Open access

Adipose-Derived Stem Cells (ASCs) for Tissue Engineering

Written By

Mathias Tremp, Souzan Salemi, Rita Gobet, Tullio Sulser and Daniel Eberli

Submitted: 17 November 2010 Published: 29 August 2011

DOI: 10.5772/22120

From the Edited Volume

Regenerative Medicine and Tissue Engineering - Cells and Biomaterials

Edited by Daniel Eberli

Chapter metrics overview

5,430 Chapter Downloads

View Full Metrics

1. Introduction

A current challenge in the field of tissue engineering is soft tissue replacement. Techniques for soft tissue reconstruction include use of autologous fat transplantation, alloplastic implants and autologous tissue flaps. However, these approaches have disadvantages, including donor-site morbidity, implant migration, foreign body reaction and immune system rejection. The use of autologous stem cells expanded in vitro and combined with novel biomaterials for organ reconstruction offers a potential solution for replacement of tissue or whole organs. Stem cells, first identified in embryonic tissue and later in numerous adult tissues, possess the unique capacity to differentiate into wide range of tissue types. However, although embryonic cells are the most flexible of stem cell lines, they raise the problem of ethical issues. For tissue engineering, candidates of stem cells include embryonic stem cells (Ahn, et al.), induced pluripotent stem cells (iPS) (Crisan, et al.;2008, Parker, et al.;2007) and adult stem cells. The ability of adult stem cells to divide or self-renewal make them attractive source of stem cells for use in tissue engineering. A significant amount of current interest has focused on the possibility that adult human stem cells are the therapeutic alternative to embryonic stem cells because of their plasticity (Aoki, et al.;2010).

The presence of self-renewing cells within the bone marrow of mice was reported in 1963 which was later known as hematopoietic stem cells (HSCs) (Becker, et al.;1963, Zhang, et al.;1999). Several years later, HSCs were identified in umbilical cord blood by other investigators (Aust, et al.; 2004, Dellavalle, et al.;2007). Furthermore, several other adult stem cell types such as neural stem cells (Dellavalle, et al.; 2007, Guilak, et al.;2010), were isolated and identified. Moreover, a population of plastic adherent cells were isolated from collagenase digests of adipose tissue.

Adipose tissue derived stem cells were termed as: adipose derived stem/stromal cells (ASCs), adipose-derived adult stem (ADAS) cells, adipose-derived adult stromal cells, adipose-derived stromal cells (ADSCs), adipose stromal cells (ASCs), adipose mesenchymal stem cells (AdMSCs), lipoblast, pericyte, preadipocyte, and processed lipoaspirate (PLA) cells. To prevent the confusion in the literature, the International Fat Applied Technology Society reached a consensus to adopt the term “adipose-derived stem cells” (ASCs) to identify the isolated, plastic-adherent, multipotent cell population (Gimble, et al.;2007). Adipose tissue derives from the mesodermal layer of the embryo and develops both pre- and postnatally (Gonda, et al.;2008, Kakudo, et al.;2008). Researchers studying the development of adipose tissue have long worked with a fibroblastic cell line, known as preadipocytes, isolated from adipose tissue (Cowherd, et al.;1999, Gregoire, et al.;1998, Kirkland and Hollenberg;1998, Nam and Lobie;2000, Sorisky;1999).

The reason why adipose tissue would contain a stem cell population is not still clear. There is some discussion whether the cells are subpopulation of fibroblasts reside within the fat tissue or are perhaps mesenchymal or peripheral blood stem cells passing through the fat tissue (Crisan, et al.; 2008, Dellavalle, et al.; 2007).

The ASCs represent a readily available source for isolation of potentially useful stem cells (Sterodimas, et al.; 2010). In culture, they have shown to have an impressive developmental plasticity, including the ability to undergo multilineage differentiation and self-renewal (Liu, et al.; 2009). When ASCs are compared with BM-MSCs, further similarities have been demonstrated in regards to their growth kinetics, cell senescence, gene transduction efficiency (De Ugarte, et al.; 2003), as well as CD surface marker expression (Gronthos, et al.; 2001, Katz, et al.; 2005, Zuk, et al.; 2002) and gene transcription (Katz, et al.; 2005). Compared to bone marrow MSCs, ASCs have potential advantages for tissue engineering application, because of the tissue accessibility, multipotency and ease of isolation without painful procedures or donor site injury.

In this chapter we will discuss the potential use of adipose-derived stem cells in the field of tissue engineering.

Advertisement

2. Isolation and expansion of ASCs

The initial methods to isolate cells from adipose tissue were pioneered by Rodbell and colleagues in the 1960s (Rodbell; 1966, Rodbell; 1966, Rodbell and Jones; 1966) using rat fat tissue. These methods were further adapted for human tissues by several other groups (Deslex, et al.; 1987, Engfeldt, et al.; 1980, Ho, et al.; 2010). The current methods for isolating ASCs rely on a collagenase digestion followed by centrifugal separation to isolate the stromal/vascular cells from primary adipocytes. The pellet is resuspended with a basal medium containing 10% foetal bovine serum (Estes, et al.; 2008). The cell suspension is filtered through 100 µm cell strainer and the cells are plated and incubated at 37ºC in the presence of 5% CO2. The medium is changed every second day until the cells reach 80-90% confluence. A large number of ASCs can be harvested in this manner, with yields of approximately 250,000 cells per gram of tissue (Aust, et al.; 2004, Guilak, et al.; 2010). In order to remove the use of animal products in human ASC cultures, a very low human serum expansion medium and a completely serum-free medium have been recently reported (Parker, et al.; 2007). Furthermore it was reported that use of platelet- rich plasma can enhance the proliferation of human ASCs. These results can support the clinical application of platelet-rich plasma for cell based, soft-tissue engineering and wound healing (Kakudo, et al.; 2008).

ASCs should be harvested at 80% confluence for freezing. Cryopreservation medium consists of 80% fetal bovine serum, 10% dimethylsulfoxide (DMSO) and 10% DMEM/Ham’s F-12. The cells should be stored in a final concentration of 1–2 million viable cells per milliliter of cryopreservation medium. Aliqouted vials are first frozen in an alcohol freezing container and store and are stored at -80ºC overnight. On the next day, the frozen vials can be transferred to a liquid nitrogen container for long-term storage. Successful storage of ASCs more than 6 months has been shown. This ensures the availability of autologous banked ASCs for clinical applications in the future (De Rosa, et al.;2009, Gonda, et al.;2008).

