201
|
Prasad A, Manivannan J, Loong DTB, Chua SM, Gharibani PM, All AH. A review of induced pluripotent stem cell, direct conversion by trans-differentiation, direct reprogramming and oligodendrocyte differentiation. Regen Med 2016; 11:181-91. [DOI: 10.2217/rme.16.5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Rapid progress in the field of stem cell therapy and cellular reprogramming provides convincing evidence of its feasibility in treating a wide range of pathologies through autologous cell replacement therapy. This review article describes in detail on three widely used approaches of somatic cell reprogramming: induced pluripotent stem cells, direct conversion and direct reprogramming, in the context of demyelination in the CNS. The potential limitations of each reprogramming technique are reviewed along with their distinct molecular approach to reprogramming. This is followed by an analysis on the scopes and challenges of its translational applications in deriving oligodendrocyte progenitor cells and oligodendrocytes for cell replacement treatment of demyelinating conditions in the CNS.
Collapse
Affiliation(s)
- Ankshita Prasad
- Department of Biomedical Engineering, National University of Singapore, Singapore
| | - Janani Manivannan
- Department of Orthopedic Surgery, National University of Singapore, Singapore
| | - Daniel TB Loong
- Department of Biomedical Engineering, National University of Singapore, Singapore
| | - Soo M Chua
- Department of Biomedical Engineering, National University of Singapore, Singapore
| | - Payam M Gharibani
- Singapore Institute of Neurotechnology, National University of Singapore, Singapore
| | - Angelo H All
- Department of Biomedical Engineering, National University of Singapore, Singapore
- Department of Orthopedic Surgery, National University of Singapore, Singapore
- Singapore Institute of Neurotechnology, National University of Singapore, Singapore
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Medicine, Division of Nuerology, National University of Singapore, Singapore
| |
Collapse
|
202
|
Yao Z, Yang C, Sun G. [Research progress of induced pluripotent stem cells in treatment of muscle atrophy]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2016; 45:147-51. [PMID: 27273988 PMCID: PMC10396904 DOI: 10.3785/j.issn.1008-9292.2016.03.07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 01/24/2016] [Indexed: 06/06/2023]
Abstract
Muscle atrophy caused by nerve injury is a common and difficult clinical problem. The development of stem cell researches has opened up a new way for the treatment of nerve injury-induced muscle atrophy. The induced pluripotent stem cells(iPSCs)can differentiate into various types of cells and have more advantages than embryonic stem cells (ESCs). After being transplanted into the damaged area, iPSCs are guided by neurogenic signals to the lesion sites, to repair the damaged nerve, promote generation of axon myelination, rebuild neural circuits and restore physiological function. Meanwhile, iPSCs can also differentiate into muscle cells and promote muscle tissue regeneration. Therefore, it would be possible to attenuate muscle atrophy caused by nerve injury with iPSCs treatment.
Collapse
Affiliation(s)
- Zhongkai Yao
- Department of Trauma Surgery, East Hospital Affiliated to Tongji University, Shanghai 200120, China
| | - Chensong Yang
- Department of Trauma Surgery, East Hospital Affiliated to Tongji University, Shanghai 200120, China
| | - Guixin Sun
- Department of Trauma Surgery, East Hospital Affiliated to Tongji University, Shanghai 200120, China.
| |
Collapse
|
203
|
Zhu B, Caldwell M, Song B. Development of stem cell-based therapies for Parkinson's disease. Int J Neurosci 2016; 126:955-62. [DOI: 10.3109/00207454.2016.1148034] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
204
|
Petersen GF, Strappe PM. Generation of diverse neural cell types through direct conversion. World J Stem Cells 2016; 8:32-46. [PMID: 26981169 PMCID: PMC4766249 DOI: 10.4252/wjsc.v8.i2.32] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 11/18/2015] [Accepted: 01/04/2016] [Indexed: 02/06/2023] Open
Abstract
A characteristic of neurological disorders is the loss of critical populations of cells that the body is unable to replace, thus there has been much interest in identifying methods of generating clinically relevant numbers of cells to replace those that have been damaged or lost. The process of neural direct conversion, in which cells of one lineage are converted into cells of a neural lineage without first inducing pluripotency, shows great potential, with evidence of the generation of a range of functional neural cell types both in vitro and in vivo, through viral and non-viral delivery of exogenous factors, as well as chemical induction methods. Induced neural cells have been proposed as an attractive alternative to neural cells derived from embryonic or induced pluripotent stem cells, with prospective roles in the investigation of neurological disorders, including neurodegenerative disease modelling, drug screening, and cellular replacement for regenerative medicine applications, however further investigations into improving the efficacy and safety of these methods need to be performed before neural direct conversion becomes a clinically viable option. In this review, we describe the generation of diverse neural cell types via direct conversion of somatic cells, with comparison against stem cell-based approaches, as well as discussion of their potential research and clinical applications.
Collapse
|
205
|
Abstract
The past 10 years have seen great advances in our ability to manipulate cell fate, including the induction of pluripotency in vitro to generate induced pluripotent stem cells (iPSCs). This process proved to be remarkably simple from a technical perspective, only needing the host cell and a defined cocktail of transcription factors, with four factors - octamer-binding protein 3/4 (OCT3/4), SOX2, Krüppel-like factor 4 (KLF4) and MYC (collectively referred to as OSKM) - initially used. The mechanisms underlying transcription factor-mediated reprogramming are still poorly understood; however, several mechanistic insights have recently been obtained. Recent years have also brought significant progress in increasing the efficiency of this technique, making it more amenable to applications in the fields of regenerative medicine, disease modelling and drug discovery.
Collapse
|
206
|
TAKAGI Y. History of Neural Stem Cell Research and Its Clinical Application. Neurol Med Chir (Tokyo) 2016; 56:110-24. [PMID: 26888043 PMCID: PMC4791305 DOI: 10.2176/nmc.ra.2015-0340] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 01/15/2016] [Indexed: 12/11/2022] Open
Abstract
"Once development was ended…in the adult centers, the nerve paths are something fixed and immutable. Everything may die, nothing may be regenerated," wrote Santiago Ramón y Cajal, a Spanish neuroanatomist and Nobel Prize winner and the father of modern neuroscience. This statement was the central dogma in neuroscience for a long time. However, in the 1960s, neural stem cells (NSCs) were discovered. Since then, our knowledge about NSCs has continued to grow. This review focuses on our current knowledge about NSCs and their surrounding microenvironment. In addition, the clinical application of NSCs for the treatment of various central nervous system diseases is also summarized.
Collapse
Affiliation(s)
- Yasushi TAKAGI
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Sakyo, Kyoto
| |
Collapse
|
207
|
Factor-Reduced Human Induced Pluripotent Stem Cells Efficiently Differentiate into Neurons Independent of the Number of Reprogramming Factors. Stem Cells Int 2016; 2016:4736159. [PMID: 26977154 PMCID: PMC4763001 DOI: 10.1155/2016/4736159] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 12/29/2015] [Accepted: 01/06/2016] [Indexed: 02/04/2023] Open
Abstract
Reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) by overexpression of the transcription factors OCT4, SOX2, KLF4, and c-Myc holds great promise for the development of personalized cell replacement therapies. In an attempt to minimize the risk of chromosomal disruption and to simplify reprogramming, several studies demonstrated that a reduced set of reprogramming factors is sufficient to generate iPSC. We recently showed that a reduction of reprogramming factors in murine cells not only reduces reprogramming efficiency but also may worsen subsequent differentiation. To prove whether this is also true for human cells, we compared the efficiency of neuronal differentiation of iPSC generated from fetal human neural stem cells with either one (OCT4; hiPSC1F-NSC) or two (OCT4, KLF4; hiPSC2F-NSC) reprogramming factors with iPSC produced from human fibroblasts using three (hiPSC3F-FIB) or four reprogramming factors (hiPSC4F-FIB). After four weeks of coculture with PA6 stromal cells, neuronal differentiation of hiPSC1F-NSC and hiPSC2F-NSC was as efficient as iPSC3F-FIB or iPSC4F-FIB. We conclude that a reduction of reprogramming factors in human cells does reduce reprogramming efficiency but does not alter subsequent differentiation into neural lineages. This is of importance for the development of future application of iPSC in cell replacement therapies.
Collapse
|
208
|
Terraf P, Babaloo H, Kouhsari SM. Directed Differentiation of Dopamine-Secreting Cells from Nurr1/GPX1 Expressing Murine Embryonic Stem Cells Cultured on Matrigel-Coated PCL Scaffolds. Mol Neurobiol 2016; 54:1119-1128. [DOI: 10.1007/s12035-016-9726-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 01/13/2016] [Indexed: 12/01/2022]
|
209
|
Ali SA, Yin N, Rehman A, Justilien V. Parkinson Disease-Mediated Gastrointestinal Disorders and Rational for Combinatorial Therapies. Med Sci (Basel) 2016; 4:medsci4010001. [PMID: 29083365 PMCID: PMC5635767 DOI: 10.3390/medsci4010001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 12/24/2015] [Accepted: 01/13/2016] [Indexed: 12/29/2022] Open
Abstract
A gradual loss of dopamine-producing nerve cells gives rise to a common neurodegenerative Parkinson’s disease (PD). This disease causes a neurotransmitter imbalance in the brain and initiates a cascade of complications in the rest of the body that appears as distressing symptoms which include gait problems, tremor, gastrointestinal (GI) disorders and cognitive decline. To aid dopamine deficiency, treatment in PD patients includes oral medications, in addition to other methods such as deep brain stimulation and surgical lesioning. Scientists are extensively studying molecular and signaling mechanisms, particularly those involving phenotypic transcription factors and their co-regulatory proteins that are associated with neuronal stem cell (SC) fate determination, maintenance and disease state, and their role in the pathogenesis of PD. Advancement in scientific research and “personalized medicine” to augment current therapeutic intervention and minimize the side effects of chemotherapy may lead to the development of more effective therapeutic strategies in the near future. This review focuses on PD and associated GI complications and summarizes the current therapeutic modalities that include stem cell studies and combinatorial drug treatment.
Collapse
Affiliation(s)
- Syed A Ali
- Department of Cancer Biology, Mayo Clinic Cancer Center, Jacksonville, FL 32224, USA.
| | - Ning Yin
- Department of Cancer Biology, Mayo Clinic Cancer Center, Jacksonville, FL 32224, USA.
| | - Arkam Rehman
- Department of Pain Medicine, Baptist Medical Center, Jacksonville, FL 32258, USA.
| | - Verline Justilien
- Department of Cancer Biology, Mayo Clinic Cancer Center, Jacksonville, FL 32224, USA.
| |
Collapse
|
210
|
Mitochondrial resetting and metabolic reprogramming in induced pluripotent stem cells and mitochondrial disease modeling. Biochim Biophys Acta Gen Subj 2016; 1860:686-93. [PMID: 26779594 DOI: 10.1016/j.bbagen.2016.01.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 01/13/2016] [Accepted: 01/14/2016] [Indexed: 01/19/2023]
Abstract
BACKGROUND Nuclear reprogramming with pluripotency factors enables somatic cells to gain the properties of embryonic stem cells. Mitochondrial resetting and metabolic reprogramming are suggested to be key early events in the induction of human skin fibroblasts to induced pluripotent stem cells (iPSCs). SCOPE OF REVIEW We review recent advances in the study of the molecular basis for mitochondrial resetting and metabolic reprogramming in the regulation of the formation of iPSCs. In particular, the recent progress in using iPSCs for mitochondrial disease modeling was discussed. MAJOR CONCLUSIONS iPSCs rely on glycolysis rather than oxidative phosphorylation as a major supply of energy. Mitochondrial resetting and metabolic reprogramming thus play crucial roles in the process of generation of iPSCs from somatic cells. GENERAL SIGNIFICANCE Neurons, myocytes, and cardiomyocytes are cells containing abundant mitochondria in the human body, which can be differentiated from iPSCs or trans-differentiated from fibroblasts. Generating these cells from iPSCs derived from skin fibroblasts of patients with mitochondrial diseases or by trans-differentiation with cell-specific transcription factors will provide valuable insights into the role of mitochondrial DNA heteroplasmy in mitochondrial disease modeling and serves as a novel platform for screening of drugs to treat patients with mitochondrial diseases.
