1
|
Xu J, Hsu SH. Self-healing hydrogel as an injectable implant: translation in brain diseases. J Biomed Sci 2023; 30:43. [PMID: 37340481 DOI: 10.1186/s12929-023-00939-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/13/2023] [Indexed: 06/22/2023] Open
Abstract
Tissue engineering biomaterials are aimed to mimic natural tissue and promote new tissue formation for the treatment of impaired or diseased tissues. Highly porous biomaterial scaffolds are often used to carry cells or drugs to regenerate tissue-like structures. Meanwhile, self-healing hydrogel as a category of smart soft hydrogel with the ability to automatically repair its own structure after damage has been developed for various applications through designs of dynamic crosslinking networks. Due to flexibility, biocompatibility, and ease of functionalization, self-healing hydrogel has great potential in regenerative medicine, especially in restoring the structure and function of impaired neural tissue. Recent researchers have developed self-healing hydrogel as drug/cell carriers or tissue support matrices for targeted injection via minimally invasive surgery, which has become a promising strategy in treating brain diseases. In this review, the development history of self-healing hydrogel for biomedical applications and the design strategies according to different crosslinking (gel formation) mechanisms are summarized. The current therapeutic progress of self-healing hydrogels for brain diseases is described as well, with an emphasis on the potential therapeutic applications validated by in vivo experiments. The most recent aspect as well as the design rationale of self-healing hydrogel for different brain diseases is also addressed.
Collapse
Affiliation(s)
- Junpeng Xu
- Institute of Polymer Science and Engineering, National Taiwan University, No. 1, Sec. 4 Roosevelt Road, Taipei, 106319, Taiwan, Republic of China
| | - Shan-Hui Hsu
- Institute of Polymer Science and Engineering, National Taiwan University, No. 1, Sec. 4 Roosevelt Road, Taipei, 106319, Taiwan, Republic of China.
- Institute of Cellular and System Medicine, National Health Research Institutes, No. 35 Keyan Road, Miaoli, 350401, Taiwan, Republic of China.
| |
Collapse
|
2
|
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
|
3
|
Yue C, Feng S, Chen Y, Jing N. The therapeutic prospects and challenges of human neural stem cells for the treatment of Alzheimer's Disease. CELL REGENERATION (LONDON, ENGLAND) 2022; 11:28. [PMID: 36050613 PMCID: PMC9437172 DOI: 10.1186/s13619-022-00128-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/15/2022] [Indexed: 06/15/2023]
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disorder associated with aging. Due to its insidious onset, protracted progression, and unclear pathogenesis, it is considered one of the most obscure and intractable brain disorders, and currently, there are no effective therapies for it. Convincing evidence indicates that the irreversible decline of cognitive abilities in patients coincides with the deterioration and degeneration of neurons and synapses in the AD brain. Human neural stem cells (NSCs) hold the potential to functionally replace lost neurons, reinforce impaired synaptic networks, and repair the damaged AD brain. They have therefore received extensive attention as a possible source of donor cells for cellular replacement therapies for AD. Here, we review the progress in NSC-based transplantation studies in animal models of AD and assess the therapeutic advantages and challenges of human NSCs as donor cells. We then formulate a promising transplantation approach for the treatment of human AD, which would help to explore the disease-modifying cellular therapeutic strategy for the treatment of human AD.
Collapse
Affiliation(s)
- Chunmei Yue
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, 215000, China.
| | - Su Feng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
- Bioland Laboratory/Guangzhou Laboratory, Guangzhou, 510005, China
| | - Yingying Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Naihe Jing
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Bioland Laboratory/Guangzhou Laboratory, Guangzhou, 510005, China.
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
4
|
Yoo SH, Lee SH, Lee S, Park JH, Lee S, Jin H, Park HJ. The effect of human mesenchymal stem cell injection on pain behavior in chronic post-ischemia pain mice. Korean J Pain 2020; 33:23-29. [PMID: 31888314 PMCID: PMC6944374 DOI: 10.3344/kjp.2020.33.1.23] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 10/29/2019] [Accepted: 11/01/2019] [Indexed: 12/30/2022] Open
Abstract
Background Neuropathic pain (NP) is considered a clinically incurable condition despite various treatment options due to its diverse causes and complicated disease mechanisms. Since the early 2000s, multipotent human mesenchymal stem cells (hMSCs) have been used in the treatment of NP in animal models. However, the effects of hMSC injections have not been studied in chronic post-ischemia pain (CPIP) mice models. Here, we investigated whether intrathecal (IT) and intrapaw (IP) injections of hMSCs can reduce mechanical allodynia in CPIP model mice. Methods Seventeen CPIP C57/BL6 mice were selected and randomized into four groups: IT sham (n = 4), IT stem (n = 5), IP sham (n = 4), and IP stem (n = 4). Mice in the IT sham and IT stem groups received an injection of 5 μL saline and 2 × 104 hMSCs, respectively, while mice in the IP sham and IP stem groups received an injection of 5 μL saline and 2 × 105 hMSCs, respectively. Mechanical allodynia was assessed using von Frey filaments from pre-injection to 30 days post-injection. Glial fibrillary acidic protein (GFAP) expression in the spinal cord and dorsal root ganglia were also evaluated. Results IT and IP injections of hMSCs improved mechanical allodynia. GFAP expression was decreased on day 25 post-injection compared with the sham group. Injections of hMSCs improved allodynia and GFAP expression was decreased compared with the sham group. Conclusions These results suggested that hMSCs may be also another treatment modality in NP model by ischemia-reperfusion.
