1
|
Cheng Q, Ma X, Liu J, Feng X, Liu Y, Wang Y, Ni W, Song M. Pharmacological Inhibition of the Asparaginyl Endopeptidase (AEP) in an Alzheimer's Disease Model Improves the Survival and Efficacy of Transplanted Neural Stem Cells. Int J Mol Sci 2023; 24:ijms24097739. [PMID: 37175445 PMCID: PMC10178525 DOI: 10.3390/ijms24097739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/21/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Stem-cell-based therapy is very promising for Alzheimer's disease (AD), yet has not become a reality. A critical challenge is the transplantation microenvironment, which impacts the therapeutic effect of stem cells. In AD brains, amyloid-beta (Aβ) peptides and inflammatory cytokines continuously poison the tissue microenvironment, leading to low survival of grafted cells and restricted efficacy. It is necessary to create a growth-supporting microenvironment for transplanted cells. Recent advances in AD studies suggest that the asparaginyl endopeptidase (AEP) is a potential intervention target for modifying pathological changes. We here chose APP/PS1 mice as an AD model and employed pharmacological inhibition of the AEP for one month to improve the brain microenvironment. Thereafter, we transplanted neural stem cells (NSCs) into the hippocampus and maintained therapy for one more month. We found that inhibition of AEPs resulted in a significant decrease of Aβ, TNF-α, IL-6 and IL-1β in their brains. In AD mice receiving NSC transplantation alone, the survival of NSCs was at a low level, while in combination with AEP inhibition pre-treatment the survival rate of engrafted cells was doubled. Within the 2-month treatment period, implantation of NSCs plus pre-inhibition of the AEP significantly enhanced neural plasticity of the hippocampus and rescued cognitive impairment. Neither NSC transplantation alone nor AEP inhibition alone achieved significant efficacy. In conclusion, pharmacological inhibition of the AEP ameliorated brain microenvironment of AD mice, and thus improved the survival and therapeutic efficacy of transplanted stem cells.
Collapse
Affiliation(s)
- Qing Cheng
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Xiaoli Ma
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Jingjing Liu
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Xuemei Feng
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Yan Liu
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Yanxia Wang
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Wenwen Ni
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Mingke Song
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| |
Collapse
|
2
|
Puranik N, Arukha AP, Yadav SK, Yadav D, Jin JO. Exploring the Role of Stem Cell Therapy in Treating Neurodegenerative Diseases: Challenges and Current Perspectives. Curr Stem Cell Res Ther 2022; 17:113-125. [PMID: 35135462 DOI: 10.2174/1574888x16666210810103838] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 05/18/2021] [Accepted: 06/01/2021] [Indexed: 11/22/2022]
Abstract
:
Several human neurological disorders, such as Parkinson’s disease, Alzheimer’s disease,
amyotrophic lateral sclerosis, Huntington’s disease, spinal cord injury, multiple sclerosis, and brain
stroke, are caused by the injury to neurons or glial cells. The recent years have witnessed the successful
generation of neurons and glia cells driving efforts to develop stem-cell-based therapies for
patients to combat a broad spectrum of human neurological diseases. The inadequacy of suitable
cell types for cell replacement therapy in patients suffering from neurological disorders has hampered
the development of this promising therapeutic approach. Attempts are thus being made to reconstruct
viable neurons and glial cells from different stem cells, such as embryonic stem cells,
mesenchymal stem cells, and neural stem cells. Dedicated research to cultivate stem cell-based
brain transplantation therapies has been carried out. We aim at compiling the breakthroughs in the
field of stem cell-based therapy for the treatment of neurodegenerative maladies, emphasizing the
shortcomings faced, victories achieved, and the future prospects of the therapy in clinical settings.
Collapse
Affiliation(s)
- Nidhi Puranik
- Department of Biological Science, Bharathiar University, Coimbatore, Tamil Nadu-641046, India
| | - Ananta Prasad Arukha
- Comparative Diagnostic
and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville- 32608, U.S.A
| | - Shiv Kumar Yadav
- Department of Botany, Government Lal Bahadur Shastri PG college, Sironj, Vidisha, Madhya Pradesh, India
| | - Dhananjay Yadav
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 712-749, Korea
| | - Jun O. Jin
- Department
of Medical Biotechnology, Yeungnam University, Gyeongsan 712-749, Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea
| |
Collapse
|
3
|
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
|
4
|
Rezaie J, Mehranjani MS, Rahbarghazi R, Shariatzadeh MA. Angiogenic and Restorative Abilities of Human Mesenchymal Stem Cells Were Reduced Following Treatment With Serum From Diabetes Mellitus Type 2 Patients. J Cell Biochem 2017; 119:524-535. [PMID: 28608561 DOI: 10.1002/jcb.26211] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 06/12/2017] [Indexed: 12/13/2022]
Abstract
This experiment investigated the impact of serum from patients with type 2 diabetes mellitus on the angiogenic behavior of human mesenchymal stem cells in vitro. Changes in the level of Ang-1, Ang-2, cell migration, and trans-differentiation into pericytes and endothelial lineage were monitored after 7 days. The interaction of mesenchymal stem cells with endothelial cells were evaluated using surface plasmon resonance technique. Paracrine restorative effect of diabetic stem cells was tested on pancreatic β cells. Compared to data from FBS and normal serum, diabetic serum reduced the stem cell survival and chemotaxis toward VEGF and SDF-1α (P < 0.05). Diabetic condition were found to decline cell migration rate and the activity of MMP-2 and -9 (P < 0.05). The down-regulation of VEGFR-2 and CXCR-4 was observed with an increase in the level of miR-1-3p and miR-15b-5p at the same time. The paracrine angiogenic potential of diabetic stem cells was disturbed via the changes in the dynamic of Ang-1, Ang-2, and VEGF. Surface plasmon resonance analysis showed that diabetes could induce an aberrant increase in the interaction of stem cells with endothelial cells. After treatment with diabetic serum, the expression of VE-cadherin and NG2 and ability for uptake of Dil-Ac-LDL were reduced (P < 0.01). Conditioned media prepared from diabetic stem cells were unable to decrease fatty acid accumulation in β-cells (P < 0.05). The level of insulin secreted by β-cells was not affected after exposure to supernatant from diabetic or non-diabetic mesenchymal stem cells. Data suggest diabetes could decrease angiogenic and restorative effect of stem cells in vitro. J. Cell. Biochem. 119: 524-535, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jafar Rezaie
- Department of Biology, Faculty of Sciences, Arak University, Arak, Iran
| | | | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
5
|
Abstract
During vertebrate embryonic development, the spinal cord is formed by the neural derivatives of a neuromesodermal population that is specified at early stages of development and which develops in concert with the caudal regression of the primitive streak. Several processes related to spinal cord specification and maturation are coupled to this caudal extension including neurogenesis, ventral patterning and neural crest specification and all of them seem to be crucially regulated by Fibroblast Growth Factor (FGF) signaling, which is prominently active in the neuromesodermal region and transiently in its derivatives. Here we review the role of FGF signaling in those processes, trying to separate its different functions and highlighting the interactions with other signaling pathways. Finally, these early functions of FGF signaling in spinal cord development may underlay partly its ability to promote regeneration in the lesioned spinal cord as well as its action promoting specific fates in neural stem cell cultures that may be used for therapeutical purposes.
Collapse
Affiliation(s)
- Ruth Diez Del Corral
- Department of Cellular, Molecular and Developmental Neurobiology, Cajal Institute, Consejo Superior de Investigaciones CientíficasMadrid, Spain.,Champalimaud Research, Champalimaud Centre for the UnknownLisbon, Portugal
| | - Aixa V Morales
- Department of Cellular, Molecular and Developmental Neurobiology, Cajal Institute, Consejo Superior de Investigaciones CientíficasMadrid, Spain
| |
Collapse
|
6
|
Abdanipour A, Noori-Zadeh A, Mesbah-Namin SA, Bakhtiyari S, Nejatbakhsh R, Anarkooli IJ. Di-(2-ethylhexyl) Phthalate-Induced Hippocampus-Derived Neural Stem Cells Proliferation. CELL JOURNAL 2016; 19:166-172. [PMID: 28367427 PMCID: PMC5241513 DOI: 10.22074/cellj.2016.4862] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 06/08/2016] [Indexed: 12/13/2022]
Abstract
The brain and spinal cord have a limited capacity for self-repair under damaged conditions. One of the best options to overcome these limitations involves the use of phytochemicals as potential therapeutic agents. In this study, we have aimed to investigate the
effects of di-(2-ethylhexyl) phthalate (DEHP) on hippocampus-derived neural stem cells
(NSCs) proliferation to search phytochemical candidates for possible treatment of neurological diseases using endogenous capacity.
In this experimental study, neonatal rat hippocampus-derived NSCs were cultured and
treated with various concentrations of DEHP (0, 100, 200, 400 and 600 µM) and Cirsium
vulgare (C. vulgare) hydroethanolic extract (0, 200, 400, 600, 800 and 1000 µg/ml) for 48
hours under in vitro conditions. Cell proliferation rates and quantitative Sox2 gene expression were evaluated using MTT assay and real-time reverse transcription polymerase
chain reaction (RT-PCR).
