Systemically Injected Mesenchymal Stem Cells Traffic to Myelomatous Bone and Inhibit Bone Disease, and Intralesionally Promotes Bone Formation and Delays Myeloma Progression During the Disease Active Stage or Remission

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 980-980
Author(s):  
Xin Li ◽  
Wen Ling ◽  
Sharmin Khan ◽  
Yuping Wang ◽  
Angela Pennisi ◽  
...  

Abstract Abstract 980 Mesenchymal stem cells (MSCs) cytotherapy has been clinically tested in various applications including bone regeneration, autoimmune diseases, and cancer. The aims of the study were to test the effect of MSCs cytotherapy on myeloma (MM) bone disease and tumor growth, and determine their ability to traffic into myelomatous bone during active disease stage and induction of remission by melphalan. We exploited the SCID-rab model for MM and a novel human myeloma cell line, Hg, established through our previously reported procedure (Xin et al., BJH 2007). Hg myeloma cells do not grow in culture but are capable of sequential passaging in SCID-rab mice. Microarray analysis revealed similar gene expression profiling between the Hg cell line and the original patient's myeloma plasma cells, and that these cells are classified in the MMSET subgroup and express DKK1, indicating their authenticity and clinical relevancy. The human fetal MSCs were transduced with a luciferase/GFP reporter in a lentiviral vector for in vivo tracking. In Hg-bearing hosts (5 mice/group), intra-bone injection of MSCs (1×106 cells/mouse) increased bone mineral density (BMD) by 21±4% while in control, diluent injected bones it was reduced by 14±5% (p<0.001). Increased bone formation by MSCs was associated with reduced tumor growth by 50% (p<0.01). Bioluminescence analysis revealed disappearance of the majority of the intralesionally injected MSCs within 4 weeks, suggesting that MSCs exert their effects on bone remodeling as a bystander cells (trophic effect). To test effect on relapse, remission was induced by treating Hg-bearing hosts with a total of 4 subcutaneous injections of melphalan (10 mg/kg/4 days), followed by intra-bone injection of diluent or MSCs (10 mice/group). Three weeks post-cytotherapy BMD was increased by 23±5% in bones injected with MSCs and reduced by 23±3% in bones injected with diluent (p<0.001). Eleven weeks post-cytotherapy, BMD was reduced by 33±6% and 9±7% in bones injected with diluent and MSCs, respectively (p<0.03). Following melphalan treatment circulating immunoglobulin (Ig) level (MM burden) was undetected while at 2 weeks after cytotherapy it was detected in 80% and 30% of hosts injected with diluent and MSCs, respectively. At experiment's end, Ig levels were significantly lower by 6 folds in hosts treated with MSCs than diluent (p<0.01). To further validate clinical relevancy, MSCs or diluent were intravenously injected into Hg-bearing hosts. In contrast to a single injection, 4 weekly, intravenous injections of MSCs (10 mice/group) prevented reduction of the BMD of the myelomatous bone while in control hosts the BMD was reduced by 14±3% (p<0.006 vs. pretreatment). MM growth was not affected by single or multiple intravenous injections of MSCs. Ex vivo imaging of tissues from hosts with active MM or treated with melphalan detected MSCs in implanted bones and murine lungs indicating that myeloma cells or conditions induced by MM or melphalan attract MSCs to myelomatous bones. We conclude that intra-bone injection of MSCs effectively promotes bone formation and delays MM progression during the disease active stage or remission. We also conclude that exogenous MSCs are capable of trafficking to myelomatous bone and that systemic, weekly injections of MSCs inhibit MM bone disease. Disclosures: No relevant conflicts of interest to declare.

2021 ◽  
Vol 11 ◽  
Author(s):  
Zhenqing Xie ◽  
Yan Xu ◽  
Xiaojing Wei ◽  
Gang An ◽  
Mu Hao ◽  
...  