Collapse
|
211
|
Wnt/β-catenin signaling plays an ever-expanding role in stem cell self-renewal, tumorigenesis and cancer chemoresistance. Genes Dis 2016; 3:11-40. [PMID: 27077077 PMCID: PMC4827448 DOI: 10.1016/j.gendis.2015.12.004] [Citation(s) in RCA: 208] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Wnt signaling transduces evolutionarily conserved pathways which play important roles in initiating and regulating a diverse range of cellular activities, including cell proliferation, calcium homeostasis, and cell polarity. The role of Wnt signaling in controlling cell proliferation and stem cell self-renewal is primarily carried out through the canonical pathway, which is the best-characterized the multiple Wnt signaling branches. The past 10 years has seen a rapid expansion in our understanding of the complexity of this pathway, as many new components of Wnt signaling have been identified and linked to signaling regulation, stem cell functions, and adult tissue homeostasis. Additionally, a substantial body of evidence links Wnt signaling to tumorigenesis of cancer types and implicates it in the development of cancer drug resistance. Thus, a better understanding of the mechanisms by which dysregulation of Wnt signaling precedes the development and progression of human cancer may hasten the development of pathway inhibitors to augment current therapy. This review summarizes and synthesizes our current knowledge of the canonical Wnt pathway in development and disease. We begin with an overview of the components of the canonical Wnt signaling pathway and delve into the role this pathway has been shown to play in stemness, tumorigenesis, and cancer drug resistance. Ultimately, we hope to present an organized collection of evidence implicating Wnt signaling in tumorigenesis and chemoresistance to facilitate the pursuit of Wnt pathway modulators that may improve outcomes of cancers in which Wnt signaling contributes to aggressive disease and/or treatment resistance.
Collapse
|
212
|
Xu X, Huang J, Li J, Liu L, Han C, Shen Y, Zhang G, Jiang H, Lin Z, Xiong N, Wang T. Induced pluripotent stem cells and Parkinson's disease: modelling and treatment. Cell Prolif 2016; 49:14-26. [PMID: 26748765 DOI: 10.1111/cpr.12229] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 08/23/2015] [Indexed: 02/06/2023] Open
Abstract
Many neurodegenerative disorders, such as Parkinson's disease (PD), are characterized by progressive neuronal loss in different regions of the central nervous system, contributing to brain dysfunction in the relevant patients. Stem cell therapy holds great promise for PD patients, including with foetal ventral mesencephalic cells, human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs). Moreover, stem cells can be used to model neurodegenerative diseases in order to screen potential medication and explore their mechanisms of disease. However, related ethical issues, immunological rejection and lack of canonical grafting protocols limit common clinical use of stem cells. iPSCs, derived from reprogrammed somatic cells, provide new hope for cell replacement therapy. In this review, recent development in stem cell treatment for PD, using hiPSCs, as well as the potential value of hiPSCs in modelling for PD, have been summarized for application of iPSCs technology to clinical translation for PD treatment.
Collapse
Affiliation(s)
- Xiaoyun Xu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jinsha Huang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jie Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ling Liu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chao Han
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yan Shen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Guoxin Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Haiyang Jiang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhicheng Lin
- Department of Psychiatry, Harvard Medical School, Division of Alcohol and Drug Abuse, Mailman Neuroscience Research Center, McLean Hospital, Belmont, MA, USA
| | - Nian Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
213
|
Abstract
Tissue engineering of Schwann cells (SCs) can serve a number of purposes, such as in vitro SC-related disease modeling, treatment of peripheral nerve diseases or peripheral nerve injury, and, potentially, treatment of CNS diseases. SCs can be generated from autologous stem cells in vitro by recapitulating the various stages of in vivo neural crest formation and SC differentiation. In this review, we survey the cellular and molecular mechanisms underlying these in vivo processes. We then focus on the current in vitro strategies for generating SCs from two sources of pluripotent stem cells, namely embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). Different methods for SC engineering from ESCs and iPSCs are reviewed and suggestions are proposed for optimizing the existing protocols. Potential safety issues regarding the clinical application of iPSC-derived SCs are discussed as well. Lastly, we will address future aspects of SC engineering.
Collapse
|
214
|
The Application of Human iPSCs in Neurological Diseases: From Bench to Bedside. Stem Cells Int 2016; 2016:6484713. [PMID: 26880979 PMCID: PMC4736583 DOI: 10.1155/2016/6484713] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 11/23/2015] [Accepted: 11/26/2015] [Indexed: 12/20/2022] Open
Abstract
In principle, induced pluripotent stem cells (iPSCs) are generated from somatic cells by reprogramming and gaining the capacity to self-renew indefinitely as well as the ability to differentiate into cells of different lineages. Human iPSCs have absolute advantages over human embryonic stem cells (ESCs) and animal models in disease modeling, drug screening, and cell replacement therapy. Since Takahashi and Yamanaka first described in 2007 that iPSCs can be generated from human adult somatic cells by retroviral transduction of the four transcription factors, Oct3/4, Sox2, Klf4, and c-Myc, disease specific iPSC lines have sprung up worldwide like bamboo shoots after a spring rain, making iPSC one of the hottest and fastest moving topics in modern science. The craze for iPSCs has spread throughout main branches of clinical medicine, covering neurology, hematology, cardiology, endocrinology, hepatology, ophthalmology, and so on. Here in this paper, we will focus on the clinical application of human iPSCs in disease modeling, drug screening, and cell replacement therapy for neurological diseases.
Collapse
|
215
|
Karanfil I, Bagci-Onder T. Derivation of Neural Stem Cells from Mouse Induced Pluripotent Stem Cells. Methods Mol Biol 2016; 1357:329-338. [PMID: 25863785 DOI: 10.1007/7651_2015_227] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Neural stem cells (NSCs) derived from induced pluripotent stem cells offer therapeutic tools for neurodegenerative diseases. This review focuses on embryoid body (EB)-mediated stem cell culture techniques used to derive NSCs from mouse induced pluripotent stem cells (iPSCs). Generation of healthy and stable NSCs from iPSCs heavily depends on standardized in vitro cell culture systems that mimic the embryonic environments utilized during neural development. Specifically, the neural induction and expansion methods after EB formation are described in this review.
Collapse
Affiliation(s)
- Işıl Karanfil
- Brain Cancer Research and Therapy Laboratory, Koç University School of Medicine, Rumelifeneri Yolu, Sarıyer, Istanbul, 34450, Turkey
| | - Tugba Bagci-Onder
- Brain Cancer Research and Therapy Laboratory, Koç University School of Medicine, Rumelifeneri Yolu, Sarıyer, Istanbul, 34450, Turkey.
| |
Collapse
|
216
|
Neuroprotective Transcription Factors in Animal Models of Parkinson Disease. Neural Plast 2015; 2016:6097107. [PMID: 26881122 PMCID: PMC4736191 DOI: 10.1155/2016/6097107] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 07/10/2015] [Accepted: 07/14/2015] [Indexed: 11/28/2022] Open
Abstract
A number of transcription factors, including En1/2, Foxa1/2, Lmx1a/b, Nurr1, Otx2, and Pitx3, with key roles in midbrain dopaminergic (mDA) neuron development, also regulate adult mDA neuron survival and physiology. Mouse models with targeted disruption of some of these genes display several features reminiscent of Parkinson disease (PD), in particular the selective and progressive loss of mDA neurons in the substantia nigra pars compacta (SNpc). The characterization of these animal models has provided valuable insights into various mechanisms of PD pathogenesis. Therefore, the dissection of the mechanisms and survival signalling pathways engaged by these transcription factors to protect mDA neuron from degeneration can suggest novel therapeutic strategies. The work on En1/2-mediated neuroprotection also highlights the potential of protein transduction technology for neuroprotective approaches in PD.
Collapse
|
217
|
Epigenetic Research of Neurodegenerative Disorders Using Patient iPSC-Based Models. Stem Cells Int 2015; 2016:9464591. [PMID: 26697081 PMCID: PMC4677257 DOI: 10.1155/2016/9464591] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 06/18/2015] [Indexed: 01/15/2023] Open
Abstract
Epigenetic mechanisms play a role in human disease but their involvement in pathologies from the central nervous system has been hampered by the complexity of the brain together with its unique cellular architecture and diversity. Until recently, disease targeted neural types were only available as postmortem materials after many years of disease evolution. Current in vitro systems of induced pluripotent stem cells (iPSCs) generated by cell reprogramming of somatic cells from patients have provided valuable disease models recapitulating key pathological molecular events. Yet whether cell reprogramming on itself implies a truly epigenetic reprogramming, the epigenetic mechanisms governing this process are only partially understood. Moreover, elucidating epigenetic regulation using patient-specific iPSC-derived neural models is expected to have a great impact to unravel the pathophysiology of neurodegenerative diseases and to hopefully expand future therapeutic possibilities. Here we will critically review current knowledge of epigenetic involvement in neurodegenerative disorders focusing on the potential of iPSCs as a promising tool for epigenetic research of these diseases.
Collapse
|
218
|
Dynamic Trk and G Protein Signalings Regulate Dopaminergic Neurodifferentiation in Human Trophoblast Stem Cells. PLoS One 2015; 10:e0143852. [PMID: 26606046 PMCID: PMC4659658 DOI: 10.1371/journal.pone.0143852] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 11/09/2015] [Indexed: 12/18/2022] Open
Abstract
Understanding the mechanisms in the generation of neural stem cells from pluripotent stem cells is a fundamental step towards successful management of neurodegenerative diseases in translational medicine. Albeit all-trans retinoic acid (RA) has been associated with axon outgrowth and nerve regeneration, the maintenance of differentiated neurons, the association with degenerative disease like Parkinson's disease, and its regulatory molecular mechanism from pluripotent stem cells to neural stem cells remain fragmented. We have previously reported that RA is capable of differentiation of human trophoblast stem cells to dopamine (DA) committed progenitor cells. Intracranial implantation of such neural progenitor cells into the 6-OHDA-lesioned substantia nigra pars compacta successfully regenerates dopaminergic neurons and integrity of the nigrostriatal pathway, ameliorating the behavioral deficits in the Parkinson’s disease rat model. Here, we demonstrated a dynamic molecular network in systematic analysis by addressing spatiotemporal molecular expression, intracellular protein-protein interaction and inhibition, imaging study, and genetic expression to explore the regulatory mechanisms of RA induction in the differentiation of human trophoblast stem cells to DA committed progenitor cells. We focused on the tyrosine receptor kinase (Trk), G proteins, canonical Wnt2B/β-catenin, genomic and non-genomic RA signaling transductions with Tyrosine hydroxylase (TH) gene expression as the differentiation endpoint. We found that at the early stage, integration of TrkA and G protein signalings aims for axonogenesis and morphogenesis, involving the novel RXRα/Gαq/11 and RARβ/Gβ signaling pathways. While at the later stage, five distinct signaling pathways together with epigenetic histone modifications emerged to regulate expression of TH, a precursor of dopamine. RA induction generated DA committed progenitor cells in one day. Our results provided substantial mechanistic evidence that human trophoblast stem cell-derived neural stem cells can potentially be used for neurobiological study, drug discovery, and as an alternative source of cell-based therapy in neurodegenerative diseases like Parkinson’s disease.
Collapse
|
219
|
Donadeu FX, Esteves CL. Prospects and Challenges of Induced Pluripotent Stem Cells in Equine Health. Front Vet Sci 2015; 2:59. [PMID: 26664986 PMCID: PMC4672244 DOI: 10.3389/fvets.2015.00059] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/02/2015] [Indexed: 01/12/2023] Open
Abstract
Pluripotent stem cells (PSCs) hold, through the capacity to differentiate into virtually all body cell types, unprecedented promise for human and animal medicine. PSCs are naturally found in the early embryo, and in rodents and humans they can be robustly harvested and grown in culture in the form of embryonic stem cells (ESCs); however, the availability of ESCs from horses is limited. ES-like cells named induced pluripotent stem cells (iPSCs) can be derived in vitro by transcription factor-mediated reprogramming of adult cells. As such, iPSCs can be generated in a patient-specific manner providing unmatched potential for tissue transplantation and in vitro disease modeling. In humans, clinical trials using iPSC-derived cells are already taking place and the use of in vitro iPSC models has identified novel mechanisms of disease and therapeutic targets. Although to a more limited extent, iPSCs have also been generated from horses, a species in which, after humans, these cells are likely to hold the greatest potential in regenerative medicine. Before a clinical use can be envisioned, however, significant challenges will need to be addressed in relation to the robust derivation, long-term culture, differentiation, and clinical safety of equine iPSCs. Toward this objective, recent studies have reported significant improvement in culture conditions and the successful derivation for the first time of functional cell types from equine iPSCs. Given the wide range of exciting applications they could have, it is hoped future research will make the biomedical promise of iPSCs a reality not only for humans but also horses.
