Current Strategies to Enhance Adipose Stem Cell Function: An Update
Abstract
:1. Introduction
2. Adipose Stem Cells
2.1. Characteristics of ASCs
2.2. ASC Applications and Limitations
3. Function Enhancement Strategies
3.1. Pre-Conditioning (Priming or Licensing)
3.1.1. Pre-Conditioning with Cytokines
3.1.2. Pre-Conditioning with TLR Agonists
3.1.3. Pre-Conditioning with Other Pharmacological/Bioactive Molecules
3.2. Genetic Manipulation
Priming Regimen | In Vitro Effects | In Vivo Effects | Model/Condition | Reference |
---|---|---|---|---|
IFN-γ | IDO ↑ T-cell suppression ↑ | - | - | [80] |
IFN-γ | PD-L1 ↑ COX-2/PGE2 ↑ PBMC proliferation ↓ | - | - | [81] |
IFN-γ | IDO ↑ | T-cell infiltration ↓ Treg induction ↑ | Obliterative bronchiolitis model | [82] |
TNF-α | Proliferation ↑ Mobilization ↑ Osteogenesis ↑ | - | - | [83] |
TNF-α | IL-6 secretion ↑ IL-8 secretion ↑ | EP cell homing ↑ Angiogenesis ↑ | Ischemic hind limb model | [84] |
IFN-γ/ TNF-α | PGE2 ↑ IL-10 ↑ CCL2 ↑ | - | - | [85] |
IFN-γ/ TNF-α IL-17 | iNOS ↑ | T-cell suppression ↑ | ConA-induced hepatitis model | [88] |
IFN-γ/ TNF-α/ IL-6 | IDO ↑ Proliferation ↓ Cell diameter ↑ PBMC proliferation ↓ | - | - | [89] |
IL-1β/ IL-6/ IL-23 | No morphologic change CD45 expression ↑ Differentiation ↑ Allogeneic T-cell proliferation ↓ TGF-β ↑, IL-4 ↓ | - | - | [90] |
TGF-β | CXCR4 ↑ | Cancer homing ↑ Tumor volume ↓ Prolonged survival time ↑ | Glioblastoma | [91] |
LPS | IL-6, TNF-α, HGF ↑ VEGF ↑ | Liver regeneration ↑ Serum AST, ALT ↓ | Partial hepatectomy model | [101] |
PolyI:C | PGE2 ↑ IDO activity ↑ Restoring ASC inhibitory effect on pre-stimulated T cells | - | - | [102] |
3.2.1. Genetic Modification to Enhance Retention and Migration
3.2.2. Genetic Engineering to Improve Immunomodulation
3.2.3. Genetic Manipulation to Induce Lineage Transdifferentiation
3.3. Modification of Cell Culture Conditions
3.3.1. 3D Spheroid Formation
3.3.2. Hypoxic Treatment
3.3.3. Other Culture Environmental Modifications
3.4. Application of Extracellular Vesicles (EVs)
4. Conclusions
Author Contributions
Funding
Conflicts of Interest
Abbreviations
2D | Two-dimensional |
3D | Three-dimensional |
AD | Alzheimer’s disease |
ASC | Adipose-tissue derived mesenchymal stem cell |
BM-MSC | Bone marrow-derived mesenchymal stem cell |
COX-2 | Cyclooxygenase 2 |
CD | Cluster of differentiation |
CXCR | C-X-C chemokine receptor |
EAE | Experimental autoimmune encephalomyelitis |
ECM | Extracellular matrix |
EV | Extracellular vesicles |
fALS | Familial amyotrophic lateral sclerosis |
GvHD | Graft-versus-host disease |
HGF | Hepatocyte growth factor |
IDO | Indoleamin2,3-dioxygenase |
IFN-γ | Interferon gamma |
iNOS | Inducible nitric oxide synthase |
LPS | Lipopolysaccharide |
MHC | Major histocompatibility complex |
MSC | Mesenchymal stem cell |
NO | Nitric oxide |
PBMCs | Peripheral blood mononuclear cells |
PGE2 | Prostaglandin E2 |
Poly I:C | Polyinosinic–polycytidylic acid |
RA | Rheumatoid arthritis |
ROS | Reactive oxygen species |
SDF | Stromal-derived factor |
SOD | Superoxide dismutase |
SVF | Stromal vascular fraction |
TGF-β | Transforming growth factor-beta |
TLR | Toll-like receptor |
TNF-α | Tumor necrosis factor-alpha |
Tregs | Regulatory T cells |
VEGF | Vascular endothelial growth factor |
UC | Ultracentrifugation |
OE | Overexpressed |
ALS | Amyotrophic lateral sclerosis |
MS | Multiple sclerosis |
HCC | Hepatocellular carcinoma |
EP | Endothelial progenitor |
ConA | Concanavalin A |
AST | Aspartate transaminase |
ALT | Alanine transaminase |
References
- Caplan, A.I. Mesenchymal stem cells. J. Orthop. Res. 1991, 9, 641–650. [Google Scholar] [CrossRef] [PubMed]
- Caplan, A.I. Why are MSCs therapeutic? New data: New insight. J. Pathol. 2009, 217, 318–324. [Google Scholar] [CrossRef] [PubMed]
- Bartholomew, A.; Sturgeon, C.; Siatskas, M.; Ferrer, K.; McIntosh, K.; Patil, S.; Hardy, W.; Devine, S.; Ucker, D.; Deans, R.; et al. Mesenchymal stem cells suppress lymphocyte proliferation in vitro and prolong skin graft survival in vivo. Exp. Hematol. 2002, 30, 42–48. [Google Scholar] [CrossRef]
- Nauta, A.J.; Fibbe, W.E. Immunomodulatory properties of mesenchymal stromal cells. Blood 2007, 110, 3499–3506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Venkataiah, V.S.; Handa, K.; Njuguna, M.M.; Hasegawa, T.; Maruyama, K.; Nemoto, E.; Yamada, S.; Sugawara, S.; Lu, L.; Takedachi, M.; et al. Periodontal Regeneration by Allogeneic Transplantation of Adipose Tissue Derived Multi-Lineage Progenitor Stem Cells in vivo. Sci. Rep. 2019, 9, 921. [Google Scholar] [CrossRef] [PubMed]
- Tassi, S.A.; Sergio, N.Z.; Misawa, M.Y.O.; Villar, C.C. Efficacy of stem cells on periodontal regeneration: Systematic review of pre-clinical studies. J. Periodontal. Res. 2017, 52, 793–812. [Google Scholar] [CrossRef]
- Bassir, S.H.; Wisitrasameewong, W.; Raanan, J.; Ghaffarigarakani, S.; Chung, J.; Freire, M.; Andrada, L.C.; Intini, G. Potential for Stem Cell-Based Periodontal Therapy. J. Cell Physiol. 2016, 231, 50–61. [Google Scholar] [CrossRef] [PubMed]
- Le Blanc, K.; Rasmusson, I.; Sundberg, B.; Gotherstrom, C.; Hassan, M.; Uzunel, M.; Ringden, O. Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells. Lancet 2004, 363, 1439–1441. [Google Scholar] [CrossRef]
- Zappia, E.; Casazza, S.; Pedemonte, E.; Benvenuto, F.; Bonanni, I.; Gerdoni, E.; Giunti, D.; Ceravolo, A.; Cazzanti, F.; Frassoni, F.; et al. Mesenchymal stem cells ameliorate experimental autoimmune encephalomyelitis inducing T-cell anergy. Blood 2005, 106, 1755–1761. [Google Scholar] [CrossRef] [Green Version]
- Lee, R.H.; Seo, M.J.; Reger, R.L.; Spees, J.L.; Pulin, A.A.; Olson, S.D.; Prockop, D.J. Multipotent stromal cells from human marrow home to and promote repair of pancreatic islets and renal glomeruli in diabetic NOD/scid mice. Proc. Natl. Acad. Sci. USA 2006, 103, 17438–17443. [Google Scholar] [CrossRef]
- Augello, A.; Tasso, R.; Negrini, S.M.; Cancedda, R.; Pennesi, G. Cell therapy using allogeneic bone marrow mesenchymal stem cells prevents tissue damage in collagen-induced arthritis. Arthritis Rheum. 2007, 56, 1175–1186. [Google Scholar] [CrossRef] [PubMed]
- Gonzalez, M.A.; Gonzalez-Rey, E.; Rico, L.; Buscher, D.; Delgado, M. Adipose-derived mesenchymal stem cells alleviate experimental colitis by inhibiting inflammatory and autoimmune responses. Gastroenterology 2009, 136, 978–989. [Google Scholar] [CrossRef] [PubMed]
- Su, W.R.; Zhang, Q.Z.; Shi, S.H.; Nguyen, A.L.; Le, A.D. Human gingiva-derived mesenchymal stromal cells attenuate contact hypersensitivity via prostaglandin E2-dependent mechanisms. Stem Cells 2011, 29, 1849–1860. [Google Scholar] [CrossRef] [PubMed]
- Nemeth, K.; Keane-Myers, A.; Brown, J.M.; Metcalfe, D.D.; Gorham, J.D.; Bundoc, V.G.; Hodges, M.G.; Jelinek, I.; Madala, S.; Karpati, S.; et al. Bone marrow stromal cells use TGF-beta to suppress allergic responses in a mouse model of ragweed-induced asthma. Proc. Natl. Acad. Sci. USA 2010, 107, 5652–5657. [Google Scholar] [CrossRef] [PubMed]
- Kim, H.S.; Yun, J.W.; Shin, T.H.; Lee, S.H.; Lee, B.C.; Yu, K.R.; Seo, Y.; Lee, S.; Kang, T.W.; Choi, S.W.; et al. Human umbilical cord blood mesenchymal stem cell-derived PGE2 and TGF-beta1 alleviate atopic dermatitis by reducing mast cell degranulation. Stem Cells 2015, 33, 1254–1266. [Google Scholar] [CrossRef] [PubMed]
- Baer, P.C.; Geiger, H. Adipose-derived mesenchymal stromal/stem cells: Tissue localization, characterization, and heterogeneity. Stem Cells Int. 2012, 2012, 812693. [Google Scholar] [CrossRef] [PubMed]
- Constantin, G.; Marconi, S.; Rossi, B.; Angiari, S.; Calderan, L.; Anghileri, E.; Gini, B.; Bach, S.D.; Martinello, M.; Bifari, F.; et al. Adipose-derived mesenchymal stem cells ameliorate chronic experimental autoimmune encephalomyelitis. Stem Cells 2009, 27, 2624–2635. [Google Scholar] [CrossRef] [PubMed]
- Gonzalez-Rey, E.; Gonzalez, M.A.; Varela, N.; O’Valle, F.; Hernandez-Cortes, P.; Rico, L.; Buscher, D.; Delgado, M. Human adipose-derived mesenchymal stem cells reduce inflammatory and T cell responses and induce regulatory T cells in vitro in rheumatoid arthritis. Ann. Rheum. Dis. 2010, 69, 241–248. [Google Scholar] [CrossRef]
- Garcia-Olmo, D.; Garcia-Arranz, M.; Herreros, D.; Pascual, I.; Peiro, C.; Rodriguez-Montes, J.A. A phase I clinical trial of the treatment of Crohn’s fistula by adipose mesenchymal stem cell transplantation. Dis. Colon Rectum 2005, 48, 1416–1423. [Google Scholar] [CrossRef]
- Riordan, N.H.; Ichim, T.E.; Min, W.P.; Wang, H.; Solano, F.; Lara, F.; Alfaro, M.; Rodriguez, J.P.; Harman, R.J.; Patel, A.N.; et al. Non-expanded adipose stromal vascular fraction cell therapy for multiple sclerosis. J. Transl. Med. 2009, 7, 29. [Google Scholar] [CrossRef]
- Yanez, R.; Lamana, M.L.; Garcia-Castro, J.; Colmenero, I.; Ramirez, M.; Bueren, J.A. Adipose tissue-derived mesenchymal stem cells have in vivo immunosuppressive properties applicable for the control of the graft-versus-host disease. Stem Cells 2006, 24, 2582–2591. [Google Scholar] [CrossRef] [PubMed]
