51
|
Ebrahimi MJ, Aliaghaei A, Boroujeni ME, Khodagholi F, Meftahi G, Abdollahifar MA, Ahmadi H, Danyali S, Daftari M, Sadeghi Y. Human Umbilical Cord Matrix Stem Cells Reverse Oxidative Stress-Induced Cell Death and Ameliorate Motor Function and Striatal Atrophy in Rat Model of Huntington Disease. Neurotox Res 2018; 34:273-284. [PMID: 29520722 DOI: 10.1007/s12640-018-9884-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 02/11/2018] [Accepted: 02/19/2018] [Indexed: 12/13/2022]
Abstract
Huntington disease (HD) is an inherited disorder hallmarked by progressive deterioration of specific neurons, followed by movement and cognitive anomalies. Cell therapy approaches in neurodegenerative conditions have concentrated on the replenishment of lost/dying neurons with functional ones. Multipotent mesenchymal stem cells (MSCs) have been represented as a potential remedy for HD. In this study, we evaluated the in vitro and in vivo efficacy of umbilical cord matrix stem cells (UCMSCs) and their paracrine effect against oxidative stress with a specific focus on HD. To this end, UCMSCs were isolated, immunophenotypically characterized by the positive expression of MSC markers, and exhibited multilineage potentiality. Besides, synthesis of neurotrophic factors of GDNF and VEGF by UCMSC was confirmed. Initially, PC12 cells were exposed to superoxide in the presence of conditioned media (CM) collected from UCMSC (UCMSC-CM) and cell viability plus neuritogenesis were measured. Next, bilateral striatal transplantation of UCMSC in 3-nitropropionic acid (3-NP) lesioned rat models was conducted, and 1 month later, post-graft analysis was performed. According to our in vitro results, CM of UCMSC protected PC12 cells against oxidative stress and considerably enhanced cell viability and neurite outgrowth. On the other hand, transplanted UCMSC survived, decreased gliosis, and ameliorated motor coordination and muscle activity, along with an increase in striatal volume as well as in dendritic length of the striatum in HD rats. Collectively, our findings imply that UCMSCs provide an enriched platform by largely their paracrine factors, which downgrades the unfavorable effects of oxidative stress.
Collapse
Affiliation(s)
- Mohammad Javad Ebrahimi
- Cell Biology and Anatomical Sciences, School Of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Aliaghaei
- Cell Biology and Anatomical Sciences, School Of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Eskandarian Boroujeni
- Department Of Stem Cells and Rgenerative Medicine, Faculty of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mohammad Amin Abdollahifar
- Cell Biology and Anatomical Sciences, School Of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Houssein Ahmadi
- Cell Biology and Anatomical Sciences, School Of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samira Danyali
- Neurophysiology Research Center, Shahid Beheshti University Of Medical Sciences, Tehran, Iran
| | - Mahtab Daftari
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Yousef Sadeghi
- Cell Biology and Anatomical Sciences, School Of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
52
|
Simmons DA. Modulating Neurotrophin Receptor Signaling as a Therapeutic Strategy for Huntington's Disease. J Huntingtons Dis 2018; 6:303-325. [PMID: 29254102 PMCID: PMC5757655 DOI: 10.3233/jhd-170275] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Huntington’s disease (HD) is an autosomal dominant neurodegenerative disorder caused by CAG repeat expansions in the IT15 gene which encodes the huntingtin (HTT) protein. Currently, no treatments capable of preventing or slowing disease progression exist. Disease modifying therapeutics for HD would be expected to target a comprehensive set of degenerative processes given the diverse mechanisms contributing to HD pathogenesis including neuroinflammation, excitotoxicity, and transcription dysregulation. A major contributor to HD-related degeneration is mutant HTT-induced loss of neurotrophic support. Thus, neurotrophin (NT) receptors have emerged as therapeutic targets in HD. The considerable overlap between NT signaling networks and those dysregulated by mutant HTT provides strong theoretical support for this approach. This review will focus on the contributions of disrupted NT signaling in HD-related neurodegeneration and how targeting NT receptors to augment pro-survival signaling and/or to inhibit degenerative signaling may combat HD pathologies. Therapeutic strategies involving NT delivery, peptidomimetics, and the targeting of specific NT receptors (e.g., Trks or p75NTR), particularly with small molecule ligands, are discussed.
Collapse
Affiliation(s)
- Danielle A Simmons
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
53
|
Stewart AN, Matyas JJ, Welchko RM, Goldsmith AD, Zeiler SE, Hochgeschwender U, Lu M, Nan Z, Rossignol J, Dunbar GL. SDF-1 overexpression by mesenchymal stem cells enhances GAP-43-positive axonal growth following spinal cord injury. Restor Neurol Neurosci 2018; 35:395-411. [PMID: 28598857 DOI: 10.3233/rnn-160678] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE Utilizing genetic overexpression of trophic molecules in cell populations has been a promising strategy to develop cell replacement therapies for spinal cord injury (SCI). Over-expressing the chemokine, stromal derived factor-1 (SDF-1α), which has chemotactic effects on many cells of the nervous system, offers a promising strategy to promote axonal regrowth following SCI. The purpose of this study was to explore the effects of human SDF-1α, when overexpressed by mesenchymal stem cells (MSCs), on axonal growth and motor behavior in a contusive rat model of SCI. METHODS Using a transwell migration assay, the paracrine effects of MSCs, which were engineered to secrete human SDF-1α (SDF-1-MSCs), were assessed on cultured neural stem cells (NSCs). For in vivo analyses, the SDF-1-MSCs, unaltered MSCs, or Hanks Buffered Saline Solution (vehicle) were injected into the lesion epicenter of rats at 9-days post-SCI. Behavior was analyzed for 7-weeks post-injury, using the Basso, Beattie, and Bresnahan (BBB) scale of locomotor functions. Immunohistochemistry was performed to evaluate major histopathological outcomes, including gliosis, inflammation, white matter sparing, and cavitation. New axonal outgrowth was characterized using immunohistochemistry against the neuron specific growth-associated protein-43 (GAP-43). RESULTS The results of these experiments demonstrate that the overexpression of SDF-1α by MSCs can enhance the migration of NSCs in vitro. Although only modest functional improvements were observed following transplantation of SDF-1-MSCs, a significant reduction in cavitation surrounding the lesion, and an increased density of GAP-43-positive axons inside the SCI lesion/graft site were found. CONCLUSION The results from these experiments support the potential role for utilizing SDF-1α as a treatment for enhancing growth and regeneration of axons after traumatic SCI.
Collapse
Affiliation(s)
- Andrew Nathaniel Stewart
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, USA.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, USA
| | - Jessica Jane Matyas
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, USA.,Department of Psychology, Central Michigan University, Mount Pleasant, MI, USA
| | - Ryan Matthew Welchko
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, USA.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, USA
| | - Alison Delanie Goldsmith
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, USA.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, USA
| | - Sarah Elizabeth Zeiler
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, USA.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, USA
| | - Ute Hochgeschwender
- Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, USA.,College of Medicine, Central Michigan University, Mount Pleasant, MI, USA
| | - Ming Lu
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, USA.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, USA.,Department of Psychology, Central Michigan University, Mount Pleasant, MI, USA
| | - Zhenhong Nan
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, USA.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, USA.,Department of Psychology, Central Michigan University, Mount Pleasant, MI, USA
| | - Julien Rossignol
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, USA.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, USA.,College of Medicine, Central Michigan University, Mount Pleasant, MI, USA
| | - Gary Leo Dunbar
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, USA.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, USA.,Department of Psychology, Central Michigan University, Mount Pleasant, MI, USA.,Field Neurosciences Inst., 4677 Towne Centre Rd. Suite 101 Saginaw, MI, USA
| |
Collapse
|
54
|
Holley SM, Kamdjou T, Reidling JC, Fury B, Coleal-Bergum D, Bauer G, Thompson LM, Levine MS, Cepeda C. Therapeutic effects of stem cells in rodent models of Huntington's disease: Review and electrophysiological findings. CNS Neurosci Ther 2018; 24:329-342. [PMID: 29512295 DOI: 10.1111/cns.12839] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 02/13/2018] [Accepted: 02/13/2018] [Indexed: 01/01/2023] Open
Abstract
The principal symptoms of Huntington's disease (HD), chorea, cognitive deficits, and psychiatric symptoms are associated with the massive loss of striatal and cortical projection neurons. As current drug therapies only partially alleviate symptoms, finding alternative treatments has become peremptory. Cell replacement using stem cells is a rapidly expanding field that offers such an alternative. In this review, we examine recent studies that use mesenchymal cells, as well as pluripotent, cell-derived products in animal models of HD. Additionally, we provide further electrophysiological characterization of a human neural stem cell line, ESI-017, which has already demonstrated disease-modifying properties in two mouse models of HD. Overall, the field of regenerative medicine represents a viable and promising avenue for the treatment of neurodegenerative disorders including HD.
Collapse
Affiliation(s)
- Sandra M Holley
- Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Talia Kamdjou
- Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Jack C Reidling
- Institute for Memory Impairment and Neurological Disorders, University of California, Irvine, CA, USA
| | - Brian Fury
- Institute for Regenerative Cures, University of California, Davis, Sacramento, CA, USA
| | - Dane Coleal-Bergum
- Institute for Regenerative Cures, University of California, Davis, Sacramento, CA, USA
| | - Gerhard Bauer
- Institute for Regenerative Cures, University of California, Davis, Sacramento, CA, USA
| | - Leslie M Thompson
- Institute for Memory Impairment and Neurological Disorders, University of California, Irvine, CA, USA.,Department of Neurobiology & Behavior and Department of Psychiatry & Human Behavior, University of California, Irvine, CA, USA.,Sue and Bill Gross Stem Cell Center, University of California, Irvine, CA, USA
| | - Michael S Levine
- Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Carlos Cepeda
- Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| |
Collapse
|
55
|
André EM, Daviaud N, Sindji L, Cayon J, Perrot R, Montero-Menei CN. A novel ex vivo Huntington's disease model for studying GABAergic neurons and cell grafts by laser microdissection. PLoS One 2018; 13:e0193409. [PMID: 29505597 PMCID: PMC5837106 DOI: 10.1371/journal.pone.0193409] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 02/09/2018] [Indexed: 02/05/2023] Open
Abstract
Organotypic brain slice cultures have been recently used to study neurodegenerative disorders such as Parkinson’s disease and Huntington’s disease (HD). They preserve brain three-dimensional architecture, synaptic connectivity and brain cells microenvironment. Here, we developed an innovative model of Huntington’s disease from coronal rat brain slices, that include all the areas involved in the pathology. HD-like neurodegeneration was obtained in only one week, in a single step, during organotypic slice preparation, without the use of neurotoxins. HD-like histopathology was analysed and after one week, a reduction of 40% of medium spiny neurons was observed. To analyse new therapeutic approaches in this innovative HD model, we developed a novel protocol of laser microdissection to isolate and analyse by RT-qPCR, grafted cells as well as surrounding tissue of fresh organotypic slices. We determined that laser microdissection could be performed on a 400μm organotypic slice after alcohol dehydration protocol, allowing the analysis of mRNA expression in the rat tissue as well as in grafted cells. In conclusion, we developed a new approach for modeling Huntington's disease ex vivo, and provided a useful innovative method for screening new potential therapies for neurodegenerative diseases especially when associated with laser microdissection.
