Elsevier

Acta Biomaterialia

Volume 9, Issue 3, March 2013, Pages 5609-5620
Acta Biomaterialia

Preparation of an adipogenic hydrogel from subcutaneous adipose tissue

https://doi.org/10.1016/j.actbio.2012.11.003Get rights and content

Abstract

The ability to generate controlled amounts of adipose tissue would greatly ease the burden on hospitals for reconstructive surgery. We have previously shown that a tissue engineering chamber containing a vascular pedicle was capable of forming new fat; however, further refinements are required to enhance fat formation. The development and maintenance of engineered adipose tissue requires a suitable source of growth factors and a suitable scaffold. A hydrogel derived from adipose tissue may fulfil this need. Subcutaneous fat was processed into a thermosensitive hydrogel we refer to as adipose-derived matrix (ADM). Protein analysis revealed high levels of basement membrane proteins, collagens and detectable levels of growth factors. Adipose-derived stem cells exposed to this hydrogel differentiated into adipocytes with > 90% efficiency and in vivo testing in rats showed significant signs of adipogenesis after 8 weeks. ADM’s adipogenic properties combined with its simple gelation, relatively long shelf life and its tolerance to multiple freeze–thaw cycles, makes it a promising candidate for adipose engineering applications.

Introduction

Congenital, traumatic or post-surgical deformities such as mastectomy often require restoration of contour, usually involving adipose tissue. Not only does adipose tissue act as a reservoir for lipids, it also provides insulation and physical protection to the underlying tissue. Being vascularized, it makes an excellent graft bed for other tissues and can be used in complex reconstructive scenarios for which no appropriate donor tissue exists. Adipose tissue engineering has recently received much attention as it promises enhanced efficacy, reproducibility and predictability, compared with the contemporary methods used to treat disfiguring contour imperfections. Autologous free fat grafting with processed lipoaspirate has unpredictable results due to post-graft resorption with sometimes as little as 10% of the original fat volume retained [1], [2], [3]. While the use of vascularized fat flaps generally has more favourable results, complications such as flap failure, infections and pulmonary embolisms exist, along with morbidity relating to the donor site [4].

Once removed from their native environments, stem cells differentiate less efficiently [5], [6]. Therefore, when designing a suitable replacement three-dimensional (3-D) scaffold for thick tissues such as fat, it is important to incorporate characteristics of the native cellular environment to maintain optimal tissue development. For adipose tissue engineering, the ideal scaffold would be a self-gelling injectable material capable of inducing adipogenesis. Such a scaffold should contain the necessary extracellular matrix (ECM) components to initiate angiogenesis and subsequently induce resident stem cells to undergo adipogenesis. Of particular importance is the regulatory and structural role of the native extracellular matrix and associated factors [7]. The mechanotransduction between the ECM and cells plays a critical role in the regulation of angiogenesis [8] and in directing cells towards specific differentiation pathways [9]. Not only does the extracellular matrix provide structural support for the cells, it is also a reservoir for tissue-specific growth factors and signalling molecules that an entirely synthetic scaffold lacks.

Chemical crosslinking agents are successful in creating scaffolds from soluble proteins; however, the introduction of artificial linkages risks converting an otherwise native protein into something that may hinder cell infiltration and maturation [10].

Naturally derived scaffolds and hydrogels have been used for some years [11], [12], [13], [14], [15], [16], [17], [18], [19], [20], [21], [22] and have shown great potential in supporting cell growth whilst maintaining their volume. Sharma et al. demonstrated that an adipocyte-derived ECM extract supported hepatocytes with a higher metabolic activity than with Matrigel, a commercially available ECM hydrogel [14], whereas Flynn et al. showed that matrices prepared from acellular placental ECM and hyaluronic acid support the differentiation of adipose-derived stem cells (ADSCs) [18], [19], [20], [21], [22]. In another study, freeze-dried, injectable powders were prepared from human lipoaspirate [11]. This scaffolding material combined with ADSCs resulted in well-vascularized adipose tissue after implantation into nude mice. When prepared as a hydrogel, adipose-derived extracts have been shown to not only support the growth of seeded ADSCs [17], but also showed signs of inducing neoadipogenesis when implanted along the rat epigastric artery vascular pedicle [15]. However, many of the current formulations of adipose-derived hydrogels either require additional steps to initiate gelation or have inconsistent gelation. Many collagen-based extracts require acid solubilization, which may adversely affect some proteins in the extract.

From what can be gathered in the literature, it is clear that adipose-derived products are capable of promoting cellular infiltration and have the potential to help form new vascularized adipose tissue once implanted in animals. In this study our aims were twofold: first, to produce a hydrogel from adipose tissue containing intact, bioactive proteins, displaying consistent gelation under physiological conditions; and second, to determine the adipogenic potential of this product both in vitro and in vivo.