Collapse
Affiliation(s)
- Sie Hyeon Yoo
- Department of Anesthesiology and Pain Medicine, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Sung Hyun Lee
- Department of Anesthesiology and Pain Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seunghwan Lee
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jae Hong Park
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seunghyeon Lee
- Department of Anesthesiology and Pain Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Heecheol Jin
- Department of Anesthesiology and Pain Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Hue Jung Park
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
5
|
Human Neural Stem Cells with GDNF Site-Specific Integration at AAVS1 by Using AAV Vectors Retained Their Stemness. Neurochem Res 2018; 43:930-937. [PMID: 29435804 DOI: 10.1007/s11064-018-2498-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 01/05/2018] [Accepted: 02/07/2018] [Indexed: 01/30/2023]
Abstract
The neural stem cells (NSCs) have the ability to self-renew, and to migrate to pathologically altered regions of the central nervous system. Glial cell derived neurotrophic factor (GDNF) could protect dopamine neurons and rescue motor neurons in vivo, which has been proposed as a promising candidate for the treatments of degenerative neurological diseases. In order to combine the advantages of neurotrophic factors and stem cells in clinical therapy, we established the modified hNSCs that has site-specific integration of GDNF gene by using recombinant adeno-associated virus (rAAV) vectors. The hNSCs were co-infected by rAAV2-EGFP-GDNF and rAAV2-SVAV2 which provide integrase to specifically integrate GDNF gene into AAVS1 site. The GDNF-hNSCs maintained their original stem cell characteristics and the ability to differentiate into neurons in vitro. In the animal model, the GDNF-hNSCs were specifically transplanted into CA1 area of hippocampi and could migrate to the dentate gyrus region and differentiate into neuronal cells while maintaining GDNF expression. hNSCs with GDNF gene site-specific integration at AAVS1 by using AAV vectors retained their stemness and effectively expressed GDNF, which indicates the potential of employing transplanted hNPCs for treatment of brain injuries and degenerative neurological diseases.
Collapse
|
6
|
Abstract
Neural stem cells (NSCs) have been proposed as a promising cellular source for the treatment of diseases in nervous systems. NSCs can self-renew and generate major cell types of the mammalian central nervous system throughout adulthood. NSCs exist not only in the embryo, but also in the adult brain neurogenic region: the subventricular zone (SVZ) of the lateral ventricle. Embryonic stem (ES) cells acquire NSC identity with a default mechanism. Under the regulations of leukemia inhibitory factor (LIF) and fibroblast growth factors, the NSCs then become neural progenitors. Neurotrophic and differentiation factors that regulate gene expression for controlling neural cell fate and function determine the differentiation of neural progenitors in the developing mammalian brain. For clinical application of NSCs in neurodegenerative disorders and damaged neurons, there are several critical problems that remain to be resolved: 1) how to obtain enough NSCs from reliable sources for autologous transplantation; 2) how to regulate neural plasticity of different adult stem cells; 3) how to control differentiation of NSCs in the adult nervous system. In order to understand the mechanisms that control NSC differentiation and behavior, we review the ontogeny of NSCs and other stem cell plasticity of neuronal differentiation. The role of NSCs and their regulation by neurotrophic factors in CNS development are also reviewed.
Collapse
Affiliation(s)
- Yi-Chao Hsu
- Stem Cell Research Center, National Health Research Institutes, Jhunan, Taiwan
| | - Don-Ching Lee
- Stem Cell Research Center, National Health Research Institutes, Jhunan, Taiwan
| | - Ing-Ming Chiu
- Stem Cell Research Center, National Health Research Institutes, Jhunan, Taiwan
- Department of Internal Medicine, Ohio State University, Columbus, OH 43210, USA
- Institute of Medical Technology, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
7
|
Parmar MS, Mishra SR, Somal A, Pandey S, Kumar GS, Sarkar M, Chandra V, Sharma GT. Expression and secretory profile of buffalo fetal fibroblasts and Wharton's jelly feeder layers. Anim Reprod Sci 2017; 180:66-77. [PMID: 28363499 DOI: 10.1016/j.anireprosci.2017.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/16/2017] [Accepted: 02/19/2017] [Indexed: 11/25/2022]
Abstract
The present study examined the comparative expression and secretory profile of vital signaling molecules in buffalo fetal fibroblasts (BFF) and Wharton's jelly (BWJ) feeder layers at different passages. Both feeder layers were expanded up to 8th passage. Signaling molecules viz. bone morphogenetic protein 4 (BMP4), fibroblast growth factor 2 (FGF2), leukemia inhibitory factor (LIF) and transforming growth factor beta 1 (TGFB1) and pluripotency-associated transcriptional factors (POU5F1, SOX2, NANOG, KLF4, MYC and FOXD3) were immunolocalized in the both feeder types. A clear variation in the expression pattern of key signaling molecules with passaging was registered in both feeders compared to primary culture (0 passage). The conditioned media (CM) was collected from different passages (2, 4, 6, 8) of both the feeder layers and was quantified using enzyme-linked immunosorbent assay (ELISA). Concomitant to expression profile, protein quantification also revealed differences in the concentration of signaling molecules at different time points. Conjointly, expression and secretory profile revealed that 2nd passage of BFF and 6th passage of BWJ exhibit optimal levels of key signaling molecules thus may be selected as best passages for embryonic stem cells (ESCs) propagation. Further, the effect of mitomycin-C (MMC) treatment on the expression profile of signaling molecules in the selected passages of BFF and BWJ revealed that MMC modulates the expression profile of these molecules. In conclusion, the results indicate that feeder layers vary in expression and secretory pattern of vital signaling molecules with passaging. Based on these findings, the appropriate feeder passages may be selected for the quality propagation of buffalo ESCs.