2.1. Characterization of ASCs

In order to characterize the undifferentiated animal or human ASC cells cultured in vitro flow cytometric and immunohistochemical methods are widely used (Gronthos, et al.; 2001, Zuk, et al.; 2001). The cell surface phenotype of ASC is quite similar to MSCs (mesenchymal stem cells). Both ASC and MSC cells express CD29, CD44, CD71, CD90, and CD105 (Zuk, et al.; 2002). In contrast, no expression of the hematopoietic lineage markers CD31, CD34 and CD45 was observed in either of the cultures. In addition the ASC cells express the neutral endopeptidase (CD10 or common acute lymphocytic leukemia antigen CALLA), aminopeptidase (CD13), and ecto nucleotidase (CD73). Furthermore, ASC cells produce Type I and Type III collagens, osteopontin, ostenectin, Thy-1 (CD90), and MUC-18 (CD146) (Gimble and Guilak; 2003).

Different investigator have reported different pattern of expression. For example, while Gronthos et al. (Gronthos, et al.;2001) detected CD34 and VCAM (CD106) on ASC cells, Zuk et al. (Zuk, et al.;2002) did not. Likewise, while Zuk et al. (Zuk, et al.;2002) detected Stro-1, Gronthos et al. did not. These discrepancies could be due to the differences in cell isolation methods, how long the cells were cultured prior to analysis and sensitivity differences between immunohistochemical and flow cytometric detection methods (Zuk, et al.;2002, Zuk, et al.;2001).

Advertisement

3. ASCs applications

Adipose tissue has proven to serve as an abundant source of adult stem cells with multipotent properties suitable for tissue engineering and regenerative medical applications. ASCs can be differentiated into variety of cell types. Differentiation is commonly induced by insulin, dexamethasone, cyclic AMP agonist, β-glycerophosphate, heparin, ascorbate and different cytokines depending on the lineage type.

ASCs like BM-MSCs, differentiate in vitro towards soft tissue such adipocytes, smooth muscle and cardiac myocytes when treated with established lineage-specific factors. In addition they can differentiate toward musculoskeletal tissues such as osteocytes, myocytes and chondrocytes. Furthermore neurogenic differentiation of these cells is reported by several investigators.

3.1. ASCs differentiation

3.1.1. Adipogenesis

ASCs in response to inductive compounds including glucocorticoid receptor ligands (dexamethasone), insulin, cyclic AMP agonist (forskolin) and peroxisome proliferator-activated receptor gamma (PPARγ) undergo adipogenic differentiation (Farmer; 2006, Hauner, et al.; 1989, Lazar; 2005, Zuk, et al.; 2001). During the differentiation process ASCs reduce their proliferation rate and undergo morphological changes. ASCs are induced in the adipocyte differentiation medium containing biotin, d-pantothenate, dexamethasone, methylisobutylxanthine, insulin and equivalent PPARγ agonist. After induction for 2 weeks in adipogenic medium the human ASC contain vacuoles filled with neutral lipid cells which can be further stained for intracellular lipid droplets accumulation using an Oil Red O stain (Preece;1972).

In addition, these cells secrete increased amounts of the adipocyte protein leptin, and transcribe adipogenic mRNAs such as the fatty acid binding protein, aP2 and lipoprotein lipase (Halvorsen, et al.; 2001, Hauner, et al.; 1989, Sen, et al.; 2001). Some of these parameters such as leptin, aP2 mRNA levels were quantified and found to be increased by several hundred-fold during the differentiation process (Halvorsen, et al.; 2001, Sen, et al.; 2001). It is reported that ASCs harvested from female mice differentiate more efficiently into adipocytes than those from male mice (Ogawa, et al.; 2004).

One of the most important uses of ASCs is for the replacement of adipose tissue itself. Large soft tissue defects are a common problem following trauma, burns and oncological resections. Several studies demonstrated the in vitro differentiation of ASCs along adipogenic lineages, including the accumulation of intracellular lipid droplets, and the expression of characteristic proteins and enzymes (Ogawa, et al.; 2004, Tchkonia, et al.; 2002, Zuk, et al.; 2002). ASCs were used to seed artificial scaffolds and were further implanted subcutaneously in mice and rats (von Heimburg, et al.; 2001, von Heimburg, et al.; 2001). The cell-seeded grafts showed significant neovascularisation of the implant, as well as penetration of the preadipocytes or ASCs into the scaffolding, and their differentiation into mature lipid-laden adipocytes.

3.1.2. Smooth muscle

ASCs can be differentiated to smooth muscle cells (SMCs) and might offer a cell source for hollow organ engineering. For myogenic differentiation ASCs at passages 3 through 5 are cultured in smooth muscle inductive medium consisting of MCDB131 supplemented with 1% FBS and 100u/ml of heparin for up to 6 weeks at 37ºC with 5% CO2. The media is changed every 3 days and cell splitting is not required (Jack, et al.; 2009).

The cellular changes after differentiation can be investigated by real- time PCR at mRNA level. As reported the expression of muscle actin (SMA), calponin and myosin heavy chain showed an increase after growth in differentiation medium (Jack, et al.; 2009). The same was observed at protein levels, induction media induced differentiation of the ASCs into a smooth muscle phenotype in which the expression of smooth muscle specific proteins SMA, caldesmon, and myosin heavy chain (MHC) was increased (Jack, et al.; 2009).

Differentiation is a complex process and has a dramatical effect on cell size, shape, membrane potential, metabolic activity and responsiveness to external signals. One of the main characteristic of SMCs is their contractility which plays important roles in angiogenesis, blood vessel maintenance, and mechanical regulation of hollow organs such as bladder.

Differentiated SMCs can show two specific phenotypes, which vary from synthetic and proliferative to contractile and quiescent (Beamish, et al.; 2010). SMCs exhibit a contractile phenotype characterized by high expression of specific contractile proteins including SMA, calponin, SM22, smoothelin, h-caldesmon and smooth muscle myosin heavy chain (SM-MHC) (Owens, et al.; 2004, Shanahan, et al.; 1993).

Since differentiated human ASCs express smooth muscle specific proteins they may prove to be of value in the repair of smooth muscle defects in the gastrointestinal and urinary tracts. Juan et al. reported that the ASCs from different sites show different myogenic differentiation abilities in vitro. ASCs from the adipose tissues of the nape of the neck and vicinity of epididymis can be used as ideal seed cells for tissue engineering of lower urinary tract (Yuan, et al.; 2010). Similar study was performed by other group using human subcutaneous and omental adipose tissues. They could show that subcutaneous adipose tissue has higher differentiation capacity than omental adipose tissue which can be a suitable cell source for use in regenerative medicine (Toyoda, et al.; 2009).

3.1.3. Osteogenesis. Bone defect repair

In the past decade, several groups isolated cells from the adipose tissue of humans and other species capable of differentiating into osteoblasts in vitro (Dragoo, et al.; 2003, Mizuno, et al.; 2002, Zuk, et al.; 2001). ASC cells differentiate into osteoblast-like cells in the presence of ascorbate, b-glycerophosphate, dexamethasone and vitamin D3.

