scholarly journals The impact of cryopreservation on bone marrow-derived mesenchymal stem cells: a systematic review

2019 ◽  
Vol 17 (1) ◽  
Author(s):  
Soukaina Bahsoun ◽  
Karen Coopman ◽  
Elizabeth C. Akam

AbstractMesenchymal stem cells (MSCs) represent an invaluable asset for the field of cell therapy. Human Bone marrow-derived MSCs (hBM-MSCs) are one of the most commonly used cell types in clinical trials. They are currently being studied and tested for the treatment of a wide range of diseases and conditions. The future availability of MSCs therapies to the public will require a robust and reliable delivery process. Cryopreservation represents the gold standard in cell storage and transportation, but its effect on BM-MSCs is still not well established. A systematic review was conducted to evaluate the impact of cryopreservation on BM-MSCs and to attempt to uncover the reasons behind some of the controversial results reported in the literature. Forty-one in vitro studies were analysed, and their results organised according to the cell attributes they assess. It was concluded that cryopreservation does not affect BM-MSCs morphology, surface marker expression, differentiation or proliferation potential. However, mixed results exist regarding the effect on colony forming ability and the effects on viability, attachment and migration, genomic stability and paracrine function are undefined mainly due to the huge variabilities governing the cryopreservation process as a whole and to the lack of standardised assays.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 4742-4742
Author(s):  
Tatiana Ribeiro ◽  
Aurelie Picard ◽  
Elfi Ducrocq ◽  
Alain Langonne ◽  
Philippe Rosset ◽  
...  

Abstract The bone marrow (BM) hematopoietic stem cell (HSC) niche is a specialized structure of the microenvironment, which supports survival and regulates HSC function (i.e. the HSC control of the self-renewal/differentiation balance and migration). The supportive cells involved in the HSC niche are usually named as “stromal cells” but their precise nature remains a matter of debate (in particular, to know whether these cells belong to osteoblastic or to vascular smooth muscle lineage). Mesenchymal stem cells (MSCs) that are present into the BM are characterized by a broad differentiation potential including adipocytic (A), osteoblastic (O) and vascular smooth muscle (V) pathways. Although MSCs are believed to be at the origin of stromal cells, their real function within the niche is unknown. The aim of this study was to investigate in vitro the hematopoietic function (HSC support and migration) of cultured adult BM MSCs non-differentiated and during induced differentiation along A, O and V lineages. MSCs were obtained from BM nucleated cells of patients undergoing orthopedic surgery by culture in expansion medium (alpha-MEM medium with 10% FCS and 1 ng/mL FGF-2). The MSCs were tested before (cultured in expansion medium) and during differentiation induction in appropriate medium for A, O or V lineages (from 3 to 21 days). Interestingly, non-differentiated MSCs already co-expressed O (PTH-receptor), A (leptin) and V (ASMA) markers as assessed by Western blotting. Capacity of MSCs to support hematopoiesis was evaluated by long-term cultures (for 5 wks) with BM CD34+ cells in limiting dilution (CAFC assay), and capacity to control CD34+ cell migration by using Transwells seeded with MSCs (trans-stromal migration assay). We showed that non-differentiated MSCs have the most important capacity to support hematopoiesis (5-week CAFC frequency) and that this capacity was quickly and dramatically lost from 3 days of differentiation towards A (36±2% of non-differentiated values), O (40±3%) and V (38±1%) lineages. This capacity was almost abolished after 14 days of A, O and V differentiation (<5%). In parallel, CD34+ cell migration was clearly reduced through 3-day A and O differentiated MSCs, while it was increased through 3-day V differentiated MSCs (5 fold). These results show that MSCs maintained in vitro in non-differentiated state (although already expressing some A, O and V markers), display the strongest hematopoietic supportive activity compared to MSCs induced to differentiate into adipocytic, osteoblastic, or vascular smooth muscle lineages. Therefore, the stromal cell function could be supported by a cell close to a non-differentiated MSC in endosteal or perivascular niches as well. In contrast, vascular smooth muscle differentiated MSCs at advanced stages could be devoted rather to HSC migration control than to HSC support.


