1
|
Gyimesi M, Oikari LE, Yu C, Sutherland HG, Nyholt DR, Griffiths LR, Van Wijnen AJ, Okolicsanyi RK, Haupt LM. CpG methylation changes in human mesenchymal and neural stem cells in response to in vitro niche modifications. Biochimie 2024; 223:147-157. [PMID: 38640996 DOI: 10.1016/j.biochi.2024.04.007] [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: 02/29/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/21/2024]
Abstract
Stem cell therapies hold promise in addressing the burden of neurodegenerative diseases with human embryonic neural stem cells (hNSC-H9s) and bone marrow-derived human mesenchymal stem cells (hMSCs) as viable candidates. The induction of hMSC neurospheres (hMSC-IN) generate a more lineage-restricted common neural progenitor-like cell population, potentially tunable by heparan sulfate proteoglycans (HSPGs). We examined CpG (5 mC) site methylation patterns using Illumina Infinium 850 K EPIC arrays in hNSC-H9, hMSCs and hMSC-IN cultures with HSPG agonist heparin at early and late phases of growth. We identified key regulatory CpG sites in syndecans (SDC2; SDC4) that potentially regulate gene expression in monolayers. Unique hMSC-IN hypomethylation in glypicans (GPC3; GPC4) underscore their significance in neural lineages with Sulfatase 1 and 2 (SULF1 &2) CpG methylation changes potentially driving the neurogenic shift. hMSC-INs methylation levels at SULF1 CpG sites and SULF2:cg25401628 were more closely aligned with hNSC-H9 cells than with hMSCs. We further suggest SOX2 regulation governed by lncSOX2-Overall Transcript (lncSOX2-OT) methylation changes with preferential activation of ENO2 over other neuronal markers within hMSC-INs. Our findings illuminate epigenetic dynamics governing neural lineage commitment of hMSC-INs offering insights for targeted mechanisms for regenerative medicine and therapeutic strategies.
Collapse
Affiliation(s)
- Martina Gyimesi
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Australia
| | - Lotta E Oikari
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Australia
| | - Chieh Yu
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Australia
| | - Heidi G Sutherland
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Australia
| | - Dale R Nyholt
- Statistical and Genomic Epidemiology Laboratory, School of Biomedical Sciences, Faculty of Health and Centre for Genomics and Personalised Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Lyn R Griffiths
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Australia
| | | | - Rachel K Okolicsanyi
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Australia; Max Planck Queensland Centre for the Materials Science of Extracellular Matrices, Australia
| | - Larisa M Haupt
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology (QUT), Australia; Max Planck Queensland Centre for the Materials Science of Extracellular Matrices, Australia; Centre for Biomedical Technologies, Queensland University of Technology (QUT), 60 Musk Ave., Kelvin Grove, QLD 4059, Australia.
| |
Collapse
|
2
|
He XY, Zhou YR, Mu T, Liao YF, Jiang L, Qin Y, Cai JH. Magnetic resonance imaging focused on the ferritin heavy chain 1 reporter gene detects neuronal differentiation in stem cells. Neural Regen Res 2023; 18:1563-1569. [PMID: 36571363 PMCID: PMC10075097 DOI: 10.4103/1673-5374.358608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The neuronal differentiation of mesenchymal stem cells offers a new strategy for the treatment of neurological disorders. Thus, there is a need to identify a noninvasive and sensitive in vivo imaging approach for real-time monitoring of transplanted stem cells. Our previous study confirmed that magnetic resonance imaging, with a focus on the ferritin heavy chain 1 reporter gene, could track the proliferation and differentiation of bone marrow mesenchymal stem cells that had been transduced with lentivirus carrying the ferritin heavy chain 1 reporter gene. However, we could not determine whether or when bone marrow mesenchymal stem cells had undergone neuronal differentiation based on changes in the magnetic resonance imaging signal. To solve this problem, we identified a neuron-specific enolase that can be differentially expressed before and after neuronal differentiation in stem cells. In this study, we successfully constructed a lentivirus carrying the neuron-specific enolase promoter and expressing the ferritin heavy chain 1 reporter gene; we used this lentivirus to transduce bone marrow mesenchymal stem cells. Cellular and animal studies showed that the neuron-specific enolase promoter effectively drove the expression of ferritin heavy chain 1 after neuronal differentiation of bone marrow mesenchymal stem cells; this led to intracellular accumulation of iron and corresponding changes in the magnetic resonance imaging signal. In summary, we established an innovative magnetic resonance imaging approach focused on the induction of reporter gene expression by a neuron-specific promoter. This imaging method can be used to noninvasively and sensitively detect neuronal differentiation in stem cells, which may be useful in stem cell-based therapies.
Collapse
Affiliation(s)
- Xiao-Ya He
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yi-Rui Zhou
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Tong Mu
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing; Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yi-Fan Liao
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics; Department of Nuclear Medicine, The Second Hospital of the Army Medical University, Chongqing, China
| | - Li Jiang
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yong Qin
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Jin-Hua Cai
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
3
|
Chen J, Li D, Li H, Zhu K, Shi L, Fu X. Cell membrane-targeting NIR fluorescent probes with large Stokes shifts for ultralong-term transplanted neural stem cell tracking. Front Bioeng Biotechnol 2023; 11:1139668. [PMID: 36845195 PMCID: PMC9948019 DOI: 10.3389/fbioe.2023.1139668] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 01/20/2023] [Indexed: 02/11/2023] Open
Abstract
There is an emerging therapeutic strategy to transplant stem cells into diseased host tissue for various neurodegenerative diseases, owing to their self-renewal ability and pluripotency. However, the traceability of long-term transplanted cells limits the further understanding of the mechanism of the therapy. Herein, we designed and synthesized a quinoxalinone scaffold-based near-infrared (NIR) fluorescent probe named QSN, which exhibits ultra-strong photostability, large Stokes shift, and cell membrane-targeting capacity. It could be found that QSN-labeled human embryonic stem cells showed strong fluorescent emission and photostability both in vitro and in vivo. Additionally, QSN would not impair the pluripotency of embryonic stem cells, indicating that QSN did not perform cytotoxicity. Moreover, it is worth mentioning that QSN-labeled human neural stem cells held cellular retention for at least 6 weeks in the mouse brain striatum post transplantation. All these findings highlight the potential application of QSN for ultralong-term transplanted cell tracking.
Collapse
Affiliation(s)
- Jing Chen
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Dan Li
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Hongfu Li
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Kongkai Zhu
- Advanced Medical Research Institute, Shandong University, Jinan, China,*Correspondence: Kongkai Zhu, ; Leilei Shi, ; Xuemei Fu,
| | - Leilei Shi
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China,*Correspondence: Kongkai Zhu, ; Leilei Shi, ; Xuemei Fu,
| | - Xuemei Fu
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China,*Correspondence: Kongkai Zhu, ; Leilei Shi, ; Xuemei Fu,
| |
Collapse
|
4
|
Fan H, Duan H, Hao P, Gao Y, Zhao W, Hao F, Li X, Yang Z. Cellular regeneration treatments for traumatic brain injury. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2022. [DOI: 10.1016/j.medntd.2022.100182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
5
|
Liu T, Li Z, Li X, Zhao R, Wei X, Wang Z, Xin SX. In vivo visualization of murine melanoma cells B16-derived exosomes through magnetic resonance imaging. Biochim Biophys Acta Gen Subj 2022; 1866:130062. [PMID: 34822924 DOI: 10.1016/j.bbagen.2021.130062] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 10/22/2021] [Accepted: 11/17/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Numerous studies demonstrated that exosomes play a powerful role in mediating intercellular communication to induce a pro-tumoral environment to promote tumor progression, including pre-metastatic niche formation and metastasis. Noninvasive imaging could determine the in vivo kinetics of exosomes in real time to provide better understanding of the mechanisms of the tumor formation, progression and metastasis. Magnetic resonance imaging (MRI) is an ideal technique which provides excellent anatomical resolution, intrinsic soft tissue contrast, unlimited penetration depth and no radiation exposure. METHODS A fusion protein composed of ferritin heavy chain (FTH1) and lactadherin was designed for visualizing exosomes through MRI. FTH1 was served as MRI reporter protein and lactadherin is a membrane-associated protein that is distributed on exosome surface. The characterizations of labeled exosomes were validated through transmission electron microscopy, western blot, nanoparticle tracking analysis and finally visualized in vitro and in vivo through MRI. RESULTS MR imaging showed that the labeled exosomes are able to be visualized in vitro and in vivo. Verification of the characterizations of exosomes observed no significant difference between labeled and unlabeled exosomes. CONCLUSION The proposed FTH1 labeling method was useful for visualizing exosomes through MRI. GENERAL SIGNIFICANCE The present study first reported a novel self-label method for imaging labeled exosomes of tumor cells in vivo through MR with cell endogenous MRI reporter protein. It may be further used as a tool to enhance understanding the role of exosomes in various pathophysiological conditions.
Collapse
Affiliation(s)
- Tianqi Liu
- School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Zhenlin Li
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Xiaodong Li
- School of Medicine, South China University of Technology, Guangzhou 510006, Guangdong, China
| | - Ruiting Zhao
- School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Xinhua Wei
- Department of Radiology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, Guangdong, China
| | - Zixin Wang
- School of Electronics and Information Technology, Sun Yat-Sen University, Xingang Xi Road 135, Guangzhou 510275, Guangdong, China
| | - Sherman Xuegang Xin
- School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, Guangdong, China; School of Medicine, South China University of Technology, Guangzhou 510006, Guangdong, China.
| |
Collapse
|
6
|
Van der Linden A, Hoehn M. Monitoring Neuronal Network Disturbances of Brain Diseases: A Preclinical MRI Approach in the Rodent Brain. Front Cell Neurosci 2022; 15:815552. [PMID: 35046778 PMCID: PMC8761853 DOI: 10.3389/fncel.2021.815552] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/06/2021] [Indexed: 12/20/2022] Open
Abstract
Functional and structural neuronal networks, as recorded by resting-state functional MRI and diffusion MRI-based tractography, gain increasing attention as data driven whole brain imaging methods not limited to the foci of the primary pathology or the known key affected regions but permitting to characterize the entire network response of the brain after disease or injury. Their connectome contents thus provide information on distal brain areas, directly or indirectly affected by and interacting with the primary pathological event or affected regions. From such information, a better understanding of the dynamics of disease progression is expected. Furthermore, observation of the brain's spontaneous or treatment-induced improvement will contribute to unravel the underlying mechanisms of plasticity and recovery across the whole-brain networks. In the present review, we discuss the values of functional and structural network information derived from systematic and controlled experimentation using clinically relevant animal models. We focus on rodent models of the cerebral diseases with high impact on social burdens, namely, neurodegeneration, and stroke.