We observed the highest average growth rate in the 400 µM DEHP and 800 µg/ml C.
vulgare extract treated groups. Sox2 expression in the DEHP-treated NSCs significantly
increased compared to the control group. Gas chromatography/mass spectrometry (GC/
MS) results demonstrated that the active ingredients that naturally occurred in the C. vulgare hydroethanolic extract were 2-ethyl-1-hexanamine, n-heptacosane, 1-cyclopentanecarboxylic acid, 1-heptadecanamine, 2,6-octadien-1-ol,2,6,10,14,18,22-tetracosahexaene, and DEHP. DEHP profoundly stimulated NSCs proliferation through Sox2 gene
overexpression.
These results provide and opportunity for further use of the C. vulgure phytochemicals for
prevention and/or treatment of neurological diseases via phytochemical mediated-proliferation of endogenous adult NSCs.
Collapse
Affiliation(s)
- Alireza Abdanipour
- Department of Anatomy, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Noori-Zadeh
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed Alireza Mesbah-Namin
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Salar Bakhtiyari
- Student Research Committee Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Reza Nejatbakhsh
- Department of Anatomy, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Iraj Jafari Anarkooli
- Department of Anatomy, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
7
|
Gholizadeh-Ghaleh Aziz S, Fathi E, Rahmati-Yamchi M, Akbarzadeh A, Fardyazar Z, Pashaiasl M. An update clinical application of amniotic fluid-derived stem cells (AFSCs) in cancer cell therapy and tissue engineering. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2016; 45:765-774. [DOI: 10.1080/21691401.2016.1216857] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Shiva Gholizadeh-Ghaleh Aziz
- Women?s Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Molecular Medicine, School of Advanced Medical Science, Tabriz University of Medical Science, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ezzatollah Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Iran
| | | | - Abolfazl Akbarzadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Fardyazar
- Women?s Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Molecular Medicine, School of Advanced Medical Science, Tabriz University of Medical Science, Tabriz, Iran
| | - Maryam Pashaiasl
- Women?s Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
8
|
Learning-induced synaptic potentiation in implanted neural precursor cell-derived neurons. Sci Rep 2015; 5:17796. [PMID: 26634434 PMCID: PMC4669478 DOI: 10.1038/srep17796] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 11/05/2015] [Indexed: 12/17/2022] Open
Abstract
Neuronal loss caused by neurodegenerative diseases, traumatic brain injury and stroke results in cognitive dysfunctioning. Implantation of neural stem/precursor cells (NPCs) can improve the brain function by replacing lost neurons. Proper synaptic integration following neuronal differentiation of implanted cells is believed to be a prerequisite for the functional recovery. In the present study, we characterized the functional properties of immortalized neural progenitor HiB5 cells implanted into the rat hippocampus with chemically induced lesion. The implanted HiB5 cells migrated toward CA1 pyramidal layer and differentiated into vGluT1-positive glutamatergic neurons with morphological and electrophysiological properties of endogenous CA1 pyramidal cells. Functional synaptic integration of HiB5 cell-derived neurons was also evidenced by immunohistochemical and electrophysiological data. Lesion-caused memory deficit was significantly recovered after the implantation when assessed by inhibitory avoidance (IA) learning. Remarkably, IA learning preferentially produced long-term potentiation (LTP) at the synapses onto HiB5 cell-derived neurons, which occluded paring protocol-induced LTP ex vivo. We conclude that the implanted HiB5 cell-derived neurons actively participate in learning process through LTP formation, thereby counteracting lesion-mediated memory impairment.
Collapse
|
9
|
Chen Q, Zhang M, Li Y, Xu D, Wang Y, Song A, Zhu B, Huang Y, Zheng JC. CXCR7 Mediates Neural Progenitor Cells Migration to CXCL12 Independent of CXCR4. Stem Cells 2015; 33:2574-85. [PMID: 25833331 DOI: 10.1002/stem.2022] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 03/02/2015] [Indexed: 12/26/2022]
Abstract
Neural progenitor cell (NPC) migration is an essential process for brain development, adult neurogenesis, and neuroregeneration after brain injury. Stromal cell-derived factor-1 (SDF-1, CXCL12) and its traditional receptor CXCR4 are well known to regulate NPC migration. However, the discovery of CXCR7, a newly identified CXCL12 receptor, adds to the dynamics of the existing CXCL12/CXCR4 pair. Antagonists for either CXCR4 or CXCR7 blocked CXCL12-mediated NPC migration in a transwell chemotaxis assay, suggesting that both receptors are required for CXCL12 action. We derived NPC cultures from Cxcr4 knockout (KO) mice and used transwell and stripe assays to determine the cell migration. NPCs derived from Cxcr4 KO mice polarized and migrated in response to CXCL12 gradient, suggesting that CXCR7 could serve as an independent migration receptor. Furthermore, Cxcr4 KO NPCs transplanted into the adult mouse striatum migrated in response to the adjacent injection of CXCL12, an effect that was blocked by a CXCR7 antagonist, suggesting that CXCR7 also mediates NPC migration in vivo. Molecular mechanism studies revealed that CXCR7 interact with Rac1 in the leading edge of the polarized NPCs in the absence of CXCR4. Both CXCR7 and Rac1 are required for extracellular signal-regulated kinases (ERK) 1/2 activation and subsequent NPC migration, indicating that CXCR7 could serve as a functional receptor in CXCL12-mediated NPC migration independent of CXCR4. Together these results reveal an essential role of CXCR7 for CXCL12-mediated NPC migration that will be important to understand neurogenesis during development and in adulthood.
Collapse
Affiliation(s)
- Qiang Chen
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Department of Pharmacology and Experimental Neuroscience and, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Min Zhang
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Yuju Li
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Department of Pharmacology and Experimental Neuroscience and, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Dongsheng Xu
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Department of Pharmacology and Experimental Neuroscience and, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Yi Wang
- Department of Pharmacology and Experimental Neuroscience and, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Aihong Song
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Bing Zhu
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Department of Pharmacology and Experimental Neuroscience and, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Yunlong Huang
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Department of Pharmacology and Experimental Neuroscience and, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Jialin C Zheng
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Department of Pharmacology and Experimental Neuroscience and, University of Nebraska Medical Center, Omaha, Nebraska, USA.,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
10
|
Torres EM, Trigano M, Dunnett TB. Translation of Cell Therapies to the Clinic: Characteristics of Cell Suspensions in Large-Diameter Injection Cannulae. Cell Transplant 2015; 24:737-49. [DOI: 10.3727/096368914x685429] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
With the use of cell replacement therapies as a realistic prospect for conditions such as Parkinson's and Huntington's diseases, the logistics of the delivery of cell suspensions to deep brain targets is a topic for consideration. Because of the large cannulae required for such procedures, we need to consider the behavior of cell suspensions within the cannulae if we are to ensure that the injected cells are distributed as intended within the target tissue. We have investigated the behavior of primary embryonic cell suspensions of neural tissue, in cannulae of different diameters, using a protocol designed to mimic the handling and injection of cells during clinical application. Internal cannula diameter had a large effect on the distribution of cells during their dispensation from the syringe. In vertical or near vertical cannulae, cells settled toward the tip of the needle, and were dispensed unevenly, with the majority of cells emerging in the first 10-20% of the injectate. In horizontal or near-horizontal cannulae, we observed the opposite effect, such that few cells were dispensed in the first 80% of the injectate, and the majority emerged in the final 10-20%. Use of a glass cannula showed that the results obtained using the horizontal cannula were caused by settling and adherence of the cells on the side of the cannulae, such that during dispensation, the overlying, cell-free solution was dispensed first, prior to the emergence of the cells. We show that the behavior of cells in such cannulae is affected by the cannula diameter, and by the material of the cannula itself. In horizontal cannulae, uneven expulsion of cells from the needle can be ameliorated by regular rotation of the cannula during the procedure. We discuss the potential impact of these observations on the translation of cell therapies to the clinic.
Collapse
|
11
|
Sakthiswary R, Raymond AA. Stem cell therapy in neurodegenerative diseases: From principles to practice. Neural Regen Res 2015; 7:1822-31. [PMID: 25624807 PMCID: PMC4302533 DOI: 10.3969/j.issn.1673-5374.2012.23.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2012] [Accepted: 06/13/2012] [Indexed: 12/11/2022] Open
Abstract
The lack of curative therapies for neurodegenerative diseases has high economic impact and places huge burden on the society. The contribution of stem cells to cure neurodegenerative diseases has been unraveled and explored extensively over the past few years. Beyond substitution of the lost neurons, stem cells act as immunomodulators and neuroprotectors. A large number of preclinical and a small number of clinical studies have shown beneficial outcomes in this context. In this review, we have summarized the current concepts of stem cell therapy in neurodegenerative diseases and the recent advances in this field, particularly between 2010 and 2012. Further studies should be encouraged to resolve the clinical issues and vague translational findings for maximum optimization of the efficacy of stem cell therapy in neurodegenerative diseases.
