Hostname: page-component-76fb5796d-25wd4 Total loading time: 0 Render date: 2024-04-25T16:02:29.669Z Has data issue: false hasContentIssue false

Human adipose tissue stem cells: relevance in the pathophysiology of obesity and metabolic diseases and therapeutic applications

Published online by Cambridge University Press:  10 December 2012

Angelo Cignarelli
Affiliation:
Department of Emergency and Organ Transplantation – Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
Sebastio Perrini
Affiliation:
Department of Emergency and Organ Transplantation – Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
Romina Ficarella
Affiliation:
Department of Emergency and Organ Transplantation – Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
Alessandro Peschechera
Affiliation:
Department of Emergency and Organ Transplantation – Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
Pasquale Nigro
Affiliation:
Department of Emergency and Organ Transplantation – Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
Francesco Giorgino*
Affiliation:
Department of Emergency and Organ Transplantation – Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
*
*Corresponding author: Francesco Giorgino, Department of Emergency and Organ Transplantation – Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, I-70124 Bari, Italy. E-mail: francesco.giorgino@uniba.it

Abstract

Stem cells are unique cells exhibiting self-renewing properties and the potential to differentiate into multiple specialised cell types. Totipotent or pluripotent stem cells are generally abundant in embryonic or fetal tissues, but the use of discarded embryos as sources of these cells raises challenging ethical problems. Adult stem cells can also differentiate into a wide variety of cell types. In particular, adult adipose tissue contains a pool of abundant and accessible multipotent stem cells, designated as adipose-derived stem cells (ASCs), that are able to replicate as undifferentiated cells, to develop as mature adipocytes and to differentiate into multiple other cell types along the mesenchymal lineage, including chondrocytes, myocytes and osteocytes, and also into cells of endodermal and neuroectodermal origin, including beta-cells and neurons, respectively. An impairment in the differentiation potential and biological functions of ASCs may contribute to the development of obesity and related comorbidities. In this review, we summarise different aspects of the ASCs with special reference to the isolation and characterisation of these cell populations, their relation to the biochemical features of the adipose tissue depot of origin and to the metabolic characteristics of the donor subject and discuss some prospective therapeutic applications.