Collapse
Affiliation(s)
- Annemie Van der Linden
- Bio-Imaging Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- μNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Mathias Hoehn
- Research Center Jülich, Institute 3 for Neuroscience and Medicine, Jülich, Germany
- *Correspondence: Mathias Hoehn
| |
Collapse
|
7
|
Hamblin MH, Murad R, Yin J, Vallim G, Lee JP. Modulation of gene expression on a transcriptome-wide level following human neural stem cell transplantation in aged mouse stroke brains. Exp Neurol 2022; 347:113913. [PMID: 34752785 PMCID: PMC8647207 DOI: 10.1016/j.expneurol.2021.113913] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 10/29/2021] [Accepted: 11/01/2021] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Neural stem cell (NSC) transplantation offers great potential for treating ischemic stroke. Clinically, ischemia followed by reperfusion results in robust cerebrovascular injury that upregulates proinflammatory factors, disrupts neurovascular units, and causes brain cell death. NSCs possess multiple actions that can be exploited for reducing the severity of neurovascular injury. Our previous studies in young adult mice showed that human NSC transplantation during the subacute stage diminishes stroke pathophysiology and improves behavioral outcome. METHODS We employed a well-established and commonly used stroke model, middle cerebral artery occlusion with subsequent reperfusion (MCAO/R). Here, we assessed the outcomes of hNSC transplantation 48 h post-MCAO (24 h post-transplant) in aged mouse brains in response to stroke because aging is a crucial risk factor for cerebral ischemia. Next, we tested whether administration of the integrin α5β1 inhibitor, ATN-161, prior to hNSC transplantation further affects stoke outcome as compared with NSCs alone. RNA sequencing (RNA-seq) was used to assess the impact of hNSC transplantation on differentially expressed genes (DEGs) on a transcriptome-wide level. RESULTS Here, we report that hNSC-engrafted brains with or without ATN-161 showed significantly reduced infarct size, and attenuated the induction of proinflammatory factors and matrix metalloproteases. RNA-seq analysis revealed DEGs and molecular pathways by which hNSCs induce a beneficial post-stroke outcome in aged stroke brains. 811 genes were differentially expressed (651 downregulated and 160 upregulated) in hNSC-engrafted stroke brains. Functional pathway analysis identified enriched and depleted pathways in hNSC-engrafted aged mouse stroke brains. Depletion of pathways following hNSC-engraftment included signaling involving neuroinflammation, acute phase response, leukocyte extravasation, and phagosome formation. On the other hand, enrichment of pathways in hNSC-engrafted brains was associated with PPAR signaling, LXR/RXR activation, and inhibition of matrix metalloproteases. Hierarchical cluster analysis of DEGs in hNSC-engrafted brains indicate decreased expression of genes encoding TNF receptors, proinflammatory factors, apoptosis factors, adhesion and leukocyte extravasation, and Toll-like receptors. CONCLUSIONS Our study is the first to show global transcripts differentially expressed following hNSC transplantation in the subacute phase of stroke in aged mice. The outcome of our transcriptome study would be useful to develop new therapies ameliorating early-stage stroke injury.
Collapse
Affiliation(s)
- Milton H Hamblin
- Tulane University Health Sciences Center, Tulane University, New Orleans, LA 70112, USA.
| | - Rabi Murad
- Bioinformatics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Jun Yin
- Bioinformatics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Gustavo Vallim
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jean-Pyo Lee
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| |
Collapse
|
8
|
Mousavinejad M, Skidmore S, Barone FG, Tyers P, Pisupati V, Poptani H, Plagge A, Barker RA, Murray P, Taylor A, Hill CJ. Assessing Human Embryonic Stem Cell-Derived Dopaminergic Neuron Progenitor Transplants Using Non-invasive Imaging Techniques. Mol Imaging Biol 2021; 22:1244-1254. [PMID: 32378000 PMCID: PMC7497430 DOI: 10.1007/s11307-020-01499-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
PURPOSE Human pluripotent stem cell (hPSC)-derived dopaminergic neuron progenitor cells (DAPCs) are a potential therapy for Parkinson's disease (PD). However, their intracranial administration raises safety concerns including uncontrolled proliferation, migration and inflammation. Here, we apply a bimodal imaging approach to investigate the fate of DAPC transplants in the rat striatum. PROCEDURES DAPCs co-expressing luciferase and ZsGreen or labelled with micron-sized particles of iron oxide (MPIOs) were transplanted in the striatum of RNU rats (n = 6 per group). DAPCs were tracked in vivo using bioluminescence and magnetic resonance (MR) imaging modalities. RESULTS Transgene silencing in differentiating DAPCs accompanied with signal attenuation due to animal growth rendered the bioluminescence undetectable by week 2 post intrastriatal transplantation. However, MR imaging of MPIO-labelled DAPCs showed that transplanted cells remained at the site of injection for over 120 days. Post-mortem histological analysis of DAPC transplants demonstrated that labelling with either luciferase/ZsGreen or MPIOs did not affect the ability of cells to differentiate into mature dopaminergic neurons. Importantly, labelled cells did not elicit increased glial reactivity compared to non-labelled cells. CONCLUSIONS In summary, our findings support the transplantation of hPSC-derived DAPCs as a safe treatment for PD.
Collapse
Affiliation(s)
- M Mousavinejad
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK
| | - S Skidmore
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK.,WT-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - F G Barone
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK
| | - P Tyers
- John van Geest Centre for Brain Repair & Department of Neurology, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - V Pisupati
- WT-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - H Poptani
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK
| | - A Plagge
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK
| | - R A Barker
- WT-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.,John van Geest Centre for Brain Repair & Department of Neurology, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - P Murray
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK
| | - A Taylor
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK.
| | - C J Hill
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK. .,Centre for Women's Health Research, Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, L8 7SS, UK.
| |
Collapse
|
9
|
Huang D, Cao Y, Yang X, Liu Y, Zhang Y, Li C, Chen G, Wang Q. A Nanoformulation-Mediated Multifunctional Stem Cell Therapy with Improved Beta-Amyloid Clearance and Neural Regeneration for Alzheimer's Disease. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2006357. [PMID: 33624894 DOI: 10.1002/adma.202006357] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 12/21/2020] [Indexed: 06/12/2023]
Abstract
Alzheimer's disease (AD) is a common dementia that is currently incurable. The existing treatments can only moderately relieve the symptoms of AD to slow down its progress. How to achieve effective neural regeneration to ameliorate cognitive impairments is a major challenge for current AD treatment. Here, the therapeutic potential of a nanoformulation-mediated neural stem cell (NSC) therapy capable of simultaneous Aβ clearance and neural regeneration is investigated in a murine model. Genetically engineered NSCs capable of stably and continuously expressing neprilysin (NEP) are developed to enhance Aβ degradation and NSC survival in the brain. A PBAE-PLGA-Ag2 S-RA-siSOX9 (PPAR-siSOX9) nanoformulation with high gene/drug deliverability is synthesized to overcome AD microenvironment-associated adverse effects and to promote neuronal differentiation of the NEP-expressing NSCs. For achieving accurate stereotactic transplantation, Ag2 S quantum-dot-based fluorescence imaging is used to guide NSC transplantation in real time. This strategy shows numerous benefits, including efficient and long-lasting Aβ degradation, improved neural regeneration, and accurate cell transplantation. It is shown that a single administration of this therapy achieves long-term efficacy (6 months) with respect to memory reversal and improvement of learning deficits.
Collapse
Affiliation(s)
- Dehua Huang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Yuheng Cao
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Xue Yang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Yongyang Liu
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Yejun Zhang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Chunyan Li
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Guangcun Chen
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Qiangbin Wang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
- College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
10
|
Pottmeier P, Doszyn O, Peuckert C, Jazin E. Increased Expression of Y-Encoded Demethylases During Differentiation of Human Male Neural Stem Cells. Stem Cells Dev 2020; 29:1497-1509. [PMID: 33040644 DOI: 10.1089/scd.2020.0138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Human neural stem cells (hNSCs) have long been used as an in vitro model to study neurogenesis and as candidates for nervous system therapy. Many parameters have been considered when evaluating the success of transplantation, but sex of donor and recipients is often not discussed. We investigated two commercial NSC lines, the female hNSC-H9 and male hNSC-H14, and we observed faster growth rates in the male cells. At 4 days of differentiation, male cells presented a significant increase in expression of DCX, an immature neuronal marker, while female cells showed a significant increase in RMST, a long noncoding RNA, which is indispensable during neurogenesis. In addition, expression of neural markers MAP2, PSD95, SYP, DCX, and TUJ1 at day 14 of differentiation suggested a similar differentiation potential in both lines. The most significant differences at day 14 of differentiation were the expression levels of RELN, with almost 100-fold difference between the sexes, and MASH1, with more than 1,000-fold increase in male cells. To evaluate whether some of the observed differences may be sex related, we measured the expression of gametologous genes located on the X- and Y-chromosome. Most noticeable was the increase of Y-encoded demethylases KDM6C (UTY) and KDM5D during differentiation of male cells. Our results indicate that attention should be paid to sex when planning neurogenesis and transplantation experiments.
Collapse
Affiliation(s)
- Philipp Pottmeier
- Department of Organismal Biology, EBC, Uppsala University, Uppsala, Sweden
| | - Olga Doszyn
- Department of Organismal Biology, EBC, Uppsala University, Uppsala, Sweden
| | - Christiane Peuckert
- Department of Organismal Biology, EBC, Uppsala University, Uppsala, Sweden.,Department of Molecular Biology, Stockholm University, Stockholm, Sweden
| | - Elena Jazin
- Department of Organismal Biology, EBC, Uppsala University, Uppsala, Sweden
| |
Collapse
|
11
|
Madsen SD, Giler MK, Bunnell BA, O'Connor KC. Illuminating the Regenerative Properties of Stem Cells In Vivo with Bioluminescence Imaging. Biotechnol J 2020; 16:e2000248. [PMID: 33089922 DOI: 10.1002/biot.202000248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 10/17/2020] [Indexed: 11/10/2022]
Abstract
Preclinical animal studies are essential to the development of safe and effective stem cell therapies. Bioluminescence imaging (BLI) is a powerful tool in animal studies that enables the real-time longitudinal monitoring of stem cells in vivo to elucidate their regenerative properties. This review describes the application of BLI in preclinical stem cell research to address critical challenges in producing successful stem cell therapeutics. These challenges include stem cell survival, proliferation, homing, stress response, and differentiation. The applications presented here utilize bioluminescence to investigate a variety of stem and progenitor cells in several different in vivo models of disease and implantation. An overview of luciferase reporters is provided, along with the advantages and disadvantages of BLI. Additionally, BLI is compared to other preclinical imaging modalities and potential future applications of this technology are discussed in emerging areas of stem cell research.
