, Morsani College of Medicine 12901 Bruce B. Downs Blvd., Tampa, FL 33612
| |
Collapse
|
12
|
Redmond DE, McEntire CRS, Kingsbery JP, Leranth C, Elsworth JD, Bjugstad KB, Roth RH, Samulski RJ, Sladek JR. Comparison of fetal mesencephalic grafts, AAV-delivered GDNF, and both combined in an MPTP-induced nonhuman primate Parkinson's model. Mol Ther 2013; 21:2160-8. [PMID: 23913185 DOI: 10.1038/mt.2013.180] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Accepted: 07/20/2013] [Indexed: 02/08/2023] Open
Abstract
We combined viral vector delivery of human glial-derived neurotrophic factor (GDNF) with the grafting of dopamine (DA) precursor cells from fetal ventral mesencephalon (VM) to determine whether these strategies would improve the anti-Parkinson's effects in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated monkeys, an animal model for Parkinson's disease (PD). Both strategies have been reported as individually beneficial in animal models of PD, leading to clinical studies. GDNF delivery has also been reported to augment VM tissue implants, but no combined studies have been done in monkeys. Monkeys were treated with MPTP and placed into four balanced treatment groups receiving only recombinant adeno-associated virus serotype 5 (rAAV5)/hu-GDNF, only fetal DA precursor cells, both together, or a buffered saline solution (control). The combination of fetal precursors with rAAV5/hu-GDNF showed significantly higher striatal DA concentrations compared with the other treatments, but did not lead to greater functional improvement in this study. For the first time under identical conditions in primates, we show that all three treatments lead to improvement compared with control animals.
Collapse
Affiliation(s)
- D Eugene Redmond
- 1] Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA [2] Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by the degeneration of the dopamine producing neurons projecting from the substantia nigra into the corpus striatum. Current medical therapy is limited and cannot stop or reverse the degeneration. Over the past 30 years, attempts were made to change the course of the disease by replacing the lost neurons with grafts from various sources. Recent controlled clinical trials of fetal cell transplantation for PD have had disappointing results. These events present an opportunity to examine the past developments and future direction of cell transplantation for PD.
Collapse
Affiliation(s)
- Ben Roitberg
- Department of Neurosurgery, University of Illinois at Chicago, Chicago, Illinois 60612, USA.
| | | | | |
Collapse
|
14
|
Shinozuka K, Staples M, Borlongan CV. Melatonin-based therapeutics for neuroprotection in stroke. Int J Mol Sci 2013; 14:8924-47. [PMID: 23698756 PMCID: PMC3676765 DOI: 10.3390/ijms14058924] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 04/10/2013] [Accepted: 04/11/2013] [Indexed: 01/25/2023] Open
Abstract
The present review paper supports the approach to deliver melatonin and to target melatonin receptors for neuroprotection in stroke. We discuss laboratory evidence demonstrating neuroprotective effects of exogenous melatonin treatment and transplantation of melatonin-secreting cells in stroke. In addition, we describe a novel mechanism of action underlying the therapeutic benefits of stem cell therapy in stroke, implicating the role of melatonin receptors. As we envision the clinical entry of melatonin-based therapeutics, we discuss translational experiments that warrant consideration to reveal an optimal melatonin treatment strategy that is safe and effective for human application.
Collapse
Affiliation(s)
- Kazutaka Shinozuka
- Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, USA.
| | | | | |
Collapse
|
15
|
Redmond DE. Using monkeys to understand and cure Parkinson disease. Hastings Cent Rep 2013; Suppl:S7-S11. [PMID: 23138432 DOI: 10.1002/hast.100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
16
|
Smith GA, Breger LS, Lane EL, Dunnett SB. Pharmacological modulation of amphetamine-induced dyskinesia in transplanted hemi-parkinsonian rats. Neuropharmacology 2012; 63:818-28. [DOI: 10.1016/j.neuropharm.2012.06.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 05/29/2012] [Accepted: 06/06/2012] [Indexed: 01/09/2023]
|
17
|
Li X, Katsanevakis E, Liu X, Zhang N, Wen X. Engineering neural stem cell fates with hydrogel design for central nervous system regeneration. Prog Polym Sci 2012. [DOI: 10.1016/j.progpolymsci.2012.02.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
18
|
Shi B, Deng L, Shi X, Dai S, Zhang H, Wang Y, Bi J, Guo M. The enhancement of neural stem cell survival and growth by coculturing with expanded Sertoli cells in vitro. Biotechnol Prog 2011; 28:196-205. [PMID: 22109810 DOI: 10.1002/btpr.720] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 08/20/2011] [Indexed: 12/19/2022]
Abstract
Sertoli cells (SCs) have been described as the "nurse cells" of testis to provide essential growth factors and to create a proper environment for the development of other cells (e.g., germinal and neural stem cell). However, the physiological functions of the SCs obtained from different culture conditions are different in a coculturing system, and thus the optimal SC culturing condition should be investigated in vitro. In this paper, primary Sertoli cells were isolated from a 12-day-old mouse and expanded in two different culture conditions: a two dimensional (2D) plastic tissue disc and a three dimensional (3D) microcarrier culture system. They were then cocultured with neural stem cells (NSCs) isolated from 14-day-old mouse embryos. The metabolic activities of SCs(2D) (SCs in 2D) and SCs(3D) (SCs in 3D) and the amount of proteins secreted from two culturing systems were compared. The results show that the metabolic activity and the amount of secreted proteins from SCs(3D) were higher than both from SCs(2D). Three coculturing groups: NSCs+SC(2D), NSCs+SC(3D), and NSCs +SC-conditioned medium (SCCM, control group) were also compared regarding cell morphology and the numbers of neurons, neural outgrowths and neurospheres. The quantity of neurons, neural outgrowths and neurospheres were the highest in the NSCs+SC(3D) group. SCs cultured in the 3D system had a strong trophic effect on NSCs and enhanced their survival and growth. Besides, the mRNA of trophic and nutritive factors such as Glial-cell-line-derived neurotrophic factor (GDNF) and Interleukin-1 α (IL-1 α) secreted by the SCs from both 2D and 3D culture system were analyzed by real time-PCR and gel assay. The mRNA transcription of GDNF and IL-1α is more apparent in the 3D culture system than that from the 2D one. The coculturing system of NSCs+SC(3D) is a promising candidate for future neural stem cell transplantation.