For osteogenic differentiation confluent ASCs cells are incubated for 3 weeks in DMEM containing 10% FBS, 100 nM dexamethasone, 10 mM β-glycerophosphate and 50 μM L-ascorbic acid-2-phosphate. After fixation cells are incubated at 37ºC for 1 hour with 0.16% naphthol AS-TR phosphate and 0.8% Fast Blue BB dissolved in 0.1 M tris buffer (pH 9.0). For osteogenic differentiation cells were also incubated in 1% alizarin red S for 3 minutes to detect calcium deposition (Sakuma, et al.; 2009).

Over a 2- 4 week period in vitro, both human and rat ASC cells deposit calcium phosphate mineral within their extracellular matrix, and express osteogenic genes. Under osteogenic conditions ASCs are observed to express genes and proteins associated with osteoblasts phenotypes such as osteopontin, osteonectin, osteocalcin collagen type I, BMP-2 and BMP-4 (Halvorsen, et al.; 2001, Zuk, et al.; 2001). In addition ASCs are able to form mineralized matrix in vitro in both long term 2-D or 3-D osteogenic cultures.

In vivo, ASC cells embedded in porous cubes of hydroxyapatite/tricalcium phosphate form bone were used as implants in immunodeficient mice (Hicok, et al.; 2004). New osteoid, derived from the human ASC cells, is present within a 6-week incubation period (Hicok, et al.; 2004). This finding indicates that ASCs cells will have therapeutic applications in bone repair. The first case of autologous ASC use for osseous repair has been reported in the treatment of a calvarial defect in a7-year-old girl (Lendeckel, et al.; 2004). Using different type of scaffolds, human ASC can form bone in immunodefficient mice (Hicok, et al.; 2004, Lee, et al.; 2003).

3.1.4. Myogenesis: skeletal muscle repair

There is several line of evidence that ASC cells can differentiate along each of the myocyte lineage pathways when cultured in myogenic induction medium containing 0.1 mM dexamethasone, 50 mM hydrocortisone, 10% FBS and 5% horse serum. ASCs express MyoD and myogenin, transcription factors regulating skeletal muscle differentiation (Pittenger, et al.; 1999, Zuk, et al.; 2001).

Skeletal myogenesis is characterized by a period of myoblast proliferation, followed by the expression of muscle-specific proteins and fusion to form multinucleated myotubules. Early myogenic differentiation is characterized by the expression of several myogenic regulatory factors including myogenic determination factor MyoD1 (Weintraub, et al.; 1991). Terminally differentiated myoblasts can be characterized by the expression of myosin and the presence of multiple nuclei (Silberstein, et al.; 1986).

In the first in vivo report, F Bacou et al injected ASCs into the anterior tibialis muscle of rabbits following cardiotoxin-induced injury. Consistent with prior work using satellite cells treated muscles were found to be heavier, have an increased fibre area cross-section and exert greater maximal force (Boubaker el Andalousi, et al.; 2002).

3.1.5. Chondrogenesis

ASCs display chondrogenic characteristics following induction with ascorbate, dexamethasone and transforming growth factor-β (Awad, et al.; 2003, Huang, et al.; 2004, Zuk, et al.; 2001). Under inductive conditions ASCs express aggrecan, chondroitin sulphate, collagen type II and IV and proteoglycans associated with chondrogenic phenotype.(Awad, et al.; 2004, Erickson, et al.; 2002, Wickham, et al.; 2003, Zuk, et al.; 2001)

For chondrogenic differentiation, ASCs cells are grown to confluency in 30-mm dishes and incubated for 3 weeks in DMEM containing 1% FBS, 50 mM L-ascorbic acid-2-phosphate, 40 mg/ml proline, 100 mg/ml pyruvate, 10 ng/ml transforming growth factor (Harriman, et al.)-b3, and 1x ITS. Induction medium is replaced every 3 days. At the indicated time points, differentiated cells are fixed for 1h with 4% paraformaldehyde and rinsed with PBS. Accumulation of chondrocyte matrix is detected with alcian blue staining (pH 2.5, Wako) (Matsumoto, et al.; 2008).

3.1.6. Neuronal differentiation

There is preliminary evidence suggesting that human ASCs can display neuronal and/or oligodendrocytic markers. ADSC at passages 2–5 are seeded in six-well plates at 40%–60%

confluence. After three washes with PBS, the cells are induced with NIM (DMEM supplemented with 500 mM IBMX, 200 mM INDO, and 5 mg/ml insulin) for 1 hr. The cells are then examined for the expression of neuronal markers S100, NF70, and nestin followed by hematoxylin-eosin (HE) staining (Ning, et al.; 2006).

The in vivo test for the therapeutic potential of ASCs looked at their effects when injected intraventricularly in rats. ASCs survived with increased engraftment at the site of injury compared with controls. Neural lineage markers microtubule-associated protein-2 and glial fibrillary acidic protein were expressed in some engrafted cells. Behavioural tests of the motor and sensory systems showed clear improvements in those treated with ASCs after infarct (Kang, et al.; 2003). It is not clear whether transplanted cells replaced the lost neurons or provided a support role for existing stem cells and injured neurons. Furthermore, in a co-culture model, Kang et al. studied the interactions between neural stem cells (NSCs) and ASCs. In comparison to laminin-coated dishes, ASC feeder layers showed ability to support the differentiation and survival of NSCs over 14 days in culture.

Advertisement

4. Biomaterials in tissue engineering with ASCs

Currently, autologous and allogenic adipose tissues represent a ubiquitous source of material for fat reconstructive therapies. However, these approaches are limited, and often accompanied by a 40–60% reduction in graft volume following transplantation. A number of factors including a stable scaffold support structure and vascularisation is necessary to support de novo adipogenesis and long-term maintenance of adipose tissue formation within adipose tissue engineered constructs (Patrick, et al.; 2002). Recently, cell-based approaches utilizing adipogenic progenitor cells in combination with biomaterial carriers for fat tissue engineering have been developed and were reported to promote both short-term in vivo adipogenesis and to repair defect sites (Borges, et al.; 2003, Patrick, et al.; 1999). To date, however the efficacy of exogenously delivered stem cell populations to support the generation of long-term volume stable adipose tissue in vivo is restricted by suboptimal properties of their biomaterial carriers including insufficient biocompatibility and rapid scaffold degradation rates (Patrick, et al.; 2002).

