gms | German Medical Science

GMS German Plastic, Reconstructive and Aesthetic Surgery – Burn and Hand Surgery

Deutsche Gesellschaft der Plastischen, Rekonstruktiven und Ästhetischen Chirurgen (DGPRÄC)
Deutsche Gesellschaft für Verbrennungsmedizin (DGV)

ISSN 2193-7052

Supplementation of fat grafts with adipose-derived regenerative cells in reconstructive surgery

Research Article

  • corresponding author C. Herold - Department of Plastic, Hand, and Reconstructive Surgery, Medizinische Hochschule Hannover, Germany
  • P.M. Vogt - Department of Plastic, Hand, and Reconstructive Surgery, Medizinische Hochschule Hannover, Germany
  • M.N. Busche - Department of Plastic, Hand, and Reconstructive Surgery, Medizinische Hochschule Hannover, Germany
  • A. Jokuszies - Department of Plastic, Hand, and Reconstructive Surgery, Medizinische Hochschule Hannover, Germany
  • J. Park - Aesthetic & Plastic Surgery Institute, University of California at Irvine, CA, USA
  • H.O. Rennekampff - Department of Plastic, Hand, and Reconstructive Surgery, Medizinische Hochschule Hannover, Germany

GMS Ger Plast Reconstr Aesthet Surg 2012;2:Doc07

doi: 10.3205/gpras000009, urn:nbn:de:0183-gpras0000099

This is the English version of the article.
The German version can be found at: http://www.egms.de/de/journals/gpras/2012-2/gpras000009.shtml

Published: September 25, 2012

© 2012 Herold et al.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by-nc-nd/3.0/deed.en). You are free: to Share – to copy, distribute and transmit the work, provided the original author and source are credited.


Abstract

Introduction: The fraction of regenerative cells in adipose tissue has been described to be even higher than in bone marrow. Adipose tissue itself is excessively available in most patients. Given that adipose tissue is abundant in majority of patients adipose derrived stem cells (ASCs) have come under scrutiny for regenerative procedures in reconstructive surgery.

Material and methods: ASCs were extracted by the Celution® system for enrichment of fat grafts that were administered in patients with decreased wound healing, soft tissue or scar defects.
Results: All patients were satisfied after reconstruction with ASCs augmented fat grafts and no side effects were observed.

Discussion: The Celution® system provides fast recovery of ASCs which can be immediately utilized for appropriate application. Since a high number of stem cells are harvested from fat tissue no expansion of cells is needed as described for bone marrow derived stem cells. Enrichment of fat graft with ASCs is of great interest due to their reported angiogenetic effect. The reported cases demonstrate the potential of ASCs in the field of regenerative medicine and encourage further application in reconstructive surgery.


Introduction

Stem cell based therapy offers promising therapeutic options in regenerative medicine and various tissues have been described for their stem cell niches. Since Zuk et al. first described stem cells from adipose tissue ten years ago [1] many studies demonstrated their potential to differentiate into various cell lines. The fraction of regenerative cells in adipose tissue has been described to be similar to bone marrow or even exceed it by the factor of 200–300 [2], [3]. An average of 106 adipose derived stem cells (ASCs) can be isolated from 100 cc of aspirated adipose tissue [4]. The main advantage of ASCs is the abundant availability of the adipose tissue in most patients as well less limited morbidity associated with collection of the source tissue [5].


ASCs have been reported to secrete angiogentic and anti-apoptotice factors [6], and have the potential to differentiate into endothelial cells and become incorporated into vessels to promote neovascularisation [7], [8].


A positive effect in postmyocardial heart function of ASCs has been found in animal experiments [9]. Furthermore it has been described that ASCs are able to differentiate into chondrocytes [10]. An ASC-based regenerative therapy has also been shown to ameliorate soft tissue defects and improve skin quality [11]. Radiation dermatopathy has also been successfully treated in humans by stem cell augmented fat transplantation [12].

In order to use autogenous stem cells from adipose tissue under consideration of current regulations, a closed isolation system is needed located next to the patient.