Collapse
Affiliation(s)
- Bingyang Shi
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Lampe KJ, Kern DS, Mahoney MJ, Bjugstad KB. The administration of BDNF and GDNF to the brain via PLGA microparticles patterned within a degradable PEG-based hydrogel: Protein distribution and the glial response. J Biomed Mater Res A 2011; 96:595-607. [PMID: 21254391 DOI: 10.1002/jbm.a.33011] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2010] [Revised: 10/09/2010] [Accepted: 11/01/2010] [Indexed: 12/18/2022]
Abstract
Tailored delivery of neurotrophic factors (NFs) is a critical challenge that continues to inhibit strategies for guidance of axonal growth in vivo. Of particular importance is the ability to recreate innervation of distant brain regions by transplant tissue, for instance rebuilding the nigrostriatal track, one focus in Parkinson's disease research. Many strategies have utilized polymer drug delivery to target NF release in space and time, but combinatorial approaches are needed to deliver multiple NFs at relevant therapeutic times and locations without toxic side effects. Here we engineered a paradigm of PLGA microparticles entrapped within a degradable PEG-based hydrogel device to locally release two different types of NFs with two different release profiles. Hydrogel/microparticle devices were developed and analyzed for their ability to release GDNF in the caudal area of the brain, near the substantia nigra, or BDNF in the rostral area, near the striatum. The devices delivered their respective NFs in a region localized to within 100 μm of the bridge, but not exclusively to the targeted rostral or caudal ends. BDNF was slowly released over a 56-day period, whereas a bolus of GDNF was released around 28 days. The timed delivery of NFs from implanted devices significantly reduced the microglial response relative to sham surgeries. Given the coordinated drug delivery ability and reduced localized inflammatory response, this multifaceted PEG hydrogel/PLGA microparticle strategy may be a useful tool for further development in combining tissue engineering and drug delivery, and recreating the nigrostriatal track.
Collapse
Affiliation(s)
- Kyle J Lampe
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado, USA
| | | | | | | |
Collapse
|
20
|
Yasuhara T, Hara K, Maki M, Xu L, Yu G, Ali MM, Masuda T, Yu SJ, Bae EK, Hayashi T, Matsukawa N, Kaneko Y, Kuzmin-Nichols N, Ellovitch S, Cruz EL, Klasko SK, Sanberg CD, Sanberg PR, Borlongan CV. Mannitol facilitates neurotrophic factor up-regulation and behavioural recovery in neonatal hypoxic-ischaemic rats with human umbilical cord blood grafts. J Cell Mol Med 2010; 14:914-21. [PMID: 20569276 PMCID: PMC3823123 DOI: 10.1111/j.1582-4934.2008.00671.x] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
We recently demonstrated that blood–brain barrier permeabilization using mannitol enhances the therapeutic efficacy of systemically administered human umbilical cord blood (HUCB) by facilitating the entry of neurotrophic factors from the periphery into the adult stroke brain. Here, we examined whether the same blood–brain barrier manipulation approach increases the therapeutic effects of intravenously delivered HUCB in a neonatal hypoxic-ischaemic (HI) injury model. Seven-day-old Sprague–Dawley rats were subjected to unilateral HI injury and then at day 7 after the insult, animals intravenously received vehicle alone, mannitol alone, HUCB cells (15k mononuclear fraction) alone or a combination of mannitol and HUCB cells. Behavioural tests at post-transplantation days 7 and 14 showed that HI animals that received HUCB cells alone or when combined with mannitol were significantly less impaired in motor asymmetry and motor coordination compared with those that received vehicle alone or mannitol alone. Brain tissues from a separate animal cohort from the four treatment conditions were processed for enzyme-linked immunosorbent assay at day 3 post-transplantation, and revealed elevated levels of GDNF, NGF and BDNF in those that received HUCB cells alone or when combined with mannitol compared with those that received vehicle or mannitol alone, with the combined HUCB cells and mannitol exhibiting the most robust neurotropic factor up-regulation. Histological assays revealed only sporadic detection of HUCB cells, suggesting that the trophic factor–mediated mechanism, rather than cell replacement per se, principally contributed to the behavioural improvement. These findings extend the utility of blood–brain barrier permeabilization in facilitating cell therapy for treating neonatal HI injury.
Collapse
Affiliation(s)
- T Yasuhara
- Department of Neurology, Medical College of Georgia, Augusta, GA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Redmond DE, Weiss S, Elsworth JD, Roth RH, Wakeman DR, Bjugstad KB, Collier TJ, Blanchard BC, Teng YD, Synder EY, Sladek JR. Cellular repair in the parkinsonian nonhuman primate brain. Rejuvenation Res 2010; 13:188-94. [PMID: 20370501 DOI: 10.1089/rej.2009.0960] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Parkinson disease (PD) is a neurodegenerative disorder that provides a useful model for testing cell replacement strategies to rejuvenate the affected dopaminergic neural systems, which have been destroyed by aging and the disease. We first showed that grafts of fetal dopaminergic neurons can reverse parkinsonian motor deficits induced by the toxin, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), validating the feasibility of cellular repair in a primate nervous system. Subsequent clinical trials in Parkinson patients showed encouraging results, including long-term improvement of neurological signs and reduction of medications in some patients. However, many experienced little therapeutic benefit, and some recipients experienced dyskinesias, suggesting a lack of regulated control of the grafts. We have since attempted to improve cell replacements by placing grafts in their correct anatomical location in the substantia nigra and using strategies such as co-grafting fetal striatal tissue or growth factors into the physiologic striatal targets. Moreover, the use of fetal cells depends on a variable supply of donor material, making it difficult to standardize cell quality and quantity. Therefore, we have also explored possibilities of using human neural stem cells (hNSCs) to ameliorate parkinsonism in nonhuman primates with encouraging results. hNSCs implanted into the striatum showed a remarkable migratory ability and were found in the substantia nigra, where a small number appeared to differentiate into dopamine neurons. The majority became growth factor-producing glia that could provide beneficial effects on host dopamine neurons. Studies to determine the optimum stage of differentiation from embryonic stem cells and to derive useful cells from somatic cell sources are in progress.