Collapse
Affiliation(s)
- E. M. André
- CRCINA, INSERM, Université de Nantes, Université d’Angers, Angers, France
| | - N. Daviaud
- CRCINA, INSERM, Université de Nantes, Université d’Angers, Angers, France
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - L. Sindji
- CRCINA, INSERM, Université de Nantes, Université d’Angers, Angers, France
| | - J. Cayon
- PACEM, Angers University, Angers, France
| | - R. Perrot
- SCIAM, Angers University, Angers, France
| | - C. N. Montero-Menei
- CRCINA, INSERM, Université de Nantes, Université d’Angers, Angers, France
- * E-mail:
| |
Collapse
|
56
|
De la Rosa MB, Kozik EM, Sakaguchi DS. Adult Stem Cell-Based Strategies for Peripheral Nerve Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1119:41-71. [PMID: 30151648 DOI: 10.1007/5584_2018_254] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Peripheral nerve injuries (PNI) occur as the result of sudden trauma and can lead to life-long disability, reduced quality of life, and heavy economic and social burdens. Although the peripheral nervous system (PNS) has the intrinsic capacity to regenerate and regrow axons to a certain extent, current treatments frequently show incomplete recovery with poor functional outcomes, particularly for large PNI. Many surgical procedures are available to halt the propagation of nerve damage, and the choice of a procedure depends on the extent of the injury. In particular, recovery from large PNI gaps is difficult to achieve without any therapeutic intervention or some form of tissue/cell-based therapy. Autologous nerve grafting, considered the "gold standard" is often implemented for treatment of gap formation type PNI. Although these surgical procedures provide many benefits, there are still considerable limitations associated with such procedures as donor site morbidity, neuroma formation, fascicle mismatch, and scarring. To overcome such restrictions, researchers have explored various avenues to improve post-surgical outcomes. The most commonly studied methods include: cell transplantation, growth factor delivery to stimulate regenerating axons and implanting nerve guidance conduits containing replacement cells at the site of injury. Replacement cells which offer maximum benefits for the treatment of PNI, are Schwann cells (SCs), which are the peripheral glial cells and in part responsible for clearing out debris from the site of injury. Additionally, they release growth factors to stimulate myelination and axonal regeneration. Both primary SCs and genetically modified SCs enhance nerve regeneration in animal models; however, there is no good source for extracting SCs and the only method to obtain SCs is by sacrificing a healthy nerve. To overcome such challenges, various cell types have been investigated and reported to enhance nerve regeneration.In this review, we have focused on cell-based strategies aimed to enhance peripheral nerve regeneration, in particular the use of mesenchymal stem cells (MSCs). Mesenchymal stem cells are preferred due to benefits such as autologous transplantation, routine isolation procedures, and paracrine and immunomodulatory properties. Mesenchymal stem cells have been transplanted at the site of injury either directly in their native form (undifferentiated) or in a SC-like form (transdifferentiated) and have been shown to significantly enhance nerve regeneration. In addition to transdifferentiated MSCs, some studies have also transplanted ex-vivo genetically modified MSCs that hypersecrete growth factors to improve neuroregeneration.
Collapse
Affiliation(s)
- Metzere Bierlein De la Rosa
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA.,Veterinary Specialty Center, Buffalo Grove, IL, USA
| | - Emily M Kozik
- Biology Program, Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, USA.,Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, USA
| | - Donald S Sakaguchi
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA. .,Biology Program, Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, USA. .,Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, USA. .,Neuroscience Program, Iowa State University, Ames, IA, USA.
| |
Collapse
|
57
|
Stem Cell-Based Therapies for Polyglutamine Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1049:439-466. [DOI: 10.1007/978-3-319-71779-1_21] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
58
|
Connor B. Concise Review: The Use of Stem Cells for Understanding and Treating Huntington's Disease. Stem Cells 2017; 36:146-160. [PMID: 29178352 DOI: 10.1002/stem.2747] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 10/13/2017] [Indexed: 12/20/2022]
Abstract
Two decades ago, researchers identified that a CAG expansion mutation in the huntingtin (HTT) gene was involved in the pathogenesis of Huntington's disease (HD). However, since the identification of the HTT gene, there has been no advance in the development of therapeutic strategies to prevent or reduce the progression of HD. With the recent advances in stem cell biology and human cell reprogramming technologies, several novel and exciting pathways have emerged allowing researchers to enhance their understanding of the pathogenesis of HD, to identify and screen potential drug targets, and to explore alternative donor cell sources for cell replacement therapy. This review will discuss the role of compensatory neurogenesis in the HD brain, the use of stem cell-based therapies for HD to replace or prevent cell loss, and the recent advance of cell reprogramming to model and/or treat HD. These new technologies, coupled with advances in genome editing herald a promising new era for HD research with the potential to identify a therapeutic strategy to alleviate this debilitating disorder. Stem Cells 2018;36:146-160.
Collapse
Affiliation(s)
- Bronwen Connor
- Department of Pharmacology and Clinical Pharmacology, Centre for Brain Research, School of Medical Science, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
59
|
Al-Gharaibeh A, Culver R, Stewart AN, Srinageshwar B, Spelde K, Frollo L, Kolli N, Story D, Paladugu L, Anwar S, Crane A, Wyse R, Maiti P, Dunbar GL, Rossignol J. Induced Pluripotent Stem Cell-Derived Neural Stem Cell Transplantations Reduced Behavioral Deficits and Ameliorated Neuropathological Changes in YAC128 Mouse Model of Huntington's Disease. Front Neurosci 2017; 11:628. [PMID: 29209158 PMCID: PMC5701605 DOI: 10.3389/fnins.2017.00628] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/27/2017] [Indexed: 01/01/2023] Open
Abstract
Huntington's disease (HD) is a genetic neurodegenerative disorder characterized by neuronal loss and motor dysfunction. Although there is no effective treatment, stem cell transplantation offers a promising therapeutic strategy, but the safety and efficacy of this approach needs to be optimized. The purpose of this study was to test the potential of intra-striatal transplantation of induced pluripotent stem cell-derived neural stem cells (iPS-NSCs) for treating HD. For this purpose, we developed mouse adenovirus-generated iPSCs, differentiated them into neural stem cells in vitro, labeled them with Hoechst, and transplanted them bilaterally into striata of 10-month old wild type (WT) and HD YAC128 mice. We assessed the efficiency of these transplanted iPS-NSCs to reduce motor deficits in YAC128 mice by testing them on an accelerating rotarod task at 1 day prior to transplantation, and then weekly for 10 weeks. Our results showed an amelioration of locomotor deficits in YAC128 mice that received iPS-NSC transplantations. Following testing, the mice were sacrificed, and their brains were analyzed using immunohistochemistry and Western blot (WB). The results from our histological examinations revealed no signs of tumors and evidence that many iPS-NSCs survived and differentiated into region-specific neurons (medium spiny neurons) in both WT and HD mice, as confirmed by co-labeling of Hoechst-labeled transplanted cells with NeuN and DARPP-32. Also, counts of Hoechst-labeled cells revealed that a higher proportion were co-labeled with DARPP-32 and NeuN in HD-, compared to WT- mice, suggesting a dissimilar differentiation pattern in HD mice. Whereas significant decreases were found in counts of NeuN- and DARPP-32-labeled cells, and for neuronal density measures in striata of HD vehicle controls, such decrements were not observed in the iPS-NSCs-transplanted-HD mice. WB analysis showed increase of BDNF and TrkB levels in striata of transplanted HD mice compared to HD vehicle controls. Collectively, our data suggest that iPS-NSCs may provide an effective option for neuronal replacement therapy in HD.
Collapse
Affiliation(s)
- Abeer Al-Gharaibeh
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States
| | - Rebecca Culver
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States
| | - Andrew N Stewart
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States
| | - Bhairavi Srinageshwar
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States
| | - Kristin Spelde
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States
| | - Laura Frollo
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States
| | - Nivya Kolli
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States
| | - Darren Story
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States.,Department of Psychology, Central Michigan University, Mount Pleasant, MI, United States
| | - Leela Paladugu
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States
| | - Sarah Anwar
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States
| | - Andrew Crane
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States
| | - Robert Wyse
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States
| | - Panchanan Maiti
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States.,Department of Psychology, Central Michigan University, Mount Pleasant, MI, United States.,Field Neurosciences Institute, St. Mary's of Michigan, Saginaw, MI, United States
| | - Gary L Dunbar
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States.,Department of Psychology, Central Michigan University, Mount Pleasant, MI, United States.,Field Neurosciences Institute, St. Mary's of Michigan, Saginaw, MI, United States
| | - Julien Rossignol
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States.,Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States.,College of Medicine, Central Michigan University, Mt Pleasant, MI, United States
| |
Collapse
|
60
|
D’Souza GX, Waldvogel HJ. Targeting the Cholinergic System to Develop a Novel Therapy for Huntington's Disease. J Huntingtons Dis 2017; 5:333-342. [PMID: 27983560 PMCID: PMC5181681 DOI: 10.3233/jhd-160200] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In this review, we outline the role of the cholinergic system in Huntington’s disease, and briefly describe the dysfunction of cholinergic transmission, cholinergic neurons, cholinergic receptors and cholinergic survival factors observed in post-mortem human brains and animal models of Huntington’s disease. We postulate how the dysfunctional cholinergic system can be targeted to develop novel therapies for Huntington’s disease, and discuss the beneficial effects of cholinergic therapies in pre-clinical and clinical studies.
Collapse
Affiliation(s)
| | - Henry J. Waldvogel
- Correspondence to: Associate Professor Henry J. Waldvogel, Centre for Brain Research and Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand. Tel.: +64 9 923 6051; E-mail:
| |
Collapse
|
61
|
Lo Furno D, Mannino G, Giuffrida R. Functional role of mesenchymal stem cells in the treatment of chronic neurodegenerative diseases. J Cell Physiol 2017; 233:3982-3999. [PMID: 28926091 DOI: 10.1002/jcp.26192] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 09/15/2017] [Indexed: 12/14/2022]
Abstract
Mesenchymal stem cells (MSCs) can differentiate into not only cells of mesodermal lineages, but also into endodermal and ectodermal derived elements, including neurons and glial cells. For this reason, MSCs have been extensively investigated to develop cell-based therapeutic strategies, especially in pathologies whose pharmacological treatments give poor results, if any. As in the case of irreversible neurological disorders characterized by progressive neuronal death, in which behavioral and cognitive functions of patients inexorably decline as the disease progresses. In this review, we focus on the possible functional role exerted by MSCs in the treatment of some disabling neurodegenerative disorders such as Alzheimer's Disease, Amyotrophic Lateral Sclerosis, Huntington's Disease, and Parkinson's Disease. Investigations have been mainly performed in vitro and in animal models by using MSCs generally originated from umbilical cord, bone marrow, or adipose tissue. Positive results obtained have prompted several clinical trials, the number of which is progressively increasing worldwide. To date, many of them have been primarily addressed to verify the safety of the procedures but some improvements have already been reported, fortunately. Although the exact mechanisms of MSC-induced beneficial activities are not entirely defined, they include neurogenesis and angiogenesis stimulation, antiapoptotic, immunomodulatory, and anti-inflammatory actions. Most effects would be exerted through their paracrine expression of neurotrophic factors and cytokines, mainly delivered at damaged regions, given the innate propensity of MSCs to home to injured sites. Hopefully, in the near future more efficacious cell-replacement therapies will be developed to substantially restore disease-disrupted brain circuitry.