Collapse
Affiliation(s)
- Mehtab S Parmar
- Reproductive Physiology Laboratory, Division of Physiology and Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar-243 122, Bareilly, UP, India
| | - Smruti Ranjan Mishra
- Reproductive Physiology Laboratory, Division of Physiology and Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar-243 122, Bareilly, UP, India
| | - Anjali Somal
- Reproductive Physiology Laboratory, Division of Physiology and Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar-243 122, Bareilly, UP, India
| | - Sriti Pandey
- Reproductive Physiology Laboratory, Division of Physiology and Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar-243 122, Bareilly, UP, India
| | - G Sai Kumar
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar-243 122, Bareilly, UP, India
| | - Mihir Sarkar
- Reproductive Physiology Laboratory, Division of Physiology and Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar-243 122, Bareilly, UP, India
| | - Vikash Chandra
- Reproductive Physiology Laboratory, Division of Physiology and Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar-243 122, Bareilly, UP, India
| | - G Taru Sharma
- Reproductive Physiology Laboratory, Division of Physiology and Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar-243 122, Bareilly, UP, India.
| |
Collapse
|
8
|
Chiang CY, Liu SA, Sheu ML, Chen FC, Chen CJ, Su HL, Pan HC. Feasibility of Human Amniotic Fluid Derived Stem Cells in Alleviation of Neuropathic Pain in Chronic Constrictive Injury Nerve Model. PLoS One 2016; 11:e0159482. [PMID: 27441756 PMCID: PMC4956194 DOI: 10.1371/journal.pone.0159482] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 07/05/2016] [Indexed: 01/01/2023] Open
Abstract
Purpose The neurobehavior of neuropathic pain by chronic constriction injury (CCI) of sciatic nerve is very similar to that in humans, and it is accompanied by a profound local inflammation response. In this study, we assess the potentiality of human amniotic fluid derived mesenchymal stem cells (hAFMSCs) for alleviating the neuropathic pain in a chronic constriction nerve injury model. Methods and Methods This neuropathic pain animal model was conducted by four 3–0 chromic gut ligatures loosely ligated around the left sciatic nerve in Sprague—Dawley rats. The intravenous administration of hAFMSCs with 5x105 cells was conducted for three consecutive days. Results The expression IL-1β, TNF-α and synaptophysin in dorsal root ganglion cell culture was remarkably attenuated when co-cultured with hAFMSCs. The significant decrease of PGP 9.5 in the skin after CCI was restored by administration of hAFMSCs. Remarkably increased expression of CD 68 and TNF-α and decreased S-100 and neurofilament expression in injured nerve were rescued by hAFMSCs administration. Increases in synaptophysin and TNF-α over the dorsal root ganglion were attenuated by hAFMSCs. Significant expression of TNF-α and OX-42 over the dorsal spinal cord was substantially attenuated by hAFMSCs. The increased amplitude of sensory evoked potential as well as expression of synaptophysin and TNF-α expression was alleviated by hAFMSCs. Human AFMSCs significantly improved the threshold of mechanical allodynia and thermal hyperalgesia as well as various parameters of CatWalk XT gait analysis. Conclusion Human AFMSCs administration could alleviate the neuropathic pain demonstrated in histomorphological alteration and neurobehavior possibly through the modulation of the inflammatory response.
Collapse
Affiliation(s)
- Chien-Yi Chiang
- Institute of Biomedical Sciences, National Chung-Hsing University, Taichung, Taiwan
| | - Shih-An Liu
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Meei-Ling Sheu
- Institute of Biomedical Sciences, National Chung-Hsing University, Taichung, Taiwan
| | - Fu-Chou Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Hong-Lin Su
- Institute of Life Sciences, National Chung-Hsing University, Taichung, Taiwan
| | - Hung-Chuan Pan
- Department of Neurosurgery, Taichung Veterans General Hospital, Taichung, Taiwan
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
9
|
Verma V, Samanthapudi K, Raviprakash R. Classic Studies on the Potential of Stem Cell Neuroregeneration. JOURNAL OF THE HISTORY OF THE NEUROSCIENCES 2015; 25:123-141. [PMID: 26308908 DOI: 10.1080/0964704x.2015.1039904] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The 1990s and 2000s were the beginning of an exciting time period for developmental neuroscience and neural stem cell research. By better understanding brain plasticity and the birth of new neurons in the adult brain, contrary to established dogma, hope for therapy from devastating neurological diseases was generated. The potential for stem cells to provide functional recovery in humans remains to be further tested and to further move into the clinical trial realm. The future certainly has great promise on stem cells to assist in alleviation of difficult-to-treat neurologic disorders. This article reviews classic studies of the 1990s and 2000s that paved the way for the advances of today, which can in turn lead to tomorrow's therapies.
