51
|
Bexell D, Svensson A, Bengzon J. Stem cell-based therapy for malignant glioma. Cancer Treat Rev 2012; 39:358-65. [PMID: 22795538 DOI: 10.1016/j.ctrv.2012.06.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 06/13/2012] [Accepted: 06/17/2012] [Indexed: 12/25/2022]
Abstract
Stem cells have been extensively investigated as tumour-tropic vectors for gene delivery to solid tumours. In this review, we discuss the potential for using stem cells as cellular vector systems in gene therapy for malignant gliomas, with a focus on neural stem cells, and multipotent mesenchymal stromal cells. Tumour cell-derived substances and factors associated with tumour-induced inflammation and tumour neovascularisation can specifically attract stem cells to invasive gliomas. Injected stem cells engineered to produce anti-tumour substances have shown strong therapeutic effects in experimental glioma models. However, the potential caveats include the immunosuppressive functions of multipotent mesenchymal stromal cells, the contribution of stem cells to the pro-tumourigenic stroma, and the malignant transformation of implanted stem cells. In addition, it is not yet known which stem cell types and therapeutic genes will be most effective for the treatment of glioma patients. Here, we highlight the possibilities and problems for translating promising experimental findings in glioma models into the clinic.
Collapse
Affiliation(s)
- Daniel Bexell
- Lund Stem Cell Center, BMC B10, Lund University, Lund, Sweden; Molecular Medicine, Center for Molecular Pathology, Lund University, Skåne University Hospital, Malmö, Sweden.
| | | | | |
Collapse
|
52
|
He W, Qu T, Yu Q, Wang Z, Lv H, Zhang J, Zhao X, Wang P. LPS induces IL-8 expression through TLR4, MyD88, NF-kappaB and MAPK pathways in human dental pulp stem cells. Int Endod J 2012; 46:128-36. [PMID: 22788664 DOI: 10.1111/j.1365-2591.2012.02096.x] [Citation(s) in RCA: 132] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 05/29/2012] [Indexed: 01/09/2023]
Abstract
AIM To evaluate the effects of lipopolysaccharide (LPS) on interleukin-8 (IL-8) and related intracellular signalling pathways in human dental pulp stem cells (hDPSCs). METHODOLOGY Human pulp tissues were isolated from human impacted third molars, and the hDPSCs were cultured and characterized. The effects of LPS on IL-8 and Toll-like receptor 4 (TLR4) gene expression in hDPSCs were investigated using real-time quantitative RT-PCR and ELISA. Whether TLR4/MyD88/NF-кB was involved in the LPS-induced up-regulation of IL-8 in hDPSCs was determined using transient transfection, luciferase assay and ELISA. The involvement of MAPKs in the LPS-induced up-regulation of IL-8 in hDPSCs was investigated via transient transfection, luciferase assay, ELISA and western blot. The data were statistically analysed using Student's t-test or one-way anova followed by the Student-Neumann-Keuls test. RESULTS Cells exposed to LPS not only displayed an enhanced expression of TLR4 but also showed an elevated IL-8 gene expression; exposure to LPS also resulted in the induction of IL-8 gene transcription via promoter activation. The LPS-induced IL-8 promoter activation was inhibited through dominant-negative mutations in TLR4 and MyD88, but not in TLR2. The LPS-induced IL-8 protein release was attenuated through the administration of TLR4-neutralizing antibody or MyD88 inhibitory peptide and a dominant-negative mutation in IκBα. In contrast, IL-8 protein release was enhanced through the expression of NF-κB p65. Treatment with PDTC, TPCK or Bay117082 effectively antagonized LPS-induced IL-8 protein release. Moreover, both the promoter activity and the LPS-induced release of IL-8 were diminished upon the administration of U0126 and SB203580, but not SP600125. Moreover, the exposure to LPS activated ERK1/2 and p38 MAPK phosphorylation in cells. CONCLUSIONS This study reports the LPS-mediated transcriptional and post-translational up-regulation of IL-8, which is a process that also involves TLR4, MyD88, NF-κB and MAPK.