Author(s):  
Dhanasekaran Marappagounder ◽  
Indumathi Somasundaram ◽  
Sudarsanam Dorairaj ◽  
Rajkumar Sankaran

AbstractAlthough stem cells are present in various adult tissues and body fluids, bone marrow has been the most popular source of stem cells for treatment of a wide range of diseases. Recent results for stem cells from adipose tissue have put it in a position to compete for being the leading therapeutic source. The major advantage of these stem cells over their counterparts is their amazing proliferative and differentiation potency. However, their pancreatic lineage transdifferentiation competence was not compared to that for bone marrow-derived stem cells. This study aims to identify an efficient source for transdifferentiation into pancreatic islet-like clusters, which would increase potential application in curative diabetic therapy. The results reveal that mesenchymal stem cells (MSC) derived from bone marrow and subcutaneous adipose tissue can differentiate into pancreatic islet-like clusters, as evidenced by their islet-like morphology, positive dithizone staining and expression of genes such as Nestin, PDX1, Isl 1, Ngn 3, Pax 4 and Insulin. The pancreatic lineage differentiation was further corroborated by positive results in the glucose challenge assay. However, the results indicate that bone marrow-derived MSCs are superior to those from subcutaneous adipose tissue in terms of differentiation into pancreatic islet-like clusters. In conclusion, bone marrow-derived MSC might serve as a better alternative in the treatment of diabetes mellitus than those from adipose tissue.


Cancers ◽  
2020 ◽  
Vol 13 (1) ◽  
pp. 62
Author(s):  
Elise Aasebø ◽  
Even Birkeland ◽  
Frode Selheim ◽  
Frode Berven ◽  
Annette K. Brenner ◽  
...  

Mesenchymal stem cells (MSCs) and osteoblasts are bone marrow stromal cells that contribute to the formation of stem cell niches and support normal hematopoiesis, leukemogenesis and development of metastases from distant cancers. This support is mediated through cell–cell contact, release of soluble mediators and formation of extracellular matrix. By using a proteomic approach, we characterized the protein release by in vitro cultured human MSCs (10 donors) and osteoblasts (nine donors). We identified 1379 molecules released by these cells, including 340 proteins belonging to the GO-term Extracellular matrix. Both cell types released a wide range of functionally heterogeneous proteins including extracellular matrix molecules (especially collagens), several enzymes and especially proteases, cytokines and soluble adhesion molecules, but also several intracellular molecules including chaperones, cytoplasmic mediators, histones and non-histone nuclear molecules. The levels of most proteins did not differ between MSCs and osteoblasts, but 82 proteins were more abundant for MSC (especially extracellular matrix proteins and proteases) and 36 proteins more abundant for osteoblasts. Finally, a large number of exosomal proteins were identified. To conclude, MSCs and osteoblasts show extracellular release of a wide range of functionally diverse proteins, including several extracellular matrix molecules known to support cancer progression (e.g., metastases from distant tumors, increased relapse risk for hematological malignancies), and the large number of identified exosomal proteins suggests that exocytosis is an important mechanism of protein release.


2011 ◽  
Vol 23 (1) ◽  
pp. 253 ◽  
Author(s):  
E. Monaco ◽  
M. Bionaz ◽  
A. Lima ◽  
W. L. Hurley ◽  
M. B. Wheeler