Collapse
Affiliation(s)
- Vivek Verma
- a Department of Neuroscience , University of Pittsburgh , Pittsburgh , PA , USA
| | | | - Ratujit Raviprakash
- a Department of Neuroscience , University of Pittsburgh , Pittsburgh , PA , USA
| |
Collapse
|
10
|
Selection of appropriate isolation method based on morphology of blastocyst for efficient derivation of buffalo embryonic stem cells. Cytotechnology 2013; 66:239-50. [PMID: 23553019 DOI: 10.1007/s10616-013-9561-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2012] [Accepted: 03/26/2013] [Indexed: 10/27/2022] Open
Abstract
The efficiency of embryonic stem cell (ESC) derivation from all species except for rodents and primates is very low. There are however, multiple interests in obtaining pluripotent cells from these animals with main expectations in the fields of transgenesis, cloning, regenerative medicine and tissue engineering. Researches are being carried out in laboratories throughout the world to increase the efficiency of ESC isolation for their downstream applications. Thus, the present study was undertaken to study the effect of different isolation methods based on the morphology of blastocyst for efficient derivation of buffalo ESCs. Embryos were produced in vitro through the procedures of maturation, fertilization and culture. Hatched blastocysts or isolated inner cell masses (ICMs) were seeded on mitomycin-C inactivated buffalo fetal fibroblast monolayer for the development of ESC colonies. The ESCs were analyzed for alkaline phosphatase activity, expression of pluripotency markers and karyotypic stability. Primary ESC colonies were obtained after 2-5 days of seeding hatched blastocysts or isolated ICMs on mitomycin-C inactivated feeder layer. Mechanically isolated ICMs attached and formed primary cell colonies more efficiently than ICMs isolated enzymatically. For derivation of ESCs from poorly defined ICMs intact hatched blastocyst culture was the most successful method. Results of this study implied that although ESCs can be obtained using all three methods used in this study, efficiency varies depending upon the morphology of blastocyst and isolation method used. So, appropriate isolation method must be selected depending on the quality of blastocyst for efficient derivation of ESCs.
Collapse
|
11
|
Muñetón-Gómez VC, Doncel-Pérez E, Fernandez AP, Serrano J, Pozo-Rodrigálvarez A, Vellosillo-Huerta L, Taylor JS, Cardona-Gómez GP, Nieto-Sampedro M, Martínez-Murillo R. Neural differentiation of transplanted neural stem cells in a rat model of striatal lacunar infarction: light and electron microscopic observations. Front Cell Neurosci 2012; 6:30. [PMID: 22876219 PMCID: PMC3410634 DOI: 10.3389/fncel.2012.00030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 07/12/2012] [Indexed: 12/14/2022] Open
Abstract
The increased risk and prevalence of lacunar stroke and Parkinson's disease (PD) makes the search for better experimental models an important requirement for translational research. In this study we assess ischemic damage of the nigrostriatal pathway in a model of lacunar stroke evoked by damaging the perforating arteries in the territory of the substantia nigra (SN) of the rat after stereotaxic administration of endothelin-1 (ET-1), a potent vasoconstrictor peptide. We hypothesized that transplantation of neural stem cells (NSCs) with the capacity of differentiating into diverse cell types such as neurons and glia, but with limited proliferation potential, would constitute an alternative and/or adjuvant therapy for lacunar stroke. These cells showed neuritogenic activity in vitro and a high potential for neural differentiation. Light and electron microscopy immunocytochemistry was used to characterize GFP-positive neurons derived from the transplants. 48 h after ET-1 injection, we characterized an area of selective degeneration of dopaminergic neurons within the nigrostriatal pathway characterized with tissue necrosis and glial scar formation, with subsequent behavioral signs of Parkinsonism. Light microscopy showed that grafted cells within the striatal infarction zone differentiated with a high yield into mature glial cells (GFAP-positive) and neuron types present in the normal striatum. Electron microscopy revealed that NSCs-derived neurons integrated into the host circuitry establishing synaptic contacts, mostly of the asymmetric type. Astrocytes were closely associated with normal small-sized blood vessels in the area of infarct, suggesting a possible role in the regulation of the blood brain barrier and angiogenesis. Our results encourage the use of NSCs as a cell-replacement therapy for the treatment of human vascular Parkinsonism.
Collapse
Affiliation(s)
- Vilma C Muñetón-Gómez
- Neurovascular Research Group, Department of Molecular, Cellular, and Developmental Neurobiology, Spanish Council for Scientific Research (CSIC), Instituto Cajal Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Kim HJ, Jin CY. Stem cells in drug screening for neurodegenerative disease. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2012; 16:1-9. [PMID: 22416213 PMCID: PMC3298819 DOI: 10.4196/kjpp.2012.16.1.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 01/20/2012] [Accepted: 01/25/2012] [Indexed: 12/26/2022]
Abstract
Because the average human life span has recently increased, the number of patients who are diagnosed with neurodegenerative diseases has escalated. Recent advances in stem cell research have given us access to unlimited numbers of multi-potent or pluripotent cells for screening for new drugs for neurodegenerative diseases. Neural stem cells (NSCs) are a good model with which to screen effective drugs that increase neurogenesis. Recent technologies for human embryonic stem cells (ESCs) or induced pluripotent stem cells (iPSCs) can provide human cells that harbour specific neurodegenerative disease. This article discusses the use of NSCs, ESCs and iPSCs for neurodegenerative drug screening and toxicity evaluation. In addition, we introduce drugs or natural products that are recently identified to affect the stem cell fate to generate neurons or glia.
