1
|
Yao D, Li S, You M, Chen Y, Yan S, Li B, Wang Y. Developmental exposure to nonylphenol leads to depletion of the neural precursor cell pool in the hippocampal dentate gyrus. Chem Biol Interact 2024; 401:111187. [PMID: 39111523 DOI: 10.1016/j.cbi.2024.111187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/24/2024] [Accepted: 08/04/2024] [Indexed: 08/10/2024]
Abstract
Developmental exposure to nonylphenol (NP) results in irreversible impairments of the central nervous system (CNS). The neural precursor cell (NPC) pool located in the subgranular zone (SGZ), a substructure of the hippocampal dentate gyrus, is critical for the development of hippocampal circuits and some hippocampal functions such as learning and memory. However, the effects of developmental exposure to NP on this pool remain unclear. Thus, our aim was to clarify the impacts of developmental exposure to NP on this pool and to explore the potential mechanisms. Animal models of developmental exposure to NP were created by treating Wistar rats with NP during pregnancy and lactation. Our data showed that developmental exposure to NP decreased Sox2-and Ki67-positive cells in the SGZ of offspring. Inhibited activation of Shh signaling and decreased levels of its downstream mediators, E2F1 and cyclins, were also observed in pups developmentally exposed to NP. Moreover, we established the in vitro model in the NE-4C cells, a neural precursor cell line, to further investigate the effect of NP exposure on NPCs and the underlying mechanisms. Purmorphamine, a small purine-derived hedgehog agonist, was used to specifically modulate the Shh signaling. Consistent with the in vivo results, exposure to NP reduced cell proliferation by inhibiting the Shh signaling in NE-4C cells, and purmorphamine alleviated this reduction in cell proliferation by restoring this signaling. Altogether, our findings support the idea that developmental exposure to NP leads to inhibition of the NPC proliferation and the NPC pool depletion in the SGZ located in the dentate gyrus. Furthermore, we also provided the evidence that suppressed activation of Shh signaling may contribute to the effects of developmental exposure to NP on the NPC pool.
Collapse
Affiliation(s)
- Dianqi Yao
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, PR China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, PR China
| | - Siyao Li
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, PR China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, PR China
| | - Mingdan You
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, PR China; School of Public Health, Guizhou Medical University, Guiyang, Guizhou, PR China
| | - Yin Chen
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, PR China
| | - Siyu Yan
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, PR China
| | - Bing Li
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, PR China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, PR China.
| | - Yi Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, PR China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, PR China.
| |
Collapse
|
2
|
Rathore R, Kaul S, Sharma JB, Mathur SR. Exploring the role of SOX 2 and OCT 4 in the pathogenesis of gliomatosis peritonei: the clinicopathological profile of eleven cases. J Turk Ger Gynecol Assoc 2024; 25:66-73. [PMID: 38867687 DOI: 10.4274/jtgga.galenos.2024.2023-9-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024] Open
Abstract
Objective Gliomatosis peritonei (GP) is a rare entity characterized by multiple mature glial tissue implants in association with ovarian teratomas in the peritoneum and omentum. To date, only 100 cases have been published. Not much is known about the origin, clinicopathological profile or prognosis of GP. SOX2 and OCT4 are recently recognized markers of embryonic stem cell differentiation. Here, the role of SOX2 and OCT4 in the pathogenesis of 11 cases of GP are reported and clinicopathological factors are described. Material and Methods This was a retrospective study of six years duration (2017-2022). All the cases of GP were retrieved from archives, the diagnosis was confirmed and clinicopathological factors were noted. Immunohistochemical (IHC) investigation for glial fibrillary acid protein (GFAP) and S100 was noted wherever available. IHC for SOX2 and OCT4 was performed using an avidin-biotin technique. Results There were 11 cases of GP identified. The median age was 29 years and 1/11 cases had nodal gliomatosis as well. There were eight cases of immature teratoma and three cases of mature cystic teratoma. SOX2 was positive in all foci of GP, while OCT4 was negative. These foci were also positive for GFAP and S100. Conclusion A possibility of GP should be considered as a differential, clinically and radiologically, in cases of omental nodularity. Adequate sampling at the time of surgery is essential to rule out metastasis or growing teratoma syndrome. SOX2, a stem cell marker inducing neural differentiation, may play a crucial role in the development of GP in association with other transcription factors.
Collapse
Affiliation(s)
- Ruchi Rathore
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Shreya Kaul
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, New Delhi, India
| | - Jai Bhagwan Sharma
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, New Delhi, India
| | - Sandeep R Mathur
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
3
|
Gancheva MR, Kremer K, Breen J, Arthur A, Hamilton-Bruce A, Thomas P, Gronthos S, Koblar S. Effect of Octamer-Binding Transcription Factor 4 Overexpression on the Neural Induction of Human Dental Pulp Stem Cells. Stem Cell Rev Rep 2024; 20:797-815. [PMID: 38316679 PMCID: PMC10984899 DOI: 10.1007/s12015-024-10678-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2024] [Indexed: 02/07/2024]
Abstract
Stem cell-based therapy is a potential alternative strategy for brain repair, with neural stem cells (NSC) presenting as the most promising candidates. Obtaining sufficient quantities of NSC for clinical applications is challenging, therefore alternative cell types, such as neural crest-derived dental pulp stem cells (DPSC), may be considered. Human DPSC possess neurogenic potential, exerting positive effects in the damaged brain through paracrine effects. However, a method for conversion of DPSC into NSC has yet to be developed. Here, overexpression of octamer-binding transcription factor 4 (OCT4) in combination with neural inductive conditions was used to reprogram human DPSC along the neural lineage. The reprogrammed DPSC demonstrated a neuronal-like phenotype, with increased expression levels of neural markers, limited capacity for sphere formation, and enhanced neuronal but not glial differentiation. Transcriptomic analysis further highlighted the expression of genes associated with neural and neuronal functions. In vivo analysis using a developmental avian model showed that implanted DPSC survived in the developing central nervous system and respond to endogenous signals, displaying neuronal phenotypes. Therefore, OCT4 enhances the neural potential of DPSC, which exhibited characteristics aligning with neuronal progenitors. This method can be used to standardise DPSC neural induction and provide an alternative source of neural cell types.
Collapse
Affiliation(s)
- Maria R Gancheva
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia.
- School of Biological Sciences, Faculty of Science, Engineering and Technology, The University of Adelaide, Adelaide, 5005, Australia.
| | - Karlea Kremer
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia
| | - James Breen
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia
| | - Agnes Arthur
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia
| | - Anne Hamilton-Bruce
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia
- Stroke Research Programme, Basil Hetzel Institute, The Queen Elizabeth Hospital, Central Adelaide Local Health Network, Woodville South, 5011, Australia
| | - Paul Thomas
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia
- South Australian Health and Medical Research Institute, Adelaide, 5000, Australia
| | - Stan Gronthos
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia
- South Australian Health and Medical Research Institute, Adelaide, 5000, Australia
| | - Simon Koblar
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia
| |
Collapse
|
4
|
Marzoog BA. Transcription Factors in Brain Regeneration: A Potential Novel Therapeutic Target. Curr Drug Targets 2024; 25:46-61. [PMID: 38444255 DOI: 10.2174/0113894501279977231210170231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 03/07/2024]
Abstract
Transcription factors play a crucial role in providing identity to each cell population. To maintain cell identity, it is essential to balance the expression of activator and inhibitor transcription factors. Cell plasticity and reprogramming offer great potential for future therapeutic applications, as they can regenerate damaged tissue. Specific niche factors can modify gene expression and differentiate or transdifferentiate the target cell to the required fate. Ongoing research is being carried out on the possibilities of transcription factors in regenerating neurons, with neural stem cells (NSCs) being considered the preferred cells for generating new neurons due to their epigenomic and transcriptome memory. NEUROD1/ASCL1, BRN2, MYTL1, and other transcription factors can induce direct reprogramming of somatic cells, such as fibroblasts, into neurons. However, the molecular biology of transcription factors in reprogramming and differentiation still needs to be fully understood.
Collapse
Affiliation(s)
- Basheer Abdullah Marzoog
- World-Class Research Center, Digital Biodesign and Personalized Healthcare», I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| |
Collapse
|
5
|
Du J, Liu X, Wong CWY, Wong KKY, Yuan Z. Direct cellular reprogramming and transdifferentiation of fibroblasts on wound healing-Fantasy or reality? Chronic Dis Transl Med 2023; 9:191-199. [PMID: 37711868 PMCID: PMC10497843 DOI: 10.1002/cdt3.77] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/15/2023] [Accepted: 05/23/2023] [Indexed: 09/16/2023] Open
Abstract
Induced pluripotent stem cell (iPSC) technology is one of the de novo approaches in regeneration medicine and has led to new research applications for wound healing in recent years. Fibroblasts have attracted wide attention as the first cell line used for differentiation into iPSCs. Researchers have found that fibroblasts can be induced into different types of cells in variable mediums or microenvironments. This indicates the potential "stem" characteristics of fibroblasts in terms of direct cellular reprogramming compared with the iPSC detour. In this review, we described the morphology and biological function of fibroblasts. The stem cell characteristics and activities of fibroblasts, including transdifferentiation into myofibroblasts, osteogenic cells, chondrogenic cells, neurons, and vascular tissue, are discussed. The biological values of fibroblasts are then briefly reviewed. Finally, we discussed the potential applications of fibroblasts in clinical practice.
Collapse
Affiliation(s)
- Juan Du
- Diabetic Foot Diagnosis and Treatment CentreJilin Province People HospitalChangchunJilinChina
| | - Xuelai Liu
- Department of SurgeryCapital Institute of Pediatrics Affiliated Children HospitalBeijingChina
| | - Carol Wing Yan Wong
- Department of Surgery, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
| | - Kenneth Kak Yuen Wong
- Department of Surgery, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
| | - Zhixin Yuan
- Department of Emergency SurgeryJilin Province People HospitalChangchunJilinChina
| |
Collapse
|
6
|
Specific Activation of Yamanaka Factors via HSF1 Signaling in the Early Stage of Zebrafish Optic Nerve Regeneration. Int J Mol Sci 2023; 24:ijms24043253. [PMID: 36834675 PMCID: PMC9961437 DOI: 10.3390/ijms24043253] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/30/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
In contrast to the case in mammals, the fish optic nerve can spontaneously regenerate and visual function can be fully restored 3-4 months after optic nerve injury (ONI). However, the regenerative mechanism behind this has remained unknown. This long process is reminiscent of the normal development of the visual system from immature neural cells to mature neurons. Here, we focused on the expression of three Yamanaka factors (Oct4, Sox2, and Klf4: OSK), which are well-known inducers of induced pluripotent stem (iPS) cells in the zebrafish retina after ONI. mRNA expression of OSK was rapidly induced in the retinal ganglion cells (RGCs) 1-3 h after ONI. Heat shock factor 1 (HSF1) mRNA was most rapidly induced in the RGCs at 0.5 h. The activation of OSK mRNA was completely suppressed by the intraocular injection of HSF1 morpholino prior to ONI. Furthermore, the chromatin immunoprecipitation assay showed the enrichment of OSK genomic DNA bound to HSF1. The present study clearly showed that the rapid activation of Yamanaka factors in the zebrafish retina was regulated by HSF1, and this sequential activation of HSF1 and OSK might provide a key to unlocking the regenerative mechanism of injured RGCs in fish.