- Galipeau, J.; Sensebe, L. Mesenchymal Stromal Cells: Clinical Challenges and Therapeutic Opportunities. Cell Stem Cell 2018, 22, 824–833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Eggenhofer, E.; Benseler, V.; Kroemer, A.; Popp, F.C.; Geissler, E.K.; Schlitt, H.J.; Baan, C.C.; Dahlke, M.H.; Hoogduijn, M.J. Mesenchymal stem cells are short-lived and do not migrate beyond the lungs after intravenous infusion. Front. Immunol. 2012, 3, 297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, R.H.; Pulin, A.A.; Seo, M.J.; Kota, D.J.; Ylostalo, J.; Larson, B.L.; Semprun-Prieto, L.; Delafontaine, P.; Prockop, D.J. Intravenous hMSCs improve myocardial infarction in mice because cells embolized in lung are activated to secrete the anti-inflammatory protein TSG-6. Cell Stem Cell 2009, 5, 54–63. [Google Scholar] [CrossRef]
- McLeod, C.M.; Mauck, R.L. On the origin and impact of mesenchymal stem cell heterogeneity: New insights and emerging tools for single cell analysis. Eur. Cell Mater. 2017, 34, 217–231. [Google Scholar] [CrossRef] [PubMed]
- Ho, A.D.; Wagner, W.; Franke, W. Heterogeneity of mesenchymal stromal cell preparations. Cytotherapy 2008, 10, 320–330. [Google Scholar] [CrossRef] [PubMed]
- Bourin, P.; Bunnell, B.A.; Casteilla, L.; Dominici, M.; Katz, A.J.; March, K.L.; Redl, H.; Rubin, J.P.; Yoshimura, K.; Gimble, J.M. Stromal cells from the adipose tissue-derived stromal vascular fraction and culture expanded adipose tissue-derived stromal/stem cells: A joint statement of the International Federation for Adipose Therapeutics and Science (IFATS) and the International Society for Cellular Therapy (ISCT). Cytotherapy 2013, 15, 641–648. [Google Scholar] [PubMed]
- Phinney, D.G. Functional heterogeneity of mesenchymal stem cells: Implications for cell therapy. J. Cell Biochem. 2012, 113, 2806–2812. [Google Scholar] [CrossRef] [PubMed]
- Barbatelli, G.; Murano, I.; Madsen, L.; Hao, Q.; Jimenez, M.; Kristiansen, K.; Giacobino, J.P.; de Matteis, R.; Cinti, S. The emergence of cold-induced brown adipocytes in mouse white fat depots is determined predominantly by white to brown adipocyte transdifferentiation. Am. J. Physiol. Endocrinol. Metab. 2010, 298, 1244–1253. [Google Scholar] [CrossRef] [PubMed]
- Palumbo, P.; Miconi, G.; Cinque, B.; La Torre, C.; Lombardi, F.; Zoccali, G.; Orsini, G.; Leocata, P.; Giuliani, M.; Cifone, M.G. In vitro evaluation of different methods of handling human liposuction aspirate and their effect on adipocytes and adipose derived stem cells. J. Cell Physiol. 2015, 230, 1974–1981. [Google Scholar] [CrossRef] [PubMed]
- Vallee, M.; Cote, J.F.; Fradette, J. Adipose-tissue engineering: Taking advantage of the properties of human adipose-derived stem/stromal cells. Pathol. Biol. 2009, 57, 309–317. [Google Scholar] [CrossRef] [PubMed]
- Melief, S.M.; Zwaginga, J.J.; Fibbe, W.E.; Roelofs, H. Adipose tissue-derived multipotent stromal cells have a higher immunomodulatory capacity than their bone marrow-derived counterparts. Stem Cells Transl. Med. 2013, 2, 455–463. [Google Scholar] [CrossRef] [PubMed]
- Hass, R.; Kasper, C.; Bohm, S.; Jacobs, R. Different populations and sources of human mesenchymal stem cells (MSC): A comparison of adult and neonatal tissue-derived MSC. Cell Commun. Signal. 2011, 9, 12. [Google Scholar] [CrossRef] [PubMed]
- Bajek, A.; Gurtowska, N.; Olkowska, J.; Maj, M.; Kazmierski, L.; Bodnar, M.; Marszalek, A.; Debski, R.; Drewa, T. Does the Harvesting Technique Affect the Properties of Adipose-Derived Stem Cells?-The Comparative Biological Characterization. J. Cell Biochem. 2017, 118, 1097–1107. [Google Scholar] [CrossRef] [PubMed]
- Zuk, P.A.; Zhu, M.; Mizuno, H.; Huang, J.; Futrell, J.W.; Katz, A.J.; Benhaim, P.; Lorenz, H.P.; Hedrick, M.H. Multilineage cells from human adipose tissue: Implications for cell-based therapies. Tissue Eng. 2001, 7, 211–228. [Google Scholar] [CrossRef]
- Shah, F.S.; Wu, X.; Dietrich, M.; Rood, J.; Gimble, J.M. A non-enzymatic method for isolating human adipose tissue-derived stromal stem cells. Cytotherapy 2013, 15, 979–985. [Google Scholar] [CrossRef]
- Li, S.H.; Liao, X.; Zhou, T.E.; Xiao, L.L.; Chen, Y.W.; Wu, F.; Wang, J.R.; Cheng, B.; Song, J.X.; Liu, H.W. Evaluation of 2 Purification Methods for Isolation of Human Adipose-Derived Stem Cells Based on Red Blood Cell Lysis With Ammonium Chloride and Hypotonic Sodium Chloride Solution. Ann. Plast Surg. 2017, 78, 83–90. [Google Scholar] [CrossRef] [PubMed]
- Cinti, S. The adipose organ at a glance. Dis. Model Mech. 2012, 5, 588–594. [Google Scholar] [CrossRef] [Green Version]
- Kishi, K.; Imanishi, N.; Ohara, H.; Ninomiya, R.; Okabe, K.; Hattori, N.; Kubota, Y.; Nakajima, H.; Nakajima, T. Distribution of adipose-derived stem cells in adipose tissues from human cadavers. J. Plast Reconstr. Aesthet. Surg. 2010, 63, 1717–1722. [Google Scholar] [CrossRef]
- Zimmerlin, L.; Donnenberg, V.S.; Rubin, J.P.; Donnenberg, A.D. Mesenchymal markers on human adipose stem/progenitor cells. Cytometry A 2013, 83, 134–140. [Google Scholar] [CrossRef]
- Strem, B.M.; Hicok, K.C.; Zhu, M.; Wulur, I.; Alfonso, Z.; Schreiber, R.E.; Fraser, J.K.; Hedrick, M.H. Multipotential differentiation of adipose tissue-derived stem cells. Keio J. Med. 2005, 54, 132–141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wagner, W.; Wein, F.; Seckinger, A.; Frankhauser, M.; Wirkner, U.; Krause, U.; Blake, J.; Schwager, C.; Eckstein, V.; Ansorge, W.; et al. Comparative characteristics of mesenchymal stem cells from human bone marrow, adipose tissue, and umbilical cord blood. Exp. Hematol. 2005, 33, 1402–1416. [Google Scholar] [CrossRef] [PubMed]
- Gonzalez-Garza, M.T.; Cruz-Vega, D.E.; Cardenas-Lopez, A.; de la Rosa, R.M.; Moreno-Cuevas, J.E. Comparing stemness gene expression between stem cell subpopulations from peripheral blood and adipose tissue. Am. J. Stem Cells 2018, 7, 38–47. [Google Scholar] [PubMed]
- Lin, G.; Garcia, M.; Ning, H.; Banie, L.; Guo, Y.L.; Lue, T.F.; Lin, C.S. Defining stem and progenitor cells within adipose tissue. Stem Cells Dev. 2008, 17, 1053–1063. [Google Scholar] [CrossRef] [PubMed]
- Oedayrajsingh-Varma, M.J.; van Ham, S.M.; Knippenberg, M.; Helder, M.N.; Klein-Nulend, J.; Schouten, T.E.; Ritt, M.J.; van Milligen, F.J. Adipose tissue-derived mesenchymal stem cell yield and growth characteristics are affected by the tissue-harvesting procedure. Cytotherapy 2006, 8, 166–177. [Google Scholar] [CrossRef] [PubMed]
- Shah, F.S.; Li, J.; Zanata, F.; Curley, J.L.; Martin, E.C.; Wu, X.; Dietrich, M.; Devireddy, R.V.; Wade, J.W.; Gimble, J.M. The Relative Functionality of Freshly Isolated and Cryopreserved Human Adipose-Derived Stromal/Stem Cells. Cells Tissues Organs 2015, 201, 436–444. [Google Scholar] [CrossRef] [PubMed]
- Gronthos, S.; Franklin, D.M.; Leddy, H.A.; Robey, P.G.; Storms, R.W.; Gimble, J.M. Surface protein characterization of human adipose tissue-derived stromal cells. J. Cell Physiol. 2001, 189, 54–63. [Google Scholar] [CrossRef] [PubMed]
- Zannettino, A.C.; Paton, S.; Arthur, A.; Khor, F.; Itescu, S.; Gimble, J.M.; Gronthos, S. Multipotential human adipose-derived stromal stem cells exhibit a perivascular phenotype in vitro and in vivo. J. Cell Physiol. 2008, 214, 413–421. [Google Scholar] [CrossRef] [PubMed]
- Cuevas-Diaz Duran, R.; Gonzalez-Garza, M.T.; Cardenas-Lopez, A.; Chavez-Castilla, L.; Cruz-Vega, D.E.; Moreno-Cuevas, J.E. Age-related yield of adipose-derived stem cells bearing the low-affinity nerve growth factor receptor. Stem Cells Int. 2013, 2013, 372164. [Google Scholar] [CrossRef]
- Efimenko, A.Y.; Kochegura, T.N.; Akopyan, Z.A.; Parfyonova, Y.V. Autologous Stem Cell Therapy: How Aging and Chronic Diseases Affect Stem and Progenitor Cells. Biores. Open Access 2015, 4, 26–38. [Google Scholar] [CrossRef] [PubMed]
- Schipper, B.M.; Marra, K.G.; Zhang, W.; Donnenberg, A.D.; Rubin, J.P. Regional anatomic and age effects on cell function of human adipose-derived stem cells. Ann. Plast Surg. 2008, 60, 538–544. [Google Scholar] [CrossRef]
- Siegel, G.; Kluba, T.; Hermanutz-Klein, U.; Bieback, K.; Northoff, H.; Schafer, R. Phenotype, donor age and gender affect function of human bone marrow-derived mesenchymal stromal cells. BMC Med. 2013, 11, 146. [Google Scholar] [CrossRef]
- Shi, Y.Y.; Nacamuli, R.P.; Salim, A.; Longaker, M.T. The osteogenic potential of adipose-derived mesenchymal cells is maintained with aging. Plast. Reconstr. Surg. 2005, 116, 1686–1696. [Google Scholar] [CrossRef]
- De Girolamo, L.; Lopa, S.; Arrigoni, E.; Sartori, M.F.; Baruffaldi Preis, F.W.; Brini, A.T. Human adipose-derived stem cells isolated from young and elderly women: Their differentiation potential and scaffold interaction during in vitro osteoblastic differentiation. Cytotherapy 2009, 11, 793–803. [Google Scholar] [CrossRef]
