1
|
Wang Y, Wang S, Wu H, Liu X, Ma J, Khan MA, Riaz A, Wang L, Qiu HJ, Sun Y. Compartmentalized Neuronal Culture for Viral Transport Research. Front Microbiol 2020; 11:1470. [PMID: 32760359 PMCID: PMC7373733 DOI: 10.3389/fmicb.2020.01470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/05/2020] [Indexed: 01/12/2023] Open
Abstract
Neuron-invading viruses usually enter via the peripheral organs/tissues of their mammalian hosts and are transported to the neurons. Virus trafficking is critical for transport or spread within the nervous system. Primary culture of neurons is a valuable and indispensable method for neurobiological research, allowing researchers to investigate basic mechanisms of diverse neuronal functions as well as retrograde and anterograde virus transport in neuronal axons. Primary ganglion sensory neurons from mice can be cultured in a compartmentalized culture device, which allows spatial fluidic separation of cell bodies and distal axons. These neurons serve as an important model for investigating the transport of viruses between the neuronal soma and distal axons. Alphaherpesviruses are fascinating and important human and animal pathogens, they replicate and establish lifelong latent infection in the peripheral nervous system, the mechanism of the viral transport along the axon is the key to understand the virus spread in the nervous system. In this review, we briefly introduce and evaluate the most frequently used compartmentalization tools in viral transport research, with particular emphasis on alphaherpesviruses.
Collapse
Affiliation(s)
- Yimin Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Shan Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Hongxia Wu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xinxin Liu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Jinyou Ma
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Muhammad Akram Khan
- Department of Veterinary Pathology, Faculty of Veterinary and Animal Science, PMAS-Arid Agriculture University, Rawalpindi, Pakistan
| | - Aayesha Riaz
- Department of Parasitology and Microbiology, Faculty of Veterinary and Animal Science, PMAS-Arid Agriculture University, Rawalpindi, Pakistan
| | - Lei Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Hua-Ji Qiu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yuan Sun
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
2
|
Guo X, Gu X, Hareshwaree S, Rong X, Li L, Chu M. Induced pluripotent stem cell-conditional medium inhibits H9C2 cardiomyocytes apoptosis via autophagy flux and Wnt/β-catenin pathway. J Cell Mol Med 2019; 23:4358-4374. [PMID: 30957422 PMCID: PMC6533467 DOI: 10.1111/jcmm.14327] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/11/2019] [Accepted: 03/21/2019] [Indexed: 12/13/2022] Open
Abstract
Induced pluripotent stem cell‐derived conditioned medium (iPS‐CM) could improve cell viability in many types of cells and may be a better alternative for the treatment of myocardial infarction. This study aimed to examine the influence of iPS‐CM on anti‐apoptosis and the proliferation of H9C2 cardiomyocytes and investigate the underlying mechanisms. H9C2 cardiomyocytes were exposed to 200 μmol/L hydrogen peroxide (H2O2) for 24 hours with or without pre‐treatment with iPS‐CM. The ratio of apoptotic cells, the loss of mitochondrial membrane potential (△Ψm) and the levels of intracellular reactive oxygen species were analysed by flow cytometric analysis. The expression levels of BCL‐2 and BAX proteins were analysed by Western blot. Cell proliferation was assessed using cell cycle and EdU staining assays. To study cell senescence, senescence‐associated β‐galactosidase (SA‐β‐gal) staining was conducted. The levels of malondialdehyde, superoxide dismutase and glutathione were also quantified using commercially available enzymatic kits. The results showed that iPS‐CM containing basic fibroblast growth factor significantly reduced H2O2‐induced H9C2 cardiomyocyte apoptosis by activating the autophagy flux pathway, promoted cardiomyocyte proliferation by up‐regulating the Wnt/β‐catenin pathway and inhibited oxidative stress and cell senescence. In conclusion, iPS‐CM effectively enhanced the cell viability of H9C2 cardiomyocytes and could potentially be used to inhibit cardiomyocytes apoptosis to treat myocardial infarction in the future.