Type
Review Article
Copyright
Copyright © Cambridge University Press 2012

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

References

1Giorgino, F., Laviola, L. and Eriksson, J. (2005) Regional differences of insulin action in adipose tissue: insights from in vivo and in vitro studies. Acta Physiologica Scandinavica 183, 13-30Google Scholar
2Laviola, L. et al. (2006) Insulin signalling in human adipose tissue. Archives of Physiology and Biochemistry 112, 82-88Google Scholar
3Tchkonia, T. et al. (2007) Identification of depot-specific human fat cell progenitors through distinct expression profiles and developmental gene patterns. American Journal of Physiology. Endocrinology and Metabolism 292, E298-E307Google Scholar
4Katz, A.J. et al. (2005) Cell surface and transcriptional characterization of human adipose-derived adherent stromal (hADAS) cells. Stem Cells 23, 412-423Google Scholar
5Perrini, S. et al. (2008) Fat depot-related differences in gene expression, adiponectin secretion, and insulin action and signalling in human adipocytes differentiated in vitro from precursor stromal cells. Diabetologia 51, 155-164Google Scholar
6Miyazaki, T. et al. (2005) Isolation of two human fibroblastic cell populations with multiple but distinct potential of mesenchymal differentiation by ceiling culture of mature fat cells from subcutaneous adipose tissue. Differentiation 73, 69-78CrossRefGoogle ScholarPubMed
7Matsumoto, T. et al. (2008) Fracture induced mobilization and incorporation of bone marrow-derived endothelial progenitor cells for bone healing. Journal of Cellular Physiology 215, 234-242Google Scholar
8Perrini, S. et al. (2009) Human adipose tissue precursor cells: a new factor linking regulation of fat mass to obesity and type 2 diabetes? Archives of Physiology and Biochemistry 115, 218-226Google Scholar
9Gimble, J.M. and Guilak, F. (2003) Differentiation potential of adipose derived adult stem (ADAS) cells. Current Topics in Developmental Biology 58, 137-160Google Scholar
10Zuk, P.A. et al. (2002) Human adipose tissue is a source of multipotent stem cells. Molecular Biology of the Cell 13, 4279-4295Google Scholar
11Zuk, P. et al. (2001) Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Engineering 7, 211-228Google Scholar
12Rodriguez, A. et al. (2005) The human adipose tissue is a source of multipotent stem cells. Biochimie 87, 125-128CrossRefGoogle ScholarPubMed
13Gimble, J.M. and Guilak, F. (2003) Adipose-derived adult stem cells: isolation, characterization, and differentiation potential. Cytotherapy 5, 362-369Google Scholar
14Lee, J.A. et al. (2003) Biological alchemy: engineering bone and fat from fat-derived stem cells. Annals of Plastic Surgery 50, 610-617Google Scholar
15Gronthos, S. et al. (2001) Surface protein characterization of human adipose tissue-derived stromal cells. Journal of Cellular Physiology 189, 54-63Google Scholar
16International Fat Applied Technology Society (IFATS) (2004). International Fat Applied Technology Society Determines That Stem Cells From Fat Offer Promising Clinical Opportunities for Future Research. The Free Library. Retrieved October 4, 2011 from http://www.thefreelibrary.com/InternationalFatAppliedTechnologySocietyDeterminesThatStem...-a0122826380Google Scholar
17Sugihara, H. et al. (1986) Primary cultures of unilocular fat cells: characteristics of growth in vitro and changes in differentiation properties. Differentiation 31, 42-49Google Scholar
18Tang, W. et al. (2008) White fat progenitor cells reside in the adipose vasculature. Science 322, 583-586CrossRefGoogle ScholarPubMed
19Gupta, R.K. et al. (2012) Zfp423 expression identifies committed preadipocytes and localizes to adipose endothelial and perivascular cells. Cell Metabolism 15, 230-239Google Scholar
20Zimmerlin, L. et al. (2010) Stromal vascular progenitors in adult human adipose tissue. Cytometry. Part A 77, 22-30Google Scholar
21Cai, X. et al. (2011) Adipose stem cells originate from perivascular cells. Biology of the Cell 103, 435-447Google Scholar
22Mareschi, K. et al. (2006) Neural differentiation of human mesenchymal stem cells: evidence for expression of neural markers and eag K+ channel types. Experimental Hematology 34, 1563-1572Google Scholar
23Ashjian, P.H. et al. (2003) In vitro differentiation of human processed lipoaspirate cells into early neural progenitors. Plastic and Reconstructive Surgery 111, 1922-1931Google Scholar