Collapse
Affiliation(s)
- Debora Lo Furno
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Giuliana Mannino
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Rosario Giuffrida
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| |
Collapse
|
62
|
Co-transplantation of mesenchymal and neural stem cells and overexpressing stromal-derived factor-1 for treating spinal cord injury. Brain Res 2017; 1672:91-105. [DOI: 10.1016/j.brainres.2017.07.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/12/2017] [Accepted: 07/05/2017] [Indexed: 12/17/2022]
|
63
|
Tartaglione AM, Popoli P, Calamandrei G. Regenerative medicine in Huntington's disease: Strengths and weaknesses of preclinical studies. Neurosci Biobehav Rev 2017; 77:32-47. [PMID: 28223129 DOI: 10.1016/j.neubiorev.2017.02.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/26/2017] [Accepted: 02/17/2017] [Indexed: 01/22/2023]
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disorder, characterized by impairment in motor, cognitive and psychiatric domains. Currently, there is no specific therapy to act on the onset or progression of HD. The marked neuronal death observed in HD is a main argument in favour of stem cells (SCs) transplantation as a promising therapeutic perspective to replace the population of lost neurons and restore the functionality of the damaged circuitry. The availability of rodent models of HD encourages the investigation of the restorative potential of SCs transplantation longitudinally. However, the results of preclinical studies on SCs therapy in HD are so far largely inconsistent; this hampers the individuation of the more appropriate model and precludes the comparative analysis of transplant efficacy on behavioural end points. Thus, this review will describe the state of the art of in vivo research on SCs therapy in HD, analysing in a translational perspective the strengths and weaknesses of animal studies investigating the therapeutic potential of cell transplantation on HD progression.
Collapse
Affiliation(s)
- A M Tartaglione
- Centre for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - P Popoli
- National Centre for Medicines Research and Preclinical/Clinical Evaluation, Rome, Italy
| | - G Calamandrei
- Centre for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
64
|
Gowing G, Svendsen S, Svendsen CN. Ex vivo gene therapy for the treatment of neurological disorders. PROGRESS IN BRAIN RESEARCH 2017; 230:99-132. [PMID: 28552237 DOI: 10.1016/bs.pbr.2016.11.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ex vivo gene therapy involves the genetic modification of cells outside of the body to produce therapeutic factors and their subsequent transplantation back into patients. Various cell types can be genetically engineered. However, with the explosion in stem cell technologies, neural stem/progenitor cells and mesenchymal stem cells are most often used. The synergy between the effect of the new cell and the additional engineered properties can often provide significant benefits to neurodegenerative changes in the brain. In this review, we cover both preclinical animal studies and clinical human trials that have used ex vivo gene therapy to treat neurological disorders with a focus on Parkinson's disease, Huntington's disease, Alzheimer's disease, ALS, and stroke. We highlight some of the major advances in this field including new autologous sources of pluripotent stem cells, safer ways to introduce therapeutic transgenes, and various methods of gene regulation. We also address some of the remaining hurdles including tunable gene regulation, in vivo cell tracking, and rigorous experimental design. Overall, given the current outcomes from researchers and clinical trials, along with exciting new developments in ex vivo gene and cell therapy, we anticipate that successful treatments for neurological diseases will arise in the near future.
Collapse
Affiliation(s)
- Genevieve Gowing
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States; Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Soshana Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States; Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Clive N Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States; Cedars-Sinai Medical Center, Los Angeles, CA, United States.
| |
Collapse
|
65
|
Mariga A, Mitre M, Chao MV. Consequences of brain-derived neurotrophic factor withdrawal in CNS neurons and implications in disease. Neurobiol Dis 2017; 97:73-79. [PMID: 27015693 PMCID: PMC5295364 DOI: 10.1016/j.nbd.2016.03.009] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 02/20/2016] [Accepted: 03/09/2016] [Indexed: 01/07/2023] Open
Abstract
Growth factor withdrawal has been studied across different species and has been shown to have dramatic consequences on cell survival. In the nervous system, withdrawal of nerve growth factor (NGF) from sympathetic and sensory neurons results in substantial neuronal cell death, signifying a requirement for NGF for the survival of neurons in the peripheral nervous system (PNS). In contrast to the PNS, withdrawal of central nervous system (CNS) enriched brain-derived neurotrophic factor (BDNF) has little effect on cell survival but is indispensible for synaptic plasticity. Given that most early events in neuropsychiatric disorders are marked by a loss of synapses, lack of BDNF may thus be an important part of a cascade of events that leads to neuronal degeneration. Here we review reports on the effects of BDNF withdrawal on CNS neurons and discuss the relevance of the loss in disease.
Collapse
Affiliation(s)
- Abigail Mariga
- Department of Cell Biology, New York University School of Medicine, New York, NY, 10016, United States; Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY, 10016, United States
| | - Mariela Mitre
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, 10016, United States; Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY, 10016, United States
| | - Moses V Chao
- Department of Cell Biology, New York University School of Medicine, New York, NY, 10016, United States; Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, 10016, United States; Department of Psychiatry, New York University School of Medicine, New York, NY, 10016, United States; Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY, 10016, United States
| |
Collapse
|
66
|
Li W, Tong HI, Gorantla S, Poluektova LY, Gendelman HE, Lu Y. Neuropharmacologic Approaches to Restore the Brain's Microenvironment. J Neuroimmune Pharmacol 2016; 11:484-94. [PMID: 27352074 PMCID: PMC4985494 DOI: 10.1007/s11481-016-9686-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 05/25/2016] [Indexed: 12/15/2022]
Abstract
Maintaining the central nervous system microenvironment after injury, infection, inflammatory and degenerative diseases is contingent upon adequate control of glial homeostatic functions. Disease is caused by microbial, environmental and endogenous factors that compromise ongoing nervous system functions. The final result is neuronal injury, dropout and nerve connection loss, and these underlie the pathobiology of Alzheimer's and Parkinson's disease, amyotrophic lateral sclerosis, stroke, and bacterial, parasitic and viral infections. However, what promotes disease are homeostatic changes in the brain's microenvironment affected by innate glial immune pro-inflammatory and adaptive immune responses. These events disturb the brain's metabolic activities and communication abilities. How the process affects the brain's regulatory functions that can be harnessed for therapeutic gain is the subject at hand. Specific examples are provided that serve to modulate inflammation and improve disease outcomes specifically for HIV-associated neurocognitive disorders.
Collapse
Affiliation(s)
- Weizhe Li
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Hsin-I Tong
- Department of Public Health Sciences, Environmental Health Laboratory, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Santhi Gorantla
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Larisa Y Poluektova
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Yuanan Lu
- Department of Public Health Sciences, Environmental Health Laboratory, University of Hawaii at Manoa, Honolulu, HI, 96822, USA.
| |
Collapse
|
67
|
Deng P, Torrest A, Pollock K, Dahlenburg H, Annett G, Nolta JA, Fink KD. Clinical trial perspective for adult and juvenile Huntington's disease using genetically-engineered mesenchymal stem cells. Neural Regen Res 2016; 11:702-5. [PMID: 27335539 PMCID: PMC4904446 DOI: 10.4103/1673-5374.182682] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Progress to date from our group and others indicate that using genetically-engineered mesenchymal stem cells (MSC) to secrete brain-derived neurotrophic factor (BDNF) supports our plan to submit an Investigational New Drug application to the Food and Drug Administration for the future planned Phase 1 safety and tolerability trial of MSC/BDNF in patients with Huntington's disease (HD). There are also potential applications of this approach beyond HD. Our biological delivery system for BDNF sets the precedent for adult stem cell therapy in the brain and could potentially be modified for other neurodegenerative disorders such as amyotrophic lateral sclerosis (ALS), spinocerebellar ataxia (SCA), Alzheimer's disease, and some forms of Parkinson's disease. The MSC/BDNF product could also be considered for studies of regeneration in traumatic brain injury, spinal cord and peripheral nerve injury. This work also provides a platform for our future gene editing studies, since we will again use MSCs to deliver the needed molecules into the central nervous system.
Collapse
Affiliation(s)
- Peter Deng
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, CA, USA
| | - Audrey Torrest
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, CA, USA
| | - Kari Pollock
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, CA, USA
| | - Heather Dahlenburg
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, CA, USA
| | - Geralyn Annett
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, CA, USA
| | - Jan A Nolta
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, CA, USA
| | - Kyle D Fink
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, CA, USA
| |
Collapse
|
68
|
Deng P, Anderson JD, Yu AS, Annett G, Fink KD, Nolta JA. Engineered BDNF producing cells as a potential treatment for neurologic disease. Expert Opin Biol Ther 2016; 16:1025-33. [PMID: 27159050 DOI: 10.1080/14712598.2016.1183641] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Brain-derived neurotrophic factor (BDNF) has been implicated in wide range of neurological diseases and injury. This neurotrophic factor is vital for neuronal health, survival, and synaptic connectivity. Many therapies focus on the restoration or enhancement of BDNF following injury or disease progression. AREAS COVERED The present review will focus on the mechanisms in which BDNF exerts its beneficial functioning, current BDNF therapies, issues and potential solutions for delivery of neurotrophic factors to the central nervous system, and other disease indications that may benefit from overexpression or restoration of BDNF. EXPERT OPINION Due to the role of BDNF in neuronal development, maturation, and health, BDNF is implicated in numerous neurological diseases making it a prime therapeutic agent. Numerous studies have shown the therapeutic potential of BDNF in a number of neurodegenerative disease models and in acute CNS injury, however clinical translation has fallen short due to issues in delivering this molecule. The use of MSC as a delivery platform for BDNF holds great promise for clinical advancement of neurotrophic factor restoration. The ease with which MSC can be engineered opens the door to the possibility of using this cell-based delivery system to advance a BDNF therapy to the clinic.