Collapse
Affiliation(s)
- F Xavier Donadeu
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh , Midlothian , UK
| | - Cristina L Esteves
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh , Midlothian , UK
| |
Collapse
|
220
|
Tyleckova J, Valekova I, Zizkova M, Rakocyova M, Marsala S, Marsala M, Gadher SJ, Kovarova H. Surface N-glycoproteome patterns reveal key proteins of neuronal differentiation. J Proteomics 2015; 132:13-20. [PMID: 26581640 DOI: 10.1016/j.jprot.2015.11.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 09/03/2015] [Accepted: 11/09/2015] [Indexed: 01/23/2023]
Abstract
UNLABELLED Pluripotent stem cell-derived committed neural precursors are an important source of cells to treat neurodegenerative diseases including spinal cord injury. There remains an urgency to identify markers for monitoring of neural progenitor specificity, estimation of neural fate and follow-up correlation with therapeutic effect in preclinical studies using animal disease models. Cell surface capture technology was used to uncover the cell surface exposed N-glycoproteome of neural precursor cells upon neuronal differentiation as well as post-mitotic mature hNT neurons. The data presented depict an extensive study of surfaceome during neuronal differentiation, confirming glycosylation at a particular predicted site of many of the identified proteins. Quantitative changes detected in cell surface protein levels reveal a set of proteins that highlight the complexity of the neuronal differentiation process. Several of these proteins including the cell adhesion molecules ICAM1, CHL1, and astrotactin1 as well as LAMP1 were validated by SRM. Combination of immunofluorescence staining of ICAM1 and flow cytometry indicated a possible direction for future scrutiny of such proteins as targets for enrichment of the neuronal subpopulation from mixed cultures after differentiation of neural precursor cells. These surface proteins hold an important key for development of safe strategies in cell-replacement therapies of neuronal disorders. BIOLOGICAL SIGNIFICANCE Neural stem and/or precursor cells have a great potential for cell-replacement therapies of neuronal diseases. Availability of well characterised and expandable neural cell lineage specific populations is critical for addressing such a challenge. In our study we identified and relatively quantified several hundred surface N-glycoproteins in the course of neuronal differentiation. We further confirmed the abundant changes for several cell adhesion proteins by SRM and outlined a strategy for utilisation of such N-glycoproteins in antibody based cell sorting. The comprehensive dataset presented here demonstrates the molecular background of neuronal differentiation highly useful for development of new plasma membrane markers to identify and select neuronal subpopulation from mixed neural cell cultures.
Collapse
Affiliation(s)
- Jirina Tyleckova
- Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Laboratory of Applied Proteome Analyses, Libechov, CZ 27721, Czech Republic; Research Center PIGMOD, Laboratory of Applied Proteome Analyses, Libechov, CZ 27721, Czech Republic
| | - Ivona Valekova
- Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Laboratory of Applied Proteome Analyses, Libechov, CZ 27721, Czech Republic; Research Center PIGMOD, Laboratory of Applied Proteome Analyses, Libechov, CZ 27721, Czech Republic; Department of Cell Biology, Faculty of Science, Charles University, CZ 128 43 Prague, Czech Republic
| | - Martina Zizkova
- Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Laboratory of Applied Proteome Analyses, Libechov, CZ 27721, Czech Republic; Research Center PIGMOD, Laboratory of Applied Proteome Analyses, Libechov, CZ 27721, Czech Republic; Department of Cell Biology, Faculty of Science, Charles University, CZ 128 43 Prague, Czech Republic
| | - Michaela Rakocyova
- Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Laboratory of Applied Proteome Analyses, Libechov, CZ 27721, Czech Republic; Research Center PIGMOD, Laboratory of Applied Proteome Analyses, Libechov, CZ 27721, Czech Republic
| | - Silvia Marsala
- University of California, San Diego, Department of Anesthesiology, Neuroregeneration Laboratory, Sanford Consortium for Regenerative Medicine, La Jolla, CA-92037, USA
| | - Martin Marsala
- University of California, San Diego, Department of Anesthesiology, Neuroregeneration Laboratory, Sanford Consortium for Regenerative Medicine, La Jolla, CA-92037, USA
| | | | - Hana Kovarova
- Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Laboratory of Applied Proteome Analyses, Libechov, CZ 27721, Czech Republic; Research Center PIGMOD, Laboratory of Applied Proteome Analyses, Libechov, CZ 27721, Czech Republic.
| |
Collapse
|
221
|
Rapid and robust generation of long-term self-renewing human neural stem cells with the ability to generate mature astroglia. Sci Rep 2015; 5:16321. [PMID: 26541394 PMCID: PMC4635383 DOI: 10.1038/srep16321] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 10/12/2015] [Indexed: 02/07/2023] Open
Abstract
Induced pluripotent stem cell bear the potential to differentiate into any desired cell type and hold large promise for disease-in-a-dish cell-modeling approaches. With the latest advances in the field of reprogramming technology, the generation of patient-specific cells has become a standard technology. However, directed and homogenous differentiation of human pluripotent stem cells into desired specific cell types remains an experimental challenge. Here, we report the development of a novel hiPSCs-based protocol enabling the generation of expandable homogenous human neural stem cells (hNSCs) that can be maintained under self-renewing conditions over high passage numbers. Our newly generated hNSCs retained differentiation potential as evidenced by the reliable generation of mature astrocytes that display typical properties as glutamate up-take and expression of aquaporin-4. The hNSC-derived astrocytes showed high activity of pyruvate carboxylase as assessed by stable isotope assisted metabolic profiling. Moreover, using a cell transplantation approach, we showed that grafted hNSCs were not only able to survive but also to differentiate into astroglial in vivo. Engraftments of pluripotent stem cells derived from somatic cells carry an inherent tumor formation potential. Our results demonstrate that hNSCs with self-renewing and differentiation potential may provide a safer alternative strategy, with promising applications especially for neurodegenerative disorders.
Collapse
|
222
|
Gershon ES, Alliey-Rodriguez N, Grennan K. Ethical and public policy challenges for pharmacogenomics. DIALOGUES IN CLINICAL NEUROSCIENCE 2015. [PMID: 25733960 PMCID: PMC4336925 DOI: 10.31887/dcns.2014.16.4/egershon] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
It is timely to consider the ethical and social questions raised by progress in pharmacogenomics, based on the current importance of pharmacogenomics for avoidance of predictable side effects of drugs, and for correct choice of medications in certain cancers. It has been proposed that the entire population be genotyped for drug-metabolizing enzyme polymorphisms, as a measure that would prevent many untoward and dangerous drug reactions. Pharmacologic treatment targeting based on genomics of disease can be expected to increase greatly in the coming years. Policy and ethical issues exist on consent for large-scale genomic pharmacogenomic data collection, public vs corporate ownership of genomic research results, testing efficacy and safety of drugs used for rare genomic indications, and accessibility of treatments based on costly research that is applicable to relatively few patients. In major psychiatric disorders and intellectual deficiency, rare and de novo deletion or duplication of chromosomal segments (copy number variation), in the aggregate, are common causes of increased risk. This implies that the policy problems of pharmacogenomics will be particularly important for the psychiatric disorders.
Collapse
Affiliation(s)
- Elliot S Gershon
- Department of Psychiatry and Behavioral Neuroscience; Department of Human Genetics; University of Chicago, Illinois, USA
| | - Ney Alliey-Rodriguez
- Department of Psychiatry and Behavioral Neuroscience; University of Chicago, Illinois, USA
| | - Kay Grennan
- Department of Psychiatry and Behavioral Neuroscience; University of Chicago, Illinois, USA
| |
Collapse
|
223
|
Kim TH, Yea CH, Chueng STD, Yin PTT, Conley B, Dardir K, Pak Y, Jung GY, Choi JW, Lee KB. Large-Scale Nanoelectrode Arrays to Monitor the Dopaminergic Differentiation of Human Neural Stem Cells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2015; 27:6356-62. [PMID: 26390254 PMCID: PMC4767621 DOI: 10.1002/adma.201502489] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Revised: 07/17/2015] [Indexed: 05/21/2023]
Abstract
A novel cell-based biosensing platform is developed using a combination of sequential laser interference lithography and electrochemical deposition methods. This enables the sensitive discrimination of dopaminergic cells from other types of neural cells in a completely nondestructive manner. This platform and detection strategy may become an effective noninvasive in situ monitoring tool that can be used to determine stem cell fate for various regenerative applications.
Collapse
Affiliation(s)
- Tae-Hyung Kim
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 (USA)
| | - Cheol-Heon Yea
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 (USA). Department of Chemical & Biomolecular Engineering, Sogang University, Seoul 121-742 (Republic of Korea)
| | - Sy-Tsong Dean Chueng
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 (USA)
| | - Perry To-Tien Yin
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 (USA)
| | - Brian Conley
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 (USA)
| | - Kholud Dardir
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 (USA)
| | - Yusin Pak
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 500-712 (Republic of Korea)
| | - Gun Young Jung
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 500-712 (Republic of Korea)
| | - Jeong-Woo Choi
- Department of Chemical & Biomolecular Engineering, Sogang University, Seoul 121-742 (Republic of Korea), Fax: (+82) 2-3273-0331
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 (USA), Fax: (+1) 732-445-5312, http://kblee.rutgers.edu/. Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 (USA)
| |
Collapse
|
224
|
Son Y, Jenny Lee H, Kim J, Shin H, Choi N, Justin Lee C, Yoon ES, Yoon E, Wise KD, Geun Kim T, Cho IJ. In vivo optical modulation of neural signals using monolithically integrated two-dimensional neural probe arrays. Sci Rep 2015; 5:15466. [PMID: 26494437 PMCID: PMC4616027 DOI: 10.1038/srep15466] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 09/17/2015] [Indexed: 11/09/2022] Open
Abstract
Integration of stimulation modalities (e.g. electrical, optical, and chemical) on a large array of neural probes can enable an investigation of important underlying mechanisms of brain disorders that is not possible through neural recordings alone. Furthermore, it is important to achieve this integration of multiple functionalities in a compact structure to utilize a large number of the mouse models. Here we present a successful optical modulation of in vivo neural signals of a transgenic mouse through our compact 2D MEMS neural array (optrodes). Using a novel fabrication method that embeds a lower cladding layer in a silicon substrate, we achieved a thin silicon 2D optrode array that is capable of delivering light to multiple sites using SU-8 as a waveguide core. Without additional modification to the microelectrodes, the measured impedance of the multiple microelectrodes was below 1 MΩ at 1 kHz. In addition, with a low background noise level (± 25 μV), neural spikes from different individual neurons were recorded on each microelectrode. Lastly, we successfully used our optrodes to modulate the neural activity of a transgenic mouse through optical stimulation. These results demonstrate the functionality of the 2D optrode array and its potential as a next-generation tool for optogenetic applications.
