Stem Cell Research-Sci Forschen

Full Text

Review Article
Fat Grafting with Adipose Stem Cells: The Successes and Challenges

  Yuping Ren1      Mikhail G Kolonin2,3      Yong Li3,4*   

1Division of Plastic Surgery, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
2Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston (UT Health), TX77030, USA
3Center of Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston (UT Health), TX77030, USA
4Department of Pediatric Surgery, University of Texas Medical School, Houston, Texas 77030, USA

*Corresponding author: Yong Li, Center of Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston (UT Health), TX77030, USA, Tel: 7135002438; E-mail: Li.1@uth.tmc.edu


Abstract

In recent years, fat grafting has become a popular method for clinically correcting soft tissue deformities. In order to increase fat graft viability a variety of modifications have been made to current grafting procedures including cell-assisted lipotransfer (CAL), structural fat grafting, fat grafting mixed with adipose stem cells (ASCs), fat grafting mixed with platelet rich plasma (PRP) and fat grafting mixed with both ASCs and PRP; however, the persistence of the grafts after transplantation remains in consistent. In this review, the biological fundamentals of lipotransfer procedures are discussed, the current progress of clinical fat grafting applications in plastic and reconstructive surgery are summarized, and the challenges facing fat transplantation are outlined.

Keywords

Fat grafting; Adipose stem/stromal cells (ASCs); Platelet rich plasma (PRP); Transplantation; Regeneration; Challenges

Correction of soft tissue contour deformities remains technically challenging for plastic and reconstructive surgeons. The ideal fillers for augmenting contour deformities should be physically and chemically stable, nontoxic, non-immunogenic, noninfectious, non-immunoreactive, and should be inexpensive to obtain and easily stored. Autologous material aspirated from white adipose tissue (WAT) has been recognized as an excellent injectablefiller for defect correction based on its biological characteristics. Through liposuction and minimal processing, large amounts of autologous adipose tissue can be harvested without significant donor-site morbidity. Currently, the use of fat grafting to fill soft tissue voids has become a very popular technique in plastic and reconstructive surgical clinics.

Historical Review of Clinically Applied Fat Grafting

The first use of free autologous fat transplantation in a clinical setting was performed by Dr. Gustav Neuber in 1893. He obtained excellent aesthetic results when he implanted detached segments of fat under the eyes of patients; however, when he tried to transplant larger grafts, the grafts failed. Harvesting the fat, used in the previous procedure resulted in a high degree of donor site morbidity and other complications which have been considered a major limitation for successful fat grafting. In 1987, Bircoll et al. [1] reported their use of liposuction for obtaining fat for breast augmentation and correcting dog-bite injury defects of the right thigh. The autologous fat obtained was transplanted using a small syringe and two and an half years after surgery, the patients presented with excellent aesthetic results. In 1988, Asken [2] described a new method for fat grafting using liposuction and micro lipo injection which resulted in excellent improvements in the contours of the patients’ face after injection. These results encouraged more clinicians to consider the use of lipo-transfer and fat grafting as a method to treat soft tissue deformities. Because the popularity of liposuction grew tremendously during the 1980s, the use of fat transplantation to augment or reconstruct soft tissue defects developed rapidly. Coleman et al. [3,4] optimized methods for harvesting fat and fat transfer in order to provide pure, intact parcels of fat, thus establishing the term” structural fat grafting”. This theory recommends that only a small quantity of fat should be injected in thin layers to increase the surface area the fat has contact with the receptor bed. This method of grafting fat is accepted by numerous plastic surgeons and has generated tremendous interest in the area of fat grafting, which has resulted in long-lasting and naturally-appearing grafts.

Today, fat grafting is utilized as a therapeutic method for the treatment of soft tissue volume loss due to aging, infection (e.g. facial lipoatrophy from human immunodeficiency virus infection), various types of trauma (mainly skin burns), idiopathic abnormalities (e.g. hemifacial microsomia) [5], tumor resection (e.g. breast reconstruction after mastectomy) [6], immune disease [7], and many other causes of soft tissue deficiency resulting in asymmetry or contour irregularity. Some clinicians have also used fat grafts for cosmetic breast augmentation [8] and for treating pressure ulcers [9], painful scars [10] and radiation-induced soft tissue injuries [11] (Table 1). Moreover, Covarrubias et al. [12] reported that patients with skin defects grafted with fat displayed evidence of regeneration including increased dermal thickness, collagen neo formation, and the presence of increased vascularity in skin adjacent to the lipofilling. Current results of fat grafting are impressive and bring significant benefits to patients who have undergone plastic and reconstructive surgery [13].