Collapse
|
7
|
Li N, Pang Y, Sang J, Sun Y, Hou W. The controversial expression of SOX2 in gastric cancer and its correlation with Helicobacter pylori infection: A meta-analysis. Medicine (Baltimore) 2022; 101:e30886. [PMID: 36221360 PMCID: PMC9542901 DOI: 10.1097/md.0000000000030886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND The expression of sex-determining region Y (SRY)-like high-mobility group (HMG) box 2 (SOX2) in gastric cancer and the prognosis of patients are controversial. This study analyzed the relationship between SOX2 expression and baseline data, clinicopathological parameters, prognosis, and Helicobacter pylori infection in patients with gastric cancer, and provided new supplements for the diagnosis and treatment of gastric cancer. METHODS The articles which reported SOX2 expression in gastric cancer from medical database was collected. The literature search was conducted in PubMed, Google Scholar, Cochrane library, SpringerLink, China National Knowledge Infrastructure, Web of Science, and Wanfang databases, which were written in English and Chinese. RESULTS A total of 32 articles, including 4641 gastric cancer patients. The results showed that SOX2 expression in gastric cancer group was lower than that in the para-cancerous control group (P < .001). Statistical difference was found between the SOX2 expression and differentiation (Well/Moderate vs Poor), TNM stage (I/II vs III/IV), lymphatic invasion (N0 vs N+), edge infiltration (R0 vs R1), and H pylori infection in the pathological parameters. The prognosis analysis showed that the level of SOX2 expression was unrelated to the overall survival of patients (P = .329). No statistical difference was observed between the SOX2 expression and the baseline data of the patients (all P > .05). CONCLUSIONS Although downregulation expression of SOX2 are related to clinicopathological parameters in gastric cancer, which is not correlated with prognosis. This controversy over the expression of SOX2 will provide a new idea for the study of gastric cancer.
Collapse
Affiliation(s)
- Ning Li
- Department of Pathology, Tai’an City Central Hospital, Tai’an, Shandong, China
| | - Yu Pang
- Department of Pathology, Tai’an City Central Hospital, Tai’an, Shandong, China
| | - Jing Sang
- Department of Pathology, Tai’an City Central Hospital, Tai’an, Shandong, China
| | - Yong Sun
- Department of Pediatrics, Tai’an City Central Hospital, Tai’an, Shandong, China
| | - Weiwei Hou
- Department of Pathology, Tai’an City Central Hospital, Tai’an, Shandong, China
- *Correspondence: Weiwei Hou, Department of Pathology, Tai’an City Central Hospital, Tai’an, Shandong 271000, China (e-mail: )
| |
Collapse
|
8
|
Fernandez-Muñoz B, Garcia-Delgado AB, Arribas-Arribas B, Sanchez-Pernaute R. Human Neural Stem Cells for Cell-Based Medicinal Products. Cells 2021; 10:2377. [PMID: 34572024 PMCID: PMC8469920 DOI: 10.3390/cells10092377] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 12/15/2022] Open
Abstract
Neural stem cells represent an attractive tool for the development of regenerative therapies and are being tested in clinical trials for several neurological disorders. Human neural stem cells can be isolated from the central nervous system or can be derived in vitro from pluripotent stem cells. Embryonic sources are ethically controversial and other sources are less well characterized and/or inefficient. Recently, isolation of NSC from the cerebrospinal fluid of patients with spina bifida and with intracerebroventricular hemorrhage has been reported. Direct reprogramming may become another alternative if genetic and phenotypic stability of the reprogrammed cells is ensured. Here, we discuss the advantages and disadvantages of available sources of neural stem cells for the production of cell-based therapies for clinical applications. We review available safety and efficacy clinical data and discuss scalability and quality control considerations for manufacturing clinical grade cell products for successful clinical application.
Collapse
Affiliation(s)
- Beatriz Fernandez-Muñoz
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
| | - Ana Belen Garcia-Delgado
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
| | - Blanca Arribas-Arribas
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Sevilla, 41012 Sevilla, Spain
| | - Rosario Sanchez-Pernaute
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
| |
Collapse
|
9
|
Baklaushev VP, Durov OV, Kalsin VA, Gulaev EV, Kim SV, Gubskiy IL, Revkova VA, Samoilova EM, Melnikov PA, Karal-Ogly DD, Orlov SV, Troitskiy AV, Chekhonin VP, Averyanov AV, Ahlfors JE. Disease modifying treatment of spinal cord injury with directly reprogrammed neural precursor cells in non-human primates. World J Stem Cells 2021; 13:452-469. [PMID: 34136075 PMCID: PMC8176843 DOI: 10.4252/wjsc.v13.i5.452] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/20/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The development of regenerative therapy for human spinal cord injury (SCI) is dramatically restricted by two main challenges: the need for a safe source of functionally active and reproducible neural stem cells and the need of adequate animal models for preclinical testing. Direct reprogramming of somatic cells into neuronal and glial precursors might be a promising solution to the first challenge. The use of non-human primates for preclinical studies exploring new treatment paradigms in SCI results in data with more translational relevance to human SCI. AIM To investigate the safety and efficacy of intraspinal transplantation of directly reprogrammed neural precursor cells (drNPCs). METHODS Seven non-human primates with verified complete thoracic SCI were divided into two groups: drNPC group (n = 4) was subjected to intraspinal transplantation of 5 million drNPCs rostral and caudal to the lesion site 2 wk post injury, and lesion control (n = 3) was injected identically with the equivalent volume of vehicle. RESULTS Follow-up for 12 wk revealed that animals in the drNPC group demonstrated a significant recovery of the paralyzed hindlimb as well as recovery of somatosensory evoked potential and motor evoked potential of injured pathways. Magnetic resonance diffusion tensor imaging data confirmed the intraspinal transplantation of drNPCs did not adversely affect the morphology of the central nervous system or cerebrospinal fluid circulation. Subsequent immunohistochemical analysis showed that drNPCs maintained SOX2 expression characteristic of multipotency in the transplanted spinal cord for at least 12 wk, migrating to areas of axon growth cones. CONCLUSION Our data demonstrated that drNPC transplantation was safe and contributed to improvement of spinal cord function after acute SCI, based on neurological status assessment and neurophysiological recovery within 12 wk after transplantation. The functional improvement described was not associated with neuronal differentiation of the allogeneic drNPCs. Instead, directed drNPCs migration to the areas of active growth cone formation may provide exosome and paracrine trophic support, thereby further supporting the regeneration processes.
Collapse
Affiliation(s)
- Vladimir P Baklaushev
- Biomedical Research, Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA of Russia, Moscow 115682, Moskva, Russia.
| | - Oleg V Durov
- Department of Neurosurgery, Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA, Moscow 115682, Moskva, Russia
| | - Vladimir A Kalsin
- Biomedical Research, Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA of Russia, Moscow 115682, Moskva, Russia
| | - Eugene V Gulaev
- Department of Neurosurgery, Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA, Moscow 115682, Moskva, Russia
| | - Sergey V Kim
- Department of Anesthesiology, N.N.Blokhin Russian Cancer Research Centre, Moscow 115478, Moskva, Russia
| | - Ilya L Gubskiy
- Ilya L Gubskiy, Radiology and Clinical Physiology Scientific Research Center, Federal center of brain research and neurotechnologies of the Federal Medical Biological Agency, Moscow 117997, Russia
| | - Veronika A Revkova
- Biomedical Research, Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA of Russia, Moscow 115682, Moskva, Russia
| | - Ekaterina M Samoilova
- Biomedical Research, Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA of Russia, Moscow 115682, Moskva, Russia
| | - Pavel A Melnikov
- Department of Neurobiology, Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119992, Moskva, Russia
| | - Dzhina D Karal-Ogly
- Department of Primatology, Russian Acad Med Sci, Research Institute of Medical Primatology, Sochi 119992, Sochi, Russia
| | - Sergey V Orlov
- Department of Primatology, Russian Acad Med Sci, Research Institute of Medical Primatology, Sochi 119992, Sochi, Russia
| | - Alexander V Troitskiy
- Department of Vascular Surgery, Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA of Russia, Moscow 115682, Moskva, Russia
| | - Vladimir P Chekhonin
- Department of Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Center for Psychiatry and Narcology, Russia
| | - Alexander V Averyanov
- Biomedical Research, Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA of Russia, Moscow 115682, Moskva, Russia
| | | |
Collapse
|
10
|
Zhang Y, Xie X, Hu J, Afreen KS, Zhang CL, Zhuge Q, Yang J. Prospects of Directly Reprogrammed Adult Human Neurons for Neurodegenerative Disease Modeling and Drug Discovery: iN vs. iPSCs Models. Front Neurosci 2020; 14:546484. [PMID: 33328842 PMCID: PMC7710799 DOI: 10.3389/fnins.2020.546484] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 10/12/2020] [Indexed: 12/11/2022] Open
Abstract
A reliable disease model is critical to the study of specific disease mechanisms as well as for the discovery and development of new drugs. Despite providing crucial insights into the mechanisms of neurodegenerative diseases, translation of this information to develop therapeutics in clinical trials have been unsuccessful. Reprogramming technology to convert adult somatic cells to induced Pluripotent Stem Cells (iPSCs) or directly reprogramming adult somatic cells to induced Neurons (iN), has allowed for the creation of better models to understand the molecular mechanisms and design of new drugs. In recent times, iPSC technology has been commonly used for modeling neurodegenerative diseases and drug discovery. However, several technological challenges have limited the application of iN. As evidence suggests, iN for the modeling of neurodegenerative disorders is advantageous compared to those derived from iPSCs. In this review, we will compare iPSCs and iN models for neurodegenerative diseases and their potential applications in the future.
Collapse
Affiliation(s)
- Ying Zhang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xinyang Xie
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,International Department of The Affiliated High School of South China Normal University (HFI), Guangzhou, China
| | - Jiangnan Hu
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Kazi Sabrina Afreen
- Department of Microbiology & Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Chun-Li Zhang
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, United States
| | - Qichuan Zhuge
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jianjing Yang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
11
|
Xu Z, Su S, Zhou S, Yang W, Deng X, Sun Y, Li L, Li Y. How to reprogram human fibroblasts to neurons. Cell Biosci 2020; 10:116. [PMID: 33062254 PMCID: PMC7549215 DOI: 10.1186/s13578-020-00476-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 09/15/2020] [Indexed: 12/13/2022] Open
Abstract
Destruction and death of neurons can lead to neurodegenerative diseases. One possible way to treat neurodegenerative diseases and damage of the nervous system is replacing damaged and dead neurons by cell transplantation. If new neurons can replace the lost neurons, patients may be able to regain the lost functions of memory, motor, and so on. Therefore, acquiring neurons conveniently and efficiently is vital to treat neurological diseases. In recent years, studies on reprogramming human fibroblasts into neurons have emerged one after another, and this paper summarizes all these studies. Scientists find small molecules and transcription factors playing a crucial role in reprogramming and inducing neuron production. At the same time, both the physiological microenvironment in vivo and the physical and chemical factors in vitro play an essential role in the induction of neurons. Therefore, this paper summarized and analyzed these relevant factors. In addition, due to the unique advantages of physical factors in the process of reprogramming human fibroblasts into neurons, such as safe and minimally invasive, it has a more promising application prospect. Therefore, this paper also summarizes some successful physical mechanisms of utilizing fibroblasts to acquire neurons, which will provide new ideas for somatic cell reprogramming.