Collapse
Affiliation(s)
- Sean D Madsen
- Department of Chemical and Biomolecular Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, 70118, USA.,Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Margaret K Giler
- Department of Chemical and Biomolecular Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, 70118, USA.,Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Bruce A Bunnell
- Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA.,Department of Pharmacology, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Kim C O'Connor
- Department of Chemical and Biomolecular Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, 70118, USA.,Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| |
Collapse
|
12
|
Boese AC, Eckert A, Hamblin MH, Lee JP. Human neural stem cells improve early stage stroke outcome in delayed tissue plasminogen activator-treated aged stroke brains. Exp Neurol 2020; 329:113275. [PMID: 32147438 PMCID: PMC7609039 DOI: 10.1016/j.expneurol.2020.113275] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/26/2020] [Accepted: 03/03/2020] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Clinically, significant stroke injury results from ischemia-reperfusion (IR), which induces a deleterious biphasic opening of the blood-brain barrier (BBB). Tissue plasminogen activator (tPA) remains the sole pharmacological agent to treat ischemic stroke. However, major limitations of tPA treatment include a narrow effective therapeutic window of 4.5 h in most patients after initial stroke onset and off-target non-thrombolytic effects (e.g., the risk of increased IR injury). We hypothesized that ameliorating BBB damage with exogenous human neural stem cells (hNSCs) would improve stroke outcome to a greater extent than treatment with delayed tPA alone in aged stroke mice. METHODS We employed middle cerebral artery occlusion to produce focal ischemia with subsequent reperfusion (MCAO/R) in aged mice and administered tPA at a delayed time point (6 h post-stroke) via tail vein. We transplanted hNSCs intracranially in the subacute phase of stroke (24 h post-stroke). We assessed the outcomes of hNSC transplantation on pathophysiological markers of stroke 48 h post-stroke (24 h post-transplant). RESULTS Delayed tPA treatment resulted in more extensive BBB damage and inflammation relative to MCAO controls. Notably, transplantation of hNSCs ameliorated delayed tPA-induced escalated stroke damage; decreased expression of proinflammatory factors (tumor necrosis factor-alpha (TNF-α) and interleukin (IL)-6), decreased the level of matrix metalloprotease-9 (MMP-9), increased the level of brain-derived neurotrophic factor (BDNF), and reduced BBB damage. CONCLUSIONS Aged stroke mice that received delayed tPA treatment in combination with hNSC transplantation exhibited reduced stroke pathophysiology in comparison to non-transplanted stroke mice with delayed tPA. This suggests that hNSC transplantation may synergize with already existing stroke therapies to benefit a larger stroke patient population.
Collapse
Affiliation(s)
- Austin C Boese
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Auston Eckert
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Milton H Hamblin
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jean-Pyo Lee
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70112, USA.
| |
Collapse
|
13
|
Schomann T, Iljas JD, Que I, Li Y, Suidgeest E, Cruz LJ, Frijns JHM, Chan A, Löwik CMWG, Huisman MA, Mezzanotte L. Multimodal imaging of hair follicle bulge-derived stem cells in a mouse model of traumatic brain injury. Cell Tissue Res 2020; 381:55-69. [PMID: 32036485 PMCID: PMC7306043 DOI: 10.1007/s00441-020-03173-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 01/20/2020] [Indexed: 01/01/2023]
Abstract
Traumatic brain injury (TBI) is a devastating event for which current therapies are limited. Stem cell transplantation may lead to recovery of function via different mechanisms, such as cell replacement through differentiation, stimulation of angiogenesis and support to the microenvironment. Adult hair follicle bulge-derived stem cells (HFBSCs) possess neuronal differentiation capacity, are easy to harvest and are relatively immune-privileged, which makes them potential candidates for autologous stem cell-based therapy. In this study, we apply in vivo multimodal, optical and magnetic resonance imaging techniques to investigate the behavior of mouse HFBSCs in a mouse model of TBI. HFBSCs expressed Luc2 and copGFP and were examined for their differentiation capacity in vitro. Subsequently, transduced HFBSCs, preloaded with ferumoxytol, were transplanted next to the TBI lesion (cortical region) in nude mice, 2 days after injury. Brains were fixed for immunohistochemistry 58 days after transplantation. Luc2- and copGFP-expressing, ferumoxytol-loaded HFBSCs showed adequate neuronal differentiation potential in vitro. Bioluminescence of the lesioned brain revealed survival of HFBSCs and magnetic resonance imaging identified their localization in the area of transplantation. Immunohistochemistry showed that transplanted cells stained for nestin and neurofilament protein (NF-Pan). Cells also expressed laminin and fibronectin but extracellular matrix masses were not detected. After 58 days, ferumoxytol could be detected in HFBSCs in brain tissue sections. These results show that HFBSCs are able to survive after brain transplantation and suggest that cells may undergo differentiation towards a neuronal cell lineage, which supports their potential use for cell-based therapy for TBI.
Collapse
Affiliation(s)
- Timo Schomann
- Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, Leiden, the Netherlands
- Percuros B.V, Leiden, the Netherlands
| | - Juvita D Iljas
- Percuros B.V, Leiden, the Netherlands
- Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Ivo Que
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Yuedan Li
- Percuros B.V, Leiden, the Netherlands
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ernst Suidgeest
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Luis J Cruz
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Johan H M Frijns
- Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, Leiden, the Netherlands
- Leiden Institute for Brain and Cognition, Leiden University, Leiden, the Netherlands
| | - Alan Chan
- Percuros B.V, Leiden, the Netherlands
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Clemens M W G Löwik
- Optical Molecular Imaging, Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
- Department of Molecular Genetics, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Margriet A Huisman
- Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, Leiden, the Netherlands
- Hair Science Institute, Maastricht, the Netherlands
| | - Laura Mezzanotte
- Optical Molecular Imaging, Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, the Netherlands.
- Department of Molecular Genetics, Erasmus Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
14
|
Minassian A, Green C, Diedenhofen M, Vogel S, Hess S, Stoeber M, Radmilovic MD, Wiedermann D, Kloppenburg P, Hoehn M. Human Neural Stem Cell Induced Functional Network Stabilization After Cortical Stroke: A Longitudinal Resting-State fMRI Study in Mice. Front Cell Neurosci 2020; 14:86. [PMID: 32317940 PMCID: PMC7155295 DOI: 10.3389/fncel.2020.00086] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 03/20/2020] [Indexed: 12/12/2022] Open
Abstract
Most stroke studies dealing with functional deficits and assessing stem cell therapy produce extensive hemispheric damage and can be seen as a model for severe clinical strokes. However, mild strokes have a better prospect for functional recovery. Recently, anatomic and behavioral changes have been reported for distal occlusion of the middle cerebral artery (MCA), generating a well-circumscribed and small cortical lesion, which can thus be proposed as mild to moderate cortical stroke. Using this cortical stroke model of moderate severity in the nude mouse, we have studied the functional networks with resting-state functional magnetic resonance imaging (fMRI) for 12 weeks following stroke induction. Further, human neural stem cells (hNSCs) were implanted adjacent to the ischemic lesion, and the stable graft vitality was monitored with bioluminescence imaging (BLI). Differentiation of the grafted neural stem cells was analyzed by immunohistochemistry and by patch-clamp electrophysiology. Following stroke induction, we found a pronounced and continuously rising hypersynchronicity of the sensorimotor networks including both hemispheres, in contrast to the severe stroke filament model where profound reduction of the functional connectivity had been reported by us earlier. The vitality of grafted neural stem cells remained stable throughout the whole 12 weeks observation period. In the stem cell treated animals, functional connectivity did not show hypersynchronicity but was globally slightly reduced below baseline at 2 weeks post-stroke, normalizing thereafter completely. Our resting-state fMRI (rsfMRI) studies on cortical stroke reveal for the first time a hypersynchronicity of the functional brain networks. This hypersynchronicity appears as a hallmark of mild cortical strokes, in contrast to severe strokes with striatal involvement where exclusively hyposynchronicity has been reported. The effect of the stem cell graft was an early and persistent normalization of the functional sensorimotor networks across the whole brain. These novel functional results may help interpret future outcome investigations after stroke and demonstrate the highly promising potential of stem cell treatment for functional outcome improvement after stroke.
