Awakened by cellular stress: isolation and characterization of a novel population of pluripotent stem cells derived from human adipose tissue

Saleh Heneidi, Ariel A Simerman, Erica Keller, Prapti Singh, Xinmin Li, Daniel A Dumesic, Gregorio Chazenbalk, Saleh Heneidi, Ariel A Simerman, Erica Keller, Prapti Singh, Xinmin Li, Daniel A Dumesic, Gregorio Chazenbalk

Abstract

Advances in stem cell therapy face major clinical limitations, particularly challenged by low rates of post-transplant cell survival. Hostile host factors of the engraftment microenvironment such as hypoxia, nutrition deprivation, pro-inflammatory cytokines, and reactive oxygen species can each contribute to unwanted differentiation or apoptosis. In this report, we describe the isolation and characterization of a new population of adipose tissue (AT) derived pluripotent stem cells, termed Multilineage Differentiating Stress-Enduring (Muse) Cells, which are isolated using severe cellular stress conditions, including long-term exposure to the proteolytic enzyme collagenase, serum deprivation, low temperatures and hypoxia. Under these conditions, a highly purified population of Muse-AT cells is isolated without the utilization of cell sorting methods. Muse-AT cells grow in suspension as cell spheres reminiscent of embryonic stem cell clusters. Muse-AT cells are positive for the pluripotency markers SSEA3, TR-1-60, Oct3/4, Nanog and Sox2, and can spontaneously differentiate into mesenchymal, endodermal and ectodermal cell lineages with an efficiency of 23%, 20% and 22%, respectively. When using specific differentiation media, differentiation efficiency is greatly enhanced in Muse-AT cells (82% for mesenchymal, 75% for endodermal and 78% for ectodermal). When compared to adipose stem cells (ASCs), microarray data indicate a substantial up-regulation of Sox2, Oct3/4, and Rex1. Muse-ATs also exhibit gene expression patterns associated with the down-regulation of genes involved in cell death and survival, embryonic development, DNA replication and repair, cell cycle and potential factors related to oncogenecity. Gene expression analysis indicates that Muse-ATs and ASCs are mesenchymal in origin; however, Muse-ATs also express numerous lymphocytic and hematopoietic genes, such as CCR1 and CXCL2, encoding chemokine receptors and ligands involved in stem cell homing. Being highly resistant to severe cellular stress, Muse-AT cells have the potential to make a critical impact on the field of regenerative medicine and cell-based therapy.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Isolation and morphologic characterization of…
Figure 1. Isolation and morphologic characterization of Muse-ATs.
(A) Schematic of Muse-AT isolation and activation from their quiescent state by exposure to cellular stress. Muse-AT cells were obtained after 16 hours, with incubation with collagenase in DMEM medium without FCS at 4°C under very low O2 (See Methods). (B) FACS analysis demonstrates that 90% of isolated cells are both SSEA3 and CD105 positive. (C) Muse-AT cells can grow in suspension, forming spheres or cell clusters as well as individual cells (see red arrows) or (D) Muse-AT cells can adhere to the dish and form cell aggregates. Under both conditions, individual Muse-AT cells reached a diameter of approximately 10µm and cell clusters reached a diameter of up to 50µm, correlating to stem cell proliferative size capacity.
Figure 2. Muse-ATs express pluripotent stem cell…
Figure 2. Muse-ATs express pluripotent stem cell markers.
Immunofluorescence microscopy demonstrates that Muse-AT aggregates, along with individual Muse-AT cells, express characteristic pluripotent stem cell markers, including SSEA3, Oct3/4, Nanog, Sox2, and TRA1-60. Comparatively, ASCs (right panel) derived from the same lipoaspirate under standard conditions (see above, were negative for these pluripotent stem cell markers. Nuclei were stained with DAPI (blue). Original magnification, 600 X.
Figure 3. Muse-AT cells can differentiate to…
Figure 3. Muse-AT cells can differentiate to mesodermal cell lineages.
