1
|
Krishnan V, Xu J, Mendoza AG, Koretsky A, Anderson SA, Pelled G. High-resolution MEMRI characterizes laminar specific ascending and descending spinal cord pathways in rats. J Neurosci Methods 2020; 340:108748. [PMID: 32335077 PMCID: PMC7281828 DOI: 10.1016/j.jneumeth.2020.108748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 04/19/2020] [Accepted: 04/19/2020] [Indexed: 11/17/2022]
Abstract
BACKGROUND The spinal cord is composed of nine distinct cellular laminae that currently can only be visualized by histological methods. Developing imaging methods that can visualize laminar architecture in-vivo is of significant interest. Manganese enhanced magnetic resonance imaging (MEMRI) yields valuable architectural and functional information about the brain and has great potential in characterizing neural pathways in the spinal cord. Here we apply MEMRI to visualize laminae architecture in the thoracic region of the spinal cord with ultra-high resolution. NEW METHOD Manganese chloride (MnCl2) was delivered systemically and imaging of the lumbar and thoracic spinal cord levels was acquired in high field, 11.7 T MRI scanner, 48 h following MnCl2 administration. RESULTS Here we demonstrate laminar specific signal enhancement in the spinal cord of rats administered with MnCl2 with 69 μm in-plane resolution. We also report reduced T1 values over time in MnCl2 groups across laminae IIX. COMPARISONS WITH EXISTING METHODS This is the first study to demonstrate that MEMRI is capable of identifying spinal laminae at a high resolution of 69 μm in a living animal. This would enable the visualization of architecture and function of distinct regions with improved resolution, in healthy and diseased animal models. CONCLUSIONS The regions with the largest T1 enhancements were observed to correspond to laminae that contain either high cell density or large motor neurons, making MEMRI an excellent tool for studying spinal cord architecture, physiology and function in different animal models.
Collapse
Affiliation(s)
- Vijai Krishnan
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, United States; The Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - Jiadi Xu
- Johns Hopkins Medicine Department of Radiology and Radiological Science, Baltimore, MD, United States
| | - Albert German Mendoza
- Johns Hopkins Medicine Department of Radiology and Radiological Science, Baltimore, MD, United States
| | - Alan Koretsky
- Laboratory of Functional and Molecular Imaging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Stasia A Anderson
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Galit Pelled
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, United States; The Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States; Department of Radiology, Michigan State University, East Lansing, MI, United States; Johns Hopkins Medicine Department of Radiology and Radiological Science, Baltimore, MD, United States.
| |
Collapse
|
2
|
Song Y, Su X, Firouzian KF, Fang Y, Zhang T, Sun W. Engineering of brain-like tissue constructs via 3D Cell-printing technology. Biofabrication 2020; 12:035016. [DOI: 10.1088/1758-5090/ab7d76] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
3
|
Xiao LY, Wang XR, Yang Y, Yang JW, Cao Y, Ma SM, Li TR, Liu CZ. Applications of Acupuncture Therapy in Modulating Plasticity of Central Nervous System. Neuromodulation 2018; 21:762-776. [PMID: 29111577 DOI: 10.1111/ner.12724] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 09/25/2017] [Accepted: 09/26/2017] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Acupuncture is widely applied for treatment of various neurological disorders. This manuscript will review the preclinical evidence of acupuncture in mediating neural plasticity, the mechanisms involved. MATERIALS AND METHODS We searched acupuncture, plasticity, and other potential related words at the following sites: PubMed, EMBASE, Cochrane Library, Chinese National Knowledge Infrastructure (CNKI), and VIP information data base. The following keywords were used: acupuncture, electroacupuncture, plasticity, neural plasticity, neuroplasticity, neurogenesis, neuroblast, stem cell, progenitor cell, BrdU, synapse, synapse structure, synaptogenesis, axon, axon regeneration, synaptic plasticity, LTP, LTD, neurotrophin, neurotrophic factor, BDNF, GDNF, VEGF, bFGF, EGF, NT-3, NT-4, NT-5, p75NTR, neurotransmitter, acetylcholine, norepinephrine, noradrenaline, dopamine, monamine. We assessed the effects of acupuncture on plasticity under pathological conditions in this review. RESULTS Relevant references were reviewed and presented to reflect the effects of acupuncture on neural plasticity. The acquired literatures mainly focused on neurogenesis, alterations of synapses, neurotrophins (NTs), and neurotranimitters. Acupuncture methods mentioned in this article include manual acupuncture and electroacupuncture. CONCLUSIONS The cumulative evidences demonstrated that acupuncture could induce neural plasticity in rodents exposed to cerebral ischemia. Neural plasticity mediated by acupuncture in other neural disorders, such as Alzheimer's disease, Parkinson's disease, and depression, were also investigated and there is evidence of positive role of acupuncture induced plasticity in these disorders as well. Mediation of neural plasticity by acupuncture is likely associated with its modulation on NTs and neurotransmitters. The exact mechanisms underlying acupuncture's effects on neural plasticity remain to be elucidated. Neural plasticity may be the potential bridge between acupuncture and the treatment of various neurological diseases.
Collapse
Affiliation(s)
- Ling-Yong Xiao
- Beijing University of Chinese Medicine, Beijing, China
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine, Beijing Key Laboratory of Acupuncture Neuromodulation, Capital Medical University, Beijing, China
| | - Xue-Rui Wang
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine, Beijing Key Laboratory of Acupuncture Neuromodulation, Capital Medical University, Beijing, China
| | - Ye Yang
- Beijing University of Chinese Medicine, Beijing, China
| | - Jing-Wen Yang
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine, Beijing Key Laboratory of Acupuncture Neuromodulation, Capital Medical University, Beijing, China
| | - Yan Cao
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine, Beijing Key Laboratory of Acupuncture Neuromodulation, Capital Medical University, Beijing, China
| | - Si-Ming Ma
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine, Beijing Key Laboratory of Acupuncture Neuromodulation, Capital Medical University, Beijing, China
| | - Tian-Ran Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Cun-Zhi Liu
- Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
4
|
Liang Y, Zhang J, Walczak P, Bulte JWM. Quantification of motor neuron loss and muscular atrophy in ricin-induced focal nerve injury. J Neurosci Methods 2018; 308:142-150. [PMID: 30056087 PMCID: PMC6251705 DOI: 10.1016/j.jneumeth.2018.07.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 07/07/2018] [Accepted: 07/18/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Intrasciatic nerve injection of the Ricinus communis agglutinin (RCA or ricin) causes degeneration of motor neurons (MNs) with functional deficits, such as those that occur in amyotrophic lateral sclerosis (ALS). The objective of this study was to develop a new comprehensive platform for quantitative evaluation of MN loss, muscular atrophy and behavioral deficits using different ricin injection regimens. NEW METHOD Fluorogold (FG)-guided stereological quantification of MNs, in vivo magnetic resonance imaging (MRI) of muscular atrophy, and CatWalk behavioral testing were used to evaluate the outcome of rats treated with different ricin regimens (RCA60 0.5 μg, RCA60 3 μg, and RCA120 6 μg) as animal models of MN degeneration. RESULTS FG-guided stereological counting of MNs enabled identification, dissection and robust quantification of ricin-induced MN loss. The RCA60 0.5 μg and RCA120 6 μg regimens were found to be best suited as preclinical MN depletion models, with a low mortality and a reproducible MN loss, accompanied by muscle atrophy and functional deficits evaluated by MRI and the CatWalk method, respectively. COMPARISON WITH EXISTING METHODS 1) Fluorogold neuronal tracing provides a robust and straightforward means for quantifying MN loss in the spinal cord; 2) MRI is well-suited to non-invasively assess muscle atrophy; and 3) The CatWalk method is more flexible than rotarod test for studying motor deficits. CONCLUSION Intrasciatic injection of RCA60 or RCA120 induces nerve injury and muscle atrophy, which can be properly evaluated by a comprehensive platform using FG-guided quantitative 3D topographic histological analysis, MRI and the CatWalk behavioral test.
Collapse
Affiliation(s)
- Yajie Liang
- Russell H. Morgan Dept. of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Jiangyang Zhang
- Russell H. Morgan Dept. of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Piotr Walczak
- Russell H. Morgan Dept. of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Department of Neurology and Neurosurgery, Faculty of Medical Sciences, University of Warmia and Mazury, Olsztyn, Poland
| | - Jeff W M Bulte
- Russell H. Morgan Dept. of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Dept. of Chemical & Biomolecular Engineering, The Johns Hopkins University Whiting School of Engineering, Baltimore, MD, 21205, USA; Dept. of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Dept of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
5
|
Therapeutic approaches for spinal muscular atrophy (SMA). Gene Ther 2017; 24:514-519. [DOI: 10.1038/gt.2017.45] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 05/09/2017] [Indexed: 12/13/2022]
|
6
|
McGinley LM, Sims E, Lunn JS, Kashlan ON, Chen KS, Bruno ES, Pacut CM, Hazel T, Johe K, Sakowski SA, Feldman EL. Human Cortical Neural Stem Cells Expressing Insulin-Like Growth Factor-I: A Novel Cellular Therapy for Alzheimer's Disease. Stem Cells Transl Med 2016; 5:379-91. [PMID: 26744412 PMCID: PMC4807660 DOI: 10.5966/sctm.2015-0103] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 11/19/2015] [Indexed: 01/05/2023] Open
Abstract
Alzheimer's disease (AD) is the most prevalent age-related neurodegenerative disorder and a leading cause of dementia. Current treatment fails to modify underlying disease pathologies and very little progress has been made to develop effective drug treatments. Cellular therapies impact disease by multiple mechanisms, providing increased efficacy compared with traditional single-target approaches. In amyotrophic lateral sclerosis, we have shown that transplanted spinal neural stem cells (NSCs) integrate into the spinal cord, form synapses with the host, improve inflammation, and reduce disease-associated pathologies. Our current goal is to develop a similar "best in class" cellular therapy for AD. Here, we characterize a novel human cortex-derived NSC line modified to express insulin-like growth factor-I (IGF-I), HK532-IGF-I. Because IGF-I promotes neurogenesis and synaptogenesis in vivo, this enhanced NSC line offers additional environmental enrichment, enhanced neuroprotection, and a multifaceted approach to treating complex AD pathologies. We show that autocrine IGF-I production does not impact the cell secretome or normal cellular functions, including proliferation, migration, or maintenance of progenitor status. However, HK532-IGF-I cells preferentially differentiate into gamma-aminobutyric acid-ergic neurons, a subtype dysregulated in AD; produce increased vascular endothelial growth factor levels; and display an increased neuroprotective capacity in vitro. We also demonstrate that HK532-IGF-I cells survive peri-hippocampal transplantation in a murine AD model and exhibit long-term persistence in targeted brain areas. In conclusion, we believe that harnessing the benefits of cellular and IGF-I therapies together will provide the optimal therapeutic benefit to patients, and our findings support further preclinical development of HK532-IGF-I cells into a disease-modifying intervention for AD.