Collapse
Affiliation(s)
- W He
- Department of Operative Dentistry and Endodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, China.
| | | | | | | | | | | | | | | |
Collapse
|
53
|
Motaln H, Gruden K, Hren M, Schichor C, Primon M, Rotter A, Lah TT. Human Mesenchymal Stem Cells Exploit the Immune Response Mediating Chemokines to Impact the Phenotype of Glioblastoma. Cell Transplant 2012; 21:1529-45. [DOI: 10.3727/096368912x640547] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
In contrast to the application of human mesenchymal stem cells (hMSCs) in regenerative medicine, only a limited number of studies are addressing their use in anticancer therapy. As the latter may represent a new hope to improve the survival of patients with glioblastoma multiformae (GBM), the most common and malignant form of the brain tumors, we aimed to investigate the interactions of hMSCs and GBM cells under in vitro conditions. Four hMSC clones and three different GBM cell lines were used to study their mutual paracrine interactions in cocultures compared to their monocultures, where cells were grown under the same experimental conditions. The effects on cell growth, proliferation, and invasion in Matrigel were quantified. Further, bioinformatics tools were used to relate these results to the data obtained from cytokine macroarrays and cDNA microarrays that revealed proteins and genes significantly involved in cellular cross-talk. We showed that hMSCs are responsible for the impairment of GBM cell invasion and growth, possibly via induction of their senescence. On the other hand, GBM cells inversely affected some of these characteristics in hMSCs. We found CCL2/MCP-1 to be the most significantly regulated chemokine during hMSC and U87-MG paracrine signaling in addition to several chemokines that may account for changed cocultured cells' phenotype by affecting genes associated with proliferation ( Pmepa-1, NF-κ B, IL-6, IL-1b), invasion ( EphB2, Sod2, Pcdh18, Col7A1, Gja1, Mmp1/2), and senescence ( Kiaa1199, SerpinB2). As we functionally confirmed the role of CCL2/MCP-1 in GBM cell invasion we thereby propose a novel mechanism of CCL2/MCP-1 antimigratory effects on GBM cells, distinct from its immunomodulatory role. Significant alterations of GBM phenotype in the presence of hMSCs should encourage the studies on the naive hMSC use for GBM treatment.
Collapse
Affiliation(s)
- Helena Motaln
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Kristina Gruden
- Department of Biotechnology and Systems Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Matjaž Hren
- Department of Biotechnology and Systems Biology, National Institute of Biology, Ljubljana, Slovenia
- Bioinstrumentation Laboratory, Centre of Excellence for Biosensors, Instrumentation and Process Control, Solkan, Slovenia
| | - Christian Schichor
- Tumorbiological Laboratory, Neurosurgical Department, Ludwig-Maximilians-University, Munich, Germany
| | - Monika Primon
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Ana Rotter
- Department of Biotechnology and Systems Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Tamara T. Lah
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| |
Collapse
|
54
|
Velpula KK, Dasari VR, Rao JS. The homing of human cord blood stem cells to sites of inflammation: unfolding mysteries of a novel therapeutic paradigm for glioblastoma multiforme. Cell Cycle 2012; 11:2303-13. [PMID: 22684297 DOI: 10.4161/cc.20766] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Efficient homing of human umbilical cord blood mesenchymal stem cells (hUCBSC) to inflammation sites is crucial for therapeutic use. In glioblastoma multiforme, soluble factors released by the tumor facilitate the migratory capacity of mesenchymal stem cells toward glioma cells. These factors include chemokines and growth inducers. Nonetheless, the mechanistic details of these factors involved in hUCBSC homing have not been clearly delineated. The present study is aimed to deduce specific factors involved in hUCBSC homing by utilizing a glioma stem cell-induced inflammatory lesion model in the mouse brain. Our results show that hUCBSC do not form tumors in athymic nude mice brains and do not elicit immune responses in immunocompetent SKH1 mice. Further, hUCBSC spheroids migrate and invade glioma spheroids, while no effect was observed on rat fetal brain aggregates. Several cytokines, including GRO, MCP-1, IL-8, IL-3, IL-10, Osteopontin and TGF-β2, were constitutively secreted in the naive hUCBSC-conditioned medium, while significant increases of IL-8, GRO, GRO-α, MCP-1 and MCP-2 were observed in glioma stem cell-challenged hUCBSC culture filtrates. Furthermore, hUCBSC showed a stronger migration capacity toward glioma stem cells in vitro and exhibited enhanced migration to glioma stem cells in an intracranial human malignant glioma xenograft model. Our results indicate that multiple cytokines are involved in recruitment of hUCBSC toward glioma stem cells, and that hUCBSC are a potential candidate for glioma therapy.
