1
|
Atkinson EA, Gregory HN, Carter LN, Evans RE, Roberton VH, Dickman R, Phillips JB. An immunomodulatory encapsulation system to deliver human iPSC-derived dopaminergic neuron progenitors for Parkinson's disease treatment. Biomater Sci 2025; 13:2012-2025. [PMID: 40013398 DOI: 10.1039/d4bm01566e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Parkinson's disease is a neurodegenerative condition associated with the progressive loss of dopaminergic neurons. This leads to neurological impairments with heightening severity and is globally increasing in prevalence due to population ageing. Cell transplantation has demonstrated significant promise in altering the disease course in the clinic, and stem cell-derived grafts are being investigated. Current clinical protocols involve systemic immunosuppression to prevent graft rejection, which could potentially be avoided by encapsulating the therapeutic cells in a locally immunosuppressive biomaterial matrix before delivery. Here we report the progression of an immunomodulatory encapsulation system employing ultrapure alginate hydrogel beads alongside tacrolimus-loaded microparticles in the encapsulation of dopaminergic neuron progenitors derived from human induced pluripotent stem cells (hiPSCs). The hiPSC-derived progenitors were characterised and displayed robust viability after encapsulation within alginate beads, producing dopamine as they matured in vitro. The encapsulation system effectively reduced T cell activation (3-fold) and protected progenitors from cytotoxicity in vitro. The alginate bead diameter was optimised using microfluidics to yield spherical and monodisperse hydrogels with a median size of 215.6 ± 0.5 μm, suitable for delivery to the brain through a surgical cannula. This technology has the potential to advance cell transplantation by locally protecting grafts from the host immune system.
Collapse
Affiliation(s)
- Emily A Atkinson
- UCL School of Pharmacy, University College London, London, UK.
- UCL Centre for Nerve Engineering, University College London, London, UK
| | - Holly N Gregory
- UCL School of Pharmacy, University College London, London, UK.
- UCL Centre for Nerve Engineering, University College London, London, UK
| | - Lara N Carter
- UCL School of Pharmacy, University College London, London, UK.
- UCL Centre for Nerve Engineering, University College London, London, UK
| | - Rachael E Evans
- UCL School of Pharmacy, University College London, London, UK.
- UCL Centre for Nerve Engineering, University College London, London, UK
| | - Victoria H Roberton
- UCL School of Pharmacy, University College London, London, UK.
- UCL Centre for Nerve Engineering, University College London, London, UK
| | - Rachael Dickman
- UCL School of Pharmacy, University College London, London, UK.
| | - James B Phillips
- UCL School of Pharmacy, University College London, London, UK.
- UCL Centre for Nerve Engineering, University College London, London, UK
| |
Collapse
|
2
|
Li C, Luo Y, Li S. The roles of neural stem cells in myelin regeneration and repair therapy after spinal cord injury. Stem Cell Res Ther 2024; 15:204. [PMID: 38978125 PMCID: PMC11232222 DOI: 10.1186/s13287-024-03825-x] [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: 02/12/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024] Open
Abstract
Spinal cord injury (SCI) is a complex tissue injury that results in a wide range of physical deficits, including permanent or progressive disabilities of sensory, motor and autonomic functions. To date, limitations in current clinical treatment options can leave SCI patients with lifelong disabilities. There is an urgent need to develop new therapies for reconstructing the damaged spinal cord neuron-glia network and restoring connectivity with the supraspinal pathways. Neural stem cells (NSCs) possess the ability to self-renew and differentiate into neurons and neuroglia, including oligodendrocytes, which are cells responsible for the formation and maintenance of the myelin sheath and the regeneration of demyelinated axons. For these properties, NSCs are considered to be a promising cell source for rebuilding damaged neural circuits and promoting myelin regeneration. Over the past decade, transplantation of NSCs has been extensively tested in a variety of preclinical models of SCI. This review aims to highlight the pathophysiology of SCI and promote the understanding of the role of NSCs in SCI repair therapy and the current advances in pathological mechanism, pre-clinical studies, as well as clinical trials of SCI via NSC transplantation therapeutic strategy. Understanding and mastering these frontier updates will pave the way for establishing novel therapeutic strategies to improve the quality of recovery from SCI.
Collapse
Affiliation(s)
- Chun Li
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Neurology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, Tongji University School of Medicine, Shanghai, 200092, China
| | - Yuping Luo
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Neurology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Siguang Li
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Neurology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
| |
Collapse
|
3
|
Spathopoulou A, Podlesnic M, De Gaetano L, Kirsch EM, Tisch M, Finotello F, Aigner L, Günther K, Edenhofer F. Single-cell Profiling of Reprogrammed Human Neural Stem Cells Unveils High Similarity to Neural Progenitors in the Developing Central Nervous System. Stem Cell Rev Rep 2024; 20:1325-1339. [PMID: 38519702 PMCID: PMC11222274 DOI: 10.1007/s12015-024-10698-3] [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] [Accepted: 02/14/2024] [Indexed: 03/25/2024]
Abstract
BACKGROUND Similar to induced pluripotent cells (iPSCs), induced neural stem cells (iNSCs) can be directly converted from human somatic cells such as dermal fibroblasts and peripheral blood monocytes. While previous studies have demonstrated the resemblance of iNSCs to neural stem cells derived from primary sources and embryonic stem cells, respectively, a comprehensive analysis of the correlation between iNSCs and their physiological counterparts remained to be investigated. METHODS Nowadays, single-cell sequencing technologies provide unique opportunities for in-depth cellular benchmarking of complex cell populations. Our study involves the comprehensive profiling of converted human iNSCs at a single-cell transcriptomic level, alongside conventional methods, like flow cytometry and immunofluorescence stainings. RESULTS Our results show that the iNSC conversion yields a homogeneous cell population expressing bona fide neural stem cell markers. Extracting transcriptomic signatures from published single cell transcriptomic atlas data and comparison to the iNSC transcriptome reveals resemblance to embryonic neuroepithelial cells of early neurodevelopmental stages observed in vivo at 5 weeks of development. CONCLUSION Our data underscore the physiological relevance of directly converted iNSCs, making them a valuable in vitro system for modeling human central nervous system development and establishing translational applications in cell therapy and compound screening.
Collapse
Affiliation(s)
- Angeliki Spathopoulou
- Department of Molecular Biology & CMBI, Genomics, Stem Cell & Regenerative Medicine Group, University of Innsbruck, Technikerstraße 25, 6020, Innsbruck, Austria
| | - Martina Podlesnic
- Department of Molecular Biology & CMBI, Genomics, Stem Cell & Regenerative Medicine Group, University of Innsbruck, Technikerstraße 25, 6020, Innsbruck, Austria
| | - Laura De Gaetano
- Department of Molecular Biology & CMBI, Genomics, Stem Cell & Regenerative Medicine Group, University of Innsbruck, Technikerstraße 25, 6020, Innsbruck, Austria
| | - Elena Marie Kirsch
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Center for Stroke Research, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marcel Tisch
- Department of Molecular Biology & CMBI, Genomics, Stem Cell & Regenerative Medicine Group, University of Innsbruck, Technikerstraße 25, 6020, Innsbruck, Austria
| | - Francesca Finotello
- Department of Molecular Biology, Digital Science Center (DiSC), University of Innsbruck, Innsbruck, Austria
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Katharina Günther
- Department of Molecular Biology & CMBI, Genomics, Stem Cell & Regenerative Medicine Group, University of Innsbruck, Technikerstraße 25, 6020, Innsbruck, Austria
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Frank Edenhofer
- Department of Molecular Biology & CMBI, Genomics, Stem Cell & Regenerative Medicine Group, University of Innsbruck, Technikerstraße 25, 6020, Innsbruck, Austria.