Collapse
Affiliation(s)
- Peter Deng
- a Stem Cell Program and Institute for Regenerative Cures , University of California Davis Health Systems , Sacramento , CA , USA.,b Genome Center, MIND Institute, and Biochemistry and Molecular Medicine , University of California , Davis , CA , USA
| | - Johnathon D Anderson
- a Stem Cell Program and Institute for Regenerative Cures , University of California Davis Health Systems , Sacramento , CA , USA
| | - Abigail S Yu
- b Genome Center, MIND Institute, and Biochemistry and Molecular Medicine , University of California , Davis , CA , USA
| | - Geralyn Annett
- a Stem Cell Program and Institute for Regenerative Cures , University of California Davis Health Systems , Sacramento , CA , USA
| | - Kyle D Fink
- a Stem Cell Program and Institute for Regenerative Cures , University of California Davis Health Systems , Sacramento , CA , USA
| | - Jan A Nolta
- a Stem Cell Program and Institute for Regenerative Cures , University of California Davis Health Systems , Sacramento , CA , USA
| |
Collapse
|
69
|
Pollock K, Dahlenburg H, Nelson H, Fink KD, Cary W, Hendrix K, Annett G, Torrest A, Deng P, Gutierrez J, Nacey C, Pepper K, Kalomoiris S, D Anderson J, McGee J, Gruenloh W, Fury B, Bauer G, Duffy A, Tempkin T, Wheelock V, Nolta JA. Human Mesenchymal Stem Cells Genetically Engineered to Overexpress Brain-derived Neurotrophic Factor Improve Outcomes in Huntington's Disease Mouse Models. Mol Ther 2016; 24:965-77. [PMID: 26765769 PMCID: PMC4881765 DOI: 10.1038/mt.2016.12] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 12/05/2015] [Indexed: 12/15/2022] Open
Abstract
Huntington's disease (HD) is a fatal degenerative autosomal dominant neuropsychiatric disease that causes neuronal death and is characterized by progressive striatal and then widespread brain atrophy. Brain-derived neurotrophic factor (BDNF) is a lead candidate for the treatment of HD, as it has been shown to prevent cell death and to stimulate the growth and migration of new neurons in the brain in transgenic mouse models. BDNF levels are reduced in HD postmortem human brain. Previous studies have shown efficacy of mesenchymal stem/stromal cells (MSC)/BDNF using murine MSCs, and the present study used human MSCs to advance the therapeutic potential of the MSC/BDNF platform for clinical application. Double-blinded studies were performed to examine the effects of intrastriatally transplanted human MSC/BDNF on disease progression in two strains of immune-suppressed HD transgenic mice: YAC128 and R6/2. MSC/BDNF treatment decreased striatal atrophy in YAC128 mice. MSC/BDNF treatment also significantly reduced anxiety as measured in the open-field assay. Both MSC and MSC/BDNF treatments induced a significant increase in neurogenesis-like activity in R6/2 mice. MSC/BDNF treatment also increased the mean lifespan of the R6/2 mice. Our genetically modified MSC/BDNF cells set a precedent for stem cell-based neurotherapeutics and could potentially be modified for other neurodegenerative disorders such as amyotrophic lateral sclerosis, Alzheimer's disease, and some forms of Parkinson's disease. These cells provide a platform delivery system for future studies involving corrective gene-editing strategies.
Collapse
Affiliation(s)
- Kari Pollock
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Heather Dahlenburg
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Haley Nelson
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Kyle D Fink
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Whitney Cary
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Kyle Hendrix
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Geralyn Annett
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Audrey Torrest
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Peter Deng
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Joshua Gutierrez
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Catherine Nacey
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Karen Pepper
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Stefanos Kalomoiris
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Johnathon D Anderson
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Jeannine McGee
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - William Gruenloh
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Brian Fury
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Gerhard Bauer
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| | - Alexandria Duffy
- Department of Neurology, University of California Davis Health System, Sacramento, California, USA
| | - Theresa Tempkin
- Department of Neurology, University of California Davis Health System, Sacramento, California, USA
| | - Vicki Wheelock
- Department of Neurology, University of California Davis Health System, Sacramento, California, USA
| | - Jan A Nolta
- Stem Cell Program and Institute for Regenerative Cures, University of California Davis Health System, Sacramento, California, USA
| |
Collapse
|
70
|
Matchynski-Franks JJ, Pappas C, Rossignol J, Reinke T, Fink K, Crane A, Twite A, Lowrance SA, Song C, Dunbar GL. Mesenchymal Stem Cells as Treatment for Behavioral Deficits and Neuropathology in the 5xFAD Mouse Model of Alzheimer's Disease. Cell Transplant 2016; 25:687-703. [DOI: 10.3727/096368916x690818] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by a progressive loss of memory and other cognitive disturbances. The neuropathology of AD includes the major hallmarks of toxic amyloid-β oligomer accumulation and neurofibrillary tangles, as well as increased oxidative stress, cholinergic dysfunction, synapse loss, changes in endogenous neurotrophic factors, and overall degeneration of the brain. Adult mesenchymal stem cells (MSCs) offer the potential for a readily available treatment that would be long lasting, have low likelihood of rejection, and could target a variety of pathological deficits. MSCs have been shown to be effective in alleviating symptoms in some transgenic models of AD, but the optimal location for transplanting MSCs has yet to be determined. In the present study, the behavioral effects of transplantation of MSCs into the lateral ventricles, the hippocampus, or both of these regions were compared in the 5xFAD mouse model of AD. The results indicate that MSC transplants effectively reduce learning deficits in the 5xFAD mouse model and demonstrate a clear impact of MSCs on the levels of Aβ42 in the brains of 5xFAD mice. Overall, these findings support the hypothesis that MSCs may be a viable treatment for AD, especially when injected into the lateral ventricles.
Collapse
Affiliation(s)
- Jessica J. Matchynski-Franks
- Field Neurosciences Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI, USA
- Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI, USA
- Department of Psychology and Behavioral Sciences, Rochester College, Rochester Hills, MI, USA
| | - Colleen Pappas
- Field Neurosciences Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI, USA
- Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI, USA
| | - Julien Rossignol
- Field Neurosciences Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI, USA
- Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI, USA
- College of Medicine, Central Michigan University, Mt. Pleasant, MI, USA
| | - Tiffany Reinke
- Field Neurosciences Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI, USA
- Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI, USA
| | - Kyle Fink
- Field Neurosciences Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI, USA
- Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI, USA
| | - Andrew Crane
- Field Neurosciences Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI, USA
- Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI, USA
| | - Alison Twite
- Field Neurosciences Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI, USA
- Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI, USA
| | - Steven A. Lowrance
- Field Neurosciences Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI, USA
- Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI, USA
| | - Cheng Song
- Field Neurosciences Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI, USA
- Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI, USA
| | - Gary L. Dunbar
- Field Neurosciences Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI, USA
- Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI, USA
- Department of Psychology, Central Michigan University, Mt. Pleasant, MI, USA
- St. Mary's of Michigan Field Neurosciences Institute, Saginaw MI, USA
| |
Collapse
|
71
|
André EM, Passirani C, Seijo B, Sanchez A, Montero-Menei CN. Nano and microcarriers to improve stem cell behaviour for neuroregenerative medicine strategies: Application to Huntington's disease. Biomaterials 2016; 83:347-62. [DOI: 10.1016/j.biomaterials.2015.12.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 12/09/2015] [Accepted: 12/13/2015] [Indexed: 12/22/2022]
|
72
|
Corey-Bloom J, Jia H, Aikin AM, Thomas EA. Disease Modifying Potential of Glatiramer Acetate in Huntington's Disease. J Huntingtons Dis 2016; 3:311-6. [PMID: 25300334 DOI: 10.3233/jhd-140110] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Deficiencies in brain-derived-neurotrophic-factor have been implicated in the pathogenesis of Huntington's disease (HD). OBJECTIVE Glatiramer acetate, an FDA- approved drug used for the treatment of multiple sclerosis, has been shown to increase brain-derived-neurotrophic-factor levels in immune cells; hence, we investigated whether it could have similar effects in striatal cells. METHODS Wild-type and HD striatal cells were treated with glatiramer acetate for 48 hrs. HD transgenic and wild-type mice were injected with glatiramer acetate (1.5 to 1.7 mg/mouse) for five days. These treatments were followed by protein measurements for brain-derived-neurotrophic-factor. RESULTS Glatiramer acetate elicited concentration-dependent increases in brain-derived-neurotrophic-factor protein levels in wild-type and HD striatal cells and in striatal tissue from N171-82Q transgenic mice. Glatiramer acetate also improved metabolic activity of HD striatal cells, and significantly reduced the early hyperactivity phenotype exhibited by N171-82Q transgenic mice. CONCLUSIONS These findings suggest that glatiramer acetate may represent a useful therapeutic approach for HD. The excellent safety and tolerability record of this compound makes it an ideal candidate for drug repurposing efforts.
Collapse
Affiliation(s)
- Jody Corey-Bloom
- Department of Neurosciences, University of California, San Diego, CA, USA
| | - Haiqun Jia
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Alaina M Aikin
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Elizabeth A Thomas
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
73
|
Nowakowski A, Walczak P, Janowski M, Lukomska B. Genetic Engineering of Mesenchymal Stem Cells for Regenerative Medicine. Stem Cells Dev 2015; 24:2219-42. [PMID: 26140302 DOI: 10.1089/scd.2015.0062] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs), which can be obtained from various organs and easily propagated in vitro, are one of the most extensively used types of stem cells and have been shown to be efficacious in a broad set of diseases. The unique and highly desirable properties of MSCs include high migratory capacities toward injured areas, immunomodulatory features, and the natural ability to differentiate into connective tissue phenotypes. These phenotypes include bone and cartilage, and these properties predispose MSCs to be therapeutically useful. In addition, MSCs elicit their therapeutic effects by paracrine actions, in which the metabolism of target tissues is modulated. Genetic engineering methods can greatly amplify these properties and broaden the therapeutic capabilities of MSCs, including transdifferentiation toward diverse cell lineages. However, cell engineering can also affect safety and increase the cost of therapy based on MSCs; thus, the advantages and disadvantages of these procedures should be discussed. In this review, the latest applications of genetic engineering methods for MSCs with regenerative medicine purposes are presented.
Collapse
Affiliation(s)
- Adam Nowakowski
- 1 NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences , Warsaw, Poland
| | - Piotr Walczak
- 2 Division of Magnetic Resonance Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine , Baltimore, Maryland.,3 Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland.,4 Department of Radiology, Faculty of Medical Sciences, University of Warmia and Mazury , Olsztyn, Poland
| | - Miroslaw Janowski
- 1 NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences , Warsaw, Poland .,2 Division of Magnetic Resonance Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine , Baltimore, Maryland.,3 Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Barbara Lukomska
- 1 NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences , Warsaw, Poland
| |
Collapse
|
74
|
D'souza N, Rossignoli F, Golinelli G, Grisendi G, Spano C, Candini O, Osturu S, Catani F, Paolucci P, Horwitz EM, Dominici M. Mesenchymal stem/stromal cells as a delivery platform in cell and gene therapies. BMC Med 2015; 13:186. [PMID: 26265166 PMCID: PMC4534031 DOI: 10.1186/s12916-015-0426-0] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 07/17/2015] [Indexed: 02/07/2023] Open
Abstract
Regenerative medicine relying on cell and gene therapies is one of the most promising approaches to repair tissues. Multipotent mesenchymal stem/stromal cells (MSC), a population of progenitors committing into mesoderm lineages, are progressively demonstrating therapeutic capabilities far beyond their differentiation capacities. The mechanisms by which MSC exert these actions include the release of biomolecules with anti-inflammatory, immunomodulating, anti-fibrogenic, and trophic functions. While we expect the spectra of these molecules with a therapeutic profile to progressively expand, several human pathological conditions have begun to benefit from these biomolecule-delivering properties. In addition, MSC have also been proposed to vehicle genes capable of further empowering these functions. This review deals with the therapeutic properties of MSC, focusing on their ability to secrete naturally produced or gene-induced factors that can be used in the treatment of kidney, lung, heart, liver, pancreas, nervous system, and skeletal diseases. We specifically focus on the different modalities by which MSC can exert these functions. We aim to provide an updated understanding of these paracrine mechanisms as a prerequisite to broadening the therapeutic potential and clinical impact of MSC.
