eISSN: 1896-9151
ISSN: 1734-1922
Archives of Medical Science
Current issue Archive Special issues Subscription
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
2/2009
vol. 5
 
Share:
Share:

In vitro osteogenesis of rat adipose-derived stem cells: comparison with bone marrow stem cells

Iraj Ragerdi Kashani
,
Arash Zaminy
,
Mohammad Barbarestani
,
Azim Hedayatpour
,
Reza Mahmoudi
,
Safoura Vardasbi
,
Ahmadreza Farzaneh Nejad
,
Mohammad ali Naraghi

Arch Med Sci 2009; 5, 2: 149-155
Online publish date: 2009/07/23
Article file
Get citation
 
 
Introduction
Various methods have been proposed for the repair of bone defects in the field of plastic and orthopaedic surgery. A previous approach to this problem focused on the development of various artificial materials that might be used instead of autogenous bone [1]. Current studies, however, rely mainly on the use of fresh autograft bone, various bone graft substitutes [2] and tissue-engineering techniques that incorporate appropriate implants seeded with cells having osteogenic potential [3]. Mesenchymal stem cells have recently received widespread attention because of their potential use in tissue-engineering applications [4]. Mesenchymal stem cells (MSCs), also known as marrow stromal cells or mesenchymal progenitor cells, are defined as self-renewable, multipotent progenitor cells with the capacity to differentiate into several distinct mesenchymal lineages [5]. To date, MSCs of various adult vertebrate species have been demonstrated to differentiate into lineage-specific cells that form the bone, cartilage, fat, tendon, and muscle tissue [6-8]. Bone marrow derived mesenchymal stem cells have been shown to be multipotential in that they differentiate in culture [8] or after implantation in vivo into osteoblasts [9]. Although bone marrow provides the most universal source of MSCs, other tissues such as periosteum [10], muscle [11], synovial membrane [12] and adipose tissue also appear to possess MSCs [13]. Adipose tissue is particularly attractive because of its easy accessibility and abundance [14-16]. Adipose tissue-derived mesen-chymal stem cells (ADSCs) obtained from lipoaspirates have been shown to have the multi-lineage potential to differentiate into adipogenic, chondrogenic, myogenic and osteogenic cells [13, 14]. Adipose-derived stem cells mineralized their extracellular matrix (ECM), and increased the expression of osteocalcin and alkaline phosphatase (ALP) [16]. However, it is not well established whether ADSCs have the same potential for osteogenesis and chondrogenesis as bone marrow-derived mesenchymal stem cells (BMSCs). Thus, the purpose of this study is to investigate whether ADSCs are equal to BMSCs with regard to their osteogenesis potential.

