1
|
Boopathy K, Palaniyandi T, Ravi M, Wahab MRA, Baskar G, Rab SO, Saeed M, Balaramnavar VM. Exploring the potential of stem cell therapy: Applications, types, and future directions. Acta Histochem 2025; 127:152237. [PMID: 40020616 DOI: 10.1016/j.acthis.2025.152237] [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: 12/23/2024] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 03/03/2025]
Abstract
One of the most significant treatment approaches now accessible is stem cell therapy. Over the last few decades, a lot of study has been done in this field, and this fascinating feature of plasticity could have therapeutic uses. The potential of stem cells to restore function lost as a result of disease, trauma, congenital defects, and age has made stem cell research a key priority for scientific and medical organizations across the world. Stem cells are a crucial topic of study in regenerative medicine because of their capacity to replace, repair, or regenerate damaged cells, tissues, or organs. As a result, stem cell therapy is being used as a treatment strategy for a number of illnesses. Because stem cells may proliferate indefinitely and generate vast quantities of differentiated cells needed for transplantation, they hold enormous promise for regenerative medicine. Stem cells can be reprogrammed from adult cell types or originate from embryonic or fetal origins. Depending on their availability and place of origin, stem cells can be totipotent, pluripotent, multipotent, oligopotent, or unipotent. With stem cell treatment, many ailments, including diabetes, liver disease, infertility, wounds and traumas, neurological disorders, cardiovascular disease, and cancer, might be cured. Various types of stem cell treatment are described in this review along with their applications in different therapeutic fields, ethical considerations, and advantages and disadvantages.
Collapse
Affiliation(s)
- KeerthiShri Boopathy
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Chennai 600095, India
| | - Thirunavukkarasu Palaniyandi
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Chennai 600095, India; ACS-Advanced Medical Research Institute, Dr. M.G.R Educational and Research Institute, Chennai 600077, India.
| | - Maddaly Ravi
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu 600 116, India
| | | | - Gomathy Baskar
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Chennai 600095, India
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Mohd Saeed
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| | - Vishal M Balaramnavar
- School of Pharmacy and Research Centre, Sanskriti University, Chhata, Mathura, Uttar Pradesh 281401, India
| |
Collapse
|
2
|
Kim WK, Son YS, Lim JH, Kim WH, Kang BJ. Neural stem/progenitor cells from adult canine cervical spinal cord have the potential to differentiate into neural lineage cells. BMC Vet Res 2023; 19:193. [PMID: 37803301 PMCID: PMC10557334 DOI: 10.1186/s12917-023-03757-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 09/27/2023] [Indexed: 10/08/2023] Open
Abstract
BACKGROUND • Neural stem/progenitor cells (NSPCs) are multipotent self-renewing cells that can be isolated from the brain or spinal cord. As they need to be isolated from neural tissues, it is difficult to study human NSPCs. To facilitate NSPC research, we attempted to isolate NSPCs from dogs, as dogs share the environment and having many similar diseases with humans. We collected and established primary cultures of ependymal and subependymal cells from the central canal of the cervical spinal cord of adult dogs. To isolate pure NSPCs, we employed the monolayer culture and selective medium culture methods. We further tested the ability of the NSPCs to form neurospheres (using the suspension culture method) and evaluated their differentiation potential. RESULTS • The cells had the ability to grow as cultures for up to 10 passages; the growth curves of the cells at the 3rd, 6th, and 9th passages showed similar patterns. The NSPCs were able to grow as neurospheres as well as monolayers, and immunostaining at the 3rd, 6th, and 9th passages showed that these cells expressed NSPC markers such as nestin and SOX2 (immunofluorescent staining). Monolayer cultures of NSPCs at the 3rd, 6th, and 9th passages were cultured for approximately 14 days using a differentiation medium and were observed to successfully differentiate into neural lineage and glial cells (astrocytes, neurons, and oligodendrocytes) at all the three passages tested. CONCLUSION • It is feasible to isolate and propagate (up to at least 10 passages) canine cervical spinal cord-derived NSPCs with the capacity to differentiate into neuronal and glial cells. To the best of our knowledge this is the first study to successfully isolate, propagate, and differentiate canine NSPCs derived from cervical spinal cord in the adult canine, and we believe that these cells will contribute to the field of spinal cord regeneration in veterinary and comparative medicine.
Collapse
Affiliation(s)
- Woo Keyoung Kim
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Korea
- BK21 FOUR Future Veterinary Medicine Leading Education and Research Center, Seoul National University, Seoul, 08826, Korea
| | - Yeon Sung Son
- Medical Research Center, College of Medicine, Seoul National University, Seoul, 03080, South Korea
| | - Ji-Hey Lim
- Department of Neurology/Neurosurgery, College of Veterinary Medicine, University of Missouri, Columbia, 65211, USA
| | - Wan Hee Kim
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Korea
| | - Byung-Jae Kang
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Korea.
- BK21 FOUR Future Veterinary Medicine Leading Education and Research Center, Seoul National University, Seoul, 08826, Korea.
| |
Collapse
|
3
|
Andolfi A, Arnaldi P, Lisa DD, Pepe S, Frega M, Fassio A, Lagazzo A, Martinoia S, Pastorino L. A micropatterned thermoplasmonic substrate for neuromodulation of in vitro neuronal networks. Acta Biomater 2023; 158:281-291. [PMID: 36563774 DOI: 10.1016/j.actbio.2022.12.036] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 12/05/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
Understanding how the spatial organization of a neural network affects its activity represents a leading issue in neuroscience. Thanks to their accessibility and easy handling, in vitro studies remain an essential tool to investigate the relationship between the structure and function of a neuronal network. Among all the patterning techniques, ink-jet printing acquired great interest thanks to its direct-write approach, which allows the patterned substrate realization without mold, leading to a considerable saving of both cost and time. However, the inks commonly used give the possibility to control only the structure of a neuronal network, leaving aside the functional aspect. In this work, we synthesize a photosensitive ink combining the rheological and bioadhesive properties of chitosan with the plasmonic properties of gold nanorods, obtaining an ink able to control both the spatial organization of a two-dimensional neuronal network and its activity through photothermal effect. After the ink characterization, we demonstrate that it is possible to print, with high precision, different geometries on a microelectrode array. In this way, it is possible obtaining a patterned device to control the structure of a neuronal network, to record its activity and to modulate it via photothermal effect. Finally, to our knowledge, we report the first evidence of photothermal inhibition of human neurons activity. STATEMENT OF SIGNIFICANCE: Patterned cell cultures remain the most efficient and simple tool for linking structural and functional studies, especially in the neuronal field. Ink-jet printing is the technique with which it is possible to realize patterned structures in the fastest, simple, versatile and low-cost way. However, the inks currently used permit the control only of the neuronal network structure but do not allow the control-modulation of the network activity. In this study, we realize and characterize a photosensitive bioink with which it is possible to drive both the structure and the activity of a neuronal network. Moreover, we report the first evidence of activity inhibition by the photothermal effect on human neurons as far as we know.
Collapse
Affiliation(s)
- Andrea Andolfi
- Department of Informatics, Bioengineering, Robotics and Systems Engineering (DIBRIS), University of Genoa, Genoa, Italy.
| | - Pietro Arnaldi
- Department of Informatics, Bioengineering, Robotics and Systems Engineering (DIBRIS), University of Genoa, Genoa, Italy.
| | - Donatella Di Lisa
- Department of Informatics, Bioengineering, Robotics and Systems Engineering (DIBRIS), University of Genoa, Genoa, Italy.
| | - Sara Pepe
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.
| | - Monica Frega
- Department of Clinical Neurophysiology, University of Twente, Enschede, the Netherlands.
| | - Anna Fassio
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.
| | - Alberto Lagazzo
- Department of Civil, Chemical and Environmental Engineering (DICCA), University of Genoa, Genoa, Italy.
| | - Sergio Martinoia
- Department of Informatics, Bioengineering, Robotics and Systems Engineering (DIBRIS), University of Genoa, Genoa, Italy.
| | - Laura Pastorino
- Department of Informatics, Bioengineering, Robotics and Systems Engineering (DIBRIS), University of Genoa, Genoa, Italy.
| |
Collapse
|
4
|
Suk Y, Kieliszek A, Mobilio D, Venugopal C, Singh SK. Derivation and culturing of neural stem cells from human embryonic brain tissue. STAR Protoc 2022; 3:101628. [PMID: 36035806 PMCID: PMC9405532 DOI: 10.1016/j.xpro.2022.101628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Human neural stem cells (hNSCs) are a valuable tool in brain cancer research since they are used as a normal control for multiple assays, mainly pertaining to toxicity. Here, we present a protocol to safely and successfully derive and culture hNSCs in vitro from human embryonic brain tissue. We describe the steps to dissociate embryonic brain tissue and culture hNSCs, followed by the procedure to expand hNSCs. These cells can be used for downstream applications including RNA-seq and omics studies. For complete details on the use and execution of this protocol, please refer to Venugopal et al. (2012b), Bakhshinyan et al. (2019), and Venugopal et al. (2012a). Derivation and cryopreservation of hNSCs from human embryonic tissue Long-term culture and expansion of primary hNSC cells Feasible to generate up to 200 million human neural stem cells in 1 to 2 months Can be used for downstream applications such as RNAseq and omics techniques
Publisher’s note: Undertaking any experimental protocol requires adherence to local institutional guidelines for laboratory safety and ethics.
Collapse
Affiliation(s)
- Yujin Suk
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada; Michael G DeGroote School of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada.
| | - Agata Kieliszek
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Daniel Mobilio
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Chitra Venugopal
- Department of Surgery, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Sheila K Singh
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada; Department of Surgery, McMaster University, Hamilton, ON L8S 4L8, Canada.
| |
Collapse
|
5
|
Luo L, Zhang W, Chen W, Fu X, Wang X, Xu R, Hu D. Based on a Self-Feeder Layer, a Novel 3D Culture Model of Human ADSCs Facilitates Trans-Differentiation of the Spheroid Cells into Neural Progenitor-Like Cells Using siEID3 with a Laminin/Poly-d-lysine Matrix. Cells 2021; 10:493. [PMID: 33668931 PMCID: PMC7996540 DOI: 10.3390/cells10030493] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/18/2021] [Accepted: 02/21/2021] [Indexed: 12/21/2022] Open
Abstract
Human adipose-derived stromal cells (ADSCs) are receiving unprecedented attention as a potential cellular source for regenerative medicine-based therapies against various diseases and conditions. However, there still have significant issues concerning the translational development of ADSC-based therapies, such as its heterogeneity and being prone to aging. We developed a new simple and economical 3D semi-suspended expansion method in which 3D spheroids reside on an ADSC-derived self-feeder cell layer, producing cells with increased population homogeneity and strong stemness and ensuring that the proliferation and differentiation potency of the cells does not become notably reduced after at least ten passages in culture. To check the potential application of the 3D ADSC spheroids, we discovered that the combination of siEID3, which is a small interfering RNA of EP300 inhibitor of differentiation 3 (EID3), and laminin/poly-d-lysine matrix can rapidly result in trans-differentiation of the 3D spheroid cells to neural progenitor-like cells (NPLCs) in approximately 9 days in vitro. This approach provides a multidisciplinary tool for stem cell research and production in mesenchymal stem cell-related fields.
Collapse
Affiliation(s)
- Liang Luo
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi’an 710003, China; (W.Z.); (X.W.)