Previous data support adipose-derived stem cells as an alternative to bone marrow as a source of adult stem cells for therapeutic purposes. The aim of the present study was to directly compare the transcriptome of adipose-derived (ADSC) and bone marrow-derived (BMSC) mesenchymal stem cells prior to differentiation and during in vitro osteogenic and adipogenic differentiation. The ADSC and BMSC were harvested from 3 adult pigs and differentiated in vitro into adipocytes and osteocytes for up to 4 weeks. Prior to differentiation and at differentiation day 2, 7, and 21, cells were harvested and RNA extracted for transcriptomics analysis by a 13 263 oligo 70-mers array (Sus scrofa AROS V1.0 with extension; Operon). Data were normalized by Lowess and statistical analysis was run using ANOVA with Benjamini-Hochberg false discovery rate (FDR) correction. Data mining was carried out using Ingenuity Pathway Analysis and David. Using an FDR of <0.05 for overall tissue effect and a post-hoc correction of P < 0.001, we observed 65 differentially expressed genes (DEG) between ADSC and BMSC before starting differentiation (0.66% of unique genes in the array). Functional analysis uncovered significant enrichment of extracellular matrix genes with direct roles in cell adhesion, migration, movement, and morphology. When the interaction cell type × differentiation × time was assessed, we observed >2 000 DEG with an FDR <0.05. This large number was mostly due to time effects. When pair-wise comparisons between cell types for each time point during the same differentiation were performed (post-hoc P < 0.001), we observed a strikingly low number of DEG. The number of DEG was lower between cell types in osteogenic (<100 DEG) compared with adipogenic (<200 DEG) differentiation. We observed significant enrichment (FDR-corrected P-value cut-off <0.05) of functions related to metabolism, antigen presentation, angiogenesis, and cell cycle in both differentiation conditions. We also observed an overall greater induction of the enriched functions in ADSC and a decrease in BMSC during adipogenic differentiation and the opposite during osteogenic differentiation except for metabolism, which appeared to be larger in ADSC in all cases. Among the significant enriched functions of DEG between the 2 differentiations, we observed enrichment of genes involved in metabolism, cell death, cell-to-cell signalling, and antigen presentation in ADSC during adipogenic compared with osteogenic differentiation. In BMSC we observed enrichment of functions related to cell death, antigen presentation, and lipid metabolism in osteogenic v. adipogenic differentiation. Overall data uncovered a high similarity at the transcriptional level between ADSC and BMSC both prior to differentiation and during differentiation. Those data support ADSC being particularly similar to BMSC. This work was support by the Illinois Regenerative Medicine Institute (IDPH # 63080017).


Cells ◽  
2020 ◽  
Vol 9 (9) ◽  
pp. 2072 ◽  
Author(s):  
Maria V. Guijarro ◽  
Laura S. Danielson ◽  
Marta Cañamero ◽  
Akbar Nawab ◽  
Carolina Abrahan ◽  
...  

TSC1 is a tumor suppressor that inhibits cell growth via negative regulation of the mammalian target of rapamycin complex (mTORC1). TSC1 mutations are associated with Tuberous Sclerosis Complex (TSC), characterized by multiple benign tumors of mesenchymal and epithelial origin. TSC1 modulates self-renewal and differentiation in hematopoietic stem cells; however, its effects on mesenchymal stem cells (MSCs) are unknown. We investigated the impact of Tsc1 inactivation in murine bone marrow (BM)-MSCs, using tissue-specific, transgelin (Tagln)-mediated cre-recombination, targeting both BM-MSCs and smooth muscle cells. Tsc1 mutants were viable, but homozygous inactivation led to a dwarfed appearance with TSC-like pathologies in multiple organs and reduced survival. In young (28 day old) mice, Tsc1 deficiency-induced significant cell expansion of non-hematopoietic BM in vivo, and MSC colony-forming potential in vitro, that was normalized upon treatment with the mTOR inhibitor, everolimus. The hyperproliferative BM-MSC phenotype was lost in aged (1.5 yr) mice, and Tsc1 inactivation was also accompanied by elevated ROS and increased senescence. ShRNA-mediated knockdown of Tsc1 in BM-MSCs replicated the hyperproliferative BM-MSC phenotype and led to impaired adipogenic and myogenic differentiation. Our data show that Tsc1 is a negative regulator of BM-MSC proliferation and support a pivotal role for the Tsc1-mTOR axis in the maintenance of the mesenchymal progenitor pool.