Collapse
Affiliation(s)
- Yoojin Son
- Centre for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), 5 Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 136-791, Korea
- Department of Electrical Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 136-701, Korea
| | - Hyunjoo Jenny Lee
- Centre for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), 5 Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 136-791, Korea
- School of Electrical Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 305-701, Korea
| | - Jeongyeon Kim
- Centre for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology (KIST), 5 Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 136-791, Korea
| | - Hyogeun Shin
- Centre for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), 5 Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 136-791, Korea
- Department of Biomedical Engineering, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, 305-350, Korea
| | - Nakwon Choi
- Centre for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), 5 Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 136-791, Korea
| | - C. Justin Lee
- Centre for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology (KIST), 5 Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 136-791, Korea
| | - Eui-Sung Yoon
- Centre for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), 5 Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 136-791, Korea
| | - Euisik Yoon
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, Michigan, 48105, USA
| | - Kensall D. Wise
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, Michigan, 48105, USA
| | - Tae Geun Kim
- Department of Electrical Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 136-701, Korea
| | - Il-Joo Cho
- Centre for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), 5 Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 136-791, Korea
- Department of Biomedical Engineering, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, 305-350, Korea
| |
Collapse
|
225
|
Li C, Ruan J, Yang M, Pan F, Gao G, Qu S, Shen YL, Dang YJ, Wang K, Jin WL, Cui DX. Human induced pluripotent stem cells labeled with fluorescent magnetic nanoparticles for targeted imaging and hyperthermia therapy for gastric cancer. Cancer Biol Med 2015; 12:163-74. [PMID: 26487961 PMCID: PMC4607817 DOI: 10.7497/j.issn.2095-3941.2015.0040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Objective Human induced pluripotent stem (iPS) cells exhibit great potential for generating functional human cells for medical therapies. In this paper, we report for use of human iPS cells labeled with fluorescent magnetic nanoparticles (FMNPs) for targeted imaging and synergistic therapy of gastric cancer cells in vivo. Methods Human iPS cells were prepared and cultured for 72 h. The culture medium was collected, and then was co-incubated with MGC803 cells. Cell viability was analyzed by the MTT method. FMNP-labeled human iPS cells were prepared and injected into gastric cancer-bearing nude mice. The mouse model was observed using a small-animal imaging system. The nude mice were irradiated under an external alternating magnetic field and evaluated using an infrared thermal mapping instrument. Tumor sizes were measured weekly. Results iPS cells and the collected culture medium inhibited the growth of MGC803 cells. FMNP-labeled human iPS cells targeted and imaged gastric cancer cells in vivo, as well as inhibited cancer growth in vivo through the external magnetic field. Conclusion FMNP-labeled human iPS cells exhibit considerable potential in applications such as targeted dual-mode imaging and synergistic therapy for early gastric cancer.
Collapse
Affiliation(s)
- Chao Li
- 1 Institute of Nano Biomedicine and Engineering, Key Laboratory for Thin Film and Microfabrication of Ministry of Education, Department of Instrument Science and Engineering, National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, Shanghai 200240, China ; 2 Basic Medical Sciences Department of Biochemistry & Molecular Biology Key Laboratory of Molecular Medicine, Fudan University, Shanghai 200032, China ; 3 Department of Imaging and Nuclear Medicine, Shanghai Sixth People's Hospital, Shanghai 20006, China
| | - Jing Ruan
- 1 Institute of Nano Biomedicine and Engineering, Key Laboratory for Thin Film and Microfabrication of Ministry of Education, Department of Instrument Science and Engineering, National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, Shanghai 200240, China ; 2 Basic Medical Sciences Department of Biochemistry & Molecular Biology Key Laboratory of Molecular Medicine, Fudan University, Shanghai 200032, China ; 3 Department of Imaging and Nuclear Medicine, Shanghai Sixth People's Hospital, Shanghai 20006, China
| | - Meng Yang
- 1 Institute of Nano Biomedicine and Engineering, Key Laboratory for Thin Film and Microfabrication of Ministry of Education, Department of Instrument Science and Engineering, National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, Shanghai 200240, China ; 2 Basic Medical Sciences Department of Biochemistry & Molecular Biology Key Laboratory of Molecular Medicine, Fudan University, Shanghai 200032, China ; 3 Department of Imaging and Nuclear Medicine, Shanghai Sixth People's Hospital, Shanghai 20006, China
| | - Fei Pan
- 1 Institute of Nano Biomedicine and Engineering, Key Laboratory for Thin Film and Microfabrication of Ministry of Education, Department of Instrument Science and Engineering, National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, Shanghai 200240, China ; 2 Basic Medical Sciences Department of Biochemistry & Molecular Biology Key Laboratory of Molecular Medicine, Fudan University, Shanghai 200032, China ; 3 Department of Imaging and Nuclear Medicine, Shanghai Sixth People's Hospital, Shanghai 20006, China
| | - Guo Gao
- 1 Institute of Nano Biomedicine and Engineering, Key Laboratory for Thin Film and Microfabrication of Ministry of Education, Department of Instrument Science and Engineering, National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, Shanghai 200240, China ; 2 Basic Medical Sciences Department of Biochemistry & Molecular Biology Key Laboratory of Molecular Medicine, Fudan University, Shanghai 200032, China ; 3 Department of Imaging and Nuclear Medicine, Shanghai Sixth People's Hospital, Shanghai 20006, China
| | - Su Qu
- 1 Institute of Nano Biomedicine and Engineering, Key Laboratory for Thin Film and Microfabrication of Ministry of Education, Department of Instrument Science and Engineering, National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, Shanghai 200240, China ; 2 Basic Medical Sciences Department of Biochemistry & Molecular Biology Key Laboratory of Molecular Medicine, Fudan University, Shanghai 200032, China ; 3 Department of Imaging and Nuclear Medicine, Shanghai Sixth People's Hospital, Shanghai 20006, China
| | - You-Lan Shen
- 1 Institute of Nano Biomedicine and Engineering, Key Laboratory for Thin Film and Microfabrication of Ministry of Education, Department of Instrument Science and Engineering, National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, Shanghai 200240, China ; 2 Basic Medical Sciences Department of Biochemistry & Molecular Biology Key Laboratory of Molecular Medicine, Fudan University, Shanghai 200032, China ; 3 Department of Imaging and Nuclear Medicine, Shanghai Sixth People's Hospital, Shanghai 20006, China
| | - Yong-Jun Dang
- 1 Institute of Nano Biomedicine and Engineering, Key Laboratory for Thin Film and Microfabrication of Ministry of Education, Department of Instrument Science and Engineering, National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, Shanghai 200240, China ; 2 Basic Medical Sciences Department of Biochemistry & Molecular Biology Key Laboratory of Molecular Medicine, Fudan University, Shanghai 200032, China ; 3 Department of Imaging and Nuclear Medicine, Shanghai Sixth People's Hospital, Shanghai 20006, China
| | - Kan Wang
- 1 Institute of Nano Biomedicine and Engineering, Key Laboratory for Thin Film and Microfabrication of Ministry of Education, Department of Instrument Science and Engineering, National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, Shanghai 200240, China ; 2 Basic Medical Sciences Department of Biochemistry & Molecular Biology Key Laboratory of Molecular Medicine, Fudan University, Shanghai 200032, China ; 3 Department of Imaging and Nuclear Medicine, Shanghai Sixth People's Hospital, Shanghai 20006, China
| | - Wei-Lin Jin
- 1 Institute of Nano Biomedicine and Engineering, Key Laboratory for Thin Film and Microfabrication of Ministry of Education, Department of Instrument Science and Engineering, National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, Shanghai 200240, China ; 2 Basic Medical Sciences Department of Biochemistry & Molecular Biology Key Laboratory of Molecular Medicine, Fudan University, Shanghai 200032, China ; 3 Department of Imaging and Nuclear Medicine, Shanghai Sixth People's Hospital, Shanghai 20006, China
| | - Da-Xiang Cui
- 1 Institute of Nano Biomedicine and Engineering, Key Laboratory for Thin Film and Microfabrication of Ministry of Education, Department of Instrument Science and Engineering, National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, Shanghai 200240, China ; 2 Basic Medical Sciences Department of Biochemistry & Molecular Biology Key Laboratory of Molecular Medicine, Fudan University, Shanghai 200032, China ; 3 Department of Imaging and Nuclear Medicine, Shanghai Sixth People's Hospital, Shanghai 20006, China
| |
Collapse
|
226
|
Fu MH, Li CL, Lin HL, Chen PC, Calkins MJ, Chang YF, Cheng PH, Yang SH. Stem cell transplantation therapy in Parkinson's disease. SPRINGERPLUS 2015; 4:597. [PMID: 26543732 PMCID: PMC4628010 DOI: 10.1186/s40064-015-1400-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 10/06/2015] [Indexed: 02/06/2023]
Abstract
Ineffective therapeutic treatments and inadequate repair ability in the central nervous system are disturbing problems for several neurological diseases. Fortunately, the development of clinically applicable populations of stem cells has provided an avenue to overcome the failure of endogenous repair systems and substitute new cells into the damaged brain. However, there are still several existing obstacles to translating into clinical application. Here we review the stem-cell based therapies for Parkinson’s disease and discuss the potential advantages and drawbacks. We hope this review may provide suggestions for viable strategies to overcome the current technical and biological issues associated with the application of stem cells in Parkinson’s disease.
Collapse
Affiliation(s)
- Mu-Hui Fu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101 Taiwan.,Department of Neurology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, 83301 Taiwan
| | - Chia-Ling Li
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101 Taiwan
| | - Hsiu-Lien Lin
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101 Taiwan.,Division of Breeding and Genetics, Livestock Research Institute, Council of Agriculture, Tainan, 71246 Taiwan
| | - Pei-Chun Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101 Taiwan.,Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, 70101 Taiwan
| | - Marcus J Calkins
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 70101 Taiwan
| | - Yu-Fan Chang
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, 70101 Taiwan
| | - Pei-Hsun Cheng
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, 70101 Taiwan
| | - Shang-Hsun Yang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101 Taiwan.,Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, 70101 Taiwan
| |
Collapse
|
227
|
Zomer HD, Vidane AS, Gonçalves NN, Ambrósio CE. Mesenchymal and induced pluripotent stem cells: general insights and clinical perspectives. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2015; 8:125-34. [PMID: 26451119 PMCID: PMC4592031 DOI: 10.2147/sccaa.s88036] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cells have awakened a great deal of interest in regenerative medicine due to their plasticity, and immunomodulatory and anti-inflammatory properties. They are high-yield and can be acquired through noninvasive methods from adult tissues. Moreover, they are nontumorigenic and are the most widely studied. On the other hand, induced pluripotent stem (iPS) cells can be derived directly from adult cells through gene reprogramming. The new iPS technology avoids the embryo destruction or manipulation to generate pluripotent cells, therefore, are exempt from ethical implication surrounding embryonic stem cell use. The pre-differentiation of iPS cells ensures the safety of future approaches. Both mesenchymal stem cells and iPS cells can be used for autologous cell transplantations without the risk of immune rejection and represent a great opportunity for future alternative therapies. In this review we discussed the therapeutic perspectives using mesenchymal and iPS cells.
Collapse
Affiliation(s)
- Helena D Zomer
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| | - Atanásio S Vidane
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| | - Natalia N Gonçalves
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| | - Carlos E Ambrósio
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo, Pirassununga, SP, Brazil
| |
Collapse
|
228
|
Li W, Chen S, Li JY. Human induced pluripotent stem cells in Parkinson's disease: A novel cell source of cell therapy and disease modeling. Prog Neurobiol 2015; 134:161-77. [PMID: 26408505 DOI: 10.1016/j.pneurobio.2015.09.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 09/15/2015] [Accepted: 09/17/2015] [Indexed: 12/16/2022]
Abstract
Human induced pluripotent stem cells (hiPSCs) and human embryonic stem cells (hESCs) are two novel cell sources for studying neurodegenerative diseases. Dopaminergic neurons derived from hiPSCs/hESCs have been implicated to be very useful in Parkinson's disease (PD) research, including cell replacement therapy, disease modeling and drug screening. Recently, great efforts have been made to improve the application of hiPSCs/hESCs in PD research. Considerable advances have been made in recent years, including advanced reprogramming strategies without the use of viruses or using fewer transcriptional factors, optimized methods for generating highly homogeneous neural progenitors with a larger proportion of mature dopaminergic neurons and better survival and integration after transplantation. Here we outline the progress that has been made in these aspects in recent years, particularly during the last year, and also discuss existing issues that need to be addressed.