- Onate, B.; Vilahur, G.; Camino-Lopez, S.; Diez-Caballero, A.; Ballesta-Lopez, C.; Ybarra, J.; Moscatiello, F.; Herrero, J.; Badimon, L. Stem cells isolated from adipose tissue of obese patients show changes in their transcriptomic profile that indicate loss in stemcellness and increased commitment to an adipocyte-like phenotype. BMC Genom. 2013, 14, 625. [Google Scholar] [CrossRef]
- Serena, C.; Keiran, N.; Ceperuelo-Mallafre, V.; Ejarque, M.; Fradera, R.; Roche, K.; Nunez-Roa, C.; Vendrell, J.; Fernandez-Veledo, S. Obesity and Type 2 Diabetes Alters the Immune Properties of Human Adipose Derived Stem Cells. Stem Cells 2016, 34, 2559–2573. [Google Scholar] [CrossRef]
- Pak, J.; Lee, J.H.; Kartolo, W.A.; Lee, S.H. Cartilage Regeneration in Human with Adipose Tissue-Derived Stem Cells: Current Status in Clinical Implications. Biomed. Res. Int. 2016, 2016, 4702674. [Google Scholar] [CrossRef]
- Zimmerlin, L.; Donnenberg, V.S.; Pfeifer, M.E.; Meyer, E.M.; Peault, B.; Rubin, J.P.; Donnenberg, A.D. Stromal vascular progenitors in adult human adipose tissue. Cytometry A 2010, 77, 22–30. [Google Scholar] [CrossRef]
- Koh, K.S.; Oh, T.S.; Kim, H.; Chung, I.W.; Lee, K.W.; Lee, H.B.; Park, E.J.; Jung, J.S.; Shin, I.S.; Ra, J.C.; et al. Clinical application of human adipose tissue-derived mesenchymal stem cells in progressive hemifacial atrophy (Parry-Romberg disease) with microfat grafting techniques using 3-dimensional computed tomography and 3-dimensional camera. Ann. Plast Surg. 2012, 69, 331–337. [Google Scholar] [CrossRef]
- Rigotti, G.; Charles-de-Sa, L.; Gontijo-de-Amorim, N.F.; Takiya, C.M.; Amable, P.R.; Borojevic, R.; Benati, D.; Bernardi, P.; Sbarbati, A. Expanded Stem Cells, Stromal-Vascular Fraction, and Platelet-Rich Plasma Enriched Fat: Comparing Results of Different Facial Rejuvenation Approaches in a Clinical Trial. Aesthet. Surg. J. 2016, 36, 261–270. [Google Scholar] [CrossRef]
- Pak, J.; Lee, J.H.; Pak, N.; Pak, Y.; Park, K.S.; Jeon, J.H.; Jeong, B.C.; Lee, S.H. Cartilage Regeneration in Humans with Adipose Tissue-Derived Stem Cells and Adipose Stromal Vascular Fraction Cells: Updated Status. Int. J. Mol. Sci. 2018, 19, 2146. [Google Scholar] [CrossRef]
- Im, G.I. Regeneration of articular cartilage using adipose stem cells. J. Biomed. Mater. Res. A 2016, 104, 1830–1844. [Google Scholar] [CrossRef]
- Bai, X.; Yan, Y.; Song, Y.H.; Seidensticker, M.; Rabinovich, B.; Metzele, R.; Bankson, J.A.; Vykoukal, D.; Alt, E. Both cultured and freshly isolated adipose tissue-derived stem cells enhance cardiac function after acute myocardial infarction. Eur. Heart J. 2010, 31, 489–501. [Google Scholar] [CrossRef]
- Ebrahimian, T.G.; Pouzoulet, F.; Squiban, C.; Buard, V.; Andre, M.; Cousin, B.; Gourmelon, P.; Benderitter, M.; Casteilla, L.; Tamarat, R. Cell therapy based on adipose tissue-derived stromal cells promotes physiological and pathological wound healing. Arterioscler. Thromb. Vasc. Biol. 2009, 29, 503–510. [Google Scholar] [CrossRef]
- Ivanova-Todorova, E.; Bochev, I.; Mourdjeva, M.; Dimitrov, R.; Bukarev, D.; Kyurkchiev, S.; Tivchev, P.; Altunkova, I.; Kyurkchiev, D.S. Adipose tissue-derived mesenchymal stem cells are more potent suppressors of dendritic cells differentiation compared to bone marrow-derived mesenchymal stem cells. Immunol. Lett. 2009, 126, 37–42. [Google Scholar] [CrossRef]
- Montespan, F.; Deschaseaux, F.; Sensebe, L.; Carosella, E.D.; Rouas-Freiss, N. Osteodifferentiated mesenchymal stem cells from bone marrow and adipose tissue express HLA-G and display immunomodulatory properties in HLA-mismatched settings: Implications in bone repair therapy. J. Immunol. Res. 2014, 2014, 230346. [Google Scholar] [CrossRef]
- Najar, M.; Raicevic, G.; Boufker, H.I.; Fayyad Kazan, H.; De Bruyn, C.; Meuleman, N.; Bron, D.; Toungouz, M.; Lagneaux, L. Mesenchymal stromal cells use PGE2 to modulate activation and proliferation of lymphocyte subsets: Combined comparison of adipose tissue, Wharton’s Jelly and bone marrow sources. Cell Immunol. 2010, 264, 171–179. [Google Scholar] [CrossRef]
- Stepien, A.; Dabrowska, N.L.; Maciagowska, M.; Macoch, R.P.; Zolocinska, A.; Mazur, S.; Siennicka, K.; Frankowska, E.; Kidzinski, R.; Chalimoniuk, M.; et al. Clinical Application of Autologous Adipose Stem Cells in Patients with Multiple Sclerosis: Preliminary Results. Mediators Inflamm. 2016, 2016, 5302120. [Google Scholar] [CrossRef]
- Alvaro-Gracia, J.M.; Jover, J.A.; Garcia-Vicuna, R.; Carreno, L.; Alonso, A.; Marsal, S.; Blanco, F.; Martinez-Taboada, V.M.; Taylor, P.; Martin-Martin, C.; et al. Intravenous administration of expanded allogeneic adipose-derived mesenchymal stem cells in refractory rheumatoid arthritis (Cx611): Results of a multicentre, dose escalation, randomised, single-blind, placebo-controlled phase Ib/IIa clinical trial. Ann. Rheum. Dis. 2017, 76, 196–202. [Google Scholar] [CrossRef]
- Bernardo, M.E.; Fibbe, W.E. Mesenchymal stromal cells: Sensors and switchers of inflammation. Cell Stem. Cell 2013, 13, 392–402. [Google Scholar] [CrossRef]
- Waterman, R.S.; Tomchuck, S.L.; Henkle, S.L.; Betancourt, A.M. A new mesenchymal stem cell (MSC) paradigm: Polarization into a pro-inflammatory MSC1 or an Immunosuppressive MSC2 phenotype. PLoS ONE 2010, 5, e10088. [Google Scholar] [CrossRef]
- Ryan, J.M.; Barry, F.; Murphy, J.M.; Mahon, B.P. Interferon-gamma does not break, but promotes the immunosuppressive capacity of adult human mesenchymal stem cells. Clin. Exp. Immunol. 2007, 149, 353–363. [Google Scholar] [CrossRef]
- Krampera, M. Mesenchymal stromal cell ’licensing’: A multistep process. Leukemia 2011, 25, 1408–1414. [Google Scholar] [CrossRef]
- Krampera, M.; Cosmi, L.; Angeli, R.; Pasini, A.; Liotta, F.; Andreini, A.; Santarlasci, V.; Mazzinghi, B.; Pizzolo, G.; Vinante, F.; et al. Role for interferon-gamma in the immunomodulatory activity of human bone marrow mesenchymal stem cells. Stem Cells 2006, 24, 386–398. [Google Scholar] [CrossRef]
- Croitoru-Lamoury, J.; Lamoury, F.M.; Caristo, M.; Suzuki, K.; Walker, D.; Takikawa, O.; Taylor, R.; Brew, B.J. Interferon-gamma regulates the proliferation and differentiation of mesenchymal stem cells via activation of indoleamine 2,3 dioxygenase (IDO). PLoS ONE 2011, 6, e14698. [Google Scholar] [CrossRef]
- Noone, C.; Kihm, A.; English, K.; O’Dea, S.; Mahon, B.P. IFN-gamma stimulated human umbilical-tissue-derived cells potently suppress NK activation and resist NK-mediated cytotoxicity in vitro. Stem Cells Dev. 2013, 22, 3003–3014. [Google Scholar] [CrossRef]
- Yamamoto, M.; Ota, A.; Hori, T.; Imai, S.; Sohma, H.; Suzuki, N.; Hatakeyama, N.; Inazawa, N.; Ito, Y.M.; Kimura, H.; et al. Early expression of plasma CCL8 closely correlates with survival rate of acute graft-vs.-host disease in mice. Exp. Hematol. 2011, 39, 1101–1112. [Google Scholar] [CrossRef]
- Muller, M.; Carter, S.; Hofer, M.J.; Campbell, I.L. Review: The chemokine receptor CXCR3 and its ligands CXCL9, CXCL10 and CXCL11 in neuroimmunity--a tale of conflict and conundrum. Neuropathol. Appl. Neurobiol. 2010, 36, 368–387. [Google Scholar] [CrossRef]
- Schoenborn, J.R.; Wilson, C.B. Regulation of interferon-gamma during innate and adaptive immune responses. Adv. Immunol. 2007, 96, 41–101. [Google Scholar]
- DelaRosa, O.; Lombardo, E.; Beraza, A.; Mancheno-Corvo, P.; Ramirez, C.; Menta, R.; Rico, L.; Camarillo, E.; Garcia, L.; Abad, J.L.; et al. Requirement of IFN-gamma-mediated indoleamine 2,3-dioxygenase expression in the modulation of lymphocyte proliferation by human adipose-derived stem cells. Tissue Eng. Part. A 2009, 15, 2795–2806. [Google Scholar] [CrossRef]
- Kronsteiner, B.; Wolbank, S.; Peterbauer, A.; Hackl, C.; Redl, H.; van Griensven, M.; Gabriel, C. Human mesenchymal stem cells from adipose tissue and amnion influence T-cells depending on stimulation method and presence of other immune cells. Stem Cells Dev. 2011, 20, 2115–2126. [Google Scholar] [CrossRef]
- Zheng, G.; Qiu, G.; Ge, M.; He, J.; Huang, L.; Chen, P.; Wang, W.; Xu, Q.; Hu, Y.; Shu, Q.; et al. Human adipose-derived mesenchymal stem cells alleviate obliterative bronchiolitis in a murine model via IDO. Respir Res. 2017, 18, 119. [Google Scholar] [CrossRef] [Green Version]
- Lu, Z.; Wang, G.; Dunstan, C.R.; Chen, Y.; Lu, W.Y.; Davies, B.; Zreiqat, H. Activation and promotion of adipose stem cells by tumour necrosis factor-alpha preconditioning for bone regeneration. J. Cell Physiol. 2013, 228, 1737–1744. [Google Scholar] [CrossRef]
- Kwon, Y.W.; Heo, S.C.; Jeong, G.O.; Yoon, J.W.; Mo, W.M.; Lee, M.J.; Jang, I.H.; Kwon, S.M.; Lee, J.S.; Kim, J.H. Tumor necrosis factor-alpha-activated mesenchymal stem cells promote endothelial progenitor cell homing and angiogenesis. Biochim. Biophys. Acta 2013, 1832, 2136–2144. [Google Scholar] [CrossRef]
- Prasanna, S.J.; Gopalakrishnan, D.; Shankar, S.R.; Vasandan, A.B. Pro-inflammatory cytokines, IFNgamma and TNFalpha, influence immune properties of human bone marrow and Wharton jelly mesenchymal stem cells differentially. PLoS ONE 2010, 5, e9016. [Google Scholar] [CrossRef]