Collapse
Affiliation(s)
- Naomi D'souza
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Filippo Rossignoli
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Giulia Golinelli
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Giulia Grisendi
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Carlotta Spano
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Olivia Candini
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Satoru Osturu
- The Division of Hematology/Oncology/BMT, Nationwide Children's Hospital, Departments of Pediatrics and Medicine, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Fabio Catani
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Paolo Paolucci
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Edwin M Horwitz
- The Division of Hematology/Oncology/BMT, Nationwide Children's Hospital, Departments of Pediatrics and Medicine, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Massimo Dominici
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy.
| |
Collapse
|
75
|
Chen S, Zhang XJ, Xie WJ, Qiu HY, Liu H, Le WD. A New VMAT-2 Inhibitor NBI-641449 in the Treatment of Huntington Disease. CNS Neurosci Ther 2015; 21:662-71. [PMID: 26122704 PMCID: PMC6495663 DOI: 10.1111/cns.12425] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 05/22/2015] [Accepted: 05/24/2015] [Indexed: 12/20/2022] Open
Abstract
AIMS To evaluate the effectiveness of a new VMAT-2 inhibitor NBI-641449 in controlling hyperkinetic movements of Huntington disease (HD) and to investigate its possible therapeutic effects. METHODS We applied three different doses of NBI-641449 (1, 10, 100 mg/kg/day) for 2 weeks in 4-month-old YAC128 mice and wild-type (WT) mice. Rotarod performance and locomotive activities were tested during the administration of the drug. The concentration of dopamine (DA) and its metabolites was quantified in the striatal tissues by high-performance liquid chromatography (HPLC). Neuron survival in striatum and huntingtin protein aggregates were assessed with immunostaining. Expression levels of endoplasmic reticulum (ER) stress proteins were detected by immunoblotting. RESULTS Rotarod performance was significantly improved after treatment with low or middle dose of NBI-641449 in YAC128 mice. Open field test showed that NBI-641449 treatment could attenuate the increased horizontal activity (HACTV), total vertical movement, moving time, and moving distance in YAC128 mice. High dose of NBI-641449 might cause sedative effects in WT and YAC128 mice. HPLC showed that NBI-641449 caused a dose-dependent decrease of DA, 3,4-dihydroxyphenylacetic acid, and homovanillic acid levels in the striatum. NeuN and DARPP-32 immunostaining revealed that NBI-641449 had no significant effect on the neuron survival in the striatum. However, NBI-641449 treatment reduced the huntingtin protein aggregates in the cortex of YAC128 mice. In addition, the levels of ER stress proteins were increased in YAC128 mice, which can be suppressed by NBI-641449. CONCLUSIONS These findings suggest that this new VMAT-2 inhibitor NBI-641449 may have therapeutic potential for the treatment of HD.
Collapse
Affiliation(s)
- Sheng Chen
- Institute of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Neurology, Baylor College of medicine, Houston, TX, USA
| | - Xiao-Jie Zhang
- Institute of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wen-Jie Xie
- Diana Helis Henry Medical Research Foundation, New Orleans, LA, USA
| | - Hong-Yan Qiu
- Institute of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hui Liu
- Institute of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wei-Dong Le
- Institute of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Center for Translational Research of Neurology Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
76
|
Siska EK, Koliakos G, Petrakis S. Stem cell models of polyglutamine diseases and their use in cell-based therapies. Front Neurosci 2015; 9:247. [PMID: 26236184 PMCID: PMC4501170 DOI: 10.3389/fnins.2015.00247] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 06/30/2015] [Indexed: 12/20/2022] Open
Abstract
Polyglutamine diseases are fatal neurological disorders that affect the central nervous system. They are caused by mutations in disease genes that contain CAG trinucleotide expansions in their coding regions. These mutations are translated into expanded glutamine chains in pathological proteins. Mutant proteins induce cytotoxicity, form intranuclear aggregates and cause neuronal cell death in specific brain regions. At the moment there is no cure for these diseases and only symptomatic treatments are available. Here, we discuss novel therapeutic approaches that aim in neuronal cell replacement using induced pluripotent or adult stem cells. Additionally, we present the beneficial effect of genetically engineered mesenchymal stem cells and their use as disease models or RNAi/gene delivery vehicles. In combination with their paracrine and cell-trophic properties, such cells may prove useful for the development of novel therapies against polyglutamine diseases.
Collapse
Affiliation(s)
| | - George Koliakos
- Biohellenika Biotechnology Company Thessaloniki, Greece ; Laboratory of Biochemistry, AHEPA University Hospital, Medical School, Aristotle University of Thessaloniki Thessaloniki, Greece
| | | |
Collapse
|
77
|
Rossignol J, Fink KD, Crane AT, Davis KK, Bombard MC, Clerc S, Bavar AM, Lowrance SA, Song C, Witte S, Lescaudron L, Dunbar GL. Reductions in behavioral deficits and neuropathology in the R6/2 mouse model of Huntington's disease following transplantation of bone-marrow-derived mesenchymal stem cells is dependent on passage number. Stem Cell Res Ther 2015; 6:9. [PMID: 25971780 PMCID: PMC4429666 DOI: 10.1186/scrt545] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 01/16/2015] [Accepted: 01/16/2015] [Indexed: 02/06/2023] Open
Abstract
Introduction Huntington’s disease (HD) is an autosomal dominant disorder caused by an expanded CAG repeat (greater than 38) on the short arm of chromosome 4, resulting in loss and dysfunction of neurons in the neostriatum and cortex, leading to cognitive decline, motor dysfunction, and death, typically occurring 15 to 20 years after the onset of motor symptoms. Although an effective treatment for HD has remained elusive, current studies using transplants of bone-marrow-derived mesenchymal stem cells provides considerable promise. This study further investigates the efficacy of these transplants with a focus on comparing how passage number of these cells may affect subsequent efficacy following transplantation. Methods In this study, mesenchymal stem cells isolated from the bone-marrow of mice (BM MSCs), were labeled with Hoechst after low (3 to 8) or high (40 to 50) numbers of passages and then transplanted intrastriatally into 5-week-old R6/2 mice, which carries the N-terminal fragment of the human HD gene (145 to 155 repeats) and rapidly develops symptoms analogous to the human form of the disease. Results It was observed that the transplanted cells survived and the R6/2 mice displayed significant behavioral and morphological sparing compared to untreated R6/2 mice, with R6/2 mice receiving high passage BM MSCs displaying fewer deficits than those receiving low-passage BM MSCs. These beneficial effects are likely due to trophic support, as an increase in brain derived neurotrophic factor mRNA expression was observed in the striatum following transplantation of BM MSCs. Conclusion The results from this study demonstrate that BM MSCs hold significant therapeutic value for HD, and that the amount of time the cells are exposed to in vitro culture conditions can alter their efficacy.
Collapse
Affiliation(s)
- Julien Rossignol
- Field Neurosciences Laboratory for Restorative Neurology, Brain Research and Integrative Neuroscience Center, Program in Neuroscience, 1280 East Campus Drive, HP Building Room 2336, Mount Pleasant, MI, 48859, USA. .,College of Medicine, Central Michigan University, Mount Pleasant, MI, 48859, USA.
| | - Kyle D Fink
- Field Neurosciences Laboratory for Restorative Neurology, Brain Research and Integrative Neuroscience Center, Program in Neuroscience, 1280 East Campus Drive, HP Building Room 2336, Mount Pleasant, MI, 48859, USA. .,Faculté des Science et des Techniques, Université de Nantes, 44300, Nantes, France. .,INSERM U1064, ITUN, 44093, Nantes, France.
| | - Andrew T Crane
- Field Neurosciences Laboratory for Restorative Neurology, Brain Research and Integrative Neuroscience Center, Program in Neuroscience, 1280 East Campus Drive, HP Building Room 2336, Mount Pleasant, MI, 48859, USA.
| | - Kendra K Davis
- Field Neurosciences Laboratory for Restorative Neurology, Brain Research and Integrative Neuroscience Center, Program in Neuroscience, 1280 East Campus Drive, HP Building Room 2336, Mount Pleasant, MI, 48859, USA.
| | - Matthew C Bombard
- Field Neurosciences Laboratory for Restorative Neurology, Brain Research and Integrative Neuroscience Center, Program in Neuroscience, 1280 East Campus Drive, HP Building Room 2336, Mount Pleasant, MI, 48859, USA.
| | - Steven Clerc
- Field Neurosciences Laboratory for Restorative Neurology, Brain Research and Integrative Neuroscience Center, Program in Neuroscience, 1280 East Campus Drive, HP Building Room 2336, Mount Pleasant, MI, 48859, USA.
| | - Angela M Bavar
- Field Neurosciences Laboratory for Restorative Neurology, Brain Research and Integrative Neuroscience Center, Program in Neuroscience, 1280 East Campus Drive, HP Building Room 2336, Mount Pleasant, MI, 48859, USA.
| | - Steven A Lowrance
- Field Neurosciences Laboratory for Restorative Neurology, Brain Research and Integrative Neuroscience Center, Program in Neuroscience, 1280 East Campus Drive, HP Building Room 2336, Mount Pleasant, MI, 48859, USA.
| | - Cheng Song
- Field Neurosciences Laboratory for Restorative Neurology, Brain Research and Integrative Neuroscience Center, Program in Neuroscience, 1280 East Campus Drive, HP Building Room 2336, Mount Pleasant, MI, 48859, USA.
| | - Steven Witte
- Field Neurosciences Laboratory for Restorative Neurology, Brain Research and Integrative Neuroscience Center, Program in Neuroscience, 1280 East Campus Drive, HP Building Room 2336, Mount Pleasant, MI, 48859, USA.
| | - Laurent Lescaudron
- Faculté des Science et des Techniques, Université de Nantes, 44300, Nantes, France. .,INSERM U791, Laboratoire d'Ingenierie Osteo-Articulaire et Dentaire (LIOAD), 44042, Nantes, France.
| | - Gary L Dunbar
- Field Neurosciences Laboratory for Restorative Neurology, Brain Research and Integrative Neuroscience Center, Program in Neuroscience, 1280 East Campus Drive, HP Building Room 2336, Mount Pleasant, MI, 48859, USA. .,Field Neurosciences Institute, Saginaw, MI, 48604, USA.
| |
Collapse
|
78
|
Sakthiswary R, Raymond AA. Stem cell therapy in neurodegenerative diseases: From principles to practice. Neural Regen Res 2015; 7:1822-31. [PMID: 25624807 PMCID: PMC4302533 DOI: 10.3969/j.issn.1673-5374.2012.23.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2012] [Accepted: 06/13/2012] [Indexed: 12/11/2022] Open
Abstract
The lack of curative therapies for neurodegenerative diseases has high economic impact and places huge burden on the society. The contribution of stem cells to cure neurodegenerative diseases has been unraveled and explored extensively over the past few years. Beyond substitution of the lost neurons, stem cells act as immunomodulators and neuroprotectors. A large number of preclinical and a small number of clinical studies have shown beneficial outcomes in this context. In this review, we have summarized the current concepts of stem cell therapy in neurodegenerative diseases and the recent advances in this field, particularly between 2010 and 2012. Further studies should be encouraged to resolve the clinical issues and vague translational findings for maximum optimization of the efficacy of stem cell therapy in neurodegenerative diseases.