Collapse
Affiliation(s)
- Ziran Xu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021 People's Republic of China
| | - Shengnan Su
- The Second Hospital of Jilin University, Jilin, Changchun, 130041 China
| | - Siyan Zhou
- Department of Stomatology, The First Hospital of Jilin University, Changchun, 130021 People's Republic of China
| | - Wentao Yang
- Norman Bethune College of Medicine, Jilin University, Changchun, 130021 People's Republic of China
| | - Xin Deng
- Norman Bethune College of Medicine, Jilin University, Changchun, 130021 People's Republic of China
| | - Yingying Sun
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021 People's Republic of China.,Department of Stomatology, The First Hospital of Jilin University, Changchun, 130021 People's Republic of China
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021 People's Republic of China
| | - Yulin Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021 People's Republic of China
| |
Collapse
|
12
|
Holmes ZE, Hamilton DJ, Hwang T, Parsonnet NV, Rinn JL, Wuttke DS, Batey RT. The Sox2 transcription factor binds RNA. Nat Commun 2020; 11:1805. [PMID: 32286318 PMCID: PMC7156710 DOI: 10.1038/s41467-020-15571-8] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 03/18/2020] [Indexed: 01/06/2023] Open
Abstract
Certain transcription factors are proposed to form functional interactions with RNA to facilitate proper regulation of gene expression. Sox2, a transcription factor critical for maintenance of pluripotency and neurogenesis, has been found associated with several lncRNAs, although it is unknown whether these interactions are direct or via other proteins. Here we demonstrate that human Sox2 interacts directly with one of these lncRNAs with high affinity through its HMG DNA-binding domain in vitro. These interactions are primarily with double-stranded RNA in a non-sequence specific fashion, mediated by a similar but not identical interaction surface. We further determined that Sox2 directly binds RNA in mouse embryonic stem cells by UV-cross-linked immunoprecipitation of Sox2 and more than a thousand Sox2-RNA interactions in vivo were identified using fRIP-seq. Together, these data reveal that Sox2 employs a high-affinity/low-specificity paradigm for RNA binding in vitro and in vivo. Some transcription factors have been proposed to functionally interact with RNA to facilitate proper regulation of gene expression. Here the authors demonstrate that human Sox2 interact directly and with high affinity to RNAs through its HMG DNA-binding domain.
Collapse
Affiliation(s)
- Zachariah E Holmes
- Department of Biochemistry, University of Colorado at Boulder, Campus Box 596, Boulder, CO, 80309, USA
| | - Desmond J Hamilton
- Department of Biochemistry, University of Colorado at Boulder, Campus Box 596, Boulder, CO, 80309, USA
| | - Taeyoung Hwang
- Department of Biochemistry, University of Colorado at Boulder, Campus Box 596, Boulder, CO, 80309, USA
| | - Nicholas V Parsonnet
- Department of Biochemistry, University of Colorado at Boulder, Campus Box 596, Boulder, CO, 80309, USA
| | - John L Rinn
- Department of Biochemistry, University of Colorado at Boulder, Campus Box 596, Boulder, CO, 80309, USA.,BioFrontiers Institute, University of Colorado at Boulder, Campus Box 596, Boulder, CO, 80309, USA
| | - Deborah S Wuttke
- Department of Biochemistry, University of Colorado at Boulder, Campus Box 596, Boulder, CO, 80309, USA.
| | - Robert T Batey
- Department of Biochemistry, University of Colorado at Boulder, Campus Box 596, Boulder, CO, 80309, USA.
| |
Collapse
|
13
|
Xia X, Li C, Wang Y, Deng X, Ma Y, Ding L, Zheng J. Reprogrammed astrocytes display higher neurogenic competence, migration ability and cell death resistance than reprogrammed fibroblasts. Transl Neurodegener 2020; 9:6. [PMID: 32071715 PMCID: PMC7011554 DOI: 10.1186/s40035-020-0184-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 01/30/2020] [Indexed: 12/20/2022] Open
Abstract
The direct reprogramming of somatic cells into induced neural progenitor cells (iNPCs) has been envisioned as a promising approach to overcome ethical and clinical issues of pluripotent stem cell transplantation. We previously reported that astrocyte-derived induced pluripotent stem cells (iPSCs) have more tendencies for neuronal differentiation than fibroblast-derived iPSCs. However, the differences of neurogenic potential between astrocyte-derived iNPCs (AiNPCs) and iNPCs from non-neural origins, such as fibroblast-derived iNPCs (FiNPCs), and the underlying mechanisms remain unclear. Our results suggested that AiNPCs exhibited higher differentiation efficiency, mobility and survival capacities, compared to FiNPCs. The whole transcriptome analysis revealed higher activities of TGFβ signaling in AiNPCs, versus FiNPCs, following a similar trend between astrocytes and fibroblasts. The higher neurogenic competence, migration ability, and cell death resistance of AiNPCs could be abrogated using TGFβ signaling inhibitor LY2157299. Hence, our study demonstrates the difference between iNPCs generated from neural and non-neural cells, together with the underlying mechanisms, which, provides valuable information for donor cell selection in the reprogramming approach.
Collapse
Affiliation(s)
- Xiaohuan Xia
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Chunhong Li
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Yi Wang
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Xiaobei Deng
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Yizhao Ma
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Lu Ding
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Jialin Zheng
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China.,2Collaborative Innovation Center for Brain Science, Tongji University, Shanghai, 200092 China.,3Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA.,4Department of Pathology and Microbiology, University of Nebraska Medical Center,, Omaha, NE 68198-5930 USA
| |
Collapse
|
14
|
Han F, Lu P. Future Challenges and Perspectives for Stem Cell Therapy of Neurodegenerative Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1266:141-145. [PMID: 33105500 DOI: 10.1007/978-981-15-4370-8_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Stem cell-based therapy has shown exciting efficacy in pre-clinical studies on different neurodegenerative diseases (NDs). However, no clinically applicable stem-cell-derived neurons are available to the patients with NDs. There exist some obstacles associated with stem cell therapy, which need to be overcome in future clinical studies. In this chapter, more challenges and new strategies will be explored to accelerate the clinical translation of a human embryonic stem cell (hESC)/induced pluripotent stem cell (iPSC)-derived neural cell product to patients with NDs.
Collapse
Affiliation(s)
- Fabin Han
- The Institute for Translational Medicine, Affiliated Second Hospital, Shandong University, Jinan, Shandong, China. .,The Institute for Tissue Engineering and Regenerative Medicine, Liaocheng University/Liaocheng People's Hospital, Liaocheng, Shandong, China.
| | - Paul Lu
- Veterans Administration San Diego Healthcare System, San Diego, CA, USA.,Department of Neurosciences, University of California - San Diego, La Jolla, CA, USA
| |
Collapse
|
15
|
Hussein MNA, Cao X, Elokil AA, Huang S. Characterisation of stem and proliferating cells on the retina and lens of loach Misgurnus anguillicaudatus. JOURNAL OF FISH BIOLOGY 2020; 96:102-110. [PMID: 31674006 DOI: 10.1111/jfb.14189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 10/30/2019] [Indexed: 06/10/2023]
Abstract
The eye of the fish has a lifelong persistent neurogenesis unlike eye of mammals, so it's highly interesting to study retinal neurogenesis and its genetic control to give complete knowledge about the cause of this property in fish in comparison to mammals. We performed fluorescent in situ hybridisation for loach Misgurnus anguillicaudatus bmi1, msi1 and sox2 genes, which are used as an indicator of the sites of multipotent stem cells. Proliferating cell nuclear antigen (PCNA), bromodeoxyuridine (BRDU) and KI67 markers were used as indicators of proliferating cells and glial fibrillary acidic protein (GFAP) immunofluorescence was used for detection of the glial property of cells, as well as, immunohistochemistry detected the role of peroxisome proliferator-activated receptor (PPAR)α and γ in retinal neurogenesis. Our results determined that the lens and the retina of loach M. anguillicaudatus contain proliferative and pluripotent stem cells that have both glial and neuroepithelial properties, which add new cells continuously throughout life even without injury-induced proliferation. The PPARα has an essential function in providing energy supply for retinal neurogenesis more than PPARγ.
Collapse
Affiliation(s)
- Mona N A Hussein
- College of Fisheries, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, China
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Benha University, Benha, Egypt
| | - Xiaojuan Cao
- College of Fisheries, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, China
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, Hubei, China
| | - Abdelmotaleb A Elokil
- College of Fisheries, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, China
- Animal Productions Department, Faculty of Agriculture, Benha University, Benha, Egypt
| | - Songqian Huang
- College of Fisheries, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, China
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Japan
| |
Collapse
|
16
|
Erharter A, Rizzi S, Mertens J, Edenhofer F. Take the shortcut - direct conversion of somatic cells into induced neural stem cells and their biomedical applications. FEBS Lett 2019; 593:3353-3369. [PMID: 31663609 PMCID: PMC6916337 DOI: 10.1002/1873-3468.13656] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 10/12/2019] [Accepted: 10/14/2019] [Indexed: 12/20/2022]
Abstract
Second-generation reprogramming of somatic cells directly into the cell type of interest avoids induction of pluripotency and subsequent cumbersome differentiation procedures. Several recent studies have reported direct conversion of human somatic cells into stably proliferating induced neural stem cells (iNSCs). Importantly, iNSCs are easier, faster, and more cost-efficient to generate than induced pluripotent stem cells (iPSCs), and also have a higher level of clinical safety. Stably, self-renewing iNSCs can be derived from different cellular sources, such as skin fibroblasts and peripheral blood mononuclear cells, and readily differentiate into neuronal and glial lineages that are indistinguishable from their iPSC-derived counterparts or from NSCs isolated from primary tissues. This review focuses on the derivation and characterization of iNSCs and their biomedical applications. We first outline different approaches to generate iNSCs and then discuss the underlying molecular mechanisms. Finally, we summarize the preclinical validation of iNSCs to highlight that these cells are promising targets for disease modeling, autologous cell therapy, and precision medicine.