Collapse
Affiliation(s)
- Kiran Kumar Velpula
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | | | | |
Collapse
|
55
|
A review of the role of stem cells in the development and treatment of glioma. Acta Neurochir (Wien) 2012; 154:951-69; discussion 969. [PMID: 22527576 DOI: 10.1007/s00701-012-1338-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 03/16/2012] [Indexed: 12/21/2022]
Abstract
The neurosurgical management of patients with intrinsic glial cancers is one of the most rapidly evolving areas of practice. This has been fuelled by advances in surgical technique not only in cytoreduction but also in drug delivery. Further innovation will depend on a deeper understanding of the biology of the disease and an appreciation of the limitations of current knowledge. Here we review the controversial topic of cancer stem cells applied to glioma to provide neurosurgeons with a working overview. It is now recognised that the adult human brain contains regionally specified cell populations capable of self-renewal that may contribute to tumour growth and maintenance following accumulated mutational change. Tumour cells adapted to maintain growth demonstrate some stem-like characteristics and as such constitute a legitimate therapeutic target. Cellular reprogramming technologies raise the potential of developing stem cells as novel surgical tools to target disease and possibly ameliorate some of the consequences of treatment. Achieving these goals remains a significant challenge to neurosurgical oncologists, not least in challenging how we think about treating brain cancer. This review will briefly examine our understanding of adult stem cells within the brain, the evidence that they contribute to the development of brain tumours as tumour-initiating cells, and the potential implications for therapy. It will also look at the role stem cells may play in the future management of glioma.
Collapse
|
56
|
Kim JY, Kim DH, Kim JH, Lee D, Jeon HB, Kwon SJ, Kim SM, Yoo YJ, Lee EH, Choi SJ, Seo SW, Lee JI, Na DL, Yang YS, Oh W, Chang JW. Soluble intracellular adhesion molecule-1 secreted by human umbilical cord blood-derived mesenchymal stem cell reduces amyloid-β plaques. Cell Death Differ 2011; 19:680-91. [PMID: 22015609 PMCID: PMC3307982 DOI: 10.1038/cdd.2011.140] [Citation(s) in RCA: 147] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Presently, co-culture of human umbilical cord blood mesenchymal stem cells (hUCB-MSCs) with BV2 microglia under amyloid-β42 (Aβ42) exposure induced a reduction of Aβ42 in the medium as well as an overexpression of the Aβ-degrading enzyme neprilysin (NEP) in microglia. Cytokine array examinations of co-cultured media revealed elevated release of soluble intracellular adhesion molecule-1 (sICAM-1) from hUCB-MSCs. Administration of human recombinant ICAM-1 in BV2 cells and wild-type mice brains induced NEP expression in time- and dose-dependent manners. In co-culturing with BV2 cells under Aβ42 exposure, knockdown of ICAM-1 expression on hUCB-MSCs by small interfering RNA (siRNA) abolished the induction of NEP in BV2 cells as well as reduction of added Aβ42 in the co-cultured media. By contrast, siRNA-mediated inhibition of the sICAM-1 receptor, lymphocyte function-associated antigen-1 (LFA-1), on BV2 cells reduced NEP expression by ICAM-1 exposure. When hUCB-MSCs were transplanted into the hippocampus of a 10-month-old transgenic mouse model of Alzheimer's disease for 10, 20, or 40 days, NEP expression was increased in the mice brains. Moreover, Aβ42 plaques in the hippocampus and other regions were decreased by active migration of hUCB-MSCs toward Aβ deposits. These data suggest that hUCB-MSC-derived sICAM-1 decreases Aβ plaques by inducing NEP expression in microglia through the sICAM-1/LFA-1 signaling pathway.