24Chandra, V. et al. (2011) Islet-like cell aggregates generated from human adipose tissue derived stem cells ameliorate experimental diabetes in mice. PLoS ONE 6, e20615Google Scholar
25Choi, B. et al. (2010) Cell behavior on extracellular matrix mimic materials based on mussel adhesive protein fused with functional peptides. Biomaterials 31, 8980-8988Google Scholar
26Choi, Y.S. et al. (2010) Differentiation of human adipose-derived stem cells into beating cardiomyocytes. Journal of Cellular and Molecular Medicine 14, 878-889Google Scholar
27Auxenfans, C. et al. (2011) Adipose-derived stem cells (ASCs) as a source of endothelial cells in the reconstruction of endothelialized skin equivalents. Journal of Tissue Engineering and Regenerative Medicine 6, 512-518Google Scholar
28Zhai, Y. et al. (2011) [In vitro effect of N-(6-aminohexyl)-5-chloro-1-naphthalenesulfonamide on differentiation from human adipose-derived mesenchymal stem cells to endothelial cells]. Zhongguo Yi Xue Ke Xue Yuan Xue Bao. Acta Academiae Medicinae Sinicae 33, 306-312Google Scholar
29Kuri-Harcuch, W. and Green, H. (1978) Adipose conversion of 3T3 cells depends on a serum factor. Proceedings of the National Academy of Sciences of the United States of America 75, 6107-6109Google Scholar
30Ailhaud, G., Grimaldi, P. and Négrel, R. (1992) Cellular and molecular aspects of adipose tissue development. Annual Review of Nutrition 12, 207-233Google Scholar
31Bernlohr, D. et al. (1985) Tissue specific expression of p422 protein, a putative lipid carrier, in mouse adipocytes. Biochemical and Biophysical Research Communications 132, 850-855Google Scholar
32Kim, K. et al. (2007) Pref-1 (preadipocyte factor 1) activates the MEK/extracellular signal-regulated kinase pathway to inhibit adipocyte differentiation. Molecular and Cellular Biology 27, 2294-2308Google Scholar
33Jing, K. et al. (2009) Expression regulation and function of Pref-1 during adipogenesis of human mesenchymal stem cells (MSCs). Biochimica et Biophysica Acta 1791, 816-826Google Scholar
34Boquest, A.C. et al. (2006) Epigenetic programming of mesenchymal stem cells from human adipose tissue. Stem Cell Reviews 2, 319-329Google Scholar
35Nedergaard, J. et al. (2007) Unexpected evidence for active brown adipose tissue in adult humans. American Journal of Physiology. Endocrinology and Metabolism 293, E444-E452Google Scholar
36Seale, P. et al. (2008) PRDM16 controls a brown fat/skeletal muscle switch. Nature 454, 961-967Google Scholar
37Timmons, J.A. et al. (2007) Myogenic gene expression signature establishes that brown and white adipocytes originate from distinct cell lineages. Proceedings of the National Academy of Sciences of the United States of America 104, 4401-4406Google Scholar
38Murano, I. et al. (2005) The adipose organ of Sv129 mice contains a prevalence of brown adipocytes and shows plasticity after cold exposure. Adipocytes 1, 121-130Google Scholar
39Himms-Hagen, J. et al. (2000) Multilocular fat cells in WAT of CL-316243-treated rats derive directly from white adipocytes. American Journal of Physiology. Cell Physiology 279, 670-681CrossRefGoogle ScholarPubMed
40Granneman, J.G. et al. (2005) Metabolic and cellular plasticity in white adipose tissue I: effects of beta3-adrenergic receptor activation. American Journal of Physiology. Endocrinology and Metabolism 289, 608-616Google Scholar
41Cinti, S. (2009) Transdifferentiation properties of adipocytes in the adipose organ. American Journal of Physiology. Endocrinology and Metabolism 297, 977-986CrossRefGoogle ScholarPubMed
42Petrovic, N. et al. (2010) Chronic peroxisome proliferator-activated receptor gamma (PPARgamma) activation of epididymally derived white adipocyte cultures reveals a population of thermogenically competent, UCP1-containing adipocytes molecularly distinct from classic brown adipocytes. Journal of Biological Chemistry 285, 7153-7164Google Scholar
43Halvorsen, Y. et al. (2001) Extracellular matrix mineralization and osteoblast gene expression by human adipose tissue-derived stromal cells. Tissue Engineering 7, 729-741Google Scholar
44Dragoo, J. et al. (2003) Tissue-engineered cartilage and bone using stem cells from human infrapatellar fat pads. Journal of Bone and Joint Surgery – British Volume 85, 740-747Google Scholar
45Gastaldi, G. et al. (2010) Human adipose-derived stem cells (hASCs) proliferate and differentiate in osteoblast-like cells on trabecular titanium scaffolds. Journal of Biomedical Materials Research. Part A 94, 790-799Google Scholar