Material and methods
Isolation and culture of mesenchymal stem cells
About 6- to 8-week-old male Wistar rats of the albino strain were killed using diethyl ether and the bones were collected under sterile conditions, then all the bones were cut at both ends. The bone marrow from each bone was collected by flushing the bone with Dulbecco’s Modified Eagle’s Medium (Sigma) containing 1000 U/ml Penicillin G. The cells after filtering were centrifuged at 3,000 rpm for 5 min. The purified cells were finally dispersed in DMEM with 15% fetal bovine serum (Sigma) containing 100 U/ml penicillin, 100 mg/ml streptomycin [17].
Primary adipose-derived stem cells were harvested from the scrotal fat pad of the same age rats. Epididymal adipose tissue was excised, placed on a sterile glass surface, and finely minced. The minced tissue was placed in a 50 ml conical tube (Greiner, Germany) containing 0.05% tissue culture grade collagenase type 1 (Sigma) and 5% bovine serum albumin (Sigma). The tube was incubated at 37°C for 1 h, being shaken every 5 min. The tube contents, after filtering through a sterile 250 mm nylon mesh, were centrifuged at 250 γ for 5 min. The cell pellet was resuspended in adipose-derived stem cell medium: DMEM/F12 (Sigma), 10% fetal bovine serum (Gibco), and 100 U/ml penicillin, 100 mg/ml streptomycin (Sigma). Cell count was determined with a haemacytometer [18].
Cell culture and expansion
The isolated stem cells were plated in T75 tissue culture flasks, containing appropriate stem cell medium at a density of 10 × 105 cells per flask. The flasks were maintained in a tissue culture incubator at 37°C and 5% carbon dioxide. The medium was replaced every third day afterwards. Cell viability was confirmed by continued cell division and the cells were subcultured using 3 ml of trypsin/EDTA (Sigma) when the flasks reached 90% confluence.
Osteogenic differentiation
Both types of cells were used to assess melatonin’s effect on osteogenic differentiation. Cells (passage 3) were then seeded at an initial density of 10 × 105 cells per flask (25 cm2) in osteogenic medium (OS) containing 0.05 mM ascorbate, 1 mM dexamethasone and 10 mM b-glycerophosphate for 4 weeks [18].
Confirmation of osteogenic differentiation
Confirmation of osteogenesis was confirmed by means of Von Kossa and Alizarin Red S staining (highlights extracellular matrix calcification) and assessment of alkaline phosphatase activity and expression of osteocalcin gene.
Von Kossa Staining
Cells in flasks (25 cm2) were rinsed with phosphate-buffered saline and were fixed in 4% paraformaldehyde for 20 min. The cells were incubated in 5% silver nitrate in the dark, then the flasks were exposed to ultraviolet light for 1 h. Secretion of calcified extracellular matrix was observed as black nodules with von Kossa staining [1].
Alizarin Red S Staining
Cells in flasks (25 cm2) were washed with phosphate-buffered saline (PBS) and fixed in 10% (v/v) formaldehyde (Sigma-Aldrich). After 15 min ARS 2% (pH = 4.1) was added to each flask. The flasks were incubated at room temperature for 20 min. Then the flasks were washed four times with dH2O while being shaken for 5 min [19].
Quantification of mineralization
The analysis of the amount of calcium deposition in osteogenic media was modified from a previous report [19]. In brief, 2 ml 10% (v/v) acetic acid was added to each flask. After 30 min the monolayer was scraped off the plate with a cell scraper and transferred with 10% (v/v) acetic acid to a 15 ml microcentrifuge tube. After vortexing for 30 s, the slurry was overlaid with 1.25 ml mineral oil (Sigma-Aldrich), heated to exactly 85°C for 10 min, and transferred to ice for 5 min. The slurry was then centrifuged at 20,000 × γ for 15 min and 500 ml of the supernatant was removed to a new 1.5 ml microcentrifuge tube. Then, 200 ml of 10% (v/v) ammonium hydroxide was added to neutralize the acid. Aliquots (150 ml) of the supernatant were read in triplicate at 405 nm in 96-well format using opaque-walled, transparent-bottomed plates.
ALP activity
The cells were lysed by sonication for three cycles, and then protein solutions were centrifuged at 2000 × γ for 15 min at 4°C. The total protein content of each sample was determined according to Bradford [20]. ALP was performed using an ALP kit (Ziest Chem, Tehran, Iran) and following the manufacturer’s instructions. The levels of activity were neutralized with an amount of protein in cell lysate solution (units/mg protein).
RNA extraction and reverse transcription-polymerase chain reaction analysis of gene expression
After extraction of total RNA, reverse trans-criptase polymerase chain reaction assays were performed as described [21].
Flow cytometry
DNA fragmentation, as a late feature of apoptosis, was evaluated using a flow cytometer. PI staining was performed as previously described [22]. Sample acquisition was performed by a FACScan flow cytometer equipped with Cell Quest software.
Cell viability assay
The MTT (Sigma) test measures the mito-chondrial (metabolic) activity in the cell culture, which reflects the number of viable cells [23]. In brief, the cultures (5 × 104 were seeded to a 96-well plate) were washed in PBS, and 200 ml of MTT reagent were added. Following incubation for 3 h in the incubator (in 5% CO2 at 37°C) the absorption of the medium was measured in an ELISA Reader (Anthos 2020) at 440 nm.
Statistical analysis
The results are listed as the mean ± SD. The statistical difference was analyzed by one-way ANOVA followed by Dennett’s test. The p value < 0.05 was considered to be significant. All assays were performed in triplicate.