Collapse
Affiliation(s)
- Anita Erharter
- Department of Molecular Biology & CMBIGenomics, Stem Cell Biology & Regenerative MedicineLeopold‐Franzens‐University InnsbruckAustria
| | - Sandra Rizzi
- Department of Molecular Biology & CMBIGenomics, Stem Cell Biology & Regenerative MedicineLeopold‐Franzens‐University InnsbruckAustria
- Institute of PharmacologyMedical University InnsbruckAustria
| | - Jerome Mertens
- Department of Molecular Biology & CMBIGenomics, Stem Cell Biology & Regenerative MedicineLeopold‐Franzens‐University InnsbruckAustria
| | - Frank Edenhofer
- Department of Molecular Biology & CMBIGenomics, Stem Cell Biology & Regenerative MedicineLeopold‐Franzens‐University InnsbruckAustria
| |
Collapse
|
17
|
Boese AC, Hamblin MH, Lee JP. Neural stem cell therapy for neurovascular injury in Alzheimer's disease. Exp Neurol 2019; 324:113112. [PMID: 31730762 DOI: 10.1016/j.expneurol.2019.113112] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/02/2019] [Accepted: 11/11/2019] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD), the most common form of dementia, is characterized by progressive neurodegeneration leading to severe cognitive decline and eventual death. AD pathophysiology is complex, but neurotoxic accumulation of amyloid-β (Aβ) and hyperphosphorylation of Tau are believed to be main drivers of neurodegeneration in AD. The formation and deposition of Aβ plaques occurs in the brain parenchyma as well as in the cerebral vasculature. Thus, proper blood-brain barrier (BBB) and cerebrovascular functioning are crucial for clearance of Aβ from the brain, and neurovascular dysfunction may be a critical component of AD development. Further, neuroinflammation and dysfunction of angiogenesis, neurogenesis, and neurorestorative capabilities play a role in AD pathophysiology. Currently, there is no effective treatment to prevent or restore loss of brain tissue and cognitive decline in patients with AD. Based on multifactorial and complex pathophysiological cascades in multiple Alzheimer's disease stages, effective AD therapies need to focus on targeting early AD pathology and preserving cerebrovascular function. Neural stem cells (NSCs) participate extensively in mammalian brain homeostasis and repair and exhibit pleiotropic intrinsic properties that likely make them attractive candidates for the treatment of AD. In the review, we summarize the current advances in knowledge regarding neurovascular aspects of AD-related neurodegeneration and discuss multiple actions of NSCs from preclinical studies of AD to evaluate their potential for future clinical treatment of AD.
Collapse
Affiliation(s)
- Austin C Boese
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Milton H Hamblin
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jean-Pyo Lee
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70112, USA.
| |
Collapse
|
18
|
Dorsett KA, Jones RB, Ankenbauer KE, Hjelmeland AB, Bellis SL. Sox2 promotes expression of the ST6Gal-I glycosyltransferase in ovarian cancer cells. J Ovarian Res 2019; 12:93. [PMID: 31610800 PMCID: PMC6792265 DOI: 10.1186/s13048-019-0574-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 09/25/2019] [Indexed: 12/15/2022] Open
Abstract
Background The ST6Gal-I glycosyltransferase, which adds α2–6-linked sialic acids to N-glycosylated proteins is upregulated in a wide range of malignancies including ovarian cancer. Prior studies have shown that ST6Gal-I-mediated sialylation of select surface receptors remodels intracellular signaling to impart cancer stem cell (CSC) characteristics. However, the mechanisms that contribute to ST6Gal-I expression in stem-like cancer cells are poorly understood. Results Herein, we identify the master stem cell transcription factor, Sox2, as a novel regulator of ST6Gal-I expression. Interestingly, SOX2 and ST6GAL1 are located within the same tumor-associated amplicon, 3q26, and these two genes exhibit coordinate gains in copy number across multiple cancers including ~ 25% of ovarian serious adenocarcinomas. In conjunction with genetic co-amplification, our studies suggest that Sox2 directly binds the ST6GAL1 promoter to drive transcription. ST6Gal-I expression is directed by at least four distinct promoters, and we identified the P3 promoter as the predominant promoter utilized by ovarian cancer cells. Chromatin Immunoprecipitation (ChIP) assays revealed that Sox2 binds regions proximal to the P3 promoter. To confirm that Sox2 regulates ST6Gal-I expression, Sox2 was either overexpressed or knocked-down in various ovarian cancer cell lines. Sox2 overexpression induced an increase in ST6Gal-I mRNA and protein, as well as surface α2–6 sialylation, whereas Sox2 knock-down suppressed levels of ST6Gal-I mRNA, protein and surface α2–6 sialylation. Conclusions These data suggest a process whereby SOX2 and ST6GAL1 are coordinately amplified in cancer cells, with the Sox2 protein then binding the ST6GAL1 promoter to further augment ST6Gal-I expression. Our collective results provide new insight into mechanisms that upregulate ST6Gal-I expression in ovarian cancer cells, and also point to the possibility that some of the CSC characteristics commonly attributed to Sox2 may, in part, be mediated through the sialyltransferase activity of ST6Gal-I.
Collapse
Affiliation(s)
- Kaitlyn A Dorsett
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, MCLM 350, 1918 University Boulevard, Birmingham, AL, 35294, USA
| | - Robert B Jones
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, MCLM 350, 1918 University Boulevard, Birmingham, AL, 35294, USA
| | - Katherine E Ankenbauer
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, MCLM 350, 1918 University Boulevard, Birmingham, AL, 35294, USA
| | - Anita B Hjelmeland
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, MCLM 350, 1918 University Boulevard, Birmingham, AL, 35294, USA
| | - Susan L Bellis
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, MCLM 350, 1918 University Boulevard, Birmingham, AL, 35294, USA.
| |
Collapse
|
19
|
Gancheva MR, Kremer KL, Gronthos S, Koblar SA. Using Dental Pulp Stem Cells for Stroke Therapy. Front Neurol 2019; 10:422. [PMID: 31110489 PMCID: PMC6501465 DOI: 10.3389/fneur.2019.00422] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 04/08/2019] [Indexed: 12/26/2022] Open
Abstract
Stroke is a leading cause of permanent disability world-wide, but aside from rehabilitation, there is currently no clinically-proven pharmaceutical or biological agent to improve neurological disability. Cell-based therapies using stem cells, such as dental pulp stem cells, are a promising alternative for treatment of neurological diseases, including stroke. The ischaemic environment in stroke affects multiple cell populations, thus stem cells, which act through cellular and molecular mechanisms, are promising candidates. The most common stem cell population studied in the neurological setting has been mesenchymal stem cells due to their accessibility. However, it is believed that neural stem cells, the resident stem cell of the adult brain, would be most appropriate for brain repair. Using reprogramming strategies, alternative sources of neural stem and progenitor cells have been explored. We postulate that a cell of closer origin to the neural lineage would be a promising candidate for reprogramming and modification towards a neural stem or progenitor cell. One such candidate population is dental pulp stem cells, which reside in the root canal of teeth. This review will focus on the neural potential of dental pulp stem cells and their investigations in the stroke setting to date, and include an overview on the use of different sources of neural stem cells in preclinical studies and clinical trials of stroke.
Collapse
Affiliation(s)
- Maria R. Gancheva
- Stroke Research Programme Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Karlea L. Kremer
- Stroke Research Programme Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Stan Gronthos
- Mesenchymal Stem Cell Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Simon A. Koblar
- Stroke Research Programme Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Central Adelaide Local Health Network, Adelaide, SA, Australia
| |
Collapse
|
20
|
Baklaushev VP, Bogush VG, Kalsin VA, Sovetnikov NN, Samoilova EM, Revkova VA, Sidoruk KV, Konoplyannikov MA, Timashev PS, Kotova SL, Yushkov KB, Averyanov AV, Troitskiy AV, Ahlfors JE. Tissue Engineered Neural Constructs Composed of Neural Precursor Cells, Recombinant Spidroin and PRP for Neural Tissue Regeneration. Sci Rep 2019; 9:3161. [PMID: 30816182 PMCID: PMC6395623 DOI: 10.1038/s41598-019-39341-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 01/17/2019] [Indexed: 02/07/2023] Open
Abstract
We have designed a novel two-component matrix (SPRPix) for the encapsulation of directly reprogrammed human neural precursor cells (drNPC). The matrix is comprised of 1) a solid anisotropic complex scaffold prepared by electrospinning a mixture of recombinant analogues of the spider dragline silk proteins - spidroin 1 (rS1/9) and spidroin 2 (rS2/12) - and polycaprolactone (PCL) (rSS-PCL), and 2) a "liquid matrix" based on platelet-rich plasma (PRP). The combination of PRP and spidroin promoted drNPC proliferation with the formation of neural tissue organoids and dramatically activated neurogenesis. Differentiation of drNPCs generated large numbers of βIII-tubulin and MAP2 positive neurons as well as some GFAP-positive astrocytes, which likely had a neuronal supporting function. Interestingly the SPRPix microfibrils appeared to provide strong guidance cues as the differentiating neurons oriented their processes parallel to them. Implantation of the SPRPix matrix containing human drNPC into the brain and spinal cord of two healthy Rhesus macaque monkeys showed good biocompatibility: no astroglial and microglial reaction was present around the implanted construct. Importantly, the human drNPCs survived for the 3 month study period and differentiated into MAP2 positive neurons. Tissue engineered constructs based on SPRPix exhibits important attributes that warrant further examination in spinal cord injury treatment.
Collapse
Affiliation(s)
- V P Baklaushev
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies FMBA of Russia 28 Orekhovy Blvd., 115682, Moscow, Russia.
| | - V G Bogush
- Scientific Center "Kurchatov Institute" - Research Institute for Genetics and Selection of Industrial Microorganisms", 1-st Dorozhniy pr., 1, 117545, Moscow, Russia
| | - V A Kalsin
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies FMBA of Russia 28 Orekhovy Blvd., 115682, Moscow, Russia
| | - N N Sovetnikov
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies FMBA of Russia 28 Orekhovy Blvd., 115682, Moscow, Russia
| | - E M Samoilova
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies FMBA of Russia 28 Orekhovy Blvd., 115682, Moscow, Russia
| | - V A Revkova
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies FMBA of Russia 28 Orekhovy Blvd., 115682, Moscow, Russia
| | - K V Sidoruk
- Scientific Center "Kurchatov Institute" - Research Institute for Genetics and Selection of Industrial Microorganisms", 1-st Dorozhniy pr., 1, 117545, Moscow, Russia
| | - M A Konoplyannikov
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies FMBA of Russia 28 Orekhovy Blvd., 115682, Moscow, Russia
- Institute for Regenerative Medicine, I. M. Sechenov First Moscow State Medical University, 8 Trubetskaya St., 119991, Moscow, Russia
| | - P S Timashev
- Federal Research Center "Crystallography and Photonics", Institute of Photonic Technology of the Russian Academy of Sciences, 2 Pionerskaya St., Troitsk, 142190, Moscow, Russia
- Institute for Regenerative Medicine, I. M. Sechenov First Moscow State Medical University, 8 Trubetskaya St., 119991, Moscow, Russia
- N.N.Semenov Institute of Chemical Physics, 4 Kosygin St., 119991, Moscow, Russia
| | - S L Kotova
- Institute for Regenerative Medicine, I. M. Sechenov First Moscow State Medical University, 8 Trubetskaya St., 119991, Moscow, Russia
- N.N.Semenov Institute of Chemical Physics, 4 Kosygin St., 119991, Moscow, Russia
| | - K B Yushkov
- National University of Science and Technology "MISIS", 4 Leninsky Prospekt, 119049, Moscow, Russia
| | - A V Averyanov
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies FMBA of Russia 28 Orekhovy Blvd., 115682, Moscow, Russia
| | - A V Troitskiy
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies FMBA of Russia 28 Orekhovy Blvd., 115682, Moscow, Russia
| | - J-E Ahlfors
- New World Laboratories Inc., Laval, Quebec, Canada.