The Celution® system has been invented by Cytori Therapeutics Inc. (San Diego, CA, USA) and is available on the European market. Under sterile conditions the Celution® system allows processing of lipoaspirate, gained from liposuction, in the operating room; the risk of contamination and destruction of the cells like on the transport to an outside laboratory is minimized. The automated process offers the possibility for standardized and reproducible isolation. In addition to augmented fat grafts in aesthetic surgery, adipose derived stem cells via such a process also open the door for other novel reconstructive applications such as:

1.
contracted or adherent scars,
2.
contour irregularities of the body surface,
3.
chronic wounds.

In this paper, we present our preliminary experience with ASC augmented fat grafting in reconstructive surgery.


Material and methods

Patients

We present four cases of ASC augmented fat grafting in reconstructive patients with volume deficits, scars or wound breakdown.

Liposuction – retrieval of fat grafts

Liposuction was used for retrieval of fat grafts and adipose tissue for isolation of ASC and stromal fraction. Tumescent solution containing 1 cc Epinephrine 1:1,000 and 10 cc sodiumbicarbonate 8.4% each 1,000 cc saline 0.9% was injected 20 minutes before liposuction. The suction lipectomy was performed manually using blunt cannulas with three holes (Figure 1 [Fig. 1]).

Celution® system and isolation of stromal fraction

The protocol for isolation of the ASC rich stromal fraction is based on the method described by Zuk et al , and employs Celution® by Cytori Therapeutics Inc., San Diego, CA, USA (Figure 2 [Fig. 2]). The lipoaspirate is transferred directly into the Celution® System (Figure 3 [Fig. 3]). After washing and depletion of erythrocytes, enzymatic digestion with several collagenases (Celase®) is started inside the chamber of the system. In the beginning a maximum of 260 cc of lipoaspirate (now after an update 360 cc) can be processed per aliquot in the Celution system. The enzymatically freed stromal fraction, containing the ASCs, is then transferred automatically to the integrated centrifuge. Several cycles of centrifugation and washing of the cells follow, resulting in approximately 5 cc of ASC rich stromal fraction no matter how much lipoaspirate has been injected [13].

Stem cell – stromal cell transfer

The required volume of fat graft for fat transplantation is gained once more by liposuction and placed in the system to be washed and mixed with the 5 cc of ASCs and stromal fraction (Figure 4 [Fig. 4], Figure 5 [Fig. 5]). The ASC augmented fat graft may now be taken from the system to be transplanted. Besides adipocytes, ACSs, endothelial progenitor cells and endothelial cells, pericytes, vascular smooth muscle cells, hematopoietic like stem cells, leukocytes and lymphocytes are transplanted [13].

Techique of injection

Using a special applicator, called Celbrush® (Figure 6 [Fig. 6]), the ASC augmented fat graft is injected in small aliquots in a fan shape pattern into the recipient tissue via 1–3 mm drops per 3 mm injection cannulas.


Results

Patient 1

A 58-year-old female patient after skin sparing mastectomy and adjuvant radiotherapy had persistent contour deformity and scarring of the right breast (Figure 7 [Fig. 7]) after failed attempts of reconstruction with a TRAM flap and a pedicled latissimus dorsi muscle flap elsewhere. There was a preoperative volume deficiency of approximately 150 cc on the right side which was addressed by fat grafting in the above describe manner in addition to corrective plastic surgery of the breast with reconstruction of the infra mammary fold. Adipose tissue was aspirated from bilateral gluteal regions and 260 cc of lipoaspirate were loaded into the Celution® system and processed to 5 cc of stemcell-stromacell solution. This 5 cc were mixed with 145 cc of freshly aspirated liposaspirate and a total of 150 cc were injected into the patient. Postoperatively the shape of her breast was ameliorated (Figure 8 [Fig. 8]).

Patient 2

A 47-year-old female patient with sclerodermia presented with a chronic ulcer overlying the partially necrotic achilles tendon. Attempts of reconstruction with skin grafts and a microvascular parascapular flap elsewhere had failed. After stabilisation of the wound bed by applying allogenic skin grafts (Figure 9 [Fig. 9]), the wound was laser debrided and pure 5 cc stromal cell fraction processed from 260 cc of lipoaspirate was applied without resuspension in additional fat graft. In this case the stromal fraction was mixed with autologous keratinocytes processed intraoperatively with the ReCell® Kit. This cell suspension was sprayed on the wound bed and covered with allogenous skin to limit mechanical shearing of the cells.