Results
Cultivation and passaging of human mesenchymal stem cell culture
No adherent cells were removed from the dish during medium changes and the subsequent passaging. Typically 80-90% of confluence was reached by day 10 in BMSC cultures and by day 5 for ADSC cultures. Cells at third passage were used for the experiments.
Rat bone marrow stem cells and adipose-derived stem cells grown in culture appeared spindle-shaped. Cells cultured in osteogenic media demonstrated a dramatic change in cell morphology from day 5 induction, with the cells changing morphology from an elongated fibroblastic appearance to polygonal, more cuboidal shape (Figure 1).
The cells of the two cell populations were cultured for 28 days in osteogenic media. After this period, samples were taken for analyses.
Quantitative estimation of ALP and mineralization
As a marker for BMSCs and ADSCs diffe-rentiation into osteoblasts, ALP levels were measured in both groups. In our study, ALP activity was measured in BMSCs and ADSCs in osteogenic medium after 14 and 28 days (Figure 2). The data showed that BMSCs underwent a much higher increase in ALP activity compared with ADSCs.
The cells were stained positively for extracellular mineralization after 2 and 4 weeks of culture in osteogenic media, as confirmed by Von Kossa and Alizarin Red S staining (Figure 3). Calcium level quantification was measured in both groups after 2 and 4 weeks after osteogenic induction in ADSCs and BMSCs (Figure 4).
The calcium measurement indicated more calcium for the BMSCs. The ADSC groups revealed a low level.
Osteocalcin gene expression
To determine gene expression of osteocalcin mRNA in both MSCs, RT-PCR was carried out with primer specific for osteocalcin. Figure 5 shows the results. The presence of the osteocalcin mRNA band indicated osteogenesis.
Apoptosis and cell viability
In individual experiments, the incidence of apoptotic cells among ADSCs was higher than BMSCs (Figure 6). The flow cytometry proves that cell growth reduction is due to a decrease of cells entering the S phase of the cell cycle. These data were demonstrated by MTT assay (Figure 7). These data indicated that viable cells among ADSCs were lower than BMSCs in the control groups.

Discussion
In the present study, we confirm that both BMSCs and ADSCs have the potential to differentiate into an osteogenic lineage. However, BMSCs have a greater osteogenesis potential as evidenced by greater matrix production when compared with ADSCs.
Over recent years, stem cells have generated great interest given their potential therapeutic use. Recent reports have provided clear evidence that multipotent adult stem cells exist in many more organs and tissues than previously expected. Mesenchymal cells capable of differentiation into a variety of specific cell phenotypes have been isolated from tissues such as bone marrow, muscle, fat, periosteum and synovial membrane both from rodents and humans [8, 12, 13, 24, 25].
Although bone marrow provides a universal source of MSCs, adipose tissue also possesses abundant and easily accessible MSCs. Recent advances in cosmetic surgery add to its advantage with a huge amount of available fatty tissue. It may have a further advantage when the morbidity associated with large volume bone marrow harvests is taken into consideration [26, 27].
Regarding comparison between BMSCs and ADSCs there are similar findings [28-30] but the overall results of the present study do not corroborate the results of the previous studies that suggested equal or comparable capacity of ADSCs for osteogenesis [13, 31, 32]. There are several possible reasons for such a difference. The reason suggested by Gun et al. [28] was that ADSC isolates may represent a fairly heterogeneous population of cell types with only a small number of progenitor cells capable of osteogenic differentiation. This would be consistent with the results of some studies which indicated differences in cell surface antigen expression between cellular preparations of adipose-derived and bone marrow-derived stromal cells [33-35]. Another explanation is that ADSCs may represent distinctly different cell populations that are at different stages of lineage-specific commitment from BMSCs [36]. Generally, studies have shown that BMSCs and ADSCs are not a homogeneous population of multilineage progenitors; rather, they are made up of a heterogeneous population of pluripotent stem cells and tripotent, bipotent, and unipotent progenitors [13, 37, 38].
Therefore, the differences between BMSCs and ADSCs observed here may not be due to the inherent difference between multipotent BMSCs and multipotent ADSCs. Rather, it could be due to the fact that BMSC cultures may be dominated by osteogenic and chondrogenic progenitors, whereas ADSCs have mainly adipogenic progenitors [38].
There are limitations of this study that preclude definite conclusions. First, we had different rats for BMSCs and ADSCs. Second, the gene expression profiles were not thoroughly investigated using a quantitative PCR technique. This will be pursued in the following study.
In conclusion, the results of this study show that ADSCs differ from BMSCs in their osteogenic potential. When equal amounts of bioactive factors are given, ADSCs have inferior capacity to differentiate into bone, suggesting the limited utility of ADSCs as a source of cells needed for tissue engineering of bone. As a further step forward, a search for the culture conditions that would induce successful osteogenesis from ADSCs is warranted.