Collapse
Affiliation(s)
- Lisa M McGinley
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Erika Sims
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - J Simon Lunn
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Osama N Kashlan
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Kevin S Chen
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Elizabeth S Bruno
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Crystal M Pacut
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Tom Hazel
- Neuralstem, Inc., Germantown, Maryland, USA
| | - Karl Johe
- Neuralstem, Inc., Germantown, Maryland, USA
| | - Stacey A Sakowski
- A. Alfred Taubman Medical Research Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA A. Alfred Taubman Medical Research Institute, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
7
|
Bonaventura G, Chamayou S, Liprino A, Guglielmino A, Fichera M, Caruso M, Barcellona ML. Different Tissue-Derived Stem Cells: A Comparison of Neural Differentiation Capability. PLoS One 2015; 10:e0140790. [PMID: 26517263 PMCID: PMC4627815 DOI: 10.1371/journal.pone.0140790] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 09/30/2015] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Stem cells are capable of self-renewal and differentiation into a wide range of cell types with multiple clinical and therapeutic applications. Stem cells are providing hope for many diseases that currently lack effective therapeutic methods, including strokes, Huntington's disease, Alzheimer's and Parkinson's disease. However, the paucity of suitable cell types for cell replacement therapy in patients suffering from neurological disorders has hampered the development of this promising therapeutic approach. AIM The innovative aspect of this study has been to evaluate the neural differentiation capability of different tissue-derived stem cells coming from different tissue sources such as bone marrow, umbilical cord blood, human endometrium and amniotic fluid, cultured under the same supplemented media neuro-transcription factor conditions, testing the expression of neural markers such as GFAP, Nestin and Neurofilaments using the immunofluorescence staining assay and some typical clusters of differentiation such as CD34, CD90, CD105 and CD133 by using the cytofluorimetric test assay. RESULTS Amniotic fluid derived stem cells showed a more primitive phenotype compared to the differentiating potential demonstrated by the other stem cell sources, representing a realistic possibility in the field of regenerative cell therapy suitable for neurodegenerative diseases.
Collapse
Affiliation(s)
- Gabriele Bonaventura
- Department of Pharmaceutical Science, Biochemistry Section, University of Catania, Catania, Italy
- Institute of Neurological Sciences, Italian National Research Council, Catania, Italy
| | - Sandrine Chamayou
- Unità di Medicina della Riproduzione, Fondazione Hera, Sant’Agata Li Battiati (CT), Italy
| | - Annalisa Liprino
- Department of Obstetrics and Gynecology and Radiological Sciences (OGiRA), University of Catania, Catania, Italy
| | - Antonino Guglielmino
- Unità di Medicina della Riproduzione, Fondazione Hera, Sant’Agata Li Battiati (CT), Italy
| | - Michele Fichera
- Department of Obstetrics and Gynecology and Radiological Sciences (OGiRA), University of Catania, Catania, Italy
| | - Massimo Caruso
- Department of Clinic and Molecular Biomedicine, University of Catania, Catania, Italy
| | - Maria Luisa Barcellona
- Department of Pharmaceutical Science, Biochemistry Section, University of Catania, Catania, Italy
| |
Collapse
|
8
|
Phan HC, Taylor JL, Hannon H, Howell R. Newborn screening for spinal muscular atrophy: Anticipating an imminent need. Semin Perinatol 2015; 39:217-29. [PMID: 25979781 DOI: 10.1053/j.semperi.2015.03.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Spinal muscular atrophy (SMA) is the most common genetic cause of infant mortality. Children with type I SMA typically die by the age of 2 years. Recent progress in gene modification and other innovative therapies suggest that improved outcomes may soon be forthcoming. In animal models, therapeutic intervention initiated before the loss of motor neurons alters SMA phenotype and increases lifespan. Presently, supportive care including respiratory, nutritional, physiatry, and orthopedic management can ameliorate clinical symptoms and improve survival rates if SMA is diagnosed early in life. Newborn screening could help optimize these potential benefits. A recent report demonstrated that SMA detection can be multiplexed at minimal additional cost with the assay for severe combined immunodeficiency, already implemented by many newborn screening programs. The public health community should remain alert to the rapidly changing developments in early detection and treatment of SMA.
Collapse
Affiliation(s)
- Han C Phan
- Department of Pediatrics, Emory University, Atlanta, GA.
| | | | - Harry Hannon
- Newborn Screening Consensus Committee, Clinical and Laboratory Standards Institute (CLSI), Wayne, PA
| | - Rodney Howell
- Miller School of Medicine, University of Miami, Miami, FL
| |
Collapse
|
9
|
Differentiation Potential of Human Chorion-Derived Mesenchymal Stem Cells into Motor Neuron-Like Cells in Two- and Three-Dimensional Culture Systems. Mol Neurobiol 2015; 53:1862-1872. [PMID: 25790953 DOI: 10.1007/s12035-015-9129-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 02/22/2015] [Indexed: 12/29/2022]
Abstract
Many people worldwide suffer from motor neuron-related disorders such as amyotrophic lateral sclerosis and spinal cord injuries. Recently, several attempts have been made to recruit stem cells to modulate disease progression in ALS and also regenerate spinal cord injuries. Chorion-derived mesenchymal stem cells (C-MSCs), used to be discarded as postpartum medically waste product, currently represent a class of cells with self renewal property and immunomodulatory capacity. These cells are able to differentiate into mesodermal and nonmesodermal lineages such as neural cells. On the other hand, gelatin, as a simply denatured collagen, is a suitable substrate for cell adhesion and differentiation. It has been shown that electrospinning of scaffolds into fibrous structure better resembles the physiological microenvironment in comparison with two-dimensional (2D) culture system. Since there is no report on potential of human chorion-derived MSCs to differentiate into motor neuron cells in two- and three-dimensional (3D) culture systems, we set out to determine the effect of retinoic acid (RA) and sonic hedgehog (Shh) on differentiation of human C-MSCs into motor neuron-like cells cultured on tissue culture plates (2D) and electrospun nanofibrous gelatin scaffold (3D).
Collapse
|
10
|
Knippenberg S, Rath KJ, Böselt S, Thau-Habermann N, Schwarz SC, Dengler R, Wegner F, Petri S. Intraspinal administration of human spinal cord-derived neural progenitor cells in the G93A-SOD1 mouse model of ALS delays symptom progression, prolongs survival and increases expression of endogenous neurotrophic factors. J Tissue Eng Regen Med 2015; 11:751-764. [PMID: 25641599 DOI: 10.1002/term.1972] [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: 03/14/2014] [Revised: 08/15/2014] [Accepted: 10/28/2014] [Indexed: 12/14/2022]
Abstract
Neural stem or progenitor cells are considered to be a novel therapeutic strategy for amyotrophic lateral sclerosis (ALS), based on their potential to generate a protective environment rather than to replace degenerating motor neurons. Following local injection to the spinal cord, neural progenitor cells may generate glial cells and release neurotrophic factors. In the present study, human spinal cord-derived neural progenitor cells (hscNPCs) were injected into the lumbar spinal cord of G93A-SOD1 ALS transgenic mice. We evaluated the potential effect of hscNPC treatment by survival analysis and behavioural/phenotypic assessments. Immunohistological and real-time PCR experiments were performed at a defined time point to study the underlying mechanisms. Symptom progression in hscNPC-injected mice was significantly delayed at the late stage of disease. On average, survival was only prolonged for 5 days. Animals treated with hscNPCs performed significantly better in motor function tests between weeks 18 and 19. Increased production of GDNF and IGF-1 mRNA was detectable in spinal cord tissue of hscNPC-treated mice. In summary, treatment with hscNPCs led to increased endogenous production of several growth factors and increased the preservation of innervated motor neurons but had only a small effect on overall survival. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
| | - Klaus Jan Rath
- Department of Neurology, Hannover Medical School, Germany.,Integriertes Forschungs- und Behandlungszentrum Transplantation (IFB-Tx), Hannover Medical School, Hannover, Germany
| | - Sebastian Böselt
- Department of Neurology, Hannover Medical School, Germany.,Integriertes Forschungs- und Behandlungszentrum Transplantation (IFB-Tx), Hannover Medical School, Hannover, Germany
| | - Nadine Thau-Habermann
- Department of Neurology, Hannover Medical School, Germany.,Centre for Systems Neuroscience, Hannover, Germany
| | - Sigrid C Schwarz
- German Centre for Neurodegenerative Diseases (DZNE), Technical University of Munich, Germany
| | - Reinhard Dengler
- Department of Neurology, Hannover Medical School, Germany.,Centre for Systems Neuroscience, Hannover, Germany
| | - Florian Wegner
- Department of Neurology, Hannover Medical School, Germany.,Centre for Systems Neuroscience, Hannover, Germany
| | - Susanne Petri
- Department of Neurology, Hannover Medical School, Germany.,Centre for Systems Neuroscience, Hannover, Germany.,Integriertes Forschungs- und Behandlungszentrum Transplantation (IFB-Tx), Hannover Medical School, Hannover, Germany
| |
Collapse
|
11
|
Dadon-Nachum M, Ben-Yaacov K, Ben-Zur T, Barhum Y, Yaffe D, Perlson E, Offen D. Transplanted modified muscle progenitor cells expressing a mixture of neurotrophic factors delay disease onset and enhance survival in the SOD1 mouse model of ALS. J Mol Neurosci 2014; 55:788-97. [PMID: 25330859 DOI: 10.1007/s12031-014-0426-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 09/16/2014] [Indexed: 12/14/2022]
Abstract
Neurotrophic factors (NTFs) are essential growth factor proteins that support the development, survival, and proper function of neurons. We have developed muscle progenitor cell (MPC) populations expressing brain-derived neurotrophic factor (BDNF), glial-derived neurotrophic factor (GDNF), vascular endothelial growth factor (VEGF), or insulin-like growth factor-1 (IGF-1). Transplantation of a mixture of such MPC populations (MPC-MIX) into the hind legs of SOD1 G93A transgenic mice (SOD1 mice), the commonly used model of ALS, delayed the onset of disease symptoms by 30 days and prolonged the average lifespan by 13 days. Treated mice also showed a decrease in the degeneration of neuromuscular junction and an increase in axonal survival. Cellular mechanism assays suggest a synergistic rescue effect of NTFs that involves the AKT and BAD signaling pathways. The results suggest that long-term delivery of a mixture of several NTFs by the transplantation of engineered MPC has a beneficial effect in the ALS mouse model.
