• Title/Summary/Keyword: human mesenchymal stem cell

Search Result 230, Processing Time 0.031 seconds

Evaluation of Optimal Combination of Commercially Available Superparamagnetic Iron Oxide Nanoparticles and Transfection Agents for Labelling of Human Mesenchymal Stem Cells (인체 중간엽 줄기세포의 표지를 위한 상용화 된 Superparamagnetic Iron Oxide Nanoparticle과 Tansfection Agent의 적절한 병용을 위한 연구)

  • Kim, Sung-Hun;Oh, Soon-Nam;Park, Youn-Hee;Kang, Won-Kyung;Ahn, Kook-Jin;Chung, Soo-Kyo
    • Investigative Magnetic Resonance Imaging
    • /
    • v.16 no.1
    • /
    • pp.31-39
    • /
    • 2012
  • Purpose : To determine the optimal combination of commercially available superparamagnetic iron oxide (SPIO) nanoparticles with transfection agents (TA). Materials and Methods: Protamine sulfate (Pro) and poly-L-lysin (PLL) were incubated with ferumoxide and ferucarbotran in human mesenchymal stem cells at various concentrations, and cellular viability were evaluated. Cellular iron uptake was qualitatively and quantitatively evaluated. Cell visibility was assessed via MR imaging and the T2-relaxation time was calculated. Results: The cellular viabilities with ferucarbotran were more significantly decreased than those with ferumoxide (p < 0.05). Iron uptake with ferumoxide was significantly higher than that for those with with ferucarbotran. The T2-relaxation time was observed to be shorter with ferumoxide in comparison to those with ferucarbotran (p < 0.05). Ferumoxide at a concentration of 25 ${\mu}g$/ml in combination with either Pro or PLL at a concentration of 3.0 ${\mu}g$/ml did not adversely impact cell viability, maximized iron uptake, and exhibited a lower T2-relaxation time in comparison to other combinations. Conclusion: Stem cells with ferumoxide exhibited a higher cellular viability and iron uptake in comparison to ferucarbotran-treated stem cells. A 25 ${\mu}g$/ml of ferumoxide with a 3.0 ${\mu}g$/ml of TA is sufficient to label mesenchymal stem cells.

Improvement of Neuronal Differentiation by PDE4 Inhibition in Human Bone Marrow-mesenchymal Stem Cells (인간 골수유래-중간엽 줄기세포(hBM-MSCs)에서 PDE4 억제조절을 통한 신경세포 분화 효율 개선)

  • Jeong, Da Hee;Joe, I-Seul;Cho, Goang-Won
    • Journal of Life Science
    • /
    • v.26 no.12
    • /
    • pp.1355-1359
    • /
    • 2016
  • Human bone marrow mesenchymal stem cells (hBM-MSCs) can differentiate into various cell types including osteoblasts, adipocytes, chondrocytes, and myocytes. Previous studies, including our own, have shown that MSCs can also differentiate into neuron-like cells. However, their rate of neuronal differentiation is not sufficient for application to stem cell therapy, which requires well-defined cell types. For this purpose, we first examined the expression of neuronal lineage markers (GFAP, MAP-2, KCNH1, Nestin, NF-M, and Tuj-1) by real-time PCR, western blot, and immunocytochemical staining. The expressions of the astrocyte marker GFAP and neuronal markers NF-M and Tuj-1 increased in neuronal differentiated MSCs (dMSCs). To improve the neuronal differentiation efficiency, PDE4, an important signaling intermediator in the progression of neuronal differentiation, was modulated using well-known inhibitors such as rolipram or resveratrol and then differentiated into neuronal cells (Roli- or RSV-dMSCs). The expressions of NF-M, Tuj-1 were increased while that of GFAP decreased in Roli- and RSV-dMSCs, which were examined by real-time PCR, western blot, and immunocytochemical staining. From these experiments, we have found that the neuronal differentiation efficiency can be ameliorated by the modulation of PDE4 activity.