Table 1: Current application of fat grafting: a summary

Adipose-derived Stem Cells (ASCs)

It has become apparent over the years that white adipose tissue (WAT) is the most suitable autologous injectable filler for correcting soft tissue defects.WAT contains large numbers of adult stem cells and is an excellent natural resource for human augmentation procedures. During the last decade, several novel adult stem cell populations have been identified and isolated from adipose tissue [14-16]. Te term “adipose-derived stromal/ stem cells (ASCs)” is a term coined by the International Fat Applied Technology Society (IFATS) [17]. ASCs have been shown to be capable of self-renewal, and increased proliferative and multipotent differentiation capabilities. These cells also exhibit similar properties to mesenchymal stem cells (MSCs), which were first described and characterized in the bone marrow [18]. ASCs are localized around blood vessels [19,20] and are abundantly present in WAT where they constitute as much as 1% of the cell population, where as bone marrow MSCs (BM-MSCs) only comprise 0.001–0.002% of the cell population in bone marrow [21]. There are many cell types other than adipocytes within WAT [22]. Once WAT is aspirated and digested utilizing collagenase, the adipocytes can be isolated through centrifugation. The remaining cell fraction, called the stromal vascular fraction (SVF), contains vascular endothelial cells and various types of leukocytes in addition to ASCs [23]. Based on biomarker staining, ASCs can be isolated and characterized in vitro. Though ASCs demonstrate multipotency, not all ASCs can be induced to differentiate into adipocytes. Adipocyte progenitors are a heterogeneous population of cells, with some giving rise to white adipocytes and others give rise to beige adipocytes, which constitute brown adipose tissue (BAT) [24,25]. These ASCs can be distinguished from other adipocyte progenitor populations based on their expression of a variety of surface markers [26]. The regenerative capacity of ASCs during graft setting, and their contribution to WAT regeneration or re growth remains undefined. Recent studies have indicated that ASCs can promote angiogenesis in addition to suppressing inflammation, both of which are known behaviors of BM-MSCs [18].

Matsumoto et al. [27] performed a procedure termed cell-assisted lipotransfer (CAL) where the aspirated fat and the SVF cells are subcutaneously co-injected. Using a severe combined immunodeficiency (SCID) mouse model, the CAL procedure demonstrated superior cell survival and micro vessel formation after transplantation compared to the control non-CAL fat transplanted mice. This study showed that ASCs could differentiate into vascular endothelial cells and contribute to the neovascularization within the recipient tissue quickly after transplantation. Yoshimura and his team [23] have investigated and shown convincing evidence of very dynamic remodeling of the adipose tissue after non vascularized grafting [28]. Two weeks after fat tissue transplantation, they observed three distinct zones within the graft (300 µm from the peripheral edge of the graft toward the center): the surviving area (consisting mostly of ASCs and adipocytes), the regenerating area (transplanted adipocytes dead, but replaced with new adipocytes, ASC’s survived), and the necrotic area (both adipocytes and ASCs dead). It is believed that the adipocytes that died in the graft are due to watershed ischemia in the deep areas of the graft where little vascularization occurs [28]. Most of the cells in the surviving regions were ASCs, suggesting that the ASCs have a greater tolerance to ischemic stress. These findings represent convincing evidence of the importance of ASCs for supporting the success of fat grafting [29].

Taken together, these studies demonstrated that there resident ASCs within fat grafted tissues can: 1) differentiate into adipocytes and add structure to fill the implanted tissue defect [30]; 2) secrete growth factors, cytokines, and chemo-attractants that can enhance angiogenesis and increase localvascularization and blood supply; and 3) inhibit innate immune responses after tissue transplantation [31,32]. ASCs are also involved with establishing fat homeostasis. These properties support successful tissue regeneration and the long-term survival of the fat graft. The surgical procedure, e.g. fat retrieval and the transplantation techniques, including cell-assisted lipotransfer (CAL), also determine the success of fat grafting [29].