Collapse
Affiliation(s)
- M Dadon-Nachum
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | | | | | |
Collapse
|
12
|
Zhang G, Zhao Y, Li XM, Kong J. Fetal cell microchimerism in the maternal mouse spinal cord. Neurosci Bull 2013; 30:81-9. [PMID: 24346789 DOI: 10.1007/s12264-013-1392-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 03/11/2013] [Indexed: 01/30/2023] Open
Abstract
Fetal cell microchimerism refers to the persistence of fetal cells in the maternal tissues following pregnancy. It has been detected in peripheral organs and the brain, but its existence in the spinal cord has not been reported. Our aim was to detect fetal cell microchimerism in the spinal cord of maternal mice. C57BL/6 female mice were crossed with GFP transgenic male mice and sacrificed after their first or third delivery. GFP-positive cells, which were presumably from fetuses whose fathers were GFP transgenic, were detected in the spinal cord by fluorescence microscopy and immunohistochemistry. PCR was also performed to detect GFP DNA, which must come from GFP hemizygous fetuses. We found GFP-positive cells and detectable GFP DNA in most of the maternal spinal cords. Twenty percent (1/5) of the mice that were only pregnant once had detectable fetal cells, while 80% (4/5) of those that were pregnant three times had detectable fetal cells. Some fetal cells, which not only emitted green fluorescence but also expressed NeuN, were detected in the spinal cords from maternal mice. These results indicate that fetal cells migrate into the spinal cord of a maternal mouse during and/or after the gestational period, and the fetal cells may differentiate into neurons in the spinal cord.
Collapse
Affiliation(s)
- Guohui Zhang
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, 075000, China
| | | | | | | |
Collapse
|
13
|
Meamar R, Nasr-Esfahani MH, Mousavi SA, Basiri K. Stem cell therapy in amyotrophic lateral sclerosis. J Clin Neurosci 2013; 20:1659-63. [PMID: 24148693 DOI: 10.1016/j.jocn.2013.04.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2012] [Revised: 01/08/2013] [Accepted: 04/14/2013] [Indexed: 02/07/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder of upper and lower motor neurons, characterized by progressive muscular atrophy and weakness which culminates in death within 2-5 years. Despite various hypotheses about the responsible mechanisms, the etiology of ALS remains incompletely understood. However, it has been recently postulated that stem cell therapy could potentially target several mechanisms responsible for the etiology of ALS and other nervous system disorders, and could be regarded as one of the most promising therapeutic strategies for ALS treatment. We present a brief review of different methods of stem cell therapy in ALS patients and discuss the results with different cell types and routes of administration.
Collapse
Affiliation(s)
- Rokhsareh Meamar
- Department of Medical Science, Najafabad Branch, Islamic Azad University, Isfahan, Iran
| | | | | | | |
Collapse
|
14
|
Parsons XH. Constraining the Pluripotent Fate of Human Embryonic Stem Cells for Tissue Engineering and Cell Therapy - The Turning Point of Cell-Based Regenerative Medicine. ACTA ACUST UNITED AC 2013. [PMID: 24926434 PMCID: PMC4051304 DOI: 10.9734/bbj/2013/4309] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
To date, the lack of a clinically-suitable source of engraftable human stem/progenitor cells with adequate neurogenic potential has been the major setback in developing safe and effective cell-based therapies for regenerating the damaged or lost CNS structure and circuitry in a wide range of neurological disorders. Similarly, the lack of a clinically-suitable human cardiomyocyte source with adequate myocardium regenerative potential has been the major setback in regenerating the damaged human heart. Given the limited capacity of the CNS and heart for self-repair, there is a large unmet healthcare need to develop stem cell therapies to provide optimal regeneration and reconstruction treatment options to restore normal tissues and function. Derivation of human embryonic stem cells (hESCs) provides a powerful in vitro model system to investigate molecular controls in human embryogenesis as well as an unlimited source to generate the diversity of human somatic cell types for regenerative medicine. However, realizing the developmental and therapeutic potential of hESC derivatives has been hindered by the inefficiency and instability of generating clinically-relevant functional cells from pluripotent cells through conventional uncontrollable and incomplete multi-lineage differentiation. Recent advances and breakthroughs in hESC research have overcome some major obstacles in bringing hESC therapy derivatives towards clinical applications, including establishing defined culture systems for de novo derivation and maintenance of clinical-grade pluripotent hESCs and lineage-specific differentiation of pluripotent hESCs by small molecule induction. Retinoic acid was identified as sufficient to induce the specification of neuroectoderm direct from the pluripotent state of hESCs and trigger a cascade of neuronal lineage-specific progression to human neuronal progenitors and neurons of the developing CNS in high efficiency, purity, and neuronal lineage specificity by promoting nuclear translocation of the neuronal specific transcription factor Nurr-1. Similarly, nicotinamide was rendered sufficient to induce the specification of cardiomesoderm direct from the pluripotent state of hESCs by promoting the expression of the earliest cardiac-specific transcription factor Csx/Nkx2.5 and triggering progression to cardiac precursors and beating cardiomyocytes with high efficiency. This technology breakthrough enables direct conversion of pluripotent hESCs into a large supply of high purity neuronal cells or heart muscle cells with adequate capacity to regenerate CNS neurons and contractile heart muscles for developing safe and effective stem cell therapies. Transforming pluripotent hESCs into fate-restricted therapy derivatives dramatically increases the clinical efficacy of graft-dependent repair and safety of hESC-derived cellular products. Such milestone advances and medical innovations in hESC research allow generation of a large supply of clinical-grade hESC therapy derivatives targeting for major health problems, bringing cell-based regenerative medicine to a turning point.
Collapse
Affiliation(s)
- Xuejun H Parsons
- San Diego Regenerative Medicine Institute, San Diego, CA 92109, USA. ; Xcelthera, San Diego, CA 92109, USA
| |
Collapse
|
15
|
Parsons XH. Constraining the Pluripotent Fate of Human Embryonic Stem Cells for Tissue Engineering and Cell Therapy - The Turning Point of Cell-Based Regenerative Medicine. ACTA ACUST UNITED AC 2013; 3:424-457. [PMID: 24926434 DOI: 10.9734/bbj/2013/4309#sthash.6d8rulbv.dpuf] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
To date, the lack of a clinically-suitable source of engraftable human stem/progenitor cells with adequate neurogenic potential has been the major setback in developing safe and effective cell-based therapies for regenerating the damaged or lost CNS structure and circuitry in a wide range of neurological disorders. Similarly, the lack of a clinically-suitable human cardiomyocyte source with adequate myocardium regenerative potential has been the major setback in regenerating the damaged human heart. Given the limited capacity of the CNS and heart for self-repair, there is a large unmet healthcare need to develop stem cell therapies to provide optimal regeneration and reconstruction treatment options to restore normal tissues and function. Derivation of human embryonic stem cells (hESCs) provides a powerful in vitro model system to investigate molecular controls in human embryogenesis as well as an unlimited source to generate the diversity of human somatic cell types for regenerative medicine. However, realizing the developmental and therapeutic potential of hESC derivatives has been hindered by the inefficiency and instability of generating clinically-relevant functional cells from pluripotent cells through conventional uncontrollable and incomplete multi-lineage differentiation. Recent advances and breakthroughs in hESC research have overcome some major obstacles in bringing hESC therapy derivatives towards clinical applications, including establishing defined culture systems for de novo derivation and maintenance of clinical-grade pluripotent hESCs and lineage-specific differentiation of pluripotent hESCs by small molecule induction. Retinoic acid was identified as sufficient to induce the specification of neuroectoderm direct from the pluripotent state of hESCs and trigger a cascade of neuronal lineage-specific progression to human neuronal progenitors and neurons of the developing CNS in high efficiency, purity, and neuronal lineage specificity by promoting nuclear translocation of the neuronal specific transcription factor Nurr-1. Similarly, nicotinamide was rendered sufficient to induce the specification of cardiomesoderm direct from the pluripotent state of hESCs by promoting the expression of the earliest cardiac-specific transcription factor Csx/Nkx2.5 and triggering progression to cardiac precursors and beating cardiomyocytes with high efficiency. This technology breakthrough enables direct conversion of pluripotent hESCs into a large supply of high purity neuronal cells or heart muscle cells with adequate capacity to regenerate CNS neurons and contractile heart muscles for developing safe and effective stem cell therapies. Transforming pluripotent hESCs into fate-restricted therapy derivatives dramatically increases the clinical efficacy of graft-dependent repair and safety of hESC-derived cellular products. Such milestone advances and medical innovations in hESC research allow generation of a large supply of clinical-grade hESC therapy derivatives targeting for major health problems, bringing cell-based regenerative medicine to a turning point.