Role of NFAT5 in Osteogenic Differentiation of Human Adipose Tissue-Derived Mesenchymal Stem Cells (인체 지방 유래 중간엽 줄기세포의 골분화 조절 기전에서 NFAT5의 역할)

  • Lee, Sun Young;Yang, Ji won;Jung, Jin Sup
    • Journal of Life Science
    • /
    • v.23 no.4
    • /
    • pp.471-478
    • /
    • 2013
  • Human adipose tissue-derived mesenchymal stem cells (hADSCs) have therapeutic potential, including the ability to self-renew and differentiate into multiple lineages. Understanding of molecular mechanisms of stem cell differentiation is important for improving the therapeutic efficacies of stem cell transplantation. In this study, we determined the role of nuclear factor of activated T cells (NFAT5) in the osteogenic differentiation of hADSCs. The down-regulation of NFAT5 expression by the transfection of a specific siRNA significantly inhibited osteogenic differentiation of hADSCs and decreased the activity of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-${\kappa}B$) promoter without affecting their proliferation and adipogenic differentiation. The inhibition of NFAT5 expression inhibited the basal and Tumor Necrosis Factor ${\alpha}$ (TNF-${\alpha}$) induced activation of NF-${\kappa}B$, but it did not affect TNF-${\alpha}$-induced degradation of the $I{\kappa}B$ protein. These findings indicate that NFAT5 plays an important role in the osteogenic differentiation of hADSCs through the modulation of the NF-${\kappa}B$ pathway.

Effects of Human Mesenchymal Stem Cell Transplantation Combined with Polymer on Functional Recovery Following Spinal Cord Hemisection in Rats

  • Choi, Ji Soo;Leem, Joong Woo;Lee, Kyung Hee;Kim, Sung-Soo;SuhKim, Haeyoung;Jung, Se Jung;Kim, Un Jeng;Lee, Bae Hwan
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.16 no.6
    • /
    • pp.405-411
    • /
    • 2012
  • The spontaneous axon regeneration of damaged neurons is limited after spinal cord injury (SCI). Recently, mesenchymal stem cell (MSC) transplantation was proposed as a potential approach for enhancing nerve regeneration that avoids the ethical issues associated with embryonic stem cell transplantation. As SCI is a complex pathological entity, the treatment of SCI requires a multipronged approach. The purpose of the present study was to investigate the functional recovery and therapeutic potential of human MSCs (hMSCs) and polymer in a spinal cord hemisection injury model. Rats were subjected to hemisection injuries and then divided into three groups. Two groups of rats underwent partial thoracic hemisection injury followed by implantation of either polymer only or polymer with hMSCs. Another hemisection-only group was used as a control. Behavioral, electrophysiological and immunohistochemical studies were performed on all rats. The functional recovery was significantly improved in the polymer with hMSC-transplanted group as compared with control at five weeks after transplantation. The results of electrophysiologic study demonstrated that the latency of somatosensory-evoked potentials (SSEPs) in the polymer with hMSC-transplanted group was significantly shorter than in the hemisection-only control group. In the results of immunohistochemical study, ${\beta}$-gal-positive cells were observed in the injured and adjacent sites after hMSC transplantation. Surviving hMSCs differentiated into various cell types such as neurons, astrocytes and oligodendrocytes. These data suggest that hMSC transplantation with polymer may play an important role in functional recovery and axonal regeneration after SCI, and may be a potential therapeutic strategy for SCI.

PLEIOTROPHIN EFFECTS ON BINDING AND SUBSEQUENT OSTEOGENESIS OF HUMAN MESENCHYMAL STEM CELLS (Pleiotrophin이 골수 줄기 세포의 부착 및 골형성에 미치는 효과에 대한 연구)