Collapse
Affiliation(s)
- Donald Eugene Redmond
- Department of Psychiatry and Neurosurgery, Yale University School of Medicine, New Haven, Connecticut 06511, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Evans BM, Allison SW, Fillmore HL, Broaddus WC, Dyer RL, Gillies GT. Cytometric catheter for neurosurgical applications. J Med Eng Technol 2010; 34:261-7. [DOI: 10.3109/03091901003639943] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
23
|
Notch-Induced Rat and Human Bone Marrow Stromal Cell Grafts Reduce Ischemic Cell Loss and Ameliorate Behavioral Deficits in Chronic Stroke Animals. Stem Cells Dev 2009; 18:1501-14. [DOI: 10.1089/scd.2009.0011] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
24
|
Fitzpatrick KM, Raschke J, Emborg ME. Cell-based therapies for Parkinson's disease: past, present, and future. Antioxid Redox Signal 2009; 11:2189-208. [PMID: 19485712 PMCID: PMC2861536 DOI: 10.1089/ars.2009.2654] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Parkinson's disease (PD) researchers have pioneered the use of cell-based therapies (CBTs) in the central nervous system. CBTs for PD were originally envisioned as a way to replace the dopaminergic nigral neurons lost with the disease. Several sources of catecholaminergic cells, including autografts of adrenal medulla and allografts or xenografts of mesencephalic fetal tissue, were successfully assessed in animal models, but their clinical translation has yielded poor results and much controversy. Recent breakthroughs on cell biology are helping to develop novel cell lines that could be used for regenerative medicine. Their future successful clinical application depends on identifying and solving the problems encountered in previous CBTs trials. In this review, we critically analyze past CBTs' clinical translation, the impact of the host in graft survival, and the role of preclinical studies and emerging new cell lines. We propose that the prediction of clinical results from preclinical studies requires experimental designs that allow blind data acquisition and statistical analysis, assessment of the therapy in models that parallel clinical conditions, looking for sources of complications or side effects, and limiting optimism bias when reporting outcomes.
Collapse
Affiliation(s)
- Kathleen M Fitzpatrick
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin 53715, USA
| | | | | |
Collapse
|
25
|
Redmond DE, Elsworth JD, Roth RH, Leranth C, Collier TJ, Blanchard B, Bjugstad KB, Samulski RJ, Aebischer P, Sladek JR. Embryonic substantia nigra grafts in the mesencephalon send neurites to the host striatum in non-human primate after overexpression of GDNF. J Comp Neurol 2009; 515:31-40. [PMID: 19399891 DOI: 10.1002/cne.22028] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In spite of partial success in treating Parkinson's disease by using ectopically placed grafts of dopamine-producing cells, restoration of the original neuroanatomical circuits, if possible, might work better. Previous evidence of normal anatomic projections from ventral mesencephalic (VM) grafts placed in the substantia nigra (SN) has been limited to neonatal rodents and double grafting or bridging procedures. This study attempted to determine whether injection of a potent growth-promoting factor, glial cell line-derived neurotrophic factor (GDNF), into the target regions or placement of fetal striatal co-grafts in the nigrostriatal pathway might elicit neuritic outgrowth to the caudate nucleus. Four adult St. Kitts green monkeys received embryonic VM grafts into the rostral mesencephalon near the host SN, and injections of adeno-associated virus 2 (AAV2)/GDNF or equine infectious anemia virus (EIAV)/GDNF into the caudate. Three adult monkeys were co-grafted with fetal VM tissue near the SN and fetal striatal grafts (STR) 2.5 mm rostral in the nigrostriatal pathway. Before sacrifice, the striatal target regions were injected with the retrograde tracer Fluoro-Gold (FG). FG label was found in tyrosine hydroxylase-labeled neurons in VM grafts in the SN of only those monkeys that received AAV2/GDNF vector injections into the ipsilateral striatum. All monkeys showed FG labeling in the host SN when FG labeling was injected on the same side. These data show that grafted dopaminergic neurons can extend neurites to a distant target releasing an elevated concentration of GDNF, and suggest that grafted neurons can be placed into appropriate loci for potential tract reconstruction.
Collapse
Affiliation(s)
- D E Redmond
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut 06520, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
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: 3.4] [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
|
27
|
Brunet JF, Redmond DE, Bloch J. Primate adult brain cell autotransplantation, a pilot study in asymptomatic MPTP-treated monkeys. Cell Transplant 2009; 18:787-99. [PMID: 19500480 DOI: 10.3727/096368909x470847] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Autologous brain cell transplantation might be useful for repairing lesions and restoring function of the central nervous system. We have demonstrated that adult monkey brain cells, obtained from cortical biopsy and kept in culture for a few weeks, exhibit neural progenitor characteristics that make them useful for brain repair. Following MPTP treatment, primates were dopamine depleted but asymptomatic. Autologous cultured cells were reimplanted into the right caudate nucleus of the donor monkey. Four months after reimplantation, histological analysis by stereology and TH immunolabeling showed that the reimplanted cells successfully survived, bilaterally migrated in the whole striatum, and seemed to have a neuroprotection effect over time. These results may add a new strategy to the field of brain neuroprotection or regeneration and could possibly lead to future clinical applications.