Myeloma bone disease (MBD), caused by the inhibition of osteoblast activity and the activation of osteoclast in the bone marrow environment, is the most frequent and life-threatening complication in multiple myeloma (MM) patients. Bortezomib (Bzb) was shown to promote MM-derived mesenchymal stem cells (MM-MSCs) differentiation to osteoblast in vitro and in animal models, promoting the bone formation and regeneration, may be mediated via β-catenin/T-cell factor (TCF) pathway. Further defining molecular mechanism of Bzb-enhanced bone formation in MM will be beneficial for the treatment of myeloma patients. The present study has identified for the first time four and a half LIM domains protein 2 (FHL2), a tissue-specific coregulator that interacts with many osteogenic marker molecules, as a therapeutic target to ameliorate MM bone disease. First, increased messenger RNA (mRNA) and protein levels of FHL2, and the mRNA level of main osteoblast markers (including Runx2, ALP, and Col1A1), were found in MM-patients-derived MSCs after Bzb treatment. FHL2 KD with short hairpin RNA (shRNA) reduced the expression of osteoblast marker genes and blocked the osteogenic differentiation of MM-MSCs regardless of the presence or absence of Bzb, implying that FHL2 is an important activator of the osteogenic differentiation of human MSCs under a proteasome inhibition condition. Molecular analysis showed that the enhanced expression of FHL2 was associated with the Bzb-induced upregulation of p53. No significant change at protein level of total β-catenin was observed with or without Bzb treatment. However, it was mostly enriched to nuclei in MSCs after Bzb treatment. Moreover, β-catenin was restricted to the perinuclear region in FHL2 KD cells. These data provide evidence that FHL2 is essential for promoting β-catenin nuclear enrichment in MM-MSCs. In conclusion, FHL2 is critical for Bzb-induced osteoblast differentiation of MM-MSCs and promotes the osteogenesis, through p53 signaling and β-catenin activation. Targeting FHL2 in MM may provide a new therapeutic strategy for treating MBD.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 510-510
Author(s):  
Alissa Huston ◽  
Ganwei Lu ◽  
Judy Anderson ◽  
Ken Patrene ◽  
Lily Hong ◽  
...  

Abstract MM is the most common cancer metastasizing to bone, with 85–90% of patients developing bone disease. It is unique from other forms of metastatic bone disease, as it is purely lytic. There are a multitude of factors involved in promoting bone disease in MM; however, what remains unanswered is why patients in complete remission from MM are unable to heal their bone lesions. We hypothesize that MM cells exert a permanent change in the bone marrow microenvironment, either through a change in the differentiation potential of mesenchymal stem cells or through stem cell depletion. To investigate this question we developed a new model of MM bone disease in which a herpes-simplex thymidine kinase gene (HSVtk), sensitive to ganciclovir (GCV), is expressed within a MM cell line and injected into the tibia of mice. This allows for eradication of the MM cells without the use of chemotherapy or immunotherapy, minimizes adverse effects on neighboring mesenchymal stem cells and directly involves the bone. The murine MM cell line (5TGM1) was infected, using a lentiviral system, with the tricistronic construct of HSVtk linked to green fluorescence protein (GFP) and blasticidin. Following blasticidin selection, 5TGM1tk cells stably expressing HSVtk and GFP were used for all experiments. GCV dosing curves to determine the dose resulting in 100% eradication were completed. The effects of GCV on both murine hematopoietic cells and mesenchymal stem cells induced to osteoblasts were completed using methylcellulose colony formation and alkaline phosphatase assays. Bystander effects of 5TGM1tk cells on murine hematopoietic cells and murine osteoblasts were completed using murine methylcellulose colony formation, alkaline phosphatase and colony-forming unit fibroblast (CFU-F) assays. In vivo data was obtained following intratibial injection of 5TGM1tk cells (1x105, 5x105, 1x106) or saline into NIH-III mice (3 mice per group, 12 mice total). Mice were evaluated at weekly intervals for tumor and lytic lesion development with plain radiographs and Micro QCT, and osteoblast activity evaluated by alkaline phosphatase analysis. No significant effects of GCV, at doses capable of eradicating the MM cell line (1ug/ml) were observed on murine hematopoietic cells or osteoblasts. No significant effect was observed on hematopoietic colony development when 5TGM1tk cells were cocultured with murine hematopoietic cells. A small bystander effect, approximately 30%, was identified when 5TGM1tk cells were cocultured with murine mesenchymal stem cells induced to osteoblasts in the presence of GCV. Dose-dependent CFU-F suppression was observed with increasing percentages of tumor cells (0–50%), however reversal of suppression was noted when cultured in the presence of GCV. 60% of the mice injected with 5TGM1tk cells developed tumor, and evidence of dose-dependent osteoblast suppression in vivo observed as measured by the alkaline phosphatase assay (p&lt;0.00001). The current model provides us with the capacity to specifically target MM cells without using chemotherapy or immunotherapy. It provides a model to study and dissect the interactions between MM cells and mesenchymal stem cells, within the bone marrow microenvironment, which lead to osteoblast suppression. This model should provide important insights into the mechanisms of osteoblast suppression in MM and help to identify targeted therapies that can reverse this process for patients.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3717-3717
Author(s):  
Xin Li ◽  
Wen Ling ◽  
Angela Pennisi ◽  
Yuping Wang ◽  
Sharmin Khan ◽  
...  