References
1. Huang JI, Beanes SR, Zhu M, Lorenz HP, Hedrick MH, Benhaim P. Rat extramedullary adipose tissue as a source of osteochondrogenic progenitor cells. Plast Reconstr Surg 2002; 109: 1033-41.
2. Saito N, Okada T, Horiuchi H, et al. Biodegradable poly-D, L-lactic acidpolyethylene glycol blocks copolymers as a BMP delivery system for inducing bone. J Bone Joint Surg Am 2001; 83-A Suppl 1: S92-8.
3. Dong J, Uemura T, Shirasaki Y, Tateishi T. Promotion of bone formation using highly pure porous beta-TCP combined with bone marrow-derived osteoprogenitor cells. Biomaterials 2002; 23: 4493-502.
4. Hattori H, Masuoka K, Sato M, et al. Bone formation using human adipose tissue-derived stromal cells and a biodegradable scaffold. J Biomed Mater Res B Appl Biomater 2006; 76: 230-9.
5. Caplan AI. Mesenchymal stem cells. J Orthop Res 1991; 9: 641-50.
6. Alhadlaq A, Mao JJ. Tissue-engineered neogenesis of human-shaped mandibular condyle from rat mesenchymal stem cells. J Dent Res 2003; 82: 951-6.
7. Alhadlaq A, Elisseeff J, Hong L, et al. Adult stem cell deriven genesis of human-shaped articular condyle. Ann Biomed Eng 2004; 32: 911-23.
8. Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential of adult human mesenchymal stem cells. Science 1999; 284: 143-7.
9. Richard DJ, Kassem M, Hefferan TE, Sarkar G, Spelsberg TC, Riggs BL. Isolation and characterization of osteoblast precursor cells from bone marrow. J Bone Miner Res 1996; 11: 312-24.
10. Iwasaki M, Nakata K, Nakahara H, et al. Transforming growth factor-beta 1 stimulates chondrogenesis and inhibits osteogenesis in high density culture of periosteum-derived cells. Endocrinology 1993; 132: 1603-8.
11. Bosch P, Musgrave DS, Lee JY, et al. Osteoprogenitor cells within skeletal muscle. J Orthop Res 2000; 18: 933-44.
12. De Bari C, Dell'Accio F, Tylzanowski P, Luyten FP. Multipotent mesenchymal stem cells from adult human synovial membrane. Arthritis Rheum 2001; 44: 1928-42.
13. Zuk PA, Zhu M, Ashjian P, et al. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell 2002; 13: 4279-95.
14. Zuk PA, Zhu M, Mizuno H, et al. Multilineage cells from human adipose tissue; implications for cell-based therapies. Tissue Eng 2001; 7: 211-28.
15. Nathan S, Das De S, Thambyah A, Fen C, Goh J, Lee EH. Cell-based therapy in the repair of osteochondral defects: a novel use for adipose tissue. Tissue Eng 2003; 9: 733-44.
16. Halvorsen YD, Franklin D, Bond AL, et al. Extracellular matrix mineralization and osteoblast gene expression by human adipose tissue-derived stromal cells. Tissue Eng 2001; 7: 729-41.
17. George J, Kuboki Y, Miyata T. Differentiation of mesenchymal stem cells into osteoblasts on honeycomb collagen scaffolds. Biotechnol Bioeng 2006; 95: 404-11.
18. Lee JA, Parrett BM, Conejero JA, et al. Biological alchemy: engineering bone and fat from fat-derived stem cells. Ann last Surg 2003; 50: 610-7.
19. Gregory CA, Gunn WG, Peister A, Prockop DJ. An Alizarin red-based assay of mineralization by adherent cells in culture: comparison with cetylpyridinium chloride extraction. Anal Biochem 2004; 329: 77-84.
20. Bradford MM. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 1976; 72: 248-54.
21. Roth JA, Kim BG, Lin WL, Cho MI. Melatonin promotes osteoblast differentiation and bone formation. J Biol Chem 1999; 274: 22041-7.