  • Yoon, Jung-Ho;Eune, Jung-Ju;Jang, Hyon-Seok;Rim, Jae-Suk;Lee, Eui-Seok;Kim, Dae-Sung;Kwon, Jong-Jin
    • Maxillofacial Plastic and Reconstructive Surgery
    • /
    • v.28 no.2
    • /
    • pp.111-117
    • /
    • 2006
  • An area of current research is investigating the app1ication of human mesenchymal stem cells or hMSCs as a cell-based regenerative therapy. In order to achieve effective bone regeneration, appropriate matrices functioning as cell-carriers must be identified and optimized in terms of function, efficacy and biocompatibility. Two methods of approaching optimization of matrices are to facilitate adhesion of the donor hMSCs and furthermore to facilitate recruitment of host progenitor cells to osteoblastic differentiation. Pleiotrophin is an extracellular matrix protein that was first identified in developing rat brains and believed to be associated with developing neuronal pathways. A recent publication by Imai and colleagues demonstrated that transgenic mice with upregulated pleiotrophin expression developed a greater volume of cortical as well as cancellous bone. The proposed mechanism of action of pleiotrophin is demonstrated here. Through either environmental stresses and/or intracellular regulation, there is an increase in pleiotrophin production. The pleiotrophin is released extracellularly into areas requiring bone deposition. A receptor-mediated process recruits host osteoprogenitor cells into these areas. Therefore, the aim of our study was to investigate the osteoconductive properties of pleiotrophin. We wanted to determine if pleiotrophin coating facilitates cellular adhesion and furthermore if this has any effect on hMSCs derived bone formation in an animal model. The results showed a dose dependent response of cellular adhesion in fibronectin samples, and cellular adhesion was facilitated with increasing pleiotrophin concentrations. Histologic findings taken after 5 weeks implantation in SCID mouse showed no presence of bone formation with only a dense fibrous connective tissue. Possible explanations for the results of the osteogenesis assay include inappropriate cell loading.

Trans-differentiation Induction of Human-mesenchymal Stem Cells Derived from Different Tissue Origin and Evaluation of their Potential for Differentiation into Corneal Epithelial-like Cells

  • Moon, Sun-Woung;Lee, Hyeon-Jeong;Lee, Won-Jae;Ock, Sun-A;Lee, Sung-Lim
    • Journal of Embryo Transfer
    • /
    • v.33 no.2
    • /
    • pp.85-97
    • /
    • 2018
  • The trans-differentiation potential of mesenchymal stem cells (MSCs) is employed, but there is little understanding of the cell source-dependent trans-differentiation potential of MSCs into corneal epithelial cells. In the present study, we induced trans-differentiation of MSCs derived from umbilical cord matrix (UCM-MSCs) and from dental tissue (D-MSCs), and we comparatively evaluated the in vitro trans-differentiation properties of both MSCs into corneal epithelial-like cells. Specific cell surface markers of MSC (CD44, CD73, CD90, and CD105) were detected in both UCM-MSCs and D-MSCs, but MHCII and CD119 were significantly lower (P < 0.05) in UCM-MSCs than in D-MSCs. In UCM-MSCs, not only expression levels of Oct3/4 and Nanog but also proliferation ability were significantly higher (P < 0.05) than in D-MSCs. In vitro differentiation abilities into adipocytes and osteocytes were confirmed for both MSCs. UCM-MSCs and D-MSCs were successfully trans-differentiated into corneal epithelial cells, and expression of lineage-specific markers (Cytokeratin-3, -8, and -12) were confirmed in both MSCs using immunofluorescence staining and qRT-PCR analysis. In particular, the differentiation capacity of UCM-MSCs into corneal epithelial cells was significantly higher (P < 0.05) than that of D-MSCs. In conclusion, UCM-MSCs have higher differentiation potential into corneal epithelial-like cells and have lower expression of CD119 and MHC class II than D-MSCs, which makes them a better source for the treatment of corneal opacity.

Growth and Osteoblastic Differentiation of Mesenchymal Stem Cells on Silk Scaffolds

  • Cho, Hee-Yeon;Baik, Young-Ae;Jeon, Suyeon;Kwak, Yoon-Hae;Kweon, Hae Yong;Jo, You Young;Lee, Kwang Gill;Park, Young Hwan;Kang, Dongchul
    • International Journal of Industrial Entomology and Biomaterials
    • /
    • v.27 no.2
    • /
    • pp.303-311
    • /
    • 2013
  • In this study, we compared the efficiency of osteoblast differentiation media (ODM) containing three distinct reagent combinations in osteoblastic differentiation of human bone marrow-derived mesenchymal stem cells (hBMSCs) in monolayer culture. In addition, we analyzed growth and differentiation of hBMSCs on silk scaffolds and examined the bone-forming activity of a nanofibrous silk scaffold in a tibia diaphysis defect model of a rat hind limb with intramedullary nailing. Although all three ODM increased alkaline phosphatase activity to a comparable extent, the ODM containing bone morphogenetic protein-2 (BMP-2) was found to be significantly less effective in promoting mineral deposition than the others. Growth of hBMSCs on sponge-form silk scaffolds was faster than on nanofibrous ones, while osteoblastic differentiation was apparent in the cells grown on either type of scaffold. By contrast, bone formation was observed only at the edge of the nanofibrous scaffold implanted in the tibia diaphysis defect, suggesting that use of the silk scaffold alone is not sufficient for the reconstitution of the long bone defect. Since silk scaffolds can support cell growth and differentiation in vitro, loading MSCs on scaffolds might be necessary to improve the bone-forming activity of the scaffold in the long bone defect model.