Two months postoperative the wound was reduced in size and depth, but a sufficient epithelization was not archived (Figure 10 [Fig. 10]).

Patient 3

A 27-year-old female patient presented with a scar formation of the superior aspect of the left breast and shoulder due to a thermal injury as a child. To reduce and soften the scar as well as to reconstruct the volume deficiency of the left breast, implantation of skin expanders were combined with autologous ASC augmented fat transplantation to the left breast (Figure 11 [Fig. 11]). Preoperative radiological imaging including mammography and MRI revealed no suspicious lesions or tumors within the breast tissue. The fat graft was harvested by liposuction from the abdomen and 135 cc were processed with the Celution® system. Fat grafting of 70 cc lipoaspirate augmented with 5 cc stromal fraction was performed to reconstruct the left mamma and to soften the scars. The expanders were explanted in time and resection of large areas of the scar could be done. The patients reports about softening of the scars and reduction of the contracture. Six month after surgery the asymmetry of the breasts was reduced and the volume deficiency of the left breast corrected (Figure 12 [Fig. 12]).

Patient 4

A 24-year-old female patient presented with a soft tissue defect of the left calf above the tibia (Figure 13 [Fig. 13]). A segment graft of the tibia had been performed following a resection of a osteofibrotic dysplasia in childhood. The course was complicated by an osteitis requiring several debridements. Examination shows an adherent scar and the asymmetry of her calf. Six months prior to ASC augmented fat grating, a conventional fat grafting procedure was performed, without sufficient volume augmentation (Figure 13 [Fig. 13]). In this patient 160 cc of lipoaspirate were processed with the Celution® system. After ASC augmented fat grafting with 50 cc the volume augmentation is more evident. Four months postoperatively, the improvement in volume deficiency is still maintained (Figure 14 [Fig. 14]).

The patients have been followed up for an average of 12 months (8–15 months) and no side effects of the therapy appeared.


Discussion

Treatment of our series of patients with ASC augmented fat grafting resulted in satisfying results. The angiogenetic effect of ASCs [7], [8] may play an important adjunctive role in the survival of transplanted adipose tissue. In 1893 Neuber reported, that the survival of fat grafts is related to the size of transplanted pieces [14]. Based on the finding that smaller pieces have a better survival Coleman developed his method of lipostructural autologous lipotransfer [15]. The angiogenetic potential of an ASC augmented fat graft supports the vitality and survival of the transplanted fat [16].

A major advantage of ASCs processed with the Celution® system is the immediate availability of the cells, obviating the need for time consuming processing in the laboratory as in instances of bone marrow preparations for clinical use. Such conventional cultivation methods are significantly more expensive and may in fact cause clinically relevant changes in cell biology [17]. Furthermore the amount of stem cells to be gained from bone marrow is limited, and associated with side effects for the donor. Stimulation medication before donation may not be free of any risks. These restrictions of bone marrow derived stem cells have led to an emerging interest in ASCs. More recently, adipose tissue has been recognized as a potentially bountiful source for various types of progenitor cells [18], [19].

Like bone marrow, adipose tissue develops from mesodermal tissue and gains volume by proliferation of progenitor cells and cell growth [16]. The progenitor cells, morphologically similar to fibroblast, have the ability of chondrogenous, adipogenous, neurogenous, osteogenous and myogenous differentiation [20], [21], [22].

Other factors suggest that adipose tissue may be a more viable source for progenitor cells. The harvesting of adipose tissue is identical to liposuction, a simple, widely utilized clinical procedure with a long track record of relative safety. The fraction of pluripotent cells in adipose tissue is also higher compared to that of bone marrow [4], [21], [23]. Combined with the abundance of fatty tissue in most patients makes adipose tissue an attractive source for the harvesting of abundant progenitor cells.