Collapse
Affiliation(s)
- Rajalingham Sakthiswary
- Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Bandar Tun Razak 56000, Kuala Lumpur, Malaysia
| | - Azman Ali Raymond
- Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Bandar Tun Razak 56000, Kuala Lumpur, Malaysia
| |
Collapse
|
79
|
Wong BKY, Ehrnhoefer DE, Graham RK, Martin DDO, Ladha S, Uribe V, Stanek LM, Franciosi S, Qiu X, Deng Y, Kovalik V, Zhang W, Pouladi MA, Shihabuddin LS, Hayden MR. Partial rescue of some features of Huntington Disease in the genetic absence of caspase-6 in YAC128 mice. Neurobiol Dis 2015; 76:24-36. [PMID: 25583186 DOI: 10.1016/j.nbd.2014.12.030] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 12/22/2014] [Accepted: 12/31/2014] [Indexed: 12/13/2022] Open
Abstract
Huntington Disease (HD) is a progressive neurodegenerative disease caused by an elongated CAG repeat in the huntingtin (HTT) gene that encodes a polyglutamine tract in the HTT protein. Proteolysis of the mutant HTT protein (mHTT) has been detected in human and murine HD brains and is implicated in the pathogenesis of HD. Of particular importance is the site at amino acid (aa) 586 that contains a caspase-6 (Casp6) recognition motif. Activation of Casp6 occurs presymptomatically in human HD patients and the inhibition of mHTT proteolysis at aa586 in the YAC128 mouse model results in the full rescue of HD-like phenotypes. Surprisingly, Casp6 ablation in two different HD mouse models did not completely prevent the generation of this fragment, and therapeutic benefits were limited, questioning the role of Casp6 in the disease. We have evaluated the impact of the loss of Casp6 in the YAC128 mouse model of HD. Levels of the mHTT-586 fragment are reduced but not absent in the absence of Casp6 and we identify caspase 8 as an alternate enzyme that can generate this fragment. In vivo, the ablation of Casp6 results in a partial rescue of body weight gain, normalized IGF-1 levels, a reversal of the depression-like phenotype and decreased HTT levels. In the YAC128/Casp6-/- striatum there is a concomitant reduction in p62 levels, a marker of autophagic activity, suggesting increased autophagic clearance. These results implicate the HTT-586 fragment as a key contributor to certain features of HD, irrespective of the enzyme involved in its generation.
Collapse
Affiliation(s)
- Bibiana K Y Wong
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada; Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Dagmar E Ehrnhoefer
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Rona K Graham
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada; Research Center on Aging, Department of Physiology and Biophysics, University of Sherbrooke, Sherbrooke, Quebec J1H 5N4, Canada
| | - Dale D O Martin
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Safia Ladha
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Valeria Uribe
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Lisa M Stanek
- Genzyme, a Sanofi Company, Framingham, MA 01701, USA
| | - Sonia Franciosi
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Xiaofan Qiu
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Yu Deng
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Vlad Kovalik
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Weining Zhang
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Mahmoud A Pouladi
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada; Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, 138648, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 138648, Singapore
| | | | - Michael R Hayden
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada.
| |
Collapse
|
80
|
Abstract
In this review, we explore the similarities and differences in the behavioural neurobiology found in the mouse models of Huntington's disease (HD) and the human disease state. The review is organised with a comparative focus on the functional domains of motor control, cognition and behavioural disturbance (akin to psychiatric disturbance in people) and how our knowledge of the underlying physiological changes that are manifest in the HD mouse lines correspond to those seen in the HD clinical population. The review is framed in terms of functional circuitry and neurotransmitter systems and how abnormalities in these systems impact on the behavioural readouts across the mouse lines and how these may correspond to the deficits observed in people. In addition, interpretational issues associated with the data from animal studies are discussed.
Collapse
Affiliation(s)
- Simon P Brooks
- Brain Repair Group, Division of Neuroscience, Cardiff University School of Bioscience, Museum Avenue, Cardiff, Wales, UK,
| | | |
Collapse
|
81
|
Fink KD, Deng P, Torrest A, Stewart H, Pollock K, Gruenloh W, Annett G, Tempkin T, Wheelock V, Nolta JA. Developing stem cell therapies for juvenile and adult-onset Huntington's disease. Regen Med 2015; 10:623-46. [PMID: 26237705 PMCID: PMC6785015 DOI: 10.2217/rme.15.25] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Stem cell therapies have been explored as a new avenue for the treatment of neurologic disease and damage within the CNS in part due to their native ability to mimic repair mechanisms in the brain. Mesenchymal stem cells have been of particular clinical interest due to their ability to release beneficial neurotrophic factors and their ability to foster a neuroprotective microenviroment. While early stem cell transplantation therapies have been fraught with technical and political concerns as well as limited clinical benefits, mesenchymal stem cell therapies have been shown to be clinically beneficial and derivable from nonembryonic, adult sources. The focus of this review will be on emerging and extant stem cell therapies for juvenile and adult-onset Huntington's disease.
Collapse
Affiliation(s)
- Kyle D Fink
- Stem Cell Program & Institute for Regenerative Cures, University of California Davis Health Systems, 2921 Stockton Blvd. Sacramento, CA 95817, USA
| | - Peter Deng
- Stem Cell Program & Institute for Regenerative Cures, University of California Davis Health Systems, 2921 Stockton Blvd. Sacramento, CA 95817, USA
- GenomeCenter, Biochemistry & Molecular Medicine, University of California, 451 Health Sciences Dr. Davis, CA 95616, USA
| | - Audrey Torrest
- Stem Cell Program & Institute for Regenerative Cures, University of California Davis Health Systems, 2921 Stockton Blvd. Sacramento, CA 95817, USA
| | - Heather Stewart
- Stem Cell Program & Institute for Regenerative Cures, University of California Davis Health Systems, 2921 Stockton Blvd. Sacramento, CA 95817, USA
| | - Kari Pollock
- Stem Cell Program & Institute for Regenerative Cures, University of California Davis Health Systems, 2921 Stockton Blvd. Sacramento, CA 95817, USA
| | - William Gruenloh
- Stem Cell Program & Institute for Regenerative Cures, University of California Davis Health Systems, 2921 Stockton Blvd. Sacramento, CA 95817, USA
| | - Geralyn Annett
- Stem Cell Program & Institute for Regenerative Cures, University of California Davis Health Systems, 2921 Stockton Blvd. Sacramento, CA 95817, USA
| | - Teresa Tempkin
- Department of Neurology, University of California Davis Health Systems, 4860 Y Street Sacramento, CA 95817, USA
| | - Vicki Wheelock
- Department of Neurology, University of California Davis Health Systems, 4860 Y Street Sacramento, CA 95817, USA
| | - Jan A Nolta
- Stem Cell Program & Institute for Regenerative Cures, University of California Davis Health Systems, 2921 Stockton Blvd. Sacramento, CA 95817, USA
| |
Collapse
|
82
|
Menstrual Blood Transplantation Therapy for Stroke and Other Neurological Disorders. Regen Med 2015. [DOI: 10.1007/978-1-4471-6542-2_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
83
|
Müller S. In silico analysis of regulatory networks underlines the role of miR-10b-5p and its target BDNF in huntington's disease. Transl Neurodegener 2014; 3:17. [PMID: 25210621 PMCID: PMC4160136 DOI: 10.1186/2047-9158-3-17] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 08/13/2014] [Indexed: 12/13/2022] Open
Abstract
Non-coding RNAs (ncRNAs) play various roles during central nervous system development. MicroRNAs (miRNAs) are a class of ncRNAs that exert their function together with argonaute proteins by post-transcriptional gene silencing of messenger RNAs (mRNAs). Several studies provide evidence for alterations in miRNA expression in patients with neurodegenerative diseases. Among these is huntington's disease (HD), a dominantly inherited fatal disorder characterized by deregulation of neuronal-specific mRNAs as well as miRNAs. Recently, next-generation sequencing (NGS) miRNA profiles from human HD and neurologically normal control brain tissues were reported. Five consistently upregulated miRNAs affect the expression of genes involved in neuronal differentiation, neurite outgrowth, cell death and survival. We re-analyzed the NGS data publicly available in array express and detected nineteen additional differentially expressed miRNAs. Subsequently, we connected these miRNAs to genes implicated in HD development and network analysis pointed to miRNA-mediated downregulation of twenty-two genes with roles in the pathogenesis as well as treatment of the disease. In silico prediction and reporter systems prove that levels of BDNF, a central node in the miRNA-mRNA regulatory network, can be post-transcriptionally controlled by upregulated miR-10b-5p and miR-30a-5p. Reduced BDNF expression is associated with neuronal dysfunction and death in HD. Moreover, the 3'UTR of CREB1 harbors a predicted binding site for these two miRNAs. CREB1 is similarly downregulated in HD and overexpression decreased susceptibility to 3-nitropropionic-induced toxicity in a cell model. In contradiction to these observations, it is presumed that miR-10b-5p upregulation in HD exerts a neuroprotective role in response to the mutation in the huntingtin gene. Therefore, the function of miR-10b-5p and especially its effect on BDNF expression in HD requires further academic research.
Collapse
Affiliation(s)
- Sören Müller
- Molecular BioSciences, University of Frankfurt, Marie-Curie-Str.9, 60439 Frankfurt a.M., Germany
| |
Collapse
|
84
|
Aleynik A, Gernavage KM, Mourad YSH, Sherman LS, Liu K, Gubenko YA, Rameshwar P. Stem cell delivery of therapies for brain disorders. Clin Transl Med 2014; 3:24. [PMID: 25097727 PMCID: PMC4106911 DOI: 10.1186/2001-1326-3-24] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Accepted: 06/30/2014] [Indexed: 02/06/2023] Open
Abstract
The blood brain barrier (BBB) poses a problem to deliver drugs for brain malignancies and neurodegenerative disorders. Stem cells such as neural stem cells (NSCs) and mesenchymal stem cells (MSCs) can be used to delivery drugs or RNA to the brain. This use of methods to bypass the hurdles of delivering drugs across the BBB is particularly important for diseases with poor prognosis such as glioblastoma multiforme (GBM). Stem cell treatment to deliver drugs to neural tumors is currently in clinical trial. This method, albeit in the early phase, could be an advantage because stem cells can cross the BBB into the brain. MSCs are particularly interesting because to date, the experimental and clinical evidence showed 'no alarm signal' with regards to safety. Additionally, MSCs do not form tumors as other more primitive stem cells such as embryonic stem cells. More importantly, MSCs showed pathotropism by migrating to sites of tissue insult. Due to the ability of MSCs to be transplanted across allogeneic barrier, drug-engineered MSCs can be available as off-the-shelf cells for rapid transplantation. This review discusses the advantages and disadvantages of stem cells to deliver prodrugs, genes and RNA to treat neural disorders.