Collapse
Affiliation(s)
- Xiaoling Guo
- Center of Scientific Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaohong Gu
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Sohun Hareshwaree
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xing Rong
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lei Li
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Maoping Chu
- Center of Scientific Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
3
|
Guo X, Chen Y, Hong T, Chen X, Duan Y, Li C, Ge R. Induced pluripotent stem cell-derived conditional medium promotes Leydig cell anti-apoptosis and proliferation via autophagy and Wnt/β-catenin pathway. J Cell Mol Med 2018; 22:3614-3626. [PMID: 29667777 PMCID: PMC6010900 DOI: 10.1111/jcmm.13641] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 03/14/2018] [Indexed: 12/14/2022] Open
Abstract
Leydig cell transplantation is a better alternative in the treatment of androgen-deficient males. The main purpose of this study was to investigate the effects of induced pluripotent stem cell-derived conditioned medium (iPS-CM) on the anti-apoptosis, proliferation and function of immature Leydig cells (ILCs), and illuminate the underlying mechanisms. ILCs were exposed to 200 μmol/L hydrogen peroxide (H2 O2 ) for 24 hours with or without iPS-CM treatments. Cell apoptosis was detected by flow cytometric analysis. Cell proliferation was assessed using cell cycle assays and EdU staining. The steroidogenic enzyme expressions were quantified with Western blotting. The results showed that iPS-CM significantly reduced H2 O2 -induced ILC apoptosis through down-regulation of autophagic and apoptotic proteins LC3-I/II, Beclin-1, P62, P53 and BAX as well as up-regulation of BCL-2, which could be inhibited by LY294002 (25 μmol/L). iPS-CM could also promote ILC proliferation through up-regulation of β-catenin and its target proteins cyclin D1, c-Myc and survivin, but was inhibited by XAV939 (10 μmol/L). The level of bFGF in iPS-CM was higher than that of DMEM-LG. Exogenous bFGF (20 ng/mL) or Wnt signalling agonist lithium chloride (LiCl) (20 mmol/L) added into DMEM-LG could achieve the similar effects of iPS-CM. Meanwhile, iPS-CM could improve the medium testosterone levels and up-regulation of LHCGR, SCARB1, STAR, CYP11A1, HSD3B1, CYP17A1, HSD17B3 and SF-1 in H2 O2 -induced ILCs. In conclusion, iPS-CM could reduce H2 O2 -induced ILC apoptosis through the activation of autophagy, promote proliferation through up-regulation of Wnt/β-catenin pathway and enhance testosterone production through increasing steroidogenic enzyme expressions, which might be used in regenerative medicine for future.
Collapse
Affiliation(s)
- Xiaoling Guo
- Center of Scientific ResearchThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Yong Chen
- Department of AnesthesiologyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Tingting Hong
- Center of Scientific ResearchThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Xianwu Chen
- Center of Scientific ResearchThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Yue Duan
- Center of Scientific ResearchThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Chao Li
- Center of Scientific ResearchThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Renshan Ge
- Center of Scientific ResearchThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
- Department of AnesthesiologyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| |
Collapse
|
4
|
Meyer K, Kaspar BK. Glia-neuron interactions in neurological diseases: Testing non-cell autonomy in a dish. Brain Res 2017; 1656:27-39. [PMID: 26778174 PMCID: PMC4939136 DOI: 10.1016/j.brainres.2015.12.051] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 12/17/2015] [Accepted: 12/22/2015] [Indexed: 12/30/2022]
Abstract
For the past century, research on neurological disorders has largely focused on the most prominently affected cell types - the neurons. However, with increasing knowledge of the diverse physiological functions of glial cells, their impact on these diseases has become more evident. Thus, many conditions appear to have more complex origins than initially thought. Since neurological pathologies are often sporadic with unknown etiology, animal models are difficult to create and might only reflect a small portion of patients in which a mutation in a gene has been identified. Therefore, reliable in vitro systems to studying these disorders are urgently needed. They might be a pre-requisite for improving our understanding of the disease mechanisms as well as for the development of potential new therapies. In this review, we will briefly summarize the function of different glial cell types in the healthy central nervous system (CNS) and outline their implication in the development or progression of neurological conditions. We will then describe different types of culture systems to model non-cell autonomous interactions in vitro and evaluate advantages and disadvantages. This article is part of a Special Issue entitled SI: Exploiting human neurons.