Cytori presents data that 100 cc of adipose tissue processed with the Celution® system result in 25 to 40 millions of cells. Lin et al. demonstrated that an average of 295,176 cells can be isolated from 1 cc of adipose tissue applying the Celution® system [13]. Suga et al. present data that one cm3 adipose tissue consist of 1 million adypocytes, 1 million ASCs, 1 million endothelial cells and 1 to 2 millions other cells like blood cells [24].

Results of injection of pure isolated stromal cells without accompanying fat tissue have shown insufficient volume augmentation. The Celution® approach adds the isolated stromal fraction to unprocessed plain fat grafts. The addition of additional adipose tissue to the graft not only augments the volume of the injectant, but also adds transferred adipose cells, which stand to benefit the most from the angiogenic effects of the ASCs. Animal studies have shown a 2.5 times higher fat preservation rate of this approach in comparison to unprocessed plain fat grafts [16].

Kim et al. present nice results in their actual study about fat cells differentiated from ASCs [25]. ASCs were isolated from lipoaspirates from the abdominal region and were cultivated for 24 to 26 days to correct soft tissue defects and facial scars. Progress control has been performed with 3 D scanners and appealing long tern results were demonstrated [25].

The cases presented in our series are first examples for regenerative therapies. Various therapeutic options of ASCs have been evaluated in animal models. In an ischemia reperfusion model the renoprotective ability of ASCs was demonstrated [26]. Flaps [27] or hind limb [28] perfusion could be ameliorated by ASCs and diabetic ulcera have been treated successfully [29]. Even functional improvement after myocardial [9] or cerebral infarction [30] have been described. Further research is compressively mentioned in the review papers by Mizuno [31], [32].

In clinical settings stem cell augmented fat transplantation or ASCs therapy has been performed in the treatment of facial lipoatrophy [33], breast augmentation [34], without giving a concrete volume survival rate, and perianal fistulas [35]. In a study consisting of 30 patients ASCs have been used for regenerative therapy of facial hemiatrophy, pectus excavatus, gluteal soft tissue defects and for breast reconstruction [36].

Rigotti reported promising results with ASC augmented fat grafts in the treatment of radiation-induced ulcers, although multiple fat grafts in combination with skin grafts was necessary [12]. In 19 of 20 patients an amelioration of the wounds were observed during an 18- to 31-months follow-up time. The high angiogenetic power of ASCs is discussed to be responsible for this finding. Creation of new vessels will consequently lead to an improved vascularisation, providing a more hospitable bed for the transferred adipose cells. In two other patients treated with ASC augmented fat grafts in our clinic, we were able to detect an improved tissue oxygenation by laser Doppler after treatment of radiation ulcer [37].

The angiogenetic potential of ASCs may be useful in breast augmentation and -reconstruction [16], [38] as it might lead to a superior volume survival – theoretically. This angiogenetic potential is especially in breast surgery socialized with the risk of tumour induction. There should be an awareness of this possible side effect, even if the in vitro findings of tumour induction for example in prostate tumours [39] or the interaction of ASCs with breast tumour cells [40] can not be transferred into clinical use directly.

Overall we found out that ASC augmented fat grafts are especially successful if vital adipose tissue is available at the recipient site and sufficient covering of the graft by local skin is given. It may be that there is a threshold level of native perfusion required in the recipient bed for the angiogenic effects of ASCs to take effect on the co-transplanted adipose tissue. As an extreme example, no amount of ASC-induced angiogenic augmentation would have an effect on a gangrenous wound. Furthermore, chronic inflammation likely blunts the effect of the anabolic potential of ASCs and in fact promotes resportion of any transplanted fat. This observation is clinically supported by the fact that wounds which have been made more quiescent in regards with inflammation with even temporary cadaveric skin grafts tend to do better with fat grafts in general.

The first examples of this work point up the potential of ASCs in regenerative medicine and open the door for further application in clinical reconstructive work.


Notes

Annotation

In Germany the clinical use of stem cell enhanced fat grafts for regenerative therapy and not for simple augmentation of adipose tissue is regulated by the German drug law (Arzneimittelgesetz) part 3 § 20 b and c.

Competing interests

The authors declare that they have no competing interests.