Collapse
Affiliation(s)
- Hyun-Jung Kim
- Laboratory of Stem Cell and Molecular Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 156-756, Korea
| | | |
Collapse
|
13
|
Siniscalco D, Giordano C, Galderisi U, Luongo L, de Novellis V, Rossi F, Maione S. Long-lasting effects of human mesenchymal stem cell systemic administration on pain-like behaviors, cellular, and biomolecular modifications in neuropathic mice. Front Integr Neurosci 2011. [PMID: 22164136 DOI: 10.3389/fnint.2011.00079.ecollection] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Neuropathic pain (NP) is an incurable disease caused by a primary lesion in the nervous system. NP is a progressive nervous system disease that results from poorly defined neurophysiological and neurochemical changes. Its treatment is very difficult. Current available therapeutic drugs have a generalized nature, sometime acting only on the temporal pain properties rather than targeting the several mechanisms underlying the generation and propagation of pain. METHODS Using biomolecular and immunohistochemical methods, we investigated the effect of the systemic injection of human mesenchymal stem cells (hMSCs) on NP relief. We used the spared nerve injury (SNI) model of NP in the mouse. hMSCs were injected into the tail vein of the mouse. Stem cell injection was performed 4 days after sciatic nerve surgery. Neuropathic mice were monitored every 10 days starting from day 11 until 90 days after surgery. RESULTS hMSCs were able to reduce pain-like behaviors, such as mechanical allodynia and thermal hyperalgesia, once injected into the tail vein. An anti-nociceptive effect was detectable from day 11 post surgery (7 days post cell injection). hMSCs were mainly able to home in the spinal cord and pre-frontal cortex of neuropathic mice. Injected hMSCs reduced the protein levels of the mouse pro-inflammatory interleukin IL-1β and IL-17 and increased protein levels of the mouse anti-inflammatory interleukin IL-10, and the marker of alternatively activated macrophages CD106 in the spinal cord of SNI mice. CONCLUSION As a potential mechanism of action of hMSCs in reducing pain, we suggest that they could exert their beneficial action through a restorative mechanism involving: (i) a cell-to-cell contact activation mechanism, through which spinal cord homed hMSCs are responsible for switching pro-inflammatory macrophages to anti-inflammatory macrophages; (ii) secretion of a broad spectrum of molecules to communicate with other cell types. This study could provide novel findings in MSC pre-clinical biology and their therapeutic potential in regenerative medicine.
Collapse
Affiliation(s)
- Dario Siniscalco
- Division of Pharmacology "L. Donatelli," Department of Experimental Medicine, Second University of Naples Naples, Italy
| | | | | | | | | | | | | |
Collapse
|
14
|
Siniscalco D, Giordano C, Galderisi U, Luongo L, de Novellis V, Rossi F, Maione S. Long-lasting effects of human mesenchymal stem cell systemic administration on pain-like behaviors, cellular, and biomolecular modifications in neuropathic mice. Front Integr Neurosci 2011; 5:79. [PMID: 22164136 PMCID: PMC3230031 DOI: 10.3389/fnint.2011.00079] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Accepted: 11/16/2011] [Indexed: 12/24/2022] Open
Abstract
Background: Neuropathic pain (NP) is an incurable disease caused by a primary lesion in the nervous system. NP is a progressive nervous system disease that results from poorly defined neurophysiological and neurochemical changes. Its treatment is very difficult. Current available therapeutic drugs have a generalized nature, sometime acting only on the temporal pain properties rather than targeting the several mechanisms underlying the generation and propagation of pain. Methods: Using biomolecular and immunohistochemical methods, we investigated the effect of the systemic injection of human mesenchymal stem cells (hMSCs) on NP relief. We used the spared nerve injury (SNI) model of NP in the mouse. hMSCs were injected into the tail vein of the mouse. Stem cell injection was performed 4 days after sciatic nerve surgery. Neuropathic mice were monitored every 10 days starting from day 11 until 90 days after surgery. Results: hMSCs were able to reduce pain-like behaviors, such as mechanical allodynia and thermal hyperalgesia, once injected into the tail vein. An anti-nociceptive effect was detectable from day 11 post surgery (7 days post cell injection). hMSCs were mainly able to home in the spinal cord and pre-frontal cortex of neuropathic mice. Injected hMSCs reduced the protein levels of the mouse pro-inflammatory interleukin IL-1β and IL-17 and increased protein levels of the mouse anti-inflammatory interleukin IL-10, and the marker of alternatively activated macrophages CD106 in the spinal cord of SNI mice. Conclusion: As a potential mechanism of action of hMSCs in reducing pain, we suggest that they could exert their beneficial action through a restorative mechanism involving: (i) a cell-to-cell contact activation mechanism, through which spinal cord homed hMSCs are responsible for switching pro-inflammatory macrophages to anti-inflammatory macrophages; (ii) secretion of a broad spectrum of molecules to communicate with other cell types. This study could provide novel findings in MSC pre-clinical biology and their therapeutic potential in regenerative medicine.