Collapse
Affiliation(s)
- J-Y Kim
- Biomedical Research Institute, MEDIPOST Co. Ltd., Seoul 137-874, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
57
|
Piperi C, Samaras V, Levidou G, Kavantzas N, Boviatsis E, Petraki K, Grivas A, Barbatis C, Varsos V, Patsouris E, Korkolopoulou P. Prognostic significance of IL-8-STAT-3 pathway in astrocytomas: Correlation with IL-6, VEGF and microvessel morphometry. Cytokine 2011; 55:387-95. [DOI: 10.1016/j.cyto.2011.05.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Accepted: 05/16/2011] [Indexed: 01/07/2023]
|
58
|
Ryu CH, Park SH, Park SA, Kim SM, Lim JY, Jeong CH, Yoon WS, Oh WI, Sung YC, Jeun SS. Gene Therapy of Intracranial Glioma Using Interleukin 12–Secreting Human Umbilical Cord Blood–Derived Mesenchymal Stem Cells. Hum Gene Ther 2011; 22:733-43. [DOI: 10.1089/hum.2010.187] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Chung Heon Ryu
- Department of Biomedical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sang-Hoon Park
- Division of Molecular and Life Sciences, Pohang University of Science and Technology, Pohang, Korea
| | - Soon A Park
- Department of Biomedical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seong Muk Kim
- Department of Biomedical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jung Yeon Lim
- Department of Biomedical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chang Hyun Jeong
- Department of Biomedical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Wan-Soo Yoon
- Department of Neurosurgery, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Won-il Oh
- Medipost Biomedical Research Institute, MEDIPOST Co., Ltd., Seoul, Korea
| | - Young Chul Sung
- Division of Molecular and Life Sciences, Pohang University of Science and Technology, Pohang, Korea
| | - Sin-Soo Jeun
- Department of Biomedical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Neurosurgery, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
59
|
Human amniotic membrane derived-mesenchymal stem cells induce C6 glioma apoptosis in vivo through the Bcl-2/caspase pathways. Mol Biol Rep 2011; 39:467-73. [PMID: 21556762 DOI: 10.1007/s11033-011-0760-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Accepted: 04/27/2011] [Indexed: 12/11/2022]
Abstract
High-grade gliomas are difficult to treat. We examined the therapeutic effect of intratumoral administration of human amniotic membrane derived-mesenchymal stem cells (hAMCs) on the growth of gliomas. Tumor volume of the control group was 1632±316 mm3 on day 30, and the group treated with a single intratumoral dose of hAMCs had a tumor volume of 1128±269 mm3 (P<0.05). Thus, administration of hAMCs significantly reduced tumor size. In rat glioma tissues treated with single and multiple dosages of hAMCs, there was a reduction in tumor volume of approximately 30.9 and 49.5%, respectively. We further evaluated the glioma tissue using Western blotting analysis and observed that the expression of Bax, caspase-8 and caspase-3 were greatly increased and the expression of Bcl-2 was greatly decreased in tumors treated with hAMCs. Sections of nude mice treated with hAMCs clearly showed the presence of an increase in apoptotic cells. The data collected herein confirms for the first time that hAMCs can inhibit C6 glioma growth and induce apoptosis of C6 gliomas in vivo. This demonstrates that hAMCs are a potential new therapeutic agent for the treatment of gliomas.