For functional tissue replacement such as bone, Silk-based biomaterials have previously been demonstrated to offer exceptional benefits over conventional synthetic (e.g. poly-glycolic and lactic acid copolymers) and natural (e.g. collagen type I) biomaterials (Meinel, et al.; 2004). The slow degradation and mechanical integrity of silk scaffolds in comparison with other conventional biomaterials such as collagen and PLA, above all for long-term in vivo studies, suggest that silk fibroin-based scaffolds would be an optimal biomaterial for long-term adipose tissue growth and function (Mauney, et al.; 2007). Also, previous studies have demonstrated the ability of adipocytes to secrete various paracrine factors which can positively influence both the migration and differentiation of preadipocytes (Shillabeer, et al.; 1989). Mauney et al. studied biomaterials derived from silk fibroin prepared by aqueous (AB) and organic (HFIP) solvent-based processes, along with collagen (COL) and poly-lactic acid (PLA)-based scaffolds in vitro and in vivo for their utility in adipose tissue engineering strategies (Mauney, et al.; 2007).

For in vitro studies, they used adipose-derived mesenchymal stem cells (hASCs) and seeded them on the various biomaterials and cultured them for 21 days in the presence of adipogenic stimulants (AD). In their study, hASCs (and hMSCs) cultured on all biomaterials in the presence of AD showed significant upregulation of adipogenic mRNA transcript levels (e.g. GLUT4) to similar extents when compared to noninduced controls (Mauney, et al.; 2007). Also, oil-red O analysis of hASC-seeded scaffold displayed substantial amounts of lipid accumulating adipocytes following cultivation with AD. Following a 4-week implantation period in a rat muscle pouch defect model, both AB and HFIP scaffolds supported in vivo adipogenesis either alone or seeded with hASCs (Mauney, et al.; 2007). On the other hand, COL and PLA scaffolds underwent rapid scaffold degradation and were irretrievable following the implantation period. The authors concluded that macroporous 3D AB and HFIP silk fibroin scaffolds offer an important platform for cell-based adipose tissue engineering applications, and in particular, provide longer-term structural integrity to promote the maintenance of soft tissue in vivo (Mauney, et al.; 2007).

Tissue-specific scaffolds and signalling systems are essential to differentiate stem cells into the required cells and use them effectively to construct three-dimensional (3D) tissues (Sterodimas, et al.; 2010). It has also been proved that adipose tissues engineered with ASCs and type I collagen scaffolds can serve in vivo for the replacement of damaged tissue (Lu, et al.; 2006). This has been confirmed by Zhang et al., where collagen I scaffold exhibited excellent cellular compatibility and can be used as a vehicle for adipose tissue engineering (Zhang, et al.; 2007).

Porous collagenous microbeads can be useful as injectable cell delivery vehicles for adipose-derived stem cells, allowing ex vivo proliferation and differentiation on particles that are small enough to be injected into a defect and molded into the desired shape without migration of the cells. The cell-seeded microbeads can be injected through a needle into the wound site, and agglomeration of the microbeads can retain the cells and microbeads in the site (Rubin, et al.; 2007). Furthermore, the use of natural material hold promises in tissues engineering.

Placental decellular matrix (PDM) holds potential as a scaffold for adipose tissue engineering applications. The placenta is a rich source of human extracellular matrix (ECM) components that can be harvested without harm to the donor. Constructs derived from the ECM may mimic the native environment of the body, promoting normal cellular organization and behavior. Natural materials also have advantages in terms of ease of processing, biodegradability and biocompatibility (Schmidt and Baier; 2000). Cell-adhesive placental decellular matrix scaffolds facilitate proliferation and viability, while differentiation is augmented when the cells are encapsulated in non-adhesive cross-linked hyaluronan (XLHA) scaffolds (Flynn, et al.; 2008). Incorporation of XLHA into the PDM scaffolds may improve the construct bulking properties and may influence cellular infiltration, differentiation and wound healing (Shu XZ; 2004).

Other candidate for suitable scaffolds is non-woven polyglycolic acid (PGA) and hyaluronic acid gel. In a recent study it has been shown that more adipose-tissue-like construct is regenerated when using type I collagen sponge than when the non-woven polyglycolic acid or hyaluronic acid gel are used (Itoi, et al.; 2010). In addition, significant evidence has been shown that ASCs and PLGA spheres can be used in a clinical setting to generate adipose tissue as a noninvasive soft tissue filler (Choi, et al.; 2006).

Altman et al. could show that human adipose-derived stem cells seeded on a silk fibroin-chitosan scaffold enhance wound healing and show differentiation into fibrovascular, endothelial, and epithelial components of restored tissue (Altman, et al.; 2009). In addition, it has been shown that transfection of human ASCs with liposome- enveloped xenogenic protein from a neonatal rat tissue preparation can induce differentiation of stem cells along the directed lineage (Gaustad, et al.; 2004). These observations support the hypothesis that the inductive biochemical and structural cues of the microenvironment are conserved across species and that a silk fibroin-chitosan delivery vehicle can provide a beneficial niche in supporting migration, proliferation, and differentiation of the applied cells (Altman, et al.; 2009).

Another complex biomolecule which has sparked great interest for tissue engineering is Hyaluronic acid which has been stated to support the growth and development of progenitor cells (Brun, et al.; 1999, Solchaga, et al.; 1999). The material has a progressive rate of biodegradation, lacks cytotoxicity and does not induce a systemic immune response or chronic inflammation in a human in vivo model (Stillaert, et al.; 2008). Hyaluronic acid-based (HA) scaffolds were demonstrated to be suitable materials for soft-tissue regeneration; they maintain volume when seeded with preadipocytes. Hemmrich et al. evaluated, in vitro and in vivo, human preadipocytes seeded onto plain hyaluronan benzyl ester (HYAFF®11) or HYAFF®11 coated with the extracellular matrix glycosaminoglycan hyaluronic acid and they found extensive formation of new vessels throughout the construct but with only minor adipose tissue (Hemmrich, et al.; 2005).

Long-standing, 3D predefined-shape adipose tissue from hAD-MSCs of human adipose tissue remains a challenge. Lin et al. cultured scaffolds (Gelatin sponges, monofilament polypropylene and polyglycolic acid meshes) with hAD-MSCs in adipogenic medium for 2 weeks before implantation, and implanted scaffolds were harvested after 2, 4, and 6 months in vivo. All of the successfully harvested scaffolds were filled with newly formed adipose tissue and had retained their predefined shape and dimensions (Lin, et al.; 2008). It has been shown that hAD-MSCs are not successful soft tissue filler if used alone (Lin, et al.; 2008, Moseley, et al.; 2006).

There are numerous adipose tissue engineering culture strategies in which the core tissue engineering principles comprising appropriate cells, scaffold, and microenvironment are optimized (Patrick; 2001). Specifically, there are static versus dynamic culture, co-cultivation, and addition of growth factors, vascularization, and long-term sustainability of engineered constructs. The advantage of dynamic culture includes increased nutrient and oxygen delivery to cells within a 3D construct (Frye and Patrick; 2006). Improved bioreactor designs to address direct perfusion conditions are necessary to advance dynamic culture techniques (Choi, et al.; 2010).