46Nöth, U. et al. (2007) Chondrogenic differentiation of human mesenchymal stem cells in collagen type I hydrogels. Journal of Biomedical Materials Research. Part A 83, 626-635Google Scholar
47Yoon, I. et al. (2011) Proliferation and chondrogenic differentiation of human adipose-derived mesenchymal stem cells in porous hyaluronic acid scaffold. Journal of Bioscience and Bioengineering 112, 402-408Google Scholar
48Winter, A. et al. (2003) Cartilage-like gene expression in differentiated human stem cell spheroids: a comparison of bone marrow-derived and adipose tissue-derived stromal cells. Arthritis and Rheumatism 48, 418-429Google Scholar
49Erickson, G.R. et al. (2002) Chondrogenic potential of adipose tissue-derived stromal cells in vitro and in vivo. Biochemical and Biophysical Research Communications 290, 763-769CrossRefGoogle ScholarPubMed
50Mizuno, H. et al. (2002) Myogenic differentiation by human processed lipoaspirate cells. Plastic and Reconstructive Surgery 109, 199-209Google Scholar
51Bacou, F. et al. (2004) Transplantation of adipose tissue-derived stromal cells increases mass and functional capacity of damaged skeletal muscle. Cell Transplantation 13, 103-111CrossRefGoogle ScholarPubMed
52Vieira, N.M. et al. (2008) Human multipotent adipose-derived stem cells restore dystrophin expression of Duchenne skeletal-muscle cells in vitro. Biology of the Cell 100, 231-241Google Scholar
53da Justa Pinheiro, C.H. et al. (2011) Local Injections of adipose-derived mesenchymal stem cells modulate inflammation and increase angiogenesis ameliorating the dystrophic phenotype in dystrophin-deficient skeletal muscle. Stem Cell Reviews and Reports 8, 363-374Google Scholar
54Miranville, A. et al. (2004) Improvement of postnatal neovascularization by human adipose tissue-derived stem cells. Circulation 110, 349-355Google Scholar
55Rehman, J. et al. (2004) Secretion of angiogenic and antiapoptotic factors by human adipose stromal cells. Circulation 109, 1292-1298Google Scholar
56Liqing, Y. et al. (2011) Directed differentiation of motor neuron cell-like cells from human adipose-derived stem cells in vitro. Neuroreport 22, 370-373Google Scholar
57Cardozo, A.J., Gómez, D.E. and Argibay, P.F. (2011) Transcriptional characterization of Wnt and notch signaling pathways in neuronal differentiation of human adipose tissue-derived stem cells. Journal of Molecular Neuroscience 44, 186-194Google Scholar
58Woodbury, D. et al. (2000) Adult rat and human bone marrow stromal cells differentiate into neurons. Journal of Neuroscience Research 61, 364-370Google Scholar
59Arboleda, D. et al. (2011) Transplantation of predifferentiated adipose-derived stromal cells for the treatment of spinal cord injury. Cellular and Molecular Neurobiology 31, 1113-1122Google Scholar
60Scholz, T. et al. (2011) Neuronal differentiation of human adipose tissue-derived stem cells for peripheral nerve regeneration in vivo. Archives of Surgery 146, 666-674CrossRefGoogle ScholarPubMed
61Jang, S. et al. (2010) Functional neural differentiation of human adipose tissue-derived stem cells using bFGF and forskolin. BMC Cell Biology 11, 25-25Google Scholar
62Marchand, M. et al. (2009) Mouse ES cells over-expressing the transcription factor NeuroD1 show increased differentiation towards endocrine lineages and insulin-expressing cells. International Journal of Developmental Biology 53, 569-578Google Scholar
63Nakanishi, M. et al. (2007) Pancreatic tissue formation from murine embryonic stem cells in vitro. Differentiation; Research in Biological Diversity 75, 1-11Google Scholar
64D'Amour, K.A. et al. (2006) Production of pancreatic hormone-expressing endocrine cells from human embryonic stem cells. Nature Biotechnology 24, 1392-1401Google Scholar
65Blyszczuk, P. et al. (2004) Embryonic stem cells differentiate into insulin-producing cells without selection of nestin-expressing cells. International Journal of Developmental Biology 48, 1095-1104CrossRefGoogle ScholarPubMed
66Soria, B. (2001) In-vitro differentiation of pancreatic beta-cells. Differentiation 68, 205-219Google Scholar
67Lumelsky, N. et al. (2001) Differentiation of embryonic stem cells to insulin-secreting structures similar to pancreatic islets. Science 292, 1389-1394CrossRefGoogle ScholarPubMed
68Metzele, R. et al. (2011) Human adipose tissue-derived stem cells exhibit proliferation potential and spontaneous rhythmic contraction after fusion with neonatal rat cardiomyocytes. FASEB Journal 25, 830-839Google Scholar