Use of Platelet Rich Plasma (PRP) in Combination with Fat Grafting

Platelet rich plasma (PRP) is an autologously derived biological material that has recently been used to facilitate fat graft survival. PRP was first described in 1975 by Oon and Hobbs [33] and first applied clinically in 1987 by Ferrari et al. [34] PRP is typically derived by centrifuging the patient’s whole blood to remove the erythrocytes and concentrate the platelets. The concentration of platelets in PRP is many times greater than that of peripheral blood [35]. The platelets contain α-granules, which are endowed with a great variety of growth factors including platelet-derived growth factor (PDGF), transforming growth factor-β (TGF-β), vascular endothelial growth factor (VEGF), epidermal growth factor (EGF), etc. After activation, typically after injury and coagulation, these factors are released by the platelets [36]. More than 500 clinical applications of PRP have been reported including its use for enhancing bone regeneration [37], wound healing [38], skin rejuvenation [39] and indeed fat augmentation [40].

Gentile et al. [40] performed a comparative translational study on fat grafting for the maintenance of tissue volume in breast reconstruction and found that the fat graft survival rate was 63% in patients treated via SVF-enhanced autologous fat grafts, 69% survival in patients treated with PRP-mixed fat grafts, and only 39% in patients treated with fat grafts alone. Retrospective studies from a cohort of 82 patients showed that fat grafts mixed with PRP reduced patient recovery time and improved the overall aesthetic outcome of facial lipofilling, compared to lipofilling using fat alone [41]. Moreover, numbers of animal studies have prospectively demonstrated that tissue grafting is improved with the use of PRP [42-44]. PRP can be viewed as a growth factor ‘factory’ supporting neoangiogenesis and enhancing the viability of the transplanted fat cells, which results in superior regeneration and grafting outcome.

Figure 1:The strategies of fat grafting after transplantation

Challenges in Fat Grafting

Although autologous fat grafting is a popular option for soft tissue augmentation, the technical challenges and potential complications of performing the procedure including the unpredictable and low survival rates of the graft limit its clinical application; moreover, central partial ischemia/necrosis often occurs in large areas of the transplanted fat. In addition, recent studies have shown that ASCs can promote tumor growth, [45-47] which have raised serious concerns regarding the safety of lipotransfer procedures [48,49]. Great variations with regard to fat graft survivability (40% to 80%) are also a concern which is probably due to technical procedure differences when harvesting the fat [50,51]. Other challenges include the critical maintenance of cell viability during the fat grafting procedures; however, there are no current techniques that have been developed to evaluate the donor fat for its ability to preserve the cellular viability. In addition, the extent to which ASCs or PRP can improve the outcomes of fat grafting in various procedures is still under investigation, though the results appear quite promising.

Conclusions

Fat grafting has become a popular procedure performed by plastic and reconstructive surgeons for facelifts, breast augmentation, and the treatment of other soft tissue defects. Several strategies have been performed to improve fat grafting as presented in Figure 1. Although the current grafting procedures, including cell-assisted lipotransfer (CAL), are effective methods that offer great potential for enhancing soft tissue regeneration, there remain many uncontrollable factors that can interfere with the efficacy, sustainability, and safety of current fat grafting techniques. Rigorous basic science and clinical studies involving both surgeons and research scientists are ongoing in an effort to improve the safety profile and outcomes of fat transplantation.

Acknowledgment

The authors would like to thank Dr. Kinga Vojnits for educational discussion and Mr. James Cummins for his editorial assistance.