Collapse
Affiliation(s)
- Anuka Minassian
- In-Vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Claudia Green
- In-Vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Michael Diedenhofen
- In-Vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Stefanie Vogel
- In-Vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Simon Hess
- Biocenter, Institute for Zoology, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Maren Stoeber
- In-Vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Marina Dobrivojevic Radmilovic
- In-Vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany.,Department of Histology and Embryology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Dirk Wiedermann
- In-Vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Peter Kloppenburg
- Biocenter, Institute for Zoology, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Mathias Hoehn
- In-Vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany.,Department of Radiology, Leiden University Medical Center, Leiden University, Leiden, Netherlands
| |
Collapse
|
15
|
Vogel S, Hoehn M, Aswendt M. In vivo bioluminescence imaging to elucidate stem cell graft differentiation. Neural Regen Res 2020; 15:61-62. [PMID: 31535647 PMCID: PMC6862421 DOI: 10.4103/1673-5374.264449] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Stefanie Vogel
- Technische Universität Dresden, DFG-Research Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
| | - Mathias Hoehn
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Juelich, Juelich, Germany; Leiden University Medical Center, Department of Radiology, Leiden, The Netherlands
| | - Markus Aswendt
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, Cologne, Germany
| |
Collapse
|
16
|
Pongrac IM, Radmilović MD, Ahmed LB, Mlinarić H, Regul J, Škokić S, Babič M, Horák D, Hoehn M, Gajović S. D-mannose-Coating of Maghemite Nanoparticles Improved Labeling of Neural Stem Cells and Allowed Their Visualization by ex vivo MRI after Transplantation in the Mouse Brain. Cell Transplant 2019; 28:553-567. [PMID: 31293167 PMCID: PMC7103599 DOI: 10.1177/0963689719834304] [Citation(s) in RCA: 9] [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: 06/10/2018] [Revised: 12/26/2018] [Accepted: 02/05/2019] [Indexed: 12/14/2022] Open
Abstract
Magnetic resonance imaging (MRI) of superparamagnetic iron oxide-labeled cells can be used as a non-invasive technique to track stem cells after transplantation. The aim of this study was to (1) evaluate labeling efficiency of D-mannose-coated maghemite nanoparticles (D-mannose(γ-Fe2O3)) in neural stem cells (NSCs) in comparison to the uncoated nanoparticles, (2) assess nanoparticle utilization as MRI contrast agent to visualize NSCs transplanted into the mouse brain, and (3) test nanoparticle biocompatibility. D-mannose(γ-Fe2O3) labeled the NSCs better than the uncoated nanoparticles. The labeled cells were visualized by ex vivo MRI and their localization subsequently confirmed on histological sections. Although the progenitor properties and differentiation of the NSCs were not affected by labeling, subtle effects on stem cells could be detected depending on dose increase, including changes in cell proliferation, viability, and neurosphere diameter. D-mannose coating of maghemite nanoparticles improved NSC labeling and allowed for NSC tracking by ex vivo MRI in the mouse brain, but further analysis of the eventual side effects might be necessary before translation to the clinic.
Collapse
Affiliation(s)
- Igor M. Pongrac
- University of Zagreb School of Medicine, Croatian Institute for Brain
Research, Zagreb, Croatia
| | | | - Lada Brkić Ahmed
- University of Zagreb School of Medicine, Croatian Institute for Brain
Research, Zagreb, Croatia
| | - Hrvoje Mlinarić
- University of Zagreb School of Medicine, Croatian Institute for Brain
Research, Zagreb, Croatia
| | - Jan Regul
- University of Zagreb School of Medicine, Croatian Institute for Brain
Research, Zagreb, Croatia
| | - Siniša Škokić
- University of Zagreb School of Medicine, Croatian Institute for Brain
Research, Zagreb, Croatia
| | - Michal Babič
- Institute of Macromolecular Chemistry, Academy of Sciences, Prague, Czech
Republic
| | - Daniel Horák
- Institute of Macromolecular Chemistry, Academy of Sciences, Prague, Czech
Republic
| | - Mathias Hoehn
- Max Planck Institute for Metabolism Research, In-vivo-NMR Laboratory,
Cologne, Germany
| | - Srećko Gajović
- University of Zagreb School of Medicine, Croatian Institute for Brain
Research, Zagreb, Croatia
| |
Collapse
|
17
|
Vogel S, Schäfer C, Hess S, Folz-Donahue K, Nelles M, Minassian A, Schwarz MK, Kukat C, Ehrlich M, Zaehres H, Kloppenburg P, Hoehn M, Aswendt M. The in vivo timeline of differentiation of engrafted human neural progenitor cells. Stem Cell Res 2019; 37:101429. [DOI: 10.1016/j.scr.2019.101429] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/18/2019] [Accepted: 03/22/2019] [Indexed: 01/19/2023] Open
|
18
|
Green C, Minassian A, Vogel S, Diedenhofen M, Wiedermann D, Hoehn M. Persistent Quantitative Vitality of Stem Cell Graft Is Necessary for Stabilization of Functional Brain Networks After Stroke. Front Neurol 2019; 10:335. [PMID: 31024429 PMCID: PMC6460358 DOI: 10.3389/fneur.2019.00335] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 03/19/2019] [Indexed: 11/13/2022] Open
Abstract
Stem cell treatment after stroke has demonstrated substantial outcome improvement. However, monitoring of stem cell fate in vivo is still challenging and not routinely performed, yet important to quantify the role of the implanted stem cells on lesion improvement; in several studies even mortality of the graft has been reported. Resting state functional magnetic resonance imaging (rs-fMRI) is a highly sensitive imaging modality to monitor the brain-wide functional network alterations of many brain diseases in vivo. We monitor for 3 months the functional connectivity changes after intracortical stem cell engraftment in large, cortico-striatal (n = 9), and in small, striatal (n = 6) ischemic lesions in the mouse brain with non-invasive rs-fMRI on a 9.4T preclinical MRi scanner with GE-EPI sequence. Graft vitality is continuously recorded by bioluminescence imaging (BLI) roughly every 2 weeks after implantation of 300 k neural stem cells. In cortico-striatal lesions, the lesion extension induces graft vitality loss, in consequence leading to a parallel decrease of functional connectivity strength after a few weeks. In small, striatal lesions, the graft vitality is preserved for the whole observation period and the functional connectivity is stabilized at values as in the pre-stroke situation. But even here, at the end of the observation period of 3 months, the functional connectivity strength is found to decrease despite preserved graft vitality. We conclude that quantitative graft viability is a necessary but not sufficient criterion for functional neuronal network stabilization after stroke. Future studies with even longer time periods after stroke induction will need to identify additional players which have negative influence on the functional brain networks.
Collapse
Affiliation(s)
- Claudia Green
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Anuka Minassian
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Stefanie Vogel
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Michael Diedenhofen
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Dirk Wiedermann
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Mathias Hoehn
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany.,Department of Radiology, Leiden University Medical Center, Leiden, Netherlands.,Percuros B.V., Enschede, Netherlands
| |
Collapse
|
19
|
Aswendt M, Vogel S, Schäfer C, Jathoul A, Pule M, Hoehn M. Quantitative in vivo dual-color bioluminescence imaging in the mouse brain. NEUROPHOTONICS 2019; 6:025006. [PMID: 31093514 PMCID: PMC6504011 DOI: 10.1117/1.nph.6.2.025006] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 04/15/2019] [Indexed: 05/03/2023]
Abstract
Bioluminescence imaging (BLI) is an optical imaging method that can be translated from the cell culture dish in vitro to cell tracking in small animal models in vivo. In contrast to the more widely used fluorescence imaging, which requires light excitation, in BLI the light is exclusively generated by the enzyme luciferase. The luciferase gene can be engineered to target and monitor almost every cell and biological process quantitatively in vitro and even from deep tissue in vivo. While initially used for tumor imaging, bioluminescence was recently optimized for mouse brain imaging of neural cells and monitoring of viability or differentiation of grafted stem cells. Here, we describe the use of bright color-shifted firefly luciferases (Flucs) based on the thermostable x5 Fluc that emit red and green for effective and quantitative unmixing of two human cell populations in vitro and after transplantation into the mouse brain in vivo. Spectral unmixing predicts the ratio of luciferases in vitro and a mixture of cells precisely for cortical grafts, however, with less accuracy for striatal grafts. This dual-color approach enables the simultaneous visualization and quantification of two cell populations on the whole brain scale, with particular relevance for translational studies of neurological disorders providing information on stem cell survival and differentiation in one imaging session in vivo.
Collapse
Affiliation(s)
- Markus Aswendt
- Max Planck Institute for Metabolism Research, In-vivo-NMR Laboratory, Cologne, Germany
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, Cologne, Germany
- Address all correspondence to Markus Aswendt, E-mail:
| | - Stefanie Vogel
- Max Planck Institute for Metabolism Research, In-vivo-NMR Laboratory, Cologne, Germany
| | - Cordula Schäfer
- Max Planck Institute for Metabolism Research, In-vivo-NMR Laboratory, Cologne, Germany
| | - Amit Jathoul
- Cardiff School of Biosciences, Molecular Biosciences, Cardiff, United Kingdom
| | - Martin Pule
- University College London, Cancer Institute, Department of Haematology, London, United Kingdom
| | - Mathias Hoehn
- Max Planck Institute for Metabolism Research, In-vivo-NMR Laboratory, Cologne, Germany
- Leiden University Medical Center, Department of Radiology, Leiden, The Netherlands
| |
Collapse
|
20
|
Crane AT, Voth JP, Shen FX, Low WC. Concise Review: Human-Animal Neurological Chimeras: Humanized Animals or Human Cells in an Animal? Stem Cells 2019; 37:444-452. [PMID: 30629789 DOI: 10.1002/stem.2971] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/16/2018] [Accepted: 12/03/2018] [Indexed: 12/24/2022]
Abstract
Blastocyst complementation is an emerging methodology in which human stem cells are transferred into genetically engineered preimplantation animal embryos eventually giving rise to fully developed human tissues and organs within the animal host for use in regenerative medicine. The ethical issues surrounding this method have caused the National Institutes of Health to issue a moratorium on funding for blastocyst complementation citing the potential for human cells to substantially contribute to the brain of the chimeric animal. To address this concern, we performed an in-depth review of the neural transplantation literature to determine how the integration of human cells into the nonhuman neural circuitry has altered the behavior of the host. Despite reports of widespread integration of human cell transplants, our review of 150 transplantation studies found no evidence suggestive of humanization of the animal host, and we thus conclude that, at present, concerns over humanization should not prevent research on blastocyst complementation to continue. We suggest proceeding in a controlled and transparent manner, however, and include recommendations for future research with careful consideration for how human cells may contribute to the animal host nervous system. Stem Cells 2019;37:444-452.