Collapse
|
60
|
Kim SM, Oh JH, Park SA, Ryu CH, Lim JY, Kim DS, Chang JW, Oh W, Jeun SS. Irradiation enhances the tumor tropism and therapeutic potential of tumor necrosis factor-related apoptosis-inducing ligand-secreting human umbilical cord blood-derived mesenchymal stem cells in glioma therapy. Stem Cells 2011; 28:2217-28. [PMID: 20945331 DOI: 10.1002/stem.543] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Irradiation is a standard therapy for gliomas and many other cancers. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is one of the most promising candidates for cancer gene therapy. Here, we show that tumor irradiation enhances the tumor tropism of human umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs) and the therapeutic effect of TRAIL delivered by UCB-MSCs. The sequential treatment with irradiation followed by TRAIL-secreting UCB-MSCs (MSC-TRAIL) synergistically enhanced apoptosis in either TRAIL-sensitive or TRAIL-resistant glioma cells by upregulating the death receptor 5 and by inducing caspase activation. Migration assays showed greater MSC migration toward irradiated glioma cells and the tumor site in glioma-bearing mice compared with unirradiated tumors. Irradiated glioma cells had increased expression of interleukin-8 (IL-8), which leads to the upregulation of the IL-8 receptor on MSCs. This upregulation, which is involved in the migratory capacity of UCB-MSCs, was confirmed by siRNA inhibition and an antibody-neutralizing assay. In vivo survival experiments in orthotopic xenografted mice showed that MSC-based TRAIL gene delivery to irradiated tumors had greater therapeutic efficacy than a single treatment. These results suggest that clinically relevant tumor irradiation increases the therapeutic efficacy of MSC-TRAIL by increasing tropism of MSCs and TRAIL-induced apoptosis, which may be a more useful strategy for cancer gene therapy.
Collapse
Affiliation(s)
- Seong Muk Kim
- Department of Biomedical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Kim SM, Kim DS, Jeong CH, Kim DH, Kim JH, Jeon HB, Kwon SJ, Jeun SS, Yang YS, Oh W, Chang JW. CXC chemokine receptor 1 enhances the ability of human umbilical cord blood-derived mesenchymal stem cells to migrate toward gliomas. Biochem Biophys Res Commun 2011; 407:741-6. [PMID: 21439934 DOI: 10.1016/j.bbrc.2011.03.093] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Accepted: 03/20/2011] [Indexed: 12/14/2022]
Abstract
In this study, we showed that knocking-down interleukin-8 (IL-8) in glioma cells, or its receptor, CXC chemokine receptor 1 (CXCR1) in hUCB-MSCs reduced hUCB-MSC migration toward glioma cells in a Transwell chamber. In contrast, CXCR1-transfected hUCB-MSCs (CXCR1-MSCs) showed a superior capacity to migrate toward glioma cells in a Transwell chamber compared to primary hUCB-MSCs. Furthermore, these transfected cells also demonstrated the same ability to migrate toward tumors in mice bearing intracranial human gliomas as shown by histological and in vivo imaging analysis. Our findings indicate that overexpression of CXCR1 could be a useful tool for MSC-based gene therapy to achieve a sufficient quantity of therapeutic MSCs that are localized within tumors.
Collapse
Affiliation(s)
- Seong Muk Kim
- Department of Neurosurgery, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Mesenchymal stem cells and progenitor cells in connective tissue engineering and regenerative medicine: is there a future for transplantation? Langenbecks Arch Surg 2011; 396:489-97. [PMID: 21373941 DOI: 10.1007/s00423-011-0762-2] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Accepted: 02/21/2011] [Indexed: 12/14/2022]
|
63
|
|
64
|
Dasari VR, Velpula KK, Kaur K, Fassett D, Klopfenstein JD, Dinh DH, Gujrati M, Rao JS. Cord blood stem cell-mediated induction of apoptosis in glioma downregulates X-linked inhibitor of apoptosis protein (XIAP). PLoS One 2010; 5:e11813. [PMID: 20676365 PMCID: PMC2911373 DOI: 10.1371/journal.pone.0011813] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2010] [Accepted: 06/27/2010] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND XIAP (X-linked inhibitor of apoptosis protein) is one of the most important members of the apoptosis inhibitor family. XIAP is upregulated in various malignancies, including human glioblastoma. It promotes invasion, metastasis, growth and survival of malignant cells. We hypothesized that downregulation of XIAP by human umbilical cord blood mesenchymal stem cells (hUCBSC) in glioma cells would cause them to undergo apoptotic death. METHODOLOGY/PRINCIPAL FINDINGS We observed the effect of hUCBSC on two malignant glioma cell lines (SNB19 and U251) and two glioma xenograft cell lines (4910 and 5310). In co-cultures of glioma cells with hUCBSC, proliferation of glioma cells was significantly inhibited. This is associated with increased cytotoxicity of glioma cells, which led to glioma cell death. Stem cells induced apoptosis in glioma cells, which was evaluated by TUNEL assay, FACS