Collapse
Affiliation(s)
- Dario Siniscalco
- Division of Pharmacology "L. Donatelli," Department of Experimental Medicine, Second University of Naples Naples, Italy
| | | | | | | | | | | | | |
Collapse
|
15
|
Patkar S, Tate R, Modo M, Plevin R, Carswell HVO. Conditionally immortalised neural stem cells promote functional recovery and brain plasticity after transient focal cerebral ischaemia in mice. Stem Cell Res 2011; 8:14-25. [PMID: 22099017 DOI: 10.1016/j.scr.2011.07.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Revised: 05/27/2011] [Accepted: 07/19/2011] [Indexed: 11/27/2022] Open
Abstract
Cell therapy has enormous potential to restore neurological function after stroke. The present study investigated effects of conditionally immortalised neural stem cells (ciNSCs), the Maudsley hippocampal murine neural stem cell line clone 36 (MHP36), on sensorimotor and histological outcome in mice subjected to transient middle cerebral artery occlusion (MCAO). Adult male C57BL/6 mice underwent MCAO by intraluminal thread or sham surgery and MHP36 cells or vehicle were implanted into ipsilateral cortex and caudate 2 days later. Functional recovery was assessed for 28 days using cylinder and ladder rung tests and tissue analysed for plasticity, differentiation and infarct size. MHP36-implanted animals showed accelerated and augmented functional recovery and an increase in neurons (MAP-2), synaptic plasticity (synaptophysin) and axonal projections (GAP-43) but no difference in astrocytes (GFAP), oligodendrocytes (CNPase), microglia (IBA-1) or lesion volumes when compared to vehicle group. This is the first study showing a potential functional benefit of the ciNSCs, MHP36, after focal MCAO in mice, which is probably mediated by promoting neuronal differentiation, synaptic plasticity and axonal projections and opens up opportunities for future exploitation of genetically altered mice for dissection of mechanisms of stem cell based therapy.
Collapse
Affiliation(s)
- Shalmali Patkar
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK
| | | | | | | | | |
Collapse
|
16
|
Markiewicz I, Sypecka J, Domanska-Janik K, Wyszomirski T, Lukomska B. Cellular environment directs differentiation of human umbilical cord blood-derived neural stem cells in vitro. J Histochem Cytochem 2011; 59:289-301. [PMID: 21378283 DOI: 10.1369/0022155410397997] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cord blood-derived neural stem cells (NSCs) are proposed as an alternative cell source to repair brain damage upon transplantation. However, there is a lack of data showing how these cells are driven to generate desired phenotypes by recipient nervous tissue. Previous research indicates that local environment provides signals driving the fate of stem cells. To investigate the impact of these local cues interaction, the authors used a model of cord blood-derived NSCs co-cultured with different rat brain-specific primary cultures, creating the neural-like microenvironment conditions in vitro. Neuronal and astro-, oligo-, and microglia cell cultures were obtained by the previously described methods. The CMFDA-labeled neural stem cells originated from, non-transformed human umbilical cord blood cell line (HUCB-NSCs) established in a laboratory. The authors show that the close vicinity of astrocytes and oligodendrocytes promotes neuronal differentiation of HUCB-NSCs, whereas postmitotic neurons induce oligodendrogliogenesis of these cells. In turn, microglia or endothelial cells do not favor any phenotypes of their neural commitment. Studies have confirmed that HUCB-NSCs can read cues from the neurogenic microenvironment, attaining features of neurons, astrocytes, or oligodendrocytes. The specific responses of neurally committed cord blood-derived cells, reported in this work, are very much similar to those described previously for NSCs derived from other "more typical" sources. This further proves their genuine neural nature. Apart from having a better insight into the neurogenesis in the adult brain, these findings might be important when predicting cord blood cell derivative behavior after their transplantation for neurological disorders.
Collapse
Affiliation(s)
- Inga Markiewicz
- Neurorepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | | | | | | | | |
Collapse
|
17
|
Siniscalco D, Giordano C, Galderisi U, Luongo L, Alessio N, Di Bernardo G, de Novellis V, Rossi F, Maione S. Intra-brain microinjection of human mesenchymal stem cells decreases allodynia in neuropathic mice. Cell Mol Life Sci 2010; 67:655-69. [PMID: 19937263 PMCID: PMC11115751 DOI: 10.1007/s00018-009-0202-4] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2009] [Revised: 10/30/2009] [Accepted: 11/03/2009] [Indexed: 12/16/2022]
Abstract
Neuropathic pain is a very complex disease, involving several molecular pathways. Current available drugs are usually not acting on the several mechanisms underlying the generation and propagation of pain. We used spared nerve injury model of neuropathic pain to assess the possible use of human mesenchymal stem cells (hMSCs) as anti-neuropathic tool. Human MSCs were transplanted in the mouse lateral cerebral ventricle. Stem cells injection was performed 4 days after sciatic nerve surgery. Neuropathic mice were monitored 7, 10, 14, 17, and 21 days after surgery. hMSCs were able to reduce pain-like behaviors, such as mechanical allodynia and thermal hyperalgesia, once transplanted in cerebral ventricle. Anti-nociceptive effect was detectable from day 10 after surgery (6 days post cell injection). Human MSCs reduced the mRNA levels of the pro-inflammatory interleukin IL-1beta mouse gene, as well as the neural beta-galactosidase over-activation in prefrontal cortex of SNI mice. Transplanted hMSCs were able to reduce astrocytic and microglial cell activation.