Collapse
Affiliation(s)
- Rajalingham Sakthiswary
- Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Bandar Tun Razak 56000, Kuala Lumpur, Malaysia
| | - Azman Ali Raymond
- Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Bandar Tun Razak 56000, Kuala Lumpur, Malaysia
| |
Collapse
|
12
|
Mesenchymal stromal cells enhance wound healing by ameliorating impaired metabolism in diabetic mice. Cytotherapy 2014; 16:1467-1475. [DOI: 10.1016/j.jcyt.2014.05.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 05/14/2014] [Accepted: 05/18/2014] [Indexed: 01/09/2023]
|
13
|
Abstract
Spinal cord injury is a complex pathology often resulting in functional impairment and paralysis. Gene therapy has emerged as a possible solution to the problems of limited neural tissue regeneration through the administration of factors promoting axonal growth, while also offering long-term local delivery of therapeutic molecules at the injury site. Of note, gene therapy is our response to the requirements of neural and glial cells following spinal cord injury, providing, in a time-dependent manner, growth substances for axonal regeneration and eliminating axonal growth inhibitors. Herein, we explore different gene therapy strategies, including targeting gene expression to modulate the presence of neurotrophic growth or survival factors and increase neural tissue plasticity. Special attention is given to describing advances in viral and non-viral gene delivery systems, as well as the available routes of gene delivery. Finally, we discuss the future of combinatorial gene therapies and give consideration to the implementation of gene therapy in humans.
Collapse
|
14
|
Faravelli I, Bucchia M, Rinchetti P, Nizzardo M, Simone C, Frattini E, Corti S. Motor neuron derivation from human embryonic and induced pluripotent stem cells: experimental approaches and clinical perspectives. Stem Cell Res Ther 2014; 5:87. [PMID: 25157556 PMCID: PMC4100331 DOI: 10.1186/scrt476] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Motor neurons are cells located in specific areas of the central nervous system, such as brain cortex (upper motor neurons), brain stem, and spinal cord (lower motor neurons), which maintain control over voluntary actions. Motor neurons are affected primarily by a wide spectrum of neurological disorders, generally indicated as motor neuron diseases (MNDs): these disorders share symptoms related to muscular atrophy and paralysis leading to death. No effective treatments are currently available. Stem cell-derived motor neurons represent a promising research tool in disease modeling, drug screening, and development of therapeutic approaches for MNDs and spinal cord injuries. Directed differentiation of human pluripotent stem cells - human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs) - toward specific lineages is the first crucial step in order to extensively employ these cells in early human development investigation and potential clinical applications. Induced pluripotent stem cells (iPSCs) can be generated from patients' own somatic cells (for example, fibroblasts) by reprogramming them with specific factors. They can be considered embryonic stem cell-like cells, which express stem cell markers and have the ability to give rise to all three germ layers, bypassing the ethical concerns. Thus, hiPSCs constitute an appealing alternative source of motor neurons. These motor neurons might be a great research tool, creating a model for investigating the cellular and molecular interactions underlying early human brain development and pathologies during neurodegeneration. Patient-specific iPSCs may also provide the premises for autologous cell replacement therapies without related risks of immune rejection. Here, we review the most recent reported methods by which hESCs or iPSCs can be differentiated toward functional motor neurons with an overview on the potential clinical applications.
Collapse
|
15
|
Biodegradable microparticles for strictly regulating the release of neurotrophic factors. J Control Release 2013; 168:307-16. [PMID: 23578846 DOI: 10.1016/j.jconrel.2013.03.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 03/26/2013] [Accepted: 03/30/2013] [Indexed: 10/27/2022]
Abstract
A lot of research has been carried out in the last decade to find a cure for neurodegenerative diseases especially Parkinson's disease but to little avail. In this study we have demonstrated the use of poly(lactic-co-glycolic acid) (PLGA)/collagen biodegradable microparticles formed using water-in-oil-in-water (W/O/W) double emulsion method, as a neurotrophic factor delivery vehicle. The microparticles were encapsulated with glial cell-derived neurotrophic factor (GDNF) fused with collagen binding peptide (CBP) immobilized to the inner collagen phase. The novelty lies in the strict regulation of release of GDNF-CBP from the microparticles as compared to a burst release from standard microparticles. The microparticles were demonstrated to be non-cytotoxic till 300 μg/2 × 10⁵ cells and revealed a maximum release of 250 ng GDNF-CBP/mg microparticles in 0.3% collagenase. Differentiation of neural progenitor cells (NPCs) into mature neurons was demonstrated by co-culturing microparticles with cells in a medium containing collagenase which enabled the release of encapsulated GDNF-CBP, signaling the differentiation of NPCs into microtubule-associated protein 2 (MAP2)-expressing neurons. The successful ability of these microparticles to deliver neurotrophic factors and allow differentiation of NPCs into mature neurons provides some scope in its use for the treatment of Parkinson's disease and other neurodegenerative diseases.
Collapse
|
16
|
Kim W, Kim JH, Kong SY, Park MH, Sohn UD, Kim HJ. Comparison of ectopic gene expression methods in rat neural stem cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2013; 17:23-30. [PMID: 23439859 PMCID: PMC3579101 DOI: 10.4196/kjpp.2013.17.1.23] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 11/19/2012] [Accepted: 11/29/2012] [Indexed: 12/21/2022]
Abstract
Neural stem cells (NSCs) have the ability to proliferate and differentiate into various types of cells that compose the nervous system. To study functions of genes in stem cell biology, genes or siRNAs need to be transfected. However, it is difficult to transfect ectopic genes into NSCs. Thus to identify the suitable method to achieve high transfection efficiency, we compared lipid transfection, electroporation, nucleofection and retroviral transduction. Among the methods that we tested, we found that nucleofection and retroviral transduction showed significantly increased transfection efficiency. In addition, with retroviral transduction of Ngn2 that is known to induce neurogenesis in various types of cells, we observed facilitated final cell division in rat NSCs. These data suggest that nucleofection and retroviral transduction provide high efficiency of gene delivery system to study functions of genes in rat NSCs.
Collapse
Affiliation(s)
- Woosuk Kim
- Laboratory of Stem Cell and Molecular Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 156-756, Korea
| | | | | | | | | | | |
Collapse
|
17
|
Hirth F. Stem Cells and Asymmetric Cell Division. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
18
|
De Vocht N, Lin D, Praet J, Hoornaert C, Reekmans K, Le Blon D, Daans J, Pauwels P, Goossens H, Hens N, Berneman Z, Van der Linden A, Ponsaerts P. Quantitative and phenotypic analysis of mesenchymal stromal cell graft survival and recognition by microglia and astrocytes in mouse brain. Immunobiology 2012; 218:696-705. [PMID: 22944251 DOI: 10.1016/j.imbio.2012.08.266] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Revised: 08/02/2012] [Accepted: 08/05/2012] [Indexed: 01/18/2023]
Abstract
Although cell transplantation is increasingly suggested to be beneficial for the treatment of various neurodegenerative diseases, the therapeutic application of such intervention is currently hindered by the limited knowledge regarding central nervous system (CNS) transplantation immunology. In this study, we aimed to investigate the early post transplantation innate immune events following grafting of autologous mesenchymal stromal cells (MSC) in the CNS of immune competent mice. First, the survival of grafted Luciferase/eGFP-expressing MSC (MSC-Luc/eGFP) was demonstrated to be stable from on day 3 post implantation using in vivo bioluminescence imaging (BLI), which was further confirmed by quantitative histological analysis of MSC-Luc/eGFP graft survival. Additional histological analyses at week 1 and week 2 post grafting revealed the appearance of (i) graft-surrounding/-invading Iba1+ microglia and (ii) graft-surrounding GFAP+ astrocytes, as compared to day 0 post grafting. While the density of graft-surrounding astrocytes and microglia did not change between week 1 and week 2 post grafting, the density of graft-invading microglia significantly decreased between week 1 and week 2 post implantation. However, despite the observed decrease in microglial density within the graft site, additional phenotypic analysis of graft-invading microglia, based on CD11b- and MHCII-expression, revealed >50% of graft-invading microglia at week 2 post implantation to display an activated status. Although microglial expression of CD11b and MHCII is already suggestive for a pro-inflammatory M1-oriented phenotype, the latter was further confirmed by: (i) the expression of NOS2 by microglia within the graft site, and (ii) the absence of arginase 1-expression, an enzyme known to suppress NO activity in M2-oriented microglia, on graft-surrounding and -invading microglia. In summary, we here provide a detailed phenotypic analysis of post transplantation innate immune events in the CNS of mice, and warrant that such intervention is associated with an M1-oriented microglia response and severe astrogliosis.