analyses and immunoblotting. The induction of apoptosis is associated with inhibition of XIAP in co-cultures of hUCBSC. Similar results were obtained by the treatment of glioma cells with shRNA to downregulate XIAP (siXIAP). Downregulation of XIAP resulted in activation of caspase-3 and caspase-9 to trigger apoptosis in glioma cells. Apoptosis is characterized by the loss of mitochondrial membrane potential and upregulation of mitochondrial apoptotic proteins Bax and Bad. Cell death of glioma cells was marked by downregulation of Akt and phospho-Akt molecules. We observed similar results under in vivo conditions in U251- and 5310-injected nude mice brains, which were treated with hUCBSC. Under in vivo conditions, Smac/DIABLO was found to be colocalized in the nucleus, showing that hUCBSC induced apoptosis is mediated by inhibition of XIAP and activation of Smac/DIABLO. CONCLUSIONS/SIGNIFICANCE Our results indicate that downregulation of XIAP by hUCBSC treatment induces apoptosis, which led to the death of the glioma cells and xenograft cells. This study demonstrates the therapeutic potential of XIAP and hUCBSC to treat malignant gliomas.
Collapse
Affiliation(s)
- Venkata Ramesh Dasari
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
| | - Kiran Kumar Velpula
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
| | - Kiranpreet Kaur
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
| | - Daniel Fassett
- Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
| | - Jeffrey D. Klopfenstein
- Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
| | - Dzung H. Dinh
- Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
| | - Meena Gujrati
- Department of Pathology, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
| | - Jasti S. Rao
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
- Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
- * E-mail:
| |
Collapse
|
65
|
Kim JY, Kim DH, Kim DS, Kim JH, Jeong SY, Jeon HB, Lee EH, Yang YS, Oh W, Chang JW. Galectin-3 secreted by human umbilical cord blood-derived mesenchymal stem cells reduces amyloid-beta42 neurotoxicity in vitro. FEBS Lett 2010; 584:3601-8. [PMID: 20655311 DOI: 10.1016/j.febslet.2010.07.028] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Revised: 07/13/2010] [Accepted: 07/16/2010] [Indexed: 01/07/2023]
Abstract
In this study, we found that expression and secretion of galectin-3 (GAL-3) were upregulated by amyloid-beta42 (Abeta42) exposure in human umbilical cord blood-derived mesenchymal stem cell (hUCB-MSC) without cell death. Abeta42-exposed rat primary cortical neuronal cells co-treated with recombinant GAL-3 were protected from neuronal death in a dose-dependent manner. hUCB-MSCs were cocultured with Abeta42-exposed rat primary neuronal cells or the neuroblastoma cell line, SH-SY5Y in a Transwell chamber. Coculture of hUCB-MSCs reduced cell death of Abeta42-exposed neurons and SH-SY5Y cells. This neuroprotective effect of hUCB-MSCs was reduced significantly by GAL-3 siRNA. These data suggested that hUCB-MSC-derived GAL-3 is a survival factor against Abeta42 neurotoxicity.
Collapse
Affiliation(s)
- Ju-Yeon Kim
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seoul 137-874, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Lee HJ, Lee JK, Lee H, Shin JW, Carter JE, Sakamoto T, Jin HK, Bae JS. The therapeutic potential of human umbilical cord blood-derived mesenchymal stem cells in Alzheimer's disease. Neurosci Lett 2010; 481:30-5. [PMID: 20600610 DOI: 10.1016/j.neulet.2010.06.045] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2010] [Revised: 06/09/2010] [Accepted: 06/15/2010] [Indexed: 12/15/2022]
Abstract
The neuropathological hallmarks of Alzheimer's disease (AD) include the presence of extracellular amyloid-beta peptide (Abeta) in the form of amyloid plaques in the brain parenchyma and neuronal loss. The mechanism associated with neuronal death by amyloid plaques is unclear but oxidative stress and glial activation has been implicated. Human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) are being scrutinized as a potential therapeutic tool to prevent various neurodegenerative diseases including AD. However, the therapeutic impact of hUCB-MSCs in AD has not yet been reported. Here we undertook in vitro work to examine the potential impact of hUCB-MSCs treatment on neuronal loss using a paradigm of cultured hippocampal neurons treated with Abeta. We confirmed that hUCB-MSCs co-culture reduced the hippocampal apoptosis induced by Abeta treatment. Moreover, in an acute AD mouse model to directly test the efficacy of hUCB-MSCs treatment on AD-related cognitive and neuropathological outcomes, we demonstrated that markers of glial activation, oxidative stress and apoptosis levels were decreased in AD mouse brain. Interestingly, hUCB-MSCs treated AD mice demonstrated cognitive rescue with restoration of learning/memory function. These data suggest that hUCB-MSCs warrant further investigation as a potential therapeutic agent in AD.