| |
Collapse
|
21
|
McCaughey-Chapman A, Connor B. Human Cortical Neuron Generation Using Cell Reprogramming: A Review of Recent Advances. Stem Cells Dev 2018; 27:1674-1692. [DOI: 10.1089/scd.2018.0122] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Amy McCaughey-Chapman
- Department of Pharmacology and Clinical Pharmacology, Centre for Brain Research, School of Medical Science, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Bronwen Connor
- Department of Pharmacology and Clinical Pharmacology, Centre for Brain Research, School of Medical Science, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
22
|
Connor B, Firmin E, McCaughey-Chapman A, Monk R, Lee K, Liot S, Geiger J, Rudolph C, Jones K. Conversion of adult human fibroblasts into neural precursor cells using chemically modified mRNA. Heliyon 2018; 4:e00918. [PMID: 30450440 PMCID: PMC6226601 DOI: 10.1016/j.heliyon.2018.e00918] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 10/11/2018] [Accepted: 11/02/2018] [Indexed: 12/14/2022] Open
Abstract
Direct reprogramming offers a unique approach by which to generate neural lineages for the study and treatment of neurological disorders. Our objective is to develop a clinically viable reprogramming strategy to generate neural precursor cells for the treatment of neurological disorders through cell replacement therapy. We initially developed a method for directly generating neural precursor cells (iNPs) from adult human fibroblasts by transient expression of the neural transcription factors, SOX2 and PAX6 using plasmid DNA. This study advances these findings by examining the use of chemically modified mRNA (cmRNA) for direct-to-iNP reprogramming. Chemically modified mRNA has the benefit of being extremely stable and non-immunogenic, offering a clinically suitable gene delivery system. The use of SOX2 and PAX6 cmRNA resulted in high co-transfection efficiency and cell viability compared with plasmid transfection. Neural positioning and fate determinant genes were observed throughout reprogramming with ion channel and synaptic marker genes detected during differentiation. Differentiation of cmRNA-derived iNPs generated immature GABAergic or glutamatergic neuronal phenotypes in conjunction with astrocytes. This represents the first time a cmRNA approach has been used to directly reprogram adult human fibroblasts to iNPs, potentially providing an efficient system by which to generate human neurons for both research and clinical application.
Collapse
Affiliation(s)
- Bronwen Connor
- Department of Pharmacology & Clinical Pharmacology, Centre for Brain Research, School of Medical Science, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Erin Firmin
- Department of Pharmacology & Clinical Pharmacology, Centre for Brain Research, School of Medical Science, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Amy McCaughey-Chapman
- Department of Pharmacology & Clinical Pharmacology, Centre for Brain Research, School of Medical Science, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Ruth Monk
- Department of Pharmacology & Clinical Pharmacology, Centre for Brain Research, School of Medical Science, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Kevin Lee
- Department of Physiology, Centre for Brain Research, School of Medical Science, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Sophie Liot
- Department of Pharmacology & Clinical Pharmacology, Centre for Brain Research, School of Medical Science, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | | | | | - Kathryn Jones
- Department of Pharmacology & Clinical Pharmacology, Centre for Brain Research, School of Medical Science, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
23
|
Lv K, Chen Z, Zhang X, Zhang Q, Liu L. Selective enrichment of CD133 +/SOX2 + glioblastoma stem cells via adherent culture. Oncol Lett 2018; 16:4567-4576. [PMID: 30197675 DOI: 10.3892/ol.2018.9154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 06/08/2018] [Indexed: 11/06/2022] Open
Abstract
Most of the brain tumors are malignant with an extremely poor survival rate. Recent progress in identifying cancer stem cells (CSCs) within the brain tumors is starting to revolutionize our understanding in the imitation and progression of tumors as well as relapse and the development of therapeutic strategies. Suspension spheroid body culture paradigm is a routine method in enriching CSCs. While, it was reported recently that CSCs within the brain tumor may also be enriched through adherent monolayer culture with optimized properties. In the present study, 18 surgically resected brain tumors were used for analyzing the feasibility of adherent enrichment of CSCs. The results indicated that 50% of glioblastomas were able to generate adherent CSCs, which were uniformly positive for Sox2, CD133, GFAP and Nestin. However, adherent culture paradigm failed to yield CSCs in secondary brain tumors, including neurocytomas, ependymomas, germ cell tumors or low-grade astrocytomas, which is most likely due to a lack of CD133+/Sox2+ cells within the original biopsies. Therefore, it was concluded that the adherent culture paradigm may serve as a reliable method in enriching brain CSCs, but this method is more suitable for enriching CD133+/Sox2+ CSCs in glioblastomas.
Collapse
Affiliation(s)
- Ke Lv
- Neurosurgical Department, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200092, P.R. China.,Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, P.R. China
| | - Zhenyu Chen
- Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, P.R. China.,Institute of Translational Research, Tongji Hospital, Tongji University School of Medicine, Shanghai 200092, P.R. China.,The Collaborative Innovation Center for Brain Science, Tongji University, Shanghai 200092, P.R. China
| | - Xiaoqing Zhang
- Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, P.R. China.,Institute of Translational Research, Tongji Hospital, Tongji University School of Medicine, Shanghai 200092, P.R. China.,The Collaborative Innovation Center for Brain Science, Tongji University, Shanghai 200092, P.R. China
| | - Quanbin Zhang
- Neurosurgical Department, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200092, P.R. China
| | - Ling Liu
- Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, P.R. China.,Institute of Translational Research, Tongji Hospital, Tongji University School of Medicine, Shanghai 200092, P.R. China
| |
Collapse
|
24
|
Direct reprogramming of fibroblasts into neural stem cells by single non-neural progenitor transcription factor Ptf1a. Nat Commun 2018; 9:2865. [PMID: 30030434 PMCID: PMC6054649 DOI: 10.1038/s41467-018-05209-1] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 06/23/2018] [Indexed: 01/29/2023] Open
Abstract
Induced neural stem cells (iNSCs) reprogrammed from somatic cells have great potentials in cell replacement therapies and in vitro modeling of neural diseases. Direct conversion of fibroblasts into iNSCs has been shown to depend on a couple of key neural progenitor transcription factors (TFs), raising the question of whether such direct reprogramming can be achieved by non-neural progenitor TFs. Here we report that the non-neural progenitor TF Ptf1a alone is sufficient to directly reprogram mouse and human fibroblasts into self-renewable iNSCs capable of differentiating into functional neurons, astrocytes and oligodendrocytes, and improving cognitive dysfunction of Alzheimer’s disease mouse models when transplanted. The reprogramming activity of Ptf1a depends on its Notch-independent interaction with Rbpj which leads to subsequent activation of expression of TF genes and Notch signaling required for NSC specification, self-renewal, and homeostasis. Together, our data identify a non-canonical and safer approach to establish iNSCs for research and therapeutic purposes. Fibroblasts can be reprogrammed into induced neural stem cells (iNSCs) using transcription factors expressed in neural progenitors. Here the authors show that Ptf1a, which is normally expressed in postmitotic neurons, can reprogram fibroblasts to iNSCs through Notch independent interaction with Rbpj.
Collapse
|
25
|
Menzel-Severing J, Zenkel M, Polisetti N, Sock E, Wegner M, Kruse FE, Schlötzer-Schrehardt U. Transcription factor profiling identifies Sox9 as regulator of proliferation and differentiation in corneal epithelial stem/progenitor cells. Sci Rep 2018; 8:10268. [PMID: 29980721 PMCID: PMC6035181 DOI: 10.1038/s41598-018-28596-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/26/2018] [Indexed: 02/08/2023] Open
Abstract
Understanding transcription factor (TF) regulation of limbal epithelial stem/progenitor cells (LEPCs) may aid in using non-ocular cells to regenerate the corneal surface. This study aimed to identify and characterize TF genes expressed specifically in LEPCs isolated from human donor eyes by laser capture microdissection. Using a profiling approach, preferential limbal expression was found for SoxE and SoxF genes, particularly for Sox9, which showed predominantly cytoplasmic localization in basal LEPCs and nuclear localization in suprabasal and corneal epithelial cells, indicating nucleocytoplasmic translocation and activation during LEPC proliferation and differentiation. Increased nuclear localization of Sox9 was also observed in activated LEPCs following clonal expansion and corneal epithelial wound healing. Knockdown of SOX9 expression in cultured LEPCs by RNAi led to reduced expression of progenitor cell markers, e.g. keratin 15, and increased expression of differentiation markers, e.g. keratin 3. Furthermore, SOX9 silencing significantly suppressed the proliferative capacity of LEPCs and reduced levels of glycogen synthase kinase 3 beta (GSK-3ß), a negative regulator of Wnt/ß-catenin signaling. Sox9 expression, in turn, was significantly suppressed by treatment of LEPCs with exogenous GSK-3ß inhibitors and enhanced by small molecule inhibitors of Wnt signaling. Our results suggest that Sox9 and Wnt/ß-catenin signaling cooperate in mutually repressive interactions to achieve a balance between quiescence, proliferation and differentiation of LEPCs in the limbal niche. Future molecular dissection of Sox9-Wnt interaction and mechanisms of nucleocytoplasmic shuttling of Sox9 may aid in improving the regenerative potential of LEPCs and the reprogramming of non-ocular cells for corneal surface regeneration.
Collapse
Affiliation(s)
- Johannes Menzel-Severing
- Department of Ophthalmology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Zenkel
- Department of Ophthalmology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Naresh Polisetti
- Department of Ophthalmology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Elisabeth Sock
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Wegner
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Friedrich E Kruse
- Department of Ophthalmology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | |
Collapse
|
26
|
Kim BE, Choi SW, Shin JH, Kim JJ, Kang I, Lee BC, Lee JY, Kook MG, Kang KS. Single-Factor SOX2 Mediates Direct Neural Reprogramming of Human Mesenchymal Stem Cells via Transfection of In Vitro Transcribed mRNA. Cell Transplant 2018; 27:1154-1167. [PMID: 29909688 PMCID: PMC6158546 DOI: 10.1177/0963689718771885] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 02/20/2018] [Accepted: 02/27/2018] [Indexed: 12/28/2022] Open
Abstract
Neural stem cells (NSCs) are a prominent cell source for understanding neural pathogenesis and for developing therapeutic applications to treat neurodegenerative disease because of their regenerative capacity and multipotency. Recently, a variety of cellular reprogramming technologies have been developed to facilitate in vitro generation of NSCs, called induced NSCs (iNSCs). However, the genetic safety aspects of established virus-based reprogramming methods have been considered, and non-integrating reprogramming methods have been developed. Reprogramming with in vitro transcribed (IVT) mRNA is one of the genetically safe reprogramming methods because exogenous mRNA temporally exists in the cell and is not integrated into the chromosome. Here, we successfully generated expandable iNSCs from human umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs) via transfection with IVT mRNA encoding SOX2 (SOX2 mRNA) with properly optimized conditions. We confirmed that generated human UCB-MSC-derived iNSCs (UM-iNSCs) possess characteristics of NSCs, including multipotency and self-renewal capacity. Additionally, we transfected human dermal fibroblasts (HDFs) with SOX2 mRNA. Compared with human embryonic stem cell-derived NSCs, HDFs transfected with SOX2 mRNA exhibited neural reprogramming with similar morphologies and NSC-enriched mRNA levels, but they showed limited proliferation ability. Our results demonstrated that human UCB-MSCs can be used for direct reprogramming into NSCs through transfection with IVT mRNA encoding a single factor, which provides an integration-free reprogramming tool for future therapeutic application.