| |
Collapse
|
4
|
Guo L, Zou D, Qiu W, Fei F, Chen L, Chen W, Xiong H, Li X, Wang Y, Gao M, Zhu J, Zhang J, He Y, Gao M, Xu R. Linc-NSC affects cell differentiation, apoptosis and proliferation in mouse neural stem cells and embryonic stem cells in vitro and in vivo. Cell Mol Life Sci 2024; 81:182. [PMID: 38615283 PMCID: PMC11016521 DOI: 10.1007/s00018-024-05224-0] [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: 07/20/2023] [Revised: 12/12/2023] [Accepted: 03/18/2024] [Indexed: 04/15/2024]
Abstract
BACKGROUND Stem cell therapy is a promising therapeutic strategy. In a previous study, we evaluated tumorigenicity by the stereotactic transplantation of neural stem cells (NSCs) and embryonic stem cells (ESCs) from experimental mice. Twenty-eight days later, there was no evidence of tumor formation or long-term engraftment in the NSCs transplantation group. In contrast, the transplantation of ESCs caused tumor formation; this was due to their high proliferative capacity. Based on transcriptome sequencing, we found that a long intergenic non-coding RNA (named linc-NSC) with unknown structure and function was expressed at 1100-fold higher levels in NSCs than in ESCs. This finding suggested that linc-NSC is negatively correlated with stem cell pluripotency and tumor development, but positively correlated with neurogenesis. In the present study, we investigated the specific role of linc-NSC in NSCs/ESCs in tumor formation and neurogenesis. METHODS Whole transcriptome profiling by RNA sequencing and bioinformatics was used to predict lncRNAs that are widely associated with enhanced tumorigenicity. The expression of linc-NSC was assessed by quantitative real-time PCR. We also performed a number of in vitro methods, including cell proliferation assays, differentiation assays, immunofluorescence assays, flow cytometry, along with in vivo survival and immunofluorescence assays to investigate the impacts of linc-NSC on tumor formation and neurogenesis in NSCs and ESCs. RESULTS Following the knockdown of linc-NSC in NSCs, NSCs cultured in vitro and those transplanted into the cortex of mice showed stronger survival ability (P < 0.0001), enhanced proliferation(P < 0.001), and reduced apoptosis (P < 0.05); the opposite results were observed when linc-NSC was overexpressed in ESCs. Furthermore, the overexpression of linc-NSC in ECSs induced enhanced apoptosis (P < 0.001) and differentiation (P < 0.01), inhibited tumorigenesis (P < 0.05) in vivo, and led to a reduction in tumor weight (P < 0.0001). CONCLUSIONS Our analyses demonstrated that linc-NSC, a promising gene-edited target, may promote the differentiation of mouse NSCs and inhibit tumorigenesis in mouse ESCs. The knockdown of linc-NSC inhibited the apoptosis in NSCs both in vitro and in vivo, and prevented tumor formation, revealing a new dimension into the effect of lncRNA on low survival NSCs and providing a prospective gene manipulation target prior to transplantation. In parallel, the overexpression of linc-NSC induced apoptosis in ESCs both in vitro and in vivo and attenuated the tumorigenicity of ESCs in vivo, but did not completely prevent tumor formation.
Collapse
Affiliation(s)
- Lili Guo
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China
| | - Dan Zou
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China
| | - Wenqiao Qiu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China
| | - Fan Fei
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China
| | - Lihua Chen
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China
| | - Wenjin Chen
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China
| | - Huan Xiong
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China
| | - Xinda Li
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China
| | - Yangyang Wang
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China
| | - Mingjun Gao
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China
| | - Jianwei Zhu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China
| | - Jin Zhang
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China
| | - Yunsen He
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China
| | - Mou Gao
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Ruxiang Xu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China.
| |
Collapse
|
5
|
Gao M, Dong Q, Yang Z, Zou D, Han Y, Chen Z, Xu R. Long non-coding RNA H19 regulates neurogenesis of induced neural stem cells in a mouse model of closed head injury. Neural Regen Res 2024; 19:872-880. [PMID: 37843223 PMCID: PMC10664125 DOI: 10.4103/1673-5374.382255] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/08/2023] [Accepted: 07/04/2023] [Indexed: 10/17/2023] Open
Abstract
Stem cell-based therapies have been proposed as a potential treatment for neural regeneration following closed head injury. We previously reported that induced neural stem cells exert beneficial effects on neural regeneration via cell replacement. However, the neural regeneration efficiency of induced neural stem cells remains limited. In this study, we explored differentially expressed genes and long non-coding RNAs to clarify the mechanism underlying the neurogenesis of induced neural stem cells. We found that H19 was the most downregulated neurogenesis-associated lncRNA in induced neural stem cells compared with induced pluripotent stem cells. Additionally, we demonstrated that H19 levels in induced neural stem cells were markedly lower than those in induced pluripotent stem cells and were substantially higher than those in induced neural stem cell-derived neurons. We predicted the target genes of H19 and discovered that H19 directly interacts with miR-325-3p, which directly interacts with Ctbp2 in induced pluripotent stem cells and induced neural stem cells. Silencing H19 or Ctbp2 impaired induced neural stem cell proliferation, and miR-325-3p suppression restored the effect of H19 inhibition but not the effect of Ctbp2 inhibition. Furthermore, H19 silencing substantially promoted the neural differentiation of induced neural stem cells and did not induce apoptosis of induced neural stem cells. Notably, silencing H19 in induced neural stem cell grafts markedly accelerated the neurological recovery of closed head injury mice. Our results reveal that H19 regulates the neurogenesis of induced neural stem cells. H19 inhibition may promote the neural differentiation of induced neural stem cells, which is closely associated with neurological recovery following closed head injury.
Collapse
Affiliation(s)
- Mou Gao
- Department of Neurosurgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China
- Zhongsai Stem Cell Genetic Engineering Co., Ltd., Sanmenxia, Henan Province, China
| | - Qin Dong
- Department of Neurology, Fu Xing Hospital, Capital Medical University, Beijing, China
| | - Zhijun Yang
- Zhongsai Stem Cell Genetic Engineering Co., Ltd., Sanmenxia, Henan Province, China
| | - Dan Zou
- Department of Neurosurgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Yajuan Han
- Zhongsai Stem Cell Genetic Engineering Co., Ltd., Sanmenxia, Henan Province, China
| | - Zhanfeng Chen
- Zhongsai Stem Cell Genetic Engineering Co., Ltd., Sanmenxia, Henan Province, China
| | - Ruxiang Xu
- Department of Neurosurgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| |
Collapse
|
6
|
Nie L, Yao D, Chen S, Wang J, Pan C, Wu D, Liu N, Tang Z. Directional induction of neural stem cells, a new therapy for neurodegenerative diseases and ischemic stroke. Cell Death Discov 2023; 9:215. [PMID: 37393356 DOI: 10.1038/s41420-023-01532-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/16/2023] [Accepted: 06/22/2023] [Indexed: 07/03/2023] Open
Abstract
Due to the limited capacity of the adult mammalian brain to self-repair and regenerate, neurological diseases, especially neurodegenerative disorders and stroke, characterized by irreversible cellular damage are often considered as refractory diseases. Neural stem cells (NSCs) play a unique role in the treatment of neurological diseases for their abilities to self-renew and form different neural lineage cells, such as neurons and glial cells. With the increasing understanding of neurodevelopment and advances in stem cell technology, NSCs can be obtained from different sources and directed to differentiate into a specific neural lineage cell phenotype purposefully, making it possible to replace specific cells lost in some neurological diseases, which provides new approaches to treat neurodegenerative diseases as well as stroke. In this review, we outline the advances in generating several neuronal lineage subtypes from different sources of NSCs. We further summarize the therapeutic effects and possible therapeutic mechanisms of these fated specific NSCs in neurological disease models, with special emphasis on Parkinson's disease and ischemic stroke. Finally, from the perspective of clinical translation, we compare the strengths and weaknesses of different sources of NSCs and different methods of directed differentiation, and propose future research directions for directed differentiation of NSCs in regenerative medicine.