- Stem Cell Research Center, Neurosurgery Institute of PLA Army, Beijing 100700, China; (W.C.); (X.F.)
| | - Wei Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi’an 710003, China; (W.Z.); (X.W.)
- Department of Plastics and Aesthetic Surgery, the First Affiliated Hospital of Xi’an Medical University, Xi’an 710077, China
| | - Wenjin Chen
- Stem Cell Research Center, Neurosurgery Institute of PLA Army, Beijing 100700, China; (W.C.); (X.F.)
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510000, China
| | - Xiaojun Fu
- Stem Cell Research Center, Neurosurgery Institute of PLA Army, Beijing 100700, China; (W.C.); (X.F.)
- Bayi Brain Hospital, General Hospital of PLA Army, Beijing 100700, China
| | - Xujie Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi’an 710003, China; (W.Z.); (X.W.)
| | - Ruxiang Xu
- Stem Cell Research Center, Neurosurgery Institute of PLA Army, Beijing 100700, China; (W.C.); (X.F.)
- Department of Plastics and Aesthetic Surgery, the First Affiliated Hospital of Xi’an Medical University, Xi’an 710077, China
- The Department of Neurosurgery, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu 610072, China
| | - Dahai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi’an 710003, China; (W.Z.); (X.W.)
| |
Collapse
|
6
|
Middelkamp HHT, Verboven AHA, De Sá Vivas AG, Schoenmaker C, Klein Gunnewiek TM, Passier R, Albers CA, 't Hoen PAC, Nadif Kasri N, van der Meer AD. Cell type-specific changes in transcriptomic profiles of endothelial cells, iPSC-derived neurons and astrocytes cultured on microfluidic chips. Sci Rep 2021; 11:2281. [PMID: 33500551 PMCID: PMC7838281 DOI: 10.1038/s41598-021-81933-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 01/13/2021] [Indexed: 01/30/2023] Open
Abstract
In vitro neuronal models are essential for studying neurological physiology, disease mechanisms and potential treatments. Most in vitro models lack controlled vasculature, despite its necessity in brain physiology and disease. Organ-on-chip models offer microfluidic culture systems with dedicated micro-compartments for neurons and vascular cells. Such multi-cell type organs-on-chips can emulate neurovascular unit (NVU) physiology, however there is a lack of systematic data on how individual cell types are affected by culturing on microfluidic systems versus conventional culture plates. This information can provide perspective on initial findings of studies using organs-on-chip models, and further optimizes these models in terms of cellular maturity and neurovascular physiology. Here, we analysed the transcriptomic profiles of co-cultures of human induced pluripotent stem cell (hiPSC)-derived neurons and rat astrocytes, as well as one-day monocultures of human endothelial cells, cultured on microfluidic chips. For each cell type, large gene expression changes were observed when cultured on microfluidic chips compared to conventional culture plates. Endothelial cells showed decreased cell division, neurons and astrocytes exhibited increased cell adhesion, and neurons showed increased maturity when cultured on a microfluidic chip. Our results demonstrate that culturing NVU cell types on microfluidic chips changes their gene expression profiles, presumably due to distinct surface-to-volume ratios and substrate materials. These findings inform further NVU organ-on-chip model optimization and support their future application in disease studies and drug testing.
Collapse
Affiliation(s)
- H H T Middelkamp
- Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands.
- BIOS/Lab on a Chip, University of Twente, Enschede, The Netherlands.
| | - A H A Verboven
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands.
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands.
- Centre for Molecular and Biomolecular Informatics, Radboudumc, Radboud Institute for Molecular Life Sciences, 6500 HB, Nijmegen, The Netherlands.
| | - A G De Sá Vivas
- Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands
- BIOS/Lab on a Chip, University of Twente, Enschede, The Netherlands
| | - C Schoenmaker
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
| | - T M Klein Gunnewiek
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - R Passier
- Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - C A Albers
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Department of Molecular Developmental Biology, Radboud University, Nijmegen, The Netherlands
| | - P A C 't Hoen
- Centre for Molecular and Biomolecular Informatics, Radboudumc, Radboud Institute for Molecular Life Sciences, 6500 HB, Nijmegen, The Netherlands
| | - N Nadif Kasri
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
- Department of Cognitive Neurosciences, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - A D van der Meer
- Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands.
| |
Collapse
|
7
|
Wang L, Ye Z, Jang YY. Convergence of human pluripotent stem cell, organoid, and genome editing technologies. Exp Biol Med (Maywood) 2021; 246:861-875. [PMID: 33467883 DOI: 10.1177/1535370220985808] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The last decade has seen many exciting technological breakthroughs that greatly expanded the toolboxes for biological and biomedical research, yet few have had more impact than induced pluripotent stem cells and modern-day genome editing. These technologies are providing unprecedented opportunities to improve physiological relevance of experimental models, further our understanding of developmental processes, and develop novel therapies. One of the research areas that benefit greatly from these technological advances is the three-dimensional human organoid culture systems that resemble human tissues morphologically and physiologically. Here we summarize the development of human pluripotent stem cells and their differentiation through organoid formation. We further discuss how genetic modifications, genome editing in particular, were applied to answer basic biological and biomedical questions using organoid cultures of both somatic and pluripotent stem cell origins. Finally, we discuss the potential challenges of applying human pluripotent stem cell and organoid technologies for safety and efficiency evaluation of emerging genome editing tools.
Collapse
Affiliation(s)
- Lin Wang
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Zhaohui Ye
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Yoon-Young Jang
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, John Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
8
|
Amores de Sousa MC, Rodrigues CAV, Ferreira IAF, Diogo MM, Linhardt RJ, Cabral JMS, Ferreira FC. Functionalization of Electrospun Nanofibers and Fiber Alignment Enhance Neural Stem Cell Proliferation and Neuronal Differentiation. Front Bioeng Biotechnol 2020; 8:580135. [PMID: 33195141 PMCID: PMC7649414 DOI: 10.3389/fbioe.2020.580135] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 09/24/2020] [Indexed: 01/29/2023] Open
Abstract
Neural stem cells (NSCs) have the potential to generate the cells of the nervous system and, when cultured on nanofiber scaffolds, constitute a promising approach for neural tissue engineering. In this work, the impact of combining nanofiber alignment with functionalization of the electrospun poly-ε-caprolactone (PCL) nanofibers with biological adhesion motifs on the culture of an NSC line (CGR8-NS) is evaluated. A five-rank scale for fiber density was introduced, and a 4.5 level, corresponding to 70–80% fiber density, was selected for NSC in vitro culture. Aligned nanofibers directed NSC distribution and, especially in the presence of laminin (PCL-LN) and the RGD-containing peptide GRGDSP (PCL-RGD), promoted higher cell elongation, quantified by the eccentricity and axis ratio. In situ differentiation resulted in relatively higher percentage of cells expressing Tuj1 in PCL-LN, as well as significantly longer neurite development (41.1 ± 1.0 μm) than PCL-RGD (32.0 ± 1.0 μm), pristine PCL (25.1 ± 1.2 μm), or PCL-RGD randomly oriented fibers (26.5 ± 1.4 μm), suggesting that the presence of LN enhances neuronal differentiation. This study demonstrates that aligned nanofibers, functionalized with RGD, perform as well as PCL-LN fibers in terms of cell adhesion and proliferation. The presence of the full LN protein improves neuronal differentiation outcomes, which may be important for the use of this system in tissue engineering applications.
Collapse
Affiliation(s)
- Miriam C Amores de Sousa
- Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Carlos A V Rodrigues
- Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Inês A F Ferreira
- Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Maria Margarida Diogo
- Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Robert J Linhardt
- Center for Biotechnology and Interdisciplinary Studies, Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, NY, United States
| | - Joaquim M S Cabral
- Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Frederico Castelo Ferreira
- Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
9
|
Gautam J, Alaref A, Hassan A, Sharma Kandel R, Mishra R, Jahan N. Safety and Efficacy of Stem Cell Therapy in Patients With Ischemic Stroke. Cureus 2020; 12:e9917. [PMID: 32968578 PMCID: PMC7505641 DOI: 10.7759/cureus.9917] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 08/21/2020] [Indexed: 01/08/2023] Open
Abstract
Stem cell therapy is emerging as a promising treatment strategy to treat patients with stroke. While there are established modes of treatment for stroke patients such as thrombolysis and endovascular intervention, most of the stroke patients frequently end up with major residual deficits or even death. The use of stem cells to treat stroke has been found to be beneficial in the animal models but strict evidence for the same in humans is still lacking. We reviewed 13 clinical trials of stem cell therapy in stroke patients conducted between 2014 and 2020 based on the search using the database PubMed, and the clinical trial registry (www.clinicaltrials.gov). We aimed to assess the safety and efficacy of stem cell treatment in stroke patients who participated in the trials. Quality assessment of the clinical trials revealed a sub-optimal score. We found mixed results regarding the efficacy of stem cells in the treatment of ischemic stroke although we could not do a quantitative analysis of the effect outcomes. Assessment for safety revealed promising results as there were only minor side effects related to cell therapy. Although stem cell therapy seems to be a promising strategy to treat stroke patients in the future, we concluded that the field needs more evidence regarding the safety and efficacy of the use of stem cells in stroke patients before we use them in the clinic.
Collapse
Affiliation(s)
- Jeevan Gautam
- Neurology, California Instititute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Amer Alaref
- Diagnostic Radiology, California institute of Behavioural Neurosciences and Psychology, Fairfield, USA
- Diagnostic Radiology, Thunder Bay Regional Health Sciences Centre, Thunder Bay, CAN
- Diagnostic Imaging, Northern Ontario School of Medicine, Sudbury, CAN
- Breast Imaging Services, Linda Buchan Centre, Thunder Bay, CAN
| | - Abdallah Hassan
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Rajan Sharma Kandel
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Rohi Mishra
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Nusrat Jahan
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
10
|
Sex Stratified Treatment of Neurological Disorders: Challenges and Perspectives. Brain Sci 2020; 10:brainsci10020103. [PMID: 32075025 PMCID: PMC7071507 DOI: 10.3390/brainsci10020103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 02/13/2020] [Indexed: 11/17/2022] Open
Abstract
Despite the obvious sex differences in many of the most common neuropsychiatric and neurodegenerative disorders, males and females are still often treated the same from a clinical perspective. Why is that? The simple answer is that there is still too little known about this very complex matter. Sex hormone signaling, genetic sex, sex-biased comorbidities, and social gender perceptions all interact, making discrimination between the impacts of each of these factors hard to evaluate. Adding to the complexity is that important species differences must be taken into account when interpreting data from animal models. Clearly, to overcome this, larger efforts are needed that incorporate epidemiological, experimental, and clinical data to provide a solid scientific base for more personalized and informed clinical decisions that will benefit both men and women suffering from neurological disorders.
