Endogenous Expression of Functional Factor VIII by Human Mesenchymal Stem Cells In Culture

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2216-2216
Author(s):  
Chad Sanada ◽  
Evan J Colletti ◽  
Melisa Soland ◽  
Chung-Jung Kuo ◽  
Christopher D Porada ◽  
...  

Abstract Abstract 2216 The liver is considered to be the primary site of factor VIII (FVIII) production in the body; however, evidence is mounting that suggests there are secondary sites in which considerable synthesis of FVIII takes place. Studies of FVIII mRNA expression in various human tissues have revealed that FVIII message can be found throughout the body. Additionally, acute liver failure correlates with an increase in circulating FVIII levels. Some reports have identified endothelial cells as a significant extra-hepatic source of FVIII, possibly explaining both the widespread presence of FVIII mRNA and the increase in FVIII levels upon liver failure. However, the possibility exists that other cell types present throughout the body also produce FVIII and contribute to circulating FVIII levels. Mesenchymal Stem Cells (MSCs) represent a potential alternative; they are a diverse group of stromal cells which can be found in the perivascular regions of multiple tissues throughout the body. Previous studies demonstrated that MSCs are capable of efficiently producing and secreting high levels of FVIII in vitro when transduced with FVIII-encoding viral vectors, but to date, the innate ability of MSCs to produce FVIII has not been explored. As such, we investigated the potential for MSCs to produce endogenous FVIII message and secrete functional protein. MSCs isolated based on Stro-1 positivity from human lung, liver, brain, and bone marrow (BM) were grown in cell culture and assayed for production of FVIII message by both microarray analysis and RT-PCR. Microarray data showed that there were significant amounts of FVIII message in all four cell types tested and that the amount of message in BM MSCs was three-fold higher than each of the other three cell types. RT-PCR analysis confirmed the presence of FVIII message in all four MSC populations. Secretion of functional FVIII protein was subsequently measured using a chromogenic assay. MSC culture supernatants were collected for either 24 or 48 hours, and FVIII activity was determined using pooled normal human plasma as a control to create a standard curve. FVIII activity in the supernatants of MSCs was in the range of 0.6 to 2.0 mU/1×10^6 cells/ 24hr. Moreover, MSCs continued to express and produce FVIII during time in culture until our last evaluation at passage 20, indicating that there is an innate ability of these cells to continually produce FVIII. Taken together, these data demonstrate that human MSCs are capable of producing and secreting functional FVIII in vitro, and given their widespread location throughout the body, this finding raises the possibility that, in vivo, these cells might significantly contribute to the total FVIII pool. This is the first report, to our knowledge, that implicates MSCs as a potential endogenous source for circulating FVIII. Further studies of in vivo FVIII expression by MSCs are warranted and may provide a clearer understanding of extra-hepatic FVIII production in the body while aiding in the discovery of novel therapies for hemophilia A. Disclosures: No relevant conflicts of interest to declare.

Biomolecules ◽  
2020 ◽  
Vol 10 (9) ◽  
pp. 1306
Author(s):  
Ann-Kristin Afflerbach ◽  
Mark D. Kiri ◽  
Tahir Detinis ◽  
Ben M. Maoz

The human-relevance of an in vitro model is dependent on two main factors—(i) an appropriate human cell source and (ii) a modeling platform that recapitulates human in vivo conditions. Recent years have brought substantial advancements in both these aspects. In particular, mesenchymal stem cells (MSCs) have emerged as a promising cell source, as these cells can differentiate into multiple cell types, yet do not raise the ethical and practical concerns associated with other types of stem cells. In turn, advanced bioengineered in vitro models such as microfluidics, Organs-on-a-Chip, scaffolds, bioprinting and organoids are bringing researchers ever closer to mimicking complex in vivo environments, thereby overcoming some of the limitations of traditional 2D cell cultures. This review covers each of these advancements separately and discusses how the integration of MSCs into novel in vitro platforms may contribute enormously to clinical and fundamental research.


2018 ◽  
Vol 2018 ◽  
pp. 1-10 ◽  
Author(s):  
Li Chen ◽  
Jiexin Zhang ◽  
Lu Yang ◽  
Guoying Zhang ◽  
Yingjie Wang ◽  
...  