Collapse
Affiliation(s)
- Xuejun H Parsons
- San Diego Regenerative Medicine Institute, San Diego, CA 92109, USA. ; Xcelthera, San Diego, CA 92109, USA
| |
Collapse
|
16
|
Abstract
The nervous system is characterized by its complex network of highly specialized cells that enable us to perceive stimuli from the outside world and react accordingly. The computational integration enabled by these networks remains to be elucidated, but appropriate sensory input, processing, and motor control are certainly essential for survival. Consequently, loss of nervous tissue due to injury or disease represents a considerable biomedical challenge. Stem cell research offers the promise to provide cells for nervous system repair to replace lost and damaged neural tissue and alleviate disease. We provide a protocol-based chapter on fundamental principles and procedures of pluripotent stem cell (PSC) differentiation and neural transplantation. Rather than detailed methodological step-by-step descriptions of these procedures, we provide an overview and highlight the most critical aspects and key steps of PSC neural induction, subtype specification in different in vitro systems, as well as neural cell transplantation to the central nervous system. We conclude with a summary of suitable readout methods including in vitro phenotypic analysis, histology, and functional analysis in vivo.
Collapse
|
17
|
Su H, Zhang W, Yang X, Qin D, Sang Y, Wu C, Wong WM, Yuan Q, So KF, Wu W. Neural Progenitor Cells Generate Motoneuron-Like Cells to Form Functional Connections with Target Muscles after Transplantation into the Musculocutaneous Nerve. Cell Transplant 2012; 21:2651-63. [DOI: 10.3727/096368912x654975] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Neural progenitor cells (NPCs) are suggested to be a valuable source of cell transplant in treatment of various neurological diseases because of their distinct attributes. They can be expanded and induced to differentiate in vitro. However, it remains uncertain whether in vitro expanded NPCs have the capacity to give rise to functional motoneurons after transplantation in vivo. Here, we showed that in vitro expanded NPCs, when transplanted into the musculocutaneous nerve, generated motoneuron-like cells that exhibited typical morphology with large cell bodies, expressed specific molecules, and extended axons to form functional connections with the target muscle. In contrast, transplanted NPCs failed to yield motoneurons in the injured ventral horn of the spinal cord. The results of the study demonstrate that NPCs have the potential to generate functional motoneurons in an appropriate environment. The distinct differentiating fate of NPCs in the musculocutaneous nerve and the injured ventral horn suggests the importance and necessity of modifying the host microenvironment in use of NPCs for cell replacement therapies for motoneuron diseases.
Collapse
Affiliation(s)
- Huanxing Su
- Department of Anatomy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Wenming Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Fujian Medical University, Fujian, China
| | - Xiaoying Yang
- Department of Anatomy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Dajiang Qin
- Department of Anatomy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Yanhua Sang
- Department of Anatomy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Chaoyang Wu
- Department of Orthopaedics, The First Affiliated Hospital of Fujian Medical University, Fujian, China
| | - Wai-Man Wong
- Department of Anatomy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Qiuju Yuan
- Department of Anatomy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Kwok-Fai So
- Department of Anatomy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- State Key Laboratory of Brain and Cognitive Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Joint Laboratory for Brain Function and Health (BFAH), Jinan University and The University of Hong Kong, Jinan University, Guangzhou, China
| | - Wutian Wu
- Department of Anatomy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- State Key Laboratory of Brain and Cognitive Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Joint Laboratory for Brain Function and Health (BFAH), Jinan University and The University of Hong Kong, Jinan University, Guangzhou, China
- Research Center of Reproduction, Development and Growth, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| |
Collapse
|
18
|
Teng YD, Yu D, Ropper AE, Li J, Kabatas S, Wakeman DR, Wang J, Sullivan MP, Redmond DE, Langer R, Snyder EY, Sidman RL. Functional multipotency of stem cells: a conceptual review of neurotrophic factor-based evidence and its role in translational research. Curr Neuropharmacol 2012; 9:574-85. [PMID: 22654717 PMCID: PMC3263453 DOI: 10.2174/157015911798376299] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Revised: 09/28/2010] [Accepted: 09/28/2010] [Indexed: 12/14/2022] Open
Abstract
We here propose an updated concept of stem cells (SCs), with an emphasis on neural stem cells (NSCs). The conventional view, which has touched principally on the essential property of lineage multipotency (e.g., the ability of NSCs to differentiate into all neural cells), should be broadened to include the emerging recognition of biofunctional multipotency of SCs to mediate systemic homeostasis, evidenced in NSCs in particular by the secretion of neurotrophic factors. Under this new conceptual context and taking the NSC as a leading example, one may begin to appreciate and seek the “logic” behind the wide range of molecular tactics the NSC appears to serve at successive developmental stages as it integrates into and prepares, modifies, and guides the surrounding CNS micro- and macro-environment towards the formation and self-maintenance of a functioning adult nervous system. We suggest that embracing this view of the “multipotency” of the SCs is pivotal for correctly, efficiently, and optimally exploiting stem cell biology for therapeutic applications, including reconstitution of a dysfunctional CNS.
Collapse
Affiliation(s)
- Yang D Teng
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Neuromuscular junction protection for the potential treatment of amyotrophic lateral sclerosis. Neurol Res Int 2012; 2012:379657. [PMID: 22919482 PMCID: PMC3423938 DOI: 10.1155/2012/379657] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Revised: 06/15/2012] [Accepted: 06/15/2012] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neuromuscular disease characterized by the progressive degeneration of upper and lower motor neurons (MNs), leading to muscular atrophy and eventual respiratory failure. ALS research has primarily focused on mechanisms regarding MN cell death; however, degenerative processes in the skeletal muscle, particularly involving neuromuscular junctions (NMJs), are observed in the early stages of and throughout disease progression. According to the "dying-back" hypothesis, NMJ degeneration may not only precede, but actively cause upper and lower MN loss. The importance of NMJ pathology has relatively received little attention in ALS, possibly because compensatory mechanisms mask NMJ loss for prolonged periods. Many mechanisms explaining NMJ degeneration have been proposed such as the disruption of anterograde/retrograde axonal transport, irregular cellular metabolism, and changes in muscle gene and protein expression. Neurotrophic factors, which are known to have neuroprotective and regenerative properties, have been intensely investigated for their therapeutic potential in both the preclinical and clinical setting. Additional research should focus on the potential of preserving NMJs in order to delay or prevent disease progression.
Collapse
|
20
|
Knippenberg S, Thau N, Dengler R, Brinker T, Petri S. Intracerebroventricular injection of encapsulated human mesenchymal cells producing glucagon-like peptide 1 prolongs survival in a mouse model of ALS. PLoS One 2012; 7:e36857. [PMID: 22745655 PMCID: PMC3380029 DOI: 10.1371/journal.pone.0036857] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Accepted: 04/15/2012] [Indexed: 12/11/2022] Open
Abstract
Background As pharmacological therapies have largely failed so far, stem cell therapy has recently come into the focus of ALS research. Neuroprotective potential was shown for several types of stem and progenitor cells, mainly due to release of trophic factors. In the present study, we assessed the effects of intracerebroventricular injection of glucagon-like peptide 1 (GLP-1) releasing mesenchymal stromal cells (MSC) in mutant SOD1 (G93A) transgenic mice. Methodology/Principal Findings To improve the neuroprotective effects of native MSC, they had been transfected with a plasmid vector encoding a GLP-1 fusion gene prior to the injection, as GLP-1 was shown to exhibit neuroprotective properties before. Cells were encapsulated and therefore protected against rejection. After intracerebroventricular injection of these GLP-1 MSC capsules in presymptomatic SOD1 (G93A) mice, we assessed possible protective effects by survival analysis, measurement of body weight, daily monitoring and evaluation of motor performance by rotarod and footprint analyses. Motor neuron numbers in the spinal cord as well as the amount of astrocytosis, microglial activation, heat shock response and neuronal nitric oxide synthase (nNOS) expression were analyzed by immunohistological methods. Treatment with GLP-1 producing MSC capsules significantly prolonged survival by 13 days, delayed symptom onset by 15 days and weight loss by 14 days and led to significant improvements in motor performance tests compared to vehicle treated controls. Histological data are mainly in favour of anti-inflammatory effects of GLP-1 producing MSC capsules with reduced detection of inflammatory markers and a significant heat shock protein increase. Conclusion/Significance Intracerebroventricular injection of GLP-1 MSC capsules shows neuroprotective potential in the SOD1 (G93A) mouse model.