Collapse
Affiliation(s)
- Dario Siniscalco
- Division of Pharmacology L. Donatelli, Department of Experimental Medicine, Second University of Naples, 80138 Naples, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
Systemic neurotransplantation (SNT) was introduced in the laboratory in 2000 and currently it is being widely examined in animal models of neurological disorders. The aim of this systematic review was to evaluate the current state of knowledge in the field of experimental SNT and the premise for the introduction of clinical trials. PubMed was searched and 60 articles utilizing an SNT approach were found and subjected to analysis. The time window for cell transplantation was addressed in only two studies, with contradictory results. Immunosuppression was applied in 25% of studies. No study addressed the justification for immunosuppression. Bone marrow was the most frequent source of grafted cells, followed by cord blood and then by cells of embryonic origin. Studies investigating dose-dependency revealed no satisfactory results with transplantation of less than 10(6) cells/animal; the efficient dose most frequently ranged from 10(6)-10(7) cells/animal (mice and rats). The behavioral effects of cell transplantation were assessed in 75% of all studies; significant improvement was achieved in 95% of them. Morphological effect was evaluated in half of the studies; significant positive effect was achieved in 73% of them. Experimental attempts to elucidate the mechanisms mediating cell-dependent effect were not undertaken in half of the studies. In the other half, the most frequent mechanisms were growth factors, neurogenesis and immunomodulation. SNT still seems to be at the very initial stage of development. Many critical factors have not been sufficiently addressed in laboratory studies and they must be clarified before the introduction of clinical trials.
Collapse
Affiliation(s)
- Miroslaw Janowski
- Department of NeuroRepair, Medical Research Center, Polish Academy of Science, Warsaw, Poland.
| | | |
Collapse
|
19
|
Thomas M, Tyers P, Lazic SE, Barker RA, Beazley L, Ziman M. Graft outcomes influenced by co-expression of Pax7 in graft and host tissue. J Anat 2009; 214:396-405. [PMID: 19245506 PMCID: PMC2673790 DOI: 10.1111/j.1469-7580.2009.01049.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2008] [Indexed: 11/26/2022] Open
Abstract
Cell replacement therapies offer promise in the treatment of neurotrauma and neurodegenerative disorders and have concentrated on the use of primary fetal brain tissue. However, there is a growing promise of using neural stem cells, in which case other factors may be important in their successful engraftment. We therefore investigated whether the co-expression of the major developmental transcription factor (Pax7 in this study) of donor tissue to graft site influences transplant survival and differentiation in the rat midbrain. Neural progenitor cells were prepared from either the Pax7-expressing dorsal (DM) or non-Pax7-expressing ventral mesencephalon (VM) of embryonic EGFP(+/+) rats. Cells were dissociated and grafted into the adult rat superior colliculus (SC) lesioned with quinolinic acid 3 days previously, a time shown to be associated with the up-regulation of Pax7. Grafts were then examined 4 weeks later. Our results suggest the origin of the graft tissue did not alter graft survival in the SC; however, dorsal grafts appear to have a higher incidence of neuronal survival, whereas ventral grafts have a higher incidence of astrocytic survivors.
Collapse
Affiliation(s)
- Meghan Thomas
- School of Biomedical Science, Edith Cowan University, Joondalup DriveWestern Australia, Australia
- School of Surgery and PathologyPerth, Australia
- School of Animal Biology, University of Western AustraliaPerth, Australia
| | - Pam Tyers
- Centre for Brain Repair, Cambridge UniversityCambridge, UK
| | | | - Roger A Barker
- School of Biomedical Science, Edith Cowan University, Joondalup DriveWestern Australia, Australia
- Centre for Brain Repair, Cambridge UniversityCambridge, UK
| | - Lyn Beazley
- School of Animal Biology, University of Western AustraliaPerth, Australia
- Western Australian Institute for Medical Research, University of Western AustraliaPerth, Australia
| | - Mel Ziman
- School of Biomedical Science, Edith Cowan University, Joondalup DriveWestern Australia, Australia
- School of Surgery and PathologyPerth, Australia
| |
Collapse
|
20
|
Lysosulfatide Regulates the Motility of a Neural Precursor Cell Line Via Calcium-mediated Process Collapse. Neurochem Res 2008; 34:508-17. [DOI: 10.1007/s11064-008-9813-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2008] [Accepted: 07/17/2008] [Indexed: 11/26/2022]
|
21
|
Yamaguchi H, Kidachi Y, Umetsu H, Ryoyama K. Differentiation of serum-free mouse embryo cells into an astrocytic lineage is associated with the asymmetric production of early neural, neuronal and glial markers. Biol Pharm Bull 2008; 31:1008-12. [PMID: 18451536 DOI: 10.1248/bpb.31.1008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Serum-free mouse embryo (SFME) cells, the astrocyte progenitor cells in the central nervous system (CNS), were exposed to 10 ng/ml leukemia inhibitory factor (LIF) and 10 ng/ml bone morphogenic protein 2 (BMP2) to induce differentiation, and expression of cell-type specific markers. Nestin, a marker of early neural lineage, betaIII-tubulin, a marker of neuronal lineage, oligodendrocyte marker O4 (O4), a marker of oligodendrocytic lineage and glial fibrillary acidic protein (GFAP), a marker of astrocytic lineage, were analyzed. Characteristics of SFME cells, as a CNS progenitor, were identified and a possible mechanism, underlying SFME cell specification into an astrocytic lineage upon differentiation, was investigated. These markers were present, both at the initial proliferative phase and after induction of differentiation. GFAP expression increased strongly upon differentiation, while expression of the other markers changed very little. These results indicate that astrocytic differentiation is associated with the asymmetric production of these markers, rather than through induction of astrocytic markers.