Collapse
Affiliation(s)
- Bo-Eun Kim
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- College of Medicine, Seoul National University, Seoul, Republic of Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Soon Won Choi
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Ji-Hee Shin
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Jae-Jun Kim
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- College of Medicine, Seoul National University, Seoul, Republic of Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Insung Kang
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Byung-Chul Lee
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Jin Young Lee
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Myoung Geun Kook
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
27
|
Samoilova EM, Kalsin VA, Kushnir NM, Chistyakov DA, Troitskiy AV, Baklaushev VP. Adult Neural Stem Cells: Basic Research and Production Strategies for Neurorestorative Therapy. Stem Cells Int 2018; 2018:4835491. [PMID: 29760724 PMCID: PMC5901847 DOI: 10.1155/2018/4835491] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 02/01/2018] [Indexed: 12/24/2022] Open
Abstract
Over many decades, constructing genetically and phenotypically stable lines of neural stem cells (NSC) for clinical purposes with the aim of restoring irreversibly lost functions of nervous tissue has been one of the major goals for multiple research groups. The unique ability of stem cells to maintain their own pluripotent state even in the adult body has made them into the choice object of study. With the development of the technology for induced pluripotent stem cells (iPSCs) and direct transdifferentiation of somatic cells into the desired cell type, the initial research approaches based on the use of allogeneic NSCs from embryonic or fetal nervous tissue are gradually becoming a thing of the past. This review deals with basic molecular mechanisms for maintaining the pluripotent state of embryonic/induced stem and reprogrammed somatic cells, as well as with currently existing reprogramming strategies. The focus is on performing direct reprogramming while bypassing the stage of iPSCs which is known for genetic instability and an increased risk of tumorigenesis. A detailed description of various protocols for obtaining reprogrammed neural cells used in the therapy of the nervous system pathology is also provided.
Collapse
Affiliation(s)
- E. M. Samoilova
- Federal Research Clinical Center of the Federal Biomedical Agency of Russian Federation, 28 Orekhovy Blvd, Moscow 115682, Russia
| | - V. A. Kalsin
- Federal Research Clinical Center of the Federal Biomedical Agency of Russian Federation, 28 Orekhovy Blvd, Moscow 115682, Russia
| | - N. M. Kushnir
- Federal Research Clinical Center of the Federal Biomedical Agency of Russian Federation, 28 Orekhovy Blvd, Moscow 115682, Russia
| | - D. A. Chistyakov
- Department of Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Center for Psychiatry and Narcology, Moscow, Russia
| | - A. V. Troitskiy
- Federal Research Clinical Center of the Federal Biomedical Agency of Russian Federation, 28 Orekhovy Blvd, Moscow 115682, Russia
- Institute for Advanced Studies, Federal Biomedical Agency of Russian Federation, Moscow, Russia
| | - V. P. Baklaushev
- Federal Research Clinical Center of the Federal Biomedical Agency of Russian Federation, 28 Orekhovy Blvd, Moscow 115682, Russia
- Institute for Advanced Studies, Federal Biomedical Agency of Russian Federation, Moscow, Russia
| |
Collapse
|
28
|
Panaliappan TK, Wittmann W, Jidigam VK, Mercurio S, Bertolini JA, Sghari S, Bose R, Patthey C, Nicolis SK, Gunhaga L. Sox2 is required for olfactory pit formation and olfactory neurogenesis through BMP restriction and Hes5 upregulation. Development 2018; 145:145/2/dev153791. [PMID: 29352015 PMCID: PMC5825848 DOI: 10.1242/dev.153791] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 12/11/2017] [Indexed: 12/26/2022]
Abstract
The transcription factor Sox2 is necessary to maintain pluripotency of embryonic stem cells, and to regulate neural development. Neurogenesis in the vertebrate olfactory epithelium persists from embryonic stages through adulthood. The role Sox2 plays for the development of the olfactory epithelium and neurogenesis within has, however, not been determined. Here, by analysing Sox2 conditional knockout mouse embryos and chick embryos deprived of Sox2 in the olfactory epithelium using CRISPR-Cas9, we show that Sox2 activity is crucial for the induction of the neural progenitor gene Hes5 and for subsequent differentiation of the neuronal lineage. Our results also suggest that Sox2 activity promotes the neurogenic domain in the nasal epithelium by restricting Bmp4 expression. The Sox2-deficient olfactory epithelium displays diminished cell cycle progression and proliferation, a dramatic increase in apoptosis and finally olfactory pit atrophy. Moreover, chromatin immunoprecipitation data show that Sox2 directly binds to the Hes5 promoter in both the PNS and CNS. Taken together, our results indicate that Sox2 is essential to establish, maintain and expand the neuronal progenitor pool by suppressing Bmp4 and upregulating Hes5 expression. Summary: Analysis of Sox2 mutant mouse and Sox2 CRISPR-targeted chick embryos reveals that Sox2 controls the establishment of sensory progenitors in the olfactory epithelium by suppressing Bmp4 and upregulating Hes5 expression.
Collapse
Affiliation(s)
| | - Walter Wittmann
- Umeå Centre for Molecular Medicine, Umeå University, 901 87 Umeå, Sweden
| | - Vijay K Jidigam
- Umeå Centre for Molecular Medicine, Umeå University, 901 87 Umeå, Sweden
| | - Sara Mercurio
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy
| | - Jessica A Bertolini
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy
| | - Soufien Sghari
- Umeå Centre for Molecular Medicine, Umeå University, 901 87 Umeå, Sweden
| | - Raj Bose
- Umeå Centre for Molecular Medicine, Umeå University, 901 87 Umeå, Sweden
| | - Cedric Patthey
- Umeå Centre for Molecular Medicine, Umeå University, 901 87 Umeå, Sweden
| | - Silvia K Nicolis
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy
| | - Lena Gunhaga
- Umeå Centre for Molecular Medicine, Umeå University, 901 87 Umeå, Sweden
| |
Collapse
|
29
|
Kwon D, Ahn HJ, Kang KS. Generation of Human Neural Stem Cells by Direct Phenotypic Conversion. Results Probl Cell Differ 2018; 66:103-121. [PMID: 30209656 DOI: 10.1007/978-3-319-93485-3_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Human neural stem cells (hNSC) are multipotent adult stem cells. Various studies are underway worldwide to identify new methods for treatment of neurological diseases using hNSC. This chapter summarizes the latest research trends in and fields for application of patient-specific hNSC using direct phenotypic conversion technology. The aim of the study was to analyze the advantages and disadvantages of current technology and to suggest relevant directions for future hNSC research.
Collapse
Affiliation(s)
- Daekee Kwon
- Stem Cells and Regenerative Bioengineering Institute in Kangstem Biotech, Seoul National University, Seoul, South Korea
| | - Hee-Jin Ahn
- Stem Cells and Regenerative Bioengineering Institute in Kangstem Biotech, Seoul National University, Seoul, South Korea
| | - Kyung-Sun Kang
- Stem Cells and Regenerative Bioengineering Institute in Kangstem Biotech, Seoul National University, Seoul, South Korea.
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, South Korea.
| |
Collapse
|
30
|
Connor B. Concise Review: The Use of Stem Cells for Understanding and Treating Huntington's Disease. Stem Cells 2017; 36:146-160. [PMID: 29178352 DOI: 10.1002/stem.2747] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 10/13/2017] [Indexed: 12/20/2022]
Abstract
Two decades ago, researchers identified that a CAG expansion mutation in the huntingtin (HTT) gene was involved in the pathogenesis of Huntington's disease (HD). However, since the identification of the HTT gene, there has been no advance in the development of therapeutic strategies to prevent or reduce the progression of HD. With the recent advances in stem cell biology and human cell reprogramming technologies, several novel and exciting pathways have emerged allowing researchers to enhance their understanding of the pathogenesis of HD, to identify and screen potential drug targets, and to explore alternative donor cell sources for cell replacement therapy. This review will discuss the role of compensatory neurogenesis in the HD brain, the use of stem cell-based therapies for HD to replace or prevent cell loss, and the recent advance of cell reprogramming to model and/or treat HD. These new technologies, coupled with advances in genome editing herald a promising new era for HD research with the potential to identify a therapeutic strategy to alleviate this debilitating disorder. Stem Cells 2018;36:146-160.
Collapse
Affiliation(s)
- Bronwen Connor
- Department of Pharmacology and Clinical Pharmacology, Centre for Brain Research, School of Medical Science, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
31
|
Playne R, Connor B. Understanding Parkinson's Disease through the Use of Cell Reprogramming. Stem Cell Rev Rep 2017; 13:151-169. [PMID: 28083784 DOI: 10.1007/s12015-017-9717-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Recent progress in the field of somatic cell reprogramming offers exciting new possibilities for the study and treatment of Parkinson's disease (PD). Reprogramming technology offers the ability to untangle the diverse contributing risk factors for PD, such as ageing, genetics and environmental toxins. In order to gain novel insights into such a complex disease, cell-based models of PD should represent, as closely as possible, aged human dopaminergic neurons of the substantia nigra. However, the generation of high yields of functionally mature, authentic ventral midbrain dopamine (vmDA) neurons has not been easy to achieve. Furthermore, ensuring cells represent aged rather than embryonic neurons has presented a significant challenge. To date, induced pluripotent stem (iPS) cells have received much attention for modelling PD. Nonetheless, direct reprogramming strategies (either to a neuronal or neural stem/progenitor fate) represent a valid alternative that are yet to be extensively explored. Direct reprogramming is faster and more efficient than iPS cell reprogramming, and appears to conserve age-related markers. At present, however, protocols aiming to derive authentic, mature vmDA neurons by direct reprogramming of adult human somatic cells are sorely lacking. This review will discuss the strategies that have been employed to generate vmDA neurons and their potential for the study and treatment of PD.
Collapse
Affiliation(s)
- Rebecca Playne
- Department of Pharmacology and Clinical Pharmacology, Centre for Brain Research, School of Medical Science, FMHS, University of Auckland, Auckland, 1023, New Zealand
| | - Bronwen Connor
- Department of Pharmacology and Clinical Pharmacology, Centre for Brain Research, School of Medical Science, FMHS, University of Auckland, Auckland, 1023, New Zealand.
| |
Collapse
|
32
|
Chen J, Wang Q, Zhou J, Deng W, Yu Q, Cao X, Wang J, Shao F, Li Y, Ma P, Spector M, Yu J, Xu X. Porphyra polysaccharide-derived carbon dots for non-viral co-delivery of different gene combinations and neuronal differentiation of ectodermal mesenchymal stem cells. NANOSCALE 2017; 9:10820-10831. [PMID: 28726952 DOI: 10.1039/c7nr03327c] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
In this study, multifunctional fluorescent carbon dots (CDs) were synthesized using a one-pot hydrothermal carbonization reaction, with the naturally-occurring porphyra polysaccharide (PPS) serving as a single carbon source for the first time and ethylenediamine (Ed) acting as the surface passivation agent. The resulting CDs enjoyed a high quantum yield (56.3%), excitation-dependent fluorescence, small size (<10 nm), spherical shape, uniform distribution, positive surface charge, low cytotoxicity and excellent ability to condense macromolecular plasmid DNA. The synthesized CDs were employed for neuronal induction from ectodermal mesenchymal stem cells for the first time via highly efficient non-viral gene delivery. The optimal combination of factors (Ascl1 and Brn2) was selected from seven different combinations out of Ascl1, Brn2 and Sox2 according to the expression of neuronal markers (Tuj1, Map2 and Tau). The results of qRT-PCR demonstrated that the CDs possessed a significantly higher transfection efficiency than the commercially available transfection reagents PEI (25 kDa) and Lipofectamine2000. Moreover, the CDs/pDNA nanoparticles exhibited more efficient neuronal differentiation of the EMSCs than the AT-RA-containing induction medium. Furthermore, the CDs/pDNA nanoparticles could enter cells via both caveolae- and clathrin-mediated endocytosis. Taken together, the natural polysaccharide PPS-derived CDs enriched the current application of CDs by employing the CDs as a novel non-viral gene carrier for neuronal differentiation of adult stem cells, which held great promise in tissue engineering and bioimaging.