- Cuerquis, J.; Romieu-Mourez, R.; Francois, M.; Routy, J.P.; Young, Y.K.; Zhao, J.; Eliopoulos, N. Human mesenchymal stromal cells transiently increase cytokine production by activated T cells before suppressing T-cell proliferation: Effect of interferon-gamma and tumor necrosis factor-alpha stimulation. Cytotherapy 2014, 16, 191–202. [Google Scholar] [CrossRef]
- Domenis, R.; Cifu, A.; Quaglia, S.; Pistis, C.; Moretti, M.; Vicario, A.; Parodi, P.C.; Fabris, M.; Niazi, K.R.; Soon-Shiong, P.; et al. Pro inflammatory stimuli enhance the immunosuppressive functions of adipose mesenchymal stem cells-derived exosomes. Sci. Rep. 2018, 8, 13325. [Google Scholar] [CrossRef]
- Han, X.; Yang, Q.; Lin, L.; Xu, C.; Zheng, C.; Chen, X.; Han, Y.; Li, M.; Cao, W.; Cao, K.; et al. Interleukin-17 enhances immunosuppression by mesenchymal stem cells. Cell Death Differ. 2014, 21, 1758–1768. [Google Scholar] [CrossRef]
- Crop, M.J.; Baan, C.C.; Korevaar, S.S.; Ijzermans, J.N.; Pescatori, M.; Stubbs, A.P.; van Ijcken, W.F.; Dahlke, M.H.; Eggenhofer, E.; Weimar, W.; et al. Inflammatory conditions affect gene expression and function of human adipose tissue-derived mesenchymal stem cells. Clin. Exp. Immunol. 2010, 162, 474–486. [Google Scholar] [CrossRef]
- Pourgholaminejad, A.; Aghdami, N.; Baharvand, H.; Moazzeni, S.M. The effect of pro-inflammatory cytokines on immunophenotype, differentiation capacity and immunomodulatory functions of human mesenchymal stem cells. Cytokine 2016, 85, 51–60. [Google Scholar] [CrossRef]
- Li, M.; Zeng, L.; Liu, S.; Dangelmajer, S.; Kahlert, U.D.; Huang, H.; Han, Y.; Chi, X.; Zhu, M.; Lei, T. Transforming Growth Factor-beta Promotes Homing and Therapeutic Efficacy of Human Mesenchymal Stem Cells to Glioblastoma. J. Neuropathol. Exp. Neurol. 2019, 78, 315–325. [Google Scholar] [CrossRef]
- Galipeau, J. Reply: “Function of Cryopreserved Mesenchymal Stromal Cells With and Without Interferon-gamma Prelicensing Is Context Dependent”. Stem Cells 2017, 35, 1440–1441. [Google Scholar] [CrossRef]
- Hwa Cho, H.; Bae, Y.C.; Jung, J.S. Role of toll-like receptors on human adipose-derived stromal cells. Stem Cells 2006, 24, 2744–2752. [Google Scholar] [CrossRef]
- Liotta, F.; Angeli, R.; Cosmi, L.; Fili, L.; Manuelli, C.; Frosali, F.; Mazzinghi, B.; Maggi, L.; Pasini, A.; Lisi, V.; et al. Toll-like receptors 3 and 4 are expressed by human bone marrow-derived mesenchymal stem cells and can inhibit their T-cell modulatory activity by impairing Notch signaling. Stem Cells 2008, 26, 279–289. [Google Scholar] [CrossRef]
- Raicevic, G.; Rouas, R.; Najar, M.; Stordeur, P.; Boufker, H.I.; Bron, D.; Martiat, P.; Goldman, M.; Nevessignsky, M.T.; Lagneaux, L. Inflammation modifies the pattern and the function of Toll-like receptors expressed by human mesenchymal stromal cells. Hum. Immunol. 2010, 71, 235–244. [Google Scholar] [CrossRef]
- Raicevic, G.; Najar, M.; Stamatopoulos, B.; De Bruyn, C.; Meuleman, N.; Bron, D.; Toungouz, M.; Lagneaux, L. The source of human mesenchymal stromal cells influences their TLR profile as well as their functional properties. Cell Immunol. 2011, 270, 207–216. [Google Scholar] [CrossRef]
- Van den Berk, L.C.; Jansen, B.J.; Siebers-Vermeulen, K.G.; Netea, M.G.; Latuhihin, T.; Bergevoet, S.; Raymakers, R.A.; Kogler, G.; Figdor, C.C.; Adema, G.J.; et al. Toll-like receptor triggering in cord blood mesenchymal stem cells. J. Cell Mol. Med. 2009, 13, 3415–3426. [Google Scholar] [CrossRef]
- Lombardo, E.; DelaRosa, O.; Mancheno-Corvo, P.; Menta, R.; Ramirez, C.; Buscher, D. Toll-like receptor-mediated signaling in human adipose-derived stem cells: Implications for immunogenicity and immunosuppressive potential. Tissue Eng. Part. A 2009, 15, 1579–1589. [Google Scholar] [CrossRef]
- Raicevic, G.; Najar, M.; Pieters, K.; de Bruyn, C.; Meuleman, N.; Bron, D.; Toungouz, M.; Lagneaux, L. Inflammation and Toll-like receptor ligation differentially affect the osteogenic potential of human mesenchymal stromal cells depending on their tissue origin. Tissue Eng. Part. A 2012, 18, 1410–1418. [Google Scholar] [CrossRef]
- Bunnell, B.A.; Betancourt, A.M.; Sullivan, D.E. New concepts on the immune modulation mediated by mesenchymal stem cells. Stem Cell Res. Ther. 2010, 1, 34. [Google Scholar] [CrossRef]
- Lee, S.C.; Jeong, H.J.; Lee, S.K.; Kim, S.J. Lipopolysaccharide preconditioning of adipose-derived stem cells improves liver-regenerating activity of the secretome. Stem Cell Res. Ther. 2015, 6, 75. [Google Scholar] [CrossRef] [Green Version]
- Mancheno-Corvo, P.; Menta, R.; del Rio, B.; Franquesa, M.; Ramirez, C.; Hoogduijn, M.J.; DelaRosa, O.; Dalemans, W.; Lombardo, E. T Lymphocyte Prestimulation Impairs in a Time-Dependent Manner the Capacity of Adipose Mesenchymal Stem Cells to Inhibit Proliferation: Role of Interferon gamma, Poly I:C, and Tryptophan Metabolism in Restoring Adipose Mesenchymal Stem Cell Inhibitory Effect. Stem Cells Dev. 2015, 24, 2158–2170. [Google Scholar]
- Opitz, C.A.; Litzenburger, U.M.; Lutz, C.; Lanz, T.V.; Tritschler, I.; Koppel, A.; Tolosa, E.; Hoberg, M.; Anderl, J.; Aicher, W.K.; et al. Toll-like receptor engagement enhances the immunosuppressive properties of human bone marrow-derived mesenchymal stem cells by inducing indoleamine-2,3-dioxygenase-1 via interferon-beta and protein kinase R. Stem Cells 2009, 27, 909–919. [Google Scholar] [CrossRef]
- Serejo, T.R.T.; Silva-Carvalho, A.E.; Braga, L.; Neves, F.A.R.; Pereira, R.W.; Carvalho, J.L.; Saldanha-Araujo, F. Assessment of the Immunosuppressive Potential of INF-gamma Licensed Adipose Mesenchymal Stem Cells, Their Secretome and Extracellular Vesicles. Cells 2019, 8, 22. [Google Scholar] [CrossRef]
- Gu, W.; Song, L.; Li, X.M.; Wang, D.; Guo, X.J.; Xu, W.G. Mesenchymal stem cells alleviate airway inflammation and emphysema in COPD through down-regulation of cyclooxygenase-2 via p38 and ERK MAPK pathways. Sci. Rep. 2015, 5, 8733. [Google Scholar] [CrossRef]
- Liu, J.; Zhu, P.; Song, P.; Xiong, W.; Chen, H.; Peng, W.; Wang, S.; Li, S.; Fu, Z.; Wang, Y.; et al. Pretreatment of Adipose Derived Stem Cells with Curcumin Facilitates Myocardial Recovery via Antiapoptosis and Angiogenesis. Stem Cells Int. 2015, 2015, 638153. [Google Scholar] [CrossRef]
- Kang, H.; Kim, K.H.; Lim, J.; Kim, Y.S.; Heo, J.; Choi, J.; Jeong, J.; Kim, Y.; Kim, S.W.; Oh, Y.M.; et al. The Therapeutic Effects of Human Mesenchymal Stem Cells Primed with Sphingosine-1 Phosphate on Pulmonary Artery Hypertension. Stem Cells Dev. 2015, 24, 1658–1671. [Google Scholar] [CrossRef] [Green Version]
- Marofi, F.; Vahedi, G.; Biglari, A.; Esmaeilzadeh, A.; Athari, S.S. Mesenchymal Stromal/Stem Cells: A New Era in the Cell-Based Targeted Gene Therapy of Cancer. Front. Immunol. 2017, 8, 1770. [Google Scholar] [CrossRef] [Green Version]
- Hossain, M.M.; Murali, M.R.; Kamarul, T. Genetically modified mesenchymal stem/stromal cells transfected with adiponectin gene can stably secrete adiponectin. Life Sci. 2017, 182, 50–56. [Google Scholar] [CrossRef]
- Altaner, C.; Altanerova, V.; Cihova, M.; Ondicova, K.; Rychly, B.; Baciak, L.; Mravec, B. Complete regression of glioblastoma by mesenchymal stem cells mediated prodrug gene therapy simulating clinical therapeutic scenario. Int. J. Cancer 2014, 134, 1458–1465. [Google Scholar] [CrossRef]
- Asahara, T.; Kalka, C.; Isner, J.M. Stem cell therapy and gene transfer for regeneration. Gene Ther. 2000, 7, 451–457. [Google Scholar] [CrossRef] [Green Version]
- Han, S.M.; Han, S.H.; Coh, Y.R.; Jang, G.; Chan Ra, J.; Kang, S.K.; Lee, H.W.; Youn, H.Y. Enhanced proliferation and differentiation of Oct4- and Sox2-overexpressing human adipose tissue mesenchymal stem cells. Exp. Mol. Med. 2014, 46, e101. [Google Scholar] [CrossRef]
- Taha, M.F.; Javeri, A.; Rohban, S.; Mowla, S.J. Upregulation of pluripotency markers in adipose tissue-derived stem cells by miR-302 and leukemia inhibitory factor. Biomed. Res. Int. 2014, 2014, 941486. [Google Scholar] [CrossRef]
- Kim, J.Y.; Shin, K.K.; Lee, A.L.; Kim, Y.S.; Park, H.J.; Park, Y.K.; Bae, Y.C.; Jung, J.S. MicroRNA-302 induces proliferation and inhibits oxidant-induced cell death in human adipose tissue-derived mesenchymal stem cells. Cell Death Dis. 2014, 5, e1385. [Google Scholar] [CrossRef]
- Schonitzer, V.; Wirtz, R.; Ulrich, V.; Berger, T.; Karl, A.; Mutschler, W.; Schieker, M.; Bocker, W. Sox2 is a potent inhibitor of osteogenic and adipogenic differentiation in human mesenchymal stem cells. Cell Reprogram 2014, 16, 355–365. [Google Scholar] [CrossRef]
- Baldari, S.; Di Rocco, G.; Trivisonno, A.; Samengo, D.; Pani, G.; Toietta, G. Promotion of Survival and Engraftment of Transplanted Adipose Tissue-Derived Stromal and Vascular Cells by Overexpression of Manganese Superoxide Dismutase. Int. J. Mol. Sci. 2016, 17, 1082. [Google Scholar] [CrossRef]