Collapse
Affiliation(s)
- Luwei Nie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Dabao Yao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Shiling Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Jingyi Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Chao Pan
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Dongcheng Wu
- Department of Biochemistry and Molecular Biology, Wuhan University School of Basic Medical Sciences, Wuhan, 430030, China
- Wuhan Hamilton Biotechnology Co., Ltd., Wuhan, 430030, China
| | - Na Liu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Zhouping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
7
|
Zheng L, Zhang L, Guo Y, Xu X, Liu Z, Yan Z, Fu R. The immunological role of mesenchymal stromal cells in patients with myelodysplastic syndrome. Front Immunol 2022; 13:1078421. [PMID: 36569863 PMCID: PMC9767949 DOI: 10.3389/fimmu.2022.1078421] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 11/24/2022] [Indexed: 12/13/2022] Open
Abstract
Myelodysplastic syndrome (MDS) is a common hematological malignant disease, characterized by malignant hematopoietic stem cell proliferation in the bone marrow (BM); clinically, it mainly manifests clinically mainly by as pathological hematopoiesis, hemocytopenia, and high-risk transformation to acute leukemia. Several studies have shown that the BM microenvironment plays a critical role in the progression of MDS. In this study, we specifically evaluated mesenchymal stromal cells (MSCs) that exert immunomodulatory effects in the BM microenvironment. This immunomodulatory effect occurs through direct cell-cell contact and the secretion of soluble cytokines or micro vesicles. Several researchers have compared MSCs derived from healthy donors to low-risk MDS-associated bone mesenchymal stem cells (BM-MSCs) and have found no significant abnormalities in the MDS-MSC phenotype; however, these cells have been observed to exhibit altered function, including a decline in osteoblastic function. This altered function may promote MDS progression. In patients with MDS, especially high-risk patients, MSCs in the BM microenvironment regulate immune cell function, such as that of T cells, B cells, natural killer cells, dendritic cells, neutrophils, myeloid-derived suppressor cells (MDSCs), macrophages, and Treg cells, thereby enabling MDS-associated malignant cells to evade immune cell surveillance. Alterations in MDS-MSC function include genomic instability, microRNA production, histone modification, DNA methylation, and abnormal signal transduction and cytokine secretion.
Collapse
Affiliation(s)
- Likun Zheng
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China,Department of Hematology, North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei, China
| | - Lei Zhang
- Department of Orthopedics, Kailuan General Hospital, Tangshan, Hebei, China
| | - Yixuan Guo
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xintong Xu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhaoyun Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhenyu Yan
- Department of Hematology, North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei, China
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China,*Correspondence: Rong Fu,
| |
Collapse
|
8
|
Wang J, Sun N, Ju Y, Ni N, Tang Z, Zhang D, Dai X, Chen M, Wang Y, Gu P, Ji J. miR-381-3p Cooperated With Hes1 to Regulate the Proliferation and Differentiation of Retinal Progenitor Cells. Front Cell Dev Biol 2022; 10:853215. [PMID: 35281083 PMCID: PMC8914042 DOI: 10.3389/fcell.2022.853215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/11/2022] [Indexed: 01/01/2023] Open
Abstract
Retinal progenitor cells (RPCs) transplantation has become a promising therapy for retinal degeneration, which is a major kind of ocular diseases causing blindness. Since RPCs have limited proliferation and differentiation abilities toward retinal neurons, it is urgent to resolve these problems. MicroRNAs have been reported to have vital effects on stem cell fate. In our study, the data showed that overexpression of miR-381-3p repressed Hes1 expression, which promoted RPCs differentiation, especially toward neuronal cells, and inhibited RPCs proliferation. Knockdown of endogenous miR-381-3p increased Hes1 expression to inhibit RPCs differentiation and promote proliferation. In addition, a luciferase assay demonstrated that miR-381-3p directly targeted the Hes1 3’ untranslated region (UTR). Taken together, our study demonstrated that miR-381-3p regulated RPCs proliferation and differentiation by targeting Hes1, which provides an experimental basis of RPCs transplantation therapy for retinal degeneration.
Collapse
Affiliation(s)
- Jiajing Wang
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Na Sun
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
- Department of Ophthalmology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yahan Ju
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Ni Ni
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Zhimin Tang
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Dandan Zhang
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Xiaochan Dai
- Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Moxin Chen
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Yiqi Wang
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Ping Gu
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
- *Correspondence: Jing Ji, ; Ping Gu,
| | - Jing Ji
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
- *Correspondence: Jing Ji, ; Ping Gu,
| |
Collapse
|
9
|
Chakritbudsabong W, Sariya L, Jantahiran P, Chaisilp N, Chaiwattanarungruengpaisan S, Rungsiwiwut R, Ferreira JN, Rungarunlert S. Generation of Porcine Induced Neural Stem Cells Using the Sendai Virus. Front Vet Sci 2022; 8:806785. [PMID: 35097051 PMCID: PMC8790232 DOI: 10.3389/fvets.2021.806785] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/13/2021] [Indexed: 12/21/2022] Open
Abstract
The reprogramming of cells into induced neural stem cells (iNSCs), which are faster and safer to generate than induced pluripotent stem cells, holds tremendous promise for fundamental and frontier research, as well as personalized cell-based therapies for neurological diseases. However, reprogramming cells with viral vectors increases the risk of tumor development due to vector and transgene integration in the host cell genome. To circumvent this issue, the Sendai virus (SeV) provides an alternative integration-free reprogramming method that removes the danger of genetic alterations and enhances the prospects of iNSCs from bench to bedside. Since pigs are among the most successful large animal models in biomedical research, porcine iNSCs (piNSCs) may serve as a disease model for both veterinary and human medicine. Here, we report the successful generation of piNSC lines from pig fibroblasts by employing the SeV. These piNSCs can be expanded for up to 40 passages in a monolayer culture and produce neurospheres in a suspension culture. These piNSCs express high levels of NSC markers (PAX6, SOX2, NESTIN, and VIMENTIN) and proliferation markers (KI67) using quantitative immunostaining and western blot analysis. Furthermore, piNSCs are multipotent, as they are capable of producing neurons and glia, as demonstrated by their expressions of TUJ1, MAP2, TH, MBP, and GFAP proteins. During the reprogramming of piNSCs with the SeV, no induced pluripotent stem cells developed, and the established piNSCs did not express OCT4, NANOG, and SSEA1. Hence, the use of the SeV can reprogram porcine somatic cells without first going through an intermediate pluripotent state. Our research produced piNSCs using SeV methods in novel, easily accessible large animal cell culture models for evaluating the efficacy of iNSC-based clinical translation in human medicine. Additionally, our piNSCs are potentially applicable in disease modeling in pigs and regenerative therapies in veterinary medicine.
Collapse
Affiliation(s)
- Warunya Chakritbudsabong
- Laboratory of Cellular Biomedicine and Veterinary Medicine, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
- Department of Preclinic and Applied Animal Science, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Ladawan Sariya
- The Monitoring and Surveillance Center for Zoonotic Disease in Wildlife and Exotic Animals (MoZWE), Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Phakhin Jantahiran
- Laboratory of Cellular Biomedicine and Veterinary Medicine, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
- Department of Preclinic and Applied Animal Science, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Nattarun Chaisilp
- The Monitoring and Surveillance Center for Zoonotic Disease in Wildlife and Exotic Animals (MoZWE), Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Somjit Chaiwattanarungruengpaisan
- The Monitoring and Surveillance Center for Zoonotic Disease in Wildlife and Exotic Animals (MoZWE), Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Ruttachuk Rungsiwiwut
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Bangkok, Thailand
| | - Joao N. Ferreira
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| | - Sasitorn Rungarunlert
- Laboratory of Cellular Biomedicine and Veterinary Medicine, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
- Department of Preclinic and Applied Animal Science, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
- *Correspondence: Sasitorn Rungarunlert
| |
Collapse
|
10
|
Lei C, Mei S, Zhou C, Xia C. Decellularized tracheal scaffolds in tracheal reconstruction: An evaluation of different techniques. J Appl Biomater Funct Mater 2021; 19:22808000211064948. [PMID: 34903089 DOI: 10.1177/22808000211064948] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
In humans, the trachea is a conduit for ventilation connecting the throat and lungs. However, certain congenital or acquired diseases may cause long-term tracheal defects that require replacement. Tissue engineering is considered a promising method to reconstruct long-segment tracheal lesions and restore the structure and function of the trachea. Decellularization technology retains the natural structure of the trachea, has good biocompatibility and mechanical properties, and is currently a hotspot in tissue engineering studies. This article lists various recent representative protocols for the generation of decellularized tracheal scaffolds (DTSs), as well as their validity and limitations. Based on the advancements in decellularization methods, we discussed the impact and importance of mechanical properties, revascularization, recellularization, and biocompatibility in the production and implantation of DTS. This review provides a basis for future research on DTS and its application in clinical therapy.