Collapse
Affiliation(s)
- Wen Li
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Rui Jin Er Road, Shanghai 200025, China; Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Lund University, BMC A10, 221 84 Lund, Sweden
| | - Shengdi Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Rui Jin Er Road, Shanghai 200025, China.
| | - Jia-Yi Li
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, China; Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Lund University, BMC A10, 221 84 Lund, Sweden.
| |
Collapse
|
229
|
Singh A, Gutekunst CA, Uthayathas S, Finberg JPM, Mewes K, Gross RE, Papa SM, Feld Y. Effects of fibroblast transplantation into the internal pallidum on levodopa-induced dyskinesias in parkinsonian non-human primates. Neurosci Bull 2015; 31:705-13. [PMID: 26373985 DOI: 10.1007/s12264-015-1559-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Accepted: 06/25/2015] [Indexed: 10/23/2022] Open
Abstract
Recent studies have shown that fibroblast transplantation can modify the activity of basal ganglia networks in models of Parkinson's disease. To determine its effects on parkinsonian motor symptoms, we performed autologous dermal fibroblast transplantation into the internal pallidum (GPi) in two parkinsonian rhesus monkeys with stable levodopa-induced dyskinesias (LIDs). Levodopa responses were assessed every week after transplantation for three months. A reduction of between 58% and 64% in total LIDs on the contralateral side was observed in both animals. No clear LID changes were observed on the ipsilateral side. These effects lasted the entire 3-month period in one monkey, but declined after 6-8 weeks in the other. The antiparkinsonian effects of levodopa did not diminish. The results of this pilot study indicate that fibroblast transplantation into the GPi may have beneficial effects on LIDs and warrant further investigation for potential therapeutic use.
Collapse
Affiliation(s)
- Arun Singh
- Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Claire A Gutekunst
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Subramaniam Uthayathas
- Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - John P M Finberg
- Molecular Pharmacology Department, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Klaus Mewes
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Robert E Gross
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Stella M Papa
- Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | | |
Collapse
|
230
|
Current Neurogenic and Neuroprotective Strategies to Prevent and Treat Neurodegenerative and Neuropsychiatric Disorders. Neuromolecular Med 2015; 17:404-22. [PMID: 26374113 DOI: 10.1007/s12017-015-8369-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 08/22/2015] [Indexed: 12/31/2022]
Abstract
The adult central nervous system is commonly known to have a very limited regenerative capacity. The presence of functional stem cells in the brain can therefore be seen as a paradox, since in other organs these are known to counterbalance cell loss derived from pathological conditions. This fact has therefore raised the possibility to stimulate neural stem cell differentiation and proliferation or survival by either stem cell replacement therapy or direct administration of neurotrophic factors or other proneurogenic molecules, which in turn has also originated regenerative medicine for the treatment of otherwise incurable neurodegenerative and neuropsychiatric disorders that take a huge toll on society. This may be facilitated by the fact that many of these disorders converge on similar pathophysiological pathways: excitotoxicity, oxidative stress, neuroinflammation, mitochondrial failure, excessive intracellular calcium and apoptosis. This review will therefore focus on the most promising achievements in promoting neuroprotection and neuroregeneration reported to date.
Collapse
|
231
|
Han F, Baremberg D, Gao J, Duan J, Lu X, Zhang N, Chen Q. Development of stem cell-based therapy for Parkinson's disease. Transl Neurodegener 2015; 4:16. [PMID: 26339485 PMCID: PMC4559356 DOI: 10.1186/s40035-015-0039-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 08/24/2015] [Indexed: 12/31/2022] Open
Abstract
Parkinson’s disease (PD) is one of the most common neurodegenerative disorders of aging, characterized by the degeneration of dopamine neurons (DA neurons) in the substantial nigra, leading to the advent of both motor symptoms and non-motor symptoms. Current treatments include electrical stimulation of the affected brain areas and dopamine replacement therapy. Even though both categories are effective in treating PD patients, the disease progression cannot be stopped. The research advance into cell therapies provides exciting potential for the treatment of PD. Current cell sources include neural stem cells (NSCs) from fetal brain tissues, human embryonic stem cells (hESCs), induced pluripotent stem cells (iPSCs) and directly induced dopamine neurons (iDA neurons). Here, we evaluate the research progress in different cell sources with a focus on using iPSCs as a valuable source and propose key challenges for developing cells suitable for large-scale clinical applications in the treatment of PD.
Collapse
Affiliation(s)
- Fabin Han
- Centre for Stem Cells and Regenerative Medicine, The Liaocheng People's Hospital/Affiliated Liaocheng Hospital, Taishan Medical University, Shandong, 252000 China
| | - Deborah Baremberg
- Centre for Stem Cells and Regenerative Medicine, The Liaocheng People's Hospital/Affiliated Liaocheng Hospital, Taishan Medical University, Shandong, 252000 China
| | - Junyu Gao
- Centre for Stem Cells and Regenerative Medicine, The Liaocheng People's Hospital/Affiliated Liaocheng Hospital, Taishan Medical University, Shandong, 252000 China
| | - Jing Duan
- Centre for Stem Cells and Regenerative Medicine, The Liaocheng People's Hospital/Affiliated Liaocheng Hospital, Taishan Medical University, Shandong, 252000 China
| | - Xianjie Lu
- Centre for Stem Cells and Regenerative Medicine, The Liaocheng People's Hospital/Affiliated Liaocheng Hospital, Taishan Medical University, Shandong, 252000 China
| | - Nan Zhang
- Centre for Stem Cells and Regenerative Medicine, The Liaocheng People's Hospital/Affiliated Liaocheng Hospital, Taishan Medical University, Shandong, 252000 China
| | - Qingfa Chen
- Centre for Stem Cells and Regenerative Medicine, The Liaocheng People's Hospital/Affiliated Liaocheng Hospital, Taishan Medical University, Shandong, 252000 China
| |
Collapse
|
232
|
Staudt MD, Di Sebastiano AR, Xu H, Jog M, Schmid S, Foster P, Hebb MO. Advances in Neurotrophic Factor and Cell-Based Therapies for Parkinson's Disease: A Mini-Review. Gerontology 2015; 62:371-80. [PMID: 26330171 DOI: 10.1159/000438701] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 07/14/2015] [Indexed: 12/28/2022] Open
Abstract
Parkinson's disease (PD) affects an estimated 7-10 million people worldwide and remains without definitive or disease-modifying treatment. There have been many recent developments in cell-based therapy (CBT) to replace lost circuitry and provide chronic biological sources of therapeutic agents to the PD-affected brain. Early neural transplantation studies underscored the challenges of immune compatibility, graft integration and the need for renewable, autologous graft sources. Neurotrophic factors (NTFs) offer a potential class of cytoprotective pharmacotherapeutics that may complement dopamine (DA) replacement and CBT strategies in PD. Chronic NTF delivery may be an integral goal of CBT, with grafts consisting of autologous drug-producing (e.g., DA, NTF) cells that are capable of integration and function in the host brain. In this mini-review, we outline the past experience and recent advances in NTF technology and CBT as promising and integrated approaches for the treatment of PD.
Collapse
Affiliation(s)
- Michael D Staudt
- Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western University, London, Ont., Canada
| | | | | | | | | | | | | |
Collapse
|
233
|
Lu MY, Li Z, Hwang SM, Linju Yen B, Lee GB. Generation of murine induced pluripotent stem cells by using high-density distributed electrodes network. BIOMICROFLUIDICS 2015; 9:054107. [PMID: 26396661 PMCID: PMC4567575 DOI: 10.1063/1.4930866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 08/31/2015] [Indexed: 06/05/2023]
Abstract
This study reports a robust method of gene transfection in a murine primary cell model by using a high-density electrodes network (HDEN). By demonstrating high cell viability after gene transfection and successful expression of transgenes including fluorescent proteins, the HDEN device shows great promise as a solution in which reprogramming efficiency using non-viral induction for generation of murine induced pluripotent stem cells (iPSCs) is optimized. High and steady transgene expression levels in host cells of iPSCs can be demonstrated using this method. Moreover, the HDEN device achieved successful gene transfection with a low voltage of less than 180 V while requiring relatively low cell numbers (less than 1.5 × 10(4) cells). The results are comparable to current conventional methods, demonstrating a reasonable fluorescent-plasmid transfection rate (42.4% in single transfection and 24.5% in triple transfection) and high cell viability of over 95%. The gene expression levels of each iPSC factor was measured to be over 10-fold higher than that reported in previous studies using a single mouse embryonic fibroblast cell. Our results demonstrate that the generation of iPSCs using HDEN transfection of plasmid DNA may be a feasible and safe alternative to using viral transfection methods in the near future.
Collapse
Affiliation(s)
- Ming-Yu Lu
- Department of Power Mechanical Engineering, National Tsing Hua University , Hsinchu, Taiwan
| | - Zhihong Li
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Institute of Microelectronics, Peking University , Beijing, China
| | - Shiaw-Min Hwang
- Bioresource Collection and Research Center, Food Industry Research and Development Institute , Hsinchu, Taiwan
| | - B Linju Yen
- Regenerative Medicine Research Group, Institute of Cellular and System Medicine, National Health Research Institutes , Zhunan, Taiwan
| | | |
Collapse
|
234
|
Aravantinou-Fatorou K, Ortega F, Chroni-Tzartou D, Antoniou N, Poulopoulou C, Politis PK, Berninger B, Matsas R, Thomaidou D. CEND1 and NEUROGENIN2 Reprogram Mouse Astrocytes and Embryonic Fibroblasts to Induced Neural Precursors and Differentiated Neurons. Stem Cell Reports 2015; 5:405-18. [PMID: 26321141 PMCID: PMC4618597 DOI: 10.1016/j.stemcr.2015.07.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 07/28/2015] [Accepted: 07/30/2015] [Indexed: 01/10/2023] Open
Abstract
Recent studies demonstrate that astroglia from non-neurogenic brain regions can be reprogrammed into functional neurons through forced expression of neurogenic factors. Here we explored the effect of CEND1 and NEUROG2 on reprogramming of mouse cortical astrocytes and embryonic fibroblasts. Forced expression of CEND1, NEUROG2, or both resulted in acquisition of induced neuronal cells expressing subtype-specific markers, while long-term live-cell imaging highlighted the existence of two different modes of neuronal trans-differentiation. Of note, a subpopulation of CEND1 and NEUROG2 double-transduced astrocytes formed spheres exhibiting neural stem cell properties. mRNA and protein expression studies revealed a reciprocal feedback loop existing between the two molecules, while knockdown of endogenous CEND1 demonstrated that it is a key mediator of NEUROG2-driven neuronal reprogramming. Our data suggest that common reprogramming mechanisms exist driving the conversion of lineage-distant somatic cell types to neurons and reveal a critical role for CEND1 in NEUROG2-driven astrocytic reprogramming. CEND1 reprograms astrocytes and fibroblasts to GABAergic neurons Neurospheres are formed from CEND1+ and NEUROG2+ cells through the β-catenin pathway CEND1 and NEUROG2 participate in a reciprocal feedback loop leading to neurogenesis CEND1 is a key mediator of NEUROG2 reprogramming function
Collapse
Affiliation(s)
| | - Felipe Ortega
- Research Group Adult Neurogenesis and Cellular Reprogramming, Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, 55128 Mainz, Germany
| | - Dafni Chroni-Tzartou
- Department of Neurobiology, Hellenic Pasteur Institute, 127 Vasilissis Sofias Avenue, Athens 11521, Greece; Department of Neurology, Laboratory of Experimental Neurophysiology, University of Athens Medical School, Eginition Hospital, 72-74 Vasilissis Sofias Avenue, Athens 11521, Greece
| | - Nasia Antoniou
- Department of Neurobiology, Hellenic Pasteur Institute, 127 Vasilissis Sofias Avenue, Athens 11521, Greece
| | - Cornelia Poulopoulou
- Department of Neurology, Laboratory of Experimental Neurophysiology, University of Athens Medical School, Eginition Hospital, 72-74 Vasilissis Sofias Avenue, Athens 11521, Greece
| | - Panagiotis K Politis
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efessiou Street, Athens 11527, Greece
| | - Benedikt Berninger
- Research Group Adult Neurogenesis and Cellular Reprogramming, Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, 55128 Mainz, Germany
| | - Rebecca Matsas
- Department of Neurobiology, Hellenic Pasteur Institute, 127 Vasilissis Sofias Avenue, Athens 11521, Greece
| | - Dimitra Thomaidou
- Department of Neurobiology, Hellenic Pasteur Institute, 127 Vasilissis Sofias Avenue, Athens 11521, Greece.
| |
Collapse
|
235
|
Guo XL, Chen JS. Research on induced pluripotent stem cells and the application in ocular tissues. Int J Ophthalmol 2015; 8:818-25. [PMID: 26309885 DOI: 10.3980/j.issn.2222-3959.2015.04.31] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 02/02/2015] [Indexed: 12/31/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) were firstly induced from mouse fibroblasts since 2006, and then the research on iPSCs had made great progress in the following years. iPSCs were established from different somatic cells through DNA, RNA, protein or small molecule pathways and transduction vehicles. With continuous improvement of technology on reprogramming, the induction of iPSCs became more secure and effective, and showed enormous promise for clinical applications. We reviewed different reprogramming of somatic cells, four kinds of pathways of reprogramming and three types of transduction vehicles, and discuss the research of iPSCs in ophthalmology and the prospect of iPSCs applications.