Collapse
Affiliation(s)
| | | | | | - Lauren S Sherman
- Graduate School of Biomedical Sciences, Texas, USA
- Department of Medicine – Division of Hematology/Oncology, New Jersey Medical School, Rutgers School of Biomedical Health Science, Newark, NJ 07103, USA
| | - Katherine Liu
- Department of Anesthesiology, New Jersey Medical School, Rutgers School of Biomedical Health Science, Newark, NJ 07103, USA
| | - Yuriy A Gubenko
- Department of Anesthesiology, New Jersey Medical School, Rutgers School of Biomedical Health Science, Newark, NJ 07103, USA
| | - Pranela Rameshwar
- Department of Medicine – Division of Hematology/Oncology, New Jersey Medical School, Rutgers School of Biomedical Health Science, Newark, NJ 07103, USA
| |
Collapse
|
85
|
Use of Genetically Altered Stem Cells for the Treatment of Huntington's Disease. Brain Sci 2014; 4:202-19. [PMID: 24961705 PMCID: PMC4066244 DOI: 10.3390/brainsci4010202] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 02/18/2014] [Accepted: 02/19/2014] [Indexed: 12/14/2022] Open
Abstract
Transplantation of stem cells for the treatment of Huntington’s disease (HD) garnered much attention prior to the turn of the century. Several studies using mesenchymal stem cells (MSCs) have indicated that these cells have enormous therapeutic potential in HD and other disorders. Advantages of using MSCs for cell therapies include their ease of isolation, rapid propagation in culture, and favorable immunomodulatory profiles. However, the lack of consistent neuronal differentiation of transplanted MSCs has limited their therapeutic efficacy to slowing the progression of HD-like symptoms in animal models of HD. The use of MSCs which have been genetically altered to overexpress brain derived neurotrophic factor to enhance support of surviving cells in a rodent model of HD provides proof-of-principle that these cells may provide such prophylactic benefits. New techniques that may prove useful for cell replacement therapies in HD include the use of genetically altering fate-restricted cells to produce induced pluripotent stem cells (iPSCs). These iPSCs appear to have certain advantages over the use of embryonic stem cells, including being readily available, easy to obtain, less evidence of tumor formation, and a reduced immune response following their transplantation. Recently, transplants of iPSCs have shown to differentiate into region-specific neurons in an animal model of HD. The overall successes of using genetically altered stem cells for reducing neuropathological and behavioral deficits in rodent models of HD suggest that these approaches have considerable potential for clinical use. However, the choice of what type of genetically altered stem cell to use for transplantation is dependent on the stage of HD and whether the end-goal is preserving endogenous neurons in early-stage HD, or replacing the lost neurons in late-stage HD. This review will discuss the current state of stem cell technology for treating the different stages of HD and possible future directions for stem-cell therapy in HD.
Collapse
|
86
|
Fink KD, Crane AT, Lévêque X, Dues DJ, Huffman LD, Moore AC, Story DT, Dejonge RE, Antcliff A, Starski PA, Lu M, Lescaudron L, Rossignol J, Dunbar GL. Intrastriatal transplantation of adenovirus-generated induced pluripotent stem cells for treating neuropathological and functional deficits in a rodent model of Huntington's disease. Stem Cells Transl Med 2014; 3:620-31. [PMID: 24657963 DOI: 10.5966/sctm.2013-0151] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) show considerable promise for cell replacement therapies for Huntington's disease (HD). Our laboratory has demonstrated that tail-tip fibroblasts, reprogrammed into iPSCs via two adenoviruses, can survive and differentiate into neuronal lineages following transplantation into healthy adult rats. However, the ability of these cells to survive, differentiate, and restore function in a damaged brain is unknown. To this end, adult rats received a regimen of 3-nitropropionic acid (3-NP) to induce behavioral and neuropathological deficits that resemble HD. At 7, 21, and 42 days after the initiation of 3-NP or vehicle, the rats received intrastriatal bilateral transplantation of iPSCs. All rats that received 3-NP and vehicle treatment displayed significant motor impairment, whereas those that received iPSC transplantation after 3-NP treatment had preserved motor function. Histological analysis of the brains of these rats revealed significant decreases in optical densitometric measures in the striatum, lateral ventricle enlargement, as well as an increase in striosome size in all rats receiving 3-NP when compared with sham rats. The 3-NP-treated rats given transplants of iPSCs in the 7- or 21-day groups did not exhibit these deficits. Transplantation of iPSCs at the late-stage (42-day) time point did not protect against the 3-NP-induced neuropathology, despite preserving motor function. Transplanted iPSCs were found to survive and differentiate into region-specific neurons in the striatum of 3-NP rats, at all transplantation time points. Taken together, these results suggest that transplantation of adenovirus-generated iPSCs may provide a potential avenue for therapeutic treatment of HD.
Collapse
Affiliation(s)
- Kyle D Fink
- Field Neurosciences Institute Laboratory for Restorative Neurology, Brain Research and Integrative Neuroscience Center, Program in Neuroscience, and College of Medicine, Central Michigan University, Mount Pleasant, Michigan, USA; Faculté des Sciences et des Techniques, Faculté de Médecine, and Faculté d'Odontologie, Université de Nantes, Nantes, France; INSERM U1064, ITUN, Nantes, France; INSERM U791, Laboratoire d'Ingenierie Osteo-Articulaire et Dentaire, Nantes, France; INSERM UMR 643, Nantes, France; Field Neurosciences Institute, Saginaw, Michigan, USA; Centre Hospitalier-Universitaire Hotel Dieu de Nantes, Nantes, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Wyse RD, Dunbar GL, Rossignol J. Use of genetically modified mesenchymal stem cells to treat neurodegenerative diseases. Int J Mol Sci 2014; 15:1719-45. [PMID: 24463293 PMCID: PMC3958818 DOI: 10.3390/ijms15021719] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 12/18/2013] [Accepted: 01/14/2014] [Indexed: 01/01/2023] Open
Abstract
The transplantation of mesenchymal stem cells (MSCs) for treating neurodegenerative disorders has received growing attention recently because these cells are readily available, easily expanded in culture, and when transplanted, survive for relatively long periods of time. Given that such transplants have been shown to be safe in a variety of applications, in addition to recent findings that MSCs have useful immunomodulatory and chemotactic properties, the use of these cells as vehicles for delivering or producing beneficial proteins for therapeutic purposes has been the focus of several labs. In our lab, the use of genetic modified MSCs to release neurotrophic factors for the treatment of neurodegenerative diseases is of particular interest. Specifically, glial cell-derived neurotrophic factor (GDNF), nerve growth factor (NGF), and brain derived neurotrophic factor (BDNF) have been recognized as therapeutic trophic factors for Parkinson's, Alzheimer's and Huntington's diseases, respectively. The aim of this literature review is to provide insights into: (1) the inherent properties of MSCs as a platform for neurotrophic factor delivery; (2) the molecular tools available for genetic manipulation of MSCs; (3) the rationale for utilizing various neurotrophic factors for particular neurodegenerative diseases; and (4) the clinical challenges of utilizing genetically modified MSCs.
Collapse
Affiliation(s)
- Robert D Wyse
- Field Neurosciences Institute Laboratory for Restorative Neurology, Brain Research and Integrative Neuroscience Center, Program in Neuroscience, Central Michigan University, Mount Pleasant, MI 48859, USA.
| | - Gary L Dunbar
- Field Neurosciences Institute Laboratory for Restorative Neurology, Brain Research and Integrative Neuroscience Center, Program in Neuroscience, Central Michigan University, Mount Pleasant, MI 48859, USA.
| | - Julien Rossignol
- Field Neurosciences Institute Laboratory for Restorative Neurology, Brain Research and Integrative Neuroscience Center, Program in Neuroscience, Central Michigan University, Mount Pleasant, MI 48859, USA.
| |
Collapse
|
88
|
Aguilar E, Cobo Pulido M, Martin F. Gene-modified mesenchymal stromal cells: A VIP experience. Inflamm Regen 2014. [DOI: 10.2492/inflammregen.34.176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
89
|
Fink KD, Rossignol J, Crane AT, Davis KK, Bombard MC, Bavar AM, Clerc S, Lowrance SA, Song C, Lescaudron L, Dunbar GL. Transplantation of umbilical cord-derived mesenchymal stem cells into the striata of R6/2 mice: behavioral and neuropathological analysis. Stem Cell Res Ther 2013; 4:130. [PMID: 24456799 PMCID: PMC3854759 DOI: 10.1186/scrt341] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 10/09/2013] [Indexed: 12/31/2022] Open
Abstract
Introduction Huntington’s disease (HD) is an autosomal dominant disorder caused by an expanded CAG repeat on the short arm of chromosome 4 resulting in cognitive decline, motor dysfunction, and death, typically occurring 15 to 20 years after the onset of motor symptoms. Neuropathologically, HD is characterized by a specific loss of medium spiny neurons in the caudate and the putamen, as well as subsequent neuronal loss in the cerebral cortex. The transgenic R6/2 mouse model of HD carries the N-terminal fragment of the human HD gene (145 to 155 repeats) and rapidly develops some of the behavioral characteristics that are analogous to the human form of the disease. Mesenchymal stem cells (MSCs) have shown the ability to slow the onset of behavioral and neuropathological deficits following intrastriatal transplantation in rodent models of HD. Use of MSCs derived from umbilical cord (UC) offers an attractive strategy for transplantation as these cells are isolated from a noncontroversial and inexhaustible source and can be harvested at a low cost. Because UC MSCs represent an intermediate link between adult and embryonic tissue, they may hold more pluripotent properties than adult stem cells derived from other sources. Methods Mesenchymal stem cells, isolated from the UC of day 15 gestation pups, were transplanted intrastriatally into 5-week-old R6/2 mice at either a low-passage (3 to 8) or high-passage (40 to 50). Mice were tested behaviorally for 6 weeks using the rotarod task, the Morris water maze, and the limb-clasping response. Following behavioral testing, tissue sections were analyzed for UC MSC survival, the immune response to the transplanted cells, and neuropathological changes. Results Following transplantation of UC MSCs, R6/2 mice did not display a reduction in motor deficits but there appeared to be transient sparing in a spatial memory task when compared to untreated R6/2 mice. However, R6/2 mice receiving either low- or high-passage UC MSCs displayed significantly less neuropathological deficits, relative to untreated R6/2 mice. Conclusions The results from this study demonstrate that UC MSCs hold promise for reducing the neuropathological deficits observed in the R6/2 rodent model of HD.
Collapse
|
90
|
Li Q, Liu Y, Chu Z, Chen J, Dai F, Zhu X, Hu A, Yun C. Brain-derived neurotrophic factor expression in dorsal root ganglia of a lumbar spinal stenosis model in rats. Mol Med Rep 2013; 8:1836-44. [PMID: 24127005 DOI: 10.3892/mmr.2013.1723] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 09/26/2013] [Indexed: 11/06/2022] Open
Abstract
This study aimed to investigate the expression of brain-derived neurotrophic factor (BDNF) in dorsal root ganglia (DRG) of a rat model of lumbar spinal stenosis (LSS). Adult male rats were divided into the operation and sham operation groups. The operation group was comprised of the rat models of LSS. Walking distance and BDNF expression levels in DRG were measured in the two groups at different time points. The total BDNF protein levels and positive cell mean optical density (MOD) values in the operation group were significantly higher at each time point compared with that of the sham operation and preoperative control groups (P<0.05). The total BDNF protein levels and MOD values following sport in the operation group were significantly higher compared with those prior to sport (P<0.05). In the sham operation group, BDNF protein levels and MOD values before and after sport at each time point showed no significant differences than those of the operation group (P>0.05). Moreover, BDNF protein levels and MOD values in the operation group indicated a negative correlation with walking distance. The present study demonstrated that the expression of BDNF in rat models of LSS increased with time and was associated with a decrease in walking distance. BDNF was therefore important for the process of intermittent claudication caused by LSS.