Collapse
Affiliation(s)
- Chenyang Lei
- Department of Otorhinolaryngology, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Sheng Mei
- Department of Otorhinolaryngology, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Chun Zhou
- Department of Geriatrics, The 903 Hospital of the Chinese People's Liberation Army Joint Logistics Support Force, Hangzhou, China
| | - Chen Xia
- Department of Orthopedic Surgery, Zhejiang Provincial People's Hospital, Hangzhou, China
| |
Collapse
|
11
|
Maiso P, Mogollón P, Ocio EM, Garayoa M. Bone Marrow Mesenchymal Stromal Cells in Multiple Myeloma: Their Role as Active Contributors to Myeloma Progression. Cancers (Basel) 2021; 13:2542. [PMID: 34067236 PMCID: PMC8196907 DOI: 10.3390/cancers13112542] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 05/16/2021] [Accepted: 05/19/2021] [Indexed: 01/01/2023] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy of plasma cells that proliferate and accumulate within the bone marrow (BM). Work from many groups has made evident that the complex microenvironment of the BM plays a crucial role in myeloma progression and response to therapeutic agents. Within the cellular components of the BM, we will specifically focus on mesenchymal stromal cells (MSCs), which are known to interact with myeloma cells and the other components of the BM through cell to cell, soluble factors and, as more recently evidenced, through extracellular vesicles. Multiple structural and functional abnormalities have been found when characterizing MSCs derived from myeloma patients (MM-MSCs) and comparing them to those from healthy donors (HD-MSCs). Other studies have identified differences in genomic, mRNA, microRNA, histone modification, and DNA methylation profiles. We discuss these distinctive features shaping MM-MSCs and propose a model for the transition from HD-MSCs to MM-MSCs as a consequence of the interaction with myeloma cells. Finally, we review the contribution of MM-MSCs to several aspects of myeloma pathology, specifically to myeloma growth and survival, drug resistance, dissemination and homing, myeloma bone disease, and the induction of a pro-inflammatory and immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Patricia Maiso
- University Hospital Marqués de Valdecilla (IDIVAL), University of Cantabria, 39008 Santander, Spain
| | - Pedro Mogollón
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (P.M.); (M.G.)
| | - Enrique M. Ocio
- University Hospital Marqués de Valdecilla (IDIVAL), University of Cantabria, 39008 Santander, Spain
| | - Mercedes Garayoa
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (P.M.); (M.G.)
| |
Collapse
|
12
|
Calinescu AA, Kauss MC, Sultan Z, Al-Holou WN, O'Shea SK. Stem cells for the treatment of glioblastoma: a 20-year perspective. CNS Oncol 2021; 10:CNS73. [PMID: 34006134 PMCID: PMC8162173 DOI: 10.2217/cns-2020-0026] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma, the deadliest form of primary brain tumor, remains a disease without cure. Treatment resistance is in large part attributed to limitations in the delivery and distribution of therapeutic agents. Over the last 20 years, numerous preclinical studies have demonstrated the feasibility and efficacy of stem cells as antiglioma agents, leading to the development of trials to test these therapies in the clinic. In this review we present and analyze these studies, discuss mechanisms underlying their beneficial effect and highlight experimental progress, limitations and the emergence of promising new therapeutic avenues. We hope to increase awareness of the advantages brought by stem cells for the treatment of glioblastoma and inspire further studies that will lead to accelerated implementation of effective therapies. Glioblastoma is the deadliest and most common form of brain tumor, for which there is no cure. It is very difficult to deliver medicine to the tumor cells, because they spread out widely into the normal brain, and local blood vessels represent a barrier that most medicines cannot cross. It was shown, in many studies over the last 20 years, that stem cells are attracted toward the tumor and that they can deliver many kinds of therapeutic agents directly to brain cancer cells and shrink the tumor. In this review we analyze these studies and present new discoveries that can be used to make stem cell therapies for glioblastoma more effective to prolong the life of patients with brain tumors.
Collapse
Affiliation(s)
| | - McKenzie C Kauss
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,College of Literature Science & Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Zain Sultan
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,College of Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Wajd N Al-Holou
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Sue K O'Shea
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
13
|
Islam R, Drecun S, Varga BV, Vonderwalde I, Siu R, Nagy A, Morshead CM. Transplantation of Human Cortically-Specified Neuroepithelial Progenitor Cells Leads to Improved Functional Outcomes in a Mouse Model of Stroke. Front Cell Neurosci 2021; 15:654290. [PMID: 33994947 PMCID: PMC8116536 DOI: 10.3389/fncel.2021.654290] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/08/2021] [Indexed: 12/02/2022] Open
Abstract
Stroke is a leading cause of death and long-term disability worldwide. Current therapeutic options are limited in terms of their time for implementation and efficacy in promoting recovery. Cell transplantation has been shown to have promise in several animal models however significant challenges remain, including the optimal source of cells to promote neural repair. Here, we report on the use of a population of human ESC derived, cortically specified, neuroepithelial precursor cells (cNEPs) that are neurally restricted in their lineage potential. CNEPs have the potential to give rise to mature neural cell types following transplantation, including neurons, astrocytes and oligodendrocytes. With a view towards translation, we sought to determine whether this human cell source was effective in promoting improved functional outcomes following stroke. Undifferentiated cNEPs were transplanted in a pre-clinical endothelin-1 (ET-1) model of ischemic motor cortical stroke in immunocompromised SCID-beige mice and cellular and functional outcomes were assessed. We demonstrate that cNEP transplantation in the acute phase (4 days post-stroke) improves motor function as early as 20 days post-stroke, compared to stroke-injured, non-transplanted mice. At the time of recovery, a small fraction (<6%) of the transplanted cNEPs are observed within the stroke injury site. The surviving cells expressed the immature neuronal marker, doublecortin, with no differentiation into mature neural phenotypes. At longer survival times (40 days), the majority of recovered, transplanted mice had a complete absence of surviving cNEPS. Hence, human cNEPs grafted at early times post-stroke support the observed functional recovery following ET-1 stroke but their persistence is not required, thereby supporting a by-stander effect rather than cell replacement.
Collapse
Affiliation(s)
- Rehnuma Islam
- Faculty of Medicine, Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Stasja Drecun
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Balazs V. Varga
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Ilan Vonderwalde
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Ricky Siu
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Andras Nagy
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Cindi M. Morshead
- Faculty of Medicine, Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
14
|
Latoszek E, Czeredys M. Molecular Components of Store-Operated Calcium Channels in the Regulation of Neural Stem Cell Physiology, Neurogenesis, and the Pathology of Huntington's Disease. Front Cell Dev Biol 2021; 9:657337. [PMID: 33869222 PMCID: PMC8047111 DOI: 10.3389/fcell.2021.657337] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/10/2021] [Indexed: 12/11/2022] Open
Abstract
One of the major Ca2+ signaling pathways is store-operated Ca2+ entry (SOCE), which is responsible for Ca2+ flow into cells in response to the depletion of endoplasmic reticulum Ca2+ stores. SOCE and its molecular components, including stromal interaction molecule proteins, Orai Ca2+ channels, and transient receptor potential canonical channels, are involved in the physiology of neural stem cells and play a role in their proliferation, differentiation, and neurogenesis. This suggests that Ca2+ signaling is an important player in brain development. Huntington’s disease (HD) is an incurable neurodegenerative disorder that is caused by polyglutamine expansion in the huntingtin (HTT) protein, characterized by the loss of γ-aminobutyric acid (GABA)-ergic medium spiny neurons (MSNs) in the striatum. However, recent research has shown that HD is also a neurodevelopmental disorder and Ca2+ signaling is dysregulated in HD. The relationship between HD pathology and elevations of SOCE was demonstrated in different cellular and mouse models of HD and in induced pluripotent stem cell-based GABAergic MSNs from juvenile- and adult-onset HD patient fibroblasts. The present review discusses the role of SOCE in the physiology of neural stem cells and its dysregulation in HD pathology. It has been shown that elevated expression of STIM2 underlying the excessive Ca2+ entry through store-operated calcium channels in induced pluripotent stem cell-based MSNs from juvenile-onset HD. In the light of the latest findings regarding the role of Ca2+ signaling in HD pathology we also summarize recent progress in the in vitro differentiation of MSNs that derive from different cell sources. We discuss advances in the application of established protocols to obtain MSNs from fetal neural stem cells/progenitor cells, embryonic stem cells, induced pluripotent stem cells, and induced neural stem cells and the application of transdifferentiation. We also present recent progress in establishing HD brain organoids and their potential use for examining HD pathology and its treatment. Moreover, the significance of stem cell therapy to restore normal neural cell function, including Ca2+ signaling in the central nervous system in HD patients will be considered. The transplantation of MSNs or their precursors remains a promising treatment strategy for HD.