References
  1. Bircoll M, Novack BH (1987) Autologous fat transplantation employing liposuction techniques. Ann Plast Surg 18: 327-329. [Ref.]
  2. Asken S (1988) Facial liposuction and microlipoinjection. J Dermatol Surg Oncol 14: 297-305. [Ref.]
  3. Coleman SR (1998) Structural fat grafting. Aesthet Surg J 18: 386-388. [Ref.]
  4. Coleman SR (2001) Structural fat grafts: the ideal filler? Clin Plast Surg 28: 111-119. [Ref.]
  5. Buckingham ED, Glasgold R, Kontis T, Smith SP Jr, Dolev Y, et al. (2015) Volume rejuvenation of the facial upper third. Facial Plast Surg 31: 43-54. [Ref.]
  6. Burnouf M, Buffet M, Schwarzinger M, Roman P, Bui P, et al. (2005) Evaluation of Coleman lipostructure for treatment of facial lipoatrophy in patients with human immunodeficiency virus and parameters associated with the efficiency of this technique. Arch Dermatol 141: 1220-1224. [Ref.]
  7. Pallua N, Baroncini A, Alharbi Z, Stromps JP (2014) Improvement of facial scar appearance and microcirculation by autologous lipofilling. J Plast Reconstr Aesthet Surg 67: 1033-1037. [Ref.]
  8. Liu SL, Peng YZ, Li XL, Yuan ZQ, Luo GX, et al. (2008) Clinical study on the repair of extensive deep burn wounds with autogenous fat granules and autologous microskin grafts in mixed grafting. Zhonghua Shao Shang Za Zhi 24: 122-125. [Ref.]
  9. Garland CB, Pomerantz JH (2012) Regenerative strategies for craniofacial disorders. Front Physiol 3: 453. [Ref.]
  10. Khouri RK, Rigotti G, Khouri RK Jr, Cardoso E, Marchi A, et al. (2015) Tissue-engineered breast reconstruction with brava-assisted fat grafting: a 7-year, 488-patient, multicenter experience. Plast Reconstr Surg 135: 643-658. [Ref.]
  11. Chen CC, Chen SN, Huang CL (2015) Correction of Sunken UpperEyelid Deformity in Young Asians by Minimally-Invasive Double-Eyelid Procedure and Simultaneous Orbital Fat Pad Repositioning: A OneYear Follow-up Study of 250 Cases. Aesthet Surg J 35: 359-366. [Ref.]
  12. Rivoalan F (2012) An oxycephaly deformation corrected with a fat tissue transplant. Ann Chir Plast Esthet 57: 618-621. [Ref.]
  13. Carraway JH (2010) Volume correction for nasojugal groove with blepharoplasty. Aesthet Surg J 30: 101-109. [Ref.]
  14. Balkin DM, Samra S, Steinbacher DM (2014) Immediate fat grafting in primary cleft lip repair. J Plast Reconstr Aesthet Surg 67: 1644-1650. [Ref.]
  15. Del Vecchio DA, Bucky LP (2011) Breast augmentation using preexpansion and autologous fat transplantation: a clinical radiographic study. Plast Reconstr Surg 127: 2441-2450. [Ref.]
  16. Marangi GF, Pallara T, Cagli B, Schena E, Giurazza F, et al. (2014) Treatment of early-stage pressure ulcers by using autologous adipose tissue grafts. Plast Surg Int 2014: 817283. [Ref.]
  17. Marino G, Moraci M, Armenia E, Orabona C, Sergio R, et al.(2013) Therapy with autologous adipose-derived regenerative cells for the care of chronic ulcer of lower limbs in patients with peripheral arterial disease. J Surg Res 185: 36-44. [Ref.]
  18. Baptista C, Iniesta A, Nguyen P, Legré R, Gay AM (2013) [Autologous fat grafting in the surgical management of painful scar: preliminary results]. Chir Main 32: 329-334. [Ref.]
  19. Scuderi N, Ceccarelli S, Onesti MG, Fioramonti P, Guidi C, et al. (2013) Human adipose-derived stromal cells for cell-based therapies in the treatment of systemic sclerosis. Cell Transplant 22: 779-795. [Ref.]
  20. Ibler KS, Gramkow C, Siemssen PA (2015) Autologous fat transplantation for the treatment of linear scleroderma en coup de sabre. Skinmed 13: 74-76. [Ref.]
  21. Bene MD, Pozzi MR, Rovati L, Mazzola I, Erba G, et sl. (2014) Autologous fat grafting for scleroderma-induced digital ulcers. An effective technique in patients with systemic sclerosis. Handchir Mikrochir Plast Chir 46: 242-247. [Ref.]
  22. Rigotti G, Marchi A, Galiè M, Baroni G, Benati D, et al. (2007) Clinical treatment of radiotherapy tissue damage by lipoaspirate transplant: a healing process mediated by adipose-derived adult stem cells. Plast Reconstr Surg 119: 1409-1422; discussion 1423-1404. [Ref.]