Collapse
Affiliation(s)
- Qinliang Li
- Department of Spine, The Affiliated Lianyungang Hospital of Xuzhou Medical College, Lianyungang, Jiangsu 222000, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
91
|
Binan L, Ajji A, De Crescenzo G, Jolicoeur M. Approaches for Neural Tissue Regeneration. Stem Cell Rev Rep 2013; 10:44-59. [DOI: 10.1007/s12015-013-9474-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
92
|
Kim SU, Lee HJ, Kim YB. Neural stem cell-based treatment for neurodegenerative diseases. Neuropathology 2013; 33:491-504. [PMID: 23384285 DOI: 10.1111/neup.12020] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 12/27/2012] [Accepted: 12/28/2012] [Indexed: 12/11/2022]
Abstract
Human neurodegenerative diseases such as Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS) and Alzheimer's disease (AD) are caused by a loss of neurons and glia in the brain or spinal cord. Neurons and glial cells have successfully been generated from stem cells such as embryonic stem cells (ESCs), mesenchymal stem cells (MSCs) and neural stem cells (NSCs), and stem cell-based cell therapies for neurodegenerative diseases have been developed. A recent advance in generation of a new class of pluripotent stem cells, induced pluripotent stem cells (iPSCs), derived from patients' own skin fibroblasts, opens doors for a totally new field of personalized medicine. Transplantation of NSCs, neurons or glia generated from stem cells in animal models of neurodegenerative diseases, including PD, HD, ALS and AD, demonstrates clinical improvement and also life extension of these animals. Additional therapeutic benefits in these animals can be provided by stem cell-mediated gene transfer of therapeutic genes such as neurotrophic factors and enzymes. Although further research is still needed, cell and gene therapy based on stem cells, particularly using neurons and glia derived from iPSCs, ESCs or NSCs, will become a routine treatment for patients suffering from neurodegenerative diseases and also stroke and spinal cord injury.
Collapse
Affiliation(s)
- Seung U Kim
- Medical Research Institute, Chung-Ang University College of Medicine, Seoul, Korea; Division of Neurology, Department of Medicine, UBC Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | |
Collapse
|
93
|
Yoo J, Kim HS, Hwang DY. Stem cells as promising therapeutic options for neurological disorders. J Cell Biochem 2013; 114:743-53. [PMID: 23097262 DOI: 10.1002/jcb.24427] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 10/12/2012] [Indexed: 12/13/2022]
Abstract
Due to the limitations of pharmacological and other current therapeutic strategies, stem cell therapies have emerged as promising options for treating many incurable neurologic diseases. A variety of stem cells including pluripotent stem cells (i.e., embryonic stem cells and induced pluripotent stem cells) and multipotent adult stem cells (i.e., fetal brain tissue, neural stem cells, and mesenchymal stem cells from various sources) have been explored as therapeutic options for treating many neurologic diseases, and it is becoming obvious that each type of stem cell has pros and cons as a source for cell therapy. Wise selection of stem cells with regard to the nature and status of neurologic dysfunctions is required to achieve optimal therapeutic efficacy. To this aim, the stem cell-mediated therapeutic efforts on four major neurological diseases, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and stroke, will be introduced, and current problems and future directions will be discussed.
Collapse
Affiliation(s)
- Jongman Yoo
- Department of Biological Science, CHA University, Kyeonggido, Korea
| | | | | |
Collapse
|
94
|
Maucksch C, Vazey EM, Gordon RJ, Connor B. Stem cell-based therapy for Huntington's disease. J Cell Biochem 2013; 114:754-63. [PMID: 23097329 DOI: 10.1002/jcb.24432] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 10/15/2012] [Indexed: 12/23/2022]
Abstract
Huntington's disease (HD) is a late-onset neurodegenerative disease characterized by a progressive loss of medium spiny neurons in the basal ganglia. The development of stem cell-based therapies for HD aims to replace lost neurons and/or to prevent cell death. This review will discuss pre-clinical studies which have utilized stem or progenitor cells for transplantation therapy using HD animal models. In several studies, neural stem and progenitor cells used as allotransplants and xenografts have been shown to be capable of surviving transplantation and differentiating into mature GABAergic neurons, resulting in behavioral improvements. Beneficial effects have also been reported for transplantation of stem cells derived from non-neural tissue, for example, mesenchymal- and adipose-derived stem cells, which have mainly been attributed to their secretion of growth and neurotrophic factors. Finally, we review studies using stem cells genetically engineered to over-express defined neurotrophic factors. While these studies prove the potential of stem cells for transplantation therapy in HD, it also becomes clear that technical and ethical issues regarding the availability of stem cells must be solved before human trials can be conducted.
Collapse
Affiliation(s)
- Christof Maucksch
- Department of Pharmacology and Clinical Pharmacology, Centre for Brain Research, School of Medical Science, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | | | | | | |
Collapse
|
95
|
Mesenchymal stem cells for treatment of neurological disorders: a paracrine effect. Tissue Eng Regen Med 2013. [DOI: 10.1007/s13770-013-1087-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
96
|
De Vocht N, Praet J, Reekmans K, Le Blon D, Hoornaert C, Daans J, Berneman Z, Van der Linden A, Ponsaerts P. Tackling the physiological barriers for successful mesenchymal stem cell transplantation into the central nervous system. Stem Cell Res Ther 2013; 4:101. [PMID: 23998480 PMCID: PMC3854758 DOI: 10.1186/scrt312] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Over the past decade a lot of research has been performed towards the therapeutic use of mesenchymal stem cells (MSCs) in neurodegenerative and neuroinflammatory diseases. MSCs have shown to be beneficial in different preclinical studies of central nervous system (CNS) disorders due to their immunomodulatory properties and their capacity to secrete various growth factors. Nevertheless, most of the transplanted cells die within the first hours after transplantation and induce a neuroinflammatory response. In order to increase the efficacy of MSC transplantation, it is thus imperative to completely characterise the mechanisms mediating neuroinflammation and cell death following MSC transplantation into the CNS. Consequently, different components of these cell death- and neuroinflammation-inducing pathways can be targeted in an attempt to improve the therapeutic potential of MSCs for CNS disorders.
Collapse
|
97
|
Increased Body Weight of the BAC HD Transgenic Mouse Model of Huntington's Disease Accounts for Some but Not All of the Observed HD-like Motor Deficits. PLOS CURRENTS 2013; 5. [PMID: 24042107 PMCID: PMC3770835 DOI: 10.1371/currents.hd.0ab4f3645aff523c56ecc8ccbe41a198] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
The genome of the Bacterial Artificial Chromosome (BAC) transgenic mouse model of Huntington’s Disease (BAC HD) contains the 170 kb human HTT locus modified by the addition of exon 1 with 97 mixed CAA-CAG repeats. BAC HD mice present robust behavioral deficits in both the open field and the accelerating rotarod tests, two standard behavioral assays of motor function. BAC HD mice, however, also typically present significantly increased body weights relative to wildtype littermate controls (WT) which potentially confounds the interpretation of any motor deficits associated directly with the effects of mutant huntingtin. In order to evaluate this possible confound of body weight, we directly compared the performance of BAC HD and WT female mice under food restricted versus free feeding conditions in both the open field and rotarod tasks to test the hypothesis that some of the motor deficits observed in this HTT-transgenic mouse line results solely from increased body weight. Our results suggest that the rotarod deficit exhibited by BAC HD mice is modulated by both body weight and non-body weight factors resulting from overexpression of full length mutant Htt. When body weights of WT and BAC HD transgenic mice were normalized using restricted feeding, the deficits exhibited by BAC HD mice on the rotarod task were less marked, but were still significant. Since the rotarod deficit between WT and BAC HD mice is attenuated when body weight is normalized by food restriction, utilization of this task in BAC HD mice during pre-clinical evaluation must be powered accordingly and results carefully considered as therapeutic benefit can result from decreased overall body weight and or motoric improvement that may not be related to body mass. Furthermore, after controlling for body weight differences, the hypoactive phenotype displayed by ad libitum fed BAC HD mice in the open field assay was not observed in the BAC HD mice undergoing food restriction. These findings suggest that assessment of spontaneous locomotor activity, as measured in the open field test, may not be the appropriate behavioral endpoint to evaluate the BAC HD mouse during preclinical evaluation since it appears that the apparent hypoactive phenotype in this model is driven primarily by body weight differences.
Collapse
|
98
|
Paul G, Anisimov SV. The secretome of mesenchymal stem cells: potential implications for neuroregeneration. Biochimie 2013; 95:2246-56. [PMID: 23871834 DOI: 10.1016/j.biochi.2013.07.013] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Accepted: 07/10/2013] [Indexed: 12/14/2022]
Abstract
Mesenchymal stem cells have shown regenerative properties in many tissues. This feature had originally been ascribed to their multipotency and thus their ability to differentiate into tissue-specific cells. However, many researchers consider the secretome of mesenchymal stem cells the most important player in the observed reparative effects of these cells. In this review, we specifically focus on the potential neuroregenerative effect of mesenchymal stem cells, summarize several possible mechanisms of neuroregeneration and list key factors mediating this effect. We illustrate examples of mesenchymal stem cell treatment in central nervous system disorders including stroke, neurodegenerative disorders (such as Parkinson's disease, Huntington's disease, multiple system atrophy and cerebellar ataxia) and inflammatory disease (such as multiple sclerosis). We specifically highlight studies where mesenchymal stem cells have entered clinical trials.
Collapse
Affiliation(s)
- Gesine Paul
- Translational Neurology Group, Division of Neurology, Department of Clinical Sciences, Lund University, Lund, Sweden; Department of Neurology, Scania University Hospital, Lund, Sweden.
| | | |
Collapse
|
99
|
Drago D, Cossetti C, Iraci N, Gaude E, Musco G, Bachi A, Pluchino S. The stem cell secretome and its role in brain repair. Biochimie 2013; 95:2271-85. [PMID: 23827856 PMCID: PMC4061727 DOI: 10.1016/j.biochi.2013.06.020] [Citation(s) in RCA: 240] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 06/19/2013] [Indexed: 12/16/2022]
Abstract
Compelling evidence exists that non-haematopoietic stem cells, including mesenchymal (MSCs) and neural/progenitor stem cells (NPCs), exert a substantial beneficial and therapeutic effect after transplantation in experimental central nervous system (CNS) disease models through the secretion of immune modulatory or neurotrophic paracrine factors. This paracrine hypothesis has inspired an alternative outlook on the use of stem cells in regenerative neurology. In this paradigm, significant repair of the injured brain may be achieved by injecting the biologics secreted by stem cells (secretome), rather than implanting stem cells themselves for direct cell replacement. The stem cell secretome (SCS) includes cytokines, chemokines and growth factors, and has gained increasing attention in recent years because of its multiple implications for the repair, restoration or regeneration of injured tissues. Thanks to recent improvements in SCS profiling and manipulation, investigators are now inspired to harness the SCS as a novel alternative therapeutic option that might ensure more efficient outcomes than current stem cell-based therapies for CNS repair. This review discusses the most recent identification of MSC- and NPC-secreted factors, including those that are trafficked within extracellular membrane vesicles (EVs), and reflects on their potential effects on brain repair. It also examines some of the most convincing advances in molecular profiling that have enabled mapping of the SCS.
Collapse
Affiliation(s)
- Denise Drago
- CNS Repair Unit, Institute of Experimental Neurology, Division of Neurosciences, San Raffaele Scientific Institute, 20132 Milan, Italy; Biomolecular Mass Spectrometry Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milan, Italy.
| | | | | | | | | | | | | |
Collapse
|
100
|
Role of BDNF in Central Motor Structures and Motor Diseases. Mol Neurobiol 2013; 48:783-93. [DOI: 10.1007/s12035-013-8466-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 04/22/2013] [Indexed: 12/29/2022]
|