Collapse
Affiliation(s)
- Nathalie De Vocht
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Yi BR, Hwang KA, Kang NH, Kim SU, Jeung EB, Kim HC, Choi KC. Synergistic effects of genetically engineered stem cells expressing cytosine deaminase and interferon-β via their tumor tropism to selectively target human hepatocarcinoma cells. Cancer Gene Ther 2012; 19:644-51. [PMID: 22790964 DOI: 10.1038/cgt.2012.45] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Stem cells have received a great deal of attention for their clinical and therapeutic potential for treating human diseases and disorders. Recent studies have shown that it is possible to genetically engineered stem cells (GESTECs) to produce suicide enzymes that convert non-toxic prodrugs to toxic metabolites, selectively migrate toward tumor sites and reduce tumor growth. In this study, we evaluated whether these GESTECs are capable of migrating to hepatocarcinoma cells and examined the potential therapeutic efficacy of gene-directed enzyme prodrug therapy against liver cancer cells in cellular and animal models. A modified transwell migration assay was performed to determine the migratory capacity of GESTECs to Hep3B hepatocarcinoma cells. GESTECs, that is, HB1.F3.CD or HB1.F3.CD.interferon-β (IFN-β) cells, engineered to express a suicide gene, cytosine deaminase (CD), selectively migrated toward liver cancer cells. Treatment of Hep3B, human liver cancer cells, with the prodrug 5-fluorocytosine (5-FC) in the presence of HB1.F3.CD or HB1.F3.CD.IFN-β cells resulted in the inhibition of Hep3B cell growth. In a xenografted mouse model injected with hepatocarcinoma, we investigated the therapeutic effect of these stem cells. For 9 weeks, the xenografted mice were treated with HB1.F3.CD or HB1.F3.CD.IFN-β in the presence of 5-FC. A growth of tumor mass was inhibited about 40-50% in the mice treated with GESTECs and a prodrug. In addition, we further confirmed the cytotoxic effect on tumor cells by histological analysis and migratory effect of therapeutic stem cells. Taken together, GESTECs expressing a fusion gene encoding CD and IFN-β may exert a synergistic antitumor effect on this type of tumor.
Collapse
Affiliation(s)
- B-R Yi
- Laboratory of Veterinary Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
20
|
Si Y, Zhao Y, Hao H, Liu J, Guo Y, Mu Y, Shen J, Cheng Y, Fu X, Han W. Infusion of mesenchymal stem cells ameliorates hyperglycemia in type 2 diabetic rats: identification of a novel role in improving insulin sensitivity. Diabetes 2012; 61:1616-25. [PMID: 22618776 PMCID: PMC3357293 DOI: 10.2337/db11-1141] [Citation(s) in RCA: 202] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Infusion of mesenchymal stem cells (MSCs) has been shown to effectively lower blood glucose in diabetic individuals, but the mechanism involved could not be adequately explained by their potential role in promoting islet regeneration. We therefore hypothesized that infused MSCs might also contribute to amelioration of the insulin resistance of peripheral insulin target tissues. To test the hypothesis, we induced a diabetic rat model by high-fat diet/streptozotocin (STZ) administration, performed MSC infusion during the early phase (7 days) or late phase (21 days) after STZ injection, and then evaluated the therapeutic effects of MSC infusion and explored the possible mechanisms involved. MSC infusion ameliorated hyperglycemia in rats with type 2 diabetes (T2D). Infusion of MSCs during the early phase not only promoted β-cell function but also ameliorated insulin resistance, whereas infusion in the late phase merely ameliorated insulin resistance. Infusion of MSCs resulted in an increase of GLUT4 expression and an elevation of phosphorylated insulin receptor substrate 1 (IRS-1) and Akt (protein kinase B) in insulin target tissues. This is the first report of MSC treatment improving insulin sensitivity in T2D. These data indicate that multiple roles and mechanisms are involved in the efficacy of MSCs in ameliorating hyperglycemia in T2D.
Collapse
Affiliation(s)
- Yiling Si
- Institute of Basic Medicine, College of Life Science, Chinese PLA General Hospital, Beijing, China
| | - Yali Zhao
- Institute of Basic Medicine, College of Life Science, Chinese PLA General Hospital, Beijing, China
| | - Haojie Hao
- Institute of Basic Medicine, College of Life Science, Chinese PLA General Hospital, Beijing, China
| | - Jiejie Liu
- Institute of Basic Medicine, College of Life Science, Chinese PLA General Hospital, Beijing, China
| | - Yelei Guo
- Institute of Basic Medicine, College of Life Science, Chinese PLA General Hospital, Beijing, China
| | - Yiming Mu
- Department of Endocrinology, Chinese PLA General Hospital, Beijing, China
| | - Jing Shen
- Department of Endocrinology, Chinese PLA General Hospital, Beijing, China
| | - Yu Cheng
- Department of Endocrinology, Chinese PLA General Hospital, Beijing, China
| | - Xiaobing Fu
- Institute of Basic Medicine, College of Life Science, Chinese PLA General Hospital, Beijing, China
- Corresponding author: Weidong Han, , or Xiaobing Fu,
| | - Weidong Han
- Institute of Basic Medicine, College of Life Science, Chinese PLA General Hospital, Beijing, China
- Corresponding author: Weidong Han, , or Xiaobing Fu,
| |
Collapse
|
21
|
Wang TY, Forsythe JS, Parish CL, Nisbet DR. Biofunctionalisation of polymeric scaffolds for neural tissue engineering. J Biomater Appl 2012; 27:369-90. [DOI: 10.1177/0885328212443297] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Patients who experience injury to the central or peripheral nervous systems invariably suffer from a range of dysfunctions due to the limited ability for repair and reconstruction of damaged neural tissue. Whilst some treatment strategies can provide symptomatic improvement of motor and cognitive function, they fail to repair the injured circuits and rarely offer long-term disease modification. To this end, the biological molecules, used in combination with neural tissue engineering scaffolds, may provide feasible means to repair damaged neural pathways. This review will focus on three promising classes of neural tissue engineering scaffolds, namely hydrogels, electrospun nanofibres and self-assembling peptides. Additionally, the importance and methods for presenting biologically relevant molecules such as, neurotrophins, extracellular matrix proteins and protein-derived sequences that promote neuronal survival, proliferation and neurite outgrowth into the lesion will be discussed.
Collapse
Affiliation(s)
- TY Wang
- Department of Materials Engineering, Monash University, Victoria, Australia
| | - JS Forsythe
- Department of Materials Engineering, Monash University, Victoria, Australia
| | - CL Parish
- Florey Neuroscience Institute and Centre for Neuroscience, The University of Melbourne, Victoria, Australia
| | - DR Nisbet
- Research School of Engineering, College of Engineering and Computer Science, The Australian National University, Canberra, Australia
| |
Collapse
|
22
|
Praet J, Reekmans K, Lin D, De Vocht N, Bergwerf I, Tambuyzer B, Daans J, Hens N, Goossens H, Pauwels P, Berneman Z, Van der Linden A, Ponsaerts P. Cell type-associated differences in migration, survival, and immunogenicity following grafting in CNS tissue. Cell Transplant 2012; 21:1867-81. [PMID: 22472278 DOI: 10.3727/096368912x636920] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cell transplantation has been suggested to display several neuroprotective and/or neuroregenerative effects in animal models of central nervous system (CNS) trauma. However, while most studies report on clinical observations, currently little is known regarding the actual fate of the cell populations grafted and whether or how the brain's innate immune system, mainly directed by activated microglia and astrocytes, interacts with autologous cellular implants. In this study, we grafted well-characterized neural stem cell, mouse embryonic fibroblast, dendritic cell, bone marrow mononuclear cell, and splenocyte populations, all isolated or cultured from C57BL/6-eGFP transgenic mice, below the capsula externa (CE) of healthy C57BL/6 mice and below the inflamed/demyelinated CE of cuprizone-treated C57BL/6 mice. Two weeks postgrafting, an extensive quantitative multicolor histological analysis was performed in order (i) to quantify cell graft localization, migration, survival, and toxicity and (ii) to characterize endogenous CNS immune responses against the different cell grafts. Obtained results indicate dependence on the cell type grafted: (i) a different degree of cell graft migration, survival, and toxicity and (ii) a different organization of the endogenous immune response. Based on these observations, we warrant that further research should be undertaken to understand-and eventually control-cell graft-induced tissue damage and activation of the brain's innate immune system. The latter will be inevitable before cell grafting in the CNS can be performed safely and successfully in clinical settings.
Collapse
Affiliation(s)
- Jelle Praet
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
De Vocht N, Bergwerf I, Vanhoutte G, Daans J, De Visscher G, Chatterjee S, Pauwels P, Berneman Z, Ponsaerts P, Van der Linden A. Labeling of Luciferase/eGFP-expressing bone marrow-derived stromal cells with fluorescent micron-sized iron oxide particles improves quantitative and qualitative multimodal imaging of cellular grafts in vivo. Mol Imaging Biol 2012; 13:1133-45. [PMID: 21246293 DOI: 10.1007/s11307-011-0469-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
PURPOSE Development of multimodal imaging strategies is currently of utmost importance for the validation of preclinical stem cell therapy studies. PROCEDURES We performed a combined labeling strategy for bone marrow-derived stromal cells (BMSC) based on genetic modification with the reporter genes Luciferase and eGFP (BMSC-Luc/eGFP) and physical labeling with blue fluorescent micron-sized iron oxide particles (MPIO) in order to unambiguously identify BMSC localization, survival, and differentiation following engraftment in the central nervous system of mice by in vivo bioluminescence (BLI) and magnetic resonance imaging and postmortem histological analysis. RESULTS Using this combination, a significant increase of in vivo BLI signal was observed for MPIO-labeled BMSC-Luc/eGFP. Moreover, MPIO labeling of BMSC-Luc/eGFP allows for the improved identification of implanted cells within host tissue during histological observation. CONCLUSIONS This study describes an optimized labeling strategy for multimodal stem cell imaging resulting in improved quantitative and qualitative detection of cellular grafts.