Collapse
|
11
|
Pacitti D, Privolizzi R, Bax BE. Organs to Cells and Cells to Organoids: The Evolution of in vitro Central Nervous System Modelling. Front Cell Neurosci 2019; 13:129. [PMID: 31024259 PMCID: PMC6465581 DOI: 10.3389/fncel.2019.00129] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 03/14/2019] [Indexed: 02/05/2023] Open
Abstract
With 100 billion neurons and 100 trillion synapses, the human brain is not just the most complex organ in the human body, but has also been described as "the most complex thing in the universe." The limited availability of human living brain tissue for the study of neurogenesis, neural processes and neurological disorders has resulted in more than a century-long strive from researchers worldwide to model the central nervous system (CNS) and dissect both its striking physiology and enigmatic pathophysiology. The invaluable knowledge gained with the use of animal models and post mortem human tissue remains limited to cross-species similarities and structural features, respectively. The advent of human induced pluripotent stem cell (hiPSC) and 3-D organoid technologies has revolutionised the approach to the study of human brain and CNS in vitro, presenting great potential for disease modelling and translational adoption in drug screening and regenerative medicine, also contributing beneficially to clinical research. We have surveyed more than 100 years of research in CNS modelling and provide in this review an historical excursus of its evolution, from early neural tissue explants and organotypic cultures, to 2-D patient-derived cell monolayers, to the latest development of 3-D cerebral organoids. We have generated a comprehensive summary of CNS modelling techniques and approaches, protocol refinements throughout the course of decades and developments in the study of specific neuropathologies. Current limitations and caveats such as clonal variation, developmental stage, validation of pluripotency and chromosomal stability, functional assessment, reproducibility, accuracy and scalability of these models are also discussed.
Collapse
Affiliation(s)
- Dario Pacitti
- Molecular and Clinical Sciences Research Institute, St George’s, University of London, London, United Kingdom
- College of Medicine and Health, St Luke’s Campus, University of Exeter, Exeter, United Kingdom
| | - Riccardo Privolizzi
- Gene Transfer Technology Group, Institute for Women’s Health, University College London, London, United Kingdom
| | - Bridget E. Bax
- Molecular and Clinical Sciences Research Institute, St George’s, University of London, London, United Kingdom
| |
Collapse
|
12
|
Yan Y, Bejoy J, Marzano M, Li Y. The Use of Pluripotent Stem Cell-Derived Organoids to Study Extracellular Matrix Development during Neural Degeneration. Cells 2019; 8:E242. [PMID: 30875781 PMCID: PMC6468789 DOI: 10.3390/cells8030242] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/06/2019] [Accepted: 03/08/2019] [Indexed: 12/30/2022] Open
Abstract
The mechanism that causes the Alzheimer's disease (AD) pathologies, including amyloid plaque, neurofibrillary tangles, and neuron death, is not well understood due to the lack of robust study models for human brain. Three-dimensional organoid systems based on human pluripotent stem cells (hPSCs) have shown a promising potential to model neurodegenerative diseases, including AD. These systems, in combination with engineering tools, allow in vitro generation of brain-like tissues that recapitulate complex cell-cell and cell-extracellular matrix (ECM) interactions. Brain ECMs play important roles in neural differentiation, proliferation, neuronal network, and AD progression. In this contribution related to brain ECMs, recent advances in modeling AD pathology and progression based on hPSC-derived neural cells, tissues, and brain organoids were reviewed and summarized. In addition, the roles of ECMs in neural differentiation of hPSCs and the influences of heparan sulfate proteoglycans, chondroitin sulfate proteoglycans, and hyaluronic acid on the progression of neurodegeneration were discussed. The advantages that use stem cell-based organoids to study neural degeneration and to investigate the effects of ECM development on the disease progression were highlighted. The contents of this article are significant for understanding cell-matrix interactions in stem cell microenvironment for treating neural degeneration.
Collapse
Affiliation(s)
- Yuanwei Yan
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA.
| | - Julie Bejoy
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA.
| | - Mark Marzano
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA.
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA.
| |
Collapse
|
13
|
Keller JM, Frega M. Past, Present, and Future of Neuronal Models In Vitro. ADVANCES IN NEUROBIOLOGY 2019; 22:3-17. [PMID: 31073930 DOI: 10.1007/978-3-030-11135-9_1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Over the past century, robust methods were developed that enable the isolation, culture, and dynamic observation of mammalian neuronal networks in vitro. But even if neuronal culture cannot yet fully recapitulate the normal brain, the knowledge that has been acquired from these surrogate in vitro models is invaluable. Indeed, neuronal culture has continued to propel basic neuroscience research, proving that in vitro systems have legitimacy when it comes to studying either the healthy or diseased human brain. Furthermore, scientific advancement typically parallels technical refinements in the field. A pertinent example is that a collective drive in the field of neuroscience to better understand the development, organization, and emergent properties of neuronal networks is being facilitated by progressive advances in micro-electrode array (MEA) technology. In this chapter, we briefly review the emergence of neuronal cell culture as a technique, the current trends in human stem cell-based modeling, and the technologies used to monitor neuronal communication. We conclude by highlighting future prospects that are evolving specifically out of the combination of human neuronal models and MEA technology.
Collapse
Affiliation(s)
- Jason M Keller
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen, The Netherlands
| | - Monica Frega
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen, The Netherlands. .,Department of Clinical Neurophysiology, University of Twente, Enschede, The Netherlands.
| |
Collapse
|
14
|
Mishima T, Fujioka S, Fukae J, Yuasa-Kawada J, Tsuboi Y. Modeling Parkinson's Disease and Atypical Parkinsonian Syndromes Using Induced Pluripotent Stem Cells. Int J Mol Sci 2018; 19:ijms19123870. [PMID: 30518093 PMCID: PMC6321610 DOI: 10.3390/ijms19123870] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 11/11/2018] [Accepted: 11/28/2018] [Indexed: 12/31/2022] Open
Abstract
Parkinson’s disease (PD) and atypical parkinsonian syndromes are age-dependent multifactorial neurodegenerative diseases, which are clinically characterized by bradykinesia, tremor, muscle rigidity and postural instability. Although these diseases share several common clinical phenotypes, their pathophysiological aspects vary among the disease categories. Extensive animal-based approaches, as well as postmortem studies, have provided important insights into the disease mechanisms and potential therapeutic targets. However, the exact pathological mechanisms triggering such diseases still remain elusive. Furthermore, the effects of drugs observed in animal models are not always reproduced in human clinical trials. By using induced pluripotent stem cell (iPSC) technology, it has become possible to establish patient-specific iPSCs from their somatic cells and to effectively differentiate these iPSCs into different types of neurons, reproducing some key aspects of the disease phenotypes in vitro. In this review, we summarize recent findings from iPSC-based modeling of PD and several atypical parkinsonian syndromes including multiple system atrophy, frontotemporal dementia and parkinsonism linked to chromosome 17 and Perry syndrome. Furthermore, we discuss future challenges and prospects for modeling and understanding PD and atypical parkinsonian syndromes.
Collapse
Affiliation(s)
- Takayasu Mishima
- Department of Neurology, Fukuoka University, Fukuoka 814-0180, Japan.
| | - Shinsuke Fujioka
- Department of Neurology, Fukuoka University, Fukuoka 814-0180, Japan.
| | - Jiro Fukae
- Department of Neurology, Fukuoka University, Fukuoka 814-0180, Japan.
| | | | - Yoshio Tsuboi
- Department of Neurology, Fukuoka University, Fukuoka 814-0180, Japan.
| |
Collapse
|
15
|
Ranjan VD, Qiu L, Tan EK, Zeng L, Zhang Y. Modelling Alzheimer's disease: Insights from in vivo to in vitro three-dimensional culture platforms. J Tissue Eng Regen Med 2018; 12:1944-1958. [PMID: 30011422 DOI: 10.1002/term.2728] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 06/21/2018] [Accepted: 07/04/2018] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia and is characterized by progressive memory loss, impairment of other cognitive functions, and inability to perform activities of daily life. The key to understanding AD aetiology lies in the development of effective disease models, which should ideally recapitulate all aspects pertaining to the disease. A plethora of techniques including in vivo, in vitro, and in silico platforms have been utilized in developing disease models of AD over the years. Each of these approaches has revealed certain essential characteristics of AD; however, none have managed to fully mimic the pathological hallmarks observed in the AD human brain. In this review, we will provide details into the genesis, evolution, and significance of the principal methods currently employed in modelling AD, the advantages and limitations faced in their application, including the headways made by each approach. This review will focus primarily on two-dimensional and three-dimensional in vitro modelling of AD, which during the last few years has made significant breakthroughs in the areas of AD pathology and therapeutic screening. In addition, a glimpse into state-of-the-art neural tissue engineering techniques incorporating biomaterials and microfluidics technologies is provided, which could pave the way for the development of more accurate and comprehensive AD models in the future.
Collapse
Affiliation(s)
- Vivek Damodar Ranjan
- NTU Institute for Health Technologies, Interdisciplinary Graduate School, Nanyang Technological University, Singapore.,School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore.,Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
| | - Lifeng Qiu
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
| | - Eng King Tan
- Department of Neurology, National Neuroscience Institute, Singapore.,Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore
| | - Li Zeng
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore.,Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore
| | - Yilei Zhang
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore
| |
Collapse
|
16
|
Joshi R, Thakuri PS, Buchanan JC, Li J, Tavana H. Microprinted Stem Cell Niches Reveal Compounding Effect of Colony Size on Stromal Cells-Mediated Neural Differentiation. Adv Healthc Mater 2018; 7:10.1002/adhm.201700832. [PMID: 29193846 PMCID: PMC5842135 DOI: 10.1002/adhm.201700832] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/02/2017] [Indexed: 01/30/2023]
Abstract
Microenvironmental factors have a major impact on differentiation of embryonic stem cells (ESCs). Here, a novel phenomenon that size of ESC colonies has a significant regulatory role on stromal cells induced differentiation of ESCs to neural cells is reported. Using a robotic cell microprinting technology, defined densities of ESCs are confined within aqueous nanodrops over a layer of supporting stromal cells immersed in a second, immiscible aqueous phase to generate ESC colonies of defined sizes. Temporal protein and gene expression studies demonstrate that larger ESC colonies generate disproportionally more neural cells and longer neurite processes. Unlike previous studies that attribute neural differentiation of ESCs solely to interactions with stromal cells, it is found that increased intercellular signaling of ESCs significantly enhances neural differentiation. This study offers an approach to generate neural cells with improved efficiency for potential use in translational research.
Collapse
Affiliation(s)
- Ramila Joshi
- Department of Biomedical Engineering, The University of Akron, Akron, OH, 44325, USA
| | - Pradip Shahi Thakuri
- Department of Biomedical Engineering, The University of Akron, Akron, OH, 44325, USA
| | - James C Buchanan
- Department of Biomedical Engineering, The University of Akron, Akron, OH, 44325, USA
| | - Jun Li
- Department of Mathematical Sciences, Kent State University, Kent, OH, 44242, USA
| | - Hossein Tavana
- Department of Biomedical Engineering, The University of Akron, 260 S. Forge St., Akron, OH, 44325, USA
| |
Collapse
|
17
|
McGrath EL, Gao J, Wu P. Zika Virus Infection of Cultured Human Fetal Brain Neural Stem Cells for Immunocytochemical Analysis. J Vis Exp 2018. [PMID: 29443111 DOI: 10.3791/56917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Human fetal brain neural stem cells are a unique non-genetically modified model system to study the impact of various stimuli on human developmental neurobiology. Rather than use an animal model or genetically modified induced pluripotent cells, human neural stem cells provide an effective in vitro system to examine the effects of treatments, screen drugs, or examine individual differences. Here, we provide the detailed protocols for methods used to expand human fetal brain neural stem cells in culture with serum-free media, to differentiate them into various neuronal subtypes and astrocytes via different priming procedures, and to freeze and recover these cells. Furthermore, we describe a procedure of using human fetal brain neural stem cells to study Zika virus infection.