Collapse
Affiliation(s)
- Sarah Knippenberg
- Department of Neurology, Hannover Medical School, Hannover, Germany.
| | | | | | | | | |
Collapse
|
21
|
Lindvall O, Barker RA, Brüstle O, Isacson O, Svendsen CN. Clinical translation of stem cells in neurodegenerative disorders. Cell Stem Cell 2012; 10:151-5. [PMID: 22305565 DOI: 10.1016/j.stem.2012.01.009] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Stem cells and their derivatives show tremendous potential for treating many disorders, including neurodegenerative diseases. We discuss here the challenges and potential for the translation of stem-cell-based approaches into treatments for Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Olle Lindvall
- Wallenberg Neuroscience Center, University Hospital, SE-221 84 Lund, Sweden.
| | | | | | | | | |
Collapse
|
22
|
Longitudinal tracking of human fetal cells labeled with super paramagnetic iron oxide nanoparticles in the brain of mice with motor neuron disease. PLoS One 2012; 7:e32326. [PMID: 22384217 PMCID: PMC3288077 DOI: 10.1371/journal.pone.0032326] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 01/25/2012] [Indexed: 12/20/2022] Open
Abstract
Stem Cell (SC) therapy is one of the most promising approaches for the treatment of Amyotrophic Lateral Sclerosis (ALS). Here we employed Super Paramagnetic Iron Oxide nanoparticles (SPIOn) and Hoechst 33258 to track human Amniotic Fluid Cells (hAFCs) after transplantation in the lateral ventricles of wobbler (a murine model of ALS) and healthy mice. By in vitro, in vivo and ex vivo approaches we found that: 1) the main physical parameters of SPIOn were maintained over time; 2) hAFCs efficiently internalized SPIOn into the cytoplasm while Hoechst 33258 labeled nuclei; 3) SPIOn internalization did not alter survival, cell cycle, proliferation, metabolism and phenotype of hAFCs; 4) after transplantation hAFCs rapidly spread to the whole ventricular system, but did not migrate into the brain parenchyma; 5) hAFCs survived for a long time in the ventricles of both wobbler and healthy mice; 6) the transplantation of double-labeled hAFCs did not influence mice survival.
Collapse
|
23
|
Induction of adipose-derived stem cell into motoneuron-like cells using selegiline as preinducer. Brain Res 2012; 1440:23-33. [PMID: 22284617 DOI: 10.1016/j.brainres.2011.12.051] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Revised: 12/14/2011] [Accepted: 12/26/2011] [Indexed: 12/12/2022]
Abstract
Cell therapy is one of the approaches taken to treatment of spinal cord disorders. In this study, adipose-derived stem cells (ADSCs) were induced to form motoneuron-like cells (MNLCs) using selegiline as preinducer, as well as Shh and all trans-retinoic acid (RA) as inducers. Selegiline was reported to induce the embryonic stem cells and bone marrow stromal cells into neuronal phenotype. ADSCs were evaluated using CD90, CD44, CD 49d, CD106, CD31, CD45, lipogenesis and osteogenesis. Dose response and time course studies were used in selecting the optimal concentration for selegiline using the percentage of viable cells (PVC) and percentages of immunoreactive cells (PIC) to nestin and neurofilament 68. Accordingly, such studies were used in selecting the optimal dose for RA using PVC and PIC to islet-1 and oligo-2. The expression of islet-1, oligo-2 and HLXB9 was evaluated using RT-PCR and immunocytochemistry. Real-time PCR was utilized in order to quantify the expression of islet-1, oligo-2 and HLXB9. ADSCs were immunoreactive to CD90, CD44 and CD 49d with consistent differentiation osteogenic and lipogenic cells. The optimal concentrations of selegiline and RA were 10⁻⁹ mM and 2 × 10⁻⁸ M, respectively. After two days, MNLCs showed high oligo-2 expression. MNLCs innervated myotubes; also, the release rate of synaptic vesicles using FM1-43 followed exponential decay model, and this rate in the induced MNLCs was approximately three times of that in the preinduced cells.
Collapse
|
24
|
Spitzer N, Sammons GS, Butts HM, Grover LM, Price EM. Multipotent progenitor cells derived from adult peripheral blood of swine have high neurogenic potential in vitro. J Cell Physiol 2011; 226:3156-68. [PMID: 21321934 DOI: 10.1002/jcp.22670] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Peripheral blood-derived multipotent adult progenitor cells (PBD-MAPCs) are a novel population of stem cells, isolated from venous blood of green fluorescent protein transgenic swine, which proliferate as multicellular non-adherent spheroids. Using a simple differentiation protocol, a large proportion of these cells developed one of five distinct neural cell phenotypes, indicating that these primordial cells have high neurogenic potential. Cells exhibiting neural morphologies developed within 48 h of exposure to differentiation conditions, increased in percentage over 2 weeks, and stably maintained the neural phenotype for three additional weeks in the absence of neurogenic signaling molecules. Cells exhibited dynamic neural-like behaviors including extension and retraction of processes with growth cone-like structures rich in filamentous actin, cell migration following a leading process, and various cell-cell interactions. Differentiated cells expressed neural markers, NeuN, β-tubulin III and synaptic proteins, and progenitor cells expressed the stem cell markers nestin and NANOG. Neurally differentiated PBD-MAPCs exhibited voltage-dependent inward and outward currents and expressed voltage-gated sodium and potassium channels, suggestive of neural-like membrane properties. PBD-MAPCs expressed early neural markers and developed neural phenotypes when provided with an extracellular matrix of laminin without the addition of cytokines or growth factors, suggesting that these multipotent cells may be primed for neural differentiation. PBD-MAPCs provide a model for understanding the mechanisms of neural differentiation from non-neural sources of adult stem cells. A similar population of cells, from humans or xenogeneic sources, may offer the potential of an accessible, renewable and non-tumorigenic source of stem cells for treating neural disorders.
Collapse
Affiliation(s)
- Nadja Spitzer
- Department of Biological Sciences, Marshall University, Huntington, West Virginia 25755, USA.
| | | | | | | | | |
Collapse
|
25
|
Nicaise C, Bohl D, Pochet R. [Cellular transdifferentiation in amyotrophic lateral sclerosis]. Med Sci (Paris) 2011; 27:799-801. [PMID: 22027411 DOI: 10.1051/medsci/20112710002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
26
|
Cordes AL, Jahn K, Hass R, Schwabe K, Weissinger EM, Ganser A, Götz F, Dengler R, Krauss JK, Petri S. Intramedullary spinal cord implantation of human CD34+ umbilical cord-derived cells in ALS. AMYOTROPHIC LATERAL SCLEROSIS : OFFICIAL PUBLICATION OF THE WORLD FEDERATION OF NEUROLOGY RESEARCH GROUP ON MOTOR NEURON DISEASES 2011; 12:325-330. [PMID: 21812628 DOI: 10.3109/17482968.2011.580850] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disorder with marginal therapeutic options. Degeneration of motor neurons in the primary motor cortex, brainstem and spinal cord lead to rapidly progressive paralysis and finally to death due to respiratory failure. As pharmacological therapies have failed to provide sufficient neuroprotective effects in ALS, transplantation of stem or progenitor cells is considered a promising treatment strategy. Cell transplantation approaches in ALS mainly aim to generate a neuroprotective environment for degenerating motor neurons by transplantation of non-neuronal cells, rather than to replace lost motor neurons. We present a 63-year-old male patient suffering from ALS who underwent intramedullary thoracic spinal cord implantation of human CD34(+) umbilical cord-derived haematopoietic progenitor cells with a three-year follow up after transplantation.
Collapse
|
27
|
Ojeda L, Gao J, Hooten KG, Wang E, Thonhoff JR, Dunn TJ, Gao T, Wu P. Critical role of PI3K/Akt/GSK3β in motoneuron specification from human neural stem cells in response to FGF2 and EGF. PLoS One 2011; 6:e23414. [PMID: 21887250 PMCID: PMC3160859 DOI: 10.1371/journal.pone.0023414] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Accepted: 07/16/2011] [Indexed: 12/17/2022] Open
Abstract
Fibroblast growth factor (FGF) and epidermal growth factor (EGF) are critical for the development of the nervous system. We previously discovered that FGF2 and EGF had opposite effects on motor neuron differentiation from human fetal neural stem cells (hNSCs), but the underlying mechanisms remain unclear. Here, we show that FGF2 and EGF differentially affect the temporal patterns of Akt and glycogen synthase kinase 3 beta (GSK3β) activation. High levels of phosphatidylinositol 3-kinase (PI3K)/Akt activation accompanied with GSK3β inactivation result in reduction of the motor neuron transcription factor HB9. Inhibition of PI3K/Akt by chemical inhibitors or RNA interference or overexpression of a constitutively active form of GSK3β enhances HB9 expression. Consequently, PI3K inhibition increases hNSCs differentiation into HB9+/microtubule-associated protein 2 (MAP2)+ motor neurons in vitro. More importantly, blocking PI3K not only enhances motor neuron differentiation from hNSCs grafted into the ventral horn of adult rat spinal cords, but also permits ectopic generation of motor neurons in the dorsal horn by overriding environmental influences. Our data suggest that FGF2 and EGF affect the motor neuron fate decision in hNSCs differently through a fine tuning of the PI3K/AKT/GSK3β pathway, and that manipulation of this pathway can enhance motor neuron generation.
Collapse
Affiliation(s)
- Luis Ojeda
- Department of Neuroscience and Cell Biology, University Of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Junling Gao
- Department of Neuroscience and Cell Biology, University Of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Kristopher G. Hooten
- Department of Neurosurgery, University of Florida, Gainesville, Florida, United States of America
| | - Enyin Wang
- Department of Neuroscience and Cell Biology, University Of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
- West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Jason R. Thonhoff
- Department of Neuroscience and Cell Biology, University Of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Tiffany J. Dunn
- Department of Neuroscience and Cell Biology, University Of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Tianyan Gao
- Markey Cancer Center and Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, United States of America
| | - Ping Wu
- Department of Neuroscience and Cell Biology, University Of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
- * E-mail:
| |
Collapse
|
28
|
Forostyak S, Jendelova P, Kapcalova M, Arboleda D, Sykova E. Mesenchymal stromal cells prolong the lifespan in a rat model of amyotrophic lateral sclerosis. Cytotherapy 2011; 13:1036-46. [PMID: 21736505 DOI: 10.3109/14653249.2011.592521] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND AIMS Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder characterized by the loss of brain and spinal cord motor neurons (MN). The intraspinal and systemic grafting of mesenchymal stromal cells (MSC) was used to treat symptomatic transgenic rats overexpressing human superoxide dismutase 1 (SOD1) in order to alleviate the disease course and prolong the animals' lifespan. METHODS At the age of 16 weeks (disease onset) the rats received two grafts of MSC expressing green fluorescent protein (GFP(+) MSC) on the same day, intraspinally (10(5) cells) and intravenously (2 × 10(6) cells). Sham-treated animals were injected with phosphate-buffered saline (PBS). Motor activity, grip strength and body weight were tested, followed by immunohistochemical analysis. RESULTS The combined grafting of MSC into symptomatic rats had a significant effect on motor activity and grip strength starting 4 weeks after transplantation. The lifespan of animals in the treated group was 190 ± 3.33 days compared with 179 ± 3.6 days in the control group of animals. Treated rats had a larger number of MN at the thoracic and lumbar levels; these MN were of larger size, and the intensity of terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick-end labeling (TUNEL) staining in the somas of apoptotic MN at the thoracic level was much lower than in sham-treated animals. Transplanted GFP(+) MSC survived in the spinal cord until the end stage of the disease and migrated both rostrally and caudally from the injection site. CONCLUSIONS Intraspinal and intravenous transplantation of MSC has a beneficial and possibly synergistic effect on the lifespan of ALS animals.