  23. Covarrubias P, Cárdenas-Camarena L, Guerrerosantos J, Valenzuela L, Espejo I, et al. (2013) Evaluation of the histologic changes in the fat-grafted facial skin: clinical trial. Aesthetic Plast Surg 37: 778-783. [Ref.]
  24. Lunder M, Bratkovic T, Doljak B, Kreft S, Urleb U, et al. (2005) Comparison of bacterial and phage display peptide libraries in search of target-binding motif. Appl Biochem Biotechnol 127: 125-131. [Ref.]
  25. Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, et al. (2002) Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell 13: 4279-4295. [Ref.]
  26. Erickson GR, Gimble JM, Franklin DM, Rice HE, Awad H, et al. (2002) Chondrogenic potential of adipose tissue-derived stromal cells in vitro and in vivo. Biochem Biophys Res Commun 290: 763-769. [Ref.]
  27. Nakagami H, Morishita R, Maeda K, Kikuchi Y, Ogihara T, et al. (2006) Adipose tissue-derived stromal cells as a novel option for regenerative cell therapy. J Atheroscler Thromb 13: 77-81. [Ref.]
  28. Gimble JM, Katz AJ, Bunnell BA (2007) Adipose-derived stem cells for regenerative medicine. Circulation Research 100: 1249-1260. [Ref.]
  29. Caplan AI, Correa D (2011) The MSC: an injury drugstore. Cell Stem Cell 9: 11-15. [Ref.]
  30. Lin CS, Xin ZC, Deng CH, Ning H, Lin G, et al. (2010) Defining adipose tissue-derived stem cells in tissue and in culture. Histol Histopathol 25: 807-815. [Ref.]
  31. Traktuev DO, Merfeld-Clauss S, Li J, Kolonin M, Arap W, et al. (2008) A Population of multipotent CD34-positive adipose stromal cells share pericyte and mesenchymal surface markers, reside in a periendothelial location, and stabilize endothelial networks. Circ Res 102: 77-85. [Ref.]
  32. Fraser JK, Wulur I, Alfonso Z, Hedrick MH (2006) Fat tissue: an underappreciated source of stem cells for biotechnology. Trends Biotechnol 24: 150-154. [Ref.]
  33. Kolonin MG, Evans KW, Mani SA, Gomer RH (2012) Alternative origins of stroma in normal organs and disease. Stem Cell Res 8: 312-323. [Ref.]
  34. Yoshimura K, Sato K, Aoi N, Kurita M, Hirohi T, et al. (2008) Cellassisted lipotransfer for cosmetic breast augmentation: supportive use of adipose-derived stem/stromal cells. Aesthetic Plast Surg 32: 48-55. [Ref.]
  35. Lee YH, Petkova AP, Mottillo EP, Granneman JG (2012) In vivo identification of bipotential adipocyte progenitors recruited by beta3- adrenoceptor activation and high-fat feeding. Cell Metab 15: 480-491. [Ref.]
  36. Daquinag AC, Tseng C, Salameh A, Zhang Y, Amaya-Manzanares F, et al. (2015) Depletion of white adipocyte progenitors induces beige adipocyte differentiation and suppresses obesity development. Cell Death Differ 22: 351-363. [Ref.]
  37. Berry R, Rodeheffer MS (2013) Characterization of the adipocyte cellular lineage in vivo. Nat Cell Biol 15: 302-308. [Ref.]
  38. Matsumoto D, Sato K, Gonda K, Takaki Y, Shigeura T, et al. (2006) Cell-assisted lipotransfer: supportive use of human adipose-derived cells for soft tissue augmentation with lipoinjection. Tissue Eng 12: 3375-3382. [Ref.]
  39. Eto H, Kato H, Suga H, Aoi N, Doi K, et al. (2012) The fate of adipocytes after nonvascularized fat grafting: evidence of early death and replacement of adipocytes. Plast Reconstr Surg 129: 1081-1092. [Ref.]
  40. Doi K, Ogata F, Eto H, Kato H, Kuno S, et al. (2015) Differential contributions of graft-derived and host-derived cells in tissue regeneration/remodeling after fat grafting. Plast Reconstr Surg 135: 1607-1617. [Ref.]
  41. Ogawa R, Mizuno H, Watanabe A, Migita M, Hyakusoku H, et al. (2004) Adipogenic differentiation by adipose-derived stem cells harvested from GFP transgenic mice-including relationship of sex differences. Biochem Biophys Res Commun 319: 511-517. [Ref.]
  42. Rehman J, Traktuev D, Li J, Merfeld-Clauss S, Temm-Grove CJ, et al. (2004) Secretion of angiogenic and antiapoptotic factors by human adipose stromal cells. Circulation 109: 1292-1298. [Ref.]
  43. Karnoub AE, Dash AB, Vo AP, Sullivan A, Brooks MW, et al. (2007) Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature 449: 557-563. [Ref.]