Collapse
Affiliation(s)
- Erica L McGrath
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch; Department of Neurology, Johns Hopkins University
| | - Junling Gao
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch
| | - Ping Wu
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch; Beijing Institute for Brain Disorders, Capital Medical University;
| |
Collapse
|
18
|
Sun AX, Ng HH, Tan EK. Translational potential of human brain organoids. Ann Clin Transl Neurol 2018; 5:226-235. [PMID: 29468184 PMCID: PMC5817829 DOI: 10.1002/acn3.505] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 10/07/2017] [Accepted: 10/21/2017] [Indexed: 12/11/2022] Open
Abstract
The recent technology of 3D cultures of cellular aggregates derived from human stem cells have led to the emergence of tissue‐like structures of various organs including the brain. Brain organoids bear molecular and structural resemblance with developing human brains, and have been demonstrated to recapitulate several physiological and pathological functions of the brain. Here we provide an overview of the development of brain organoids for the clinical community, focusing on the current status of the field with an critical evaluation of its translational value.
Collapse
Affiliation(s)
- Alfred X Sun
- Stem Cell and Regenerative Biology Genome Institute of Singapore 60 Biopolis
Street Singapore 138672 Singapore.,Department of Neurology National Neuroscience Institute 20 College Road Singapore 169856 Singapore
| | - Huck-Hui Ng
- Stem Cell and Regenerative Biology Genome Institute of Singapore 60 Biopolis
Street Singapore 138672 Singapore.,Graduate School for integrative Sciences and Engineering National University of Singapore Singapore 117456 Singapore
| | - Eng-King Tan
- Department of Neurology National Neuroscience Institute 20 College Road Singapore 169856 Singapore
| |
Collapse
|
19
|
Venkatesh K, Reddy LVK, Abbas S, Mullick M, Moghal ETB, Balakrishna JP, Sen D. NOTCH Signaling Is Essential for Maturation, Self-Renewal, and Tri-Differentiation of In Vitro Derived Human Neural Stem Cells. Cell Reprogram 2017; 19:372-383. [DOI: 10.1089/cell.2017.0009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Katari Venkatesh
- Cellular and Molecular Therapeutics Laboratory, Centre for Biomaterials, Cellular and Molecular Theranostics, Vellore Institute of Technology (VIT) University, Vellore, India
| | - L. Vinod Kumar Reddy
- Cellular and Molecular Therapeutics Laboratory, Centre for Biomaterials, Cellular and Molecular Theranostics, Vellore Institute of Technology (VIT) University, Vellore, India
| | - Salar Abbas
- Centre for Stem Cell Research, Christian Medical College, Vellore, India
| | - Madhubanti Mullick
- Cellular and Molecular Therapeutics Laboratory, Centre for Biomaterials, Cellular and Molecular Theranostics, Vellore Institute of Technology (VIT) University, Vellore, India
| | - Erfath Thanjeem Begum Moghal
- Cellular and Molecular Therapeutics Laboratory, Centre for Biomaterials, Cellular and Molecular Theranostics, Vellore Institute of Technology (VIT) University, Vellore, India
| | | | - Dwaipayan Sen
- Cellular and Molecular Therapeutics Laboratory, Centre for Biomaterials, Cellular and Molecular Theranostics, Vellore Institute of Technology (VIT) University, Vellore, India
| |
Collapse
|
20
|
Sarmah D, Kaur H, Saraf J, Pravalika K, Goswami A, Kalia K, Borah A, Wang X, Dave KR, Yavagal DR, Bhattacharya P. Getting Closer to an Effective Intervention of Ischemic Stroke: The Big Promise of Stem Cell. Transl Stroke Res 2017; 9:356-374. [PMID: 29075984 DOI: 10.1007/s12975-017-0580-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 10/12/2017] [Accepted: 10/17/2017] [Indexed: 12/13/2022]
Abstract
Stem cell therapy for ischemic stroke has widely been explored. Results from both preclinical and clinical studies have immensely supported the judicious use of stem cells as therapy. These provide an attractive means for preserving and replacing the damaged brain tissues following an ischemic attack. Since the past few years, researchers have used various types of stem cells to replenish insulted neuronal and glial cells in neurological disorders. In the present review, we discuss different types of stem cells employed for the treatment of ischemic stroke and mechanisms and challenges these cells face once introduced into the living system. Further, we also present different ways to maneuver and overcome challenges to translate the advances made at the preclinical level to clinics.
Collapse
Affiliation(s)
- Deepaneeta Sarmah
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, Gandhinagar, Gujarat, 382355, India
| | - Harpreet Kaur
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, Gandhinagar, Gujarat, 382355, India
| | - Jackson Saraf
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, Gandhinagar, Gujarat, 382355, India
| | - Kanta Pravalika
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, Gandhinagar, Gujarat, 382355, India
| | - Avirag Goswami
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kiran Kalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, Gandhinagar, Gujarat, 382355, India
| | - Anupom Borah
- Cellular and Molecular Neurobiology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, Assam, India
| | - Xin Wang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kunjan R Dave
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Dileep R Yavagal
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Pallab Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, Gandhinagar, Gujarat, 382355, India.
- Department of Neurosurgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
21
|
Aldana BI, Zhang Y, Lihme MF, Bak LK, Nielsen JE, Holst B, Hyttel P, Freude KK, Waagepetersen HS. Characterization of energy and neurotransmitter metabolism in cortical glutamatergic neurons derived from human induced pluripotent stem cells: A novel approach to study metabolism in human neurons. Neurochem Int 2017; 106:48-61. [DOI: 10.1016/j.neuint.2017.02.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 01/19/2017] [Accepted: 02/20/2017] [Indexed: 02/01/2023]
|
22
|
Obora K, Onodera Y, Takehara T, Frampton J, Hasei J, Ozaki T, Teramura T, Fukuda K. Inflammation-induced miRNA-155 inhibits self-renewal of neural stem cells via suppression of CCAAT/enhancer binding protein β (C/EBPβ) expression. Sci Rep 2017; 7:43604. [PMID: 28240738 PMCID: PMC5378916 DOI: 10.1038/srep43604] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/24/2017] [Indexed: 01/04/2023] Open
Abstract
Intracerebral inflammation resulting from injury or disease is implicated in disruption of neural regeneration and may lead to irreversible neuronal dysfunction. Analysis of inflammation-related microRNA profiles in various tissues, including the brain, has identified miR-155 among the most prominent miRNAs linked to inflammation. Here, we hypothesize that miR-155 mediates inflammation-induced suppression of neural stem cell (NSC) self-renewal. Using primary mouse NSCs and human NSCs derived from induced pluripotent stem (iPS) cells, we demonstrate that three important genes involved in NSC self-renewal (Msi1, Hes1 and Bmi1) are suppressed by miR-155. We also demonstrate that suppression of self-renewal genes is mediated by the common transcription factor C/EBPβ, which is a direct target of miR-155. Our study describes an axis linking inflammation and miR-155 to expression of genes related to NSC self-renewal, suggesting that regulation of miR-155 may hold potential as a novel therapeutic strategy for treating neuroinflammatory diseases.
Collapse
Affiliation(s)
- Kayoko Obora
- Department of Rehabilitation Medicine, Kindai University Faculty of Medicine, Osaka, Japan
| | - Yuta Onodera
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University Faculty of Medicine, Osaka, Japan
| | - Toshiyuki Takehara
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University Faculty of Medicine, Osaka, Japan
| | - John Frampton
- School of Biomedical Engineering, Dalhousie University. Halifax, Nova Scotia, Canada
| | - Joe Hasei
- Science of Functional Recovery and Reconstruction, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Toshifumi Ozaki
- Science of Functional Recovery and Reconstruction, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takeshi Teramura
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University Faculty of Medicine, Osaka, Japan
| | - Kanji Fukuda
- Department of Rehabilitation Medicine, Kindai University Faculty of Medicine, Osaka, Japan.,Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University Faculty of Medicine, Osaka, Japan
| |
Collapse
|
23
|
Vagaska B, Ferretti P. Toward modeling the human nervous system in a dish: recent progress and outstanding challenges. Regen Med 2016; 12:15-23. [PMID: 27900887 DOI: 10.2217/rme-2016-0106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Studying the cellular and molecular bases governing development, and normal and abnormal functions of the human CNS is hampered by its complexity and the very limited possibility of experimentally manipulating it in vivo. Development of 3D, tissue-like culture systems offers much promise for boosting our understanding of human neural development, birth defects, neurodegenerative diseases and neural injury, and for providing platforms that will more accurately predict efficacy of putative therapeutic compounds and assess responses to potentially neurotoxic agents. Although novel technological developments and a more interdisciplinary approach to modeling the human CNS are accelerating the pace of discovery, increasing the complexity of in vitro systems increases the ordeals to be overcome to establish highly reproducible models amenable to quantitative analysis.
Collapse
Affiliation(s)
- Barbora Vagaska
- Stem Cell & Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Patrizia Ferretti
- Stem Cell & Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| |
Collapse
|
24
|
The emerging role of in vitro electrophysiological methods in CNS safety pharmacology. J Pharmacol Toxicol Methods 2016; 81:47-59. [DOI: 10.1016/j.vascn.2016.03.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/30/2016] [Accepted: 03/30/2016] [Indexed: 01/16/2023]
|
25
|
Kim SM, Kim JW, Kwak TH, Park SW, Kim KP, Park H, Lim KT, Kang K, Kim J, Yang JH, Han H, Lee I, Hyun JK, Bae YM, Schöler HR, Lee HT, Han DW. Generation of Integration-free Induced Neural Stem Cells from Mouse Fibroblasts. J Biol Chem 2016; 291:14199-14212. [PMID: 27189941 DOI: 10.1074/jbc.m115.713578] [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: 12/31/2015] [Indexed: 01/10/2023] Open
Abstract
The viral vector-mediated overexpression of the defined transcription factors, Brn4/Pou3f4, Sox2, Klf4, and c-Myc (BSKM), could induce the direct conversion of somatic fibroblasts into induced neural stem cells (iNSCs). However, viral vectors may be randomly integrated into the host genome thereby increasing the risk for undesired genotoxicity, mutagenesis, and tumor formation. Here we describe the generation of integration-free iNSCs from mouse fibroblasts by non-viral episomal vectors containing BSKM. The episomal vector-derived iNSCs (e-iNSCs) closely resemble control NSCs, and iNSCs generated by retrovirus (r-iNSCs) in morphology, gene expression profile, epigenetic status, and self-renewal capacity. The e-iNSCs are functionally mature, as they could differentiate into all the neuronal cell types both in vitro and in vivo Our study provides a novel concept for generating functional iNSCs using a non-viral, non-integrating, plasmid-based system that could facilitate their biomedical applicability.