Abstract Abstract 3717 Human placenta has emerged as a valuable, uncontroversial source of transplantable cells for many cytotherapeutic purposes, including modulation of inflammation, bone repair, and cancer. Placenta-derived adherent cells (PDAC) are mesenchymal like adherent cells isolated from postpartum human placenta and capable of supporting bone formation in vivo. Multiple myeloma (MM) is closely associated with induction of bone disease and large lytic lesions, which are often not repaired and are usually the sites of relapses. The aim of the study was to evaluate the antimyeloma therapeutic potential, in vivo survival, and trafficking of PDAC in the SCID-rab model of MM-associated bone disease. SCID-rab system constructed by implanting a 4-weeks old rabbit bone into which primary human myeloma cells were directly injected (Yatta et al., Leukemia 2004; Yaccoby et al., Blood 2007). Bone disease was evaluated by measurements of bone mineral density (BMD) and X-rays. MM growth was determined by human immunoglobulin (hIg) ELISA and histologically. For in vivo tracking PDAC were transduced with a luciferase/GFP reporter in a lentiviral vector. SCID-rab mice engrafted with primary myeloma cells from 2 patients. Upon establishment of MM growth, PDAC (1×106 cells/bone) or vehicle were injected into the implanted myelomatous bone (Patient's 1, 5 mice/group; Patient's 2, 7 mice/group). While BMD of the implanted bones was significantly reduced in control hosts, intralesional PDAC cytotherapy significantly increased BMD of the implanted bones from pretreatment levels by >37% (p<0.01 versus control) and inhibited MM growth in the 2 sets of experiments (p<0.04). The bone anabolic effect of PDAC was associated with increased number of osteoblasts (p<0.003) and reduced number of osteoclasts (p<0.004). Intralesional PDAC cytotherapy also promoted bone formation in nonmyelomatous SCID-rab mice. Intralesional but not subcutaneous engraftment of PDAC inhibited bone disease and tumor growth in SCID-rab mice. In contrast to intra-bone injection in SCID-rab mice, intra-tumor injection of PDAC had no effect on subcutaneous growth of the H929 myeloma cell line in SCID mice (8 mice/group). Live-animal imaging revealed that the majority of PDAC disappeared from the injected bones within 4 weeks. To test their systemic behavior, PDAC were intravenously injected into 18 SCID-rab mice engrafted with H929 myeloma cells. The presences of PDAC in various organs were evaluated 1, 2 and 7 days after injection. Ex vivo bioluminescence analysis of the implanted myelomatous bones detected PDAC in two of five bones on day 1, in four of four bones on day 2, and in four of nine bones on day 7. Intravenously injected PDAC were also detected in lungs not in any other murine tissues. Immunohistochemical staining for GFP in myelomatous bone sections detected GFP-expressing PDAC in rabbit marrow areas infiltrated with myeloma cells, supporting bioluminescence analysis. Our study suggest that PDAC stimulate bone formation by acting as bystander cells that increase endogenous osteoblastogenesis and inhibit osteoclastogenesis, and that alteration of the bone marrow microenvironment by PDAC attenuates growth of MM. PDAC cytotherapy is a promising therapeutic approach for myeloma bone disease. Disclosures: Khan: Celgene Cellular Therapeutics: Research Funding. Heidaran:Celgene Cellular Therapeutics: Employment. Pal:Celgene Cellular Therapeutics: Employment. Zhang:Celgene Cellular Therapeutics: Employment. He:Celgene Cellular Therapeutics: Employment. Zeitlin:Celgene Cellular Therapeutics: Employment. Abbot:Celgene Cellular Therapeutics: Employment. Faleck:Celgene Cellular Therapeutics: Employment. Hariri:Celgene Cellular Therapeutics: Employment.


2020 ◽  
Vol 13 (633) ◽  
pp. eaay8203 ◽  
Author(s):  
Zhiqiang Liu ◽  
Huan Liu ◽  
Jin He ◽  
Pei Lin ◽  
Qiang Tong ◽  
...  