Collapse
Affiliation(s)
- Jean-François Brunet
- Department of Neurosurgery, Lausanne University Hospital, 1011 Lausanne, Switzerland.
| | | | | |
Collapse
|
28
|
Troche SJ, Trenkwalder C, Morelli-Canelo M, Gibbons H, Rammsayer TH. To the influence of general slowing and medication on identity- and location-based priming effects in patients with Parkinson's disease. J Neuropsychol 2009; 3:147-68. [PMID: 19338711 DOI: 10.1348/174866408x306872] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Previous studies on inhibitory mechanisms assessed by negative priming (NP) paradigms in patients suffering from Parkinson's disease (PD) have yielded highly ambiguous results. The present study examined two possible reasons for this heterogeneity: general slowing and anti-Parkinsonian medication. Their effects on identity and location NP and positive priming (PP) were investigated. Twenty medicated PD patients and 20 PD patients after drug withdrawal were compared to 20 sex- and age-matched healthy controls. The influence of PD patients' general slowing on priming effects was statistically controlled. Location NP was found not to be affected by PD, whereas identity NP was reduced in medicated PD patients compared to non-medicated PD patients and healthy controls. At first, identity and location PP appeared to be enhanced in both PD groups. After controlling for general slowing, however, differences between PD patients and healthy controls disappeared. These findings endorse the notion that uncontrolled effects of both, PD-related general slowing and anti-Parkinsonian medication may have contributed to previously conflicting results on priming effects in PD patients.
Collapse
Affiliation(s)
- S J Troche
- Institute for Psychology, University of Bern, Bern, Switzerland.
| | | | | | | | | |
Collapse
|
29
|
Xi J, Zhang SC. Stem cells in development of therapeutics for Parkinson's disease: a perspective. J Cell Biochem 2009; 105:1153-60. [PMID: 18980212 DOI: 10.1002/jcb.21916] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Using Parkinson's disease as a prototype of neurodegenerative diseases, we propose applications of human stem cells in the development of therapeutics for neurodegenerative diseases. First, in vitro differentiation of human stem cells offers a versatile model for dissecting molecular interactions underlying human dopamine (DA) neuron specification, which may form a foundation for instigating regeneration of DA neurons from progenitors that reside in the brain. Second, stem cells derived from diseased cells or through genetic modification can serve as a platform for unraveling biochemical processes that lead to the cellular pathogenesis of degeneration. This may in turn serve as a template for identifying or developing therapeutics for slowing, stopping, or reversing the disease process. And finally, stem cells, particularly those induced from patients' own cells, provide a reliable source of DA neurons for cell-based therapy.
Collapse
Affiliation(s)
- Jiajie Xi
- Department of Human Anatomy and Histology, Institute of Stem Cells and Tissue Engineering, Fudan University Shanghai Medical School, Shanghai, China
| | | |
Collapse
|
30
|
Grunwell J, Illes J, Karkazis K. Advancing Neuroregenerative Medicine: a Call for Expanded Collaboration Between Scientists and Ethicists. NEUROETHICS-NETH 2008. [DOI: 10.1007/s12152-008-9025-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
31
|
Abstract
Non-human primates have a small but important role in basic and translational biomedical research, owing to similarities with human beings in physiology, cognitive capabilities, neuroanatomy, social complexity, reproduction, and development. Although non-human primates have contributed to many areas of biomedical research, we review here their unique contributions to work in neuroscience, and focus on four domains: Alzheimer's disease, neuroAIDS, Parkinson's disease, and stress. Our discussion includes, for example, the role of non-human primates in development of new treatments (eg, stem cells, gene transfer) before phase I clinical trials in patients; basic research on disease pathogenesis; and understanding neurobehavioural outcomes resulting from genotype-environment interactions.
Collapse
Affiliation(s)
- John P Capitanio
- California National Primate Research Center and Department of Psychology, University of California, Davis, CA 95616, USA.
| | | |
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: 1.3] [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
|
Redmond DE, Bjugstad KB, Teng YD, Ourednik V, Ourednik J, Wakeman DR, Parsons XH, Gonzalez R, Blanchard BC, Kim SU, Gu Z, Lipton SA, Markakis EA, Roth RH, Elsworth JD, Sladek JR, Sidman RL, Snyder EY. Behavioral improvement in a primate Parkinson's model is associated with multiple homeostatic effects of human neural stem cells. Proc Natl Acad Sci U S A 2007; 104:12175-80. [PMID: 17586681 PMCID: PMC1896134 DOI: 10.1073/pnas.0704091104] [Citation(s) in RCA: 266] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Stem cells have been widely assumed to be capable of replacing lost or damaged cells in a number of diseases, including Parkinson's disease (PD), in which neurons of the substantia nigra (SN) die and fail to provide the neurotransmitter, dopamine (DA), to the striatum. We report that undifferentiated human neural stem cells (hNSCs) implanted into 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated Parkinsonian primates survived, migrated, and had a functional impact as assessed quantitatively by behavioral improvement in this DA-deficit model, in which Parkinsonian signs directly correlate to reduced DA levels. A small number of hNSC progeny differentiated into tyrosine hydroxylase (TH) and/or dopamine transporter (DAT) immunopositive cells, suggesting that the microenvironment within and around the lesioned adult host SN still permits development of a DA phenotype by responsive progenitor cells. A much larger number of hNSC-derived cells that did not express neuronal or DA markers was found arrayed along the persisting nigrostriatal path, juxtaposed with host cells. These hNSCs, which express DA-protective factors, were therefore well positioned to influence host TH+ cells and mediate other homeostatic adjustments, as reflected in a return to baseline endogenous neuronal number-to-size ratios, preservation of extant host nigrostriatal circuitry, and a normalizing effect on alpha-synuclein aggregation. We propose that multiple modes of reciprocal interaction between exogenous hNSCs and the pathological host milieu underlie the functional improvement observed in this model of PD.