Collapse
Affiliation(s)
- Kathrin Meyer
- The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Brian K Kaspar
- The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Molecular, Cellular & Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
5
|
Al-Ali H, Beckerman SR, Bixby JL, Lemmon VP. In vitro models of axon regeneration. Exp Neurol 2016; 287:423-434. [PMID: 26826447 DOI: 10.1016/j.expneurol.2016.01.020] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 01/20/2016] [Accepted: 01/25/2016] [Indexed: 12/31/2022]
Abstract
A variety of in vitro models have been developed to understand the mechanisms underlying the regenerative failure of central nervous system (CNS) axons, and to guide pre-clinical development of regeneration-promoting therapeutics. These range from single-cell based assays that typically focus on molecular mechanisms to organotypic assays that aim to recapitulate in vivo behavior. By utilizing a combination of models, researchers can balance the speed, convenience, and mechanistic resolution of simpler models with the biological relevance of more complex models. This review will discuss a number of models that have been used to build our understanding of the molecular mechanisms of CNS axon regeneration.
Collapse
Affiliation(s)
- Hassan Al-Ali
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Samuel R Beckerman
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - John L Bixby
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Center for Computational Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Molecular & Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Vance P Lemmon
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Center for Computational Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
6
|
Effects of induced pluripotent stem cells-derived conditioned medium on the proliferation and anti-apoptosis of human adipose-derived stem cells. Mol Cell Biochem 2016; 413:69-85. [DOI: 10.1007/s11010-015-2640-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 12/23/2015] [Indexed: 01/09/2023]
|
7
|
Hodgetts SI, Edel M, Harvey AR. The State of Play with iPSCs and Spinal Cord Injury Models. J Clin Med 2015; 4:193-203. [PMID: 26237027 PMCID: PMC4470248 DOI: 10.3390/jcm4010193] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 12/08/2014] [Indexed: 01/10/2023] Open
Abstract
The application of induced pluripotent stem cell (iPSC) technologies in cell based strategies, for the repair of the central nervous system (with particular focus on the spinal cord), is moving towards the potential use of clinical grade donor cells. The ability of iPSCs to generate donor neuronal, glial and astrocytic phenotypes for transplantation is highlighted here, and we review recent research using iPSCs in attempts to treat spinal cord injury in various animal models. Also discussed are issues relating to the production of clinical grade iPSCs, recent advances in transdifferentiation protocols for iPSC-derived donor cell populations, concerns about tumourogenicity, and whether iPSC technologies offer any advantages over previous donor cell candidates or tissues already in use as therapeutic tools in experimental spinal cord injury studies.
Collapse
Affiliation(s)
- Stuart I Hodgetts
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Western Australia 6009, Australia.
| | - Michael Edel
- Control of Pluripotency Laboratory, Department of Physiological Sciences I, Faculty of Medicine, University of Barcelona, Hospital Clinic, Casanova 143, Barcelona 08036, Spain.
- Faculty of Medicine, The University of Sydney Medical School, Division of Pediatrics and Child Health, Westmead Children's Hospital, Sydney 2010, Australia.
- School of Anatomy, Physiology and Human Biology, and the Harry Perkins Institute for Medical Research (CCTRM), The University of Western Australia, Western Australia 6009, Australia.
| | - Alan R Harvey
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Western Australia 6009, Australia.
| |
Collapse
|
8
|
Nakamura M, Kamishibahara Y, Kitazawa A, Kawaguchi H, Shimizu N. Differentiation patterns of mouse embryonic stem cells and induced pluripotent stem cells into neurons. Cytotechnology 2014; 68:409-17. [PMID: 25354731 DOI: 10.1007/s10616-014-9792-2] [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: 05/21/2014] [Accepted: 09/27/2014] [Indexed: 10/24/2022] Open
Abstract
Mouse embryonic stem (ES) cells and induced pluripotent stem (iPS) cells have the ability to differentiate in vitro into various cell lineages including neurons. The differentiation of these cells into neurons has potential applications in regenerative medicine. Previously, we reported that a chick dorsal root ganglion (DRG)-conditioned medium (CM) promoted the differentiation of mouse ES and iPS cells into neurons. Here, we used real-time PCR to investigate the differentiation patterns of ES and iPS cells into neurons when DRG-CM was added. DRG-CM promoted the expression levels of βIII-tubulin gene (a marker of postmitotic neurons) in ES and iPS cells. ES cells differentiated into neurons faster than iPS cells, and the maximum peaks of gene expression involved in motor, sensory, and dopaminergic neurons were different. Rho kinase (ROCK) inhibitors could be very valuable at numerous stages in the production and use of stem cells in basic research and eventual cell-based therapies. Thus, we investigated whether the addition of a ROCK inhibitor Y-27632 and DRG-CM on the basis of the differentiation patterns promotes the neuronal differentiation of ES cells. When the ROCK inhibitor was added to the culture medium at the initial stages of cultivation, it stimulated the neuronal differentiation of ES cells more strongly than that stimulated by DRG-CM. Moreover, the combination of the ROCK inhibitor and DRG-CM promoted the neuronal differentiation of ES cells when the ROCK inhibitor was added to the culture medium at day 3. The ROCK inhibitor may be useful for promoting neuronal differentiation of ES cells.