The suppression of bone formation is a hallmark of multiple myeloma. Myeloma cells inhibit osteoblastogenesis from mesenchymal stem cells (MSCs), which can also differentiate into adipocytes. We investigated myeloma-MSC interactions and the effects of such interactions on the differentiation of MSCs into adipocytes or osteoblasts using single-cell RNA sequencing, in vitro coculture, and subcutaneous injection of MSCs and myeloma cells into mice. Our results revealed that the α4 integrin subunit on myeloma cells stimulated vascular cell adhesion molecule–1 (VCAM1) on MSCs, leading to the activation of protein kinase C β1 (PKCβ1) signaling and repression of the muscle ring-finger protein-1 (MURF1)–mediated ubiquitylation of peroxisome proliferator–activated receptor γ2 (PPARγ2). Stabilized PPARγ2 proteins enhanced adipogenesis and consequently reduced osteoblastogenesis from MSCs, thus suppressing bone formation in vitro and in vivo. These findings reveal that suppressed bone formation is a direct consequence of myeloma-MSC contact that promotes the differentiation of MSCs into adipocytes at the expense of osteoblasts. Thus, this study provides a potential strategy for treating bone resorption in patients with myeloma by counteracting tumor-MSC interactions.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 119-119 ◽  
Author(s):  
Michaela R. Reagan ◽  
Michelle McDonald ◽  
Rachael Terry ◽  
Jessica Pettitt ◽  
Lawrence Le ◽  
...  

Abstract Multiple myeloma (MM) is a malignancy of plasma cells and is characterized by unrestricted tumor cell growth in bone marrow (BM). MM causes destructive osteolytic lesions causing bone fracture, bone pain, hypercalcaemia, and nerve-compression, resulting from increased bone resorption and suppressed bone formation. Despite the introduction of agents to inhibit bone resorption, such as bisphosphonates, which prevent further bone loss, approaches to preventing osteoblast suppression and repair bone lesions are limited and there are no agents available clinically. The wnt/β-catenin pathway plays a critical role in the regulation of bone formation. Production of the soluble wnt antagonist dickkopf1 (Dkk1) by MM cells has been implicated in MM inhibition of bone formation. As such, Anti-Dkk1 treatment prevents bone disease in pre-clinical models of MM and is in early clinical development. However, Dkk1 is not expressed by all myeloma cells; hence only a proportion of patients may respond to anti-Dkk1 therapy. Sclerostin (Scl) is a soluble wnt antagonist whose expression, unlike Dkk1, is restricted to osteocytes; therefore Scl targeted agents may have less off target effects. Anti-Sclerostin (Anti-Scl) treatment increased bone formation and bone volume in experimental models of osteoporosis, and increased bone mineral density in phase II osteoporosis clinical trials. However, Anti-Scl treatment effects on myeloma bone disease are unknown. Further, cells of the BM such as osteoblasts have been implicated in the regulation of MM cell survival and growth. Thus, in the present study we explored the potential for Anti-Scl therapy to prevent MM induced bone loss and inhibit MM growth in both murine and human xenograft MM models. Female C57BLKalwRij mice (n=8) were injected i.v, with 5TGM1/eGFP murine MM cells (1×106) and female SCID/beige mice (n=10) were injected i.v. with MM1S/Luc/eGFP human MM cells (4 × 106). 24 hours later, naïve mice (without tumor cells) or mice bearing MM cells were treated with anti-sclerostin antibody (Anti-Scl) (100mg/kg i.v) or control antibody. Mice were sacrificed at day 21 (MM1S) or day 28 (5TGM1) and the effect of Anti-Scl on bone structure in the femora and vertebrae were determined by microCT analysis. The effect of Anti-Scl on MM burden was determined by bioluminescent imaging (BLI) performed twice weekly from week 1 using a Xenogen IVIS system, whereas MM burden in 5TGM1/eGFP bearing mice was examined by FACS analysis. Anti-Scl treatment in naïve C57BLKalwRij mice increased trabecular bone volume fraction (BV/TV, 39%, p<0.01, Fig. 1B) in the femur, which was mediated by increases in trabecular thickness (Tb.Th, 42%, p<0.01). Treatment also increased cortical bone volume (22%, p<0.01) in the femur and increased trabecular BV/TV in the vertebra (32%, p<0.01). This demonstrated the potent bone anabolic effect of Anti-Scl independent of myeloma cells. Injection of 5TGM1 cells resulted in a decrease in femoral trabecular BV/TV (30%, p<0.01) through a 30% reduction in trabecular number (TbN) (p<0.01), but no effect on Tb.Th (Figs. 1A and B), whilst also reducing cortical bone volume (BV) by 6% (p<0.05). Vertebrae were also impacted by 5TGM1 tumor growth with a 29% reduction in Tb BV/TV through a 23% reduction in Tb.Th (p<0.01) and also a 15% reduction in cortical BV (p<0.01). Treatment of 5TGM1-bearing mice with Anti-Scl increased trabecular BV/TV (46%, p<0.01) and Tb.Th (30%, p<0.01) to values equivalent to femora of naïve, non-tumor bearing, control mice. Treatment with Anti-Scl also increased cortical BV by 16% (p<0.01), vertebral Tb BV/TV by 29% and cortical BV by 36% in 5TGM1 burdened mice (p<0.01). Treatment of 5TGM1-bearing mice with Anti-Scl had no effect on the proportion 5TGM1/eGFP cells in the BM or spleen. However Anti-Scl treatment significantly suppressed tumor progression in the MM1S model at 3, 3.5 and 4 weeks post cell injection, as determined by BLI imaging (p=0.02, wk 3; p=0.0019, wk 3.5, and p=0.0068, wk 4, 2-tailed t-tests, Fig. 1C). These data demonstrate that Anti-Scl antibody treatment can prevent development of myeloma bone disease. Furthermore, Anti-Scl treatment also suppressed tumor growth, supporting the possibility that targeting the BM microenvironment with this agent may slow disease progression. Our findings highlight the potential clinical application of Anti-Scl antibody treatment in patients with MM and other bone destructive cancers. Disclosures Kneissel: Novartis Institutes for Biomedical Research, Novartis Pharma AG: Employment. Kramer:Novartis Pharma AG: Employment. Brooks:Spouse works for Boston Biomedical Inc: Employment.