Collapse
Affiliation(s)
- D. Eugene Redmond
- Departments of *Psychiatry
- Neurosurgery, and
- To whom correspondence may be addressed. E-mail: , , or
| | | | - Yang D. Teng
- Departments of Neurology and Neurosurgery, Harvard Medical School, Children's Hospital and Beth Israel Deaconess Medical Center, Division of Spinal Cord Injury Research, Veterans Affairs Boston Healthcare System, Boston, MA 02115
| | - Vaclav Ourednik
- Departments of Neurology and Neurosurgery, Harvard Medical School, Children's Hospital and Beth Israel Deaconess Medical Center, Division of Spinal Cord Injury Research, Veterans Affairs Boston Healthcare System, Boston, MA 02115
| | - Jitka Ourednik
- Departments of Neurology and Neurosurgery, Harvard Medical School, Children's Hospital and Beth Israel Deaconess Medical Center, Division of Spinal Cord Injury Research, Veterans Affairs Boston Healthcare System, Boston, MA 02115
| | - Dustin R. Wakeman
- **Burnham Institute for Medical Research, La Jolla, CA 92037
- Biomedical Sciences and Molecular Pathology Programs, University of California at San Diego, La Jolla, CA 92093
| | | | - Rodolfo Gonzalez
- **Burnham Institute for Medical Research, La Jolla, CA 92037
- Biomedical Sciences and Molecular Pathology Programs, University of California at San Diego, La Jolla, CA 92093
| | | | - Seung U. Kim
- Department of Neurology, University of British Columbia, Vancouver, BC, Canada V6T 2B5; and
| | - Zezong Gu
- **Burnham Institute for Medical Research, La Jolla, CA 92037
| | | | | | - Robert H. Roth
- Departments of *Psychiatry
- Pharmacology, Yale University School of Medicine, New Haven, CT 06510
| | - John D. Elsworth
- Departments of *Psychiatry
- Pharmacology, Yale University School of Medicine, New Haven, CT 06510
| | - John R. Sladek
- Department of Psychiatry, University of Colorado, Aurora, CO 80045
| | - Richard L. Sidman
- Departments of Neurology and Neurosurgery, Harvard Medical School, Children's Hospital and Beth Israel Deaconess Medical Center, Division of Spinal Cord Injury Research, Veterans Affairs Boston Healthcare System, Boston, MA 02115
- To whom correspondence may be addressed. E-mail: , , or
| | - Evan Y. Snyder
- Departments of Neurology and Neurosurgery, Harvard Medical School, Children's Hospital and Beth Israel Deaconess Medical Center, Division of Spinal Cord Injury Research, Veterans Affairs Boston Healthcare System, Boston, MA 02115
- **Burnham Institute for Medical Research, La Jolla, CA 92037
- To whom correspondence may be addressed. E-mail: , , or
| |
Collapse
|
34
|
Wakeman DR, Crain AM, Snyder EY. Large animal models are critical for rationally advancing regenerative therapies. Regen Med 2007; 1:405-13. [PMID: 17465832 PMCID: PMC2905042 DOI: 10.2217/17460751.1.4.405] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Dustin R Wakeman
- Biomedical Sciences Graduate Program, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, USA
- Burnham Institute for Medical Research, 10901 North Torrey Pines RD, La Jolla CA 92037, USA
| | - Andrew M Crain
- Biomedical Sciences Graduate Program, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, USA
- Burnham Institute for Medical Research, 10901 North Torrey Pines RD, La Jolla CA 92037, USA
| | - Evan Y Snyder
- Burnham Institute for Medical Research, 10901 North Torrey Pines RD, La Jolla CA 92037, USA
- Correspondence: ; 858-646-3158 Fax: 858-713-6273
| |
Collapse
|
35
|
Greely HT, Cho MK, Hogle LF, Satz DM. Thinking about the human neuron mouse. THE AMERICAN JOURNAL OF BIOETHICS : AJOB 2007; 7:27-40. [PMID: 17497502 PMCID: PMC2220020 DOI: 10.1080/15265160701290371] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
|
36
|
Abstract
Nonhuman primate (NHP) models of Parkinson's disease (PD) play an essential role in the understanding of PD pathophysiology and the assessment of PD therapies. NHP research enabled the identification of environmental risk factors for the development of PD. Electrophysiological studies in NHP models of PD identified the neural circuit responsible for PD motor symptoms, and this knowledge led to the development of subthalamic surgical ablation and deep brain stimulation. Similar to human PD patients, parkinsonian monkeys are responsive to dopamine replacement therapies and present complications associated with their long-term use, a similarity that facilitated the assessment of new symptomatic treatments, such as dopaminergic agonists. New generations of compounds and novel therapies that use directed intracerebral delivery of drugs, cells, and viral vectors benefit from preclinical evaluation in NHP models of PD. There are several NHP models of PD, each with characteristics that make it suitable for the study of different aspects of the disease or potential new therapies. Investigators who use the models and peer scientists who evaluate their use need information about the strengths and limitations of the different PD models and their methods of evaluation. This article provides a critical review of available PD monkey models, their utilization, and how they compare to emerging views of PD as a multietiologic, multisystemic disease. The various models are particularly useful for representing different aspects of PD at selected time points. This conceptualization provides clues for the development of new NHP models and facilitates the clinical translation of findings. As ever, successful application of any model depends on matching the model to the scientific question to be answered. Adequate experimental designs, with multiple outcome measures of clinical relevance and an appropriate number of animals, are essential to minimize the limitations of models and increase their predictive clinical validity.