Collapse
Affiliation(s)
- Ewelina Latoszek
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Magdalena Czeredys
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| |
Collapse
|
15
|
Spellicy SE, Hess DC. The Immunomodulatory Capacity of Induced Pluripotent Stem Cells in the Post-stroke Environment. Front Cell Dev Biol 2021; 9:647415. [PMID: 33796535 PMCID: PMC8007866 DOI: 10.3389/fcell.2021.647415] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/25/2021] [Indexed: 11/13/2022] Open
Abstract
Inflammation has proven to be a key contributing factor to the pathogenesis of ischemic and hemorrhagic stroke. This sequential and progressive response, marked by proliferation of resident immune cells and recruitment of peripheral immune populations, results in increased oxidative stress, and neuronal cell death. Therapeutics aimed at quelling various stages of this post-stroke inflammatory response have shown promise recently, one of which being differentiated induced pluripotent stem cells (iPSCs). While direct repopulation of damaged tissues and enhanced neurogenesis are hypothesized to encompass some of the therapeutic potential of iPSCs, recent evidence has demonstrated a substantial paracrine effect on neuroinflammation. Specifically, investigation of iPSCs, iPSC-neural progenitor cells (iPSC-NPCs), and iPSC-neuroepithelial like stem cells (iPSC-lt-NESC) has demonstrated significant immunomodulation of proinflammatory signaling and endogenous inflammatory cell populations, such as microglia. This review aims to examine the mechanisms by which iPSCs mediate neuroinflammation in the post-stroke environment, as well as delineate avenues for further investigation.
Collapse
Affiliation(s)
- Samantha E Spellicy
- MD-Ph.D. Program, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - David C Hess
- Dean's Office, Medical College of Georgia at Augusta University, Augusta, GA, United States
| |
Collapse
|
16
|
Valerio LSA, Sugaya K. Xeno- and transgene-free reprogramming of mesenchymal stem cells toward the cells expressing neural markers using exosome treatments. PLoS One 2020; 15:e0240469. [PMID: 33048978 PMCID: PMC7553345 DOI: 10.1371/journal.pone.0240469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 09/26/2020] [Indexed: 01/03/2023] Open
Abstract
Neural stem cells (NSCs), capable of self-renew and differentiate into neural cells, hold promise for use in studies and treatments for neurological diseases. However, current approaches to obtain NSCs from a live brain are risky and invasive, since NSCs reside in the subventricular zone and the in the hippocampus dentate gyrus. Alternatively, mesenchymal stem cells (MSCs) could be a more available cell source due to their abundance in tissues and easier to access. However, MSCs are committed to producing mesenchymal tissue and are not capable of spontaneously differentiating into neural cells. Thus, the process of reprogramming of MSCs into neural cells to use in clinical and scientific settings has significantly impacted the advancement of regenerative medicine. Previously, our laboratory reported trans-differentiation of MSCs to neural cells through the induced pluripotent stem (iPS) cells state, which was produced by overexpression of the embryonic stem cell gene NANOG. In the current study, we demonstrate that treatment with exosomes derived from NSCs makes MSCs capable of expressing neural cell markers bypassing the generation of iPS cells. An epigenetic modifier, decitabine (5-aza-2'-deoxycytidine), enhanced the process. This novel Xeno and transgene-free trans-differentiation technology eliminates the issues associated with iPS cells, such as tumorigenesis. Thus, it may accelerate the development of neurodegenerative therapies and in vitro neurological disorder models for personalized medicine.
Collapse
Affiliation(s)
- Luis Sebástian Alexis Valerio
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States of America
- Institute for Scientific Research and Technology Services (INDICASAT), Panama City, Republic of Panama
- Department of Biotechnology, Acharya Nagarjuna University, Guntur, India
| | - Kiminobu Sugaya
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States of America
- Institute for Scientific Research and Technology Services (INDICASAT), Panama City, Republic of Panama
- * E-mail:
| |
Collapse
|
17
|
Sullivan GM, Knutsen AK, Peruzzotti-Jametti L, Korotcov A, Bosomtwi A, Dardzinski BJ, Bernstock JD, Rizzi S, Edenhofer F, Pluchino S, Armstrong RC. Transplantation of induced neural stem cells (iNSCs) into chronically demyelinated corpus callosum ameliorates motor deficits. Acta Neuropathol Commun 2020; 8:84. [PMID: 32517808 PMCID: PMC7285785 DOI: 10.1186/s40478-020-00960-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 05/30/2020] [Indexed: 12/19/2022] Open
Abstract
Multiple Sclerosis (MS) causes neurologic disability due to inflammation, demyelination, and neurodegeneration. Immunosuppressive treatments can modify the disease course but do not effectively promote remyelination or prevent long term neurodegeneration. As a novel approach to mitigate chronic stage pathology, we tested transplantation of mouse induced neural stem cells (iNSCs) into the chronically demyelinated corpus callosum (CC) in adult mice. Male C57BL/6 mice fed 0.3% cuprizone for 12 weeks exhibited CC atrophy with chronic demyelination, astrogliosis, and microglial activation. Syngeneic iNSCs were transplanted into the CC after ending cuprizone and perfused for neuropathology 2 weeks later. Magnetic resonance imaging (MRI) sequences for magnetization transfer ratio (MTR), diffusion-weighted imaging (T2), and diffusion tensor imaging (DTI) quantified CC pathology in live mice before and after iNSC transplantation. Each MRI technique detected progressive CC pathology. Mice that received iNSCs had normalized DTI radial diffusivity, and reduced astrogliosis post-imaging. A motor skill task that engages the CC is Miss-step wheel running, which demonstrated functional deficits from cuprizone demyelination. Transplantation of iNSCs resulted in marked recovery of running velocity. Neuropathology after wheel running showed that iNSC grafts significantly increased host oligodendrocytes and proliferating oligodendrocyte progenitors, while modulating axon damage. Transplanted iNSCs differentiated along astrocyte and oligodendrocyte lineages, without myelinating, and many remained neural stem cells. Our findings demonstrate the applicability of neuroimaging and functional assessments for pre-clinical interventional trials during chronic demyelination and detect improved function from iNSC transplantation. Directly reprogramming fibroblasts into iNSCs facilitates the future translation towards exogenous autologous cell therapies.
Collapse
|
18
|
miR-17 regulates the proliferation and differentiation of retinal progenitor cells by targeting CHMP1A. Biochem Biophys Res Commun 2020; 523:493-499. [PMID: 31894018 DOI: 10.1016/j.bbrc.2019.11.108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 11/17/2019] [Indexed: 11/22/2022]
Abstract
MicroRNAs have a vital effect on the differentiation of many types of progenitor cells. Recent studies have suggested that miR-17 plays an important role in the differentiation process of brain neural progenitor cells (NPC). Nevertheless, its detailed functions in regulating retinal progenitor cells (RPC) remain unclear. In our study, overexpression and knockdown of miR-17 were performed by transfecting RPC with mimics and inhibitors, respectively. Next, we investigated the role of miR-17 in RPC proliferation and differentiation by the following experiments: qPCR, CCK8, Edu staining, immunostaining and Western blot. The results revealed that miR-17 inhibited RPC proliferation but enhanced differentiation. Furthermore, according to a web-based database analysis, we identified charged multivesicular body protein 1A (CHMP1A) as a target gene. A dual luciferase reporter system showed that miR-17 specifically binds to the CHMP1A 3' untranslated region (UTR). Next, our data showed upregulation of miR-17 decreased CHMP1A protein level, causing reduced proliferation and enhanced differentiation of RPC. Downregulation of miR-17 led to enhanced CHMP1A protein expression, increased RPC proliferation and decreased differentiation. Taken together, our data provide a proven pathway by which miR-17 regulates RPC proliferation and differentiation by targeting CHMP1A.