Collapse
Affiliation(s)
- Mai Nakamura
- Graduate School of Life Sciences, Toyo University, 1-1-1 Izumino, Itakura-machi, Ora-gun, Gunma, 374-0193, Japan
| | - Yu Kamishibahara
- Graduate School of Life Sciences, Toyo University, 1-1-1 Izumino, Itakura-machi, Ora-gun, Gunma, 374-0193, Japan
| | - Ayako Kitazawa
- Graduate School of Life Sciences, Toyo University, 1-1-1 Izumino, Itakura-machi, Ora-gun, Gunma, 374-0193, Japan.,Bio-Nano Electronics Research Center, Toyo University, 2100 Kujirai, Kawagoe-shi, Saitama, 350-8585, Japan
| | - Hideo Kawaguchi
- Graduate School of Life Sciences, Toyo University, 1-1-1 Izumino, Itakura-machi, Ora-gun, Gunma, 374-0193, Japan
| | - Norio Shimizu
- Graduate School of Life Sciences, Toyo University, 1-1-1 Izumino, Itakura-machi, Ora-gun, Gunma, 374-0193, Japan. .,Bio-Nano Electronics Research Center, Toyo University, 2100 Kujirai, Kawagoe-shi, Saitama, 350-8585, Japan.
| |
Collapse
|
9
|
Kamishibahara Y, Kawaguchi H, Shimizu N. Promotion of mouse embryonic stem cell differentiation by Rho kinase inhibitor Y-27632. Neurosci Lett 2014; 579:58-63. [PMID: 25038419 DOI: 10.1016/j.neulet.2014.07.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 07/07/2014] [Accepted: 07/09/2014] [Indexed: 01/15/2023]
Abstract
Rho kinase (ROCK) is one of the major downstream mediators of Rho. Rho plays crucial regulatory roles in the cellular proliferation and differentiation. Because a ROCK inhibitor, Y-27632, is known to inhibit the dissociation-induced cell death in human embryonic stem (ES) cells, we investigated the effects of this ROCK inhibitor on the differentiation of the mouse ES cells. The ROCK inhibitor promoted the differentiation of the ES cells into neurons, particularly motor and sensory neurons. The addition of both ROCK inhibitor and nerve growth factor (NGF) strongly stimulated the differentiation of the ES cells into neurons. Moreover, the ROCK inhibitor promoted the differentiation of the ES cells into muscle cells. The ES cells primarily differentiated into neurons rather than muscle cells. We found that the ROCK inhibitor may promote the neuronal differentiation of the ES cells by activating the extracellular signal-regulated kinase (ERK) signaling pathway. These results suggest that the ROCK inhibitor has a significant potential to regulate the differentiation of the ES cells.