It has been shown that hASCs express pericyte lineage markers in vivo and in vitro, exhibit increased migration in response to PDGF-BB in vitro, exhibit perivascular morphology when injected in vivo, and contribute to increases in microvascular density during angiogenesis by migrating toward vessels (Amos, et al.; 2008). In cell-assisted lipotransfer (CAL), autologous ASCs are used in combination with lipoinjection (e.g. Parry-Romberg syndrome). A stromal vascular fraction (SVF) containing ASCs is freshly isolated from half of the aspirated fat and recombined with the other half. The preliminary results suggest that CAL is effective and safe for soft-tissue augmentation and superior to conventional lipoinjection and microvasculature can be detected more prominently in CAL fat (Yoshimura, et al.; 2008). Recently, adipose stem cells have proved to selectively induce neovascularisation and increase the viability of random-pattern skin flaps. This mechanism might be both due to the direct differentiation of ASCs into endothelial cells and the indirect effect of angiogenic growh factors released from ASCs (Lu, et al.; 2008).

Advertisement

5. Conclusion

ASCs provide an abundant and readily accessible source of multipotent stem cells. The use of autologous stem cells expanded in vitro and combined with novel selected biomaterials for organ reconstruction offers a potential solution for replacement of tissue or whole organs. ASC does have one important advantage over the other sources of stem cells namely easy availability. There is no human tissue as expendable as adipose tissue, making it relatively easy to isolate adequate numbers of ASCs for possible human therapies. Human ASCs can be ideal cell source for tissue engineering. They are available in large quantities of cells per individual, multipotent, are transplantable in an autologous setting.

However, further studies are needed before ASCs can be used clinically. In particular, investigators need to demonstrate the safety and efficacy of ASCs cells in animal models, either alone or in combination with novel biomaterial scaffolds.

Advertisement

Acknowledgments

Both the authors Dr. Mathias Tremp and Dr. Souzan Salemi contributed equivalently in writing this chapter.