Collapse
|
19
|
Bilodeau C, Goltsis O, Rogers IM, Post M. Limitations of recellularized biological scaffolds for human transplantation. J Tissue Eng Regen Med 2019; 14:521-538. [PMID: 31826325 DOI: 10.1002/term.3004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 12/15/2022]
Abstract
A shortage of donor organs for transplantation and the dependence of the recipients on immunosuppressive therapy have motivated researchers to consider alternative regenerative approaches. The answer may reside in acellular scaffolds generated from cadaveric human and animal tissues. Acellular scaffolds are expected to preserve the architectural and mechanical properties of the original organ, permitting cell attachment, growth, and differentiation. Although theoretically, the use of acellular scaffolds for transplantation should pose no threat to the recipient's immune system, experimental data have revealed significant immune responses to allogeneic and xenogeneic transplanted scaffolds. Herein, we review the various factors of the scaffold that could trigger an inflammatory and/or immune response, thereby compromising its use for human transplant therapy. In addition, we provide an overview of the major cell types that have been considered for recellularization of the scaffold and their potential contribution to triggering an immune response.
Collapse
Affiliation(s)
- Claudia Bilodeau
- Translational Medicine Program, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Olivia Goltsis
- Translational Medicine Program, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Ian M Rogers
- Lunenfeld Research Institute, Mount Sinai Health, Toronto, Ontario, Canada
| | - Martin Post
- Translational Medicine Program, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
20
|
Zhang S, Sun P, Lin K, Chan FHL, Gao Q, Lau WF, Roy VAL, Zhang H, Lai KWC, Huang Z, Yung KKL. Extracellular Nanomatrix-Induced Self-Organization of Neural Stem Cells into Miniature Substantia Nigra-Like Structures with Therapeutic Effects on Parkinsonian Rats. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1901822. [PMID: 31871862 PMCID: PMC6918115 DOI: 10.1002/advs.201901822] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/29/2019] [Indexed: 05/14/2023]
Abstract
Substantia nigra (SN) is a complex and critical region of the brain wherein Parkinson's disease (PD) arises from the degeneration of dopaminergic neurons. Miniature SN-like structures (mini-SNLSs) constructed from novel combination of nanomaterials and cell technologies exhibit promise as potentially curative cell therapies for PD. In this work, a rapid self-organization of mini-SNLS, with an organizational structure and neuronal identities similar to those of the SN in vivo, is achieved by differentiating neural stem cells in vitro on biocompatible silica nanozigzags (NZs) sculptured by glancing angle deposition, without traditional chemical growth factors. The differentiated neurons exhibit electrophysiological activity in vitro. Diverse physical cues and signaling pathways that are determined by the nanomatrices and lead to the self-organization of the mini-SNLSs are clarified and elucidated. In vivo, transplantation of the neurons from a mini-SNLS results in an early and progressive amelioration of PD in rats. The sculptured medical device reported here enables the rapid and specific self-organization of region-specific and functional brain-like structures without an undesirable prognosis. This development provides promising and significant insights into the screening of potentially curative drugs and cell therapies for PD.
Collapse
Affiliation(s)
- Shiqing Zhang
- Department of BiologyHong Kong Baptist University (HKBU)Kowloon TongKowloonHong Kong SAR China
- Golden Meditech Center for NeuroRegeneration SciencesHKBUKowloon TongKowloonHong Kong SAR China
- HKBU Institute of Research and Continuing Education, 9FThe Industrialization Complex of Shenzhen Virtual University ParkNo. 2 Yuexing 3rd Road, South Zone, Hi‐tech Industrial Park, Nanshan DistrictShenzhen518057Guangdong ProvinceChina
| | - Peng Sun
- Department of PhysicsHKBUKowloon TongKowloonHong Kong SAR China
- Department of Materials Science and EngineeringSouthern University of Science and TechnologyShenzhen518000Guangdong ProvinceChina
| | - Kaili Lin
- Department of BiologyHong Kong Baptist University (HKBU)Kowloon TongKowloonHong Kong SAR China
- Golden Meditech Center for NeuroRegeneration SciencesHKBUKowloon TongKowloonHong Kong SAR China
| | - Florence Hiu Ling Chan
- Department of Biomedical EngineeringCity University of Hong Kong (CityU)Tat Chee Avenue, Kowloon TongKowloonHong Kong SAR China
| | - Qi Gao
- Department of Biomedical EngineeringCity University of Hong Kong (CityU)Tat Chee Avenue, Kowloon TongKowloonHong Kong SAR China
| | - Wai Fung Lau
- Department of PhysicsHKBUKowloon TongKowloonHong Kong SAR China
| | - Vellaisamy A. L. Roy
- Department of Materials Science and EngineeringCity University of Hong KongTat Chee Avenue, Kowloon TongKowloonHong Kong SAR China
| | - Hongqi Zhang
- School of Chinese MedicineHKBUKowloon TongKowloonHong Kong SAR China
| | - King Wai Chiu Lai
- Department of Biomedical EngineeringCity University of Hong Kong (CityU)Tat Chee Avenue, Kowloon TongKowloonHong Kong SAR China
| | - Zhifeng Huang
- Golden Meditech Center for NeuroRegeneration SciencesHKBUKowloon TongKowloonHong Kong SAR China
- HKBU Institute of Research and Continuing Education, 9FThe Industrialization Complex of Shenzhen Virtual University ParkNo. 2 Yuexing 3rd Road, South Zone, Hi‐tech Industrial Park, Nanshan DistrictShenzhen518057Guangdong ProvinceChina
- Department of PhysicsHKBUKowloon TongKowloonHong Kong SAR China
- Institute of Advanced MaterialsState Key Laboratory of Environmental and Biological AnalysisHKBUKowloon TongKowloonHong Kong SAR China
| | - Ken Kin Lam Yung
- Department of BiologyHong Kong Baptist University (HKBU)Kowloon TongKowloonHong Kong SAR China
- Golden Meditech Center for NeuroRegeneration SciencesHKBUKowloon TongKowloonHong Kong SAR China
- HKBU Institute of Research and Continuing Education, 9FThe Industrialization Complex of Shenzhen Virtual University ParkNo. 2 Yuexing 3rd Road, South Zone, Hi‐tech Industrial Park, Nanshan DistrictShenzhen518057Guangdong ProvinceChina
- Institute of Advanced MaterialsState Key Laboratory of Environmental and Biological AnalysisHKBUKowloon TongKowloonHong Kong SAR China
| |
Collapse
|
21
|
Paim Á, Cardozo NSM, Tessaro IC, Pranke P. Relevant biological processes for tissue development with stem cells and their mechanistic modeling: A review. Math Biosci 2018; 301:147-158. [PMID: 29746816 DOI: 10.1016/j.mbs.2018.05.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 04/27/2018] [Accepted: 05/04/2018] [Indexed: 02/07/2023]
Abstract
A potential alternative for tissue transplants is tissue engineering, in which the interaction of cells and biomaterials can be optimized. Tissue development in vitro depends on the complex interaction of several biological processes such as extracellular matrix synthesis, vascularization and cell proliferation, adhesion, migration, death, and differentiation. The complexity of an individual phenomenon or of the combination of these processes can be studied with phenomenological modeling techniques. This work reviews the main biological phenomena in tissue development and their mathematical modeling, focusing on mesenchymal stem cell growth in three-dimensional scaffolds.