Section snippets

Preparation of adipogenic hydrogels

Frozen porcine subcutaneous adipose tissue was shaved into 1–2 g pieces and homogenized with an equal volume of phosphate-buffered saline (PBS) until it reached a smooth consistency. After centrifugation (1942g, 4 °C, 10 min), the tissue was treated with 2 U ml−1 dispase II (Roche, Australia) for 30 min in a shaking 37 °C incubator to help with decellularization. The tissue was then centrifuged (3000g, 4 °C, 10 min) and excess dispase removed. This was followed by washes with 2× volumes of salt buffer

Gelation

A thermosensitive hydrogel was prepared from subcutaneous adipose tissue using a heavily modified method based on one used to produce the skeletal muscle product, Myogel [28], and incorporating a previously reported enzymatic decellularization step [15] (Fig. 1). This gel displayed sol–gel properties similar to those of commercial purified collagen gels, remaining a viscous liquid at 4 °C and polymerizing once incubated at 37 °C. Complete gelation was reached after ∼15 min. The gelation of ADM did

Discussion

Tissue engineering is a relatively new field. At its heart, is the potential to regrow organs and regenerate lost tissues. The replacement of adipose tissue with a simple non-invasive procedure would drastically reduce the burden on hospitals for reconstructive surgery and help avoid complications associated with the implantation of synthetic materials. We have shown that adipose tissue provides all the components necessary to produce an extract capable of forming a gel under physiological

Conclusion

Human and porcine subcutaneous adipose tissue can be processed to form an extract containing high levels of ECM proteins and basement membrane components. This decellularized matrix is able to induce the adipogenic differentiation of ADSCs both in vivo and in vitro. On its own, the ADM induced significant differentiation of the resident cells to form fat, thus moving towards the establishment of a 3-D biological scaffold for clinical applications with wide ranging applications in the tissue

Acknowledgements

We wish to thank Dr. Caroline Taylor, Dr. Michael Findlay, Dr. Peter Mountford and Associate Professor Anthony Penington for their input and advice and the staff at EMSU for animal surgery. The O’Brien Institute acknowledges the Victorian State Government’s Department of Innovation, Industry and Regional Development’s Operational Infrastructure Support Program. This work was supported by the NBCF novel concept grant NC-11-01 and the DBI VSA grant Neopec.

References (59)

  • Y. Tsurutani et al.

    The roles of transforming growth factor-beta and Smad3 signaling in adipocyte differentiation and obesity

    Biochem Biophys Res Commun

    (2011)
  • S. Hirai et al.

    Activin A inhibits differentiation of 3T3-L1 preadipocyte

    Mol Cell Endocrinol

    (2005)
  • P. Pettersson et al.

    Adipocyte precursor cells in obese and nonobese humans

    Metabolism

    (1985)
  • T.L. Samuels et al.

    Pepsin as a causal agent of inflammation during nonacidic reflux

    Otolaryngol Head Neck Surg

    (2009)
  • Z.H. Li et al.

    Adipocyte differentiation factor (ADF): a protein secreted by mature fat cells that induces preadipocyte differentiation in culture

    Cell Biol Int

    (1998)
  • M.T. Boschert et al.

    Analysis of lipocyte viability after liposuction

    Plast Reconstr Surg

    (2002)
  • J.L. Mikus et al.

    Fate of liposuctioned and purified autologous fat injections in the canine vocal fold

    Laryngoscope

    (1995)
  • N. Blondeel et al.

    The donor site morbidity of free DIEP flaps and free TRAM flaps for breast reconstruction

    Br J Plast Surg

    (1997)
  • A. Chaubey et al.

    Extracellular matrix components as modulators of adult stem cell differentiation in an adipose system

    J Bioact Compat Polym

    (2008)
  • S.S. Chen et al.

    Cell-cell and cell-extracellular matrix interactions regulate embryonic stem cell differentiation

    Stem Cells

    (2007)
  • D.E. Ingber et al.

    Mechanochemical switching between growth and differentiation during fibroblast growth factor-stimulated angiogenesis in vitro: role of extracellular matrix

    J Cell Biol

    (1989)
  • K. Daniels et al.

    Modulation of chondrogenesis by the cytoskeleton and extracellular matrix

    J Cell Sci

    (1991)
  • M. Jarman-Smith et al.

    Porcine collagen crosslinking, degradation and its capability for fibroblast adhesion and proliferation

    J Mater Sci Mater Med

    (2004)
  • J.S. Choi et al.

    Human extracellular matrix (ECM) powders for injectable cell delivery and adipose tissue engineering

    J Control Release

    (2009)
  • J.S. Choi et al.

    Fabrication of porous extracellular matrix (ecm) scaffolds from human adipose tissue

    Tissue Eng Part C Methods

    (2009)
  • J.S. Choi et al.

    Fabrication of porous extracellular matrix scaffolds from human adipose tissue

    Tissue Eng Part C Methods

    (2010)
  • N.S. Sharma et al.

    Adipocyte-derived basement membrane extract with biological activity: applications in hepatocyte functional augmentation in vitro

    FASEB J

    (2010)
  • S. Uriel et al.

    Extraction and assembly of tissue-derived gels for cell culture and tissue engineering

    Tissue Eng Part C Methods

    (2009)
  • L. Flynn et al.

    Adipose tissue engineering in vivo with adipose-derived stem cells on naturally derived scaffolds

    J Biomed Mater Res A

    (2009)
  • Cited by (64)

    • Injectable human decellularized adipose tissue hydrogel containing stem cells enhances wound healing in mouse

      2020, Colloids and Surfaces A: Physicochemical and Engineering Aspects
      Citation Excerpt :

      Meanwhile, hDAT can be further processed into diversified forms including sheets, injectable powders, microcarriers, foams, beadfoams and so on [14–17]. However, for hDAT, only a few researchers have developed a form of hydrogel and just applied it to adipose tissue engineering [18–20]. Our research group has conducted a series of studies on hDAT, including its detailed physical, biological and material characterization, as well as its innovative application in wound healing.

    View all citing articles on Scopus
    1

    These authors contributed equally to this paper.

    View full text