Collapse
Affiliation(s)
- Jiaxin Chen
- Department of Pharmaceutics, School of Pharmacy, and Center for Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang 212001, P.R. China.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Julian LM, McDonald AC, Stanford WL. Direct reprogramming with SOX factors: masters of cell fate. Curr Opin Genet Dev 2017; 46:24-36. [PMID: 28662445 DOI: 10.1016/j.gde.2017.06.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 04/25/2017] [Accepted: 06/09/2017] [Indexed: 12/13/2022]
Abstract
Over the last decade significant advances have been made toward reprogramming the fate of somatic cells, typically by overexpression of cell lineage-determinant transcription factors. As key regulators of cell fate, the SOX family of transcription factors has emerged as potent drivers of direct somatic cell reprogramming into multiple lineages, in some cases as the sole overexpressed factor. The vast capacity of SOX factors, especially those of the SOXB1, E and F subclasses, to reprogram cell fate is enlightening our understanding of organismal development, cancer and disease, and offers tremendous potential for regenerative medicine and cell-based therapies. Understanding the molecular mechanisms through which SOX factors reprogram cell fate is essential to optimize the development of novel somatic cell transdifferentiation strategies.
Collapse
Affiliation(s)
- Lisa M Julian
- Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario K1L8L6, Canada
| | - Angela Ch McDonald
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children Research Institute, 686 Bay Street, Toronto, Ontario M5G0A4, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S3G9, Canada
| | - William L Stanford
- Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario K1L8L6, Canada; Department of Cellular and Molecular Medicine, Faulty of Medicine, University of Ottawa, 451 Smyth Rd, Ottawa, Ontario K1H8M5, Canada; Department of Biochemistry, Microbiology and Immunology, Faulty of Medicine, University of Ottawa, 451 Smyth Rd, Ottawa, Ontario K1H8M5, Canada; Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Rd, Ottawa, Ontario K1H8M5, Canada.
| |
Collapse
|
34
|
Panchision DM. Concise Review: Progress and Challenges in Using Human Stem Cells for Biological and Therapeutics Discovery: Neuropsychiatric Disorders. Stem Cells 2016; 34:523-36. [PMID: 26840228 DOI: 10.1002/stem.2295] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 11/29/2015] [Accepted: 12/09/2015] [Indexed: 12/12/2022]
Abstract
In facing the daunting challenge of using human embryonic and induced pluripotent stem cells to study complex neural circuit disorders such as schizophrenia, mood and anxiety disorders, and autism spectrum disorders, a 2012 National Institute of Mental Health workshop produced a set of recommendations to advance basic research and engage industry in cell-based studies of neuropsychiatric disorders. This review describes progress in meeting these recommendations, including the development of novel tools, strides in recapitulating relevant cell and tissue types, insights into the genetic basis of these disorders that permit integration of risk-associated gene regulatory networks with cell/circuit phenotypes, and promising findings of patient-control differences using cell-based assays. However, numerous challenges are still being addressed, requiring further technological development, approaches to resolve disease heterogeneity, and collaborative structures for investigators of different disciplines. Additionally, since data obtained so far is on small sample sizes, replication in larger sample sets is needed. A number of individual success stories point to a path forward in developing assays to translate discovery science to therapeutics development.
Collapse
Affiliation(s)
- David M Panchision
- Division of Neuroscience and Basic Behavioral Science, National Institute of Mental Health, Bethesda, Maryland, USA
| |
Collapse
|
35
|
Liang L, Zhang Y, Malpica A, Ramalingam P, Euscher ED, Fuller GN, Liu J. Gliomatosis peritonei: a clinicopathologic and immunohistochemical study of 21 cases. Mod Pathol 2015; 28:1613-20. [PMID: 26564007 PMCID: PMC4682736 DOI: 10.1038/modpathol.2015.116] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 08/27/2015] [Accepted: 08/29/2015] [Indexed: 02/02/2023]
Abstract
Gliomatosis peritonei, a rare condition often associated with immature ovarian teratoma, is characterized by the presence of mature glial tissue in the peritoneum. We retrospectively evaluated 21 patients with gliomatosis peritonei and studied their clinicopathologic features and immunophenotype. The patients' ages ranged from 5 to 42 years (median, 19 years). Their primary ovarian tumors consisted of immature teratoma (n=14), mixed germ cell tumors (n=6), and mature teratoma with a carcinoid tumor (n=1). Gliomatosis peritonei was diagnosed at the same time as primary ovarian neoplasm in 16 patients and secondary surgery in 5 patients. Also, 11 of 21 patients had metastatic immature teratoma (n=4), metastatic mature teratoma (n=2), or both (n=5). One patient developed glioma arising from gliomatosis peritonei. Seventeen patients had follow-up information and were alive with no evidence of disease (n=13), alive with disease (n=3), or alive with an unknown disease status (n=1). The follow-up durations ranged from 1 to 229 months (mean, 49 months; median, 23 months). Immunohistochemistry results demonstrated that SOX2 was expressed in all cases of gliomatosis peritonei and glioma with tissue available (nine of nine cases), whereas OCT4 and NANOG were negative in all cases with available tissue (eight of eight cases). In conclusion, both gliomatosis peritonei and glioma arising from it show a SOX2+/OCT4-/NANOG- immunophenotype. These findings demonstrated that gliomatosis peritonei is associated with favorable prognosis, although it is important to rule out potentially associated immature teratoma and malignant transformation. SOX2 may have an important role in the development of gliomatosis peritonei.
Collapse
Affiliation(s)
- Li Liang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yifen Zhang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA,Department of Pathology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Anais Malpica
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Preetha Ramalingam
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elizabeth D. Euscher
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gregory N. Fuller
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jinsong Liu
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA,Department of Pathology, The first Affiliated Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| |
Collapse
|
36
|
Lim MS, Chang MY, Kim SM, Yi SH, Suh-Kim H, Jung SJ, Kim MJ, Kim JH, Lee YS, Lee SY, Kim DW, Lee SH, Park CH. Generation of Dopamine Neurons from Rodent Fibroblasts through the Expandable Neural Precursor Cell Stage. J Biol Chem 2015; 290:17401-14. [PMID: 26023233 DOI: 10.1074/jbc.m114.629808] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Indexed: 01/08/2023] Open
Abstract
Recent groundbreaking work has demonstrated that combined expression of the transcription factors Brn2, Ascl1, and Myt1L (BAM; also known as Wernig factors) convert mouse fibroblasts into postmitotic neuronal cells. However, questions remain regarding whether trans-conversion is achieved directly or involves an intermediary precursor stage. Trans-conversion toward expandable neural precursor cells (NPCs) is more useful than direct one-step neuron formation with respect to yielding a sufficient number of cells and the feasibility of manipulating NPC differentiation toward certain neuron subtypes. Here, we show that co-expression of Wernig factors and Bcl-xL induces fibroblast conversion into NPCs (induced NPCs (iNPCs)) that are highly expandable for >100 passages. Gene expression analyses showed that the iNPCs exhibited high expression of common NPC genes but not genes specific to defined embryonic brain regions. This finding indicated that a regional identity of iNPCs was not established. Upon induction, iNPCs predominantly differentiated into astrocytes. However, the differentiation potential was not fixed and could be efficiently manipulated into general or specific subtypes of neurons by expression of additional genes. Specifically, overexpression of Nurr1 and Foxa2, transcription factors specific for midbrain dopamine neuron development, drove iNPCs to yield mature midbrain dopamine neurons equipped with presynaptic DA neuronal functions. We further assessed the therapeutic potential of iNPCs in Parkinson disease model rats.
Collapse
Affiliation(s)
- Mi-Sun Lim
- From the Graduate School of Biomedical Science and Engineering, the Hanyang Biomedical Research Institute, the Departments of Microbiology
| | - Mi-Yoon Chang
- the Hanyang Biomedical Research Institute, Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul 133-791, Korea
| | - Sang-Mi Kim
- the Department of Biomedical Science, Graduate School, and
| | - Sang-Hoon Yi
- the Hanyang Biomedical Research Institute, Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul 133-791, Korea
| | - Haeyoung Suh-Kim
- the Department of Anatomy and Brain Disease Research Center, College of Medicine, Ajou University, Suwon 443-749, Korea, and
| | - Sung Jun Jung
- the Hanyang Biomedical Research Institute, Physiology, and
| | - Min Jung Kim
- the Hanyang Biomedical Research Institute, Physiology, and
| | - Jin Hyuk Kim
- From the Graduate School of Biomedical Science and Engineering, the Hanyang Biomedical Research Institute, Physiology, and
| | - Yong-Sung Lee
- From the Graduate School of Biomedical Science and Engineering, the Hanyang Biomedical Research Institute, Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul 133-791, Korea
| | | | - Dong-Wook Kim
- the Department of Physiology and Cell Therapy Center, Yonsei University College of Medicine, Seoul 120-752, Korea
| | - Sang-Hun Lee
- the Hanyang Biomedical Research Institute, Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul 133-791, Korea,
| | - Chang-Hwan Park
- From the Graduate School of Biomedical Science and Engineering, the Hanyang Biomedical Research Institute, the Departments of Microbiology,
| |
Collapse
|
37
|
Li D, Zhang T, Gu W, Li P, Cheng X, Tong T, Wang W. The ALDH1⁺ subpopulation of the human NMFH-1 cell line exhibits cancer stem-like characteristics. Oncol Rep 2015; 33:2291-8. [PMID: 25760144 DOI: 10.3892/or.2015.3842] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 02/23/2015] [Indexed: 11/06/2022] Open
Abstract
Cancer stem cells (CSCs) have been reported in many tissues. However, CSCs have yet to be identified in a human malignant fibrous histiocytoma (MFH) cell line. Elevated aldehyde dehydrogenase 1 (ALDH1) has been proposed as a stem cell marker for isolating CSCs from cancer. The aim of the present study was to identify a population with elevated ALDH in the human NMFH-1 cell line. ALDH⁺ and ALDH- cell populations were isolated and compared for CSC characteristics. ALDH enzymatic activity was used as a marker to identify the cells in the NMFH-1 line. Self-renewal, differentiation capacity, and tumorigenicity of the NMFH-1 ALDH⁺ cell population were then examined using a spheroid formation assay and xenograft model in nude mice. Chemoresistance levels, ABCG2 drug transport gene expression, and stem cell-associated gene expression were compared in these NMFH-1 populations. The ALDH⁺ population was better able to form spheres in anchorage-independent serum-starved conditions. Furthermore, the mRNA expression of key stem cell-related genes was enhanced in these cells. Increased expression of the drug transporter gene, ABCG2, was detected. Compared with ALDH-, the ALDH⁺ subpopulation had higher levels of chemoresistance to doxorubicin (DXR) and cisplatin (CDDP). Additionally, the ALDH⁺ cells more efficiently formed tumors when implanted into BALB/c nude mice. ALDH1 may therefore be used as a marker for the isolation of cells that exhibit several characteristics of CSCs from the NMFH-1 cell line. This finding may lead to the development of novel therapies to specifically kill ALDH1⁺ subpopulations (CSCs).