References

1.
Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, Benhaim P, Lorenz HP, Hedrick MH. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng. 2001 Apr;7(2):211-28. DOI: 10.1089/107632701300062859 External link
2.
De Ugarte DA, Morizono K, Elbarbary A, Alfonso Z, Zuk PA, Zhu M, Dragoo JL, Ashjian P, Thomas B, Benhaim P, Chen I, Fraser J, Hedrick MH. Comparison of multi-lineage cells from human adipose tissue and bone marrow. Cells Tissues Organs. 2003;174(3):101-9.
3.
Rydén M, Dicker A, Götherström C, Aström G, Tammik C, Arner P, Le Blanc K. Functional characterization of human mesenchymal stem cell-derived adipocytes. Biochem Biophys Res Commun. 2003 Nov;311(2):391-7.
4.
Meliga E, Strem BM, Duckers HJ, Serruys PW. Adipose-derived cells. Cell Transplant. 2007;16(9):963-70.
5.
Schreml S, Babilas P, Fruth S, Orsó E, Schmitz G, Mueller MB, Nerlich M, Prantl L. Harvesting human adipose tissue-derived adult stem cells: resection versus liposuction. Cytotherapy. 2009;11(7):947-57. DOI: 10.3109/14653240903204322 External link
6.
Sadat S, Gehmert S, Song YH, Yen Y, Bai X, Gaiser S, Klein H, Alt E. The cardioprotective effect of mesenchymal stem cells is mediated by IGF-I and VEGF. Biochem Biophys Res Commun. 2007 Nov;363(3):674-9. DOI: 10.1016/j.bbrc.2007.09.058 External link
7.
Cao Y, Sun Z, Liao L, Meng Y, Han Q, Zhao RC. Human adipose tissue-derived stem cells differentiate into endothelial cells in vitro and improve postnatal neovascularization in vivo. Biochem Biophys Res Commun. 2005 Jul;332(2):370-9. DOI: 10.1016/j.bbrc.2005.04.135 External link
8.
Rehman J, Traktuev D, Li J, Merfeld-Clauss S, Temm-Grove CJ, Bovenkerk JE, Pell CL, Johnstone BH, Considine RV, March KL. Secretion of angiogenic and antiapoptotic factors by human adipose stromal cells. Circulation. 2004 Mar;109(10):1292-8. DOI: 10.1161/01.CIR.0000121425.42966.F1 External link
9.
Schenke-Layland K, Strem BM, Jordan MC, Deemedio MT, Hedrick MH, Roos KP, Fraser JK, Maclellan WR. Adipose tissue-derived cells improve cardiac function following myocardial infarction. J Surg Res. 2009 May;153(2):217-23. DOI: 10.1016/j.jss.2008.03.019 External link
10.
Dragoo JL, Carlson G, McCormick F, Khan-Farooqi H, Zhu M, Zuk PA, Benhaim P. Healing full-thickness cartilage defects using adipose-derived stem cells. Tissue Eng. 2007 Jul;13(7):1615-21. DOI: 10.1089/ten.2006.0249 External link
11.
Mojallal A, Lequeux C, Shipkov C, Breton P, Foyatier JL, Braye F, Damour O. Improvement of skin quality after fat grafting: clinical observation and an animal study. Plast Reconstr Surg. 2009 Sep;124(3):765-74. DOI: 10.1097/PRS.0b013e3181b17b8f External link
12.
Rigotti G, Marchi A, Galiè M, Baroni G, Benati D, Krampera M, Pasini A, Sbarbati A. Clinical treatment of radiotherapy tissue damage by lipoaspirate transplant: a healing process mediated by adipose-derived adult stem cells. Plast Reconstr Surg. 2007 Apr;119(5):1409-22; discussion 1423-4. DOI: 10.1097/01.prs.0000256047.47909.71 External link
13.
Lin K, Matsubara Y, Masuda Y, Togashi K, Ohno T, Tamura T, Toyoshima Y, Sugimachi K, Toyoda M, Marc H, Douglas A. Characterization of adipose tissue-derived cells isolated with the Celution system. Cytotherapy. 2008;10(4):417-26. DOI: 10.1080/14653240801982979 External link