Collapse
Affiliation(s)
- Andrew T Crane
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, USA.,Minnesota Craniofacial Research Training Program, University of Minnesota, Minneapolis, Minnesota, USA
| | - Joseph P Voth
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, USA.,Department of Neurosurgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Francis X Shen
- University of Minnesota Law School, Minneapolis, Minnesota, USA.,Graduate Program in Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA
| | - Walter C Low
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, USA.,Department of Neurosurgery, University of Minnesota, Minneapolis, Minnesota, USA.,Graduate Program in Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
21
|
Kassi AAY, Mahavadi AK, Clavijo A, Caliz D, Lee SW, Ahmed AI, Yokobori S, Hu Z, Spurlock MS, Wasserman JM, Rivera KN, Nodal S, Powell HR, Di L, Torres R, Leung LY, Rubiano AM, Bullock RM, Gajavelli S. Enduring Neuroprotective Effect of Subacute Neural Stem Cell Transplantation After Penetrating TBI. Front Neurol 2019; 9:1097. [PMID: 30719019 PMCID: PMC6348935 DOI: 10.3389/fneur.2018.01097] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 12/03/2018] [Indexed: 12/13/2022] Open
Abstract
Traumatic brain injury (TBI) is the largest cause of death and disability of persons under 45 years old, worldwide. Independent of the distribution, outcomes such as disability are associated with huge societal costs. The heterogeneity of TBI and its complicated biological response have helped clarify the limitations of current pharmacological approaches to TBI management. Five decades of effort have made some strides in reducing TBI mortality but little progress has been made to mitigate TBI-induced disability. Lessons learned from the failure of numerous randomized clinical trials and the inability to scale up results from single center clinical trials with neuroprotective agents led to the formation of organizations such as the Neurological Emergencies Treatment Trials (NETT) Network, and international collaborative comparative effectiveness research (CER) to re-orient TBI clinical research. With initiatives such as TRACK-TBI, generating rich and comprehensive human datasets with demographic, clinical, genomic, proteomic, imaging, and detailed outcome data across multiple time points has become the focus of the field in the United States (US). In addition, government institutions such as the US Department of Defense are investing in groups such as Operation Brain Trauma Therapy (OBTT), a multicenter, pre-clinical drug-screening consortium to address the barriers in translation. The consensus from such efforts including "The Lancet Neurology Commission" and current literature is that unmitigated cell death processes, incomplete debris clearance, aberrant neurotoxic immune, and glia cell response induce progressive tissue loss and spatiotemporal magnification of primary TBI. Our analysis suggests that the focus of neuroprotection research needs to shift from protecting dying and injured neurons at acute time points to modulating the aberrant glial response in sub-acute and chronic time points. One unexpected agent with neuroprotective properties that shows promise is transplantation of neural stem cells. In this review we present (i) a short survey of TBI epidemiology and summary of current care, (ii) findings of past neuroprotective clinical trials and possible reasons for failure based upon insights from human and preclinical TBI pathophysiology studies, including our group's inflammation-centered approach, (iii) the unmet need of TBI and unproven treatments and lastly, (iv) present evidence to support the rationale for sub-acute neural stem cell therapy to mediate enduring neuroprotection.
Collapse
Affiliation(s)
- Anelia A. Y. Kassi
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Anil K. Mahavadi
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Angelica Clavijo
- Neurosurgery Service, INUB-MEDITECH Research Group, El Bosque University, Bogotá, CO, United States
| | - Daniela Caliz
- Neurosurgery Service, INUB-MEDITECH Research Group, El Bosque University, Bogotá, CO, United States
| | - Stephanie W. Lee
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Aminul I. Ahmed
- Wessex Neurological Centre, University Hospitals Southampton, Southampton, United Kingdom
| | - Shoji Yokobori
- Department of Emergency and Critical Care Medicine, Nippon Medical School, Tokyo, Japan
| | - Zhen Hu
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Markus S. Spurlock
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Joseph M Wasserman
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Karla N. Rivera
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Samuel Nodal
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Henry R. Powell
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Long Di
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Rolando Torres
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Lai Yee Leung
- Branch of Brain Trauma Neuroprotection and Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Andres Mariano Rubiano
- Neurosurgery Service, INUB-MEDITECH Research Group, El Bosque University, Bogotá, CO, United States
| | - Ross M. Bullock
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Shyam Gajavelli
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
22
|
Mezzanotte L, Iljas JD, Que I, Chan A, Kaijzel E, Hoeben R, Löwik C. Optimized Longitudinal Monitoring of Stem Cell Grafts in Mouse Brain Using a Novel Bioluminescent/Near Infrared Fluorescent Fusion Reporter. Cell Transplant 2018; 26:1878-1889. [PMID: 29390874 PMCID: PMC5802635 DOI: 10.1177/0963689717739718] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Biodistribution and fate of transplanted stem cells via longitudinal monitoring has been successfully achieved in the last decade using optical imaging. However, sensitive longitudinal imaging of transplanted stem cells in deep tissue like the brain remains challenging not only due to low light penetration but because of other factors such as low or inferior expression levels of optical reporters in stem cells and stem cell death after transplantation. Here we describe an optimized imaging protocol for sensitive long-term monitoring of bone marrow-derived human mesenchymal stem cells (hMSCs) expressing a novel bioluminescent/near infrared fluorescent (NIRF) fusion reporter transplanted in mouse brain cortex. Lentivirus expressing the luc2-iRFP720 reporter, a fusion between luc2 codon-optimized firefly luciferase (luc2) and the gene encoding NIRF protein iRFP720, was generated to transduce hMSCs. These cells were analyzed for their fluorescent and bioluminescent emission and checked for their differentiation potential. In vivo experiments were performed by transplanting decreasing amounts of luc2-iRFP720 expressing hMSCs in mouse brain, followed by fluorescence and bioluminescence imaging (BLI) starting 1 wk after cell injection when the blood–brain barrier was restored. Bioluminescent images were acquired when signals peaked and used to compare different luc2 substrate performances, that is, D-luciferin (D-Luc; 25 μM/kg or 943 μM/kg) or CycLuc1 (25 μM/kg). Results showed that luc2-iRFP720 expressing hMSCs maintained a good in vitro differentiation potential toward adipocytes, chondrocytes, and osteocytes, suggesting that lentiviral transduction did not affect cell behavior. Moreover, in vivo experiments allowed us to image as low as 1 × 105 cells using both fluorescence and BLI. The highest bioluminescent signals (∼1 × 107 photons per second) were achieved 15 min after the injection of D-Luc (943 μM/kg). This allowed us to monitor as low as 1 × 105 hMSCs for the subsequent 7 wk without a significant drop in bioluminescent signals, suggesting the sustained viability of hMSCs transplanted into the cortex.
Collapse
Affiliation(s)
- Laura Mezzanotte
- 1 Department of Radiology, Optical Molecular Imaging, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Juvita Delancy Iljas
- 2 Percuros BV, Enschede, the Netherlands.,3 Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Ivo Que
- 4 Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Alan Chan
- 2 Percuros BV, Enschede, the Netherlands
| | - Eric Kaijzel
- 4 Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Rob Hoeben
- 5 Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Clemens Löwik
- 1 Department of Radiology, Optical Molecular Imaging, Erasmus Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
23
|
Collmann FM, Pijnenburg R, Schneider G, Schäfer C, Folz-Donahue K, Kukat C, Hoehn M. Imaging Reporter Strategy to Monitor Gene Activation of Microglia Polarisation States under Stimulation. J Neuroimmune Pharmacol 2018; 13:371-382. [PMID: 29790106 PMCID: PMC6096558 DOI: 10.1007/s11481-018-9789-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 05/06/2018] [Indexed: 12/24/2022]
Abstract
Microglial cells as innate immune key players have a critical and unique role in neurodegenerative disorders. They strongly interact with their microenvironment in a complex manner and react to changes by switching their phenotype and functional activation states. In order to understand the development of brain diseases, it is imperative to elucidate up- or down-regulation of genes involved in microglia polarisation in time-profile by a simple-to-use strategy. Here, we present a new imaging strategy to follow promoter activity of genes involved in microglia polarisation. We lentivirally transduced BV-2 microglia cells in culture with constructs consisting of the induced nitric oxide synthase (iNOS), Fc gamma receptor III (Fcgr3) (both resembling the pro-inflammatory M1-like phenotype) or Chitinase-like 3 (Chil3/Ym1) (resembling the anti-inflammatory M2-like phenotype) promoters and stimulated transgenic cells with potent activators for pro- or anti-inflammatory response, such as lipopolysaccharide (LPS) + interferon gamma (IFN-γ) or interleukin (IL)-4, respectively. Promoter activities upon polarisation phases were quantitatively assessed by the two imaging reporters Luc2 for bioluminescence and eGFP for fluorescence.
Collapse
Affiliation(s)
- Franziska M Collmann
- Max Planck Institute for Metabolism Research, In-vivo-NMR Laboratory, Gleueler Straße 50, D-50931, Cologne, Germany
| | - Rory Pijnenburg
- Max Planck Institute for Metabolism Research, In-vivo-NMR Laboratory, Gleueler Straße 50, D-50931, Cologne, Germany
| | - Gabriele Schneider
- Max Planck Institute for Metabolism Research, In-vivo-NMR Laboratory, Gleueler Straße 50, D-50931, Cologne, Germany
| | - Cordula Schäfer
- Max Planck Institute for Metabolism Research, In-vivo-NMR Laboratory, Gleueler Straße 50, D-50931, Cologne, Germany
| | - Kat Folz-Donahue
- Max Planck Institute for Biology of Ageing, FACS & Imaging Core Facility, Cologne, Germany
| | - Christian Kukat
- Max Planck Institute for Biology of Ageing, FACS & Imaging Core Facility, Cologne, Germany
| | - Mathias Hoehn
- Max Planck Institute for Metabolism Research, In-vivo-NMR Laboratory, Gleueler Straße 50, D-50931, Cologne, Germany. .,Leiden University Medical Center, Radiology, Leiden, The Netherlands. .,PERCUROS, Enschede, The Netherlands.
| |
Collapse
|
24
|
Lin GQ, He XF, Liang FY, Guo Y, Sunnassee G, Chen J, Cao XM, Chen YY, Pan GJ, Pei Z, Tan S. Transplanted human neural precursor cells integrate into the host neural circuit and ameliorate neurological deficits in a mouse model of traumatic brain injury. Neurosci Lett 2018; 674:11-17. [PMID: 29501684 DOI: 10.1016/j.neulet.2018.02.064] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 02/27/2018] [Accepted: 02/28/2018] [Indexed: 12/23/2022]
Abstract
Traumatic brain injury (TBI) is to date one of the major critical conditions causing death and disability worldwide. Exogenous neural stem/precursor cells (NSCs/NPCs) hold great promise for improving neurological dysfunction, but their functional properties in vivo remain unknown. Human neural precursor cells (hNPCs) carrying one fluorescent reporter gene (DsRed) can be observed directly in vivo using two-photon laser-scanning microscope. Therefore, we evaluated the neural integration and potential therapeutic effect of hNPCs on mice with TBI. Behavioral tests were performed by rotarod task and Morris Water Maze task. Neural integration was detected by fluorometric Ca2+ imaging and nerve tracing. We found that motor and cognition functions were significantly improved in mice with hNPCs injection compared to mice with vehicle treatment, and hNPCs integrated into the host circuit and differentiated toward neuronal lineage. Our study provided reliable evidence for further hNPCs transplantation in clinical practice.