Collapse
Affiliation(s)
- Sung Min Kim
- Department of Stem Cell Biology, School of Medicine, 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea; Department of Animal Biotechnology, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jong-Wan Kim
- Department of Nanobiomedical Science, Dankook University Graduate School, Cheonan 31116, Republic of Korea
| | - Tae Hwan Kwak
- Department of Stem Cell Biology, School of Medicine, 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Sang Woong Park
- Department of Physiology, School of Medicine, Konkuk University, Chungju, Chungbuk 27478, Republic of Korea
| | - Kee-Pyo Kim
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Hyunji Park
- Department of Physiology, School of Medicine, Konkuk University, Chungju, Chungbuk 27478, Republic of Korea
| | - Kyung Tae Lim
- Department of Stem Cell Biology, School of Medicine, 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Kyuree Kang
- Department of Stem Cell Biology, School of Medicine, 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jonghun Kim
- Department of Stem Cell Biology, School of Medicine, 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Ji Hun Yang
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Heonjong Han
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 04056, Korea
| | - Insuk Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 04056, Korea
| | - Jung Keun Hyun
- Department of Nanobiomedical Science, Dankook University Graduate School, Cheonan 31116, Republic of Korea
| | - Young Min Bae
- Department of Physiology, School of Medicine, Konkuk University, Chungju, Chungbuk 27478, Republic of Korea
| | - Hans R Schöler
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany,; University of Münster, Medical Faculty, Domagkstraße 3, 48149 Münster, Germany
| | - Hoon Taek Lee
- Department of Animal Biotechnology, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Dong Wook Han
- Department of Stem Cell Biology, School of Medicine, 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea; KU Open-Innovation Center, Institute of Biomedical Science & Technology, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
26
|
Using Stem Cells to Grow Artificial Tissue for Peripheral Nerve Repair. Stem Cells Int 2016; 2016:7502178. [PMID: 27212954 PMCID: PMC4861803 DOI: 10.1155/2016/7502178] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 02/17/2016] [Accepted: 03/02/2016] [Indexed: 12/17/2022] Open
Abstract
Peripheral nerve injury continues to pose a clinical hurdle despite its frequency and advances in treatment. Unlike the central nervous system, neurons of the peripheral nervous system have a greater ability to regenerate. However, due to a number of confounding factors, this is often both incomplete and inadequate. The lack of supportive Schwann cells or their inability to maintain a regenerative phenotype is a major factor. Advances in nervous system tissue engineering technology have led to efforts to build Schwann cell scaffolds to overcome this and enhance the regenerative capacity of neurons following injury. Stem cells that can differentiate along a neural lineage represent an essential resource and starting material for this process. In this review, we discuss the different stem cell types that are showing promise for nervous system tissue engineering in the context of peripheral nerve injury. We also discuss some of the biological, practical, ethical, and commercial considerations in using these different stem cells for future clinical application.
Collapse
|
27
|
Urnukhsaikhan E, Cho H, Mishig-Ochir T, Seo YK, Park JK. Pulsed electromagnetic fields promote survival and neuronal differentiation of human BM-MSCs. Life Sci 2016; 151:130-138. [PMID: 26898125 DOI: 10.1016/j.lfs.2016.02.066] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 02/15/2016] [Accepted: 02/16/2016] [Indexed: 12/11/2022]
Abstract
Pulsed electromagnetic fields (PEMF) are known to affect biological properties such as differentiation, regulation of transcription factor and cell proliferation. However, the cell-protective effect of PEMF exposure is largely unknown. The aim of this study is to understand the mechanisms underlying PEMF-mediated suppression of apoptosis and promotion of survival, including PEMF-induced neuronal differentiation. Treatment of induced human BM-MSCs with PEMF increased the expression of neural markers such as NF-L, NeuroD1 and Tau. Moreover, treatment of induced human BM-MSCs with PEMF greatly decreased cell death in a dose- and time-dependent manner. There is evidence that Akt and Ras are involved in neuronal survival and protection. Activation of Akt and Ras results in the regulation of survival proteins such as Bad and Bcl-xL. Thus, the Akt/Ras signaling pathway may be a desirable target for enhancing cell survival and treatment of neurological disease. Our analyses indicated that PEMF exposure dramatically increased the activity of Akt, Rsk, Creb, Erk, Bcl-xL and Bad via phosphorylation. PEMF-dependent cell protection was reversed by pretreatment with LY294002, a specific inhibitor of phosphatidylinositol 3-kinase (PI3K). Our data suggest that the PI3K/Akt/Bad signaling pathway may be a possible mechanism for the cell-protective effects of PEMF.
Collapse
Affiliation(s)
| | - Hyunjin Cho
- Dongguk University Research Institute of Biotechnology, Republic of Korea
| | | | - Young-Kwon Seo
- Department of Medical Biotechnology, Dongguk University, Seoul, Republic of Korea
| | - Jung-Kueg Park
- Department of Medical Biotechnology, Dongguk University, Seoul, Republic of Korea
| |
Collapse
|
28
|
Iraci N, Leonardi T, Gessler F, Vega B, Pluchino S. Focus on Extracellular Vesicles: Physiological Role and Signalling Properties of Extracellular Membrane Vesicles. Int J Mol Sci 2016; 17:171. [PMID: 26861302 PMCID: PMC4783905 DOI: 10.3390/ijms17020171] [Citation(s) in RCA: 220] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 09/24/2015] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) are a heterogeneous population of secreted membrane vesicles, with distinct biogenesis routes, biophysical properties and different functions both in physiological conditions and in disease. The release of EVs is a widespread biological process, which is conserved across species. In recent years, numerous studies have demonstrated that several bioactive molecules are trafficked with(in) EVs, such as microRNAs, mRNAs, proteins and lipids. The understanding of their final impact on the biology of specific target cells remains matter of intense debate in the field. Also, EVs have attracted great interest as potential novel cell-free therapeutics. Here we describe the proposed physiological and pathological functions of EVs, with a particular focus on their molecular content. Also, we discuss the advances in the knowledge of the mechanisms regulating the secretion of EV-associated molecules and the specific pathways activated upon interaction with the target cell, highlighting the role of EVs in the context of the immune system and as mediators of the intercellular signalling in the brain.
Collapse
Affiliation(s)
- Nunzio Iraci
- Wellcome Trust-Medical Research Council Stem Cell Institute, Clifford Allbutt Building-Cambridge Biosciences Campus, Department of Clinical Neurosciences, and NIHR Biomedical Research Centre, University of Cambridge, Hills Road CB2 0PY, UK.
| | - Tommaso Leonardi
- Wellcome Trust-Medical Research Council Stem Cell Institute, Clifford Allbutt Building-Cambridge Biosciences Campus, Department of Clinical Neurosciences, and NIHR Biomedical Research Centre, University of Cambridge, Hills Road CB2 0PY, UK.
- EMBL-European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SD, UK.
| | - Florian Gessler
- Wellcome Trust-Medical Research Council Stem Cell Institute, Clifford Allbutt Building-Cambridge Biosciences Campus, Department of Clinical Neurosciences, and NIHR Biomedical Research Centre, University of Cambridge, Hills Road CB2 0PY, UK.
| | - Beatriz Vega
- Wellcome Trust-Medical Research Council Stem Cell Institute, Clifford Allbutt Building-Cambridge Biosciences Campus, Department of Clinical Neurosciences, and NIHR Biomedical Research Centre, University of Cambridge, Hills Road CB2 0PY, UK.
| | - Stefano Pluchino
- Wellcome Trust-Medical Research Council Stem Cell Institute, Clifford Allbutt Building-Cambridge Biosciences Campus, Department of Clinical Neurosciences, and NIHR Biomedical Research Centre, University of Cambridge, Hills Road CB2 0PY, UK.
| |
Collapse
|
29
|
Lim MS, Shin MS, Lee SY, Minn YK, Hoh JK, Cho YH, Kim DW, Lee SH, Kim CH, Park CH. Noggin Over-Expressing Mouse Embryonic Fibroblasts and MS5 Stromal Cells Enhance Directed Differentiation of Dopaminergic Neurons from Human Embryonic Stem Cells. PLoS One 2015; 10:e0138460. [PMID: 26383864 PMCID: PMC4575120 DOI: 10.1371/journal.pone.0138460] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 08/31/2015] [Indexed: 11/18/2022] Open
Abstract
Directed methods for differentiating human embryonic stem cells (hESCs) into dopaminergic (DA) precursor cells using stromal cells co-culture systems are already well established. However, not all of the hESCs differentiate into DA precursors using these methods. HSF6, H1, H7, and H9 cells differentiate well into DA precursors, but CHA13 and CHA15 cells hardly differentiate. To overcome this problem, we modified the differentiation system to include a co-culturing step that exposes the cells to noggin early in the differentiation process. This was done using γ-irradiated noggin-overexpressing CF1-mouse embryonic fibroblasts (MEF-noggin) and MS5 stromal cells (MS5-noggin and MS5-sonic hedgehog). After directed differentiation, RT-PCR analyses revealed that engrailed-1 (En-1), Lmx1b, and Nurr1, which are midbrain DA markers, were expressed regardless of differentiation stage. Moreover, tyrosine hydroxylase (Th) and an A9 midbrain-specific DA marker (Girk2) were expressed during differentiation, whereas levels of Oct3/4, an undifferentiated marker, decreased. Immunocytochemical analyses revealed that protein levels of the neuronal markers TH and TuJ1 increased during the final differentiation stage. These results demonstrate that early noggin exposure may play a specific role in the directed differentiation of DA cells from human embryonic stem cells.
Collapse
Affiliation(s)
- Mi-Sun Lim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Korea
| | - Min-Seop Shin
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea
| | - Soo Young Lee
- Department of Microbiology, College of Medicine, Hanyang University, Seoul, Korea
| | - Yang-Ki Minn
- Department of Neurology, Hangang Sacred Heart Hospital, Hallym University, Seoul, Korea
| | - Jeong-Kyu Hoh
- Department of Obstetrics and Gynecology, College of Medicine, Hanyang University, Seoul, Korea
| | - Youl-Hee Cho
- Department of Medical Genetics, College of Medicine, Hanyang University, Seoul, Korea
| | - Dong-Wook Kim
- Department of Physiology and Cell Therapy Center, Yonsei University College of Medicine, Seoul, Korea
| | - Sang-Hun Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Korea
| | - Chun-Hyung Kim
- Institute of Green Bio Science and Technology, Seoul National University, Pyeongchang, Korea
- * E-mail: (CHK); (CHP)
| | - Chang-Hwan Park
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Korea
- Department of Microbiology, College of Medicine, Hanyang University, Seoul, Korea
- * E-mail: (CHK); (CHP)
| |
Collapse
|
30
|
Tennstaedt A, Aswendt M, Adamczak J, Collienne U, Selt M, Schneider G, Henn N, Schaefer C, Lagouge M, Wiedermann D, Kloppenburg P, Hoehn M. Human neural stem cell intracerebral grafts show spontaneous early neuronal differentiation after several weeks. Biomaterials 2015; 44:143-54. [DOI: 10.1016/j.biomaterials.2014.12.038] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 12/20/2014] [Indexed: 10/24/2022]
|
31
|
Hopkins AM, DeSimone E, Chwalek K, Kaplan DL. 3D in vitro modeling of the central nervous system. Prog Neurobiol 2015; 125:1-25. [PMID: 25461688 PMCID: PMC4324093 DOI: 10.1016/j.pneurobio.2014.11.003] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Revised: 10/12/2014] [Accepted: 11/15/2014] [Indexed: 12/15/2022]
Abstract
There are currently more than 600 diseases characterized as affecting the central nervous system (CNS) which inflict neural damage. Unfortunately, few of these conditions have effective treatments available. Although significant efforts have been put into developing new therapeutics, drugs which were promising in the developmental phase have high attrition rates in late stage clinical trials. These failures could be circumvented if current 2D in vitro and in vivo models were improved. 3D, tissue-engineered in vitro systems can address this need and enhance clinical translation through two approaches: (1) bottom-up, and (2) top-down (developmental/regenerative) strategies to reproduce the structure and function of human tissues. Critical challenges remain including biomaterials capable of matching the mechanical properties and extracellular matrix (ECM) composition of neural tissues, compartmentalized scaffolds that support heterogeneous tissue architectures reflective of brain organization and structure, and robust functional assays for in vitro tissue validation. The unique design parameters defined by the complex physiology of the CNS for construction and validation of 3D in vitro neural systems are reviewed here.