69Vohl, M. et al. (2004) A survey of genes differentially expressed in subcutaneous and visceral adipose tissue in men. Obesity Research 12, 1217-1222Google Scholar
70Linder, K. et al. (2004) Differentially expressed genes in visceral or subcutaneous adipose tissue of obese men and women. Journal of Lipid Research 45, 148-154Google Scholar
71Atzmon, G. et al. (2002) Differential gene expression between visceral and subcutaneous fat depots. Hormone and Metabolic Research 34, 622-628Google Scholar
72Tchkonia, T. et al. (2006) Fat depot-specific characteristics are retained in strains derived from single human preadipocytes. Diabetes 55, 2571-2578Google Scholar
73Van Harmelen, V. et al. (2004) Comparison of proliferation and differentiation capacity of human adipocyte precursor cells from the omental and subcutaneous adipose tissue depot of obese subjects. Metabolism 53, 632-637Google Scholar
74Baglioni, S. et al. (2012) Functional differences in visceral and subcutaneous fat pads originate from differences in the adipose stem cell. PLoS ONE 7, e36569Google Scholar
75Gesta, S. et al. (2006) Evidence for a role of developmental genes in the origin of obesity and body fat distribution. Proceedings of the National Academy of Sciences of the United States of America 103, 6676-6681Google Scholar
76Sepe, A. et al. (2011) Aging and regional differences in fat cell progenitors – a mini-review. Gerontology 57, 66-75CrossRefGoogle ScholarPubMed
77Schipper, B.M. et al. (2008) Regional anatomic and age effects on cell function of human adipose-derived stem cells. Annals of Plastic Surgery 60, 538-544Google Scholar
78Shahparaki, A., Grunder, L. and Sorisky, A. (2002) Comparison of human abdominal subcutaneous versus omental preadipocyte differentiation in primary culture. Metabolism 51, 1211-1215Google Scholar
79Montague, C. et al. (1998) Depot-related gene expression in human subcutaneous and omental adipocytes. Diabetes 47, 1384-1391Google Scholar
80Adams, M. et al. (1997) Activators of peroxisome proliferator-activated receptor gamma have depot-specific effects on human preadipocyte differentiation. Journal of Clinical Investigation 100, 3149-3153Google Scholar
81Tchoukalova, Y.D. et al. (2010) Sex- and depot-dependent differences in adipogenesis in normal-weight humans. Obesity 18, 1875-1880Google Scholar
82Niesler, C., Siddel, K. and Prins, J. (1998) Human preadipocytes display a depot-specific susceptibility to apoptosis. Diabetes 47, 1365-1368Google Scholar
83Kern, P.A. et al. (2001) Adipose tissue tumor necrosis factor and interleukin-6 expression in human obesity and insulin resistance. American Journal of Physiology. Endocrinology and Metabolism 280, E745-E751Google Scholar
84Hotta, K. et al. (2001) Circulating concentrations of the adipocyte protein adiponectin are decreased in parallel with reduced insulin sensitivity during the progression to type 2 diabetes in rhesus monkeys. Diabetes 50, 1126-1133Google Scholar
85Friedman, J.M. et al. (1998) Leptin and the regulation of body weight in mammals. Nature 395, 763-770Google Scholar
86Iyengar, P. et al. (2003) Adipocyte-secreted factors synergistically promote mammary tumorigenesis through induction of anti-apoptotic transcriptional programs and proto-oncogene stabilization. Oncogene 22, 6408-6423Google Scholar
87D'Esposito, V. et al. (2012) Adipocyte-released insulin-like growth factor-1 is regulated by glucose and fatty acids and controls breast cancer cell growth in vitro. Diabetologia 55, 2811-2822CrossRefGoogle ScholarPubMed
88Baker, M. et al. (2005) Association between common polymorphisms of the proopiomelanocortin gene and body fat distribution: a family study. Diabetes 54, 2492-2496Google Scholar
89Nelson, T. et al. (2000) Genetic and environmental influences on body fat distribution, fasting insulin levels and CVD: are the influences shared? Twin Research 3, 43-50Google Scholar
90Spalding, K.L. et al. (2008) Dynamics of fat cell turnover in humans. Nature 453, 783-787Google Scholar
91Arner, P. et al. (2011) Dynamics of human adipose lipid turnover in health and metabolic disease. Nature 478, 110-113Google Scholar
92Permana, P.A. et al. (2004) Subcutaneous abdominal preadipocyte differentiation in vitro inversely correlates with central obesity. American Journal of Physiology. Endocrinology and Metabolism 286, E958-E962Google Scholar
93Tchoukalova, Y., Koutsari, C. and Jensen, M. (2007) Committed subcutaneous preadipocytes are reduced in human obesity. Diabetologia 50, 151-157Google Scholar