22. Luchetti F, Canonico B, Curci R, et al. Melatonin prevents apoptosis induced by UV-B treatment in U937 cell line. J Pineal Res 2006; 40: 158-67.
23. Merklein F, Hendrich C, Nöth U, et al. Standardized tests of bone implant surfaces with an osteoblast cell culture system. I. Orthopedic standard materials [German]. Biomed Tech (Berl) 1998; 43: 354-9.
24. Yoo JU, Johnstone B. The role of osteochondral progenitor cells in fracture repair. Clin Orthop Relat Res 1998; 355: S73-81.
25. Asakura A, Seale P, Girgis-Gabardo A, Rudnicki MA. Myogenic specification of side population cells in skeletal muscle. J Cell Biol 2002; 159: 123-34.
26. Auquier P, Macquart-Moulin G, Moatti JP, et al. Comparison of anxiety, pain and discomfort in two procedures of hematopoietic stem cell collection: leukacytapheresis and bone marrow harvest. Bone Marrow Transplant 1995; 16: 541-7.
27. Nishimori M, Yamada Y, Hoshi K, et al. Health-related quality of life of unrelated bone marrow donors in Japan. Blood 2002; 99: 1995-2001.
28. Im GI, Shin YW, Lee KB. Do adipose tissue-derived mesenchymal stem cells have the same osteogenic and chondrogenic potential as bone marrow-derived cells? Osteoarthritis Cartilage 2005; 13: 845-53.
29. Yoshimura H, Muneta T, Nimura A, Yokoyama A, Koga H, Sekiya I. Comparison of rat mesenchymal stem cells derived from bone marrow, synovium, periosteum, adipose tissue, and muscle. Cell Tissue Res 2007; 327: 449-62.
30. Sakaguchi Y, Sekiya I, Yagishita K, Muneta T. Comparison of human stem cells derived from various mesenchymal tissues: superiority of synovium as a cell source. Arthritis Rheum 2005; 52: 2521-9.
31. De Ugarte DA, Alfonso Z, Zuk PA, et al. Differential expression of stem cell mobilization-associated molecules on multi-lineage cells from adipose tissue and bone marrow. Immunol Lett 2003; 89: 267-70.
32. De Ugarte DA, Morizono K, Elbarbary A, et al. Comparison of multi-lineage cells from human adipose tissue and bone marrow. Cells Tissues Organs 2003; 174: 101-9.
33. Wickham MQ, Erickson GR, Gimble JM, Vail TP, Guilak F. Multipotent stromal cells derived from the infrapatellar fat pad of the knee. Clin Orthop Relat Res 2003; 412: 196-212.
34. Winter A, Breit S, Parsch D, et al. Cartilage-like gene expression in differentiatedhuman stem cell spheroids: a comparison of bone marrow-derived and adipose tissue-derived stromal cells. Arthritis Rheum 2003; 48: 418-29.
35. Gronthos S, Franklin DM, Leddy HA, Robey PG, Storms RW, Gimble JM. Surface protein characterization of human adipose tissue-derived stromal cells. J Cell Physiol 2001; 189: 54-63.
36. De Ugarte DA, Morizono K, Elbarbary A, et al. Comparison of multi-lineage cells from human adipose tissue and bone marrow. Cells Tissues Organs 2003; 174: 101-9.
37. Muraglia A, Cancedda R, Quarto R. Clonal mesenchymal progenitors from human bone marrow differentiatiate in vitro according to a hierarchical model. J Cell Sci 2000; 113: 1161-6.
38. Liu TM, Martina M, Hutmacher DW, Hui JH, Lee EH, Lim B. Identification of common pathways mediating differentiation of bone marrow- and adipose tissue- derived human Mesenchymal stem cells into three Mesenchymal lineages. Stem Cells 2007; 25: 750-60.
Copyright: © 2009 Termedia & Banach. This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.