Collapse
|
24
|
Decimo I, Bifari F, Rodriguez FJ, Malpeli G, Dolci S, Lavarini V, Pretto S, Vasquez S, Sciancalepore M, Montalbano A, Berton V, Krampera M, Fumagalli G. Nestin- and doublecortin-positive cells reside in adult spinal cord meninges and participate in injury-induced parenchymal reaction. Stem Cells 2012; 29:2062-76. [PMID: 22038821 PMCID: PMC3468739 DOI: 10.1002/stem.766] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Adult spinal cord has little regenerative potential, thus limiting patient recovery following injury. In this study, we describe a new population of cells resident in the adult rat spinal cord meninges that express the neural stem/precursor markers nestin and doublecortin. Furthermore, from dissociated meningeal tissue a neural stem cell population was cultured in vitro and subsequently shown to differentiate into functional neurons or mature oligodendrocytes. Proliferation rate and number of nestin- and doublecortin-positive cells increased in vivo in meninges following spinal cord injury. By using a lentivirus-labeling approach, we show that meningeal cells, including nestin- and doublecortin-positive cells, migrate in the spinal cord parenchyma and contribute to the glial scar formation. Our data emphasize the multiple roles of meninges in the reaction of the parenchyma to trauma and indicate for the first time that spinal cord meninges are potential niches harboring stem/precursor cells that can be activated by injury. Meninges may be considered as a new source of adult stem/precursor cells to be further tested for use in regenerative medicine applied to neurological disorders, including repair from spinal cord injury. Stem Cells 2011;29:2062–2076.
Collapse
Affiliation(s)
- Ilaria Decimo
- Department of Public Health and Community Medicine, Section of Pharmacology, University of Verona, Verona, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Kim KY, Yi BR, Lee HR, Kang NH, Jeung EB, Kim SU, Choi KC. Stem cells with fused gene expression of cytosine deaminase and interferon-β migrate to human gastric cancer cells and result in synergistic growth inhibition for potential therapeutic use. Int J Oncol 2011; 40:1097-104. [PMID: 22159640 PMCID: PMC3584621 DOI: 10.3892/ijo.2011.1288] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 11/09/2011] [Indexed: 01/14/2023] Open
Abstract
Genetically engineered stem cells (GESTECs) producing suicide enzymes and immunotherapeutic cytokines have therapeutic effects on tumors, and may possibly reduce the side effects of toxic drugs used for treatments. Suicide enzymes can convert non-toxic pro-drugs to toxic metabolites that can reduce tumor growth. Cytosine deaminase (CD) is a suicide enzyme that metabolizes a non-toxic pro-drug, 5-fluorocytosine (5-FC), into the cytotoxic agent, 5-fluorouracil (5-FU). As an immunotherapeutic agent, human interferon-β (IFN-β) has anticancer effects. In this study, we used modified human neural stem cells (HB1.F3) expressing the Escherichia coli (E. coli) CD gene (HB1.F3.CD) or both the CD and human IFN-β genes (HB1.F3.CD.IFN-β) and evaluated their effectiveness on gastric carcinoma cells (AGS); migration of GESTECs to AGS was analyzed as well as formation of 5-FU and IFN-β. Reverse transcription-polymerase chain reaction (RT-PCR) was used to confirm the expression of CD and IFN-β genes in GESTECs along with confirming the production of chemoattractant molecules such as stem cell factor (SCF), CXCR4, c-Kit, vascular endothelial growth factor (VEGF) and VEGF receptor 2 (VEGFR2). In addition, by co-culturing GESTECs with AGS in the presence of 5-FC, we were able to confirm that cancer growth was inhibited, along with a synergistic effect when the CD and IFN-β genes (HB1.F3.CD.IFN-β) were co-expressed. Indeed a marked anticancer effect was demonstrated when the CD and IFN-β genes were expressed together compared to expression of the CD gene alone (HB1.F3.CD). According to a modified transwell migration assay, the migration of GESTECs toward AGS was confirmed. In conclusion, these data suggest potential application of GESTECs to gastric cancer therapy, due to a remarkable synergistic effect of CD and IFN-β genes in the presence of 5-FC. Additionally, the tumor-selective migration capability in vitro suggests that GESTECs are a potential anticancer therapy candidate that may result in minimal side effects compared to the conventional chemotherapy.
Collapse
Affiliation(s)
- Kyoung-Yoon Kim
- Laboratory of Veterinary Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
26
|
Guillemot F, Zimmer C. From cradle to grave: the multiple roles of fibroblast growth factors in neural development. Neuron 2011; 71:574-88. [PMID: 21867876 DOI: 10.1016/j.neuron.2011.08.002] [Citation(s) in RCA: 179] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2011] [Indexed: 01/08/2023]
Abstract
The generation of a functional nervous system involves a multitude of steps that are controlled by just a few families of extracellular signaling molecules. Among these, the fibroblast growth factor (FGF) family is particularly prominent for the remarkable diversity of its functions. FGFs are best known for their roles in the early steps of patterning of the neural primordium and proliferation of neural progenitors. However, other equally important functions have emerged more recently, including in the later steps of neuronal migration, axon navigation, and synaptogenesis. We review here these diverse functions and discuss the mechanisms that account for this unusual range of activities. FGFs are essential components of most protocols devised to generate therapeutically important neuronal populations in vitro or to stimulate neuronal repair in vivo. How FGFs promote the development of the nervous system and maintain its integrity will thus remain an important focus of research in the future.
Collapse
Affiliation(s)
- François Guillemot
- Division of Molecular Neurobiology, Medical Research Council, National Institute for Medical Research, The Ridgeway, Mill Hill, London NW71AA, UK.
| | | |
Collapse
|
27
|
Angibaud J, Baudouin SJ, Louveau A, Nerrière-Daguin V, Bonnamain V, Csaba Z, Dournaud P, Naveilhan P, Noraz N, Pellier-Monnin V, Boudin H. Ectopic expression of the immune adaptor protein CD3zeta in neural stem/progenitor cells disrupts cell-fate specification. J Mol Neurosci 2011; 46:431-41. [PMID: 21809042 DOI: 10.1007/s12031-011-9607-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Accepted: 07/20/2011] [Indexed: 12/17/2022]
Abstract
Immune signaling and neuroinflammatory mediators have recently emerged as influential variables that regulate neural precursor/stem cell (NPC) behavior and function. In this study, we investigated whether the signaling adaptor protein CD3ζ, a transmembrane protein involved in T cell differentiation and function and recently shown to regulate neuronal development in the central nervous system (CNS), may have a role in NPC differentiation. We analyzed the expression profile of CD3ζ in embryonic rat brain during neurogenic periods and in neurosphere-derived neural cells, and we investigated the action of CD3ζ on cell differentiation. We found that CD3ζ expression coincided with neuronal commitment, but its forced expression in NPCs prevented the production of neurons and oligodendrocytes, but not astroglial cells. This blockade of neuronal differentiation was operated through an ITAM-independent mechanism, but required the Asp36 of the CD3ζ transmembrane domain involved in membrane receptor interaction. Together, our findings show that ectopic CD3ζ expression in NPCs impaired their normal cell-fate specification and suggest that variations of CD3ζ expression in the developing CNS might result in neurodevelopmental anomalies.
Collapse
|
28
|
Serre A, Snyder EY, Mallet J, Buchet D. Overexpression of basic helix-loop-helix transcription factors enhances neuronal differentiation of fetal human neural progenitor cells in various ways. Stem Cells Dev 2011; 21:539-53. [PMID: 21561385 DOI: 10.1089/scd.2011.0079] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In a perspective of regenerative medicine, multipotent human neural progenitor cells (hNPCs) offer a therapeutic advantage over pluripotent stem cells in that they are already invariantly "neurally committed" and lack tumorigenicity. However, some of their intrinsic properties, such as slow differentiation and uncontrolled multipotency, remain among the obstacles to their routine use for transplantation. Although rodent NPCs have been genetically modified in vitro to overcome some of these limitations, the translation of this strategy to human cells remains in its early stages. In the present study, we compare the actions of 4 basic helix-loop-helix transcription factors on the proliferation, specification, and terminal differentiation of hNPCs isolated from the fetal dorsal telencephalon. Consistent with their proneural activity, Ngn1, Ngn2, Ngn3, and Mash1 prompted rapid commitment of the cells. The Ngns induced a decrease in proliferation, whereas Mash1 maintained committed progenitors in a proliferative state. As opposed to Ngn1 and Ngn3, which had no effect on glial differentiation, Ngn2 induced an increase in astrocytes in addition to neurons, whereas Mash1 led to both neuronal and oligodendroglial specification. GABAergic, cholinergic, and motor neuron differentiations were considerably increased by overexpression of Ngn2 and, to a lesser extent, of Ngn3 and Mash1. Thus, we provide evidence that hNPCs can be efficiently, rapidly, and safely expanded in vitro as well as rapidly differentiated toward mature neural (typically neuronal) lineages by the overexpression of select proneural genes.