Collapse
Affiliation(s)
- Amy M Hopkins
- Department of Biomedical Engineering, Tufts University, Science & Technology Center, 4 Colby Street, Medford, MA 02155, USA
| | - Elise DeSimone
- Department of Biomedical Engineering, Tufts University, Science & Technology Center, 4 Colby Street, Medford, MA 02155, USA
| | - Karolina Chwalek
- Department of Biomedical Engineering, Tufts University, Science & Technology Center, 4 Colby Street, Medford, MA 02155, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Science & Technology Center, 4 Colby Street, Medford, MA 02155, USA.
| |
Collapse
|
32
|
Stincone A, Prigione A, Cramer T, Wamelink MMC, Campbell K, Cheung E, Olin-Sandoval V, Grüning NM, Krüger A, Tauqeer Alam M, Keller MA, Breitenbach M, Brindle KM, Rabinowitz JD, Ralser M. The return of metabolism: biochemistry and physiology of the pentose phosphate pathway. Biol Rev Camb Philos Soc 2014; 90:927-63. [PMID: 25243985 PMCID: PMC4470864 DOI: 10.1111/brv.12140] [Citation(s) in RCA: 921] [Impact Index Per Article: 83.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 07/07/2014] [Accepted: 07/16/2014] [Indexed: 12/13/2022]
Abstract
The pentose phosphate pathway (PPP) is a fundamental component of cellular metabolism. The PPP is important to maintain carbon homoeostasis, to provide precursors for nucleotide and amino acid biosynthesis, to provide reducing molecules for anabolism, and to defeat oxidative stress. The PPP shares reactions with the Entner–Doudoroff pathway and Calvin cycle and divides into an oxidative and non-oxidative branch. The oxidative branch is highly active in most eukaryotes and converts glucose 6-phosphate into carbon dioxide, ribulose 5-phosphate and NADPH. The latter function is critical to maintain redox balance under stress situations, when cells proliferate rapidly, in ageing, and for the ‘Warburg effect’ of cancer cells. The non-oxidative branch instead is virtually ubiquitous, and metabolizes the glycolytic intermediates fructose 6-phosphate and glyceraldehyde 3-phosphate as well as sedoheptulose sugars, yielding ribose 5-phosphate for the synthesis of nucleic acids and sugar phosphate precursors for the synthesis of amino acids. Whereas the oxidative PPP is considered unidirectional, the non-oxidative branch can supply glycolysis with intermediates derived from ribose 5-phosphate and vice versa, depending on the biochemical demand. These functions require dynamic regulation of the PPP pathway that is achieved through hierarchical interactions between transcriptome, proteome and metabolome. Consequently, the biochemistry and regulation of this pathway, while still unresolved in many cases, are archetypal for the dynamics of the metabolic network of the cell. In this comprehensive article we review seminal work that led to the discovery and description of the pathway that date back now for 80 years, and address recent results about genetic and metabolic mechanisms that regulate its activity. These biochemical principles are discussed in the context of PPP deficiencies causing metabolic disease and the role of this pathway in biotechnology, bacterial and parasite infections, neurons, stem cell potency and cancer metabolism.
Collapse
Affiliation(s)
- Anna Stincone
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, U.K.,Cambridge Systems Biology Centre, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, U.K
| | - Alessandro Prigione
- Max Delbrueck Centre for Molecular Medicine, Robert-Rössle-Str. 10, 13092 Berlin, Germany
| | - Thorsten Cramer
- Department of Gastroenterology and Hepatology, Molekulares Krebsforschungszentrum (MKFZ), Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Mirjam M C Wamelink
- Metabolic Unit, Department of Clinical Chemistry, VU University Medical Centre Amsterdam, De Boelelaaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Kate Campbell
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, U.K.,Cambridge Systems Biology Centre, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, U.K
| | - Eric Cheung
- Cancer Research UK, Beatson Institute, Switchback Road, Glasgow G61 1BD, U.K
| | - Viridiana Olin-Sandoval
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, U.K.,Cambridge Systems Biology Centre, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, U.K
| | - Nana-Maria Grüning
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, U.K.,Cambridge Systems Biology Centre, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, U.K
| | - Antje Krüger
- Max Planck Institute for Molecular Genetics, Ihnestr 73, 14195 Berlin, Germany
| | - Mohammad Tauqeer Alam
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, U.K.,Cambridge Systems Biology Centre, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, U.K
| | - Markus A Keller
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, U.K.,Cambridge Systems Biology Centre, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, U.K
| | - Michael Breitenbach
- Department of Cell Biology, University of Salzburg, Hellbrunnerstrasse 34, A-5020 Salzburg, Austria
| | - Kevin M Brindle
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, U.K.,Cancer Research UK Cambridge Research Institute (CRI), Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge CB2 0RE, U.K
| | - Joshua D Rabinowitz
- Department of Chemistry, Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, 08544 NJ, U.S.A
| | - Markus Ralser
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, U.K.,Cambridge Systems Biology Centre, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, U.K.,Division of Physiology and Metabolism, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7, U.K
| |
Collapse
|
33
|
Wattanapanitch M, Klincumhom N, Potirat P, Amornpisutt R, Lorthongpanich C, U-pratya Y, Laowtammathron C, Kheolamai P, Poungvarin N, Issaragrisil S. Dual small-molecule targeting of SMAD signaling stimulates human induced pluripotent stem cells toward neural lineages. PLoS One 2014; 9:e106952. [PMID: 25207966 PMCID: PMC4160199 DOI: 10.1371/journal.pone.0106952] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 08/01/2014] [Indexed: 01/10/2023] Open
Abstract
Incurable neurological disorders such as Parkinson's disease (PD), Huntington's disease (HD), and Alzheimer's disease (AD) are very common and can be life-threatening because of their progressive disease symptoms with limited treatment options. To provide an alternative renewable cell source for cell-based transplantation and as study models for neurological diseases, we generated induced pluripotent stem cells (iPSCs) from human dermal fibroblasts (HDFs) and then differentiated them into neural progenitor cells (NPCs) and mature neurons by dual SMAD signaling inhibitors. Reprogramming efficiency was improved by supplementing the histone deacethylase inhibitor, valproic acid (VPA), and inhibitor of p160-Rho associated coiled-coil kinase (ROCK), Y-27632, after retroviral transduction. We obtained a number of iPS colonies that shared similar characteristics with human embryonic stem cells in terms of their morphology, cell surface antigens, pluripotency-associated gene and protein expressions as well as their in vitro and in vivo differentiation potentials. After treatment with Noggin and SB431542, inhibitors of the SMAD signaling pathway, HDF-iPSCs demonstrated rapid and efficient differentiation into neural lineages. Six days after neural induction, neuroepithelial cells (NEPCs) were observed in the adherent monolayer culture, which had the ability to differentiate further into NPCs and neurons, as characterized by their morphology and the expression of neuron-specific transcripts and proteins. We propose that our study may be applied to generate neurological disease patient-specific iPSCs allowing better understanding of disease pathogenesis and drug sensitivity assays.
Collapse
Affiliation(s)
- Methichit Wattanapanitch
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nuttha Klincumhom
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Porntip Potirat
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Rattaya Amornpisutt
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chanchao Lorthongpanich
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Yaowalak U-pratya
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chuti Laowtammathron
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pakpoom Kheolamai
- Division of Cell Biology, Department of Pre-clinical Sciences, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Niphon Poungvarin
- Division of Neurology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Surapol Issaragrisil
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- * E-mail:
| |
Collapse
|
34
|
Carlessi L, Poli EF, Bechi G, Mantegazza M, Pascucci B, Narciso L, Dogliotti E, Sala C, Verpelli C, Lecis D, Delia D. Functional and molecular defects of hiPSC-derived neurons from patients with ATM deficiency. Cell Death Dis 2014; 5:e1342. [PMID: 25032865 PMCID: PMC4123100 DOI: 10.1038/cddis.2014.310] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 05/30/2014] [Accepted: 06/16/2014] [Indexed: 11/21/2022]
Abstract
Loss of ataxia telangiectasia mutated (ATM) kinase, a key factor of the DNA damage response (DDR) pathway, causes the cancer predisposing and neurodegenerative syndrome ataxia-telangiectasia (A-T). To investigate the mechanisms of neurodegeneration, we have reprogrammed fibroblasts from ATM-null A-T patients and normal controls to pluripotency (human-induced pluripotent stem cells), and derived from these neural precursor cells able to terminally differentiate into post-mitotic neurons positive to >90% for β-tubulin III+/microtubule-associated protein 2+. We show that A-T neurons display similar voltage-gated potassium and sodium currents and discharges of action potentials as control neurons, but defective expression of the maturation and synaptic markers SCG10, SYP and PSD95 (postsynaptic density protein 95). A-T neurons exhibited defective repair of DNA double-strand breaks (DSBs) and repressed phosphorylation of ATM substrates (e.g., γH2AX, Smc1-S966, Kap1-S824, Chk2-T68, p53-S15), but normal repair of single-strand breaks, and normal short- and long-patch base excision repair activities. Moreover, A-T neurons were resistant to apoptosis induced by the genotoxic agents camptothecin and trabectedin, but as sensitive as controls to the oxidative agents. Most notably, A-T neurons exhibited abnormal accumulation of topoisomerase 1-DNA covalent complexes (Top1-ccs). These findings reveal that ATM deficiency impairs neuronal maturation, suppresses the response and repair of DNA DSBs, and enhances Top1-cc accumulation. Top1-cc could be a risk factor for neurodegeneration as they may interfere with transcription elongation and promote transcriptional decline.
Collapse
Affiliation(s)
- L Carlessi
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milano, Italy
| | - E Fusar Poli
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milano, Italy
| | - G Bechi
- Department of Neurophysiopathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Amadeo 42, 20133 Milano, Italy
| | - M Mantegazza
- Department of Neurophysiopathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Amadeo 42, 20133 Milano, Italy
- Institute of Molecular and Cellular Pharmacology (IPMC) CNRS UMR7275 and University of Nice-Sophia Antipolis, 660 Route des Lucioles, 06560 Valbonne, France
| | - B Pascucci
- CNR Institute of Crystallography, Via Salaria, Km. 29.300, 00016 Monterotondo Scalo, Roma, Italy
| | - L Narciso
- Department of Food Safety and Veterinary Public Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Roma, Italy
| | - E Dogliotti
- Department of Environment and Primary Prevention, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Roma, Italy
| | - C Sala
- CNR Institute of Neuroscience and Department of Biotechnology and Translational Medicine, Via Vanvitelli 32, 20129 Milano, Italy
| | - C Verpelli
- CNR Institute of Neuroscience and Department of Biotechnology and Translational Medicine, Via Vanvitelli 32, 20129 Milano, Italy
| | - D Lecis
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milano, Italy
| | - D Delia
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milano, Italy
| |
Collapse
|
35
|
Potential and limitation of HLA-based banking of human pluripotent stem cells for cell therapy. J Immunol Res 2014; 2014:518135. [PMID: 25126584 PMCID: PMC4121106 DOI: 10.1155/2014/518135] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 06/06/2014] [Accepted: 06/18/2014] [Indexed: 12/13/2022] Open
Abstract
Great hopes have been placed on human pluripotent stem (hPS) cells for therapy. Tissues or organs derived from hPS cells could be the best solution to cure many different human diseases, especially those who do not respond to standard medication or drugs, such as neurodegenerative diseases, heart failure, or diabetes. The origin of hPS is critical and the idea of creating a bank of well-characterized hPS cells has emerged, like the one that already exists for cord blood. However, the main obstacle in transplantation is the rejection of tissues or organ by the receiver, due to the three main immunological barriers: the human leukocyte antigen (HLA), the ABO blood group, and minor antigens. The problem could be circumvented by using autologous stem cells, like induced pluripotent stem (iPS) cells, derived directly from the patient. But iPS cells have limitations, especially regarding the disease of the recipient and possible difficulties to handle or prepare autologous iPS cells. Finally, reaching standards of good clinical or manufacturing practices could be challenging. That is why well-characterized and universal hPS cells could be a better solution. In this review, we will discuss the interest and the feasibility to establish hPS cells bank, as well as some economics and ethical issues.