Collapse
Affiliation(s)
- Yu Kamishibahara
- Graduate School of Life Sciences, Toyo University, 1-1-1 Izumino, Itakura-machi, Ora-gun, Gunma 374-0193, Japan.
| | - Hideo Kawaguchi
- Graduate School of Life Sciences, Toyo University, 1-1-1 Izumino, Itakura-machi, Ora-gun, Gunma 374-0193, Japan.
| | - Norio Shimizu
- Graduate School of Life Sciences, Toyo University, 1-1-1 Izumino, Itakura-machi, Ora-gun, Gunma 374-0193, Japan; Bio-Nano Electronics Research Center, Toyo University, 2100 Kujirai, Kawagoe-shi, Saitama 350-8585, Japan.
| |
Collapse
|
10
|
Induction of pluripotency. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 786:5-25. [PMID: 23696349 DOI: 10.1007/978-94-007-6621-1_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
The molecular and phenotypic irreversibility of mammalian cell differentiation was a fundamental principle of developmental biology at least until the 1980s, despite numerous reports dating back to the 1950s of the induction of pluripotency in amphibian cells by nuclear transfer (NT). Landmark reports in the 1980s and 1990s in sheep progressively challenged this dogmatic assumption; firstly, embryonic development of reconstructed embryos comprising whole (donor) blastomeres fused to enucleated oocytes, and famously, the cloning of Dolly from a terminally differentiated cell. Thus, the intrinsic ability of oocyte-derived factors to reverse the differentiated phenotype was confirmed. The concomitant elucidation of methods for human embryonic stem cell isolation and cultivation presented opportunities for therapeutic cell replacement strategies, particularly through NT of patient nuclei to enucleated oocytes for subsequent isolation of patient-specific (autologous), pluripotent cells from the resulting blastocysts. Associated logistical limitations of working with human oocytes, in addition to ethical and moral objections prompted exploration of alternative approaches to generate autologous stem cells for therapy, utilizing the full repertoire of factors characteristic of pluripotency, primarily through cell fusion and use of pluripotent cell extracts. Stunningly, in 2006, Japanese scientists described somatic cell reprogramming through delivery of four key factors (identified through a deductive approach from 24 candidate genes). Although less efficient than previous approaches, much of current stem cell research adopts this focused approach to cell reprogramming and (autologous) cell therapy. This chapter is a quasi-historical commentary of the various aforementioned approaches for the induction of pluripotency in lineage-committed cells, and introduces transcriptional and epigenetic changes occurring during reprogramming.
Collapse
|
11
|
Wang J, Chao F, Han F, Zhang G, Xi Q, Li J, Jiang H, Wang J, Yu G, Tian M, Zhang H. PET demonstrates functional recovery after transplantation of induced pluripotent stem cells in a rat model of cerebral ischemic injury. J Nucl Med 2013; 54:785-92. [PMID: 23503731 DOI: 10.2967/jnumed.112.111112] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
UNLABELLED The purpose of this study was to determine the functionality of the transplanted induced pluripotent stem cells (iPSCs) and embryonic stem cells (ESCs) in a rat model of cerebral ischemia with use of (18)F-FDG small-animal PET imaging. METHODS Middle cerebral artery occlusion was used to establish cerebral ischemia. Twenty-four male rats were randomly assigned to 1 of 3 groups: iPSC treatment, ESC treatment, and the control phosphate-buffered saline (PBS) injection. After neurologic function tests and baseline (18)F-FDG small-animal PET had been performed, 1.0 × 10(6) suspended iPSCs or ESCs were injected stereotactically into the left lateral ventricle. The treatment response was evaluated weekly by (18)F-FDG PET scans and neurologic function tests. Histologic analyses and autoradiographic imaging were performed 4 wk after stem cell transplantation. RESULTS Compared with the PBS injection group, higher (18)F-FDG accumulation in the ipsilateral cerebral infarction was observed in both the iPSC and the ESC treatment groups during the 4-wk period (P < 0.05). (18)F-FDG accumulation in the ipsilateral cerebral infarction increased steadily over time in the iPSC treatment group. At 1 and 2 wk after stem cell transplantation, significant recovery of glucose metabolism was found in the ESC treatment group (P < 0.05) and then decreased gradually. The neurologic score in both stem cell-treated groups was significantly lower than that in the PBS group, indicating functional improvement. Immunohistochemical analysis demonstrated that transplanted stem cells survived and migrated close to the ischemic region, and most of the stem cells expressed protein markers for cells of interest. CONCLUSION (18)F-FDG small-animal PET demonstrated metabolic recovery after iPSC and ESC transplantation in the rat model of cerebral ischemia. iPSCs could be considered a potentially better therapeutic approach than ESCs and are worthy of further translational investigation.
Collapse
Affiliation(s)
- Jiachuan Wang
- Department of Nuclear Medicine, Second Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|