Collapse
Affiliation(s)
- Angéline Serre
- Centre de Recherche de l'Institut du Cerveau et de la Moelle Epinière, Université Pierre et Marie Curie-Paris 6, UMR-S975, Paris, France
| | | | | | | |
Collapse
|
29
|
Kaeser M, Brunet JF, Wyss A, Belhaj-Saif A, Liu Y, Hamadjida A, Rouiller EM, Bloch J. Autologous Adult Cortical Cell Transplantation Enhances Functional Recovery Following Unilateral Lesion of Motor Cortex in Primates: A Pilot Study. Neurosurgery 2011; 68:1405-16; discussion 1416-7. [DOI: 10.1227/neu.0b013e31820c02c0] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
30
|
Abstract
INTRODUCTION Cell transplants to replace cells lost due to injury or degenerative diseases, for which there are currently no cures, are being pursued in a wide range of experimental models. Thus, the application of stem cell-based therapies to treat neurodegenerative and traumatic injuries is now a clinical reality. However, the monitoring of cellular grafts, non-invasively, is an important aspect of the ongoing efficacy and safety assessment of cell-based therapies. Hence, there is a need for non-invasive imaging techniques to ensure that transplants are not only administered to the relevant site, but also allow the monitoring of inappropriate cellular migration to improve our understanding of stem cell migration in the context of the whole organism. AREAS COVERED This review provides an up to date overview of molecular imaging approaches that have been used for visualizing and tracking transplanted stem cells, in vivo. EXPERT OPINION It's important to emphasize that the application of molecular imaging to interrogate transplanted cells may require one or even two imaging modalities to provide a reasonable assessment of transplanted cells in specific organs.
Collapse
Affiliation(s)
- Kishore Bhakoo
- Singapore Bioimaging Consortium - A*Star - Translational Molecular Imaging, Singapore 138667, Singapore.
| |
Collapse
|
31
|
Sørensen AT, Rogelius N, Lundberg C, Kokaia M. Activity-dependent long-term plasticity of afferent synapses on grafted stem/progenitor cell-derived neurons. Exp Neurol 2011; 229:274-81. [PMID: 21324317 DOI: 10.1016/j.expneurol.2011.02.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Revised: 02/01/2011] [Accepted: 02/09/2011] [Indexed: 01/24/2023]
Abstract
Stem cell-based cell replacement therapies aiming at restoring injured or diseased brain function ultimately rely on the capability of transplanted cells to promote functional recovery. The mechanisms by which stem cell-based therapies for neurological conditions can lead to functional recovery are uncertain, but structural and functional repair appears to depend on integration of transplanted cell-derived neurons into neuronal circuitries. The nature by which stem/progenitor cell-derived neurons synaptically integrate into neuronal circuitries is largely unexplored. Here we show that transplanted GFP-labeled neuronal progenitor cells into the rat hippocampus exhibit mature neuronal morphology following 4-10 weeks. GFP-positive cells were preferentially integrated into the principal cell layers of hippocampus, particularly CA3. Patch-clamp recordings from GFP-expressing cells revealed that they generated fast action potentials, and their intrinsic membrane properties were overall similar to endogenous host neurons recorded in same areas. As judged by occurrence of spontaneous excitatory postsynaptic currents (EPSCs), transplanted GFP-positive cells were synaptically integrated into the host circuitry. Comparable to host neurons, both paired-pulse depression and facilitation of afferent fiber stimulation-evoked EPSCs were observed in GFP-positive cells. Upon high-frequency stimulation, GFP-positive cells displayed post-tetanic potentiation of EPSCs, in some cases followed by long-term potentiation (LTP) lasting for more than 30 min. Our data show for the first time that transplanted neuronal progenitor cells can become functional neurons and their afferent synapses are capable of expressing activity-dependent short and long-term plasticity. These synaptic properties may facilitate host-to-graft interactions and regulate activity of the grafted cells promoting functional recovery of the diseased brain.
Collapse
Affiliation(s)
- Andreas Toft Sørensen
- Experimental Epilepsy Group, Wallenberg Neuroscience Center, Lund University Hospital, 221 84 Lund, Sweden.
| | | | | | | |
Collapse
|
32
|
Cell Fusion and Tissue Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 713:161-75. [DOI: 10.1007/978-94-007-0763-4_10] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
33
|
Hirth F. Stem Cells and Asymmetric Cell Division. Regen Med 2011. [DOI: 10.1007/978-90-481-9075-1_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
34
|
Akchiche N, Bossenmeyer-Pourié C, Pourié G, Koziel V, Nédélec E, Guéant JL, Daval JL. Differentiation and neural integration of hippocampal neuronal progenitors: signaling pathways sequentially involved. Hippocampus 2010; 20:949-61. [PMID: 19714568 DOI: 10.1002/hipo.20690] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In the context of their potential implication in regenerative strategies, we characterized cell mechanisms underlying the fate of embryonic rat hippocampal H19-7 progenitors in culture upon induction of their differentiation, and tested their capacities to integrate into a neuronal network in vitro. Without addition of growth factors, nearly 100% of cells expressed various neuronal markers, with a progressive rise of the expression of Synapsin I and II, suggesting that cells developed as mature neurons with synaptogenic capacities. Fully differentiated neurons were identified as glutamatergic and expressed the receptor-associated protein PSD-95. Quantification of ATP showed that 60% of cells died within 24 h after differentiation. Cell death was shown to imply Erk1/2-dependent intrinsic mitochondrial apoptosis signaling pathway, with activation of caspase-9 and -3, finally leading to single-strand DNA. Surviving neurons displayed high levels of Akt, phospho-Akt, and antiapoptotic proteins such as Bcl-2 and Bcl-XL, with decreased caspase activation. In the absence of trophic support, the proapoptotic death-associated protein (DAP) kinase was dramatically stimulated by 24 h postdifferentiation, along with increased levels of p38 and phospho-p38, and caspase reactivation. These findings show that different signaling pathways are sequentially triggered by differentiation, and highlight that ultimate cell death would involve p38 and DAP kinase activation. This was supported by the improvement of cell survival at 24-h postdifferentiation when cells were treated by PD169316, a specific inhibitor of p38. Finally, when seeded on rat hippocampal primary cultured neurons, a significant number of differentiated H19-7 cells were able to survive and to develop cell-cell communication.
Collapse
|
35
|
Voss M, Bischof F. Recurrent myelitis after allogeneic stem cell transplantation. Report of two cases. BMC Neurol 2010; 10:76. [PMID: 20809972 PMCID: PMC2936883 DOI: 10.1186/1471-2377-10-76] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2010] [Accepted: 09/01/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Allogeneic and autologous haematopoietic stem cell transplantation are established treatment options for haematological malignancies and may possibly be employed to treat a range of genetic and autoimmune diseases. CASE PRESENTATION We report two patients who developed an acute myelitis within their thoracic spinal cord after allogeneic stem cell transplantation. Myelitis in these patients was not related to graft versus host disease or immune reconstitution and was responsive to intravenous methylprednisolone and cyclophosphamide. CONCLUSIONS Myelitis is a possibly disabling consequence of haematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Martin Voss
- Hertie Institute for Clinical Brain Research, Department of General Neurology, University of Tübingen, Hoppe-Seyler-Strasse 3, 72076 Tübingen, Germany
| | | |
Collapse
|
36
|
Bifari F, Pacelli L, Krampera M. Immunological properties of embryonic and adult stem cells. World J Stem Cells 2010; 2:50-60. [PMID: 21607122 PMCID: PMC3097923 DOI: 10.4252/wjsc.v2.i3.50] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Revised: 03/17/2010] [Accepted: 03/24/2010] [Indexed: 02/06/2023] Open
Abstract
The possibility of treating degenerative diseases by stem cell-based approaches is a promising therapeutical option. Among major concerns for the clinical application of stem cells, some derive from the possibility that stem cells may be rejected by the immune system as a consequence of histoincompatibility and that stem cells themselves may interfere with the normal functions of host immune response. Therefore, the immunogenicity and the immunomodulatory properties of stem cells must be carefully addressed. Although these properties are common features of different stem cell types, some peculiarities can be recognized and characterized for their proper clinical use.
Collapse
Affiliation(s)
- Francesco Bifari
- Francesco Bifari, Luciano Pacelli, Mauro Krampera, Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Pz.le Scuro 10, 37134 Verona, Italy
| | | | | |
Collapse
|
37
|
Shihabuddin LS, Aubert I. Stem cell transplantation for neurometabolic and neurodegenerative diseases. Neuropharmacology 2010; 58:845-54. [DOI: 10.1016/j.neuropharm.2009.12.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2009] [Revised: 12/11/2009] [Accepted: 12/15/2009] [Indexed: 01/21/2023]
|
38
|
Neri M, Maderna C, Ferrari D, Cavazzin C, Vescovi AL, Gritti A. Robust generation of oligodendrocyte progenitors from human neural stem cells and engraftment in experimental demyelination models in mice. PLoS One 2010; 5:e10145. [PMID: 20405042 PMCID: PMC2853578 DOI: 10.1371/journal.pone.0010145] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2009] [Accepted: 03/16/2010] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Cell-based therapy holds great promises for demyelinating diseases. Human-derived fetal and adult oligodendrocyte progenitors (OPC) gave encouraging results in experimental models of dysmyelination but their limited proliferation in vitro and their potential immunogenicity might restrict their use in clinical applications. Virtually unlimited numbers of oligodendroglial cells could be generated from long-term self-renewing human (h)-derived neural stem cells (hNSC). However, robust oligodendrocyte production from hNSC has not been reported so far, indicating the need for improved understanding of the molecular and environmental signals controlling hNSC progression through the oligodendroglial lineage. The aim of this work was to obtain enriched and renewable cultures of hNSC-derived oligodendroglial cells by means of epigenetic manipulation. METHODOLOGY/PRINCIPAL FINDINGS We report here the generation of large numbers of hNSC-derived oligodendroglial cells by concurrent/sequential in vitro exposure to combinations of growth factors (FGF2, PDGF-AA), neurotrophins (NT3) and hormones (T3). In particular, the combination FGF2+NT3+PDGF-AA resulted in the maintenance and enrichment of an oligodendroglial cell population displaying immature phenotype (i.e., proliferation capacity and expression of PDGFRalpha, Olig1 and Sox10), limited self-renewal and increased migratory activity in vitro. These cells generate large numbers of oligodendroglial progeny at the early stages of maturation, both in vitro and after transplantation in models of CNS demyelination. CONCLUSIONS/SIGNIFICANCE We describe a reliable method to generate large numbers of oligodendrocytes from a renewable source of somatic, non-immortalized NSC from the human foetal brain. We also provide insights on the mechanisms underlying the pro-oligodendrogenic effect of the treatments in vitro and discuss potential issues responsible for the limited myelinating capacity shown by hNSC-derived oligodendrocytes in vivo.