  44. Oon CJ, Hobbs JR (1975) Clinical applications of the continuous flow blood separator machine. Clin Exp Immunol 20: 1-16. [Ref.]
  45. Ferrari M, Zia S, Valbonesi M, Henriquet F, Venere G, et al. (1987) A new technique for hemodilution, preparation of autologous plateletrich plasma and intraoperative blood salvage in cardiac surgery. Int J Artif Organs 10: 47-50. [Ref.]
  46. Nurden AT, Nurden P, Sanchez M, Andia I, Anitua E (2008) Platelets and wound healing. Front Biosci 13: 3532-3548. [Ref.]
  47. Marx RE (2004) Platelet-rich plasma: evidence to support its use. J Oral Maxillofac Surg 62: 489-496. [Ref.]
  48. Eskan MA, Greenwell H, Hill M, Morton D, Vidal R, et al. (2014) Platelet-rich plasma-assisted guided bone regeneration for ridge augmentation: a randomized, controlled clinical trial. J Periodontol 85: 661-668. [Ref.]
  49. Villela DL, Santos VL (2010) Evidence on the use of platelet-rich plasma for diabetic ulcer: a systematic review. Growth Factors 28: 111-116. [Ref.]
  50. Shin MK, Lee JH, Lee SJ, Kim NI (2012) Platelet-rich plasma combined with fractional laser therapy for skin rejuvenation. Dermatol Surg 38: 623-630. [Ref.]
  51. Gentile P, Orlandi A, Scioli MG, Di Pasquali C, Bocchini I, et al. (2012) A comparative translational study: the combined use of enhanced stromal vascular fraction and platelet-rich plasma improves fat grafting maintenance in breast reconstruction. Stem Cells Transl Med 1: 341-351. [Ref.]
  52. Willemsen JC, van der Lei B, Vermeulen KM, Stevens HP (2014) The effects of platelet-rich plasma on recovery time and aesthetic outcome in facial rejuvenation: preliminary retrospective observations. Aesthetic Plast Surg 38: 1057-1063. [Ref.]
  53. Nakamura S, Ishihara M, Takikawa M, Murakami K, Kishimoto S, et al. (2010) Platelet-rich plasma (PRP) promotes survival of fat-grafts in rats. Ann Plast Surg 65: 101-106. [Ref.]
  54. Pires Fraga MF, Nishio RT, Ishikawa RS, Perin LF, Helene A Jr, et al. (2010) Increased survival of free fat grafts with platelet-rich plasma in rabbits. J Plast Reconstr Aesthet Surg 63: e818-822. [Ref.]
  55. Oh DS, Cheon YW, Jeon YR, Lew DH (2011) Activated plateletrich plasma improves fat graft survival in nude mice: a pilot study. Dermatol Surg 37: 619-625. [Ref.]
  56. Zhang Y, Daquinag AC, Amaya-Manzanares F, Sirin O, Tseng C, et al. (2012) Stromal progenitor cells from endogenous adipose tissue contribute to pericytes and adipocytes that populate the tumor microenvironment. Cancer Res 72: 5198-5208. [Ref.]
  57. Klopp AH, Zhang Y, Solley T, Amaya-Manzanares F, Marini F, et al. (2012) Omental adipose tissue-derived stromal cells promote vascularization and growth of endometrial tumors. Clin Cancer Res 18: 771-782. [Ref.]
  58. Zhang Y, Daquinag A, Traktuev DO, Amaya-Manzanares F, Simmons PJ, et al. (2009) White adipose tissue cells are recruited by experimental tumors and promote cancer progression in mouse models. Cancer Res 69: 5259-5266. [Ref.]
  59. Bertolini F, Petit JY, Kolonin MG (2015) Stem cells from adipose tissue and breast cancer: hype, risks and hope. Br J Cancer 112: 419-23. [Ref.]
  60. Bertolini F, Orecchioni S, Petit JY, Kolonin MG (2014) Obesity, proinflammatory mediators, adipose tissue progenitors, and breast cancer. Curr Opin Oncol 26: 545-550. [Ref.]
  61. Rubin A, Hoefflin SM (2002) Fat purification: survival of the fittest. Plast Reconstr Surg 109: 1463-1464. [Ref.]
  62. Zocchi ML, Zuliani F (2008) Bicompartmental breast lipostructuring. Aesthetic Plast Surg 32: 313-328. [Ref.]

Download Provisional PDF Here

Article Information

Aritcle Type: Review Article

Citation: Ren Y, Kolonin MG, Li Y (2015) Fat Grafting with Adipose Stem Cells: The Successes and Challenges. Int J Stem Cell Res 1(2): doi http:// dx.doi.org/10.16966/2472-6990.106

Copyright: © 2015 Ren Y, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Publication history: 

  • Received date: 22 October 2015

  • Accepted date: 29 October 2015

  • Published date: 31 October 2015
  •