Collapse
Affiliation(s)
- Hideaki Yamaguchi
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Aomori University, Aomori University; 2-3-1 Kobata, Aomori 030-0943, Japan.
| | | | | | | |
Collapse
|
22
|
Walczak P, Zhang J, Gilad AA, Kedziorek DA, Ruiz-Cabello J, Young RG, Pittenger MF, van Zijl PCM, Huang J, Bulte JWM. Dual-modality monitoring of targeted intraarterial delivery of mesenchymal stem cells after transient ischemia. Stroke 2008; 39:1569-74. [PMID: 18323495 DOI: 10.1161/strokeaha.107.502047] [Citation(s) in RCA: 294] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND PURPOSE In animal models of stroke, functional improvement has been obtained after stem cell transplantation. Successful therapy depends largely on achieving a robust and targeted cell engraftment, with intraarterial (IA) injection being a potentially attractive route of administration. We assessed the suitability of laser Doppler flow (LDF) signal measurements and magnetic resonance (MR) imaging for noninvasive dual monitoring of targeted IA cell delivery. METHODS Transient cerebral ischemia was induced in adult Wistar rats (n=25) followed by IA or intravenous (IV) injection of mesenchymal stem cells (MSCs) labeled with superparamagnetic iron oxide. Cell infusion was monitored in real time with transcranial laser Doppler flowmetry while cellular delivery was assessed with MRI in vivo (4.7 T) and ex vivo (9.4 T). RESULTS Successful delivery of magnetically labeled MSCs could be readily visualized with MRI after IA but not IV injection. IA stem cell injection during acute stroke resulted in a high variability of cerebral engraftment. The amount of LDF reduction during cell infusion (up to 80%) was found to correlate well with the degree of intracerebral engraftment, with low LDF values being associated with significant morbidity. CONCLUSIONS High cerebral engraftment rates are associated with impeded cerebral blood flow. Noninvasive dual-modality imaging enables monitoring of targeted cell delivery, and through interactive adjustment may improve the safety and efficacy of stem cell therapy.
Collapse
Affiliation(s)
- Piotr Walczak
- Division of MR Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 217 Traylor, 720 Rutland Ave, Baltimore, MD 21205-2195, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Rieske P, Azizi SA, Augelli B, Gaughan J, Krynska B. A population of human brain parenchymal cells express markers of glial, neuronal and early neural cells and differentiate into cells of neuronal and glial lineages. Eur J Neurosci 2007; 25:31-7. [PMID: 17241264 DOI: 10.1111/j.1460-9568.2006.05254.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Glial fibrillary acidic protein (GFAP)-positive cells derived from the neurogenic areas of the brain can be stem/progenitor cells and give rise to new neurons in vitro and in vivo. We report here that a population of GFAP-positive cells derived from fetal human brain parenchyma coexpress markers of early neural and neuronal cells, and have neural progenitor cell characteristics. We used a monolayer culture system to expend and differentiate these cells. During the initial proliferative phase, all cells expressed GFAP, nestin and low levels of betaIII-tubulin. When these cells were cultured in serum and then basic fibroblast growth factor, they generated two distinct progenies: (i) betaIII-tubulin- and nestin-positive cells and (ii) GFAP- and nestin-positive cells. These cells, when subsequently cultured in serum-free media without growth factors, ceased to proliferate and differentiated into two major neural cell classes, neurons and glia. In the cells of neuronal lineage, nestin expression was down-regulated and betaIII-tubulin expression became robust. Cells of glial lineage differentiated by down-regulating nestin expression and up-regulating GFAP expression. These data suggest that populations of parenchymal brain cells, initially expressing both glial and neuronal markers, are capable of differentiating into single neuronal and glial lineages through asymmetric regulation of gene expression in these cells, rather than acquiring markers through differentiation.
Collapse
Affiliation(s)
- Piotr Rieske
- Department of Neurology, Temple University School of Medicine, 3401 N Broad St, Philldelphia, PA 19140, USA
| | | | | | | | | |
Collapse
|
24
|
Abstract
In spite of the commonly held belief that ‘the brain does not regenerate’, it is now accepted that postnatal neurogenesis does occur. Thus, one wonders whether cellular-replacement therapy might be used to heal the brain in diseases caused by neuronal cell loss. The existence of neural stem cells has been demonstrated by many scientists and is now generally accepted. The exact role of these cells, how their numbers are regulated and how they participate in CNS and spinal cord regeneration in postnatal life are still not well known. There are many reviews summarizing work on these cells; consequently, I will focus instead on other cells that may participate in postnatal neurogenesis: bone marrow-derived stem cells. The possibility that bone marrow-derived stem cells populate the CNS and differentiate into various neural elements is certainly not universally accepted.
Collapse
Affiliation(s)
- Eva Mezey
- CSDB, NIDCR, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|