Lactoferrin Protects Human Mesenchymal Stem Cells from Oxidative Stress-Induced Senescence and Apoptosis

  • Park, Soon Yong;Jeong, Ae-Jin;Kim, Geun-Young;Jo, Ara;Lee, Joo Eon;Leem, Sun-Hee;Yoon, Joung-Hahn;Ye, Sang Kyu;Chung, Jin Woong
    • Journal of Microbiology and Biotechnology
    • /
    • v.27 no.10
    • /
    • pp.1877-1884
    • /
    • 2017
  • Mesenchymal stem cells (MSCs) have been suggested as a primary candidate for cell therapy applications because they have self-renewal and differentiation capabilities. Although they can be expanded in ex vivo system, clinical application of these cells is still limited because they survive poorly and undergo senescence or apoptosis when transplanted and exposed to environmental factors such as oxidative stress. Thus, reducing oxidative stress is expected to improve the efficacy of MSC therapy. The milk protein lactoferrin is a multifunctional iron-binding glycoprotein that plays various roles, including reduction of oxidative stress. Thus, we explored the effect of lactoferrin on oxidative stress-induced senescence and apoptosis of human MSCs (hMSCs). Measurement of reactive oxygen species (ROS) revealed that lactoferrin inhibited the production of hydrogen peroxide-induced intracellular ROS, suggesting lactoferrin as a good candidate as an antioxidant in hMSCs. Pretreatment of lactoferrin suppressed hydrogen peroxide-induced senescence of hMSCs. In addition, lactoferrin reduced hydrogen peroxide-induced apoptosis via inhibition of caspase-3 and Akt activation. These results demonstrate that lactoferrin can be a promising factor to protect hMSCs from oxidative stress-induced senescence and apoptosis, thus increasing the efficacy of MSC therapy.

The Significance of SDF-1α-CXCR4 Axis in in vivo Angiogenic Ability of Human Periodontal Ligament Stem Cells

  • Bae, Yoon-Kyung;Kim, Gee-Hye;Lee, Jae Cheoun;Seo, Byoung-Moo;Joo, Kyeung-Min;Lee, Gene;Nam, Hyun
    • Molecules and Cells
    • /
    • v.40 no.6
    • /
    • pp.386-392
    • /
    • 2017
  • Periodontal ligament stem cells (PDLSCs) are multipotent stem cells derived from periodontium and have mesenchymal stem cell (MSC)-like characteristics. Recently, the perivascular region was recognized as the developmental origin of MSCs, which suggests the in vivo angiogenic potential of PDLSCs. In this study, we investigated whether PDLSCs could be a potential source of perivascular cells, which could contribute to in vivo angiogenesis. PDLSCs exhibited typical MSC-like characteristics such as the expression pattern of surface markers (CD29, CD44, CD73, and CD105) and differentiation potentials (osteogenic and adipogenic differentiation). Moreover, PDLSCs expressed perivascular cell markers such as NG2, ${\alpha}-smooth$ muscle actin, platelet-derived growth factor receptor ${\beta}$, and CD146. We conducted an in vivo Matrigel plug assay to confirm the in vivo angiogenic potential of PDLSCs. We could not observe significant vessel-like structures with PDLSCs alone or human umbilical vein endothelial cells (HUVECs) alone at day 7 after injection. However, when PDLSCs and HUVECs were co-injected, there were vessel-like structures containing red blood cells in the lumens, which suggested that anastomosis occurred between newly formed vessels and host circulatory system. To block the $SDF-1{\alpha}$ and CXCR4 axis between PDLSCs and HUVECs, AMD3100, a CXCR4 antagonist, was added into the Matrigel plug. After day 3 and day 7 after injection, there were no significant vessel-like structures. In conclusion, we demonstrated the perivascular characteristics of PDLSCs and their contribution to in vivo angiogenesis, which might imply potential application of PDLSCs into the neovascularization of tissue engineering and vascular diseases.