- Sen, S.; Domingues, C.C.; Rouphael, C.; Chou, C.; Kim, C.; Yadava, N. Genetic modification of human mesenchymal stem cells helps to reduce adiposity and improve glucose tolerance in an obese diabetic mouse model. Stem Cell Res. Ther. 2015, 6, 242. [Google Scholar] [CrossRef]
- Janowski, M. Functional diversity of SDF-1 splicing variants. Cell Adh. Migr. 2009, 3, 243–249. [Google Scholar] [CrossRef] [Green Version]
- Cho, H.H.; Kyoung, K.M.; Seo, M.J.; Kim, Y.J.; Bae, Y.C.; Jung, J.S. Overexpression of CXCR4 increases migration and proliferation of human adipose tissue stromal cells. Stem Cells Dev. 2006, 15, 853–864. [Google Scholar] [CrossRef]
- Kim, M.; Kim, D.I.; Kim, E.K.; Kim, C.W. CXCR4 Overexpression in Human Adipose Tissue-Derived Stem Cells Improves Homing and Engraftment in an Animal Limb Ischemia Model. Cell Transplant. 2017, 26, 191–204. [Google Scholar] [CrossRef] [Green Version]
- Kim, S.W.; Lee, D.W.; Yu, L.H.; Zhang, H.Z.; Kim, C.E.; Kim, J.M.; Park, T.H.; Cha, K.S.; Seo, S.Y.; Roh, M.S.; et al. Mesenchymal stem cells overexpressing GCP-2 improve heart function through enhanced angiogenic properties in a myocardial infarction model. Cardiovasc. Res. 2012, 95, 495–506. [Google Scholar] [CrossRef]
- Min, C.K.; Kim, B.G.; Park, G.; Cho, B.; Oh, I.H. IL-10-transduced bone marrow mesenchymal stem cells can attenuate the severity of acute graft-versus-host disease after experimental allogeneic stem cell transplantation. Bone Marrow Transplant. 2007, 39, 637–645. [Google Scholar] [CrossRef] [Green Version]
- Bian, L.; Guo, Z.K.; Wang, H.X.; Wang, J.S.; Wang, H.; Li, Q.F.; Yang, Y.F.; Xiao, F.J.; Wu, C.T.; Wang, L.S. In vitro and in vivo immunosuppressive characteristics of hepatocyte growth factor-modified murine mesenchymal stem cells. In Vivo 2009, 23, 21–27. [Google Scholar]
- Kim, D.S.; Jang, I.K.; Lee, M.W.; Ko, Y.J.; Lee, D.H.; Lee, J.W.; Sung, K.W.; Koo, H.H.; Yoo, K.H. Enhanced Immunosuppressive Properties of Human Mesenchymal Stem Cells Primed by Interferon-gamma. EBioMedicine 2018, 28, 261–273. [Google Scholar] [CrossRef]
- Qi, H.; Chen, G.; Huang, Y.; Si, Z.; Li, J. Foxp3-modified bone marrow mesenchymal stem cells promotes liver allograft tolerance through the generation of regulatory T cells in rats. J. Transl. Med. 2015, 13, 274. [Google Scholar] [CrossRef]
- Payne, N.L.; Dantanarayana, A.; Sun, G.; Moussa, L.; Caine, S.; McDonald, C.; Herszfeld, D.; Bernard, C.C.; Siatskas, C. Early intervention with gene-modified mesenchymal stem cells overexpressing interleukin-4 enhances anti-inflammatory responses and functional recovery in experimental autoimmune demyelination. Cell Adh. Migr. 2012, 6, 179–189. [Google Scholar] [CrossRef] [Green Version]
- Choi, E.W.; Shin, I.S.; Song, J.W.; Lee, M.; Yun, T.W.; Yang, J.; Choi, K.S.; Kim, S.J. Effects of Transplantation of CTLA4Ig-Overexpressing Adipose Tissue-Derived Mesenchymal Stem Cells in Mice With Sustained Severe Rheumatoid Arthritis. Cell Transplant. 2016, 25, 243–259. [Google Scholar] [CrossRef]
- Park, M.J.; Lee, S.H.; Moon, S.J.; Lee, J.A.; Lee, E.J.; Kim, E.K.; Park, J.S.; Lee, J.; Min, J.K.; Kim, S.J.; et al. Overexpression of soluble RAGE in mesenchymal stem cells enhances their immunoregulatory potential for cellular therapy in autoimmune arthritis. Sci. Rep. 2016, 6, 35933. [Google Scholar] [CrossRef]
- Martinez-Gonzalez, I.; Roca, O.; Masclans, J.R.; Moreno, R.; Salcedo, M.T.; Baekelandt, V.; Cruz, M.J.; Rello, J.; Aran, J.M. Human mesenchymal stem cells overexpressing the IL-33 antagonist soluble IL-1 receptor-like-1 attenuate endotoxin-induced acute lung injury. Am. J. Respir. Cell Mol. Biol. 2013, 49, 552–562. [Google Scholar] [CrossRef]
- Zhao, N.; Li, H.; Yan, Y.; Jiang, R.; He, X. Mesenchymal stem cells overexpressing IL-35 effectively inhibit CD4(+) T cell function. Cell Immunol. 2017, 312, 61–66. [Google Scholar] [CrossRef]
- Boulaiz, H.; Marchal, J.A.; Prados, J.; Melguizo, C.; Aranega, A. Non-viral and viral vectors for gene therapy. Cell Mol. Biol. 2005, 51, 3–22. [Google Scholar]
- Arboleda, D.; Forostyak, S.; Jendelova, P.; Marekova, D.; Amemori, T.; Pivonkova, H.; Masinova, K.; Sykova, E. Transplantation of predifferentiated adipose-derived stromal cells for the treatment of spinal cord injury. Cell Mol. Neurobiol. 2011, 31, 1113–1122. [Google Scholar] [CrossRef]
- Phinney, D.G.; Isakova, I. Plasticity and therapeutic potential of mesenchymal stem cells in the nervous system. Curr. Pharm. Des. 2005, 11, 1255–1265. [Google Scholar] [CrossRef]
- Barzilay, R.; Melamed, E.; Offen, D. Introducing transcription factors to multipotent mesenchymal stem cells: Making transdifferentiation possible. Stem Cells 2009, 27, 2509–2515. [Google Scholar] [CrossRef]
- Neshati, V.; Mollazadeh, S.; Fazly Bazzaz, B.S.; de Vries, A.A.; Mojarrad, M.; Naderi-Meshkin, H.; Neshati, Z.; Kerachian, M.A. Cardiomyogenic differentiation of human adipose-derived mesenchymal stem cells transduced with Tbx20-encoding lentiviral vectors. J. Cell Biochem. 2018, 119, 6146–6153. [Google Scholar] [CrossRef]
- Tang, L.; Lu, X.; Zhu, R.; Qian, T.; Tao, Y.; Li, K.; Zheng, J.; Zhao, P.; Li, S.; Wang, X.; et al. Adipose-Derived Stem Cells Expressing the Neurogenin-2 Promote Functional Recovery After Spinal Cord Injury in Rat. Cell Mol. Neurobiol. 2016, 36, 657–667. [Google Scholar] [CrossRef]
- Goudenege, S.; Pisani, D.F.; Wdziekonski, B.; di Santo, J.P.; Bagnis, C.; Dani, C.; Dechesne, C.A. Enhancement of myogenic and muscle repair capacities of human adipose-derived stem cells with forced expression of MyoD. Mol. Ther. 2009, 17, 1064–1072. [Google Scholar] [CrossRef]
- Cheng, F.; Zhang, Y.; Wang, Y.; Jiang, Q.; Zhao, C.J.; Deng, J.; Chen, X.; Yao, Y.; Xia, Z.; Cheng, L.; et al. Conversion of human adipose-derived stem cells into functional and expandable endothelial-like cells for cell-based therapies. Stem Cell Res. Ther. 2018, 9, 350. [Google Scholar] [CrossRef]
- Jones, D.L.; Wagers, A.J. No place like home: Anatomy and function of the stem cell niche. Nat. Rev. Mol. Cell Bio. 2008, 9, 11–21. [Google Scholar] [CrossRef]
- Voog, J.; Jones, D.L. Stem Cells and the Niche: A Dynamic Duo. Cell Stem Cell 2010, 6, 103–115. [Google Scholar] [CrossRef] [Green Version]
- Li, L.; Clevers, H. Coexistence of quiescent and active adult stem cells in mammals. Science 2010, 327, 542–545. [Google Scholar] [CrossRef]
- Ema, H.; Suda, T. Two anatomically distinct niches regulate stem cell activity. Blood 2012, 120, 2174–2181. [Google Scholar] [CrossRef] [Green Version]
- Amelian, A.; Wasilewska, K.; Megias, D.; Winnicka, K. Application of standard cell cultures and 3D in vitro tissue models as an effective tool in drug design and development. Pharmacol. Rep. 2017, 69, 861–870. [Google Scholar] [CrossRef]
- Duval, K.; Grover, H.; Han, L.H.; Mou, Y.; Pegoraro, A.F.; Fredberg, J.; Chen, Z. Modeling Physiological Events in 2D vs. 3D Cell Culture. Physiology 2017, 32, 266–277. [Google Scholar] [CrossRef]
- Edmondson, R.; Broglie, J.J.; Adcock, A.F.; Yang, L.J. Three-Dimensional Cell Culture Systems and Their Applications in Drug Discovery and Cell-Based Biosensors. Assay Drug Dev. Techn. 2014, 12, 207–218. [Google Scholar] [CrossRef] [Green Version]
- Murphy, K.C.; Hung, B.; Browne-Bourne, S.; Zhou, D.J.; Yeung, J.; Genetos, D.C.; Leach, J.K. Measurement of oxygen tension within mesenchymal stem cell spheroids. J. R. Soc. Interface 2017, 14, 20160851. [Google Scholar] [CrossRef]
- Griffith, L.G.; Swartz, M.A. Capturing complex 3D tissue physiology in vitro. Nat. Rev. Mol. Cell Biol. 2006, 7, 211–224. [Google Scholar] [CrossRef]
- Frith, J.E.; Thomson, B.; Genever, P.G. Dynamic three-dimensional culture methods enhance mesenchymal stem cell properties and increase therapeutic potential. Tissue Eng. Part. C Methods 2010, 16, 735–749. [Google Scholar] [CrossRef]
- Bartosh, T.J.; Ylostalo, J.H.; Mohammadipoor, A.; Bazhanov, N.; Coble, K.; Claypool, K.; Lee, R.H.; Choi, H.; Prockop, D.J. Aggregation of human mesenchymal stromal cells (MSCs) into 3D spheroids enhances their antiinflammatory properties. P. Natl. Acad. Sci. USA 2010, 107, 13724–13729. [Google Scholar] [CrossRef] [Green Version]
- Wang, W.; Itaka, K.; Ohba, S.; Nishiyama, N.; Chung, U.I.; Yamasaki, Y.; Kataoka, K. 3D spheroid culture system on micropatterned substrates for improved differentiation efficiency of multipotent mesenchymal stem cells. Biomaterials 2009, 30, 2705–2715. [Google Scholar] [CrossRef]
- Cheng, N.C.; Wang, S.; Young, T.H. The influence of spheroid formation of human adipose-derived stem cells on chitosan films on stemness and differentiation capabilities. Biomaterials 2012, 33, 1748–1758. [Google Scholar] [CrossRef]
- Amos, P.J.; Kapur, S.K.; Stapor, P.C.; Shang, H.; Bekiranov, S.; Khurgel, M.; Rodeheaver, G.T.; Peirce, S.M.; Katz, A.J. Human adipose-derived stromal cells accelerate diabetic wound healing: Impact of cell formulation and delivery. Tissue Eng. Part. A 2010, 16, 1595–1606. [Google Scholar] [CrossRef]