References

  1. 1. AhnH. H.LeeJ. H.KimK. S.LeeJ. Y.KimM. S.KhangG.LeeI. W.LeeH. B.2008Polyethyleneimine-mediated gene delivery into human adipose derived stem cells. Biomaterials 2915241524220142-9612
  2. 2. AltmanA. M.YanY.MatthiasN.BaiX.RiosC.MathurA. B.SongY. H.AltE. U.2009IFATS collection: Human adipose-derived stem cells seeded on a silk fibroin-chitosan scaffold enhance wound repair in a murine soft tissue injury model. Stem Cells 2712502581549-4918
  3. 3. AmosP. J.ShangH.BaileyA. M.TaylorA.KatzA. J.PeirceS. M.2008IFATS collection: The role of human adipose-derived stromal cells in inflammatory microvascular remodeling and evidence of a perivascular phenotype. Stem Cells 2610268226901549-4918
  4. 4. AokiT.OhnishiH.OdaY.TadokoroM.SasaoM.KatoH.HattoriK.OhgushiH.2010Generation of induced pluripotent stem cells from human adipose-derived stem cells without c-MYC. Tissue Eng Part A 167219722060193-7335X
  5. 5. AustL.DevlinB.FosterS. J.HalvorsenY. D.HicokK.duLaney. T.SenA.WillingmyreG. D.GimbleJ. M.2004Yield of human adipose-derived adult stem cells from liposuction aspirates. Cytotherapy 617141465-3249
  6. 6. AwadH. A.HalvorsenY. D.GimbleJ. M.GuilakF.2003Effects of transforming growth factor beta1 and dexamethasone on the growth and chondrogenic differentiation of adipose-derived stromal cells. Tissue Eng 96130113121076-3279
  7. 7. AwadH. A.WickhamM. Q.LeddyH. A.GimbleJ. M.GuilakF.2004Chondrogenic differentiation of adipose-derived adult stem cells in agarose, alginate, and gelatin scaffolds. Biomaterials 2516321132220142-9612
  8. 8. BeamishJ. A.HeP.Kottke-MarchantK.MarchantR. E.2010Molecular regulation of contractile smooth muscle cell phenotype: implications for vascular tissue engineering. Tissue Eng Part B Rev 1654674911937-3376
  9. 9. BeckerA. J.Mc C. E.TillJ. E.1963Cytological demonstration of the clonal nature of spleen colonies derived from transplanted mouse marrow cells. Nature 1974524540028-0836
  10. 10. BorgesJ.MuellerM. C.PadronN. T.TegtmeierF.LangE. M.StarkG. B.2003Engineered adipose tissue supplied by functional microvessels. Tissue Eng 96126312701076-3279
  11. 11. Boubaker elAndalousi. R.DaussinP. A.MicallefJ. P.RouxC.NouguesJ.ChammasM.ReyneY.BacouF.2002Changes in mass and performance in rabbit muscles after muscle damage with or without transplantation of primary satellite cells. Cell Transplant 1121691800963-6897
  12. 12. BrunP.CortivoR.ZavanB.VecchiatoN.AbatangeloG.1999In vitro reconstructed tissues on hyaluronan-based temporary scaffolding. J Mater Sci Mater Med 10106836880957-4530
  13. 13. ChoiJ. H.GimbleJ. M.LeeK.MarraK. G.RubinJ. P.YooJ. J.Vunjak-NovakovicG.KaplanD. L.2010Adipose tissue engineering for soft tissue regeneration. Tissue Eng Part B Rev 1644134261937-3376
  14. 14. ChoiY. S.ChaS. M.LeeY. Y.KwonS. W.ParkC. J.KimM.2006Adipogenic differentiation of adipose tissue derived adult stem cells in nude mouse. Biochem Biophys Res Commun 34526316370000-6291X
  15. 15. CowherdR. M.LyleR. E.Mc GeheeR. E.Jr 1999Molecular regulation of adipocyte differentiation. Semin Cell Dev Biol 1013101084-9521
  16. 16. CrisanM.YapS.CasteillaL.ChenC. W.CorselliM.ParkT. S.AndrioloG.SunB.ZhengB.ZhangL.NorotteC.TengP. N.TraasJ.SchugarR.DeasyB. M.BadylakS.BuhringH. J.GiacobinoJ. P.LazzariL.HuardJ.PeaultB.2008A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell 333013131875-9777
  17. 17. De RosaA.De FrancescoF.TirinoV.FerraroG. A.DesiderioV.PainoF.PirozziG.D’AndreaF.PapaccioG.2009A new method for cryopreserving adipose-derived stem cells: an attractive and suitable large-scale and long-term cell banking technology. Tissue Eng Part C Methods 1546596671937-3392
  18. 18. De UgarteD. A.MorizonoK.ElbarbaryA.AlfonsoZ.ZukP. A.ZhuM.DragooJ. L.AshjianP.ThomasB.BenhaimP.ChenI.FraserJ.HedrickM. H.2003Comparison of multi-lineage cells from human adipose tissue and bone marrow. Cells Tissues Organs 17431011091422-6405
  19. 19. DellavalleA.SampaolesiM.TonlorenziR.TagliaficoE.SacchettiB.PeraniL.InnocenziA.GalvezB. G.MessinaG.MorosettiR.LiS.BelicchiM.PerettiG.ChamberlainJ. S.WrightW. E.TorrenteY.FerrariS.BiancoP.CossuG.2007Pericytes of human skeletal muscle are myogenic precursors distinct from satellite cells. Nat Cell Biol 932552671465-7392
  20. 20. DeslexS.NegrelR.VannierC.EtienneJ.AilhaudG.1987Differentiation of human adipocyte precursors in a chemically defined serum-free medium. Int J Obes 11119270307-0565
  21. 21. DragooJ. L.SamimiB.ZhuM.HameS. L.ThomasB. J.LiebermanJ. R.HedrickM. H.BenhaimP.2003Tissue-engineered cartilage and bone using stem cells from human infrapatellar fat pads. J Bone Joint Surg Br 8557407470030-1620X
  22. 22. EngfeldtP.ArnerP.OstmanJ.1980Influence of adipocyte isolation by collagenase on phosphodiesterase activity and lipolysis in man. J Lipid Res 2144434480022-2275
  23. 23. EricksonG. R.GimbleJ. M.FranklinD. M.RiceH. E.AwadH.GuilakF.2002Chondrogenic potential of adipose tissue-derived stromal cells in vitro and in vivo. Biochem Biophys Res Commun 29027637690000-6291X
  24. 24. EstesB. T.DiekmanB. O.GuilakF.2008Monolayer cell expansion conditions affect the chondrogenic potential of adipose-derived stem cells. Biotechnol Bioeng 9949869951097-0290
  25. 25. FarmerS. R.2006Transcriptional control of adipocyte formation. Cell Metab 442632731550-4131
  26. 26. FlynnL. E.PrestwichG. D.SempleJ. L.WoodhouseK. A.2008Proliferation and differentiation of adipose-derived stem cells on naturally derived scaffolds. Biomaterials 2912186218710142-9612
  27. 27. FryeC. A.PatrickC. W.2006Three-dimensional adipose tissue model using low shear bioreactors. In Vitro Cell Dev Biol Anim 425-61091141071-2690
  28. 28. GaustadK. G.BoquestA. C.AndersonB. E.GerdesA. M.CollasP.2004Differentiation of human adipose tissue stem cells using extracts of rat cardiomyocytes. Biochem Biophys Res Commun 31424204270000-6291X
  29. 29. GimbleJ.GuilakF.2003Adipose-derived adult stem cells: isolation, characterization, and differentiation potential. Cytotherapy 553623691465-3249
  30. 30. GimbleJ. M.KatzA. J.BunnellB. A.2007Adipose-derived stem cells for regenerative medicine. Circ Res 1009124912601524-4571
  31. 31. GondaK.ShigeuraT.SatoT.MatsumotoD.SugaH.InoueK.AoiN.KatoH.SatoK.MuraseS.KoshimaI.YoshimuraK.2008Preserved proliferative capacity and multipotency of human adipose-derived stem cells after long-term cryopreservation. Plast Reconstr Surg 12124014101529-4242
  32. 32. GregoireF. M.SmasC. M.SulH. S.1998Understanding adipocyte differentiation. Physiol Rev 7837838090031-9333
  33. 33. GronthosS.FranklinD. M.LeddyH. A.RobeyP. G.StormsR. W.GimbleJ. M.2001Surface protein characterization of human adipose tissue-derived stromal cells. J Cell Physiol 189154630021-9541