Collapse
Affiliation(s)
- Hyun Ju Lee
- Stem Cell Neuroplasticity Research Group, Kyungpook National University, Daegu, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
67
|
Jin HJ, Nam HY, Bae YK, Kim SY, Im IR, Oh W, Yang YS, Choi SJ, Kim SW. GD2 expression is closely associated with neuronal differentiation of human umbilical cord blood-derived mesenchymal stem cells. Cell Mol Life Sci 2010; 67:1845-58. [PMID: 20165901 PMCID: PMC11115935 DOI: 10.1007/s00018-010-0292-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2009] [Revised: 01/08/2010] [Accepted: 01/26/2010] [Indexed: 01/09/2023]
Abstract
GD2 ganglioside has been identified as a key determinant of bone marrow-derived mesenchymal stem cells (BM-MSCs). Here, we characterized GD2 ganglioside expression and its implications in umbilical cord blood-derived MSCs (UCB-MSCs). Using immune-selection analysis, we showed that both GD2-positive and GD2-negative UCB-MSCs expressed general stem cell markers and possessed mesodermal lineage differentiation potential. Although the fraction of GD2-expressing cells was lower in UCB-MSC than in BM-MSC populations, inhibition of GD2 synthesis in UCB-MSCs suppressed neuronal differentiation and down-regulated basic helix-loop-helix (bHLH) transcription factors, which are involved in early stage neuronal differentiation. In addition, the levels of bHLH factors in neuronally induced UCB-MSCs were significantly higher in GD2-positive than GD2-negative cells. Our data demonstrate that GD2 ganglioside expression is associated with regulation of bHLH factors and identify neurogenic-capable UCB-MSCs, providing new insights into the potential clinical applications of MSC-based therapy.
Collapse
Affiliation(s)
- Hye Jin Jin
- Biomedical Research Institute, Medipost Co., Ltd, 1571-17 Seocho-3 dong, Seocho-Gu, Seoul, 136-736 Korea
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, 388-1 Pungnap-2 dong, Songpa-Gu, Seoul, 136-736 Korea
| | - Hae Yun Nam
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, 388-1 Pungnap-2 dong, Songpa-Gu, Seoul, 136-736 Korea
| | - Yun Kyong Bae
- Biomedical Research Institute, Medipost Co., Ltd, 1571-17 Seocho-3 dong, Seocho-Gu, Seoul, 136-736 Korea
| | - Soo Yeon Kim
- Mercersburg Academy, 300 East Seminary Street, Mercersburg, PA 17236 USA
| | - I. Rang Im
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, 388-1 Pungnap-2 dong, Songpa-Gu, Seoul, 136-736 Korea
| | - Wonil Oh
- Biomedical Research Institute, Medipost Co., Ltd, 1571-17 Seocho-3 dong, Seocho-Gu, Seoul, 136-736 Korea
| | - Yoon Sun Yang
- Biomedical Research Institute, Medipost Co., Ltd, 1571-17 Seocho-3 dong, Seocho-Gu, Seoul, 136-736 Korea
| | - Soo Jin Choi
- Biomedical Research Institute, Medipost Co., Ltd, 1571-17 Seocho-3 dong, Seocho-Gu, Seoul, 136-736 Korea
| | - Seong Who Kim
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, 388-1 Pungnap-2 dong, Songpa-Gu, Seoul, 136-736 Korea
| |
Collapse
|
68
|
Lee HJ, Lee JK, Lee H, Carter JE, Chang JW, Oh W, Yang YS, Suh JG, Lee BH, Jin HK, Bae JS. Human umbilical cord blood-derived mesenchymal stem cells improve neuropathology and cognitive impairment in an Alzheimer's disease mouse model through modulation of neuroinflammation. Neurobiol Aging 2010; 33:588-602. [PMID: 20471717 DOI: 10.1016/j.neurobiolaging.2010.03.024] [Citation(s) in RCA: 208] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Revised: 03/26/2010] [Accepted: 03/31/2010] [Indexed: 12/12/2022]
Abstract