94Isakson, P. et al. (2009) Impaired preadipocyte differentiation in human abdominal obesity: role of Wnt, tumor necrosis factor-alpha, and inflammation. Diabetes 58, 1550-1557Google Scholar
95Sakurai, T. et al. (2010) Effects of exercise training on adipogenesis of stromal-vascular fraction cells in rat epididymal white adipose tissue. Acta Physiologica 200, 325-338Google Scholar
96Xu, X. et al. (2012) Altered adipocyte progenitor population and adipose-related gene profile in adipose tissue by long-term high-fat diet in mice. Life Sciences 90, 1001-1009Google Scholar
97Clinicaltrials.gov – U.S. National Institutes of Health (2012) Retrieved October 4, 2011 from http://clinicaltrials.govGoogle Scholar
98Kim, M. et al. (2006) Muscle regeneration by adipose tissue-derived adult stem cells attached to injectable PLGA spheres. Biochemical and Biophysical Research Communications 348, 386-392Google Scholar
99Tseng, Y.H., Cypess, A.M. and Kahn, C.R. (2010) Cellular bioenergetics as a target for obesity therapy. Nature Reviews. Drug Discovery 9, 465-482CrossRefGoogle ScholarPubMed
100Kuhbier, J.W. et al. (2010) Isolation, characterization, differentiation, and application of adipose-derived stem cells. Advances in Biochemical Engineering/Biotechnology 123, 55-105Google Scholar
101Gavrilova, O. et al. (2000) Surgical implantation of adipose tissue reverses diabetes in lipoatrophic mice. Journal of Clinical Investigation 105, 271-278Google Scholar
102Liu, Y. et al. (2007) Flk-1+ adipose-derived mesenchymal stem cells differentiate into skeletal muscle satellite cells and ameliorate muscular dystrophy in mdx mice. Stem Cells and Development 16, 695-706Google Scholar
103Thesleff, T. et al. (2011) Cranioplasty with adipose-derived stem cells and biomaterial: a novel method for cranial reconstruction. Neurosurgery 68, 1535-1540Google Scholar
104Nathan, S. et al. (2003) Cell-based therapy in the repair of osteochondral defects: a novel use for adipose tissue. Tissue Engineering 9, 733-744Google Scholar
105Cao, Y. et al. (2005) Human adipose tissue-derived stem cells differentiate into endothelial cells in vitro and improve postnatal neovascularization in vivo. Biochemical and Biophysical Research Communications 332, 370-379Google Scholar
106Cousin, B. et al. (2003) Reconstitution of lethally irradiated mice by cells isolated from adipose tissue. Biochemical and Biophysical Research Communications 301, 1016-1022Google Scholar
107Shi, D. et al. (2011) Human adipose tissue-derived mesenchymal stem cells facilitate the immunosuppressive effect of cyclosporin A on T lymphocytes through Jagged-1-mediated inhibition of NF-κB signaling. Experimental Hematology 39, 214-224.e1Google Scholar
108Tse, W.T. et al. (2003) Suppression of allogeneic T-cell proliferation by human marrow stromal cells: implications in transplantation. Transplantation 75, 389-397Google Scholar
109Puissant, B. et al. (2005) Immunomodulatory effect of human adipose tissue-derived adult stem cells: comparison with bone marrow mesenchymal stem cells. British Journal of Haematology 129, 118-129Google Scholar
110Madonna, R., Geng, Y. and De Caterina, R. (2009) Adipose tissue-derived stem cells: characterization and potential for cardiovascular repair. Arteriosclerosis, Thrombosis, and Vascular Biology 29, 1723-1729Google Scholar
111Hwangbo, S. et al. (2010) Therapeutic potential of human adipose stem cells in a rat myocardial infarction model. Yonsei Medical Journal 51, 69-76Google Scholar
112Bai, X. et al. (2010) Both cultured and freshly isolated adipose tissue-derived stem cells enhance cardiac function after acute myocardial infarction. European Heart Journal 31, 489-501Google Scholar
113Sanz-Ruiz, R. et al. (2009) Early translation of adipose-derived cell therapy for cardiovascular disease. Cell Transplantation 18, 245-254Google Scholar
114Bel, A. et al. (2010) Composite cell sheets: a further step toward safe and effective myocardial regeneration by cardiac progenitors derived from embryonic stem cells. Circulation 122, S118-23Google Scholar
115Danoviz, M.E. et al. (2010) Rat adipose tissue-derived stem cells transplantation attenuates cardiac dysfunction post infarction and biopolymers enhance cell retention. PLoS ONE 5, e12077CrossRefGoogle ScholarPubMed
116Giorgino, F. (2009) Adipose tissue function and dysfunction: organ cross talk and metabolic risk. American Journal of Physiology. Endocrinology and Metabolism 297, E975-E976Google Scholar