Collapse
Affiliation(s)
- Xiao-Ling Guo
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Jian-Su Chen
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou 510632, Guangdong Province, China ; Eye Institute, Medical College of Jinan University, Guangzhou 510632, Guangdong Province, China ; Department of Ophthalmology, the First Clinical Medical College of Jinan University, Guangzhou 510632, Guangdong Province, China
| |
Collapse
|
236
|
Gallegos-Cárdenas A, Webb R, Jordan E, West R, West FD, Yang JY, Wang K, Stice SL. Pig Induced Pluripotent Stem Cell-Derived Neural Rosettes Developmentally Mimic Human Pluripotent Stem Cell Neural Differentiation. Stem Cells Dev 2015; 24:1901-11. [DOI: 10.1089/scd.2015.0025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Affiliation(s)
- Amalia Gallegos-Cárdenas
- Regenerative Bioscience Center, University of Georgia, Rhodes Center for Animal and Dairy Science, Athens, Georgia
- Department of Animal and Dairy Science, University of Georgia, Rhodes Center for Animal and Dairy Science, Athens, Georgia
- Departamento de Producción Animal, Facultad de Zootecnia, Universidad Nacional Agraria La Molina, Girona, Perú
| | - Robin Webb
- Regenerative Bioscience Center, University of Georgia, Rhodes Center for Animal and Dairy Science, Athens, Georgia
- Department of Animal and Dairy Science, University of Georgia, Rhodes Center for Animal and Dairy Science, Athens, Georgia
| | - Erin Jordan
- Regenerative Bioscience Center, University of Georgia, Rhodes Center for Animal and Dairy Science, Athens, Georgia
- Department of Animal and Dairy Science, University of Georgia, Rhodes Center for Animal and Dairy Science, Athens, Georgia
| | - Rachel West
- Regenerative Bioscience Center, University of Georgia, Rhodes Center for Animal and Dairy Science, Athens, Georgia
- Department of Animal and Dairy Science, University of Georgia, Rhodes Center for Animal and Dairy Science, Athens, Georgia
| | - Franklin D. West
- Regenerative Bioscience Center, University of Georgia, Rhodes Center for Animal and Dairy Science, Athens, Georgia
- Department of Animal and Dairy Science, University of Georgia, Rhodes Center for Animal and Dairy Science, Athens, Georgia
| | - Jeong-Yeh Yang
- Regenerative Bioscience Center, University of Georgia, Rhodes Center for Animal and Dairy Science, Athens, Georgia
- Department of Animal and Dairy Science, University of Georgia, Rhodes Center for Animal and Dairy Science, Athens, Georgia
| | - Kai Wang
- Regenerative Bioscience Center, University of Georgia, Rhodes Center for Animal and Dairy Science, Athens, Georgia
- Department of Animal and Dairy Science, University of Georgia, Rhodes Center for Animal and Dairy Science, Athens, Georgia
| | - Steven L. Stice
- Regenerative Bioscience Center, University of Georgia, Rhodes Center for Animal and Dairy Science, Athens, Georgia
- Department of Animal and Dairy Science, University of Georgia, Rhodes Center for Animal and Dairy Science, Athens, Georgia
| |
Collapse
|
237
|
Dehshahri A, Sadeghpour H. Surface decorations of poly(amidoamine) dendrimer by various pendant moieties for improved delivery of nucleic acid materials. Colloids Surf B Biointerfaces 2015; 132:85-102. [PMID: 26022400 DOI: 10.1016/j.colsurfb.2015.05.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Revised: 05/05/2015] [Accepted: 05/07/2015] [Indexed: 12/22/2022]
|
238
|
Cerri S, Greco R, Levandis G, Ghezzi C, Mangione AS, Fuzzati-Armentero MT, Bonizzi A, Avanzini MA, Maccario R, Blandini F. Intracarotid Infusion of Mesenchymal Stem Cells in an Animal Model of Parkinson's Disease, Focusing on Cell Distribution and Neuroprotective and Behavioral Effects. Stem Cells Transl Med 2015. [PMID: 26198165 DOI: 10.5966/sctm.2015-0023] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
UNLABELLED Mesenchymal stem cells (MSCs) have been proposed as a potential therapeutic tool for Parkinson's disease (PD) and systemic administration of these cells has been tested in preclinical and clinical studies. However, no information on survival and actual capacity of MSCs to reach the brain has been provided. In this study, we evaluated homing of intraarterially infused rat MSCs (rMSCs) in the brain of rats bearing a 6-hydroxydopamine (6-OHDA)-induced lesion of the nigrostriatal tract, to establish whether the toxin-induced damage is sufficient to grant MSC passage across the blood-brain barrier (BBB) or if a transient BBB disruption is necessary. The rMSC distribution in peripheral organs and the effects of cell infusion on neurodegenerative process and motor deficits were also investigated. rMSCs were infused 14 days after 6-OHDA injection. A hyperosmolar solution of mannitol was used to transiently permeabilize the BBB. Behavioral impairment was assessed by adjusting step test and response to apomorphine. Animals were sacrificed 7 and 28 days after cell infusion. Our work shows that appreciable delivery of rMSCs to the brain of 6-OHDA-lesioned animals can be obtained only after mannitol pretreatment. A notable percentage of infused cells accumulated in peripheral organs. Infusion of rMSCs did not modify the progression of 6-OHDA-induced damage or the motor impairment at the stepping test, but induced progressive normalization of the pathological response (contralateral turning) to apomorphine administration. These findings suggest that many aspects should be further investigated before considering any translation of MSC systemic administration into the clinical setting for PD treatment. SIGNIFICANCE This study demonstrates that mesenchymal stem cells infused through the carotid artery do not efficiently cross the blood-brain barrier in rats with a Parkinson's disease-like degeneration of nigrostriatal neurons, unless a permeabilizing agent (e.g., mannitol) is used. The infusion did not reduce the neuronal damage and associated motor impairment, but abolished the motor abnormalities these animals typically show when challenged with a dopaminergic agonist. Therefore, although arterially infused mesenchymal stem cells did not show neurorestorative effects in this study's Parkinson's disease model, they appeared to normalize the pathological responsiveness of striatal neurons to dopaminergic stimulation. This capability should be further explored in future studies.
Collapse
Affiliation(s)
- Silvia Cerri
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, and Laboratory of Neurophysiology of Integrative Autonomic Systems, "C. Mondino" National Neurological Institute, Pavia, Italy; Immunology and Transplantation Laboratory/Cell Factory/Pediatric Hematology/Oncology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Rosaria Greco
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, and Laboratory of Neurophysiology of Integrative Autonomic Systems, "C. Mondino" National Neurological Institute, Pavia, Italy; Immunology and Transplantation Laboratory/Cell Factory/Pediatric Hematology/Oncology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Giovanna Levandis
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, and Laboratory of Neurophysiology of Integrative Autonomic Systems, "C. Mondino" National Neurological Institute, Pavia, Italy; Immunology and Transplantation Laboratory/Cell Factory/Pediatric Hematology/Oncology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Cristina Ghezzi
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, and Laboratory of Neurophysiology of Integrative Autonomic Systems, "C. Mondino" National Neurological Institute, Pavia, Italy; Immunology and Transplantation Laboratory/Cell Factory/Pediatric Hematology/Oncology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Antonina Stefania Mangione
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, and Laboratory of Neurophysiology of Integrative Autonomic Systems, "C. Mondino" National Neurological Institute, Pavia, Italy; Immunology and Transplantation Laboratory/Cell Factory/Pediatric Hematology/Oncology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Marie-Therese Fuzzati-Armentero
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, and Laboratory of Neurophysiology of Integrative Autonomic Systems, "C. Mondino" National Neurological Institute, Pavia, Italy; Immunology and Transplantation Laboratory/Cell Factory/Pediatric Hematology/Oncology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Arianna Bonizzi
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, and Laboratory of Neurophysiology of Integrative Autonomic Systems, "C. Mondino" National Neurological Institute, Pavia, Italy; Immunology and Transplantation Laboratory/Cell Factory/Pediatric Hematology/Oncology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Maria Antonietta Avanzini
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, and Laboratory of Neurophysiology of Integrative Autonomic Systems, "C. Mondino" National Neurological Institute, Pavia, Italy; Immunology and Transplantation Laboratory/Cell Factory/Pediatric Hematology/Oncology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Rita Maccario
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, and Laboratory of Neurophysiology of Integrative Autonomic Systems, "C. Mondino" National Neurological Institute, Pavia, Italy; Immunology and Transplantation Laboratory/Cell Factory/Pediatric Hematology/Oncology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Fabio Blandini
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, and Laboratory of Neurophysiology of Integrative Autonomic Systems, "C. Mondino" National Neurological Institute, Pavia, Italy; Immunology and Transplantation Laboratory/Cell Factory/Pediatric Hematology/Oncology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
239
|
Mesenchymal Stromal Cells Affect Disease Outcomes via Macrophage Polarization. Stem Cells Int 2015; 2015:989473. [PMID: 26257791 PMCID: PMC4518189 DOI: 10.1155/2015/989473] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 06/30/2015] [Indexed: 12/21/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are multipotent and self-renewable cells that reside in almost all postnatal tissues. In recent years, many studies have reported the effect of MSCs on the innate and adaptive immune systems. MSCs regulate the proliferation, activation, and effector function of T lymphocytes, professional antigen presenting cells (dendritic cells, macrophages, and B lymphocytes), and NK cells via direct cell-to-cell contact or production of soluble factors including indoleamine 2,3-dioxygenase, prostaglandin E2, tumor necrosis factor-α stimulated gene/protein 6, nitric oxide, and IL-10. MSCs are also able to reprogram macrophages from a proinflammatory M1 phenotype toward an anti-inflammatory M2 phenotype capable of regulating immune response. Because of their capacity for differentiation and immunomodulation, MSCs have been used in many preclinical and clinical studies as possible new therapeutic agents for the treatment of autoimmune, degenerative, and inflammatory diseases. In this review, we discuss the central role of MSCs in macrophage polarization and outcomes of diseases such as wound healing, brain/spinal cord injuries, and diseases of heart, lung, and kidney in animal models.
Collapse
|
240
|
Wan W, Cao L, Kalionis B, Xia S, Tai X. Applications of Induced Pluripotent Stem Cells in Studying the Neurodegenerative Diseases. Stem Cells Int 2015; 2015:382530. [PMID: 26240571 PMCID: PMC4512612 DOI: 10.1155/2015/382530] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Accepted: 12/05/2014] [Indexed: 12/21/2022] Open
Abstract
Neurodegeneration is the umbrella term for the progressive loss of structure or function of neurons. Incurable neurodegenerative disorders such as Alzheimer's disease (AD) and Parkinson's disease (PD) show dramatic rising trends particularly in the advanced age groups. However, the underlying mechanisms are not yet fully elucidated, and to date there are no biomarkers for early detection or effective treatments for the underlying causes of these diseases. Furthermore, due to species variation and differences between animal models (e.g., mouse transgenic and knockout models) of neurodegenerative diseases, substantial debate focuses on whether animal and cell culture disease models can correctly model the condition in human patients. In 2006, Yamanaka of Kyoto University first demonstrated a novel approach for the preparation of induced pluripotent stem cells (iPSCs), which displayed similar pluripotency potential to embryonic stem cells (ESCs). Currently, iPSCs studies are permeating many sectors of disease research. Patient sample-derived iPSCs can be used to construct patient-specific disease models to elucidate the pathogenic mechanisms of disease development and to test new therapeutic strategies. Accordingly, the present review will focus on recent progress in iPSC research in the modeling of neurodegenerative disorders and in the development of novel therapeutic options.