2021 ◽  
Vol 12 (6) ◽  
Author(s):  
Virginia Egea ◽  
Kai Kessenbrock ◽  
Devon Lawson ◽  
Alexander Bartelt ◽  
Christian Weber ◽  
...  

AbstractBone marrow-derived human mesenchymal stem cells (hMSCs) are recruited to damaged or inflamed tissues where they contribute to tissue repair. This multi-step process involves chemokine-directed invasion of hMSCs and on-site release of factors that influence target cells or tumor tissues. However, the underlying molecular mechanisms are largely unclear. Previously, we described that microRNA let-7f controls hMSC differentiation. Here, we investigated the role of let-7f in chemotactic invasion and paracrine anti-tumor effects. Incubation with stromal cell-derived factor-1α (SDF-1α) or inflammatory cytokines upregulated let-7f expression in hMSCs. Transfection of hMSCs with let-7f mimics enhanced CXCR4-dependent invasion by augmentation of pericellular proteolysis and release of matrix metalloproteinase-9. Hypoxia-induced stabilization of the hypoxia-inducible factor 1 alpha in hMSCs promoted cell invasion via let-7f and activation of autophagy. Dependent on its endogenous level, let-7f facilitated hMSC motility and invasion through regulation of the autophagic flux in these cells. In addition, secreted let-7f encapsulated in exosomes was increased upon upregulation of endogenous let-7f by treatment of the cells with SDF-1α, hypoxia, or induction of autophagy. In recipient 4T1 tumor cells, hMSC-derived exosomal let-7f attenuated proliferation and invasion. Moreover, implantation of 3D spheroids composed of hMSCs and 4T1 cells into a breast cancer mouse model demonstrated that hMSCs overexpressing let-7f inhibited tumor growth in vivo. Our findings provide evidence that let-7f is pivotal in the regulation of hMSC invasion in response to inflammation and hypoxia, suggesting that exosomal let-7f exhibits paracrine anti-tumor effects.


2021 ◽  
Vol 25 (1) ◽  
Author(s):  
Thakoon Thitiset ◽  
Siriporn Damrongsakkul ◽  
Supansa Yodmuang ◽  
Wilairat Leeanansaksiri ◽  
Jirun Apinun ◽  
...  