Collapse
Affiliation(s)
- Ágata Paim
- Department of Chemical Engineering, Universidade Federal do Rio Grande do Sul (UFRGS), R. Eng. Luis Englert, s/n Porto Alegre, Rio Grande do Sul 90040-040, Brazil; Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul (UFRGS), Av. Ipiranga, 2752. Porto Alegre, Rio Grande do Sul 90610-000, Brazil.
| | - Nilo S M Cardozo
- Department of Chemical Engineering, Universidade Federal do Rio Grande do Sul (UFRGS), R. Eng. Luis Englert, s/n Porto Alegre, Rio Grande do Sul 90040-040, Brazil
| | - Isabel C Tessaro
- Department of Chemical Engineering, Universidade Federal do Rio Grande do Sul (UFRGS), R. Eng. Luis Englert, s/n Porto Alegre, Rio Grande do Sul 90040-040, Brazil
| | - Patricia Pranke
- Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul (UFRGS), Av. Ipiranga, 2752. Porto Alegre, Rio Grande do Sul 90610-000, Brazil; Stem Cell Research Institute, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
22
|
Iwata H, Arima Y, Tsutsui Y. Design of Bioartificial Pancreases From the Standpoint of Oxygen Supply. Artif Organs 2018; 42:E168-E185. [PMID: 29611212 DOI: 10.1111/aor.13106] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 12/11/2017] [Accepted: 12/15/2017] [Indexed: 12/24/2022]
Abstract
A bioartificial pancreas (BAP), in which islets of Langerhans (islets) are enclosed in a semipermeable membrane, has been developed to realize islet transplantation without the use of immunosuppressive drugs. Although recent progress in induced pluripotent stem (iPS) and embryonic stem (ES) cells has attracted attention owing to the potential applications of these cells as insulin-releasing cells, concerns about the safety of implantation of these cells remain. The use of the BAP has the advantage of easy removal if insulin-releasing cells derived from iPS/ES cells undesirably proliferate and form tumors in the BAP. Oxygen supply is a crucial issue for cell survival in BAPs as insufficient oxygen supply causes central necrosis of cell aggregates. In this study, we derived several simple equations considering oxygen supply in BAPs in order to provide insights into the rational design of three different types of BAPs (spherical microcapsules, cylindrical capsules, and planar capsules). The equations give (i) the thickness of a capsule membrane leading to no central necrosis of encapsulated cell aggregates as a function of the original size of the cell aggregate; (ii) the oxygen concentration profiles in BAPs; (iii) the effects of encapsulation of a cell aggregate on insulin release; (iv) the amount of encapsulated cells required to normalize blood glucose levels of a patient; and (v) the total volumes and sizes of BAPs. As an example, we used the equations in order to design three different types of BAPs for subcutaneous implantation.
Collapse
Affiliation(s)
- Hiroo Iwata
- Compass to Healthy Life Research Complex Program, RIKEN, Kobe, Japan
| | - Yusuke Arima
- Institute for Life and Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | | |
Collapse
|
23
|
Induced neural stem cell-derived astrocytes modulate complement activation and mediate neuroprotection following closed head injury. Cell Death Dis 2018; 9:101. [PMID: 29367701 PMCID: PMC5833559 DOI: 10.1038/s41419-017-0172-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 10/25/2017] [Accepted: 11/22/2017] [Indexed: 11/09/2022]
Abstract
The complement system is a crucial component of immunity, and its activation has critical roles in neuroinflammatory response and cellular damage following closed head injury (CHI). We previously demonstrated that systemically injected induced neural stem cells (iNSCs) could modulate complement activation to ameliorate neuronal apoptosis in mouse CHI models. However, it remains unknown whether iNSC derivatives can regulate complement activation. In the present study, after CHI mouse serum treatment, we found dramatic decreases in the cellular viabilities of differentiated iNSCs. Interestingly, following CHI mouse serum treatment, the death of astrocytes derived from iNSCs which were pre-treated with CHI mouse serum was significantly decreased. Meanwhile, the deposition of C3 (C3d) and C5b-9 in these astrocytes was substantially reduced. Remarkably, we detected increased expression of complement receptor type 1-related protein y (Crry) in these astrocytes. Moreover, these astrocytes could reduce the numbers of apoptotic neurons via Crry expression post-CHI mouse serum treatment. Additionally, intracerebral-transplanted iNSCs, pre-treated with CHI mouse serum, significantly increased the levels of Crry expression in astrocytes to reduce the accumulation of C3d and C9 and the death of neurons in the brains of CHI mice. In summary, iNSCs receiving CHI mouse serum pre-treatment could enhance the expression of Crry in iNSC-derived astrocytes to modulate complement activation and mediate neuroprotection following CHI.
Collapse
|
24
|
Kwon D, Ahn HJ, Kang KS. Generation of Human Neural Stem Cells by Direct Phenotypic Conversion. Results Probl Cell Differ 2018; 66:103-121. [PMID: 30209656 DOI: 10.1007/978-3-319-93485-3_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Human neural stem cells (hNSC) are multipotent adult stem cells. Various studies are underway worldwide to identify new methods for treatment of neurological diseases using hNSC. This chapter summarizes the latest research trends in and fields for application of patient-specific hNSC using direct phenotypic conversion technology. The aim of the study was to analyze the advantages and disadvantages of current technology and to suggest relevant directions for future hNSC research.
Collapse
Affiliation(s)
- Daekee Kwon
- Stem Cells and Regenerative Bioengineering Institute in Kangstem Biotech, Seoul National University, Seoul, South Korea
| | - Hee-Jin Ahn
- Stem Cells and Regenerative Bioengineering Institute in Kangstem Biotech, Seoul National University, Seoul, South Korea
| | - Kyung-Sun Kang
- Stem Cells and Regenerative Bioengineering Institute in Kangstem Biotech, Seoul National University, Seoul, South Korea.
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, South Korea.
| |
Collapse
|
25
|
Yang Y, Du T, Zhang J, Kang T, Luo L, Tao J, Gou Z, Chen S, Du Y, He J, Jiang S, Mao Q, Gou M. A 3D-Engineered Conformal Implant Releases DNA Nanocomplexs for Eradicating the Postsurgery Residual Glioblastoma. ADVANCED SCIENCE 2017; 4:1600491. [PMID: 28852611 PMCID: PMC5566247 DOI: 10.1002/advs.201600491] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 02/01/2017] [Indexed: 02/05/2023]
Abstract
Gene therapy has great promise for glioblastoma treatment; however, it remains a great challenge to efficiently deliver genes to the brain. The incomplete resection of glioblastoma always leads to poor prognosis. Here, a 3D‐engineered conformal implant for eradicating the postsurgery residual glioblastoma is designed. This implant is constructed by 3D‐printing technology to match the tumor cavity and release an oncolytic virus‐inspired DNA nanocomplex to kill glioblastoma cells through apoptosis induction. Meanwhile, a 3D‐engineered subcutaneous glioblastoma xenograft is built to mimic the resection tumor cavity in mice. Insertion of the implant into the glioblastoma resection cavity efficiently delays tumor recurrence and significantly prolongs overall survival. This study provides a proof‐of‐concept of glioblastoma therapy using a conformal implant that releases oncolytic DNA nanocomplexs. This strategy can lead to the development of future precision therapy for eradicating postsurgery residual tumors.