Collapse
Affiliation(s)
- Wenbin Wan
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Lan Cao
- State Key Laboratory of Medical Neurobiology, Department of Neurobiology and Institutes of Brain Science, School of Basic Medical Science, Fudan University, Shanghai 200032, China
| | - Bill Kalionis
- Department of Perinatal Medicine, Pregnancy Research Centre and University of Melbourne Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, VIC 3052, Australia
| | - Shijin Xia
- Shanghai Institute of Geriatrics, Huadong Hospital, Fudan University, Shanghai 200040, China
| | - Xiantao Tai
- School of Acupuncture, Massage and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming 650500, China
| |
Collapse
|
241
|
Multiple sclerosis: getting personal with induced pluripotent stem cells. Cell Death Dis 2015; 6:e1806. [PMID: 26158512 PMCID: PMC4650727 DOI: 10.1038/cddis.2015.179] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 05/27/2015] [Accepted: 06/01/2015] [Indexed: 12/13/2022]
Abstract
Human induced pluripotent stem (iPS) cells can be derived from lineage-restricted cells and represent an important tool to develop novel patient-specific cell therapies and research models for inherited and acquired diseases. Recently, patient-derived iPS cells, containing donor genetic background, have offered a breakthrough approach to study human genetics of neurodegenerative diseases. By offering an unlimited source of patient-specific disease-relevant cells, iPS cells hold great promise for understanding disease mechanisms, identifying molecular targets and developing phenotypic screens for drug discovery. This review will discuss the potential impact of using iPS cell-derived models in multiple sclerosis (MS) research and highlight some of the current challenges and prospective for generating novel therapeutic treatments for MS patients.
Collapse
|
242
|
Immunogenicity and functional evaluation of iPSC-derived organs for transplantation. Cell Discov 2015; 1:15015. [PMID: 27462414 PMCID: PMC4860825 DOI: 10.1038/celldisc.2015.15] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 05/07/2015] [Indexed: 12/19/2022] Open
Abstract
Whether physiologically induced pluripotent stem cell (iPSC)-derived organs are immunogenic and can be used for transplantation is unclear. Here, we generated iPSC-derived skin, islet, and heart representing three germ layers of the body through 4n complementation and evaluated their immunogenicity and therapeutic efficacy. Upon transplantation into recipient mice, iPSC-derived skin successfully survived and repaired local tissue wounds. In diabetic mouse models, explanted iPSC-derived islets effectively produced insulin and lowered blood glucose to basal levels. iPSC-derived heart grafts maintained normal beating for more than 3 months in syngeneic recipients. Importantly, no obvious immune rejection responses against iPSC-derived organs were detected long after transplantation. Our study not only demonstrates the fundamental immunogenicity and function of iPSC derivatives, but also provides preclinical evidence to support the feasibility of using iPSC-derived skin, islet, and heart for therapeutic use.
Collapse
|
243
|
Girlovanu M, Susman S, Soritau O, Rus-Ciuca D, Melincovici C, Constantin AM, Mihu CM. Stem cells - biological update and cell therapy progress. ACTA ACUST UNITED AC 2015; 88:265-71. [PMID: 26609255 PMCID: PMC4632881 DOI: 10.15386/cjmed-483] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 06/12/2015] [Indexed: 12/17/2022]
Abstract
In recent years, the advances in stem cell research have suggested that the human body may have a higher plasticity than it was originally expected. Until now, four categories of stem cells were isolated and cultured in vivo: embryonic stem cells, fetal stem cells, adult stem cells and induced pluripotent stem cells (hiPSCs). Although multiple studies were published, several issues concerning the stem cells are still debated, such as: the molecular mechanisms of differentiation, the methods to prevent teratoma formation or the ethical and religious issues regarding especially the embryonic stem cell research. The direct differentiation of stem cells into specialized cells: cardiac myocytes, neural cells, pancreatic islets cells, may represent an option in treating incurable diseases such as: neurodegenerative diseases, type I diabetes, hematologic or cardiac diseases. Nevertheless, stem cell-based therapies, based on stem cell transplantation, remain mainly at the experimental stages and their major limitation is the development of teratoma and cancer after transplantation. The induced pluripotent stem cells (hiPSCs) represent a prime candidate for future cell therapy research because of their significant self-renewal and differentiation potential and the lack of ethical issues. This article presents an overview of the biological advances in the study of stem cells and the current progress made in the field of regenerative medicine.
Collapse
Affiliation(s)
- Mihai Girlovanu
- Morphological Sciences Department 1, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Sergiu Susman
- Morphological Sciences Department 1, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Olga Soritau
- Research Department, Prof. Dr. I. Chiricuta Oncology Institute, Cluj-Napoca, Romania
| | - Dan Rus-Ciuca
- Department of Pathology, Karlstad Central Hospital, Sweden
| | - Carmen Melincovici
- Morphological Sciences Department 1, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Anne-Marie Constantin
- Morphological Sciences Department 1, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Carmen Mihaela Mihu
- Morphological Sciences Department 1, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
244
|
Michelsen KA, Acosta-Verdugo S, Benoit-Marand M, Espuny-Camacho I, Gaspard N, Saha B, Gaillard A, Vanderhaeghen P. Area-specific reestablishment of damaged circuits in the adult cerebral cortex by cortical neurons derived from mouse embryonic stem cells. Neuron 2015; 85:982-97. [PMID: 25741724 DOI: 10.1016/j.neuron.2015.02.001] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 12/18/2014] [Accepted: 01/27/2015] [Indexed: 01/09/2023]
Abstract
Pluripotent stem-cell-derived neurons constitute an attractive source for replacement therapies, but their utility remains unclear for cortical diseases. Here, we show that neurons of visual cortex identity, differentiated in vitro from mouse embryonic stem cells (ESCs), can be transplanted successfully following a lesion of the adult mouse visual cortex. Reestablishment of the damaged pathways included long-range and reciprocal axonal projections and synaptic connections with targets of the damaged cortex. Electrophysiological recordings revealed that some grafted neurons were functional and responsive to visual stimuli. No significant integration was observed following grafting of the same neurons in motor cortex, or transplantation of embryonic motor cortex in visual cortex, indicating that successful transplantation required a match in the areal identity of grafted and lesioned neurons. These findings demonstrate that transplantation of mouse ESC-derived neurons of appropriate cortical areal identity can contribute to the reconstruction of an adult damaged cortical circuit.
Collapse
Affiliation(s)
- Kimmo A Michelsen
- Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB), Campus Erasme, 808 Route de Lennik, 1070 Brussels, Belgium; ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Campus Erasme, 808 Route de Lennik, 1070 Brussels, Belgium
| | - Sandra Acosta-Verdugo
- Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB), Campus Erasme, 808 Route de Lennik, 1070 Brussels, Belgium; ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Campus Erasme, 808 Route de Lennik, 1070 Brussels, Belgium
| | - Marianne Benoit-Marand
- INSERM U1084, Experimental and Clinical Neurosciences Laboratory, Cellular Therapies in Brain Diseases Group, University of Poitiers, 1 rue Georges Bonnet, BP 633, 86022 Poitiers Cedex, France
| | - Ira Espuny-Camacho
- Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB), Campus Erasme, 808 Route de Lennik, 1070 Brussels, Belgium; ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Campus Erasme, 808 Route de Lennik, 1070 Brussels, Belgium
| | - Nicolas Gaspard
- Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB), Campus Erasme, 808 Route de Lennik, 1070 Brussels, Belgium; ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Campus Erasme, 808 Route de Lennik, 1070 Brussels, Belgium
| | - Bhaskar Saha
- INSERM U1084, Experimental and Clinical Neurosciences Laboratory, Cellular Therapies in Brain Diseases Group, University of Poitiers, 1 rue Georges Bonnet, BP 633, 86022 Poitiers Cedex, France
| | - Afsaneh Gaillard
- INSERM U1084, Experimental and Clinical Neurosciences Laboratory, Cellular Therapies in Brain Diseases Group, University of Poitiers, 1 rue Georges Bonnet, BP 633, 86022 Poitiers Cedex, France.
| | - Pierre Vanderhaeghen
- Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB), Campus Erasme, 808 Route de Lennik, 1070 Brussels, Belgium; ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), Campus Erasme, 808 Route de Lennik, 1070 Brussels, Belgium; WELBIO, Université Libre de Bruxelles (ULB), Campus Erasme, 808 Route de Lennik, 1070 Brussels, Belgium.
| |
Collapse
|
245
|
Smith DK, Zhang CL. Regeneration through reprogramming adult cell identity in vivo. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:2619-28. [PMID: 26056931 DOI: 10.1016/j.ajpath.2015.02.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 01/19/2015] [Accepted: 02/18/2015] [Indexed: 12/28/2022]
Abstract
The discovery and in vivo application of cell fate reprogramming concepts have jumpstarted new technologies aimed at the functional regeneration of damaged tissues. As most adult organ systems retain only a limited potential for self-regeneration after trauma, the production of fate-specific cells by in vivo transdifferentiation offers a targeted method for tissue bioengineering. Proof-of-principle studies have demonstrated the induction of neural precursor cells, neurons, cardiomyocytes, and insulin-producing β islet cells. Each of these induced cell types survive, mature, and integrate into the local environment in a functionally meaningful manner. Here, we briefly highlight recent advances in the in vivo reprogramming of cell identity and the current challenges that face the clinical relevance of these methods.
Collapse
Affiliation(s)
- Derek K Smith
- Department of Molecular Biology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chun-Li Zhang
- Department of Molecular Biology, The University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
246
|
Isobe KI, Cheng Z, Nishio N, Suganya T, Tanaka Y, Ito S. Reprint of "iPSCs, aging and age-related diseases". N Biotechnol 2015; 32:169-79. [PMID: 25479728 DOI: 10.1016/j.nbt.2014.11.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Human histocompatibility antigens are quite heterogeneous and promote the rejection of transplanted tissue. Recent advances in stem cell research that enable the use of a patient's own stem cells for transplantation are very important because rejection could be avoided. In particular, Yamanaka’s group in Japan gave new hope to patients with incurable diseases when they developed induced murine pluripotent stem cells (iPSCs) in 2006 and human iPSCs in 2007. Whereas embryonic stem cells (ESCs) are derived from the inner cell mass and are supported in culture by LIF, iPSCs are derived from fetal or adult somatic cells. Through the application of iPSC technology, adult somatic cells can develop a pluripotent state. One advantage of using iPSCs instead of ESCs in regenerative medicine is that (theoretically) immune rejection could be avoided, although there is some debate about immune rejection of a patient's own iPSCs. Many diseases occur in elderly patients. In order to use regenerative medicine with the elderly, it is important to demonstrate that iPSCs can indeed be generated from older patients. Recent findings have shown that iPSCs can be established from aged mice and aged humans. These iPSCs can differentiate to cells from all three germ layers. However, it is not known whether iPSCs from aged mice or humans show early senescence. Before clinical use of iPSCs, issues related to copy number variation, tumorigenicity and immunogenicity must be resolved. It is particularly important that researchers have succeeded in generating iPSCs that have differentiated to somatic cells related to specific diseases of the elderly, including atherosclerosis, diabetes, Alzheimer's disease and Parkinson's disease. These efforts will facilitate the use of personalized stem cell transplantation therapy for currently incurable diseases.