- Cheng, N.C.; Chen, S.Y.; Li, J.R.; Young, T.H. Short-term spheroid formation enhances the regenerative capacity of adipose-derived stem cells by promoting stemness, angiogenesis, and chemotaxis. Stem Cells Transl. Med. 2013, 2, 584–594. [Google Scholar] [CrossRef]
- Potapova, I.A.; Brink, P.R.; Cohen, I.S.; Doronin, S.V. Culturing of human mesenchymal stem cells as three-dimensional aggregates induces functional expression of CXCR4 that regulates adhesion to endothelial cells. J. Biological. Chemistry 2008, 283, 13100–13107. [Google Scholar] [CrossRef]
- Zhang, S.; Liu, P.; Chen, L.; Wang, Y.; Wang, Z.; Zhang, B. The effects of spheroid formation of adipose-derived stem cells in a microgravity bioreactor on stemness properties and therapeutic potential. Biomaterials 2015, 41, 15–25. [Google Scholar] [CrossRef]
- Tae, J.Y.; Lee, S.I.; Ko, Y.; Park, J.B. The use of adipose-derived stem cells for the fabrication of three-dimensional spheroids for the osteogenic differentiation. Biomed. Res. 2017, 28, 7098–7103. [Google Scholar]
- Cho, R.J.; Kim, Y.S.; Kim, J.Y.; Oh, Y.M. Human adipose-derived mesenchymal stem cell spheroids improve recovery in a mouse model of elastase-induced emphysema. BMB Rep. 2017, 50, 79–84. [Google Scholar] [CrossRef]
- Park, I.S.; Chung, P.S.; Ahn, J.C. Adipose-derived stem cell spheroid treated with low-level light irradiation accelerates spontaneous angiogenesis in mouse model of hindlimb ischemia. Cytotherapy 2017, 19, 1070–1078. [Google Scholar] [CrossRef]
- Park, I.S.; Chung, P.S.; Ahn, J.C. Enhancement of Ischemic Wound Healing by Spheroid Grafting of Human Adipose-Derived Stem Cells Treated with Low-Level Light Irradiation. PLoS ONE 2015, 10, e0122776. [Google Scholar] [CrossRef]
- Xu, Y.; Shi, T.P.; Xu, A.X.; Zhang, L. 3D spheroid culture enhances survival and therapeutic capacities of MSCs injected into ischemic kidney. J. Cell Mol. Med. 2016, 20, 1203–1213. [Google Scholar] [CrossRef]
- Yoon, H.H.; Bhang, S.H.; Shin, J.Y.; Shin, J.; Kim, B.S. Enhanced Cartilage Formation via Three-Dimensional Cell Engineering of Human Adipose-Derived Stem Cells. Tissue Eng. Pt. A 2012, 18, 1949–1956. [Google Scholar] [CrossRef] [Green Version]
- Lee, J.H.; Han, Y.S.; Lee, S.H. Long-Duration Three-Dimensional Spheroid Culture Promotes Angiogenic Activities of Adipose-Derived Mesenchymal Stem Cells. Biomol Ther. 2016, 24, 260–267. [Google Scholar] [CrossRef] [Green Version]
- Giaccia, A.J.; Simon, M.C.; Johnson, R. The biology of hypoxia: The role of oxygen sensing in development, normal function, and disease. Genes Dev. 2004, 18, 2183–2194. [Google Scholar] [CrossRef]
- Mohyeldin, A.; Garzon-Muvdi, T.; Quinones-Hinojosa, A. Oxygen in stem cell biology: A critical component of the stem cell niche. Cell Stem Cell 2010, 7, 150–161. [Google Scholar] [CrossRef]
- Panchision, D.M. The role of oxygen in regulating neural stem cells in development and disease. J. Cell Physiol. 2009, 220, 562–568. [Google Scholar] [CrossRef]
- Eliasson, P.; Jonsson, J.I. The Hematopoietic Stem Cell Niche: Low in Oxygen but a Nice Place to be. J. Cellular Physiol. 2010, 222, 17–22. [Google Scholar] [CrossRef]
- Goossens, G.H.; Blaak, E.E. Adipose tissue oxygen tension: implications for chronic metabolic and inflammatory diseases. Curr. Opin. Clin. Nutr. Metab. Care 2012, 15, 539–546. [Google Scholar] [CrossRef]
- Choi, J.R.; Pingguan-Murphy, B.; Wan Abas, W.A.; Noor Azmi, M.A.; Omar, S.Z.; Chua, K.H.; Wan Safwani, W.K. Impact of low oxygen tension on stemness, proliferation and differentiation potential of human adipose-derived stem cells. Biochem. Biophys Res. Commun. 2014, 448, 218–224. [Google Scholar] [CrossRef]
- Yamamoto, Y.; Fujita, M.; Tanaka, Y.; Kojima, I.; Kanatani, Y.; Ishihara, M.; Tachibana, S. Low oxygen tension enhances proliferation and maintains stemness of adipose tissue-derived stromal cells. Biores Open Access 2013, 2, 199–205. [Google Scholar] [CrossRef]
- Kakudo, N.; Morimoto, N.; Ogawa, T.; Taketani, S.; Kusumoto, K. Hypoxia Enhances Proliferation of Human Adipose-Derived Stem Cells via HIF-1 alpha Activation. PLoS ONE 2015, 10, e0139890. [Google Scholar] [CrossRef]
- Weijers, E.M.; Van Den Broek, L.J.; Waaijman, T.; Van Hinsbergh, V.W.; Gibbs, S.; Koolwijk, P. The influence of hypoxia and fibrinogen variants on the expansion and differentiation of adipose tissue-derived mesenchymal stem cells. Tissue Eng. Part. A 2011, 17, 2675–2685. [Google Scholar] [CrossRef]
- Feng, Y.; Zhu, M.; Dangelmajer, S.; Lee, Y.M.; Wijesekera, O.; Castellanos, C.X.; Denduluri, A.; Chaichana, K.L.; Li, Q.; Zhang, H.; et al. Hypoxia-cultured human adipose-derived mesenchymal stem cells are non-oncogenic and have enhanced viability, motility, and tropism to brain cancer. Cell Death Dis 2015, 6, e1567. [Google Scholar] [CrossRef]
- Choi, J.R.; Yong, K.W.; Wan Safwani, W.K.Z. Effect of hypoxia on human adipose-derived mesenchymal stem cells and its potential clinical applications. Cell Mol. Life Sci. 2017, 74, 2587–2600. [Google Scholar] [CrossRef]
- Wan Safwani, W.K.Z.; Choi, J.R.; Yong, K.W.; Ting, I.; Mat Adenan, N.A.; Pingguan-Murphy, B. Hypoxia enhances the viability, growth and chondrogenic potential of cryopreserved human adipose-derived stem cells. Cryobiology 2017, 75, 91–99. [Google Scholar] [CrossRef]
- Merceron, C.; Vinatier, C.; Portron, S.; Masson, M.; Amiaud, J.; Guigand, L.; Cherel, Y.; Weiss, P.; Guicheux, J. Differential effects of hypoxia on osteochondrogenic potential of human adipose-derived stem cells. Am. J. Physiol-Cell Ph. 2010, 298, 355–364. [Google Scholar] [CrossRef]
- Malladi, P.; Xu, Y.; Chiou, M.; Giaccia, A.J.; Longaker, M.T. Effect of reduced oxygen tension on chondrogenesis and osteogenesis in adipose-derived mesenchymal cells. Am. J. Physiol-Cell Ph. 2006, 290, 1139–1145. [Google Scholar] [CrossRef]
- Xu, Y.; Malladi, P.; Chiou, M.; Bekerman, E.; Giaccia, A.J.; Longaker, M.T. In vitro expansion of adipose-derived adult stromal cells in hypoxia enhances early chondrogenesis. Tissue Eng. 2007, 13, 2981–2993. [Google Scholar] [CrossRef]
- Valorani, M.G.; Montelatici, E.; Germani, A.; Biddle, A.; D’Alessandro, D.; Strollo, R.; Patrizi, M.P.; Lazzari, L.; Nye, E.; Otto, W.R.; et al. Pre-culturing human adipose tissue mesenchymal stem cells under hypoxia increases their adipogenic and osteogenic differentiation potentials. Cell Proliferat. 2012, 45, 225–238. [Google Scholar] [CrossRef]
- Roemeling-van Rhijn, M.; Mensah, F.K.; Korevaar, S.S.; Leijs, M.J.; van Osch, G.J.; Ijzermans, J.N.; Betjes, M.G.; Baan, C.C.; Weimar, W.; Hoogduijn, M.J. Effects of Hypoxia on the Immunomodulatory Properties of Adipose Tissue-Derived Mesenchymal Stem cells. Front. Immunol. 2013, 4, 203. [Google Scholar] [CrossRef] [Green Version]
- Efimenko, A.; Starostina, E.; Kalinina, N.; Stolzing, A. Angiogenic properties of aged adipose derived mesenchymal stem cells after hypoxic conditioning. J. Transl. Med. 2011, 9, 10. [Google Scholar] [CrossRef]
- Liu, L.; Gao, J.; Yuan, Y.; Chang, Q.; Liao, Y.; Lu, F. Hypoxia preconditioned human adipose derived mesenchymal stem cells enhance angiogenic potential via secretion of increased VEGF and bFGF. Cell Biol. Int. 2013, 37, 551–560. [Google Scholar] [CrossRef]
- Kim, J.H.; Park, S.H.; Park, S.G.; Choi, J.S.; Xia, Y.; Sung, J.H. The pivotal role of reactive oxygen species generation in the hypoxia-induced stimulation of adipose-derived stem cells. Stem. Cells Dev. 2011, 20, 1753–1761. [Google Scholar] [CrossRef]
- Hu, X.; Yu, S.P.; Fraser, J.L.; Lu, Z.; Ogle, M.E.; Wang, J.A.; Wei, L. Transplantation of hypoxia-preconditioned mesenchymal stem cells improves infarcted heart function via enhanced survival of implanted cells and angiogenesis. J. Thorac. Cardiovasc Surg. 2008, 135, 799–808. [Google Scholar] [CrossRef] [Green Version]
- Hollenbeck, S.T.; Senghaas, A.; Komatsu, I.; Zhang, Y.; Erdmann, D.; Klitzman, B. Tissue engraftment of hypoxic-preconditioned adipose-derived stem cells improves flap viability. Wound Repair Regen. 2012, 20, 872–878. [Google Scholar] [CrossRef] [Green Version]
- Rosova, I.; Dao, M.; Capoccia, B.; Link, D.; Nolta, J.A. Hypoxic preconditioning results in increased motility and improved therapeutic potential of human mesenchymal stem cells. Stem Cells 2008, 26, 2173–2182. [Google Scholar] [CrossRef]
- Leroux, L.; Descamps, B.; Tojais, N.F.; Seguy, B.; Oses, P.; Moreau, C.; Daret, D.; Ivanovic, Z.; Boiron, J.M.; Lamaziere, J.M.D.; et al. Hypoxia Preconditioned Mesenchymal Stem Cells Improve Vascular and Skeletal Muscle Fiber Regeneration After Ischemia Through a Wnt4-dependent Pathway. Molecular Therapy 2010, 18, 1545–1552. [Google Scholar] [CrossRef]
- Schive, S.W.; Mirlashari, M.R.; Hasvold, G.; Wang, M.; Josefsen, D.; Gullestad, H.P.; Korsgren, O.; Foss, A.; Kvalheim, G.; Scholz, H. Human Adipose-Derived Mesenchymal Stem Cells Respond to Short-Term Hypoxia by Secreting Factors Beneficial for Human Islets In Vitro and Potentiate Antidiabetic Effect In Vivo. Cell Med. 2017, 9, 103–116. [Google Scholar] [CrossRef]