Collapse
Affiliation(s)
- Marina E Emborg
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Court, Madison, WI 53715, USA.
| |
Collapse
|
37
|
Skinner SJM, Geaney MS, Rush R, Rogers ML, Emerich DF, Thanos CG, Vasconcellos AV, Tan PLJ, Elliott RB. Choroid plexus transplants in the treatment of brain diseases. Xenotransplantation 2006; 13:284-8. [PMID: 16768721 DOI: 10.1111/j.1399-3089.2006.00310.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The choroid plexus (CP) produces and secretes numerous biologically active neurotrophic factors into the cerebrospinal fluid (CSF). These circulate throughout the brain and spinal cord, maintaining neuronal networks and associated cells. In neurodegenerative disease and in acute brain injury there is local up-regulation of neurotrophin production close to the site of the lesion. Treatment by direct injection of neurotrophins and growth factors close to these lesion sites has repeatedly been demonstrated to improve recovery. It has therefore been proposed that transplanting viable choroid plexus cells close to the lesion might provide a novel means for continuous delivery of these molecules directly to the site of injury. Recent publications describe how transplanted CP, either free or in an immunoprotected encapsulated form, deliver therapeutic molecules to the desired site. This review briefly describes the accumulated evidence that CP cells support neuronal cells in vitro and have therapeutic properties when transplanted to treat acute and chronic brain disease and injury in animal models.
Collapse
|
38
|
Vander Borght T. Molecular imaging of cell transplantation in Parkinson's disease. Eur J Nucl Med Mol Imaging 2006; 33:403-6. [PMID: 16450137 DOI: 10.1007/s00259-005-0041-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
39
|
Morrow BA, Redmond DE, Roth RH, Elsworth JD. Development of A9/A10 dopamine neurons during the second and third trimesters in the African green monkey. J Comp Neurol 2005; 488:215-23. [PMID: 15924344 DOI: 10.1002/cne.20599] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Disruption in the development of dopamine-containing neurons has been postulated to underlie several CNS disorders. However, there have been no quantitative studies on the normal development of primate dopamine neurons. Thus, the fetal maturation of primate midbrain dopamine neurons was examined to establish changes that occur in the A9/A10 groups during the second and third trimesters. Eleven fetal African green monkey midbrains were immunostained for tyrosine hydroxylase (TH-ir) as a marker for dopamine neurons and quantified using stereological techniques (nucleator method). The number and size of defined dopamine neurons and the volume occupied by A9/A10 neurons increased in near linear fashion throughout the term. The estimated number of defined dopamine neurons in each hemisphere rose from approximately 50,000 at embryonic day (E) 70 to 225,000 at birth (E165), similar to the adult population. The size and the area occupied by them at birth were, however, well below the estimated adult levels. Additionally, the younger fetal midbrains had far less diversity in dopamine cell volumes compared with older fetuses and adult brains. Until midway through gestation (E81), clusters of apparently immature midbrain TH-ir cells were observed, but could not be counted. Even though the majority of cells destined to become dopamine neurons are generated in the first trimester, phenotypical maturation of A9 and A10 cell bodies continues steadily throughout gestation and extends well into the postnatal period. These data have relevance to transplantation studies that employ fetal dopaminergic grafts, and to disorders hypothesized to result from damage to developing midbrain dopamine neurons.
Collapse
Affiliation(s)
- Bret A Morrow
- Neuropsychopharmacology Research Unit, Departments of Psychiatry and Pharmacology, Yale University School of Medicine, New Haven, CN 06511, USA
| | | | | | | |
Collapse
|
40
|
Qiu G, Seiler MJ, Mui C, Arai S, Aramant RB, de Juan E, Sadda S. Photoreceptor differentiation and integration of retinal progenitor cells transplanted into transgenic rats. Exp Eye Res 2005; 80:515-25. [PMID: 15781279 DOI: 10.1016/j.exer.2004.11.001] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2004] [Accepted: 11/05/2004] [Indexed: 11/22/2022]
Abstract
Previous studies evaluating neural stem cells transplanted into the mature retina have demonstrated limited levels of graft-host integration and photoreceptor differentiation. The purpose of this investigation is to enhance photoreceptor cell differentiation and integration of retinal progenitor cells (RPC) following subretinal transplantation into retinal degenerate rats by optimization of isolation, expansion, and transplantation procedures. RPCs were isolated from human placental alkaline phosphatase (hPAP)-positive embryonic day 17 (E17) rat retina and expanded in serum-free defined media. RPCs at passage 2 underwent in vitro induction with all trans retinoic acid or were transplanted into the subretinal space of post-natal day (P) 17 S334ter-3 and S334ter-5 transgenic rats. Animals were examined post-operatively by ophthalmoscopy and optical coherence tomography (OCT) at weeks 1 and 4. Differentiation profiles of RPCs, both in vitro and in vivo were analysed microscopically by immunohistochemistry for various retinal cell specific markers. Our results demonstrated that the majority of passage 2 RPCs differentiated into retina-specific neurons expressing rhodopsin after in vitro induction. Following subretinal transplantation, grafted cells formed a multi-layer cellular sheet in the subretinal space in both S334ter-3 and S334ter-5 rats. Prominent retina-specific neuronal differentiation was observed in both rat lines as evidenced by recoverin or rhodopsin staining in 80% of grafted cells. Less than 5% of the grafted cells expressed glial fibrillary acidic protein. Synapsin-1 (label for nerve terminals) positive neural processes were present at the graft-host interface. Expression profiles of the grafted RPCs were similar to those of RPCs induced to differentiate in vitro using all-trans retinoic acid. In contrast to our previous study, grafted RPCs can demonstrate extensive rhodopsin expression, organize into layers, and show some features of apparent integration with the host retina following subretinal transplantation in slow and fast retinal degenerate rats. The similarity of the in vitro and in vivo RPC differentiation profiles suggests that intrinsic signals may have a significant contribution to RPC cell fate determination.