Collapse
Affiliation(s)
- Ken-ichi Isobe
- Department of Immunology, Nagoya University Graduate School of Medicine, 65 Turumai-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan
| | | | | | | | | | | |
Collapse
|
247
|
Salewski RP, Mitchell RA, Li L, Shen C, Milekovskaia M, Nagy A, Fehlings MG. Transplantation of Induced Pluripotent Stem Cell-Derived Neural Stem Cells Mediate Functional Recovery Following Thoracic Spinal Cord Injury Through Remyelination of Axons. Stem Cells Transl Med 2015; 4:743-54. [PMID: 25979861 DOI: 10.5966/sctm.2014-0236] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 03/09/2015] [Indexed: 02/06/2023] Open
Abstract
UNLABELLED : Neural stem cells (NSCs) from embryonic or fetal/adult tissue sources have shown considerable promise in regenerative strategies for traumatic spinal cord injury (SCI). However, there are limitations with their use related to the availability, immunogenicity, and uncertainty of the mechanisms involved. To address these issues, definitive NSCs derived from induced pluripotent stem (iPS) cells generated using a nonviral, piggyBac transposon approach, were investigated. Committed NSCs were generated from iPS cells using a free-floating neurosphere methodology previously described by our laboratory. To delineate the mechanism of action, specifically the role of exogenous myelination, NSCs derived from wildtype (wt) and nonmyelinating Shiverer (shi) iPS cell lines were used following thoracic SCI with subacute intraspinal transplantation. Behavioral, histological, and electrophysiological outcomes were analyzed to assess the effectiveness of this treatment. The wt- and shi-iPS-NSCs were validated and shown to be equivalent except in myelination capacity. Both iPS-NSC lines successfully integrated into the injured spinal cord and predominantly differentiated to oligodendrocytes, but only the wt-iPS-NSC treatment resulted in a functional benefit. The wt-iPS-dNSCs, which exhibited the capacity for remyelination, significantly improved neurobehavioral function (Basso Mouse Scale and CatWalk), histological outcomes, and electrophysiological measures of axonal function (sucrose gap analysis) compared with the nonmyelinating iPS-dNSCs and cell-free controls. In summary, we demonstrated that iPS cells can generate translationally relevant NSCs for applications in SCI. Although NSCs have a diverse range of functions in the injured spinal cord, remyelination is the predominant mechanism of recovery following thoracic SCI. SIGNIFICANCE Gain-of-function/loss-of-function techniques were used to examine the mechanistic importance of graft-derived remyelination following thoracic spinal cord injury (SCI). The novel findings of this study include the first use of neural stem cells (NSCs) from induced pluripotent stem cells (iPSCs) derived using the clonal neurosphere expansion conditions, for the treatment of SCI, the first characterization and in vivo application of iPSCs from Shiverer mouse fibroblasts, and the first evidence of the importance of remyelination by pluripotent-sourced NSCs for SCI repair and regeneration.
Collapse
Affiliation(s)
- Ryan P Salewski
- Division of Genetics and Development, Toronto Western Research Institute, Toronto, Ontario, Canada; Institute of Medical Science, Faculty of Medicine, Department of Obstetrics and Gynaecology, and Division of Neurosurgery, University of Toronto, Toronto, Ontario, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Spinal Program, University Health Network, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Robert A Mitchell
- Division of Genetics and Development, Toronto Western Research Institute, Toronto, Ontario, Canada; Institute of Medical Science, Faculty of Medicine, Department of Obstetrics and Gynaecology, and Division of Neurosurgery, University of Toronto, Toronto, Ontario, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Spinal Program, University Health Network, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Lijun Li
- Division of Genetics and Development, Toronto Western Research Institute, Toronto, Ontario, Canada; Institute of Medical Science, Faculty of Medicine, Department of Obstetrics and Gynaecology, and Division of Neurosurgery, University of Toronto, Toronto, Ontario, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Spinal Program, University Health Network, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Carl Shen
- Division of Genetics and Development, Toronto Western Research Institute, Toronto, Ontario, Canada; Institute of Medical Science, Faculty of Medicine, Department of Obstetrics and Gynaecology, and Division of Neurosurgery, University of Toronto, Toronto, Ontario, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Spinal Program, University Health Network, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Maria Milekovskaia
- Division of Genetics and Development, Toronto Western Research Institute, Toronto, Ontario, Canada; Institute of Medical Science, Faculty of Medicine, Department of Obstetrics and Gynaecology, and Division of Neurosurgery, University of Toronto, Toronto, Ontario, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Spinal Program, University Health Network, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Andras Nagy
- Division of Genetics and Development, Toronto Western Research Institute, Toronto, Ontario, Canada; Institute of Medical Science, Faculty of Medicine, Department of Obstetrics and Gynaecology, and Division of Neurosurgery, University of Toronto, Toronto, Ontario, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Spinal Program, University Health Network, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Michael G Fehlings
- Division of Genetics and Development, Toronto Western Research Institute, Toronto, Ontario, Canada; Institute of Medical Science, Faculty of Medicine, Department of Obstetrics and Gynaecology, and Division of Neurosurgery, University of Toronto, Toronto, Ontario, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Spinal Program, University Health Network, Toronto Western Hospital, Toronto, Ontario, Canada
| |
Collapse
|
248
|
Alvarez Palomo AB, McLenachan S, Chen FK, Da Cruz L, Dilley RJ, Requena J, Lucas M, Lucas A, Drukker M, Edel MJ. Prospects for clinical use of reprogrammed cells for autologous treatment of macular degeneration. FIBROGENESIS & TISSUE REPAIR 2015; 8:9. [PMID: 25984235 PMCID: PMC4432516 DOI: 10.1186/s13069-015-0026-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 04/24/2015] [Indexed: 12/12/2022]
Abstract
Since the discovery of induced pluripotent stem cells (iPSC) in 2006, the symptoms of many human diseases have been reversed in animal models with iPSC therapy, setting the stage for future clinical development. From the animal data it is clear that iPSC are rapidly becoming the lead cell type for cell replacement therapy and for the newly developing field of iPSC-derived body organ transplantation. The first human pathology that might be treated in the near future with iPSC is age-related macular degeneration (AMD), which has recently passed the criteria set down by regulators for phase I clinical trials with allogeneic human embryonic stem cell-derived cell transplantation in humans. Given that iPSC are currently in clinical trial in Japan (RIKEN) to treat AMD, the establishment of a set of international criteria to make clinical-grade iPSC and their differentiated progeny is the next step in order to prepare for future autologous cell therapy clinical trials. Armed with clinical-grade iPSC, we can then specifically test for their threat of cancer, for proper and efficient differentiation to the correct cell type to treat human disease and then to determine their immunogenicity. Such a rigorous approach sets a far more relevant paradigm for their intended future use than non-clinical-grade iPSC. This review focuses on the latest developments regarding the first possible use of iPSC-derived retinal pigment epithelial cells in treating human disease, covers data gathered on animal models to date and methods to make clinical-grade iPSC, suggests techniques to ensure quality control and discusses possible clinical immune responses.
Collapse
Affiliation(s)
- Ana Belen Alvarez Palomo
- Control of Pluripotency Laboratory, Department of Physiological Sciences I, Faculty of Medicine, University of Barcelona, Hospital Clinic, Casanova 143, 08036 Barcelona, Spain
| | - Samuel McLenachan
- Centre for Ophthalmology and Visual Science (Lions Eye Institute), University of Western Australia, 2 Verdun Street, Nedlands, WA 6009 Australia
| | - Fred K Chen
- Centre for Ophthalmology and Visual Science (Lions Eye Institute), University of Western Australia, 2 Verdun Street, Nedlands, WA 6009 Australia
| | - Lyndon Da Cruz
- Moorfields Eye Hospital, 162 City Road, London, EC1V 2PD England
| | - Rodney J Dilley
- Ear Sciences Centre, 1 Salvado Rd, Subiaco, WA 6008 Australia ; School of Surgery, University of Western Australia, 35 Stirling Highway, Nedlands, WA 6009 Australia
| | - Jordi Requena
- Control of Pluripotency Laboratory, Department of Physiological Sciences I, Faculty of Medicine, University of Barcelona, Hospital Clinic, Casanova 143, 08036 Barcelona, Spain
| | - Michaela Lucas
- School of Medicine and Pharmacology, University of Western Australia, 35 Stirling Highway, Nedlands, WA 6009 Australia ; PathWest, SCGH Laboratories Hospital Ave, Nedlands, WA 6009 Australia
| | - Andrew Lucas
- Institute for Immunology and Infectious Diseases, Murdoch University, Building 390, Discovery Way, Murdoch, Perth, WA 6150 Australia
| | - Micha Drukker
- Helmholtz Zentrum München, German Research Centre for Environmental Health (GmbH), Institute of Stem Cell Research, Ingolstädter Landstraße 1, D-85764 Neuherberg, Germany
| | - Michael J Edel
- Control of Pluripotency Laboratory, Department of Physiological Sciences I, Faculty of Medicine, University of Barcelona, Hospital Clinic, Casanova 143, 08036 Barcelona, Spain ; Division of Pediatrics and Child Health, Westmead Children's Hospital, Corner Hawkesbury Road and Hainsworth Street, Westmead, Sydney, NSW 2145 Australia ; School of Anatomy, Physiology & Human Biology and Centre for Cell Therapy and Regenerative Medicine (CCTRM), University of Western Australia, 35 Stirling Highway, Nedlands, WA 6009 Australia
| |
Collapse
|
249
|
Liao W, Huang N, Yu J, Jares A, Yang J, Zieve G, Avila C, Jiang X, Zhang XB, Ma Y. Direct Conversion of Cord Blood CD34+ Cells Into Neural Stem Cells by OCT4. Stem Cells Transl Med 2015; 4:755-63. [PMID: 25972144 DOI: 10.5966/sctm.2014-0289] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 04/08/2015] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED : Cellular reprogramming or conversion is a promising strategy to generate desired stem cell types from somatic cells. Neural stem cells (NSCs) have the potential to regenerate central nervous system tissue and repair damage in response to injury. However, NSCs are difficult to isolate from human tissues and expand in sufficient quantities for therapy. Here, we report a method to generate neural stem cells from cord blood CD34-positive cells by ectopic expression of OCT4 in a feeder-free system. The induced cells (iNSCs) show a characteristic NSC-like morphology and can be expanded in vitro for more than 20 passages. In addition, the iNSCs are positive for neural stem cell-specific markers such as Nestin and Musashi-1 and are similar in gene expression patterns to a human neural stem cell line. The iNSCs express distinct transcriptional factors for forebrain, hindbrain, and spinal cord regions. Upon differentiation, the iNSCs are able to commit into multilineage mature neural cells. Following in vivo introduction into NOD/SCID mice, iNSCs can survive and differentiate in the mouse brain 3 months post-transplantation. Alternatively, we were also able to derive iNSCs with an episomal vector expressing OCT4. Our results suggest a novel, efficient approach to generate neural precursor cells that can be potentially used in drug discovery or regenerative medicine for neurological disease and injury. SIGNIFICANCE This study describes a novel method to generate expandable induced neural stem cells from human cord blood cells in a feeder-free system by a single factor, OCT4. The data are promising for future applications that require the generation of large amounts of autologous neural stem cells in disease modeling and regenerative medicine.
Collapse
Affiliation(s)
- Wenbin Liao
- Departments of Pathology, Surgery, and Obstetrics & Gynecology, Stony Brook University Hospital, Stony Brook University, Stony Brook, New York, USA; Department of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Nick Huang
- Departments of Pathology, Surgery, and Obstetrics & Gynecology, Stony Brook University Hospital, Stony Brook University, Stony Brook, New York, USA; Department of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Jingxia Yu
- Departments of Pathology, Surgery, and Obstetrics & Gynecology, Stony Brook University Hospital, Stony Brook University, Stony Brook, New York, USA; Department of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Alexander Jares
- Departments of Pathology, Surgery, and Obstetrics & Gynecology, Stony Brook University Hospital, Stony Brook University, Stony Brook, New York, USA; Department of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Jianchang Yang
- Departments of Pathology, Surgery, and Obstetrics & Gynecology, Stony Brook University Hospital, Stony Brook University, Stony Brook, New York, USA; Department of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Gary Zieve
- Departments of Pathology, Surgery, and Obstetrics & Gynecology, Stony Brook University Hospital, Stony Brook University, Stony Brook, New York, USA; Department of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Cecilia Avila
- Departments of Pathology, Surgery, and Obstetrics & Gynecology, Stony Brook University Hospital, Stony Brook University, Stony Brook, New York, USA; Department of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Xun Jiang
- Departments of Pathology, Surgery, and Obstetrics & Gynecology, Stony Brook University Hospital, Stony Brook University, Stony Brook, New York, USA; Department of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Xiao-Bing Zhang
- Departments of Pathology, Surgery, and Obstetrics & Gynecology, Stony Brook University Hospital, Stony Brook University, Stony Brook, New York, USA; Department of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Yupo Ma
- Departments of Pathology, Surgery, and Obstetrics & Gynecology, Stony Brook University Hospital, Stony Brook University, Stony Brook, New York, USA; Department of Medicine, Loma Linda University, Loma Linda, California, USA
| |
Collapse
|
250
|
Donadeu FX. Equine induced pluripotent stem cells or how to turn skin cells into neurons: horse tissues a la carte? Equine Vet J 2015; 46:534-7. [PMID: 25099189 DOI: 10.1111/evj.12300] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- F X Donadeu
- The Roslin Institute and Royal Dick School of Veterinary Sciences, University of Edinburgh, UK
| |
Collapse
|