Further reading, resources and contacts

Sugii, S., Kida, Y., Berggren, W.T. & Evans, R.M. (2011) Feeder-dependent and feeder-independent iPS cell derivation from human and mouse adipose stem cells. Nature Protocols 6, 346358

This article describes a protocol for isolation, preparation and transformation of ASCs from fat tissue into mouse and human iPSCs.

Cinti, S. (2011) Between brown and white: novel aspects of adipocyte differentiation. Ann Med. 2011 43(2):104–15

This article reviews the ability of the adipose tissue of different depots to interconvert between white and brown cytotypes to meet changes in metabolic needs.

Sacks, HS, Fain, JN. (2011) Human epicardial fat: what is new and what is missing? Clin Exp Pharmacol Physiol. 38(12):879–87

This is an overview of our current knowledge on the anatomy, physiology, and pathophysiology of epicardial adipose tissue and its relationship to coronary atherosclerosis.

Gimble, JM, Bunnell, BA, Guilak, F. (2012) Human adipose-derived cells: an update on the transition to clinical translation. Regen Med. 7(2):225–35

The present article focuses on the literature of the past two years to assess the status of clinical and preclinical studies on adipose-derived cell therapies for regenerative medicine.

Peinado, JR, Pardo, M, de la Rosa, O, Malagón, MM. (2012) Proteomic characterization of adipose tissue constituents, a necessary step for understanding adipose tissue complexity. Proteomics. 12(4–5):607–20

This review discusses the recent proteomic studies on adipose tissue focused on the analysis of the distinct cellular components and their secretory products.

Sepe, A, Tchkonia, T, Thomou, T, Zamboni, M, Kirkland, JL (2011). Aging and regional differences in fat cell progenitors - a mini-review. Gerontology. 57(1):6675

This article discusses how inherent, age-related, depot-dependent alterations in preadipocyte function contribute to age-related fat tissue redistribution and metabolic dysfunction.

Registry and results database of publicly and privately supported clinical studies of human participants conducted around the world:

http://clinicaltrials.gov/

Providers of hASC

http://www.lonza.com/products-services/bio-research/primary-and-stem-cells/adult-stem-cells-and-media/human-adipose-derived-stem-cells-and-media/human-adipose-derived-stem-cells.aspx?WT.srch=1

http://products.invitrogen.com/ivgn/product/R7788115

http://www.atcc.org/ATCCAdvancedCatalogSearch/ProductDetails/tabid/452/Default.aspx?ATCCNum=PCS-500-011&Template=primaryCells

http://www.zen-bio.com/products/cells/adult_stem_cells.php

International Federation for Adipose Tissue and Science (IFATS) project aims to exchange information among researchers.

http://www.ifats.org/

COST (European Cooperation in Science and Technology) is one of the longest-running European frameworks supporting cooperation among scientists and researchers across Europe. COST Action BM0602 aims to advance knowledge on the pathogenesis and prevention of obesity and the specific role of adipose tissue in the development of the metabolic syndrome.

http://www.cost.eu/domains_actions/bmbs/Actions/BM0602