Collapse
Affiliation(s)
- Serhiy Forostyak
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | | | | | | | | |
Collapse
|
29
|
Nicaise C, Mitrecic D, Pochet R. Brain and spinal cord affected by amyotrophic lateral sclerosis induce differential growth factors expression in rat mesenchymal and neural stem cells. Neuropathol Appl Neurobiol 2011; 37:179-88. [PMID: 20846186 DOI: 10.1111/j.1365-2990.2010.01124.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
UNLABELLED Stem cell research raises hopes for incurable neurodegenerative diseases. In amyotrophic lateral sclerosis (ALS), affecting the motoneurones of the central nervous system (CNS), stem cell-based therapy aims to replace dying host motoneurones by transplantation of cells in disease-affected regions. Moreover, transplanted stem cells can serve as a source of trophic factors providing neuroprotection, slowing down neuronal degeneration and disease progression. AIM To determine the profile of seven trophic factors expressed by mesenchymal stem cells (MSC) and neural stem cells (NSC) upon stimulation with CNS protein extracts from SOD1-linked ALS rat model. METHODS Culture of rat MSC, NSC and fibroblasts were incubated with brain and spinal cord extracts from SOD1(G93A) transgenic rats and mRNA expression of seven growth factors was measured by quantitative PCR. RESULTS MSC, NSC and fibroblasts exhibited different expression patterns. Nerve growth factor and brain-derived neurotropic factor were significantly upregulated in both NSC and MSC cultures upon stimulation with SOD1(G93A) CNS extracts. Fibroblast growth factor 2, insulin-like growth factor and glial-derived neurotropic factor were upregulated in NSC, while the same factors were downregulated in MSC. Vascular endothelial growth factor A upregulation was restricted to MSC and fibroblasts. Surprisingly, SOD1(G93A) spinal cord, but not the brain extract, upregulated brain-derived neurotropic factor in MSC and glial-derived neurotropic factor in NSC. CONCLUSIONS These results suggest that inherent characteristics of different stem cell populations define their healing potential and raise the concept of ALS environment in stem cell transplantation.
Collapse
Affiliation(s)
- C Nicaise
- Laboratory of Histology, Neuroanatomy and Neuropathology, Université Libre de, Bruxelles, Brussels, Belgium.
| | | | | |
Collapse
|
30
|
Gamez J, Carmona F, Raguer N, Ferrer-Sancho J, Martín-Henao GA, Martí-Beltrán S, Badia M, Gratacós M, Rodriguez-Gónzalez E, Seoane JL, Pallero-Castillo M, Burgos R, Puiggros C, Pasarin A, Bori-Fortuny I. Cellular transplants in amyotrophic lateral sclerosis patients: an observational study. Cytotherapy 2011; 12:669-77. [PMID: 20586670 DOI: 10.3109/14653241003774037] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND AIMS Cytotherapy is a promising option for neurodegenerative disease treatment. Because of the fatal prognosis and imperative need for effective treatment, amyotrophic lateral sclerosis (ALS) patients request this therapy before its effectiveness has been verified. The increase in clinics offering cytotherapies but providing little scientific information has prompted considerable medical tourism. We present an observational study of Spanish ALS patients receiving cytotherapy, analyzing the experiences arising from the treatment (TX) and considering two progression markers, FVC and ALSFRS-R. METHODS Twelve ALS patients with a mean age of 48.6 years (SD 12.8) received cytotherapy 26.9 months (SD 15.8) after clinical onset. ALSFRS-R and FVC at TX were 32.3 (SD 6.8) and 63.4% (SD 15.3), respectively. TX involved transplants of olfactory ensheathing cells in three patients, and autologous mesenchymal stromal cells in the remainder. RESULTS One patient died 33 months post-TX after surviving for 49 months. Five required mechanical non-invasive home ventilation 7.4 months post-TX. Two required invasive ventilation 13 months post-TX. Five patients needed gastrostomy feeding 23.3 months post-TX. Survival between clinical onset and the study end date was 50 months (SD 17.2). No significant adverse events or changes in the decline of FVC and ALSFRS-R compared with the disease's natural history were observed. CONCLUSIONS Our observations suggest that these therapies do not halt the course of the disease. Cytotherapy cannot yet be considered a curative treatment for ALS.
Collapse
Affiliation(s)
- Josep Gamez
- Neurology Department, Hospital Universitari Vall d'Hebron, Autonomous University of Barcelona, Barcelona, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Kuzma-Kozakiewicz M, Kwiecinski H. New therapeutic targets for amyotrophic lateral sclerosis. Expert Opin Ther Targets 2010; 15:127-43. [PMID: 21133819 DOI: 10.1517/14728222.2011.542152] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is one of the most devastating neurological disorders, affecting approximately half a million people worldwide. Currently there is no cure or prevention for ALS. Although ALS is a rare condition, it places a tremendous socioeconomic burden on patients, family members, caregivers and health systems. AREAS COVERED The review examines the mechanisms that may contribute to motor neuron degeneration in ALS, among which oxidative damage, glutatamate excitoxicity, mitochondrial dysfunction, impaired axonal transport, apoptotic cell death, growth factor deficiency, glial cell pathology and abnormal RNA metabolism are potential targets for ALS treatment. The article provides an overview of clinical trials performed to date in attempts to treat ALS with regard to molecular mechanisms and pathways they act on. It also discusses new trials based on recently developed molecular biology techniques. EXPERT OPINION Despite significant effectiveness of several potential therapeutics observed in preclinical trials, the results were not translatable to patients with ALS. The development of effective treatments of ALS strictly depends on understanding the primary cause of the disease. This goal will only be achieved when we identify the trigger point for motor neuron death in ALS.
Collapse
|
32
|
Cova L, Silani V. Amyotrophic lateral sclerosis: applications of stem cells - an update. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2010; 3:145-56. [PMID: 24198520 PMCID: PMC3781739 DOI: 10.2147/sccaa.s8662] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neurodegenerative diseases are a growing public health challenge, and amyotrophic lateral sclerosis (ALS) remains a fatal incurable disease. The advent of stem cell therapy has opened new horizons for both researchers and ALS patients, desperately looking for a treatment. ALS must be considered a systemic disease affecting many cell phenotypes besides motor neurons, even outside the central nervous system. Cell replacement therapy needs to address the specific neurobiological issues of ALS to safely and efficiently reach clinical settings. Moreover, the enormous potential of induced pluripotent cells directly derived from patients for modeling and understanding the pathological mechanisms, in correlation with the discoveries of new genes and animal models, provides new opportunities that need to be integrated with previously described transplantation strategies. Finally, a careful evaluation of preclinical data in conjunction with wary patient choice in clinical trials needs to be established in order to generate meaningful results.
Collapse
Affiliation(s)
- Lidia Cova
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milano, Italy
| | | |
Collapse
|
33
|
Lunn JS, Pacut C, Backus C, Hong Y, Johe K, Hefferan M, Marsala M, Feldman EL. The pleotrophic effects of insulin-like growth factor-I on human spinal cord neural progenitor cells. Stem Cells Dev 2010; 19:1983-93. [PMID: 20406098 DOI: 10.1089/scd.2010.0003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Most stem cell therapies involve direct, intraparachymal placement of neural progenitor cells. These cells provide physical support to the endogenous neuronal population and may be engineered to provide in situ growth factor support. Insulin-like growth factor-I (IGF-I) has potent neurotrophic and neuroprotective properties and is expressed by human neural stem cells (hNSCs). IGF-I is implicated in multiple aspects of cell behavior, including proliferation, differentiation, and survival. Enhancing hNSC function through IGF-I overexpression may increase the benefits of stem cell therapy. As a first step to that goal, we examined the direct effects of IGF-I on hNSC behavior in vitro. We demonstrate that IGF-I treatment enhances both the number and length of hNSC neurites. This is correlated with a decrease in proliferation, suggesting that IGF-I promotes neurite outgrowth but not proliferation. While IGF-I activates both AKT and MAPK signaling in hNSCs, we demonstrate that IGF-I-mediated neurite outgrowth is dependent only on AKT signaling. Finally, we demonstrate that IGF-I is neuroprotective after glutamate exposure in a model of excitotoxic cell death.