Collapse
Affiliation(s)
- Gui-Qing Lin
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China; The Cadre Ward in Department of Neurology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Xiao-Fei He
- Department of Neurology, The First Affiliated Hospital, SunYat-sen University, Guangzhou, China
| | - Feng-Yin Liang
- Department of Neurology, The First Affiliated Hospital, SunYat-sen University, Guangzhou, China
| | - Yang Guo
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Gavin Sunnassee
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jian Chen
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiao-Min Cao
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yi-Yi Chen
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Guang-Jin Pan
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zhong Pei
- Department of Neurology, The First Affiliated Hospital, SunYat-sen University, Guangzhou, China.
| | - Sheng Tan
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
25
|
Sensorimotor Functional and Structural Networks after Intracerebral Stem Cell Grafts in the Ischemic Mouse Brain. J Neurosci 2018; 38:1648-1661. [PMID: 29321138 DOI: 10.1523/jneurosci.2715-17.2018] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 11/20/2017] [Accepted: 01/02/2018] [Indexed: 01/28/2023] Open
Abstract
Past investigations on stem cell-mediated recovery after stroke have limited their focus on the extent and morphological development of the ischemic lesion itself over time or on the integration capacity of the stem cell graft ex vivo However, an assessment of the long-term functional and structural improvement in vivo is essential to reliably quantify the regenerative capacity of cell implantation after stroke. We induced ischemic stroke in nude mice and implanted human neural stem cells (H9 derived) into the ipsilateral cortex in the acute phase. Functional and structural connectivity changes of the sensorimotor network were noninvasively monitored using magnetic resonance imaging for 3 months after stem cell implantation. A sharp decrease of the functional sensorimotor network extended even to the contralateral hemisphere, persisting for the whole 12 weeks of observation. In mice with stem cell implantation, functional networks were stabilized early on, pointing to a paracrine effect as an early supportive mechanism of the graft. This stabilization required the persistent vitality of the stem cells, monitored by bioluminescence imaging. Thus, we also observed deterioration of the early network stabilization upon vitality loss of the graft after a few weeks. Structural connectivity analysis showed fiber-density increases between the cortex and white matter regions occurring predominantly on the ischemic hemisphere. These fiber-density changes were nearly the same for both study groups. This motivated us to hypothesize that the stem cells can influence, via early paracrine effect, the functional networks, while observed structural changes are mainly stimulated by the ischemic event.SIGNIFICANCE STATEMENT In recent years, research on strokes has made a shift away from a focus on immediate ischemic effects and towards an emphasis on the long-range effects of the lesion on the whole brain. Outcome improvements in stem cell therapies also require the understanding of their influence on the whole-brain networks. Here, we have longitudinally and noninvasively monitored the structural and functional network alterations in the mouse model of focal cerebral ischemia. Structural changes of fiber-density increases are stimulated in the endogenous tissue without further modulation by the stem cells, while functional networks are stabilized by the stem cells via a paracrine effect. These results will help decipher the underlying networks of brain plasticity in response to cerebral lesions and offer clues to unravelling the mystery of how stem cells mediate regeneration.
Collapse
|
26
|
Schomann T, Mezzanotte L, De Groot JCMJ, Rivolta MN, Hendriks SH, Frijns JHM, Huisman MA. Neuronal differentiation of hair-follicle-bulge-derived stem cells co-cultured with mouse cochlear modiolus explants. PLoS One 2017; 12:e0187183. [PMID: 29084289 PMCID: PMC5662184 DOI: 10.1371/journal.pone.0187183] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 10/16/2017] [Indexed: 11/18/2022] Open
Abstract
Stem-cell-based repair of auditory neurons may represent an attractive therapeutic option to restore sensorineural hearing loss. Hair-follicle-bulge-derived stem cells (HFBSCs) are promising candidates for this type of therapy, because they (1) have migratory properties, enabling migration after transplantation, (2) can differentiate into sensory neurons and glial cells, and (3) can easily be harvested in relatively high numbers. However, HFBSCs have never been used for this purpose. We hypothesized that HFBSCs can be used for cell-based repair of the auditory nerve and we have examined their migration and incorporation into cochlear modiolus explants and their subsequent differentiation. Modiolus explants obtained from adult wild-type mice were cultured in the presence of EF1α-copGFP-transduced HFBSCs, constitutively expressing copepod green fluorescent protein (copGFP). Also, modiolus explants without hair cells were co-cultured with DCX-copGFP-transduced HFBSCs, which demonstrate copGFP upon doublecortin expression during neuronal differentiation. Velocity of HFBSC migration towards modiolus explants was calculated, and after two weeks, co-cultures were fixed and processed for immunohistochemical staining. EF1α-copGFP HFBSC migration velocity was fast: 80.5 ± 6.1 μm/h. After arrival in the explant, the cells formed a fascicular pattern and changed their phenotype into an ATOH1-positive neuronal cell type. DCX-copGFP HFBSCs became green-fluorescent after integration into the explants, confirming neuronal differentiation of the cells. These results show that HFBSC-derived neuronal progenitors are migratory and can integrate into cochlear modiolus explants, while adapting their phenotype depending on this micro-environment. Thus, HFBSCs show potential to be employed in cell-based therapies for auditory nerve repair.
Collapse
Affiliation(s)
- Timo Schomann
- Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, Leiden, South Holland, the Netherlands
| | - Laura Mezzanotte
- Optical Molecular Imaging Group, Department of Radiology, Erasmus Medical Center, Rotterdam, South Holland, the Netherlands
| | - John C. M. J. De Groot
- Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, Leiden, South Holland, the Netherlands
| | - Marcelo N. Rivolta
- Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Sheffield, England, United Kingdom
| | - Sanne H. Hendriks
- Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, Leiden, South Holland, the Netherlands
| | - Johan H. M. Frijns
- Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, Leiden, South Holland, the Netherlands
| | - Margriet A. Huisman
- Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, Leiden, South Holland, the Netherlands
- * E-mail:
| |
Collapse
|
27
|
Vogel S, Aswendt M, Nelles M, Henn N, Schneider G, Hoehn M. Initial graft size and not the innate immune response limit survival of engrafted neural stem cells. J Tissue Eng Regen Med 2017; 12:784-793. [DOI: 10.1002/term.2497] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 04/27/2017] [Accepted: 06/01/2017] [Indexed: 12/21/2022]
Affiliation(s)
- Stefanie Vogel
- In‐vivo‐NMR LaboratoryMax Planck Institute for Metabolism Research Cologne Germany
| | - Markus Aswendt
- In‐vivo‐NMR LaboratoryMax Planck Institute for Metabolism Research Cologne Germany
| | - Melanie Nelles
- In‐vivo‐NMR LaboratoryMax Planck Institute for Metabolism Research Cologne Germany
| | - Nadine Henn
- In‐vivo‐NMR LaboratoryMax Planck Institute for Metabolism Research Cologne Germany
| | - Gabriele Schneider
- In‐vivo‐NMR LaboratoryMax Planck Institute for Metabolism Research Cologne Germany
| | - Mathias Hoehn
- In‐vivo‐NMR LaboratoryMax Planck Institute for Metabolism Research Cologne Germany
- Department of RadiologyLeiden University Medical Center Leiden The Netherlands
- Percuros B.V., Enschede The Netherlands
| |
Collapse
|
28
|
Scarfe L, Brillant N, Kumar JD, Ali N, Alrumayh A, Amali M, Barbellion S, Jones V, Niemeijer M, Potdevin S, Roussignol G, Vaganov A, Barbaric I, Barrow M, Burton NC, Connell J, Dazzi F, Edsbagge J, French NS, Holder J, Hutchinson C, Jones DR, Kalber T, Lovatt C, Lythgoe MF, Patel S, Patrick PS, Piner J, Reinhardt J, Ricci E, Sidaway J, Stacey GN, Starkey Lewis PJ, Sullivan G, Taylor A, Wilm B, Poptani H, Murray P, Goldring CEP, Park BK. Preclinical imaging methods for assessing the safety and efficacy of regenerative medicine therapies. NPJ Regen Med 2017; 2:28. [PMID: 29302362 PMCID: PMC5677988 DOI: 10.1038/s41536-017-0029-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 06/30/2017] [Accepted: 07/24/2017] [Indexed: 02/08/2023] Open
Abstract
Regenerative medicine therapies hold enormous potential for a variety of currently incurable conditions with high unmet clinical need. Most progress in this field to date has been achieved with cell-based regenerative medicine therapies, with over a thousand clinical trials performed up to 2015. However, lack of adequate safety and efficacy data is currently limiting wider uptake of these therapies. To facilitate clinical translation, non-invasive in vivo imaging technologies that enable careful evaluation and characterisation of the administered cells and their effects on host tissues are critically required to evaluate their safety and efficacy in relevant preclinical models. This article reviews the most common imaging technologies available and how they can be applied to regenerative medicine research. We cover details of how each technology works, which cell labels are most appropriate for different applications, and the value of multi-modal imaging approaches to gain a comprehensive understanding of the responses to cell therapy in vivo.