Mesenchymal stem cells (MSCs) and hepatocytes are two attractive sources of cell-based therapies for acute liver failure (ALF). The cotransplantation of hepatocytes with MSCs can improve the therapeutic performance for the treatment of ALF. However, the therapeutic potential of conditioned medium (CM) derived from MSCs cocultured with hepatocytes (MSC-H-CM) remains unclear. The purpose of this study was to investigate the effects of MSC-H-CM on damaged hepatocytes in vitro and on D-galactosamine-induced ALF in vivo. D-Galactosamine-treated L02 cells cultured in MSC-H-CM exhibited higher of cell viability and total protein synthesis than L02 cells cultured in MSC-CM, CM derived from hepatocytes (H-CM), MSC-CM + H-CM, or with nonconditioned medium (NCM). Lactate dehydrogenase and aspartate aminotransferase levels were lower in the supernatant of damaged L02 cells cultured in MSC-H-CM than in that of L02 cells cultured in other types of CM. The lowest percentage of apoptotic cells was observed after the MSC-H-CM treatment. When CM was injected into the tail vein of rats with ALF, MSC-H-CM was the most successful at preventing the release of liver injury biomarkers and in promoting the recovery of liver structure. The greatest survival rate 7 days after the first treatment was observed in the MSC-H-CM-treated rats. Our results reveal that the delivery of MSC-H-CM could be a novel strategy for integrating the therapeutic potentials of hepatocytes and MSCs for the treatment of ALF.


2017 ◽  
Vol 2017 ◽  
pp. 1-11 ◽  
Author(s):  
Huan Liao ◽  
Hongxuan Wang ◽  
Xiaoming Rong ◽  
Enqin Li ◽  
Ren-He Xu ◽  
...  

Radiation-induced brain injury (RI) commonly occurs in patients who received head and neck radiotherapy. However, the mechanism of RI remains unclear. We aimed to evaluate whether pyroptosis was involved in RI and the impact of mesenchymal stem cells (MSCs) on it. BALB/c male mice (6–8 weeks) were cranially irradiated (15 Gy), and MSCs were transplanted into the bilateral cortex 2 days later; then mice were sacrificed 1 month later. Meanwhile, irradiated BV-2 microglia cells (10 Gy) were cocultured with MSCs for 24 hours. We observed that irradiated mice brains presented NLRP3 and caspase-1 activation. RT-PCR then indicated that it mainly occurred in microglia cells but not in neurons. Further, irradiated BV-2 cells showed pyroptosis and increased production of IL-18 and IL-1β. RT-PCR also demonstrated an increased expression of several inflammasome genes in irradiated BV-2 cells, including NLRP3 and AIM2. Particularly, NLRP3 was activated. Knockdown of NLRP3 resulted in decreased LDH release. Noteworthily, in vivo, MSCs transplantation alleviated radiation-induced NLRP3 and caspase-1 activation. Moreover, in vitro, MSCs could decrease caspase-1 dependent pyroptosis, NLRP3 inflammasome activation, and ROS production induced by radiation. Thus, our findings proved that microglia pyroptosis occurred in RI. MSCs may act as a potent therapeutic tool in attenuating pyroptosis.


2001 ◽  
Vol 73 (4) ◽  
pp. 533-545 ◽  
Author(s):  
ELISABETH DUPIN ◽  
CARLA REAL ◽  
NICOLE LeDOUARIN

How the considerable diversity of neural crest (NC)-derived cell types arises in the vertebrate embryo has long been a key question in developmental biology. The pluripotency and plasticity of differentiation of the NC cell population has been fully documented and it is well-established that environmental cues play an important role in patterning the NC derivatives throughout the body. Over the past decade, in vivo and in vitro cellular approaches have unravelled the differentiation potentialities of single NC cells and led to the discovery of NC stem cells. Although it is clear that the final fate of individual cells is in agreement with their final position within the embryo, it has to be stressed that the NC cells that reach target sites are pluripotent and further restrictions occur only late in development. It is therefore a heterogenous collection of cells that is submitted to local environmental signals in the various NC-derived structures. Several factors were thus identified which favor the development of subsets of NC-derived cells in vitro. Moreover, the strategy of gene targeting in mouse has led at identifying new molecules able to control one or several aspects of NC cell differentiation in vivo. Endothelin peptides (and endothelin receptors) are among those. The conjunction of recent data obtained in mouse and avian embryos and reviewed here contributes to a better understanding of the action of the endothelin signaling pathway in the emergence and stability of NC-derived cell phenotypes.