14.
Neuber F. Fetttransplantation. Chir Kongr Verhand Dtsch Gesellsch Chir. 1883;22:66.
15.
Coleman SR. Facial recontouring with lipostructure. Clin Plast Surg. 1997 Apr;24(2):347-67.
16.
Moseley TA, Zhu M, Hedrick MH. Adipose-derived stem and progenitor cells as fillers in plastic and reconstructive surgery. Plast Reconstr Surg. 2006 Sep;118(3 Suppl):121S-128S. DOI: 10.1097/01.prs.0000234609.74811.2e External link
17.
Vacanti V, Kong E, Suzuki G, Sato K, Canty JM, Lee T. Phenotypic changes of adult porcine mesenchymal stem cells induced by prolonged passaging in culture. J Cell Physiol. 2005 Nov;205(2):194-201. DOI: 10.1002/jcp.20376 External link
18.
Rodriguez AM, Elabd C, Amri EZ, Ailhaud G, Dani C. The human adipose tissue is a source of multipotent stem cells. Biochimie. 2005 Jan;87(1):125-8. DOI: 10.1016/j.biochi.2004.11.007 External link
19.
Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, Alfonso ZC, Fraser JK, Benhaim P, Hedrick MH. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002 Dec;13(12):4279-95. DOI: 10.1091/mbc.E02-02-0105 External link
20.
Gimble J, Guilak F. Adipose-derived adult stem cells: isolation, characterization, and differentiation potential. Cytotherapy. 2003;5(5):362-9. DOI: 10.1080/14653240310003026 External link
21.
Strem BM, Hicok KC, Zhu M, Wulur I, Alfonso Z, Schreiber RE, Fraser JK, Hedrick MH. Multipotential differentiation of adipose tissue-derived stem cells. Keio J Med. 2005 Sep;54(3):132-41.
22.
Tholpady SS, Aojanepong C, Llull R, Jeong JH, Mason AC, Futrell JW, Ogle RC, Katz AJ. The cellular plasticity of human adipocytes. Ann Plast Surg. 2005 Jun;54(6):651-6.
23.
Aust L, Devlin B, Foster SJ, Halvorsen YD, Hicok K, du Laney T, Sen A, Willingmyre GD, Gimble JM. Yield of human adipose-derived adult stem cells from liposuction aspirates. Cytotherapy. 2004;6(1):7-14. DOI: 10.1080/14653240310004539 External link
24.
Suga H, Eto H, Aoi N, Kato H, Araki J, Doi K, Higashino T, Yoshimura K. Adipose tissue remodeling under ischemia: death of adipocytes and activation of stem/progenitor cells. Plast Reconstr Surg. 2010 Dec;126(6):1911-23. DOI: 10.1097/PRS.0b013e3181f4468b External link
25.
Kim M, Kim I, Lee SK, Bang SI, Lim SY. Clinical trial of autologous differentiated adipocytes from stem cells derived from human adipose tissue. Dermatol Surg. 2011 Jun;37(6):750-9. DOI: 10.1111/j.1524-4725.2011.01765.x External link
26.
Feng Z, Ting J, Alfonso Z, Strem BM, Fraser JK, Rutenberg J, Kuo HC, Pinkernell K. Fresh and cryopreserved, uncultured adipose tissue-derived stem and regenerative cells ameliorate ischemia-reperfusion-induced acute kidney injury. Nephrol Dial Transplant. 2010 Dec;25(12):3874-84. DOI: 10.1093/ndt/gfq603 External link
27.
Lu F, Mizuno H, Uysal CA, Cai X, Ogawa R, Hyakusoku H. Improved viability of random pattern skin flaps through the use of adipose-derived stem cells. Plast Reconstr Surg. 2008 Jan;121(1):50-8. DOI: 10.1097/01.prs.0000293876.10700.b8 External link
28.
Moon MH, Kim SY, Kim YJ, Kim SJ, Lee JB, Bae YC, Sung SM, Jung JS. Human adipose tissue-derived mesenchymal stem cells improve postnatal neovascularization in a mouse model of hindlimb ischemia. Cell Physiol Biochem. 2006;17(5-6):279-90. DOI: 10.1159/000094140 External link
29.