Abstract Background A novel biodegradable scaffold including gelatin (G), chitooligosaccharide (COS), and demineralized bone matrix (DBM) could play a significant part in bone tissue engineering. The present study aimed to investigate the biological characteristics of composite scaffolds in combination of G, COS, and DBM for in vitro cell culture and in vivo animal bioassays. Methods Three-dimensional scaffolds from the mixture of G, COS, and DBM were fabricated into 3 groups, namely, G, GC, and GCD using a lyophilization technique. The scaffolds were cultured with mesenchymal stem cells (MSCs) for 4 weeks to determine biological responses such as cell attachment and cell proliferation, alkaline phosphatase (ALP) activity, calcium deposition, cell morphology, and cell surface elemental composition. For the in vivo bioassay, G, GC, and GCD, acellular scaffolds were implanted subcutaneously in 8-week-old male Wistar rats for 4 weeks and 8 weeks. The explants were assessed for new bone formation using hematoxylin and eosin (H&E) staining and von Kossa staining. Results The MSCs could attach and proliferate on all three groups of scaffolds. Interestingly, the ALP activity of MSCs reached the greatest value on day 7 after cultured on the scaffolds, whereas the calcium assay displayed the highest level of calcium in MSCs on day 28. Furthermore, weight percentages of calcium and phosphorus on the surface of MSCs after cultivation on the GCD scaffolds increased when compared to those on other scaffolds. The scanning electron microscopy images showed that MSCs attached and proliferated on the scaffold surface thoroughly over the cultivation time. Mineral crystal aggregation was evident in GC and greatly in GCD scaffolds. H&E staining illustrated that G, GC, and GCD scaffolds displayed osteoid after 4 weeks of implantation and von Kossa staining confirmed the mineralization at 8 weeks in G, GC, and GCD scaffolds. Conclusion The MSCs cultured in GCD scaffolds revealed greater osteogenic differentiation than those cultured in G and GC scaffolds. Additionally, the G, GC, and GCD scaffolds could promote in vivo ectopic bone formation in rat model. The GCD scaffolds exhibited maximum osteoinductive capability compared with others and may be potentially used for bone regeneration.


PLoS ONE ◽  
2012 ◽  
Vol 7 (10) ◽  
pp. e48654 ◽  
Author(s):  
Giovanna Bianchi ◽  
Fabio Morandi ◽  
Michele Cilli ◽  
Antonio Daga ◽  
Chiara Bocelli-Tyndall ◽  
...  

2021 ◽  
Vol 30 ◽  
pp. 096368972097539
Author(s):  
Akiko Toyota ◽  
Rei Shinagawa ◽  
Mikiko Mano ◽  
Kazuyuki Tokioka ◽  
Naoto Suda

Cleft lip and palate is a congenital disorder including cleft lip, and/or cleft palate, and/or alveolar cleft, with high incidence.The alveolar cleft causes morphological and functional abnormalities. To obtain bone bridge formation and continuous structure between alveolar clefts, surgical interventions are performed from infancy to childhood. However, desirable bone bridge formation is not obtained in many cases. Regenerative medicine using mesenchymal stem cells (MSCs) is expected to be a useful strategy to obtain sufficient bone bridge formation between alveolar clefts. In this study, we examined the effect of human umbilical cord-derived MSCs by transplantation into a rat experimental alveolar cleft model. Human umbilical cords were digested enzymatically and the isolated cells were collected (UC-EZ cells). Next, CD146-positive cells were enriched from UC-EZ cells by magnetic-activated cell sorting (UC-MACS cells). UC-EZ and UC-MACS cells showed MSC gene/protein expression, in vitro. Both cells had multipotency and could differentiate to osteogenic, chondrogenic, and adipogenic lineages under the differentiation-inducing media. However, UC-EZ cells lacked Sox2 expression and showed the lower ratio of MSCs than UC-MACS cells. Thus, UC-MACS cells were transplanted with hydroxyapatite and collagen (HA + Col) into alveolar cleft model to evaluate bone formation in vivo. The results of micro computed tomography and histological staining showed that UC-MACS cells with HA + Col induced more abundant bone formation between the experimental alveolar clefts than HA + Col implantation only. Cells immunopositive for osteopontin were accumulated along the bone surface and some of them were embedded in the bone. Cells immunopositive for human-specific mitochondria were aligned along the newly formed bone surface and in the new bone, suggesting that UC-MACS cells contributed to the bone bridge formation between alveolar clefts. These findings indicate that human umbilical cords are reliable bioresource and UC-MACS cells are useful for the alveolar cleft regeneration.


Sign in / Sign up

Export Citation Format

Share Document