Collapse
Affiliation(s)
- Yuan Yang
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University and Collaborative Innovation Center of Biotherapy; Chengdu P. R. China
- Department of Neurosurgery; West China Hospital; Sichuan University; Chengdu P. R. China
| | - Ting Du
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University and Collaborative Innovation Center of Biotherapy; Chengdu P. R. China
| | - Jiumeng Zhang
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University and Collaborative Innovation Center of Biotherapy; Chengdu P. R. China
| | - Tianyi Kang
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University and Collaborative Innovation Center of Biotherapy; Chengdu P. R. China
| | - Li Luo
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University and Collaborative Innovation Center of Biotherapy; Chengdu P. R. China
| | - Jie Tao
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University and Collaborative Innovation Center of Biotherapy; Chengdu P. R. China
- School of Materials Science and Engineering; Sichuan University; Chengdu Sichuan 610065 P. R. China
| | - Zhiyuan Gou
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University and Collaborative Innovation Center of Biotherapy; Chengdu P. R. China
| | - Shaochen Chen
- Department of Nanoengineering; Institute of Engineering in Medicine; 245B SME Building; MC 0448; University of California; San Diego 9500 Gilman Drive La Jolla CA 92093 USA
| | - Yanan Du
- Department of Biomedical Engineering; Tsinghua University School of Medicine; Beijing P. R. China
| | - Jiankang He
- State key laboratory for manufacturing systems engineering; Xi'an Jiaotong University; Xi'an 710049 China
| | - Shu Jiang
- Department of Neurosurgery; West China Hospital; Sichuan University; Chengdu P. R. China
| | - Qing Mao
- Department of Neurosurgery; West China Hospital; Sichuan University; Chengdu P. R. China
| | - Maling Gou
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University and Collaborative Innovation Center of Biotherapy; Chengdu P. R. China
| |
Collapse
|
26
|
Gao M, Yao H, Dong Q, Zhang Y, Yang Y, Zhang Y, Yang Z, Xu M, Xu R. Neurotrophy and immunomodulation of induced neural stem cell grafts in a mouse model of closed head injury. Stem Cell Res 2017; 23:132-142. [PMID: 28743043 DOI: 10.1016/j.scr.2017.07.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 07/10/2017] [Accepted: 07/15/2017] [Indexed: 11/26/2022] Open
Abstract
Closed head injury (CHI) usually results in severe and permanent neurological impairments, which are caused by several intertwined phenomena, such as cerebral edema, blood-brain barrier (BBB) disruption, neuronal loss, astroglial scarring and inflammation. We previously reported that induced neural stem cells (iNSCs), similar to neural stem cells (NSCs), can accelerate neurological recovery in vivo and produce neurotrophic factors in vitro. However, the effects of iNSC neurotrophy following CHI were not determined. Moreover, whether iNSCs have immunomodulatory properties is unknown. Mouse models of CHI were established using a standardized weight-drop device and assessed by neurological severity score (NSS). Although these models fail to mimic the complete spectrum of human CHI, they reproduce impairment in neurological function observed in clinical patients. Syngeneic iNSCs or NSCs were separately transplanted into the brains of CHI mice at 12h after CHI. Neurological impairment post-CHI was evaluated by several tests. Animals were sacrificed for morphological and molecular biological analyses. We discovered that iNSC administration promoted neurological functional recovery in CHI mice and reduced cerebral edema, BBB disruption, cell death and astroglial scarring following trauma. Implanted iNSCs could up-regulate brain-derived neurotrophic factor (BDNF) and glial-derived neurotrophic factor (GDNF) levels to support the survival of existing neurons after CHI. In addition, engrafted iNSCs decreased immune cell recruitment and pro-inflammatory cytokine expression in the brain post-injury. Moreover, we found significant nuclear factor-kappaB (NF-κB) inhibition in the presence of iNSC grafts. In short, iNSCs exert neurotrophic and immunomodulatory effects that mitigate CHI-induced neurological impairment.
Collapse
Affiliation(s)
- Mou Gao
- Department of Neurosurgery, The Third Affiliated Hospital of the Third Military Medical University, Chongqing 400042, China; Affiliated Bayi Brain Hospital, P.L.A Army General Hospital, Beijing 100700, China
| | - Hui Yao
- Affiliated Bayi Brain Hospital, P.L.A Army General Hospital, Beijing 100700, China
| | - Qin Dong
- Department of Neurology, Fu Xing Hospital, Capital Medical University, Beijing 100038, China
| | - Yan Zhang
- Affiliated Bayi Brain Hospital, P.L.A Army General Hospital, Beijing 100700, China
| | - Yang Yang
- Affiliated Bayi Brain Hospital, P.L.A Army General Hospital, Beijing 100700, China
| | - Yihua Zhang
- Department of Neurosurgery, The Third Affiliated Hospital of the Third Military Medical University, Chongqing 400042, China
| | - Zhijun Yang
- Affiliated Bayi Brain Hospital, P.L.A Army General Hospital, Beijing 100700, China
| | - Minhui Xu
- Department of Neurosurgery, The Third Affiliated Hospital of the Third Military Medical University, Chongqing 400042, China.
| | - Ruxiang Xu
- Affiliated Bayi Brain Hospital, P.L.A Army General Hospital, Beijing 100700, China.
| |
Collapse
|
27
|
Systemic Administration of Induced Neural Stem Cells Regulates Complement Activation in Mouse Closed Head Injury Models. Sci Rep 2017; 7:45989. [PMID: 28383046 PMCID: PMC5382667 DOI: 10.1038/srep45989] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/07/2017] [Indexed: 02/06/2023] Open
Abstract
Complement activation plays important roles in the pathogenesis of central nervous system (CNS) diseases. Patients face neurological disorders due to the development of complement activation, which contributes to cell apoptosis, brain edema, blood-brain barrier dysfunction and inflammatory infiltration. We previously reported that induced neural stem cells (iNSCs) can promote neurological functional recovery in closed head injury (CHI) animals. Remarkably, we discovered that local iNSC grafts have the potential to modulate CNS inflammation post-CHI. In this study, we aimed to explore the role of systemically delivered iNSCs in complement activation following CNS injury. Our data showed that iNSC grafts decreased the levels of sera C3a and C5a and down-regulated the expression of C3d, C9, active Caspase-3 and Bax in the brain, kidney and lung tissues of CHI mice. Furthermore, iNSC grafts decreased the levels of C3d+/NeuN+, C5b-9+/NeuN+, C3d+/Map2+ and C5b-9+/Map2+ neurons in the injured cortices of CHI mice. Subsequently, we explored the mechanisms underlying these effects. With flow cytometry analysis, we observed a dramatic increase in complement receptor type 1-related protein y (Crry) expression in iNSCs after CHI mouse serum treatment. Moreover, both in vitro and in vivo loss-of-function studies revealed that iNSCs could modulate complement activation via Crry expression.
Collapse
|
28
|
Gao M, Dong Q, Yao H, Zhang Y, Yang Y, Dang Y, Zhang H, Yang Z, Xu M, Xu R. Induced neural stem cells modulate microglia activation states via CXCL12/CXCR4 signaling. Brain Behav Immun 2017; 59:288-299. [PMID: 27650112 DOI: 10.1016/j.bbi.2016.09.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 09/16/2016] [Accepted: 09/16/2016] [Indexed: 12/25/2022] Open
Abstract
We previously reported that induced neural stem cells (iNSCs) directly reprogrammed from mouse embryonic fibroblasts can expand and differentiate into neurons, astrocytes and oligodendrocytes. Whether iNSCs have immunoregulatory properties in addition to facilitating cell replacement remains uncertain. In this study, we aimed to characterize the immunomodulatory effects of iNSCs on the activation states of microglia and to elucidate the mechanisms underlying these effects. Using a mouse model of closed head injury (CHI), we observed that iNSC grafts decreased the levels of ED1+/Iba1+ and TNF-α+/Iba1+ microglia but increased the levels of IGF1+/Iba1+ microglia in the injured cortex. Subsequently, using a Transwell co-culture system, we discovered that iNSCs could modulate LPS-pretreated microglia phenotypes in vitro via CXCL12/CXCR4 signaling, which we demonstrated through the administration of the CXCR4 antagonist AMD3100 and CXCR4-specific siRNA treatment. An in vivo loss-of-function study also revealed that iNSC grafts regulated the behavior of resident microglia via CXCL12/CXCR4 signaling, influencing their activation state such that they promoted neurological functional recovery and neuron survival. Furthermore, the beneficial effects of iNSC transplantation were significantly diminished by CXCR4 knockdown. In short, iNSCs have the potential to influence microglia activation and the acquisition of neuroprotective phenotypes via CXCL12/CXCR4 signaling.
Collapse
Affiliation(s)
- Mou Gao
- Department of Neurosurgery, The Third Affiliated Hospital of The Third Military Medical University, Chongqing 400042, China; Affiliated Bayi Brain Hospital, P.L.A Army General Hospital, Beijing 100700, China
| | - Qin Dong
- Department of Neurology, Fu Xing Hospital, Capital Medical University, Beijing 100038, China
| | - Hui Yao
- Affiliated Bayi Brain Hospital, P.L.A Army General Hospital, Beijing 100700, China
| | - Yan Zhang
- Affiliated Bayi Brain Hospital, P.L.A Army General Hospital, Beijing 100700, China
| | - Yang Yang
- Affiliated Bayi Brain Hospital, P.L.A Army General Hospital, Beijing 100700, China
| | - Yuanyuan Dang
- Affiliated Bayi Brain Hospital, P.L.A Army General Hospital, Beijing 100700, China
| | - Hongtian Zhang
- Affiliated Bayi Brain Hospital, P.L.A Army General Hospital, Beijing 100700, China
| | - Zhijun Yang
- Affiliated Bayi Brain Hospital, P.L.A Army General Hospital, Beijing 100700, China
| | - Minhui Xu
- Department of Neurosurgery, The Third Affiliated Hospital of The Third Military Medical University, Chongqing 400042, China.
| | - Ruxiang Xu
- Affiliated Bayi Brain Hospital, P.L.A Army General Hospital, Beijing 100700, China.
| |
Collapse
|