Collapse
|
36
|
Rosa V, Toh WS, Cao T, Shim W. Inducing pluripotency for disease modeling, drug development and craniofacial applications. Expert Opin Biol Ther 2014; 14:1233-40. [PMID: 24850281 DOI: 10.1517/14712598.2014.915306] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION The induced pluripotent stem cells (iPSCs) have characteristics similar to embryonic stem cells, including the capability of self-renewal and large-scale expansion and the ability to differentiate into all types of cells including germ cells, which defines pluripotency. Using iPSC avoids problems of immunological rejection and ethical controversy. The possible future uses of iPSC are diverse and go beyond the differentiation into somatic cells for regeneration of damaged tissues. AREAS COVERED A unique feature of iPSC is the potential to generate patient disease-specific tissues. Thus, cells from patients can be differentiated into relevant cells of interest for drug screening, characterization of drug effects and cytotoxic assays. This review presents key aspects related to iPSC, such as their generation, potential for disease modeling, treatment, drug development and future contributions to the craniofacial complex. EXPERT OPINION It is undisputable that the evolution in iPSC knowledge will improve the approaches for drug screening and development, help to understand and treat disease origins and mechanisms and provide new strategies to clinical treatment. However, it is necessary to fine-tune protocols to establish iPSCs that are cost-effective and safe for clinical use.
Collapse
Affiliation(s)
- Vinicius Rosa
- National University of Singapore, Faculty of Dentistry, Oral Sciences , 11 Lower Kent Ridge Road, Singapore 119083 , Singapore +65 6779 5555 ext 1650 ; + 65 6778 5742 ;
| | | | | | | |
Collapse
|
37
|
Ross CA, Akimov SS. Human-induced pluripotent stem cells: potential for neurodegenerative diseases. Hum Mol Genet 2014; 23:R17-26. [PMID: 24824217 DOI: 10.1093/hmg/ddu204] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The cell biology of human neurodegenerative diseases has been difficult to study till recently. The development of human induced pluripotent stem cell (iPSC) models has greatly enhanced our ability to model disease in human cells. Methods have recently been improved, including increasing reprogramming efficiency, introducing non-viral and non-integrating methods of cell reprogramming, and using novel gene editing techniques for generating genetically corrected lines from patient-derived iPSCs, or for generating mutations in control cell lines. In this review, we highlight accomplishments made using iPSC models to study neurodegenerative disorders such as Huntington's disease, Parkinson's disease, Amyotrophic Lateral Sclerosis, Fronto-Temporal Dementia, Alzheimer's disease, Spinomuscular Atrophy and other polyglutamine diseases. We review disease-related phenotypes shown in patient-derived iPSCs differentiated to relevant neural subtypes, often with stressors or cell "aging", to enhance disease-specific phenotypes. We also discuss prospects for the future of using of iPSC models of neurodegenerative disorders, including screening and testing of therapeutic compounds, and possibly of cell transplantation in regenerative medicine. The new iPSC models have the potential to greatly enhance our understanding of pathogenesis and to facilitate the development of novel therapeutics.
Collapse
Affiliation(s)
- Christopher A Ross
- Division of Neurobiology, Department of Psychiatry and Departments of Neurology, Neuroscience and Pharmacology, and Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MA, USA
| | | |
Collapse
|
38
|
Meamar R, Nasr-Esfahani MH, Mousavi SA, Basiri K. Stem cell therapy in amyotrophic lateral sclerosis. J Clin Neurosci 2013; 20:1659-63. [PMID: 24148693 DOI: 10.1016/j.jocn.2013.04.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2012] [Revised: 01/08/2013] [Accepted: 04/14/2013] [Indexed: 02/07/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder of upper and lower motor neurons, characterized by progressive muscular atrophy and weakness which culminates in death within 2-5 years. Despite various hypotheses about the responsible mechanisms, the etiology of ALS remains incompletely understood. However, it has been recently postulated that stem cell therapy could potentially target several mechanisms responsible for the etiology of ALS and other nervous system disorders, and could be regarded as one of the most promising therapeutic strategies for ALS treatment. We present a brief review of different methods of stem cell therapy in ALS patients and discuss the results with different cell types and routes of administration.
Collapse
Affiliation(s)
- Rokhsareh Meamar
- Department of Medical Science, Najafabad Branch, Islamic Azad University, Isfahan, Iran
| | | | | | | |
Collapse
|
39
|
Carlessi L, Fusar Poli E, Delia D. Brain and induced pluripotent stem cell-derived neural stem cells as an in vitro model of neurodegeneration in ataxia-telangiectasia. Exp Biol Med (Maywood) 2013; 238:301-7. [PMID: 23598976 DOI: 10.1177/1535370213480703] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The ataxia telangiectasia mutated (ATM) kinase is a key transducer of the cellular response to DNA double strand breaks and its deficiency causes ataxia-telangiectasia (A-T), a pleiotropic genetic disorder primarily characterized by cerebellar neuropathy, immunodeficiency and cancer predisposition. While enormous progress has been achieved in elucidating the biochemical and functional regulation of ATM in DNA damage response, and more recently in redox signalling and antioxidant defence, the factors that make neurons in A-T extremely vulnerable remain unclear. Given also that ATM knockout mice do not recapitulate the central nervous system phenotype, a number of human neural stem cell (hNSC) model systems have been developed to provide insights into the mechanisms of neurodegeneration associated with ATM dysfunction. Here we review the hNSC systems developed by us an others to model A-T.
Collapse
Affiliation(s)
- Luigi Carlessi
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Via Amadeo 42, 20133 Milan, Italy
| | | | | |
Collapse
|
40
|
Carlessi L, Fusar Poli E, De Filippis L, Delia D. ATM-deficient human neural stem cells as an in vitro model system to study neurodegeneration. DNA Repair (Amst) 2013; 12:605-11. [PMID: 23707302 PMCID: PMC3732388 DOI: 10.1016/j.dnarep.2013.04.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Loss of ATM kinase, a transducer of the DNA damage response and redox sensor, causes the neurodegenerative disorder ataxia-telangiectasia (A-T). While a great deal of progress has been made in elucidating the ATM-dependent DNA damage response (DDR) network, a key challenge remains in understanding the selective susceptibility of the nervous system to faulty DDR. Several factors appear implicated in the neurodegenerative phenotype in A-T, but which of them plays a crucial role remains unclear, especially since mouse models of A-T do not fully mirror the respective human syndrome. Therefore, a number of human neural stem cell (hNSC) systems have been developed to get an insight into the molecular mechanisms of neurodegeneration as consequence of ATM inactivation. Here we review the hNSC systems developed by us an others to model A-T.
Collapse
Affiliation(s)
- Luigi Carlessi
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Via Amadeo 42, 20133, Milan, Italy
| | | | | | | |
Collapse
|
41
|
Farmer JR, Altschaefl KM, O'Shea KS, Miller DJ. Activation of the type I interferon pathway is enhanced in response to human neuronal differentiation. PLoS One 2013; 8:e58813. [PMID: 23505563 PMCID: PMC3591356 DOI: 10.1371/journal.pone.0058813] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 02/07/2013] [Indexed: 11/29/2022] Open
Abstract
Despite the crucial role of innate immunity in preventing or controlling pathogen-induced damage in most, if not all, cell types, very little is known about the activity of this essential defense system in central nervous system neurons, especially in humans. In this report we use both an established neuronal cell line model and an embryonic stem cell-based system to examine human neuronal innate immunity and responses to neurotropic alphavirus infection in cultured cells. We demonstrate that neuronal differentiation is associated with increased expression of crucial type I interferon signaling pathway components, including interferon regulatory factor-9 and an interferon receptor heterodimer subunit, which results in enhanced interferon stimulation and subsequent heightened antiviral activity and cytoprotective responses against neurotropic alphaviruses such as western equine encephalitis virus. These results identify important differentiation-dependent changes in innate immune system function that control cell-autonomous neuronal responses. Furthermore, this work demonstrates the utility of human embryonic stem cell-derived cultures as a platform to study the interactions between innate immunity, virus infection, and pathogenesis in central nervous system neurons.
Collapse
Affiliation(s)
- Jocelyn R. Farmer
- Departments of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Kate M. Altschaefl
- Department of Epidemiology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - K. Sue O'Shea
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - David J. Miller
- Departments of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- * E-mail: .
| |
Collapse
|
42
|
Park KI, Goo K, Jung K, Kim M, Kim IS, Yun S, Lee IS, Shin JE, Yu HY, Eun HS, Kim JE, Namgung R, Lee C. Therapeutic Application of Neural Stem Cells for Neonatal Hypoxic-ischemic Brain Injury. NEONATAL MEDICINE 2013. [DOI: 10.5385/nm.2013.20.3.343] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Affiliation(s)
- Kook In Park
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Korea
| | - Kyoyeon Goo
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Korea
| | - Kwangsoo Jung
- BK21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Miri Kim
- BK21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Il-Sun Kim
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Korea
| | - Seokhwan Yun
- BK21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Il-Shin Lee
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Korea
| | - Jeong Eun Shin
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Korea
| | - Ha Yang Yu
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Korea
| | - Ho Seon Eun
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Korea
| | - Jung Eun Kim
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Korea
| | - Ran Namgung
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Korea
| | - Chul Lee
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
43
|
Vanee N, Roberts SB, Riggs MJ, Rao RR, Fong SS. Identification of metabolic changes in genetically unstable stem cells by using model analysis of gene expression. Chem Biodivers 2012; 9:911-29. [PMID: 22589092 DOI: 10.1002/cbdv.201100390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Stem-cell research seeks to address many different questions related to fundamental stem-cell function with the ultimate goal of being able to control and utilize stem cells for a broad range of therapeutic needs. While a large amount of work is focused on discovering and controlling differentiation mechanisms in stem cells, an equally interesting and important area of work is to understand the basics of stem-cell propagation and self-renewal. With high-throughput genomics and transcriptomic information on hand, it is becoming possible to address some of the detailed mechanistic processes occurring in stem cells, though interpretation of these data is often difficult. In this work, stem cells with genetic abnormalities were compared to genetically normal stem cells using gene-expression array data integrated with a large-scale metabolic model to help interpret changes in metabolism resulting in the identification of several metabolic pathways that were different in the normal and abnormal cells.