  34. 34. GuilakF.EstesB. T.DiekmanB. O.MoutosF. T.GimbleJ. M.2010Nicolas Andry Award: Multipotent adult stem cells from adipose tissue for musculoskeletal tissue engineering. Clin Orthop Relat Res 4689253025401528-1132
  35. 35. HalvorsenY. D.BondA.SenA.FranklinD. M.Lea-CurrieY. R.SujkowskiD.EllisP. N.WilkisonW. O.GimbleJ. M.2001Thiazolidinediones and glucocorticoids synergistically induce differentiation of human adipose tissue stromal cells: biochemical, cellular, and molecular analysis. Metabolism 5044074130026-0495
  36. 36. HalvorsenY. D.FranklinD.BondA. L.HittD. C.AuchterC.BoskeyA. L.PaschalisE. P.WilkisonW. O.GimbleJ. M.2001Extracellular matrix mineralization and osteoblast gene expression by human adipose tissue-derived stromal cells. Tissue Eng 767297411076-3279
  37. 37. HarrimanG. R.BogueM.RogersP.FinegoldM.PachecoS.BradleyA.ZhangY.MbawuikeI. N.1999Targeted deletion of the IgA constant region in mice leads to IgA deficiency with alterations in expression of other Ig isotypes. J Immunol 1625252125290022-1767
  38. 38. HaunerH.EntenmannG.WabitschM.GaillardD.AilhaudG.NegrelR.PfeifferE. F.1989Promoting effect of glucocorticoids on the differentiation of human adipocyte precursor cells cultured in a chemically defined medium. J Clin Invest 845166316700021-9738
  39. 39. HemmrichK.vonHeimburg. D.RendchenR.Di BartoloC.MilellaE.PalluaN.2005Implantation of preadipocyte-loaded hyaluronic acid-based scaffolds into nude mice to evaluate potential for soft tissue engineering. Biomaterials 2634702570370142-9612
  40. 40. HicokK. C.DuLaney. T. V.ZhouY. S.HalvorsenY. D.HittD. C.CooperL. F.GimbleJ. M.2004Human adipose-derived adult stem cells produce osteoid in vivo. Tissue Eng 103-43713801076-3279
  41. 41. HoJ. H.MaW. H.TsengT. C.ChenY. F.ChenM. H.LeeO. K.2010Isolation and Characterization of Multi-Potent Stem Cells from Human Orbital Fat Tissues. Tissue Eng Part A, pp., 0193-73351937335X
  42. 42. HuangJ. I.ZukP. A.JonesN. F.ZhuM.LorenzH. P.HedrickM. H.BenhaimP.2004Chondrogenic potential of multipotential cells from human adipose tissue. Plast Reconstr Surg 11325855940032-1052
  43. 43. ItoiY.TakatoriM.HyakusokuH.MizunoH.2010Comparison of readily available scaffolds for adipose tissue engineering using adipose-derived stem cells. J Plast Reconstr Aesthet Surg 6358588641878-0539
  44. 44. JackG. S.ZhangR.LeeM.XuY.WuB. M.RodriguezL. V.2009Urinary bladder smooth muscle engineered from adipose stem cells and a three dimensional synthetic composite. Biomaterials 3019325932701878-5905
  45. 45. KakudoN.MinakataT.MitsuiT.KushidaS.NotodihardjoF. Z.KusumotoK.2008Proliferation-promoting effect of platelet-rich plasma on human adipose-derived stem cells and human dermal fibroblasts. Plast Reconstr Surg 1225135213601529-4242
  46. 46. KangS. K.LeeD. H.BaeY. C.KimH. K.BaikS. Y.JungJ. S.2003Improvement of neurological deficits by intracerebral transplantation of human adipose tissue-derived stromal cells after cerebral ischemia in rats. Exp Neurol 18323553660014-4886
  47. 47. KatzA. J.TholpadyA.TholpadyS. S.ShangH.OgleR. C.2005Cell surface and transcriptional characterization of human adipose-derived adherent stromal (hADAS) cells. Stem Cells 2334124231066-5099
  48. 48. KirklandJ. L.HollenbergC. H.1998Inhibitors of preadipocyte replication: opportunities for the treatment of obesity. Prog Mol Subcell Biol 201771950079-6484
  49. 49. LazarM. A.2005PPAR gamma, 10 years later. Biochimie 8719130300-9084
  50. 50. LeeJ. A.ParrettB. M.ConejeroJ. A.LaserJ.ChenJ.KogonA. J.NandaD.GrantR. T.BreitbartA. S.2003Biological alchemy: engineering bone and fat from fat-derived stem cells. Ann Plast Surg 5066106170148-7043
  51. 51. LendeckelS.JodickeA.ChristophisP.HeidingerK.WolffJ.FraserJ. K.HedrickM. H.BertholdL.HowaldtH. P.2004Autologous stem cells (adipose) and fibrin glue used to treat widespread traumatic calvarial defects: case report. J Craniomaxillofac Surg 3263703731010-5182
  52. 52. LinS. D.WangK. H.KaoA. P.2008Engineered adipose tissue of predefined shape and dimensions from human adipose-derived mesenchymal stem cells. Tissue Eng Part A 1455715811937-3341
  53. 53. LiuZ. J.ZhugeY.VelazquezO. C.2009Trafficking and differentiation of mesenchymal stem cells. J Cell Biochem 10669849911097-4644
  54. 54. LuF.GaoJ. H.OgawaR.MizuroH.HykusokuH.2006Adipose tissues differentiated by adipose-derived stem cells harvested from transgenic mice. Chin J Traumatol 963593641008-1275
  55. 55. LuF.MizunoH.UysalC. A.CaiX.OgawaR.HyakusokuH.2008Improved viability of random pattern skin flaps through the use of adipose-derived stem cells. Plast Reconstr Surg 121150581529-4242
  56. 56. MatsumotoT.KanoK.KondoD.FukudaN.IribeY.TanakaN.MatsubaraY.SakumaT.SatomiA.OtakiM.RyuJ.MugishimaH.2008Mature adipocyte-derived dedifferentiated fat cells exhibit multilineage potential. J Cell Physiol 21512102221097-4652
  57. 57. MauneyJ. R.NguyenT.GillenK.Kirker-HeadC.GimbleJ. M.KaplanD. L.2007Engineering adipose-like tissue in vitro and in vivo utilizing human bone marrow and adipose-derived mesenchymal stem cells with silk fibroin 3D scaffolds. Biomaterials 2835528052900142-9612
  58. 58. MeinelL.KarageorgiouV.HofmannS.FajardoR.SnyderB.LiC.ZichnerL.LangerR.Vunjak-NovakovicG.KaplanD. L.2004Engineering bone-like tissue in vitro using human bone marrow stem cells and silk scaffolds. J Biomed Mater Res A 71125341549-3296
  59. 59. MizunoH.ZukP. A.ZhuM.LorenzH. P.BenhaimP.HedrickM. H.2002Myogenic differentiation by human processed lipoaspirate cells. Plast Reconstr Surg 1091199209discussion 210-1, 0032-1052
  60. 60. MoseleyT. A.ZhuM.HedrickM. H.2006Adipose-derived stem and progenitor cells as fillers in plastic and reconstructive surgery. Plast Reconstr Surg 1183Suppl, 121S128S1529-4242
  61. 61. NamS. Y.LobieP. E.2000The mechanism of effect of growth hormone on preadipocyte and adipocyte function. Obes Rev 1273861467-7881
  62. 62. NingH.LinG.LueT. F.LinC. S.2006Neuron-like differentiation of adipose tissue-derived stromal cells and vascular smooth muscle cells. Differentiation 749-105105180301-4681
  63. 63. OgawaR.MizunoH.WatanabeA.MigitaM.HyakusokuH.ShimadaT.2004Adipogenic differentiation by adipose-derived stem cells harvested from GFP transgenic mice-including relationship of sex differences. Biochem Biophys Res Commun 31925115170000-6291X
  64. 64. OwensG. K.KumarM. S.WamhoffB. R.2004Molecular regulation of vascular smooth muscle cell differentiation in development and disease. Physiol Rev 8437678010031-9333
  65. 65. ParkerA. M.ShangH.KhurgelM.KatzA. J.2007Low serum and serum-free culture of multipotential human adipose stem cells. Cytotherapy 976376461465-3249