Collapse
Affiliation(s)
- Lauren Scarfe
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
- Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Nathalie Brillant
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
- Medical Research Council Centre for Drug Safety Science, University of Liverpool, Liverpool, UK
| | - J. Dinesh Kumar
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
| | - Noura Ali
- College of Health Science, University of Duhok, Duhok, Iraq
| | - Ahmed Alrumayh
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Mohammed Amali
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Stephane Barbellion
- Medical Research Council Centre for Drug Safety Science, University of Liverpool, Liverpool, UK
| | - Vendula Jones
- GlaxoSmithKline, David Jack Centre for Research and Development, Ware, UK
| | - Marije Niemeijer
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Sophie Potdevin
- SANOFI Research and Development, Disposition, Safety and Animal Research, Alfortville, France
| | - Gautier Roussignol
- SANOFI Research and Development, Disposition, Safety and Animal Research, Alfortville, France
| | - Anatoly Vaganov
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - Ivana Barbaric
- Department of Biomedical Science, University of Sheffield, Sheffield, UK
| | - Michael Barrow
- Department of Chemistry, University of Liverpool, Liverpool, UK
| | | | - John Connell
- Centre for Advanced Biomedical Imaging, University College London, London, UK
| | - Francesco Dazzi
- Department of Haemato-Oncology, King’s College London, London, UK
| | | | - Neil S. French
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Julie Holder
- Roslin Cells, University of Cambridge, Cambridge, UK
| | - Claire Hutchinson
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
- Medical Research Council Centre for Drug Safety Science, University of Liverpool, Liverpool, UK
| | - David R. Jones
- Medicines and Healthcare Products Regulatory Agency, London, UK
| | - Tammy Kalber
- Centre for Advanced Biomedical Imaging, University College London, London, UK
| | - Cerys Lovatt
- GlaxoSmithKline, David Jack Centre for Research and Development, Ware, UK
| | - Mark F. Lythgoe
- Centre for Advanced Biomedical Imaging, University College London, London, UK
| | - Sara Patel
- ReNeuron Ltd, Pencoed Business Park, Pencoed, Bridgend, UK
| | - P. Stephen Patrick
- Centre for Advanced Biomedical Imaging, University College London, London, UK
| | - Jacqueline Piner
- GlaxoSmithKline, Medicines Research Centre, Gunnels Wood Road, Stevenage, UK
| | | | - Emanuelle Ricci
- Institute of Veterinary Science, University of Liverpool, Liverpool, UK
| | | | - Glyn N. Stacey
- UK Stem Cell Bank, Division of Advanced Therapies, National Institute for Biological Standards Control, Medicines and Healthcare Products Regulatory Agency, London, UK
| | - Philip J. Starkey Lewis
- Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Gareth Sullivan
- Department of Biochemistry, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Norwegian Center for Stem Cell Research, Blindern, Oslo, Norway
- Institute of Immunology, Oslo University Hospital-Rikshospitalet, Nydalen, Oslo, Norway
- Hybrid Technology Hub—Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Blindern, Oslo, Norway
| | - Arthur Taylor
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
- Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Bettina Wilm
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
- Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Harish Poptani
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
- Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Patricia Murray
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
- Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Chris E. P. Goldring
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
- Medical Research Council Centre for Drug Safety Science, University of Liverpool, Liverpool, UK
| | - B. Kevin Park
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
- Medical Research Council Centre for Drug Safety Science, University of Liverpool, Liverpool, UK
| |
Collapse
|
29
|
Aswendt M, Collmann FM, Hoehn M. Neurobiological insights from bioluminescence imaging. Oncotarget 2017; 8:69198-69199. [PMID: 29050195 PMCID: PMC5642470 DOI: 10.18632/oncotarget.20302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Indexed: 11/25/2022] Open
Affiliation(s)
- Markus Aswendt
- Mathias Hoehn: In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany and Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Franziska Melanie Collmann
- Mathias Hoehn: In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany and Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mathias Hoehn
- Mathias Hoehn: In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany and Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
30
|
Spurlock MS, Ahmed AI, Rivera KN, Yokobori S, Lee SW, Sam PN, Shear DA, Hefferan MP, Hazel TG, Johe KK, Gajavelli S, Tortella FC, Bullock RM. Amelioration of Penetrating Ballistic-Like Brain Injury Induced Cognitive Deficits after Neuronal Differentiation of Transplanted Human Neural Stem Cells. J Neurotrauma 2017; 34:1981-1995. [PMID: 28249550 PMCID: PMC6913783 DOI: 10.1089/neu.2016.4602] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Penetrating traumatic brain injury (PTBI) is one of the major cause of death and disability worldwide. Previous studies with penetrating ballistic-like brain injury (PBBI), a PTBI rat model revealed widespread perilesional neurodegeneration, similar to that seen in humans following gunshot wound to the head, which is unmitigated by any available therapies to date. Therefore, we evaluated human neural stem cell (hNSC) engraftment to putatively exploit the potential of cell therapy that has been seen in other central nervous system injury models. Toward this objective, green fluorescent protein (GFP) labeled hNSC (400,000 per animal) were transplanted in immunosuppressed Sprague-Dawley (SD), Fisher, and athymic (ATN) PBBI rats 1 week after injury. Tacrolimus (3 mg/kg 2 days prior to transplantation, then 1 mg/kg/day), methylprednisolone (10 mg/kg on the day of transplant, 1 mg/kg/week thereafter), and mycophenolate mofetil (30 mg/kg/day) for 7 days following transplantation were used to confer immunosuppression. Engraftment in SD and ATN was comparable at 8 weeks post-transplantation. Evaluation of hNSC differentiation and distribution revealed increased neuronal differentiation of transplanted cells with time. At 16 weeks post-transplantation, neither cell proliferation nor glial lineage markers were detected. Transplanted cell morphology was similar to that of neighboring host neurons, and there was relatively little migration of cells from the peritransplant site. By 16 weeks, GFP-positive processes extended both rostrocaudally and bilaterally into parenchyma, spreading along host white matter tracts, traversing the internal capsule, and extending ∼13 mm caudally from transplantation site reaching into the brainstem. In a Morris water maze test at 8 weeks post-transplantation, animals with transplants had shorter latency to platform than vehicle-treated animals. However, weak injury-induced cognitive deficits in the control group at the delayed time point confounded benefits of durable engraftment and neuronal differentiation. Therefore, these results justify further studies to progress towards clinical translation of hNSC therapy for PTBI.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Deborah A. Shear
- Branch of Brain Trauma Neuroprotection and Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | | | | | | | | | - Frank C. Tortella
- Branch of Brain Trauma Neuroprotection and Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | | |
Collapse
|
31
|
Tao ZY, Gao P, Yan YH, Li HY, Song J, Yang JX. Osthole Enhances the Therapeutic Efficiency of Stem Cell Transplantation in Neuroendoscopy Caused Traumatic Brain Injury. Biol Pharm Bull 2017; 40:1043-1054. [DOI: 10.1248/bpb.b17-00072] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Zhen-yu Tao
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine
| | - Peng Gao
- The First Affiliated Hospital of Dalian Medical University
| | - Yu-hui Yan
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine
| | - Hong-yan Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine
| | - Jie Song
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine
| | - Jing-xian Yang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine
| |
Collapse
|
32
|
Shokrollahi N, Shahbazzadeh D, Pooshang-Bagheri K, Habibi-Anbouhi M, Jahanian-Najafabadi A, Behdani M. A Model to Study the Phenotypic Changes of Insect Cell Transfection by Copepod Super Green Fluorescent Protein (cop-GFP) in Baculovirus Expression System. IRANIAN BIOMEDICAL JOURNAL 2016; 20:182-6. [PMID: 26518237 PMCID: PMC4949983 DOI: 10.7508/ibj.2016.03.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Background: Baculovirus expression system is one of the most attractive and powerful eukaryotic expression systems for the production of recombinant proteins. The presence of a biomarker is required to monitor transfection efficiency or protein expression levels in insect cells. Methods: The aim of this study was to construct a baculovirus expression vector encoding a copepod super green fluorescent protein (copGFP). In this light, the resultant vector was constructed and used for transfection of Spodoptera frugiperda cells. Results: Expression of the copGFP protein in insect cells was confirmed by fluorescent microscopy and Western-blot analysis. Conclusion: The application of copGFP control bacmid can be considered as an appropriate control for insect cell transfection.
Collapse
Affiliation(s)
- Narjes Shokrollahi
- Biotechnology Research Center, Venom and Biotherapeutics Molecules Lab, Pasteur Institute of Iran, Tehran, Iran
| | - Delavar Shahbazzadeh
- Biotechnology Research Center, Venom and Biotherapeutics Molecules Lab, Pasteur Institute of Iran, Tehran, Iran
| | - Kamran Pooshang-Bagheri
- Biotechnology Research Center, Venom and Biotherapeutics Molecules Lab, Pasteur Institute of Iran, Tehran, Iran
| | | | - Ali Jahanian-Najafabadi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Isfahan University of Medical Sciences and Health Services, Isfahan, Iran
| | - Mahdi Behdani
- Biotechnology Research Center, Venom and Biotherapeutics Molecules Lab, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
33
|
Ortuño-Lizarán I, Vilariño-Feltrer G, Martínez-Ramos C, Pradas MM, Vallés-Lluch A. Influence of synthesis parameters on hyaluronic acid hydrogels intended as nerve conduits. Biofabrication 2016; 8:045011. [DOI: 10.1088/1758-5090/8/4/045011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
34
|
Arulmoli J, Wright HJ, Phan DTT, Sheth U, Que RA, Botten GA, Keating M, Botvinick EL, Pathak MM, Zarembinski TI, Yanni DS, Razorenova OV, Hughes CCW, Flanagan LA. Combination scaffolds of salmon fibrin, hyaluronic acid, and laminin for human neural stem cell and vascular tissue engineering. Acta Biomater 2016; 43:122-138. [PMID: 27475528 PMCID: PMC5386322 DOI: 10.1016/j.actbio.2016.07.043] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 06/29/2016] [Accepted: 07/26/2016] [Indexed: 12/13/2022]
Abstract
UNLABELLED Human neural stem/progenitor cells (hNSPCs) are good candidates for treating central nervous system (CNS) trauma since they secrete beneficial trophic factors and differentiate into mature CNS cells; however, many cells die after transplantation. This cell death can be ameliorated by inclusion of a biomaterial scaffold, making identification of optimal scaffolds for hNSPCs a critical research focus. We investigated the properties of fibrin-based scaffolds and their effects on hNSPCs and found that fibrin generated from salmon fibrinogen and thrombin stimulates greater hNSPC proliferation than mammalian fibrin. Fibrin scaffolds degrade over the course of a few days in vivo, so we sought to develop a novel scaffold that would retain the beneficial properties of fibrin but degrade more slowly to provide longer support for hNSPCs. We found combination scaffolds of salmon fibrin with interpenetrating networks (IPNs) of hyaluronic acid (HA) with and without laminin polymerize more effectively than fibrin alone and generate compliant hydrogels matching the physical properties of brain tissue. Furthermore, combination scaffolds support hNSPC proliferation and differentiation while significantly attenuating the cell-mediated degradation seen with fibrin alone. HNSPCs express two fibrinogen-binding integrins, αVβ1 and α5β1, and several laminin binding integrins (α7β1, α6β1, α3β1) that can mediate interaction with the scaffold. Lastly, to test the ability of scaffolds to support vascularization, we analyzed human cord blood-derived endothelial cells alone and in co-culture with hNSPCs and found enhanced vessel formation and complexity in co-cultures within combination scaffolds. Overall, combination scaffolds of fibrin, HA, and laminin are excellent biomaterials for hNSPCs. STATEMENT OF SIGNIFICANCE Interest has increased recently in the development of biomaterials as neural stem cell transplantation scaffolds to treat central nervous system (CNS) injury since scaffolds improve survival and integration of transplanted cells. We report here on a novel combination scaffold composed of fibrin, hyaluronic acid, and laminin to support human neural stem/progenitor cell (hNSPC) function. This combined biomaterial scaffold has appropriate physical properties for hNSPCs and the CNS, supports hNSPC proliferation and differentiation, and attenuates rapid cell-mediated scaffold degradation. The hNSPCs and scaffold components synergistically encourage new vessel formation from human endothelial cells. This work marks the first report of a combination scaffold supporting human neural and vascular cells to encourage vasculogenesis, and sets a benchmark for biomaterials to treat CNS injury.