Collapse
Affiliation(s)
- Guanting Qiu
- Department of Ophthalmology, Doheny Retina Institute, Keck School of Medicine, University of Southern California, 1450 San Pablo St. DEI-3600, Los Angeles, CA 90033, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Alessandri G, Pagano S, Bez A, Benetti A, Pozzi S, Iannolo G, Baronio M, Invernici G, Caruso A, Muneretto C, Bisleri G, Parati E. Isolation and culture of human muscle-derived stem cells able to differentiate into myogenic and neurogenic cell lineages. Lancet 2004; 364:1872-83. [PMID: 15555667 DOI: 10.1016/s0140-6736(04)17443-6] [Citation(s) in RCA: 151] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Skeletal-muscle-derived stem cells seem to be a distinct population of immature progenitors of satellite cells, but their functional properties remain unclear, especially in human adult tissue. We investigated their differentiation in samples of skeletal muscle obtained from adults undergoing cardiovascular surgery. METHODS Samples were obtained from the brachioradialis muscle of 12 patients in whom the radial artery was the conduit for myocardial revascularisation. The stem cells were isolated by a procedure similar to that used for rat gastrocnemius and cultured in medium optimised for growth of neural stem cells. Cytometry was used for phenotypic characterisation and immunocytochemistry and RT-PCR to assess differentiation. Immunohistochemistry was used to examine engraftment of skeletal-muscle-derived stem cells into injured rat spinal cord. FINDINGS The skeletal-muscle stem cells consisted of two distinct types: one with the typical spindle morphology of satellite cells, the other of rounded cells. Some cultures could be maintained for longer than 6 months. The cells were mainly positive for desmin and to a lesser extent CD105, vimentin, and AC133/CD133, but negative for FLK-1/KDR, CD34, CD31, CD45, von Willebrand factor, Ve-cadherins, and BCL2. After in-vitro differentiation, the cells were able to organise skeletal-muscle fibres and stained positively for striated-muscle actin, smooth-muscle actin, and desmin. Moreover, they differentiated into astrocytes and neurons, as confirmed by positive staining for characteristic proteins. INTERPRETATION Adult human skeletal muscle includes a population of progenitor stem cells that can generate cells of the same lineage and cells with neurogenic properties. Muscle may therefore be a tissue source for the isolation of pluripotent stem cells for development of cell-based therapies for human myogenic and neurogenic diseases.
Collapse
Affiliation(s)
- Giulio Alessandri
- Laboratory of Neurobiology and Neuroregenerative Therapies, Carlo Besta Neurological Institute, Via Celoria 11, 20131 Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Basic science advances in spinal cord injury and regeneration research have led to a variety of novel experimental therapeutics designed to promote functionally effective axonal regrowth and sprouting. Among these interventions are cell-based approaches involving transplantation of neural and non-neural tissue elements that have potential for restoring damaged neural pathways or reconstructing intraspinal synaptic circuitries by either regeneration or neuronal/glial replacement. Notably, some of these strategies (e.g., grafts of peripheral nerve tissue, olfactory ensheathing glia, activated macrophages, marrow stromal cells, myelin-forming oligodendrocyte precursors or stem cells, and fetal spinal cord tissue) have already been translated to the clinical arena, whereas others have imminent likelihood of bench-to-bedside application. Although this progress has generated considerable enthusiasm about treating what once was thought to be a totally incurable condition, there are many issues to be considered relative to treatment safety and efficacy. The following review reflects on different experimental applications of intraspinal transplantation with consideration of the underlying pathological, pathophysiological, functional, and neuroplastic responses to spinal trauma that such treatments may target along with related issues of procedural and biological safety. The discussion then moves to an overview of ongoing and completed clinical trials to date. The pros and cons of these endeavors are considered, as well as what has been learned from them. Attention is primarily directed at preclinical animal modeling and the importance of patterning clinical trials, as much as possible, according to laboratory experiences.
Collapse
Affiliation(s)
- Paul J Reier
- College of Medicine and McKnight Brain Institute, University of Florida, Gainesville, Florida 32610, USA.
| |
Collapse
|
43
|
Cellular transplantation strategies for spinal cord injury and translational neurobiology. Neurotherapeutics 2004. [DOI: 10.1007/bf03206629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
44
|
Borlongan CV, Hadman M, Sanberg CD, Sanberg PR. Central nervous system entry of peripherally injected umbilical cord blood cells is not required for neuroprotection in stroke. Stroke 2004; 35:2385-9. [PMID: 15345799 DOI: 10.1161/01.str.0000141680.49960.d7] [Citation(s) in RCA: 322] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE To date, stem cell graft-mediated neuroprotection is equated with graft survival and secretion of neurotrophic factors in the brain. Here, we examined whether neuroprotection by systemically delivered human umbilical cord blood (HUCB) cells was dependent on their entry into the central nervous system in a rodent model of acute stroke. METHODS Adult male Sprague-Dawley rats were subjected to right middle cerebral artery occlusion for 60 minutes. During the 1-hour occlusion, animals were randomly assigned to 1 of the following treatments: intravenous injection of HUCB (a subtherapeutic dose of 200,000 cells in 10 microL) with blood-brain barrier (BBB) permeabilizer (1.1 mol/L mannitol at 4 degrees C) or vehicle, intravenous vehicle alone, or intravenous mannitol alone. Behavioral tests, using elevated body swing test and passive avoidance test, were conducted at day 3 poststroke, and thereafter, animals were euthanized for: (1) immunohistochemical examination of HUCB, which were lentivirally labeled with green fluorescent protein; (2) cerebral infarction analysis using 2,3,5-triphenyl-tetrazolium chloride; and (3) enzyme-linked immunosorbent assay of trophic factors within the striatal region. RESULTS We did not detect intravenously administered low dose of HUCB cells in the brains of animals at day 3 after stroke even when cells were coinfused with a BBB permeabilizer (mannitol). However, HUCB-mannitol treatment significantly increased brain levels of neurotrophic factors, which correlated positively with reduced cerebral infarcts and improved behavioral functions. CONCLUSIONS Our data show that central nervous system availability of grafted cells is not a prerequisite for acute neuroprotection provided that therapeutic molecules secreted by these cells could cross the BBB.