Collapse
Affiliation(s)
- Dejian Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Tao Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Wenguang Gu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Peng Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xiangyang Cheng
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Tiejun Tong
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Wenbo Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
38
|
Can the ‘neuron theory’ be complemented by a universal mechanism for generic neuronal differentiation. Cell Tissue Res 2014; 359:343-84. [DOI: 10.1007/s00441-014-2049-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 10/23/2014] [Indexed: 12/19/2022]
|
39
|
Hu Q, Chen R, Teesalu T, Ruoslahti E, Clegg DO. Reprogramming human retinal pigmented epithelial cells to neurons using recombinant proteins. Stem Cells Transl Med 2014; 3:1526-34. [PMID: 25298373 DOI: 10.5966/sctm.2014-0038] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Somatic cells can be reprogrammed to an altered lineage by overexpressing specific transcription factors. To avoid introducing exogenous genetic material into the genome of host cells, cell-penetrating peptides can be used to deliver transcription factors into cells for reprogramming. Position-dependent C-end rule (CendR) cell- and tissue-penetrating peptides provide an alternative to the conventional cell-penetrating peptides, such as polyarginine. In this study, we used a prototypic, already active CendR peptide, RPARPAR, to deliver the transcription factor SOX2 to retinal pigmented epithelial (RPE) cells. We demonstrated that RPE cells can be directly reprogrammed to a neuronal fate by introduction of SOX2. Resulting neuronal cells expressed neuronal marker mRNAs and proteins and downregulated expression of RPE markers. Cells produced extensive neurites and developed synaptic machinery capable of dye uptake after depolarization with potassium. The RPARPAR-mediated delivery of SOX2 alone was sufficient to allow cell lineage reprogramming of both fetal and stem cell-derived RPE cells to become functional neurons.
Collapse
Affiliation(s)
- Qirui Hu
- Center for Stem Cell Biology and Engineering and Center for Nanomedicine and Sanford-Burnham Medical Research Institute, University of California Santa Barbara, Santa Barbara, California, USA; Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California, USA
| | - Renwei Chen
- Center for Stem Cell Biology and Engineering and Center for Nanomedicine and Sanford-Burnham Medical Research Institute, University of California Santa Barbara, Santa Barbara, California, USA; Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California, USA
| | - Tambet Teesalu
- Center for Stem Cell Biology and Engineering and Center for Nanomedicine and Sanford-Burnham Medical Research Institute, University of California Santa Barbara, Santa Barbara, California, USA; Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California, USA
| | - Erkki Ruoslahti
- Center for Stem Cell Biology and Engineering and Center for Nanomedicine and Sanford-Burnham Medical Research Institute, University of California Santa Barbara, Santa Barbara, California, USA; Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California, USA
| | - Dennis O Clegg
- Center for Stem Cell Biology and Engineering and Center for Nanomedicine and Sanford-Burnham Medical Research Institute, University of California Santa Barbara, Santa Barbara, California, USA; Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California, USA
| |
Collapse
|
40
|
Ni Y, Zhang K, Liu X, Yang T, Wang B, Fu L, A L, Zhou Y. miR-21 promotes the differentiation of hair follicle-derived neural crest stem cells into Schwann cells. Neural Regen Res 2014; 9:828-36. [PMID: 25206896 PMCID: PMC4146246 DOI: 10.4103/1673-5374.131599] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2014] [Indexed: 12/20/2022] Open
Abstract
Hair follicle-derived neural crest stem cells can be induced to differentiate into Schwann cells in vivo and in vitro. However, the underlying regulatory mechanism during cell differentiation remains poorly understood. This study isolated neural crest stem cells from human hair follicles and induced them to differentiate into Schwann cells. Quantitative RT-PCR showed that microRNA (miR)-21 expression was gradually increased during the differentiation of neural crest stem cells into Schwann cells. After transfection with the miR-21 agonist (agomir-21), the differentiation capacity of neural crest stem cells was enhanced. By contrast, after transfection with the miR-21 antagonist (antagomir-21), the differentiation capacity was attenuated. Further study results showed that SOX-2 was an effective target of miR-21. Without compromising SOX2 mRNA expression, miR-21 can down-regulate SOX protein expression by binding to the 3′-UTR of miR-21 mRNA. Knocking out the SOX2 gene from the neural crest stem cells significantly reversed the antagomir-21 inhibition of neural crest stem cells differentiating into Schwann cells. The results suggest that miR-21 expression was increased during the differentiation of neural crest stem cells into Schwann cells and miR-21 promoted the differentiation through down-regulating SOX protein expression by binding to the 3′-UTR of SOX2 mRNA.
Collapse
Affiliation(s)
- Yuxin Ni
- Hospital of Stomatology, Jilin University, Changchun, Jilin Province, China
| | - Kaizhi Zhang
- China-Japan Union Hospital, Jilin University, Changchun, Jilin Province, China
| | - Xuejuan Liu
- First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Tingting Yang
- Hospital of Stomatology, Jilin University, Changchun, Jilin Province, China
| | - Baixiang Wang
- Hospital of Stomatology, Jilin University, Changchun, Jilin Province, China
| | - Li Fu
- Hospital of Stomatology, Jilin University, Changchun, Jilin Province, China
| | - Lan A
- Hospital of Stomatology, Jilin University, Changchun, Jilin Province, China
| | - Yanmin Zhou
- Hospital of Stomatology, Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
41
|
Reiprich S, Wegner M. From CNS stem cells to neurons and glia: Sox for everyone. Cell Tissue Res 2014; 359:111-24. [PMID: 24894327 DOI: 10.1007/s00441-014-1909-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 05/05/2014] [Indexed: 12/17/2022]
Abstract
Neuroepithelial precursor cells of the vertebrate central nervous system either self-renew or differentiate into neurons, oligodendrocytes or astrocytes under the influence of a gene regulatory network that consists in transcription factors, epigenetic modifiers and microRNAs. Sox transcription factors are central to this regulatory network, especially members of the SoxB, SoxC, SoxD, SoxE and SoxF groups. These Sox proteins are widely expressed in neuroepithelial precursor cells and in newly specified, differentiating and mature neurons, oligodendrocytes and astrocytes and influence their identity, survival and development. They exert their effect predominantly at the transcriptional level but also have substantial impact on expression at the epigenetic and posttranscriptional levels with some Sox proteins acting as pioneer factors, recruiting chromatin-modifying and -remodelling complexes or influencing microRNA expression. They interact with a large variety of other transcription factors and influence the expression of regulatory molecules and effector genes in a cell-type-specific and temporally controlled manner. As versatile regulators with context-dependent functions, they are not only indispensable for central nervous system development but might also be instrumental for the development of reprogramming and cell conversion strategies for replacement therapies and for assisted regeneration after injury or degeneration-induced cell loss in the central nervous system.
Collapse
Affiliation(s)
- Simone Reiprich
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, 91054, Erlangen, Germany,
| | | |
Collapse
|
42
|
Sharma R, Livesey MR, Wyllie DJA, Proudfoot C, Whitelaw CBA, Hay DC, Donadeu FX. Generation of functional neurons from feeder-free, keratinocyte-derived equine induced pluripotent stem cells. Stem Cells Dev 2014; 23:1524-34. [PMID: 24548115 DOI: 10.1089/scd.2013.0565] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Pluripotent stem cells (PSCs) offer unprecedented biomedical potential not only in relation to humans but also companion animals, particularly the horse. Despite this, attempts to generate bona fide equine embryonic stem cells have been unsuccessful. A very limited number of induced PSC lines have so far been generated from equine fibroblasts but their potential for directed differentiation into clinically relevant tissues has not been explored. In this study, we used retroviral vectors to generate induced pluripotent stem cells (iPSCs) with comparatively high efficiency from equine keratinocytes. Expression of endogenous PSC markers (OCT4, SOX2, LIN28, NANOG, DNMT3B, and REX1) was effectively restored in these cells, which could also form in vivo several tissue derivatives of the three germ layers, including functional neurons, keratinized epithelium, cartilage, bone, muscle, and respiratory and gastric epithelia. Comparative analysis of different reprogrammed cell lines revealed an association between the ability of iPSCs to form well-differentiated teratomas and the distinct endogenous expression of OCT4 and REX1 and reduced expression of viral transgenes. Importantly, unlike in previous studies, equine iPSCs were successfully expanded using simplified feeder-free culture conditions, constituting significant progress toward future biomedical applications. Further, under appropriate conditions equine iPSCs generated cells with features of cholinergic motor neurons including the ability to generate action potentials, providing the first report of functional cells derived from equine iPSCs. The ability to derive electrically active neurons in vitro from a large animal reveals highly conserved pathways of differentiation across species and opens the way for new and exciting applications in veterinary regenerative medicine.
Collapse
Affiliation(s)
- Ruchi Sharma
- 1 The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh , Midlothian, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
43
|
Gericota B, Anderson JS, Mitchell G, Borjesson DL, Sturges BK, Nolta JA, Sieber-Blum M. Canine epidermal neural crest stem cells: characterization and potential as therapy candidate for a large animal model of spinal cord injury. Stem Cells Transl Med 2014; 3:334-45. [PMID: 24443004 PMCID: PMC3952930 DOI: 10.5966/sctm.2013-0129] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 10/23/2013] [Indexed: 02/07/2023] Open
Abstract
The discovery of multipotent neural crest-derived stem cells, named epidermal neural crest stem cells (EPI-NCSC), that persist postnatally in an easy-to-access location-the bulge of hair follicles-opens a spectrum of novel opportunities for patient-specific therapies. We present a detailed characterization of canine EPI-NCSC (cEPI-NCSC) from multiple dog breeds and protocols for their isolation and ex vivo expansion. Furthermore, we provide novel tools for research in canines, which currently are still scarce. In analogy to human and mouse EPI-NCSC, the neural crest origin of cEPI-NCSC is shown by their expression of the neural crest stem cell molecular signature and other neural crest-characteristic genes. Similar to human EPI-NCSC, cEPI-NCSC also expressed pluripotency genes. We demonstrated that cEPI-NCSC can generate all major neural crest derivatives. In vitro clonal analyses established multipotency and self-renewal ability of cEPI-NCSC, establishing cEPI-NCSC as multipotent somatic stem cells. A critical analysis of the literature on canine spinal cord injury (SCI) showed the need for novel treatments and suggested that cEPI-NCSC represent viable candidates for cell-based therapies in dog SCI, particularly for chondrodystrophic dogs. This notion is supported by the close ontological relationship between neural crest stem cells and spinal cord stem cells. Thus, cEPI-NCSC promise to offer not only a potential treatment for canines but also an attractive and realistic large animal model for human SCI. Taken together, we provide the groundwork for the development of a novel cell-based therapy for a condition with extremely poor prognosis and no available effective treatment.
Collapse
|