Collapse
Affiliation(s)
- J Simon Lunn
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Su H, Wu Y, Yuan Q, Guo J, Zhang W, Wu W. Optimal time point for neuronal generation of transplanted neural progenitor cells in injured spinal cord following root avulsion. Cell Transplant 2010; 20:167-76. [PMID: 20719091 DOI: 10.3727/096368910x522090] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Root avulsion of the brachial plexus results in a progressive and pronounced loss of motoneurons. Cell replacement strategies have therapeutic potential in the treatment of motoneuron degenerative neurological disorders. Here, we transplanted spinal cord-derived neural progenitor cells (NPCs) into the cervical ventral horn of adult rats immediately, 2 weeks, or 6 weeks after root avulsion to determine an optimal time scale for the survival and differentiation of grafted cells. We showed that grafted NPCs survived robustly at all three time points and there was no statistical difference in survival rate. Interestingly, however, transplantation at 2 weeks postavulsion significantly increased the neuronal differentiation of transplanted NPCs compared to transplantation immediately or at 6 weeks postavulsion. Moreover, only NPCs transplanted at 2 weeks postavulsion were able to differentiate into choline acetyltransferase (ChAT)-positive neurons. Specific ELISAs and quantitative reverse transcriptase polymerase chain reaction (RT-PCR) demonstrated that expression levels of BDNF and GDNF were significantly upregulated in the ventral cord at 2 weeks postavulsion compared to immediately or at 6 weeks postavulsion. Our study suggests that the cervical ventral horn at 2 weeks postavulsion both supports neuronal differentiation and induces region-specific neuronal generation possibly because of its higher expression of BDNF and GDNF.
Collapse
Affiliation(s)
- Huanxing Su
- Department of Anatomy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | | | | | | | | | | |
Collapse
|
35
|
Vansteensel MJ, Hermes D, Aarnoutse EJ, Bleichner MG, Schalk G, van Rijen PC, Leijten FSS, Ramsey NF. Brain-computer interfacing based on cognitive control. Ann Neurol 2010; 67:809-16. [PMID: 20517943 DOI: 10.1002/ana.21985] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Brain-computer interfaces (BCIs) translate deliberate intentions and associated changes in brain activity into action, thereby offering patients with severe paralysis an alternative means of communication with and control over their environment. Such systems are not available yet, partly due to the high performance standard that is required. A major challenge in the development of implantable BCIs is to identify cortical regions and related functions that an individual can reliably and consciously manipulate. Research predominantly focuses on the sensorimotor cortex, which can be activated by imagining motor actions. However, because this region may not provide an optimal solution to all patients, other neuronal networks need to be examined. Therefore, we investigated whether the cognitive control network can be used for BCI purposes. We also determined the feasibility of using functional magnetic resonance imaging (fMRI) for noninvasive localization of the cognitive control network. METHODS Three patients with intractable epilepsy, who were temporarily implanted with subdural grid electrodes for diagnostic purposes, attempted to gain BCI control using the electrocorticographic (ECoG) signal of the left dorsolateral prefrontal cortex (DLPFC). RESULTS All subjects quickly gained accurate BCI control by modulation of gamma-power of the left DLPFC. Prelocalization of the relevant region was performed with fMRI and was confirmed using the ECoG signals obtained during mental calculation localizer tasks. INTERPRETATION The results indicate that the cognitive control network is a suitable source of signals for BCI applications. They also demonstrate the feasibility of translating understanding about cognitive networks derived from functional neuroimaging into clinical applications.
Collapse
Affiliation(s)
- Mariska J Vansteensel
- Section of Brain Function and Plasticity, Department of Neurology and Neurosurgery, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Henriques A, Pitzer C, Schneider A. Neurotrophic growth factors for the treatment of amyotrophic lateral sclerosis: where do we stand? Front Neurosci 2010; 4:32. [PMID: 20592948 PMCID: PMC2902233 DOI: 10.3389/fnins.2010.00032] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2010] [Accepted: 05/07/2010] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease that results in progressive loss of motoneurons, motor weakness and death within 3–5 years after disease onset. Therapeutic options remain limited despite substantial number of approaches that have been tested clinically. Many neurotrophic growth factors are known to promote the survival of neurons and foster regeneration in the central nervous system. Various neurotrophic factors have been investigated pre-clinically and clinically for the treatment of ALS. Although pre-clinical data appeared promising, no neurotrophic factors succeeded yet in a clinical phase III trial. In this review we discuss the rationale behind those factors, possible reasons for clinical failures, and argue for a renewal of hope in this powerful class of drugs for the treatment of ALS.
Collapse
|
37
|
Human adipose-derived stem cells enhance the glutamate uptake function of GLT1 in SOD1G93A-bearing astrocytes. Biochem Biophys Res Commun 2010; 393:481-6. [DOI: 10.1016/j.bbrc.2010.02.029] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Accepted: 02/06/2010] [Indexed: 12/13/2022]
|
38
|
Bossolasco P, Cova L, Calzarossa C, Servida F, Mencacci NE, Onida F, Polli E, Lambertenghi Deliliers G, Silani V. Metalloproteinase alterations in the bone marrow of ALS patients. J Mol Med (Berl) 2010; 88:553-64. [PMID: 20091292 DOI: 10.1007/s00109-009-0584-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2009] [Revised: 11/20/2009] [Accepted: 12/17/2009] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease, nowadays considered as suitable candidate for autologous stem therapy with bone marrow (BM). A careful characterization of BM stem cell (SC) compartment is mandatory before its extensive application to clinic. Indeed, widespread systemic involvement has been recently advocated given that non-neuronal neighboring cells actively influence the pathological neuronal loss. We therefore investigated BM samples from 21 ALS patients and reported normal hematopoietic biological properties while an atypical behavior and impaired SC capabilities affected only the mesenchymal compartment. Moreover, by quantitative real-time approach, we observed altered Collagen IV and Metalloproteinase-9 levels in patients' derived mesenchymal stem cells (MSCs). Widespread metalloproteinase (MMPs) and their tissue inhibitor (TIMPs) alterations were established by multiplex ELISA analysis, demonstrating diffuse enzymatic variations in MSC compartment. Since MMPs act as fundamental effectors of extra-cellular matrix remodeling and stem cell mobilization, their modifications in ALS may influence reparative mechanisms effective in counteracting the pathology. In conclusion, ALS is further confirmed to be a systemic disease, not restricted to the nervous system, but affecting also the BM stromal compartment, even in sporadic cases. Therefore, therapeutic implantation of autologous BM derived SC in ALS patients needs to be carefully reevaluated.
Collapse
Affiliation(s)
- Patrizia Bossolasco
- Fondazione Matarelli, Dipartimento di Farmacologia, Chemioterapia e Tossicologia Medica, Università degli Studi di Milano, Via Vanvitelli, 32-20129, Milan, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Corti S, Nizzardo M, Nardini M, Donadoni C, Salani S, Ronchi D, Simone C, Falcone M, Papadimitriou D, Locatelli F, Mezzina N, Gianni F, Bresolin N, Comi GP. Embryonic stem cell-derived neural stem cells improve spinal muscular atrophy phenotype in mice. ACTA ACUST UNITED AC 2009; 133:465-81. [PMID: 20032086 DOI: 10.1093/brain/awp318] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Spinal muscular atrophy, characterized by selective loss of lower motor neurons, is an incurable genetic neurological disease leading to infant mortality. We previously showed that primary neural stem cells derived from spinal cord can ameliorate the spinal muscular atrophy phenotype in mice, but this primary source has limited translational value. Here, we illustrate that pluripotent stem cells from embryonic stem cells show the same potential therapeutic effects as those derived from spinal cord and offer great promise as an unlimited source of neural stem cells for transplantation. We found that embryonic stem cell-derived neural stem cells can differentiate into motor neurons in vitro and in vivo. In addition, following their intrathecal transplantation into spinal muscular atrophy mice, the neural stem cells, like those derived from spinal cord, survived and migrated to appropriate areas, ameliorated behavioural endpoints and lifespan, and exhibited neuroprotective capability. Neural stem cells obtained using a drug-selectable embryonic stem cell line yielded the greatest improvements. As with cells originating from primary tissue, the embryonic stem cell-derived neural stem cells integrated appropriately into the parenchyma, expressing neuron- and motor neuron-specific markers. Our results suggest translational potential for the use of pluripotent cells in neural stem cell-mediated therapies and highlight potential safety improvements and benefits of drug selection for neuroepithelial cells.
Collapse
Affiliation(s)
- Stefania Corti
- Department of Neurological Sciences, University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Suzuki M, Klein S, Wetzel EA, Meyer M, McHugh J, Tork C, Hayes A, Svendsen CN. Acute glial activation by stab injuries does not lead to overt damage or motor neuron degeneration in the G93A mutant SOD1 rat model of amyotrophic lateral sclerosis. Exp Neurol 2009; 221:346-52. [PMID: 20005223 DOI: 10.1016/j.expneurol.2009.12.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2009] [Revised: 11/19/2009] [Accepted: 12/01/2009] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease where motor neurons within the brain and spinal cord are lost, leading to paralysis and death. Recently, a correlation between head trauma and the incidence of ALS has been reported. Furthermore, new invasive neurosurgical studies are being planned which involve inserting needles directly to the spinal cord. We therefore tested whether acute trauma to the spinal cord via a knife wound injury would lead to accelerated disease progression in rodent models of ALS (SOD1(G93A) rats). A longitudinal stab injury using a small knife was performed within the lumbar spinal cord region of presymptomatic SOD1(G93A) rats. Host glial activation was detected in the lumbar area surrounding a micro-knife lesion at 2 weeks after surgery in both wild type and SOD1(G93A) animals. However, there was no sign of motor neuron loss in the injured spinal cord of any animal and normal motor function was maintained in the ipsilateral limb. These results indicate that motor neurons in presymptomatic G93A animals are not affected by an invasive puncture wound injury involving reactive astrocytes. Furthermore, acute trauma alone does not accelerate disease onset or progression in this ALS model which is important for future strategies of gene and cell therapies directly targeting the spinal cord of ALS patients.