Author(s):  
Prithiv Kumar K R

Discrepant results due to lack of phenotype-specific species to species relevance to pharmacological results from in vitro cell testing cause inappropriate approaches to treatments to rare conditions in the brain. One such condition is Friedrich Ataxia. The prime focus is on this extreme condition, a neurodegenerative condition that sets precedence for clinical trials. Compounds such as nicotinamide and resveratrol were selected and used in the treatment of Ataxia, the condition shows no improvement. These compounds contracted various responses in iPSC derived neuron generation and more discoverable approach to drug delivery system in vitro conditions. The cell types from iPSC were considered nonrelevant for the disease derived from fibroblasts and more relevant to disease derived from neurons differentiated iPSC cells. This above illustrates the value of utilizing human iPSC cells to improve drug discovery and cure. The over the reacting theory of human iPSC cells are advanced further to discover the potent application of stem cells and further discuss the feasibility of transplantation of Friedrich Ataxia(FA) patients bone marrow-derived mesenchymal stem cells to discover the GAA expansion and reduced FXN and mRNA expression. There is scope for discussion for the assessment of the proliferative ability of pluripotency of FR from bone marrow. The characteristics of bone marrow in vitro differentiation have two types of cells, peripheral neurons, and cardiomyocytes. Also, this chapter leads to series of discussions on the need for testing applications on bone marrow mesenchymal stem cells, mutated gene correction that will play a critical role in gene-based cell therapy, and human artificial chromosomes intact with FXN genes differentiated into neurons or cardiomyocytes.


2014 ◽  
Vol 2014 ◽  
pp. 1-15 ◽  
Author(s):  
Claudia Ulbrich ◽  
Markus Wehland ◽  
Jessica Pietsch ◽  
Ganna Aleshcheva ◽  
Petra Wise ◽  
...  

How microgravity affects the biology of human cells and the formation of 3D cell cultures in real and simulated microgravity (r- and s-µg) is currently a hot topic in biomedicine. In r- and s-µg, various cell types were found to form 3D structures. This review will focus on the current knowledge of tissue engineering in space and on Earth using systems such as the random positioning machine (RPM), the 2D-clinostat, or the NASA-developed rotating wall vessel bioreactor (RWV) to create tissue from bone, tumor, and mesenchymal stem cells. To understand the development of 3D structures,in vitroexperiments using s-µgdevices can provide valuable information about modulations in signal-transduction, cell adhesion, or extracellular matrix induced by altered gravity conditions. These systems also facilitate the analysis of the impact of growth factors, hormones, or drugs on these tissue-like constructs. Progress has been made in bone tissue engineering using the RWV, and multicellular tumor spheroids (MCTS), formed in both r- and s-µg, have been reported and were analyzed in depth. Currently, these MCTS are available for drug testing and proteomic investigations. This review provides an overview of the influence ofµgon the aforementioned cells and an outlook for future perspectives in tissue engineering.


Author(s):  
Maria Camilla Ciardulli ◽  
Luigi Marino ◽  
Erwin P. Lamparelli ◽  
Maurizio Guida ◽  
Nicholas R. Forsyth ◽  
...  