Collapse
Affiliation(s)
- Cesar V Borlongan
- Department of Neurology, Medical College of Georgia, Augusta, GA 30912-3200, USA.
| | | | | | | |
Collapse
|
45
|
Modo M, Roberts TJ, Sandhu JK, Williams SCR. In vivo monitoring of cellular transplants by magnetic resonance imaging and positron emission tomography. Expert Opin Biol Ther 2004; 4:145-55. [PMID: 14998774 DOI: 10.1517/14712598.4.2.145] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Cellular loss is a common pathological observation in many disease conditions. Recent evidence that these cells can be replaced has generated huge excitement over possible clinical applications. The use of stem or progenitor cells, which can differentiate into site-appropriate phenotypes required to "repair" the damaged tissue, has already demonstrated potential in animal models, but many aspects of this novel treatment strategy require further elucidation. Most importantly, the monitoring of the safety of cellular transplants in patients remains a challenge. Traditional histological methods do not address the dynamic nature of transplant-induced recovery and highlight the necessity of in vivo imaging to probe the survival, migration and functional consequences of transplanted cells. This paper reviews how non-invasive imaging technology can be used to serially assess intact living organisms in order to visualise and monitor cellular transplants.
Collapse
Affiliation(s)
- Michel Modo
- Neuroimaging Research Group P042, Department of Neurology, Institute of Psychiatry, King's College London, De Crespigny Park, London SE5 8AF, U.K.
| | | | | | | |
Collapse
|
46
|
Borlongan CV, Sumaya I, Moss D, Kumazaki M, Sakurai T, Hida H, Nishino H. Melatonin-secreting pineal gland: a novel tissue source for neural transplantation therapy in stroke. Cell Transplant 2004; 12:225-34. [PMID: 12797377 DOI: 10.3727/000000003108746786] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Chronic systemic melatonin treatment attenuates abnormalities produced by occlusion of middle cerebral artery (MCA) in adult rats. Because the pineal gland secretes high levels of melatonin, we examined in the present study whether transplantation of pineal gland exerted similar protective effects in MCA-occluded adult rats. Animals underwent same-day MCA occlusion and either intrastriatal transplantation of pineal gland (harvested from 2-month-old rats) or vehicle infusion. Behavioral tests (from day of surgery to 3 days posttransplantation) revealed that transplanted stroke rats displayed significantly less motor asymmetrical behaviors than vehicle-infused stroke rats. Histological analysis at 3 days posttransplantation revealed that transplanted stroke rats had significantly smaller cerebral infarction than vehicle-infused rats. Additional experiments showed that pinealectomy affected transplantation outcome, in that transplantation of pineal gland only protected against stroke-induced deficits in stroke animals with intact pineal gland, but not in pinealectomized stroke rats. Interestingly, nonpinealectomized vehicle-infused stroke rats, as well as pinealectomized transplanted stroke rats, had significantly lower melatonin levels in the cerebrospinal fluid than nonpinealectomized transplanted stroke rats. We conclude that intracerebral transplantation of pineal gland, in the presence of host intact pineal gland, protected against stroke, possibly through secretion of melatonin.
Collapse
Affiliation(s)
- C V Borlongan
- Neurology/Institute of Molecular Medicine & Genetics/School of Graduate Studies, Medical College of Georgia, Augusta, GA 30912-3200, USA.
| | | | | | | | | | | | | |
Collapse
|
47
|
Emgård M, Hallin U, Karlsson J, Bahr BA, Brundin P, Blomgren K. Both apoptosis and necrosis occur early after intracerebral grafting of ventral mesencephalic tissue: a role for protease activation. J Neurochem 2003; 86:1223-32. [PMID: 12911630 DOI: 10.1046/j.1471-4159.2003.01931.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Neural transplantation is an experimental treatment for Parkinson's disease. Widespread clinical application of the grafting technique is hampered by a relatively poor survival (around 10%) of implanted embryonic dopamine neurones. Earlier animal studies have indicated that a large proportion of the grafted cells die during graft tissue preparation and within the first few days after intracerebral implantation. The present study was designed to reveal the prevalence of cell death in rat intrastriatal grafts at 90 min, 1, 3, 6 and 42 days after implantation. We examined apoptotic cell death using semi-thin and paraffin sections stained with methylene blue and an antibody against activated caspase 3, respectively. We identified abundant apoptotic cell death up to 3 days after transplantation. In addition, we studied calpain activation using an antibody specific for calpain-cleaved fodrin. We report a peak in calpain activity 90 min after grafting. Surprisingly, we did not observe any significant difference in the number of dopaminergic neurones over time. The present results imply that grafted cells may be victims of either an early necrotic or a later apoptotic cell death and that there is substantial cell death as early as 90 min after implantation.
Collapse
Affiliation(s)
- M Emgård
- Section for Neuronal Survival, Wallenberg Neuroscience Center, Lund University, Sweden.
| | | | | | | | | | | |
Collapse
|
48
|
Abstract
Understanding the bases of aging-related cognitive decline remains a central challenge in neurobiology. Quantitative studies reveal little change in the number of neurons or synapses in most of the brain but their ongoing replacement is reduced, resulting in a significant loss of neuronal plasticity with senescence. Aging also may alter neuronal function and plasticity in ways that are not evident from anatomical studies of neurons and their connections. Since the nervous system is dependent upon a consistent blood supply, any aging-related changes in the microvasculature could affect neuronal function. Several studies suggest that, as the nervous system ages, there is a rarefaction of the microvasculature in some regions of the brain, as well as changes in the structure of the remaining vessels. These changes contribute to a decline in cerebral blood flow (CBF) that reduces metabolic support for neural signaling, particularly when levels of neuronal activity are high. In addition to direct effects on the microvasculature, aging reduces microvascular plasticity and the ability of the vessels to respond appropriately to changes in metabolic demand. This loss of microvascular plasticity has significance beyond metabolic support for neuronal signaling, since neurogenesis in the adult brain is regulated coordinately with capillary growth.
Collapse
Affiliation(s)
- David R Riddle
- Department of Neurobiology and Anatomy, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157-1010, USA.
| | | | | |
Collapse
|