Human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSC) have a potential therapeutic role in the treatment of neurological disorders, but their current clinical usage and mechanism of action has yet to be ascertained in Alzheimer's disease (AD). Here we report that hUCB-MSC transplantation into amyloid precursor protein (APP) and presenilin1 (PS1) double-transgenic mice significantly improved spatial learning and memory decline. Furthermore, amyloid-β peptide (Aβ) deposition, β-secretase 1 (BACE-1) levels, and tau hyperphosphorylation were dramatically reduced in hUCB-MSC transplanted APP/PS1 mice. Interestingly, these effects were associated with reversal of disease-associated microglial neuroinflammation, as evidenced by decreased microglia-induced proinflammatory cytokines, elevated alternatively activated microglia, and increased anti-inflammatory cytokines. These findings lead us to suggest that hUCB-MSC produced their sustained neuroprotective effect by inducing a feed-forward loop involving alternative activation of microglial neuroinflammation, thereby ameliorating disease pathophysiology and reversing the cognitive decline associated with Aβ deposition in AD mice.
Collapse
Affiliation(s)
- Hyun Ju Lee
- Stem Cell Neuroplasticity Research Group, Kyungpook National University, Daegu, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Kim JY, Jeon HB, Yang YS, Oh W, Chang JW. Application of human umbilical cord blood-derived mesenchymal stem cells in disease models. World J Stem Cells 2010; 2:34-8. [PMID: 21607114 PMCID: PMC3097922 DOI: 10.4252/wjsc.v2.i2.34] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Revised: 01/28/2010] [Accepted: 02/04/2010] [Indexed: 02/07/2023] Open
Abstract
Human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) are regarded as an alternative source of bone marrow-derived mesenchymal stem cells because collection of cord blood is less invasive than that of bone marrow. hUCB-MSCs have recently been studied for evaluation of their potential as a source of cell therapy. In this review, the general characteristics of hUCB-MSCs and their therapeutic effects on various diseases in vitro and in vivo will be discussed.
Collapse
Affiliation(s)
- Ju-Yeon Kim
- Ju-Yeon Kim, Hong Bae Jeon, Yoon Sun Yang, Wonil Oh, Jong Wook Chang, Biomedical Research Institute, MEDIPOST Co., Ltd, Seoul 137-874, South Korea
| | | | | | | | | |
Collapse
|
70
|
Kosztowski T, Zaidi HA, Quiñones-Hinojosa A. Applications of neural and mesenchymal stem cells in the treatment of gliomas. Expert Rev Anticancer Ther 2009; 9:597-612. [PMID: 19445577 DOI: 10.1586/era.09.22] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In addition to stem cells providing a better understanding about the biology and origins of gliomas, new therapeutic approaches have been developed based on the use of stem cells as delivery vehicles. The unique ability of stem cells to track down tumor cells makes them a very appealing therapeutic modality. This review introduces neural and mesenchymal stem cells, discusses the advances that have been made in the utilization of these stem cells as therapies and in diagnostic imaging (to track the advancement of the stem cells towards the tumor cells), and concludes by addressing various challenges and concerns regarding these therapies.
Collapse
Affiliation(s)
- Thomas Kosztowski
- The Johns Hopkins Hospital, Department of Neurosurgery, Johns Hopkins University, CRB II, 1550 Orleans Street, Room 247, Baltimore, MD 21231, USA.
| | | | | |
Collapse
|