Nambu M, Kishimoto S, Nakamura S, Mizuno H, Yanagibayashi S, Yamamoto N, Azuma R, Nakamura S, Kiyosawa T, Ishihara M, Kanatani Y. Accelerated wound healing in healing-impaired db/db mice by autologous adipose tissue-derived stromal cells combined with atelocollagen matrix. Ann Plast Surg. 2009 Mar;62(3):317-21. DOI: 10.1097/SAP.0b013e31817f01b6 External link
30.
Kang SK, Lee DH, Bae YC, Kim HK, Baik SY, Jung JS. Improvement of neurological deficits by intracerebral transplantation of human adipose tissue-derived stromal cells after cerebral ischemia in rats. Exp Neurol. 2003 Oct;183(2):355-66.
31.
Mizuno H. Adipose-derived stem cells for tissue repair and regeneration: ten years of research and a literature review. J Nihon Med Sch. 2009 Apr;76(2):56-66.
32.
Mizuno H. Adipose-derived stem and stromal cells for cell-based therapy: current status of preclinical studies and clinical trials. Curr Opin Mol Ther. 2010 Aug;12(4):442-9.
33.
Yoshimura K, Sato K, Aoi N, Kurita M, Inoue K, Suga H, Eto H, Kato H, Hirohi T, Harii K. Cell-assisted lipotransfer for facial lipoatrophy: efficacy of clinical use of adipose-derived stem cells. Dermatol Surg. 2008 Sep;34(9):1178-85. DOI: 10.1111/j.1524-4725.2008.34256.x External link
34.
Yoshimura K, Sato K, Aoi N, Kurita M, Hirohi T, Harii K. Cell-assisted lipotransfer for cosmetic breast augmentation: supportive use of adipose-derived stem/stromal cells. Aesthetic Plast Surg. 2008 Jan;32(1):48-55; discussion 56-7. DOI: 10.1007/s00266-007-9019-4 External link
35.
Garcia-Olmo D, Herreros D, Pascual I, Pascual JA, Del-Valle E, Zorrilla J, De-La-Quintana P, Garcia-Arranz M, Pascual M. Expanded adipose-derived stem cells for the treatment of complex perianal fistula: a phase II clinical trial. Dis Colon Rectum. 2009 Jan;52(1):79-86. DOI: 10.1007/DCR.0b013e3181973487 External link
36.
Tiryaki T, Findikli N, Tiryaki D. Staged stem cell-enriched tissue (SET) injections for soft tissue augmentation in hostile recipient areas: a preliminary report. Aesthetic Plast Surg. 2011 Dec;35(6):965-71. DOI: 10.1007/s00266-011-9716-x External link
37.
Herold C, Rennekampff HO, Krämer R, Hillmer A, Knobloch K, Vogt P. Stammzellangereicherte Fetttransplantation - eine mögliche Therapieoption bei Strahlenulzera? [Stem Cell-Enhanced Fat Transplantation - A Potential Therapeutic Option for Radiation Ulcus?]. Zentralbl Chir. 2011 Feb;:. DOI: 10.1055/s-0030-1247476 External link
38.
Zhu M, Zhou Z, Chen Y, Schreiber R, Ransom JT, Fraser JK, Hedrick MH, Pinkernell K, Kuo HC. Supplementation of fat grafts with adipose-derived regenerative cells improves long-term graft retention. Ann Plast Surg. 2010 Feb;64(2):222-8. DOI: 10.1097/SAP.0b013e31819ae05c External link
39.
Prantl L, Muehlberg F, Navone NM, Song YH, Vykoukal J, Logothetis CJ, Alt EU. Adipose tissue-derived stem cells promote prostate tumor growth. Prostate. 2010 Nov;70(15):1709-15. DOI: 10.1002/pros.21206 External link
40.
Zimmerlin L, Donnenberg AD, Rubin JP, Basse P, Landreneau RJ, Donnenberg VS. Regenerative therapy and cancer: in vitro and in vivo studies of the interaction between adipose-derived stem cells and breast cancer cells from clinical isolates. Tissue Eng Part A. 2011 Jan;17(1-2):93-106. DOI: 10.1089/ten.TEA.2010.0248 External link