Collapse
Affiliation(s)
- Masatoshi Suzuki
- The Waisman Center, University of Wisconsin-Madison, Madison, WI, USA.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Motoneuron transplantation rescues the phenotype of SMARD1 (spinal muscular atrophy with respiratory distress type 1). J Neurosci 2009; 29:11761-71. [PMID: 19776263 DOI: 10.1523/jneurosci.2734-09.2009] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Spinal muscular atrophy with respiratory distress type 1 (SMARD1) is a fatal form of infantile motoneuron disease. There is currently no effective treatment, although motor neuron replacement is a possible therapeutic strategy. We transplanted purified motor neurons into the spinal cord of nmd mice, an animal model of SMARD1. We also administered pharmacological treatment targeting the induction of axonal growth toward skeletal muscle target. At the end stage of the disease, donor-derived motor neurons were detected in the nmd anterior horns, extended axons into the ventral roots, and formed new neuromuscular junctions. These data correlated with improved neuromuscular function and increased life spans. The neuroprotective effect was associated with a reduction in proinflammatory molecules in treated spinal cords. This is the first report that functional restoration of motor units with transplanted motoneurons is feasible in an animal model of a human motoneuron disease, opening up new possibilities for therapeutic intervention.
Collapse
|
42
|
Cizkova D, Cizek M, Nagyova M, Slovinska L, Novotna I, Jergova S, Radonak J, Hlucilova J, Vanicky I. Enrichment of rat oligodendrocyte progenitor cells by magnetic cell sorting. J Neurosci Methods 2009; 184:88-94. [DOI: 10.1016/j.jneumeth.2009.07.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2008] [Revised: 06/08/2009] [Accepted: 07/28/2009] [Indexed: 01/17/2023]
|
43
|
Abstract
Successful treatment of neurodegenerative diseases and CNS trauma are the most intractable problems in modern medicine. Numerous reports have shown the strong role that laminins have on the survival, regeneration and development of various types of cells, including neural cells. It would be desirable to take advantage of laminin activities for therapeutic purposes. However, there are at least ten laminin variants and the trimeric molecules are of the order of 800,000 molecular weight. Furthermore, human laminins are not available in quantity. Therefore, we and others have taken the approach of determining which domains of the laminin molecules are functional in the CNS, and whether short peptides from these regions exhibit biological activities with the intent of testing their potential for therapeutic use. Understanding the role of laminins and their small biologically active peptide domains, such as the KDI (lysine–aspartic acid–isoleucine) peptide from γ1 laminin, in neuronal development, CNS trauma (spinal cord injury and stroke) and neurodegenerative disorders (amyotrophic lateral sclerosis, Alzheimer’s disease and Parkinson’s disease) may help to develop clinically applicable methods to treat the presently untreatable CNS diseases and trauma even in the near future.
Collapse
Affiliation(s)
- Päivi Liesi
- The Brain Laboratory, Department of Biological & Environmental Sciences, University of Helsinki, PO Box 65 (Viikinkaari 1), 00014 University of Helsinki, Finland
| |
Collapse
|
44
|
Gál P, Kravcuková P, Mokrý M, Kluchová D. Chemokines as possible targets in modulation of the secondary damage after acute spinal cord injury: a review. Cell Mol Neurobiol 2009; 29:1025-35. [PMID: 19363652 PMCID: PMC11506275 DOI: 10.1007/s10571-009-9392-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2008] [Accepted: 03/10/2009] [Indexed: 12/23/2022]
Abstract
In spite of many promising experimental studies, an effective treatment dramatically eliminating the secondary damage after spinal cord injury (SCI) is still missing. Since clinical data on the therapeutical effect after methylprednisolone treatment are not conclusive, new therapeutical modalities targeting specific components of secondary spinal cord damage needs to be developed. It is known that immune cells are recruited to injury sites by chemokines, which are small, structurally similar proteins released locally at the site of inflammation. Hence, this review was aimed to summarize possible roles of chemokines in the inflammation following SCI as well as to identify possible new therapeutical targets which can potentially be effective in ameliorating individual components of this inflammatory response. Data concerning inflammation reduction together with techniques improving axonal growth, cell replacement and remyelinization, may be crucial to move a small step forward in an attempt to make paraplegic and quadriplegic patients to walk.
Collapse
Affiliation(s)
- Peter Gál
- Institute of Biology and Ecology, Pavol Jozef Safárik University, 041 80 Kosice, Slovak Republic.
| | | | | | | |
Collapse
|
45
|
Boucherie C, Schäfer S, Lavand'homme P, Maloteaux JM, Hermans E. Chimerization of astroglial population in the lumbar spinal cord after mesenchymal stem cell transplantation prolongs survival in a rat model of amyotrophic lateral sclerosis. J Neurosci Res 2009; 87:2034-46. [PMID: 19267424 DOI: 10.1002/jnr.22038] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Adult mesenchymal stem cells (MSCs) exhibit neuroprotective properties when introduced into the degenerating central nervous system through different putative mechanisms including secretion of growth factors and transdifferentiation. In the present study, we injected MSCs into the cerebrospinal fluid of symptomatic hSOD1(G93A) rats, a transgenic animal model of familial amyotrophic lateral sclerosis (ALS) expressing a mutated form of the human superoxide dismutase. MSCs were found to infiltrate the nervous parenchyma and migrate substantially into the ventral gray matter, where motor neurons degenerate. Even though overall astrogliosis was not modified, MSCs differentiated massively into astrocytes at the site of degeneration. The intrathecal delivery of MSCs and the subsequent generation of healthy astrocytes at symptomatic stage decreased motor neuron loss in the lumbar spinal cord, preserving motor functions and extending the survival of hSOD1(G93A) rats. This neuroprotection was correlated with decreased inflammation, as shown by the lower proliferation of microglial cells and the reduced expressiontion of COX-2 and NOX-2. Together, these data highlight the protective capacity of adult MSC-derived astrocytes when grafted into the central nervous system and illustrate an attractive strategy to target excessive inflammation in ALS.
Collapse
Affiliation(s)
- Cédric Boucherie
- Institute of Neuroscience (INES), Université catholique de Louvain, Brussels, Belgium
| | | | | | | | | |
Collapse
|
46
|
Mazzini L, Vercelli A, Ferrero I, Mareschi K, Boido M, Servo S, Oggioni GD, Testa L, Monaco F, Fagioli F. Stem cells in amyotrophic lateral sclerosis: state of the art. Expert Opin Biol Ther 2009; 9:1245-58. [DOI: 10.1517/14712590903186956] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
47
|
Schmidt ER, Pasterkamp RJ, van den Berg LH. Axon guidance proteins: Novel therapeutic targets for ALS? Prog Neurobiol 2009; 88:286-301. [DOI: 10.1016/j.pneurobio.2009.05.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2008] [Revised: 04/06/2009] [Accepted: 05/27/2009] [Indexed: 12/12/2022]
|
48
|
Genetic control of wayward pluripotent stem cells and their progeny after transplantation. Cell Stem Cell 2009; 4:289-300. [PMID: 19341619 DOI: 10.1016/j.stem.2009.03.010] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The proliferative capacity of pluripotent stem cells and their progeny brings a unique aspect to therapeutics, in that once a transplant is initiated the therapist no longer has control of the therapy. In the context of the recent FDA approval of a human ESC trial and report of a neuronal-stem-cell-derived tumor in a human trial, strategies need to be developed to control wayward pluripotent stem cells. Here, we focus on one approach: direct genetic modification of the cells prior to transplantation with genes that can prevent the adverse events and/or eliminate the transplanted cells and their progeny.
Collapse
|
49
|
Lunn JS, Hefferan MP, Marsala M, Feldman EL. Stem cells: comprehensive treatments for amyotrophic lateral sclerosis in conjunction with growth factor delivery. Growth Factors 2009; 27:133-40. [PMID: 19294549 DOI: 10.1080/08977190902814855] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by loss of both upper and lower motor neurons. ALS progression is complex and likely due to cellular dysfunction at multiple levels, including mitochondrial dysfunction, glutamate excitotoxicity, oxidative stress, axonal dysfunction, reactive astrocytosis, and mutant superoxide dismutase expression, therefore, treatment must provide neuronal protection from multiple insults. A significant amount of ALS research focuses on growth factor-based therapies. Growth factors including insulin-like growth factor-I, vascular endothelial growth factor, brain-derived neurotrophic factor, and glial-derived neurotrophic factor exhibit robust neuroprotective effects on motor neurons in ALS models. Issues concerning growth factor delivery, stability and unwanted side effects slow the transfer of these treatments to human ALS patients. Stem cells represent a new therapeutic approach offering both cellular replacement and trophic support for the existing population. Combination therapy consisting of stem cells expressing beneficial growth factors may provide a comprehensive treatment for ALS.
Collapse
Affiliation(s)
- J Simon Lunn
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | | | | | | |
Collapse
|
50
|
Arnett ALH, Chamberlain JR, Chamberlain JS. Therapy for neuromuscular disorders. Curr Opin Genet Dev 2009; 19:290-7. [PMID: 19411172 DOI: 10.1016/j.gde.2009.03.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2009] [Revised: 03/17/2009] [Accepted: 03/20/2009] [Indexed: 12/21/2022]
Abstract
Research into therapeutic approaches for both recessive and dominant neuromuscular disorders has made great progress over the past few years. In the field of gene therapy, antisense-mediated exon skipping is being applied to bypass deleterious mutations in the dystrophin gene and restore dystrophin expression in animal models of muscular dystrophy. Approaches for the dominant genetic muscle diseases have turned toward elimination of the mutant gene product with anti-sense oligonucleotide therapy and RNA interference techniques. Refinements of adeno-associated viral vectors and strategies for their delivery are also leading towards future clinical trials. The discovery of new, multipotent cell lineages, some of which possess the ability to successfully engraft muscle following vascular delivery, presents exciting prospects for the field of stem cell therapy. These discoveries represent steady progress towards the development of effective therapies for a wide range of neuromuscular disorders.
Collapse
Affiliation(s)
- Andrea L H Arnett
- Medical Scientist Training Program, University of Washington, Seattle, WA 98195-7720, United States.
| | | | | |
Collapse
|