- Lee, S.C.; Jeong, H.J.; Lee, S.K.; Kim, S.J. Hypoxic Conditioned Medium From Human Adipose-Derived Stem Cells Promotes Mouse Liver Regeneration Through JAK/STAT3 Signaling. Stem Cell Transl. Med. 2016, 5, 816–825. [Google Scholar] [CrossRef]
- Shin, H.S.; Lee, S.; Kim, Y.M.; Lim, J.Y. Hypoxia-Activated Adipose Mesenchymal Stem Cells Prevents Irradiation-Induced Salivary Hypofunction by Enhanced Paracrine Effect Through Fibroblast Growth Factor 10. Stem Cells 2018, 36, 1020–1032. [Google Scholar] [CrossRef] [Green Version]
- Hosogai, N.; Fukuhara, A.; Oshima, K.; Miyata, Y.; Tanaka, S.; Segawa, K.; Furukawa, S.; Tochino, Y.; Komuro, R.; Matsuda, M.; et al. Adipose tissue hypoxia in obesity and its impact on adipocytokine dysregulation. Diabetes 2007, 56, 901–911. [Google Scholar] [CrossRef]
- Oikonomopoulos, A.; van Deen, W.K.; Manansala, A.R.; Lacey, P.N.; Tomakili, T.A.; Ziman, A.; Hommes, D.W. Optimization of human mesenchymal stem cell manufacturing: The effects of animal/xeno-free media. Sci. Rep. 2015, 5, 16570. [Google Scholar] [CrossRef]
- Gstraunthaler, G.; Lindl, T.; van der Valk, J. A plea to reduce or replace fetal bovine serum in cell culture media. Cytotechnology 2013, 65, 791–793. [Google Scholar] [CrossRef] [Green Version]
- Van der Valk, J.; Brunner, D.; De Smet, K.; Fex Svenningsen, A.; Honegger, P.; Knudsen, L.E.; Lindl, T.; Noraberg, J.; Price, A.; Scarino, M.L.; et al. Optimization of chemically defined cell culture media--replacing fetal bovine serum in mammalian in vitro methods. Toxicol. In Vitro 2010, 24, 1053–1063. [Google Scholar] [CrossRef]
- Lee, M.S.; Youn, C.; Kim, J.H.; Park, B.J.; Ahn, J.; Hong, S.; Kim, Y.D.; Shin, Y.K.; Park, S.G. Enhanced Cell Growth of Adipocyte-Derived Mesenchymal Stem Cells Using Chemically-Defined Serum-Free Media. Int. J. Mol. Sci. 2017, 18, 1779. [Google Scholar] [CrossRef]
- Tatullo, M.; Marrelli, M.; Falisi, G.; Rastelli, C.; Palmieri, F.; Gargari, M.; Zavan, B.; Paduano, F.; Benagiano, V. Mechanical influence of tissue culture plates and extracellular matrix on mesenchymal stem cell behavior: A topical review. Int. J. Immunopathol. Pharmacol. 2016, 29, 3–8. [Google Scholar] [CrossRef]
- Yin, J.Q.; Zhu, J.; Ankrum, J.A. Manufacturing of primed mesenchymal stromal cells for therapy. Nat. Biomed. Eng. 2019, 3, 90–104. [Google Scholar] [CrossRef]
- Banks, J.M.; Mozdzen, L.C.; Harley, B.A.; Bailey, R.C. The combined effects of matrix stiffness and growth factor immobilization on the bioactivity and differentiation capabilities of adipose-derived stem cells. Biomaterials 2014, 35, 8951–8959. [Google Scholar] [CrossRef] [Green Version]
- Wen, J.H.; Vincent, L.G.; Fuhrmann, A.; Choi, Y.S.; Hribar, K.C.; Taylor-Weiner, H.; Chen, S.; Engler, A.J. Interplay of matrix stiffness and protein tethering in stem cell differentiation. Nat. Mater. 2014, 13, 979–987. [Google Scholar] [CrossRef] [Green Version]
- Lopez-Verrilli, M.A.; Court, F.A. Exosomes: Mediators of communication in eukaryotes. Biol. Res. 2013, 46, 5–11. [Google Scholar] [CrossRef]
- Biancone, L.; Bruno, S.; Deregibus, M.C.; Tetta, C.; Camussi, G. Therapeutic potential of mesenchymal stem cell-derived microvesicles. Nephrol. Dial. Transplant. 2012, 27, 3037–3042. [Google Scholar] [CrossRef]
- Camussi, G.; Deregibus, M.C.; Cantaluppi, V. Role of stem-cell-derived microvesicles in the paracrine action of stem cells. Biochem. Soc. Trans. 2013, 41, 283–287. [Google Scholar] [CrossRef]
- Ratajczak, M.Z.; Kucia, M.; Jadczyk, T.; Greco, N.J.; Wojakowski, W.; Tendera, M.; Ratajczak, J. Pivotal role of paracrine effects in stem cell therapies in regenerative medicine: Can we translate stem cell-secreted paracrine factors and microvesicles into better therapeutic strategies? Leukemia 2012, 26, 1166–1173. [Google Scholar] [CrossRef]
- Marconi, S.; Bonaconsa, M.; Scambi, I.; Squintani, G.M.; Rui, W.; Turano, E.; Ungaro, D.; D’Agostino, S.; Barbieri, F.; Angiari, S.; et al. Systemic treatment with adipose-derived mesenchymal stem cells ameliorates clinical and pathological features in the amyotrophic lateral sclerosis murine model. Neuroscience 2013, 248, 333–343. [Google Scholar] [CrossRef]
- Marconi, S.; Castiglione, G.; Turano, E.; Bissolotti, G.; Angiari, S.; Farinazzo, A.; Constantin, G.; Bedogni, G.; Bedogni, A.; Bonetti, B. Human adipose-derived mesenchymal stem cells systemically injected promote peripheral nerve regeneration in the mouse model of sciatic crush. Tissue Eng. Part. A 2012, 18, 1264–1272. [Google Scholar] [CrossRef]
- Phinney, D.G.; Hill, K.; Michelson, C.; DuTreil, M.; Hughes, C.; Humphries, S.; Wilkinson, R.; Baddoo, M.; Bayly, E. Biological activities encoded by the murine mesenchymal stem cell transcriptome provide a basis for their developmental potential and broad therapeutic efficacy. Stem Cells 2006, 24, 186–198. [Google Scholar] [CrossRef]
- Cho, K.A.; Park, M.; Kim, Y.H.; Woo, S.Y.; Ryu, K.H. RNA sequencing reveals a transcriptomic portrait of human mesenchymal stem cells from bone marrow, adipose tissue, and palatine tonsils. Sci. Rep. 2017, 7, 17114. [Google Scholar] [CrossRef]
- Jansen, B.J.H.; Gilissen, C.; Roelofs, H.; Schaap-Oziemlak, A.; Veltman, J.A.; Raymakers, R.A.P.; Jansen, J.H.; Kogler, G.; Figdor, C.G.; Torensma, R.; et al. Functional Differences Between Mesenchymal Stem Cell Populations Are Reflected by Their Transcriptome. Stem Cells Dev. 2010, 19, 481–489. [Google Scholar] [CrossRef]
- Tkach, M.; Thery, C. Communication by Extracellular Vesicles: Where We Are and Where We Need to Go. Cell 2016, 164, 1226–1232. [Google Scholar] [CrossRef] [Green Version]
- Bruno, S.; Deregibus, M.C.; Camussi, G. The secretome of mesenchymal stromal cells: Role of extracellular vesicles in immunomodulation. Immunol. Lett. 2015, 168, 154–158. [Google Scholar] [CrossRef]
- Hwu, P.; Du, M.X.; Lapointe, R.; Do, M.; Taylor, M.W.; Young, H.A. Indoleamine 2,3-dioxygenase production by human dendritic cells results in the inhibition of T cell proliferation. J. Immunol. 2000, 164, 3596–3599. [Google Scholar] [CrossRef]
- Spaggiari, G.M.; Capobianco, A.; Becchetti, S.; Mingari, M.C.; Moretta, L. Mesenchymal stem cell-natural killer cell interactions: Evidence that activated NK cells are capable of killing MSCs, whereas MSCs can inhibit IL-2-induced NK-cell proliferation. Blood 2006, 107, 1484–1490. [Google Scholar] [CrossRef]
- Sato, K.; Ozaki, K.; Oh, I.; Meguro, A.; Hatanaka, K.; Nagai, T.; Muroi, K.; Ozawa, K. Nitric oxide plays a critical role in suppression of T-cell proliferation by mesenchymal stem cells. Blood 2007, 109, 228–234. [Google Scholar] [CrossRef]
- Aggarwal, S.; Pittenger, M.F. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood 2005, 105, 1815–1822. [Google Scholar] [CrossRef] [Green Version]
- Vizoso, F.J.; Eiro, N.; Cid, S.; Schneider, J.; Perez-Fernandez, R. Mesenchymal Stem Cell Secretome: Toward Cell-Free Therapeutic Strategies in Regenerative Medicine. Int. J. Mol. Sci. 2017, 18, 1852. [Google Scholar] [CrossRef]
- Seo, Y.; Kim, H.S.; Hong, I.S. Stem Cell-Derived Extracellular Vesicles as Immunomodulatory Therapeutics. Stem Cells International. 2019, 2019. [Google Scholar] [CrossRef]
- Farinazzo, A.; Turano, E.; Marconi, S.; Bistaffa, E.; Bazzoli, E.; Bonetti, B. Murine adipose-derived mesenchymal stromal cell vesicles: In vitro clues for neuroprotective and neuroregenerative approaches. Cytotherapy 2015, 17, 571–578. [Google Scholar] [CrossRef]
- Bonafede, R.; Scambi, I.; Peroni, D.; Potrich, V.; Boschi, F.; Benati, D.; Bonetti, B.; Mariotti, R. Exosome derived from murine adipose-derived stromal cells: Neuroprotective effect on in vitro model of amyotrophic lateral sclerosis. Exp. Cell Res. 2016, 340, 150–158. [Google Scholar] [CrossRef]
- Katsuda, T.; Tsuchiya, R.; Kosaka, N.; Yoshioka, Y.; Takagaki, K.; Oki, K.; Takeshita, F.; Sakai, Y.; Kuroda, M.; Ochiya, T. Human adipose tissue-derived mesenchymal stem cells secrete functional neprilysin-bound exosomes. Sci. Rep. 2013, 3, 1197. [Google Scholar] [CrossRef]
- Laso-Garcia, F.; Ramos-Cejudo, J.; Carrillo-Salinas, F.J.; Otero-Ortega, L.; Feliu, A.; Gomez-de Frutos, M.; Mecha, M.; Diez-Tejedor, E.; Guaza, C.; Gutierrez-Fernandez, M. Therapeutic potential of extracellular vesicles derived from human mesenchymal stem cells in a model of progressive multiple sclerosis. PLoS ONE 2018, 13, e0202590. [Google Scholar] [CrossRef]
- Farinazzo, A.; Angiari, S.; Turano, E.; Bistaffa, E.; Dusi, S.; Ruggieri, S.; Bonafede, R.; Mariotti, R.; Constantin, G.; Bonetti, B. Nanovesicles from adipose-derived mesenchymal stem cells inhibit T lymphocyte trafficking and ameliorate chronic experimental autoimmune encephalomyelitis. Sci. Rep. 2018, 8, 7473. [Google Scholar] [CrossRef]
- Blazquez, R.; Sanchez-Margallo, F.M.; de la Rosa, O.; Dalemans, W.; Alvarez, V.; Tarazona, R.; Casado, J.G. Immunomodulatory potential of human adipose mesenchymal stem cells derived exosomes on in vitro stimulated T cells. Front. Immunol. 2014, 5, 556. [Google Scholar] [CrossRef]