Collapse
Affiliation(s)
- Janahan Arulmoli
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Heather J Wright
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Duc T T Phan
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Urmi Sheth
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Richard A Que
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA
| | - Giovanni A Botten
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mark Keating
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA
| | - Elliot L Botvinick
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA
| | - Medha M Pathak
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| | | | - Daniel S Yanni
- Disc Comfort, Inc., 351 Hospital Road, Suite 202, Newport Beach, CA 92663, USA
| | - Olga V Razorenova
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Christopher C W Hughes
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA; The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA
| | - Lisa A Flanagan
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Neurology, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
35
|
Yang C, Tian R, Liu T, Liu G. MRI Reporter Genes for Noninvasive Molecular Imaging. Molecules 2016; 21:580. [PMID: 27213309 PMCID: PMC6273230 DOI: 10.3390/molecules21050580] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 04/21/2016] [Accepted: 04/25/2016] [Indexed: 01/17/2023] Open
Abstract
Magnetic resonance imaging (MRI) is one of the most important imaging technologies used in clinical diagnosis. Reporter genes for MRI can be applied to accurately track the delivery of cell in cell therapy, evaluate the therapy effect of gene delivery, and monitor tissue/cell-specific microenvironments. Commonly used reporter genes for MRI usually include genes encoding the enzyme (e.g., tyrosinase and β-galactosidase), the receptor on the cells (e.g., transferrin receptor), and endogenous reporter genes (e.g., ferritin reporter gene). However, low sensitivity limits the application of MRI and reporter gene-based multimodal imaging strategies are common including optical imaging and radionuclide imaging. These can significantly improve diagnostic efficiency and accelerate the development of new therapies.
Collapse
Affiliation(s)
- Caixia Yang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Rui Tian
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Ting Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
36
|
Medda X, Mertens L, Versweyveld S, Diels A, Barnham L, Bretteville A, Buist A, Verheyen A, Royaux I, Ebneth A, Cabrera-Socorro A. Development of a Scalable, High-Throughput-Compatible Assay to Detect Tau Aggregates Using iPSC-Derived Cortical Neurons Maintained in a Three-Dimensional Culture Format. ACTA ACUST UNITED AC 2016; 21:804-15. [PMID: 26984927 DOI: 10.1177/1087057116638029] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 02/15/2016] [Indexed: 01/14/2023]
Abstract
Tau aggregation is the pathological hallmark that best correlates with the progression of Alzheimer's disease (AD). The presence of neurofibrillary tangles (NFTs), formed of hyperphosphorylated tau, leads to neuronal dysfunction and loss, and is directly associated with the cognitive decline observed in AD patients. The limited success in targeting β-amyloid pathologies has reinforced the hypothesis of blocking tau phosphorylation, aggregation, and/or spreading as alternative therapeutic entry points to treat AD. Identification of novel therapies requires disease-relevant and scalable assays capable of reproducing key features of the pathology in an in vitro setting. Here we use induced pluripotent stem cells (iPSCs) as a virtually unlimited source of human cortical neurons to develop a robust and scalable tau aggregation model compatible with high-throughput screening (HTS). We downscaled cell culture conditions to 384-well plate format and used Matrigel to introduce an extra physical protection against cell detachment that reduces shearing stress and better recapitulates pathological conditions. We complemented the assay with AlphaLISA technology for the detection of tau aggregates in a high-throughput-compatible format. The assay is reproducible across users and works with different commercially available iPSC lines, representing a highly translational tool for the identification of novel treatments against tauopathies, including AD.
Collapse
Affiliation(s)
- X Medda
- Janssen Research & Development, a division of Janssen Pharmaceutica N.V., Beerse, Belgium Bordeaux University, Bordeaux, France
| | - L Mertens
- Janssen Research & Development, a division of Janssen Pharmaceutica N.V., Beerse, Belgium
| | - S Versweyveld
- Janssen Research & Development, a division of Janssen Pharmaceutica N.V., Beerse, Belgium
| | - A Diels
- Janssen Research & Development, a division of Janssen Pharmaceutica N.V., Beerse, Belgium
| | - L Barnham
- Janssen Research & Development, a division of Janssen Pharmaceutica N.V., Beerse, Belgium
| | - A Bretteville
- Janssen Research & Development, a division of Janssen Pharmaceutica N.V., Beerse, Belgium
| | - A Buist
- Janssen Research & Development, a division of Janssen Pharmaceutica N.V., Beerse, Belgium
| | - A Verheyen
- Janssen Research & Development, a division of Janssen Pharmaceutica N.V., Beerse, Belgium
| | - I Royaux
- Janssen Research & Development, a division of Janssen Pharmaceutica N.V., Beerse, Belgium
| | - A Ebneth
- Janssen Research & Development, a division of Janssen Pharmaceutica N.V., Beerse, Belgium
| | - A Cabrera-Socorro
- Janssen Research & Development, a division of Janssen Pharmaceutica N.V., Beerse, Belgium
| |
Collapse
|
37
|
Tennstaedt A, Mastropietro A, Nelles M, Beyrau A, Hoehn M. In Vivo Fate Imaging of Intracerebral Stem Cell Grafts in Mouse Brain. PLoS One 2015; 10:e0144262. [PMID: 26641453 PMCID: PMC4671578 DOI: 10.1371/journal.pone.0144262] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 11/16/2015] [Indexed: 12/03/2022] Open
Abstract
We generated transgenic human neural stem cells (hNSCs) stably expressing the reporter genes Luciferase for bioluminescence imaging (BLI) and GFP for fluorescence imaging, for multimodal imaging investigations. These transgenic hNSCs were further labeled with a clinically approved perfluoropolyether to perform parallel 19F MRI studies. In vitro validation demonstrated normal cell proliferation and differentiation of the transgenic and additionally labeled hNSCs, closely the same as the wild type cell line, making them suitable for in vivo application. Labeled and unlabeled transgenic hNSCs were implanted into the striatum of mouse brain. The time profile of their cell fate after intracerebral grafting was monitored during nine days following implantation with our multimodal imaging approach, assessing both functional and anatomical condition. The 19F MRI demarcated the graft location and permitted to estimate the cell number in the graft. BLI showed a pronounce cell loss during this monitoring period, indicated by the decrease of the viability signal. The in vivo obtained cell fate results were further validated and confirmed by immunohistochemistry. We could show that the surviving cells of the graft continued to differentiate into early neurons, while the severe cell loss could be explained by an inflammatory reaction to the graft, showing the graft being surrounded by activated microglia and macrophages. These results are different from earlier cell survival studies of our group where we had implanted the identical cells into the same mouse strain but in the cortex and not in the striatum. The cortical transplanted cells did not show any loss in viability but only pronounced and continuous neuronal differentiation.
Collapse
Affiliation(s)
- Annette Tennstaedt
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Alfonso Mastropietro
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
- Scientific Direction Unit, IRCCS Foundation Neurological Institute “C. Besta”, Milan, Italy
- Politecnico di Milano, Department of Electronic Information and Bioengineering, Milan, Italy
| | - Melanie Nelles
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Andreas Beyrau
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Mathias Hoehn
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
- Department Radiology, Leiden University Medical Center, Leiden University, Leiden, Netherlands
- * E-mail:
| |
Collapse
|
38
|
Aswendt M, Henn N, Michalk S, Schneider G, Steiner MS, Bissa U, Dose C, Hoehn M. Novel bimodal iron oxide particles for efficient tracking of human neural stem cells in vivo. Nanomedicine (Lond) 2015; 10:2499-512. [PMID: 26296195 DOI: 10.2217/nnm.15.94] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
AIMS We validated novel bimodal iron oxide particles as substitute of ferumoxides for efficient labeling of human neural stem cells (NSCs). The dextrane-coated FeraTrack Direct (FTD)-Vio particles have additional far-red fluorophores for microscopic cell analysis. METHODS MR relaxometry, spectrophotometric iron determination and microscopy are used for characterization in vitro and in vivo. RESULTS Efficient uptake is not transfection agent-dependent. FTD-Vio594 labeling had no influence on viability, proliferation, migration and differentiation capacity. It allows MRI-based tracking of engrafted NSCs in mouse brain up to 11 days, complemented by bioluminescence imaging of firefly luciferase expressed by the engrafted cells. CONCLUSION Our results highlight the FTD-Vio594 particles as safe and sensitive substitute of ferumoxides for longitudinal tracking of NSCs in preclinical studies.
Collapse
Affiliation(s)
- Markus Aswendt
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, D-50931 Cologne, Germany
| | - Nadine Henn
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, D-50931 Cologne, Germany
| | - Stefanie Michalk
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, D-50931 Cologne, Germany
| | - Gabriele Schneider
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, D-50931 Cologne, Germany
| | - Mark-Steven Steiner
- Miltenyi Biotec GmbH, Friedrich-Ebert-Strasse 68, 51429 Bergisch Gladbach, Germany
| | - Ursula Bissa
- Miltenyi Biotec GmbH, Friedrich-Ebert-Strasse 68, 51429 Bergisch Gladbach, Germany
| | - Christian Dose
- Miltenyi Biotec GmbH, Friedrich-Ebert-Strasse 68, 51429 Bergisch Gladbach, Germany
| | - Mathias Hoehn
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, D-50931 Cologne, Germany.,Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands.,Percuros BV, Enschede, Drienerlolaan 5-Zuidhorst, 7522 NB Enschede, The Netherlands
| |
Collapse
|