Mesenchymal Stem Cells derived from bone marrow (hBM-MSCs) are utilized in tendon tissue‐engineering protocols while extra-embryonic cord-derived, including from Wharton&rsquo;s Jelly (hWJ-MSC), are emerging as useful alternatives. To explore the tenogenic responsiveness of hBM-MSCs and hWJ-MSCs to hGDF-5 we supplemented each at doses of 1, 10, and 100 ng/mL and determined proliferation, morphology and time-dependent expression of tenogenic markers. We evaluated expression of Collagen types 1 (COL1A1) and 3 (COL3A1), Decorin (DCN), Scleraxis A (SCX-A), Tenascin-C (TNC) and Tenomodulin (TNMD) noting the earliest and largest increase with 100 ng/mL. With 100 ng/mL, hBM-MSCs showed upregulation of SCX-A (1.7-fold) at day 1, TNC (1.3-fold) and TNMD (12-fold) at Day 8. hWJ-MSCs, at the same dose, showed up-regulation of COL1A1 (3-fold), DCN (2.7-fold), SCX (3.8-fold) and TNC (2.3-fold) after 3 days of culture. hWJ-MSCs also showed larger proliferation rate and marked aggregation into a tubular shaped system at Day 7 (with 100 ng/mL of hGDF-5). Simultaneous to this we explored expression of pro-inflammatory (IL-6, TNF, IL-12A, IL-1&beta;) and anti-inflammatory (IL-10, TGF-&beta;1) cytokines across for both cell types. hBM-MSCs exhibited a better balance of pro-inflammatory and anti-inflammatory cytokines upregulating IL-1&beta; (11-fold) and IL-10 (10-fold) at Day 8; hWJ-MSCs, had a slight expression of IL-12A (1.5-fold) but a greater up-regulation of IL-10 (2.5-fold). Collagen type I and tenomodulin proteins, detected by immunofluorescence, confirming the greater protein expression when 100 ng/mL were supplemented. In the same conditions, both cell types showed specific alignment and shape modification (fibroblast-like) with a Lenght/Width ratio increase at value higher than 1, suggesting their response in activating tenogenic commitment events, and they both potential use in 3D in vitro tissue engineering protocols.


2019 ◽  
Vol 8 (8) ◽  
pp. 397-404 ◽  
Author(s):  
Liza Osagie-Clouard ◽  
Anita Sanghani-Kerai ◽  
Melanie Coathup ◽  
Richard Meeson ◽  
Timothy Briggs ◽  
...  

Objectives Mesenchymal stem cells (MSCs) are of growing interest in terms of bone regeneration. Most preclinical trials utilize bone-marrow-derived mesenchymal stem cells (bMSCs), although this is not without isolation and expansion difficulties. The aim of this study was: to compare the characteristics of bMSCs and adipose-derived mesenchymal stem cells (AdMSCs) from juvenile, adult, and ovarectomized (OVX) rats; and to assess the effect of human parathyroid hormone (hPTH) 1-34 on their osteogenic potential and migration to stromal cell-derived factor-1 (SDF-1). Methods Cells were isolated from the adipose and bone marrow of juvenile, adult, and previously OVX Wistar rats, and were characterized with flow cytometry, proliferation assays, osteogenic and adipogenic differentiation, and migration to SDF-1. Experiments were repeated with and without intermittent hPTH 1-34. Results Juvenile and adult MSCs demonstrated significantly increased osteogenic and adipogenic differentiation and superior migration towards SDF-1 compared with OVX groups; this was the case for AdMSCs and bMSCs equally. Parathyroid hormone (PTH) increased parameters of osteogenic differentiation and migration to SDF-1. This was significant for all cell types, although it had the most significant effect on cells derived from OVX animals. bMSCs from all groups showed increased mineralization and migration to SDF-1 compared with AdMSCs. Conclusion Juvenile MSCs showed significantly greater migration to SDF-1 and significantly greater osteogenic and adipogenic differentiation compared with cells from osteopenic rats; this was true for bMSCs and AdMSCs. The addition of PTH increased these characteristics, with the most significant effect on cells derived from OVX animals, further illustrating possible clinical application of both PTH and MSCs in bone regenerative therapies. Cite this article:L. Osagie-Clouard, A. Sanghani-Kerai, M. Coathup, R. Meeson, T. Briggs, G. Blunn. The influence of parathyroid hormone 1-34 on the osteogenic characteristics of adipose- and bone-marrow-derived mesenchymal stem cells from juvenile and ovarectomized rats. Bone Joint Res 2019;8:397–404. DOI: 10.1302/2046-3758.88.BJR-2019-0018.R1.


Sign in / Sign up

Export Citation Format

Share Document