2016 ◽  
Vol 12 ◽  
pp. 38-47 ◽  
Author(s):  
Aleksandra Skubis ◽  
Bartosz Sikora ◽  
Nikola Zmarzły ◽  
Emilia Wojdas ◽  
Urszula Mazurek

This review article provides an overview on adipose-derived stem cells (ADSCs) for implications in bone tissue regeneration. Firstly this article focuses on mesenchymal stem cells (MSCs) which are object of interest in regenerative medicine. Stem cells have unlimited potential for self-renewal and develop into various cell types. They are used for many therapies such as bone tissue regeneration. Adipose tissue is one of the main sources of mesenchymal stem cells (MSCs). Regenerative medicine intends to differentiate ADSC along specific lineage pathways to effect repair of damaged or failing organs. For further clinical applications it is necessary to understand mechanisms involved in ADSCs proliferation and differentiation. Second part of manuscript based on osteogenesis differentiation of stem cells. Bones are highly regenerative organs but there are still many problems with therapy of large bone defects. Sometimes there is necessary to make a replacement or expansion new bone tissue. Stem cells might be a good solution for this especially ADSCs which manage differentiate into osteoblast in in vitro and in vivo conditions.


2014 ◽  
Vol 26 (1) ◽  
pp. 207
Author(s):  
B. Rossi ◽  
B. Merlo ◽  
E. Iacono ◽  
P. P. Pagliaro ◽  
P. L. Tazzari ◽  
...  

In recent years, fetal adnexa and fluids have been recognised as important sources of mesenchymal stem cells (MSC). The aim of this study was to characterise cell populations of bovine amniotic fluid, studying phenotypic characterisation, RNA expression, and differentiation potential of samples after in vitro culture for different lengths of time following trypsinization and expansion (passage). Amniotic fluid samples were recovered at the slaughterhouse from 25 pregnant cows and harvested cells were cultured in DMEM-TCM199 (1 : 1) plus 10% fetal bovine serum (FBS) in 5% CO2 at 38.5°C. At passages P3 and P7, a sample for each of the 4 population found was characterised. Immunophenotypic characterisation was performed for MSC (CD90, CD105, CD44) and haematopoietic (CD14, CD34) markers by flow cytometry (FACS). Immunocytochemistry (ICC) was performed for Oct4, SSEA4, and α-SMA and the ratio between positive cells and total nuclei was evaluated. Gene expression profile was analysed by RT–PCR for pluripotency markers (Oct4, Nanog, Sox2). At the same passages chondrogenic, osteogenic and adipogenic differentiation were induced and evaluated morphologically and cytologically using, respectively, Alcian blue to identify cartilage matrix, Von Kossa for extracellular calcium deposition, and Oil Red O for intracellular lipid droplets. Cell population appeared heterogeneous and we could identify 2 main cell types: round (R) and spindle-shaped (S) cells. Each isolated sample was classified into one of the following 4 types depending on percentages of R or S cells: prevalence of S-cells (S), prevalence of R-cells (R), and samples showing both morphologies with ~10% of S-cells (S10) or 40% S-cells (S40). S-cells percentage decreased with passages in S10 and S40. After FACS, all lines were positive for CD90, CD105, CD44, and CD34 and negative for CD14 both at P3 and at P7. After ICC, Oct4 was negative in all samples analysed, few S cells stained for SSEA4 (8%) at P3 but increased at P7 to 22%; R, S10, and S40 did not express SSEA4 both at P3 and at P7. α-SMA was expressed in all samples at P3 (9.4% S; 0.9% R; 2.5% S10; 27% S40) but not at P7 (27.5% S; 0% R; 0% S10; 0% S40). After RT–PCR analyses, Oct4 was negative in all samples; at P3, Nanog was clearly positive in S-cells, weak in S40, and negative in R and S10, but all samples turned negative at P7. Sox2 was weakly expressed (S) or not expressed (S10, S40, R) at P3 and it was negative in all cells at P7. Only S showed high differentiation potential into all 3 lineages at both P3 and P7, R had the lowest differentiation potential, whereas S10 and S40 were intermediate at both end points. In conclusion, bovine amniotic fluid showed heterogeneous cell populations and S-type had the characteristics of MSCs. S10 and S40 showed more MSC markers at P3, when S cells were still present, and this aspect suggests that S population is the presumptive MSC one. Although prevalent, R-type showed only some MSC characteristics. Further studies are under way to improve S-type isolation, purification, and culture, and to determine the lifespan of these cell types. This work was supported by grant PRIN2009.