  66. 66. PatrickC. W.Jr 2001Tissue engineering strategies for adipose tissue repair. Anat Rec 26343613660000-3276X
  67. 67. PatrickC. W.Jr ChauvinP. B.HobleyJ.ReeceG. P.1999Preadipocyte seeded PLGA scaffolds for adipose tissue engineering. Tissue Eng 521391511076-3279
  68. 68. PatrickC. W.Jr ZhengB.JohnstonC.ReeceG. P.2002Long-term implantation of preadipocyte-seeded PLGA scaffolds. Tissue Eng 822832931076-3279
  69. 69. PittengerM. F.MackayA. M.BeckS. C.JaiswalR. K.DouglasR.MoscaJ. D.MoormanM. A.SimonettiD. W.CraigS.MarshakD. R.1999Multilineage potential of adult human mesenchymal stem cells. Science 28454111431470036-8075
  70. 70. PreeceA.1972A Manual for Histologic Technicians. Boston, MA: Little, Brown and Co.
  71. 71. RodbellM.1966Metabolism of isolated fat cells. II. The similar effects of phospholipase C (Clostridium perfringens alpha toxin) and of insulin on glucose and amino acid metabolism. J Biol Chem 24111301390021-9258
  72. 72. RodbellM.1966The metabolism of isolated fat cells. IV. Regulation of release of protein by lipolytic hormones and insulin. J Biol Chem 24117390939170021-9258
  73. 73. RodbellM.JonesA. B.1966Metabolism of isolated fat cells. 3. The similar inhibitory action of phospholipase C (Clostridium perfringens alpha toxin) and of insulin on lipolysis stimulated by lipolytic hormones and theophylline. J Biol Chem 24111401420021-9258
  74. 74. RubinJ. P.BennettJ. M.DoctorJ. S.TebbetsB. M.MarraK. G.2007Collagenous microbeads as a scaffold for tissue engineering with adipose-derived stem cells. Plast Reconstr Surg 12024144241529-4242
  75. 75. SakumaT.MatsumotoT.KanoK.FukudaN.ObinataD.YamaguchiK.YoshidaT.TakahashiS.MugishimaH.2009Mature, adipocyte derived, dedifferentiated fat cells can differentiate into smooth muscle-like cells and contribute to bladder tissue regeneration. J Urol 18213553651527-3792
  76. 76. SchmidtC. E.BaierJ. M.2000Acellular vascular tissues: natural biomaterials for tissue repair and tissue engineering. Biomaterials 2122221522310142-9612
  77. 77. SenA.Lea-CurrieY. R.SujkowskaD.FranklinD. M.WilkisonW. O.HalvorsenY. D.GimbleJ. M.2001Adipogenic potential of human adipose derived stromal cells from multiple donors is heterogeneous. J Cell Biochem 8123123190730-2312
  78. 78. ShanahanC. M.WeissbergP. L.MetcalfeJ. C.1993Isolation of gene markers of differentiated and proliferating vascular smooth muscle cells. Circ Res 7311932040009-7330
  79. 79. ShillabeerG.FordenJ. M.LauD. C.1989Induction of preadipocyte differentiation by mature fat cells in the rat. J Clin Invest 8423813870021-9738
  80. 80. ShuX. Z. P. G.2004Therapeutic biomaterials from chemically modified hyaluronan. In: Garg HG, Hales CA, editors Chemistry and biology of hyaluronan. Amsterdam: Elsevier Press, 475504
  81. 81. SilbersteinL.WebsterS. G.TravisM.BlauH. M.1986Developmental progression of myosin gene expression in cultured muscle cells. Cell 467107510810092-8674
  82. 82. SolchagaL. A.DennisJ. E.GoldbergV. M.CaplanA. I.1999Hyaluronic acid-based polymers as cell carriers for tissue-engineered repair of bone and cartilage. J Orthop Res 1722052130736-0266
  83. 83. SoriskyA.1999From preadipocyte to adipocyte: differentiation-directed signals of insulin from the cell surface to the nucleus. Crit Rev Clin Lab Sci 3611341040-8363
  84. 84. SterodimasA.de FariaJ.NicarettaB.PitanguyI.2010Tissue engineering with adipose-derived stem cells (ADSCs): current and future applications. J Plast Reconstr Aesthet Surg 6311188618921878-0539
  85. 85. StillaertF. B.Di BartoloC.HuntJ. A.RhodesN. P.TognanaE.MonstreyS.BlondeelP. N.2008Human clinical experience with adipose precursor cells seeded on hyaluronic acid-based spongy scaffolds. Biomaterials 2929395339590142-9612
  86. 86. TchkoniaT.GiorgadzeN.PirtskhalavaT.TchoukalovaY.KaragiannidesI.ForseR. A.De PonteM.StevensonM.GuoW.HanJ.WalogaG.LashT. L.JensenM. D.KirklandJ. L.2002Fat depot origin affects adipogenesis in primary cultured and cloned human preadipocytes. Am J Physiol Regul Integr Comp Physiol 2825R1286R12960363-6119
  87. 87. ToyodaM.MatsubaraY.LinK.SugimachiK.FurueM.2009Characterization and comparison of adipose tissue-derived cells from human subcutaneous and omental adipose tissues. Cell Biochem Funct 2774404471099-0844
  88. 88. vonHeimburg. D.ZachariahS.HeschelI.KuhlingH.SchoofH.HafemannB.PalluaN.2001Human preadipocytes seeded on freeze-dried collagen scaffolds investigated in vitro and in vivo. Biomaterials 2254294380142-9612
  89. 89. vonHeimburg. D.ZachariahS.LowA.PalluaN.2001Influence of different biodegradable carriers on the in vivo behavior of human adipose precursor cells. Plast Reconstr Surg 1082411420discussion 421-2, 0032-1052
  90. 90. WeintraubH.DavisR.TapscottS.ThayerM.KrauseM.BenezraR.BlackwellT. K.TurnerD.RuppR.HollenbergS.et al.1991The myoD gene family: nodal point during specification of the muscle cell lineage. Science 25149957617660036-8075
  91. 91. WickhamM. Q.EricksonG. R.GimbleJ. M.VailT. P.GuilakF.2003Multipotent stromal cells derived from the infrapatellar fat pad of the knee. Clin Orthop Relat Res, 4121962120000-9921X
  92. 92. YoshimuraK.SatoK.AoiN.KuritaM.HirohiT.HariiK.2008Cell-assisted lipotransfer for cosmetic breast augmentation: supportive use of adipose-derived stem/stromal cells. Aesthetic Plast Surg 3214855discussion 56-7, 0036-4216X
  93. 93. YuanQ.ZengX.ChenL.PengE.YeZ.2010Comparison of myogenic differentiation ability of adipose-derived stem cells from different sites in rabbit]. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi 2410122812321002-1892
  94. 94. ZhangJ.ShehabeldinA.daCruz. L. A.ButlerJ.SomaniA. K.Mc GavinM.KozieradzkiI.dosSantos. A. O.NagyA.GrinsteinS.PenningerJ. M.SiminovitchK. A.1999Antigen receptor-induced activation and cytoskeletal rearrangement are impaired in Wiskott-Aldrich syndrome protein-deficient lymphocytes. J Exp Med 1909132913420022-1007
  95. 95. ZhangY. S.GaoJ. H.LuF.ZhuM.LiaoY. J.2007Cellular compatibility of type collagen I scaffold and human adipose-derived stem cells]. Nan Fang Yi Ke Da Xue Xue Bao 2722232251673-4254
  96. 96. ZukP. A.ZhuM.AshjianP.De UgarteD. A.HuangJ. I.MizunoH.AlfonsoZ. C.FraserJ. K.BenhaimP.HedrickM. H.2002Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell 1312427942951059-1524
  97. 97. ZukP. A.ZhuM.MizunoH.HuangJ.FutrellJ. W.KatzA. J.BenhaimP.LorenzH. P.HedrickM. H.2001Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng 722112281076-3279

Written By

Mathias Tremp, Souzan Salemi, Rita Gobet, Tullio Sulser and Daniel Eberli

Submitted: 17 November 2010 Published: 29 August 2011