Collapse
Affiliation(s)
- Margherita Neri
- San Raffaele Scientific Institute, San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), Milano, Italy
- Vita-Salute San Raffaele University, Milano, Italy
| | - Claudio Maderna
- San Raffaele Scientific Institute, San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), Milano, Italy
| | - Daniela Ferrari
- Bioscience and Biotechnology Department, University of Milano-Bicocca, Milano, Italy
| | - Chiara Cavazzin
- San Raffaele Scientific Institute, San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), Milano, Italy
| | - Angelo L. Vescovi
- Bioscience and Biotechnology Department, University of Milano-Bicocca, Milano, Italy
| | - Angela Gritti
- San Raffaele Scientific Institute, San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), Milano, Italy
| |
Collapse
|
39
|
Kim KY, Kim SU, Leung PCK, Jeung EB, Choi KC. Influence of the prodrugs 5-fluorocytosine and CPT-11 on ovarian cancer cells using genetically engineered stem cells: tumor-tropic potential and inhibition of ovarian cancer cell growth. Cancer Sci 2010; 101:955-62. [PMID: 20704576 PMCID: PMC11159652 DOI: 10.1111/j.1349-7006.2009.01485.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2024] Open
Abstract
Recent studies have shown that genetically engineered stem cells (GESTECs) to produce suicide enzymes that convert non-toxic prodrugs to toxic metabolites selectively migrate toward tumor sites and reduce tumor growth. In the present study, we evaluated whether these GESTECs were capable of migrating to human ovarian cancer cells and examined the potential therapeutic efficacy of the gene-directed enzyme prodrug therapy against ovarian cancer cells in vitro. The expression of cytosine deaminase (CD) or carboxyl esterase (CE) mRNA of GESTECs was confirmed by RT-PCR. A modified transwell migration assay was performed to determine the migratory capacity of GESTECs to ovarian cancer cells. GESTECs (HB1.F3.CD or HB1.F3.CE cells) engineered to express a suicide gene (CD or CE) selectively migrated toward ovarian cancer cells. A [(3)H] thymidine incorporation assay was conducted to measure the proliferative index. Treatment of human epithelial ovarian cancer cell line (SKOV-3, an ovarian adenocarcinoma derived from the ascites of an ovarian cancer patient) with the prodrugs 5-fluorocytosine (5-FC) or camptothecin-11 (CPT-11) in the presence of HB1.F3.CD or HB1.F3.CE cells resulted in the inhibition of ovarian cancer cell growth. Based on the data presented herein, we suggest that GESTECs expressing CD/CE may have a potent advantage to selectively treat ovarian cancers.
Collapse
Affiliation(s)
- Ki-Yon Kim
- Department of Obstetrics and Gynecology, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | |
Collapse
|
40
|
Kim KY, Kim SU, Leung PCK, Jeung EB, Choi KC. Influence of the prodrugs 5-fluorocytosine and CPT-11 on ovarian cancer cells using genetically engineered stem cells: tumor-tropic potential and inhibition of ovarian cancer cell growth. Cancer Sci 2010. [DOI: 10.1111/j.1349-7006.2010.01485.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
41
|
Ning H, Huang YC, Banie L, Hung S, Lin G, Li LC, Lue TF, Lin CS. MicroRNA regulation of neuron-like differentiation of adipose tissue-derived stem cells. Differentiation 2009; 78:253-9. [PMID: 19695767 PMCID: PMC2787638 DOI: 10.1016/j.diff.2009.07.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Revised: 07/24/2009] [Accepted: 07/30/2009] [Indexed: 01/08/2023]
Abstract
We have reported previously that adipose tissue-derived stem cells (ADSC) could be induced by isobutylmethylxanthine (IBMX) to differentiate into neuron-like cells and such differentiation was mediated by insulin-like growth factor I (IGF-I) signaling. In the present study we show that both IBMX and IGF-I upregulated neural markers beta-III-tubulin and paired-like homeobox transcription factor (Pitx3) in ADSC. Because Pitx3 is important for the maturation and function of dopaminergic neurons and has been shown to be regulated by microRNA miR-133b, we also examined the effects of IBMX and IGF-I on miR-133b expression. The results show that both IBMX and IGF-I upregulated miR-133b expression. When miR-133b was overexpressed in ADSC through transfection of a synthetic microRNA mimic, it caused a downregulation of beta-III-tubulin as evidenced by immunofluorescence staining and western blot analysis. Overexpression of miR-133b also downregulated Pitx3 and IGF-1 receptor (IGF-IR), and all such downregulation occurred at the protein but not the RNA level. The apparent posttranscriptional regulation was subsequently found to be exerted through a potential miR-133b target in the 3'-untranslated region of IGF-IR, as evidenced by luciferase assay. Thus, IGF-I signaling and miR-133b co-regulate potential neural differentiation of ADSC through a feedback mechanism, in which IGF-I upregulates miR-133b while miR-133b in turn downregulates IGF-IR.
Collapse
Affiliation(s)
- Hongxiu Ning
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University, of California, San Francisco, CA 94143-0738, USA
| | - Yun-Ching Huang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University, of California, San Francisco, CA 94143-0738, USA
- Division of Urology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Chia-Yi, Taiwan
| | - Lia Banie
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University, of California, San Francisco, CA 94143-0738, USA
| | - Steven Hung
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University, of California, San Francisco, CA 94143-0738, USA
| | - Guiting Lin
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University, of California, San Francisco, CA 94143-0738, USA
| | - Long-Cheng Li
- Department of Urology, School of Medicine, University of California, San Francisco, CA 94143-0738, USA
| | - Tom F. Lue
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University, of California, San Francisco, CA 94143-0738, USA
| | - Ching-Shwun Lin
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University, of California, San Francisco, CA 94143-0738, USA
| |
Collapse
|
42
|
Transplanted myogenic progenitor cells express neuronal markers in the CNS and ameliorate disease in Experimental Autoimmune Encephalomyelitis. J Neuroimmunol 2009; 215:73-83. [DOI: 10.1016/j.jneuroim.2009.08.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2009] [Revised: 08/23/2009] [Accepted: 08/24/2009] [Indexed: 12/27/2022]
|
43
|
Kim DW, Hirth F. Genetic mechanisms regulating stem cell self-renewal and differentiation in the central nervous system of Drosophila. Cell Adh Migr 2009; 3:402-11. [PMID: 19421003 PMCID: PMC2802756 DOI: 10.4161/cam.3.4.8690] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2009] [Accepted: 04/07/2009] [Indexed: 01/06/2023] Open
Abstract
Recent studies using the Drosophila central nervous system as a model have identified key molecules and mechanisms underlying stem cell self-renewal and differentiation. These studies suggest that proteins like Aurora-A, atypical protein kinase C, Prospero and Brain tumor act as key regulators in a tightly coordinated interplay between mitotic spindle orientation and asymmetric protein localization. These data also provide initial evidence that both processes are coupled to cell cycle progression and growth control, thereby regulating a binary switch between proliferative stem self-renewal and differentiative progenitor cell specification. Considering the evolutionary conservation of some of the mechanisms and molecules involved, these data provide a rationale and genetic model for understanding stem cell self-renewal and differentiation in general. The new data gained in Drosophila may therefore lead to conceptual advancements in understanding the aetiology and treatment of human neurological disorders such as brain tumor formation and neurodegenerative diseases.