2021 ◽  
Author(s):  
Lucile Fievet ◽  
Nicolas Serratrice ◽  
Benedicte Brulin ◽  
Laurent Giraudo ◽  
Julie Veran ◽  
...  

Bone repair induced by stem cells and biomaterials may represent an alternative to autologous bone grafting. Here, we compared the efficiency of two biomaterials - biphasic calcium phosphate (BCP) and bioactive glass (BG) - when loaded with either adult bone marrow mesenchymal stem cells (BM-MSCs) or newborn nasal ecto-mesenchymal stem cells (NE-MSCs), the latter being collected for further repair of lip cleft-associated bone loss. Both cell types display the typical stem cell surface markers CD73+/CD90+/CD105+/nestin, and exhibit the MSC-associated osteogenic, chondrogenic and adipogenic multipotency. NE-MSCs produce less collagen and alkaline phosphatase than BM-MSCs. At the transcript level, NE-MSCs express more abundantly three genes coding for bone sialoprotein, osteocalcin and osteopontin, while BM-MSCs produce extra copies of RUNX2. BM-MSCs and NE-MSCs adhere and survive on BCP and BG. In vivo experiments reveal that bone formation is only observed with BM-MSCs transplanted on BCP biomaterial.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4675-4675
Author(s):  
Jishi Wang ◽  
Dan Ma ◽  
Yan Li ◽  
Qin Fang ◽  
Shuya Chen ◽  
...  

Abstract Abstract 4675 Objective: Bone mesenchymal stem cells (BMSCs) possessing immunoregulatory activities have been evaluated in the treatment of graft-versus-host disease (GVHD). In this study, mice's heme oxygenase 1 (HO-1) was transduced into mice's bone marrow-derived mesenchymal stem cells (mBMSCs), we assessed the immuno-suppressive capacity of lentiviral vector transduced BMSCs expressing HO-1 in BALB/c mice aGVHD model, and the immuno-regulatory effect of mBMSCs on alleviating acute GVHD in vivo was measured to provide laboratory data for gene therapy for aGVHD which used mBMSCs as vehicles. Methods: We cloned mice's HO-1 cDNA from mice's bone marrow and constructed recombinant lentivirus vectors (Lentivirus-V5-D-TOPO-HO-1-EGFP/Lentivirus–V 5-D-TOPO-EGFP) which titer was 1×1011 pfu/mL. These mBMSCs were separated, cultured, purified, and detected by morphology, flow cytometry, osteogenic, adipogenic and chondrogenic induction, and the mRNA level of the neural ganglioside GD2 gene which is a surface marker for the identification of MSCs by RT-PCR. Then recombinant vectors were transferred into mBMSCs, and the expression of EGFP and HO-1 were detected by fluorescence microscope, RT-PCR and Western blot respectively. The immunsuppressive capacity of HO-1 over-expressing mBMSCs was investigated using transwell assay in vitro. And before infusion, the homing of MSC was detected by Flow cytometry. In addition, we established BALB/c mice's aGVHD model after Allo-HSCT, the recombinant vectors tranfected mBMSCs and primary mBMSCs were injected into the BALB/c mice aGVHD model in tail vein, respectively. Four groups were separated in vivo test (Group A: aGVHD control; Group B: aGVHD model injected in mBMSCs; Group C: aGVHD model injected in mBMSCs transfected with EGFP; Group D: aGVHD model injected in mBMSCs transfected with HO-1 gene.) The survival, body weight and clinical score of GVHD in transplanted mice were monitored to evaluate the severity of aGVHD. The aGVHD targeted organ, such as Liver, intestine and lung from mice in each group were obtained for histological examination and pathological score. Plasma concentrations of interleukin (IL)-2, IL-4, IL-6, IL-10, IFN-γ and TNF-γ were also determined using a Cytometric Bead Array. Results: We cloned mice HO-1 gene from mice's bone marrow and constructed the recombinant adenovirus vectors (Lentivirus-V5-D-TOPO-HO-1- EGFP/Lentivirus-V5-D-TOPO-EGFP) successfully. mBMSCs was separated and identified successfully. Fluorescence microscope detected the expression of EGFP, while both RT-PCR and Western blot detected high expression of HO-1 in gene-transfected group cells. It clearly showed that mBMSCs in Group D can migrate more through the polycarbonate filter toward bone marrow micro-environment in the lower chamber than Group A, B, C in vitro, while homing of Group D showed similar trend compared with others groups. In the mice GVHD model, treatment with HO-1 over-expressing BMSCs significantly decreased the mortality rate and attenuated clinical and pathological GVHD scores, and volume of spleen was not more obviously enlarged than others groups. Moreover, compared with control groups, the plasma IL-2, IL-6, IFN-γ and TNF-γ levels in recipients infused with HO-1 over-expressing BMSCs were significantly decreased, while those of IL-4 and IL-10 were increased. Conclusion: Lentiviral vectors carrying the HO-1 were successfully used to transduce mBMSCs. In our report, mBMSCs transferred with lentiviral vectors expressed strongerly immunoregulatory activities to alleviate aGVHD. Not only did HO-1 enhance that migration of mBMSCs, but also strengthen homing of MSCs. In vivo experiments, the evidence of survival rate, body weight, clinical score and pathological score fully proved that HO-1-transduced BMSCs effectively controlled the occurrence of mice's aGVHD following allogeneic BM transplantation, and HO-1 may be a potential target to overcome aGVHD in vivo. Disclosures: Ma: National Natural Science Foundation of China: Research Funding. Li:National Natural Science Foundation of China: Research Funding. Fang:National Natural Science Foundation of China: Research Funding. Chen:National Natural Science Foundation of China: Research Funding. Sun:National Natural Science Foundation of China: Research Funding.