Collapse
Affiliation(s)
- Niti Vanee
- VCU Life Sciences, Virginia Commonwealth University, 601 W Main Street, P.O. Box 843068, Richmond, VA 23220, USA
| | | | | | | | | |
Collapse
|
44
|
Khandekar N, Lie KH, Sachdev PS, Sidhu KS. Amyloid precursor proteins, neural differentiation of pluripotent stem cells and its relevance to Alzheimer's disease. Stem Cells Dev 2012; 21:997-1006. [PMID: 22122714 DOI: 10.1089/scd.2011.0564] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) is a leading cause of age-related dementia that is characterized by an extensive loss of neurons and synaptic transmission. The pathological hallmarks of AD are neurofibrillary tangles and deposition of β-amyloid (Aβ) plaques. Previous research has investigated how Aβ fragments disrupt synaptic mechanisms in the vulnerable regions of the brain. There is a tremendous potential for stem cell technology to extend upon this research, not only in terms of developing therapeutic applications, but also in modeling AD. Indeed, the advent of induced pluripotent stem cell technology has opened up exciting new avenues for generating patient and disease-specific cell lines from somatic cells that may be used to model AD. Amyloid precursor protein (APP) is a key protein in neuronal development and this article reviews the role of APP in AD. Stem cell technology offers the opportunity to make use of APP in the directed differentiation of induced pluripotent stem cells into functional neurons, a process that may help generate a model of AD and thereby facilitate an understanding of the mechanisms underlying this disease.
Collapse
Affiliation(s)
- Neeta Khandekar
- Faculty of Medicine, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia.
| | | | | | | |
Collapse
|
45
|
Cristini S, Alessandri G, Acerbi F, Ciusani E, Colombo A, Fascio U, Nicosia RF, Invernizzi RW, Gelati M, Parati EA, Invernici G. Three-dimensional self-organizing neural architectures: a neural stem cells reservoir and a system for neurodevelopmental studies. Tissue Eng Part C Methods 2011; 17:1109-20. [PMID: 21721991 DOI: 10.1089/ten.tec.2010.0622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Complex microenvironmental stimuli influence neural cell properties. To study this, we developed a three-dimensional (3-D) neural culture system, composed of different populations including neurons, astrocytes, and neural stem cells (NSCs). In particular, these last-mentioned cells represent a source potentially exploitable to test drugs, to study neurodevelopment and cell-therapies for neuroregenerations. On seeding on matrigel in a medium supplemented with serum and mitogens, cells obtained from human fetal brain tissue formed 3-D self-organizing neural architectures. Immunocytochemical analysis demonstrated the presence of undifferentiated nestin+ and CD133+ cells, surrounded by β-tub-III+ and GFAP+ cells, suggesting the formation of niches containing potential human NSCs (hNSCs). The presence of hNSCs was confirmed by both neurosphere assay and RT-PCR, and their multipotentiality was demonstrated by both immunofluorescent staining and RT-PCR. Flow cytometry analysis revealed that neurosphere forming cells originating from at least two different subsets expressing, respectively, CD133 and CD146 markers were endowed with different proliferative and differentiation potential. Our data implicate that the complexity of environment within niches and aggregates of heterogeneous neural cell subsets may represent an innovative platform for neurobiological and neurodevelopmental investigations and a reservoir for a rapid expansion of hNSCs.
Collapse
Affiliation(s)
- Silvia Cristini
- Laboratory of Cellular Neurobiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
The childhood leukodystrophies are characterized by neonatal or childhood deficiencies in myelin production or maintenance; these may be due to hereditary defects in genes for myelin maintenance, as in Pelizaeus-Merzbacher disease, or to enzymatic deficiencies resulting in substrate misaccumulation or misprocessing, as in the lysosomal storage disorders. Regardless of their respective etiologies, these disorders are essentially all manifested by a profound deterioration in neurological function with age. A congenital deficit in forebrain myelination is also noted in children with the periventricular leukomalacia of cerebral palsy, which yields a more static morbidity. In light of the wide range of disorders to which congenital hypomyelination or postnatal demyelination may contribute, and the relative homogeneity of oligodendrocytes and their progenitors, the leukodystrophies may be especially attractive targets for cell-based therapeutic strategies. As a result, glial progenitor cells, which can give rise to new myelinogenic oligodendrocytes, have become of great interest as potential vectors for the restoration of myelin to the dysmyelinated brain and spinal cord. In addition, by distributing throughout the neuraxis after perinatal graft, and giving rise to astrocytes as well as oligodendrocytes, glial progenitor cells may be of great utility in rectifying the dysmyelination-associated enzymatic deficiencies of the lysosomal storage disorders.
Collapse
Affiliation(s)
- Steven A Goldman
- Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA.
| |
Collapse
|
47
|
Abstract
How a single fertilized cell generates diverse neuronal populations has been a fundamental biological problem since the 19(th) century. Classical histological methods revealed that postmitotic neurons are produced in a precise temporal and spatial order from germinal cells lining the cerebral ventricles. In the 20(th) century, DNA labeling and histo- and immunohistochemistry helped to distinguish the subtypes of dividing cells and delineate their locations in the ventricular and subventricular zones. Recently, genetic and cell biological methods have provided insights into sequential gene expression and molecular and cellular interactions that generate heterogeneous populations of NSCs leading to specific neuronal classes. This precisely regulated developmental process does not tolerate significant in vivo deviation, making replacement of adult neurons by NSCs during pathology a colossal challenge. In contrast, utilizing the trophic factors emanating from the NSC or their derivatives to slow down deterioration or prevent death of degenerating neurons may be a more feasible strategy.
Collapse
Affiliation(s)
- Joshua J. Breunig
- Department of Neurobiology, 2Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Tarik F. Haydar
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, Massachusetts, 02118, USA
| | - Pasko Rakic
- Department of Neurobiology, 2Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| |
Collapse
|
48
|
Cellular models for disease exploring and drug screening. Protein Cell 2010; 1:355-362. [PMID: 21203947 DOI: 10.1007/s13238-010-0027-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2009] [Accepted: 01/21/2010] [Indexed: 01/08/2023] Open
Abstract
The biopharmaceutical industry has been greatly promoted by the application of drug and disease models, including both animal and cellular models. In particular, the emergence of induced pluripotent stem cells (iPSC) makes it possible to create a large number of disease-specific cells in vitro. This review introduces the most widely applied models and their specialties.
Collapse
|
49
|
Sareen D, McMillan E, Ebert AD, Shelley BC, Johnson JA, Meisner LF, Svendsen CN. Chromosome 7 and 19 trisomy in cultured human neural progenitor cells. PLoS One 2009; 4:e7630. [PMID: 19898616 PMCID: PMC2765070 DOI: 10.1371/journal.pone.0007630] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2009] [Accepted: 09/25/2009] [Indexed: 01/07/2023] Open
Abstract
Background Stem cell expansion and differentiation is the foundation of emerging cell therapy technologies. The potential applications of human neural progenitor cells (hNPCs) are wide ranging, but a normal cytogenetic profile is important to avoid the risk of tumor formation in clinical trials. FDA approved clinical trials are being planned and conducted for hNPC transplantation into the brain or spinal cord for various neurodegenerative disorders. Although human embryonic stem cells (hESCs) are known to show recurrent chromosomal abnormalities involving 12 and 17, no studies have revealed chromosomal abnormalities in cultured hNPCs. Therefore, we investigated frequently occurring chromosomal abnormalities in 21 independent fetal-derived hNPC lines and the possible mechanisms triggering such aberrations. Methods and Findings While most hNPC lines were karyotypically normal, G-band karyotyping and fluorescent in situ hybridization (FISH) analyses revealed the emergence of trisomy 7 (hNPC+7) and trisomy 19 (hNPC+19), in 24% and 5% of the lines, respectively. Once detected, subsequent passaging revealed emerging dominance of trisomy hNPCs. DNA microarray and immunoblotting analyses demonstrate epidermal growth factor receptor (EGFR) overexpression in hNPC+7 and hNPC+19 cells. We observed greater levels of telomerase (hTERT), increased proliferation (Ki67), survival (TUNEL), and neurogenesis (βIII-tubulin) in hNPC+7 and hNPC+19, using respective immunocytochemical markers. However, the trisomy lines underwent replicative senescence after 50–60 population doublings and never showed neoplastic changes. Although hNPC+7 and hNPC+19 survived better after xenotransplantation into the rat striatum, they did not form malignant tumors. Finally, EGF deprivation triggered a selection of trisomy 7 cells in a diploid hNPC line. Conclusions We report that hNPCs are susceptible to accumulation of chromosome 7 and 19 trisomy in long-term cell culture. These results suggest that micro-environmental cues are powerful factors in the selection of specific hNPC aneuploidies, with trisomy of chromosome 7 being the most common. Given that a number of stem cell based clinical trials are being conducted or planned in USA and a recent report in PLoS Medicine showing the dangers of grafting an inordinate number of cells, these data substantiate the need for careful cytogenetic evaluation of hNPCs (fetal or hESC-derived) before their use in clinical or basic science applications.
Collapse
Affiliation(s)
- Dhruv Sareen
- Department of Neurology, University of Wisconsin School of Medicine and Public Health, Wisconsin Institutes for Medical Research (WIMR), Madison, Wisconsin, United States of America
- The Stem Cell and Regenerative Medicine Center, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Erin McMillan
- The Waisman Center, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Allison D. Ebert
- Department of Neurology, University of Wisconsin School of Medicine and Public Health, Wisconsin Institutes for Medical Research (WIMR), Madison, Wisconsin, United States of America
- The Stem Cell and Regenerative Medicine Center, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Brandon C. Shelley
- Department of Neurology, University of Wisconsin School of Medicine and Public Health, Wisconsin Institutes for Medical Research (WIMR), Madison, Wisconsin, United States of America
- The Stem Cell and Regenerative Medicine Center, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Julie A. Johnson
- Cell Line Genetics, LLC, Madison, Wisconsin, United States of America
| | | | - Clive N. Svendsen
- Department of Neurology, University of Wisconsin School of Medicine and Public Health, Wisconsin Institutes for Medical Research (WIMR), Madison, Wisconsin, United States of America
- The Stem Cell and Regenerative Medicine Center, University of Wisconsin, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
50
|
Koch P, Kokaia Z, Lindvall O, Brüstle O. Emerging concepts in neural stem cell research: autologous repair and cell-based disease modelling. Lancet Neurol 2009; 8:819-29. [PMID: 19679274 DOI: 10.1016/s1474-4422(09)70202-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The increasing availability of human pluripotent stem cells provides new prospects for neural-replacement strategies and disease-related basic research. With almost unlimited potential for self-renewal, the use of human embryonic stem cells (ESCs) bypasses the restricted supply and expandability of primary cells that has been a major bottleneck in previous neural transplantation approaches. Translation of developmental patterning and cell-type specification techniques to human ESC cultures enables in vitro generation of various neuronal and glial cell types. The derivation of stably proliferating neural stem cells from human ESCs further facilitates standardisation and circumvents the problem of batch-to-batch variations commonly encountered in "run-through" protocols, which promote terminal differentiation of pluripotent stem cells into somatic cell types without defined intermediate precursor stages. The advent of cell reprogramming offers an opportunity to translate these advances to induced pluripotent stem cells, thereby enabling the generation of neurons and glia from individual patients. Eventually, reprogramming could provide a supply of autologous neural cells for transplantation, and could lead to the establishment of cellular model systems of neurological diseases.
Collapse
Affiliation(s)
- Philipp Koch
- Institute of Reconstructive Neurobiology, Life & Brain Center, University of Bonn and Hertie Foundation, Bonn, Germany
| | | | | | | |
Collapse
|