Collapse
Affiliation(s)
- Dongwook W Kim
- MRC Centre for Neurodegeneration Research, Institute of Psychiatry, King's College London, London, UK
| | | |
Collapse
|
44
|
Wilkins A, Kemp K, Ginty M, Hares K, Mallam E, Scolding N. Human bone marrow-derived mesenchymal stem cells secrete brain-derived neurotrophic factor which promotes neuronal survival in vitro. Stem Cell Res 2009; 3:63-70. [DOI: 10.1016/j.scr.2009.02.006] [Citation(s) in RCA: 211] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2008] [Revised: 12/03/2008] [Accepted: 02/25/2009] [Indexed: 12/19/2022] Open
|
45
|
Rey SM, Povazay B, Hofer B, Unterhuber A, Hermann B, Harwood A, Drexler W. Three- and four-dimensional visualization of cell migration using optical coherence tomography. JOURNAL OF BIOPHOTONICS 2009; 2:370-9. [PMID: 19475627 DOI: 10.1002/jbio.200910027] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Conventionally, cell chemotaxis is studied on two-dimensional (2D) transparent surfaces, due to limitations in optical and image data-collection techniques. However, surfaces that more closely mimic the natural environment of cells are often opaque. Optical coherence tomography (OCT) is a noninvasive label-free imaging technique, which offers the potential to visualize moving cells on opaque surfaces and in three dimensions (3D). Here, we demonstrate that OCT is an effective means of time-lapse videomicroscopy of Dictyostelium cells undergoing 3D (2D+time) cell migration on nitrocellulose substrates and 4D (3D+time) chemotaxis within low-density agarose gels. The generated image sequences are compatible with current computer-based image-analysis software for quantification of cell motility. This demonstrates the utility of OCT for cell tracking and analysis of cell chemotaxis in complex environments.
Collapse
Affiliation(s)
- Sara M Rey
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | | | | | | | | | | | | |
Collapse
|
46
|
Jackson J, Chapon C, Jones W, Hirani E, Qassim A, Bhakoo K. In vivo multimodal imaging of stem cell transplantation in a rodent model of Parkinson's disease. J Neurosci Methods 2009; 183:141-8. [PMID: 19559725 DOI: 10.1016/j.jneumeth.2009.06.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Revised: 06/15/2009] [Accepted: 06/17/2009] [Indexed: 12/16/2022]
Abstract
Stem cell therapy in the nervous system aims to replace the lost neurons and provide functional recovery. However, it is imperative that we understand the in vivo behaviour of these cells post-implantation. We report visualisation of iron oxide labelled bone marrow-derived stem cells (BMSCs) implanted into the striatum of hemi-parkinsonian rats by magnetic resonance imaging (MRI). Functional efficacy of the donor cells was monitored in vivo using the positron emission tomography (PET) radioligand [11C]raclopride. The cells were visible for 28 days by in vivo MRI. BMSCs provided functional recovery demonstrated by a decreased binding of [11C]raclopride. Although, histology confirmed the persistence of donor cells, no tyrosine hydroxylase positive cells were present. This suggests that BMSCs may have a limited paracrine effect and influence functional recovery. We demonstrate, using multimodal imaging, that we can not only track BMSCs but also establish their effects in a pre-clinical model of Parkinson's disease.
Collapse
Affiliation(s)
- Johanna Jackson
- Stem Cell Imaging, MRC Clinical Sciences Centre, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom.
| | | | | | | | | | | |
Collapse
|
47
|
Walking pattern analysis after unilateral 6-OHDA lesion and transplantation of foetal dopaminergic progenitor cells in rats. Behav Brain Res 2009; 199:317-25. [DOI: 10.1016/j.bbr.2008.12.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2008] [Accepted: 12/07/2008] [Indexed: 11/21/2022]
|
48
|
Increased neurogenesis after hypoxic-ischemic encephalopathy in humans is age related. Acta Neuropathol 2009; 117:525-34. [PMID: 19277687 DOI: 10.1007/s00401-009-0509-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2008] [Revised: 02/01/2009] [Accepted: 02/27/2009] [Indexed: 12/27/2022]
Abstract
The leading cause of morbidity and mortality after successful resuscitation is hypoxic-ischemic encephalopathy (HIE), which results in neuronal loss within the neocortex and the hippocampal formation. This study focuses on the impact of HIE on adult neurogenesis in the human hippocampal dentate gyrus as a potential intrinsic regenerative mechanism in response to neuronal damage. Brain sections of 22 autopsy cases with HIE and of 19 age-matched controls without neuropathological abnormalities were investigated by means of immunohistochemistry. The densities of immature granule cells during axon guidance and outgrowth (assessed by TUC-4 immunohistochemistry) and of young calretinin-expressing postmitotic neurons were increased in the granule cell layer of cases who had suffered from HIE (P = 0.0002 and P = 0.0001, respectively). Similarly, the density of apoptotic granule cells, as detected by in situ tailing and morphological criteria, was increased in HIE (P = 0.014). In cases with HIE, the increase in the density of TUC-4-labeled cells inversely correlated with age (P = 0.027). In contrast, neither the density of proliferating nor that of apoptotic cells was substantially influenced by age within the control group. Taken together, both an increase in adult neurogenesis and in neuronal apoptosis was observed in the human dentate gyrus in response to HIE. The data suggest a decrease of adult neurogenesis in older-aged cases. Whether neurogenesis can contribute to recovery after HIE remains to be determined. The stimulation of adult neurogenesis may be less efficient in older victims of HIE.
Collapse
|
49
|
Bifari F, Decimo I, Chiamulera C, Bersan E, Malpeli G, Johansson J, Lisi V, Bonetti B, Fumagalli G, Pizzolo G, Krampera M. Novel stem/progenitor cells with neuronal differentiation potential reside in the leptomeningeal niche. J Cell Mol Med 2009; 13:3195-208. [PMID: 19228261 PMCID: PMC4516477 DOI: 10.1111/j.1582-4934.2009.00706.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Stem cells capable of generating neural differentiated cells are recognized by the expression of nestin and reside in specific regions of the brain, namely, hippocampus, subventricular zone and olfactory bulb. For other brain structures, such as leptomeninges, which contribute to the correct cortex development and functions, there is no evidence so far that they may contain stem/precursor cells. In this work, we show for the first time that nestin-positive cells are present in rat leptomeninges during development up to adulthood. The newly identified nestin-positive cells can be extracted and expanded in vitro both as neurospheres, displaying high similarity with subventricular zone-derived neural stem cells, and as homogeneous cell population with stem cell features. In vitro expanded stem cell population can differentiate with high efficiency into excitable cells with neuronal phenotype and morphology. Once injected into the adult brain, these cells survive and differentiate into neurons, thus showing that their neuronal differentiation potential is operational also in vivo. In conclusion, our data provide evidence that a specific population of immature cells endowed of neuronal differentiation potential is resident in the leptomeninges throughout the life. As leptomeninges cover the entire central nervous system, these findings could have relevant implications for studies on cortical development and for regenerative medicine applied to neurological disorders.
Collapse
Affiliation(s)
- Francesco Bifari
- Department of Clinical and Experimental Medicine, Stem Cell Research Laboratory, Section of Hematology, University of Verona, Verona, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Cumberland Consensus Working Group, Cheeran B, Cohen L, Dobkin B, Ford G, Greenwood R, Howard D, Husain M, Macleod M, Nudo R, Rothwell J, Rudd A, Teo J, Ward N, Wolf S. The future of restorative neurosciences in stroke: driving the translational research pipeline from basic science to rehabilitation of people after stroke. Neurorehabil Neural Repair 2009; 23:97-107. [PMID: 19189939 PMCID: PMC3230220 DOI: 10.1177/1545968308326636] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Major advances during the past 50 years highlight the immense potential for restoration of function after neural injury, even in the damaged adult human brain. Yet, the translation of these advances into clinically useful treatments is painstakingly slow. OBJECTIVE Here, we consider why the traditional model of a "translational research pipeline" that transforms basic science into novel clinical practice has failed to improve rehabilitation practice for people after stroke. RESULTS We find that (1) most treatments trialed in vitro and in animal models have not yet resulted in obviously useful functional gains in patients; (2) most clinical trials of restorative treatments after stroke have been limited to small-scale studies; (3) patient recruitment for larger clinical trials is difficult; (4) the determinants of patient outcomes and what patients want remain complex and ill-defined, so that basic scientists have no clear view of the clinical importance of the problems that they are addressing; (5) research in academic neuroscience centers is poorly integrated with practice in front-line hospitals and the community, where the majority of patients are treated; and (6) partnership with both industry stakeholders and patient pressure groups is poorly developed, at least in the United Kingdom where research in the translational restorative neurosciences in stroke depends on public sector research funds and private charities. CONCLUSIONS We argue that interaction between patients, front-line clinicians, and clinical and basic scientists is essential so that they can explore their different priorities, skills, and concerns. These interactions can be facilitated by funding research consortia that include basic and clinical scientists, clinicians and patient/carer representatives with funds targeted at those impairments that are major determinants of patient and carer outcomes. Consortia would be instrumental in developing a lexicon of common methods, standardized outcome measures, data sharing and long-term goals. Interactions of this sort would create a research-friendly, rather than only target-led, culture in front-line stroke rehabilitation services.
Collapse
Collaborators
Ann Ashburn, Jane Burridge, John Bamford, Jean-Claude Baron, Phillip Bath, Jon Driver, Steve Dunnett, Martin Eccles, Anne Forster, Shaheen Hamdy, Peter U Heuschmann, Neville Hogan, Roger Lemon, Jon Marsden, Averil Mansfield, Robert McCrum, Isaebel Mhairi Macrae, Cathy Price, Alex Leff, Yves Rosetti, Duncan Turner, Paulette van Vliet, Alan Wing, Derick Wade, Cornelius Weiller, Richard Wise, Ulf Ziemann,
Collapse
|