2017 ◽  
Vol 204 (2) ◽  
pp. 59-83 ◽  
Author(s):  
Ahmad Oryan ◽  
Amir Kamali ◽  
Ali Moshiri ◽  
Mohamadreza Baghaban Eslaminejad

Healing and regeneration of bone injuries, particularly those that are associated with large bone defects, are a complicated process. There is growing interest in the application of osteoinductive and osteogenic growth factors and mesenchymal stem cells (MSCs) in order to significantly improve bone repair and regeneration. MSCs are multipotent stromal stem cells that can be harvested from many different sources and differentiated into a variety of cell types, such as preosteogenic chondroblasts and osteoblasts. The effectiveness of MSC therapy is dependent on several factors, including the differentiating state of the MSCs at the time of application, the method of their delivery, the concentration of MSCs per injection, the vehicle used, and the nature and extent of injury, for example. Tissue engineering and regenerative medicine, together with genetic engineering and gene therapy, are advanced options that may have the potential to improve the outcome of cell therapy. Although several in vitro and in vivo investigations have suggested the potential roles of MSCs in bone repair and regeneration, the mechanism of MSC therapy in bone repair has not been fully elucidated, the efficacy of MSC therapy has not been strongly proven in clinical trials, and several controversies exist, making it difficult to draw conclusions from the results. In this review, we update the recent advances in the mechanisms of MSC action and the delivery approaches in bone regenerative medicine. We will also review the most recent clinical trials to find out how MSCs may be beneficial for treating bone defects.


2013 ◽  
Vol 5 (3) ◽  
pp. 151
Author(s):  
Anna Meiliana ◽  
Andi Wijaya

BACKGROUND: Blood vessels are a source of stem and progenitor cells, which likely contribute to a variety of vascular processes and diseases. Emerging concepts in this field could influence therapeutic approaches to diseases of blood vessels such as atherosclerosis.CONTENT: Vascular Stem Cells (VSCs) field is only beginning to emerge, and thus, many issues regarding VSCs’s identity and function remain poorly understood. In fact, even after decades of intensive research, Mesenchymal Stem Cells (MSC), which is suggested to be VSCs, is still having many outstanding issues of its own. And, on top of this, likewise decades-long intensive pericyte research has not been able resolve the identity issue. While favors Adventitial Progenitor Cells (APCs) over pericytes as the likely VSC candidate, it should be pointed out that currently the opposite view (i.e., pericytes as VSCs) is more prevalent, and many excellent reviews, including a recent one, have discussed this issue extensively.SUMMARY: It has been postulated that, within the vasculature, APCs could differentiate into pericytes (CD34- CD31- CD140b+ SMA-), endothelial cells (CD34+ CD31+ CD140b- SMA-), and smooth muscle cells (SMCs) (CD34- CD31- CD140b- SMA+); and during tissue expansion or repair, APCs could also differentiate into tissue-specific cell types (e.g., muscle and fat) Thus, in vitro, APCs fulfill all criteria for being VSCs. Meanwhile, in vivo evidence is still limited and will require further investigation.KEYWORDS: vascular stem cells, VSC, mesenchymal stem cells, MSC, endothelial progenitor cells, EPC, adventitial progenitor cells, APC


Sign in / Sign up

Export Citation Format

Share Document