- Cho, B.S.; Kim, J.O.; Ha, D.H.; Yi, Y.W. Exosomes derived from human adipose tissue-derived mesenchymal stem cells alleviate atopic dermatitis. Stem Cell Res. Ther. 2018, 9, 187. [Google Scholar] [CrossRef]
- Hu, L.; Wang, J.; Zhou, X.; Xiong, Z.; Zhao, J.; Yu, R.; Huang, F.; Zhang, H.; Chen, L. Exosomes derived from human adipose mensenchymal stem cells accelerates cutaneous wound healing via optimizing the characteristics of fibroblasts. Sci. Rep. 2016, 6, 32993. [Google Scholar] [CrossRef]
- Cui, X.J.; He, Z.Y.; Liang, Z.H.; Chen, Z.Y.; Wang, H.F.; Zhang, J.K. Exosomes From Adipose-derived Mesenchymal Stem Cells Protect the Myocardium Against Ischemia/Reperfusion Injury Through Wnt/beta-Catenin Signaling Pathway. J. Cardiovasc. Pharm. 2017, 70, 225–231. [Google Scholar] [CrossRef]
- Liang, X.; Zhang, L.; Wang, S.; Han, Q.; Zhao, R.C. Exosomes secreted by mesenchymal stem cells promote endothelial cell angiogenesis by transferring miR-125a. J. Cell Sci. 2016, 129, 2182–2189. [Google Scholar] [CrossRef] [Green Version]
- Yu, B.; Kim, H.W.; Gong, M.; Wang, J.C.; Millard, R.W.; Wang, Y.G.; Ashraf, M.; Xu, M.F. Exosomes secreted from GATA-4 overexpressing mesenchymal stem cells serve as a reservoir of anti-apoptotic microRNAs for cardioprotection. Int. J. Cardiol. 2015, 182, 349–360. [Google Scholar] [CrossRef]
- Di Trapani, M.; Bassi, G.; Midolo, M.; Gatti, A.; Takam, P.; Cassaro, A.; Carusone, R.; Adamo, A.; Krampera, M. Differential and Transferable Modulatory Effects of Mesenchymal Stromal Cell-Derived Extracellular Vesicles on T, B and Nk Cell Functions. Haematologica 2016, 101, S37. [Google Scholar] [CrossRef]
- Lo Sicco, C.; Reverberi, D.; Balbi, C.; Ulivi, V.; Principi, E.; Pascucci, L.; Becherini, P.; Bosco, M.C.; Varesio, L.; Franzin, C.; et al. Mesenchymal Stem Cell-Derived Extracellular Vesicles as Mediators of Anti-Inflammatory Effects: Endorsement of Macrophage Polarization. Stem Cell Transl. Med. 2017, 6, 1018–1028. [Google Scholar] [CrossRef]
- Lou, G.H.; Song, X.L.; Yang, F.; Wu, S.S.; Wang, J.; Chen, Z.; Liu, Y.N. Exosomes derived from miR-122-modified adipose tissue-derived MSCs increase chemosensitivity of hepatocellular carcinoma. J. Hematol. Oncol. 2015, 8, 122. [Google Scholar] [CrossRef]
- Qu, Y.; Zhang, Q.D.; Cai, X.B.; Li, F.; Ma, Z.Z.; Xu, M.Y.; Lu, L.E. Exosomes derived from miR-181-5p-modified adipose-derived mesenchymal stem cells prevent liver fibrosis via autophagy activation. J. Cell Mol. Med. 2017, 21, 2491–2502. [Google Scholar] [CrossRef]
- Katsuda, T.; Oki, K.; Ochiya, T. Potential application of extracellular vesicles of human adipose tissue-derived mesenchymal stem cells in Alzheimer’s disease therapeutics. Methods Mol. Biol. 2015, 1212, 171–181. [Google Scholar]
Target Gene | In Vitro Effects | In Vivo Results | Model/Condition | Reference |
---|---|---|---|---|
Sox2/Oct4 | Proliferation ↑ Osteogenesis ↑ Adipogenesis ↑ | - | - | [112] |
SOD2 | Survival ↑ | Engraftment ↑ | Hypoxia | [116] |
SOD2 | ROS ↓ PPARG, FABP-4, IL-6, TNFα expression ↓ | Body weight ↓ Adipocyte inflammation ↓ Glucose tolerance ↑ | Hyperglycemia (obese diabetic mouse model) | [117] |
CXCR4 | Proliferation ↑ Apoptosis ↓ Migration ↑ | - | - | [119] |
CXCR4 | - | Long-term engraftment ↑ Muscular regeneration ↑ | Diabetic mice with hindlimb ischemia | [120] |
GCP-2/ CXCL6 | VEGFA, HGF, IL-8 ↑ IGF-1, Akt-1 ↑ Proliferation ↑ Migration ↑ Endothelial differentiation ↑ | Angiogenesis ↑ Infarct size ↓ Heart function ↑ | Myocardial infarction model | [121] |
IL-4 | T-cell suppression ↑ | MOG-specific T-cell priming ↓ EAE protective effect | Experimental autoimmune encephalomyelitis | [126] |
CTLA4Ig | - | Treg/Th17 ratio ↑ CII autoantibodies ↓ CIA therapeutic effect | Collagen-induced arthritis | [127] |
sRAGE | IL-1β, IL-6, VEGF ↓ IDO, IL-10, TGF-β, HGF ↑ Migration ↑ | Treg/Th17 ratio ↑ Inflammatory arthritis ↓ | Arthritic IL-1Ra-knockout mice | [128] |
sST2 | Immunomodulatory mediator expression ↑ | Pulmonary inflammation ↓ Alveolar architecture ↑ | Endotoxin-induced acute lung injury | [129] |
IL-33 | T-cell proliferation ↓ IL17 secretion ↓ | - | - | [130] |
Culture Protocol | Phenotypic Changes | Applied Disease Model | Reference | |||
---|---|---|---|---|---|---|
Method | Duration | Size | Seeding Density | |||
Chitosan-coated culture plate | 7 days | 150 μm | 2.5 × 104 cells/cm2 | ECM ↑ Pluripotent marker ↑ Osteogenesis ↑ Adipogenesis ↓ | - | [151] |
Chitosan-coated culture plate | 7 days | - | 2.5 × 104 cells/cm2 | Pluripotent marker ↑ Angiogenesis ↑ | Skin wound-healing model | [153] |
Silicon elastomer-based concave wells | 5 days | ~200 μm | 1 × 105, 3 × 105, 6 × 105/well | Osteogenesis ↑ Angiogenesis ↑ | - | [156] |
Polydimethylsiloxane-based concave wells | 1 day | - | 105/well | Growth factor ↑ Pro-survival signal ↑ | Elastase-induced emphysema model | [157] |
Ultra-low attachment plates | 3 days | ~50 μm | - | ECM ↑ Angiogenesis ↑ | Hindlimb Ischemia model | [158] |
Ultra-low attachment plate | 3 days | - | 7.5 × 104 cells/cm2 | Angiogenesis ↑ endothelial markers ↑ | Skin defect wound-healing model | [159] |
Hanging drop | 1 day | - | 25,000 cells/drop | ECM ↑ Antioxdative effect ↑ Angiogenesis ↑ | Acute kidney ischemia model | [160] |
Hanging drop | 1 day | ~200 μm | 25,000 cells/drop | ECM ↑ Angiogenesis ↑ | Diabetic skin wound model | [152] |
Spinner flask | 3 days | >200 μm | 106 cells/mL | Chondrogenesis ↑ | - | [161] |
Microgravity bioreactor | 5 days | 123.4 ± 26.2 μm | 106 cells/mL | ECM ↑ Osteogenesis, Adipogenesis, Chondrigenesis ↑ | Tetrachloride-induced acute liver failure | [155] |
Priming Regimen | EV Source | In Vitro Effects | In Vivo Effects | Target Disease | Ref. |
---|---|---|---|---|---|
Filtration +UC | Naïve ASCs | Oxidative stress↓ in SH-SY5Y cells and primary murine hippocampal neurons | - | - | [216] |
PureExo® Exosome isolation kit | Naïve ASCs | Oxidative stress↓ in NSC-34 cells | - | Familial ALS | [217] |
Filtration + UC | Naïve ASCs | Amyloid-β levels↓in N2a cells | AD | [218] | |
UC | Naïve ASCs | - | Cell proliferation in the SVZ↑ Anti-inflammatory brain atrophy↓ | MS | [219] |
UC | Naïve ASCs | Proliferation↓in N9 cells activation and adhesion capacity↓in CD4+ T cells | Limiting immune cell infiltration Anti-inflammatory behavior improvement | MS | [220] |
UC | Naïve ASCs | Proliferation↓in T cells inhibit IFN-γ production terminal differentiation↓in effector-memory T cells | - | - | [221] |
UC | Naïve ASCs | Clinical score↓ Inflammatory dendritic epidermal cell/mast cell infiltration↓ Serum IgE level↓ | Atopic dermatitis | [222] | |
Exoquick® Exosome isolation kit | Naïve ASCs | Migration, proliferation, collagen synthesis↑in fibroblast | Cutaneous wound healing↑ Collagen production↑ | - | [223] |
Filtration + UC | Naïve ASCs | Hypoxic damage and apoptosis↓in H9C2 cells | Infarct size↓Ischemic damage-related marker↓Myocardial apoptosis↓ | Ischemic heart disease | [224] |
Filtration + UC | miR-125a-OE ASCs | Pro-angiogenic gene expression↑in HUVEC vascular length and branch↑in tube formation assay | Vascular structure↑in Matrigel plug assay | - | [225] |
Exoquick® Exosome isolation kit | GATA4 OE ASCs | Hypoxic apoptosis↓in cardiomyocyte | Infarct size↓ Cardiac fibrosis↓ Ventricle wall thickness↑ | Regional myocardial ischemia | [226] |
UC | IFNγ/TNFα primed ASCs | Proliferation↓in B cells and NK cells Immunosuppressive effect↑in MSCs | - | - | [227] |
UC | Hypoxic(1%) cultured ASCs | Macrophagic M2 induction Macrophagic M1 inhibition | Vascular structure↑in Matrigel plug assay macrophage infiltration↓at the damaged site muscular regeneration↑ | Skeletal muscle Injury | [228] |
Exoquick® Exosome isolation kit | miR-122-OE ASCs | Chemosensitivity↑in HepG2 cells | Sensitization of HCC cells to Sorafenib tumor growth↓ | HCC | [229] |
Exoquick® Exosome isolation kit | miR-181-5p-OE ASCs | Proliferation↓in HST-T6 cells Autophagy↑in HST-T6 cells | Fibrosis↓ inflammation↓ | Liver fibrosis | [230] |
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Seo, Y.; Shin, T.-H.; Kim, H.-S. Current Strategies to Enhance Adipose Stem Cell Function: An Update. Int. J. Mol. Sci. 2019, 20, 3827. https://doi.org/10.3390/ijms20153827
Seo Y, Shin T-H, Kim H-S. Current Strategies to Enhance Adipose Stem Cell Function: An Update. International Journal of Molecular Sciences. 2019; 20(15):3827. https://doi.org/10.3390/ijms20153827
Chicago/Turabian StyleSeo, Yoojin, Tae-Hoon Shin, and Hyung-Sik Kim. 2019. "Current Strategies to Enhance Adipose Stem Cell Function: An Update" International Journal of Molecular Sciences 20, no. 15: 3827. https://doi.org/10.3390/ijms20153827
APA StyleSeo, Y., Shin, T. -H., & Kim, H. -S. (2019). Current Strategies to Enhance Adipose Stem Cell Function: An Update. International Journal of Molecular Sciences, 20(15), 3827. https://doi.org/10.3390/ijms20153827