Isolated Muse-ATs were grown as adherent cells in the presence of DMEM/10%FCS for 6 days. (A) Immunostaining indicates that Muse-AT have the capacity to spontaneously differentiate to mesodermal cell lineages. DLK is a specific marker for preadipocytes; BODIPY-C16 recognized lipid drops present in adipocytes; Myosin D is specific marker for myocytes. Comparatively, ASCs (right panel) which are mesenchymal stem cells derived from the same lipoaspirate under standard conditions (see above, were slightly positive for DLK; nuclei were stained with DAPI (blue), original magnification, 600 X. (B) Isolated Muse-AT cells were grown as adherent cells in the presence of adipocyte differentiation medium (See Methods) for 3 or 6 days and formation of adipocytes was detected using BODIPY-C16; (C) Isolated Muse-AT cells were grown as adherent cells in the presence of myocyte differentiation medium and formation of myocytes was detected using anti human-MSA antibodies. Comparatively, ASCs (right panel) which are mesenchymal stem cells derived from the same lipoaspirate under standard conditions (See above, were slightly positive for BODIPY-C16 or MSA (B and C, right panel). Nuclei were stained with DAPI (blue). Original magnification was 200 X (first three rows) or 600 X (last two rows).
Figure 4. Muse-ATs can differentiate to endodermal…
Figure 4. Muse-ATs can differentiate to endodermal cell lineages.
Isolated Muse-ATs were grown as adherent cells in the presence of DMEM/10%FCS for 6 days. (A) Immunostaining indicates that Muse-AT have the capacity to spontaneously differentiate to endodermal cell lineages. α-fetoprotein and pan-keratin are specific markers for hepatocytes; Nuclei were stained with DAPI (blue). Original magnification, 600 X. Comparatively, ASCs (right panel) derived from the same lipoaspirate under standard conditions (see above, [16]) were negative for α-fetoprotein and pan-keratin. Nuclei were stained with DAPI (blue); original magnification, 600 X; (B) Isolated Muse-AT cells were grown as adherent cells in the presence of hepatocyte differentiation medium (see methods) for 3 or 6 days and formation of hepatocytes was detected using anti-human cytokeratin 7 or α-fetoprotein antibodies. Comparatively, ASCs (right panel) were completely negative for cytokeratin 7 and α-fetoprotein (B, right panel). Nuclei were stained with DAPI (blue). Original magnification was 200 X (first three rows) or 600 X (last two rows).
Figure 5. Muse- ATs can differentiate to…
Figure 5. Muse- ATs can differentiate to ectodermal cell lineages.
(A) Isolated Muse-ATs were grown as adherent cells in the presence of DMEM/10%FCS for 6 days. Immunostaining indicates that Muse-AT cells have the capacity to spontaneously differentiate to ectodermal cell lineages. NSE, Glu-R and Neuro D are specific markers for neural progenitor or neural-like cells. Comparatively, ASCs (right panel) were negative for NSE, Glu-R and NeuroD. Nuclei were stained with DAPI (blue); original magnification was 600X. (B) Isolated Muse-AT cells were grown as non-adherent cells in the presence of Neurobasal medium/B-27 supplement serum free/kanamycin/glutamine/bFGF and EGF for 7 days. Formation of neural cell spheres was detected at different times during this incubation. Cells were then grown as adherent cells for an additional 7 days in a DMEM 2% FCS/bFGF/BDNF. Formation of neural-like cells is indicated by red arrows. (C) Isolated Muse-AT cells were grown for 7 days as non-adherent cells and then cultured for another 7 days as adherent cells (See Methods). Neural-like cells were detected by immunofluorescence using anti-human nestin and MAP2 antibodies. Comparatively, ASCs (right panel) were negative for nestin and MAP2 (B, right panel). Nuclei were stained with DAPI (blue). Original magnification was 200 X (first three rows) or 600X (last two rows).
Figure 6. Identification of functional groups and…
Figure 6. Identification of functional groups and canonical pathways in Muse-ATs vs ASCs.
Identification of the top ten (A) functional groups (B) canonical pathways of all differentially expressed genes (2 fold or higher) in Muse-AT cells versus ASCs. Fischer’s exact test was used to calculate a p-value determining the probability of the association between the genes in the data set with functional groups and canonical pathways. Both (A) functional groups (B) canonical pathways are displayed along the x-axis, while the y-axis displays logarithm of p values calculated by Fisher exact between the ratio of the number of genes differentially expressed genes (2 fold or higher) in Muse-AT versus ASCs in a given functional group or pathway divided by total number of genes that make up that functional group or pathway with a threshold for statistical significance set at 0.05. The analysis was performed by Ingenuity Pathways analysis software.

References

    1. Dayen AA, Choi H, Kim J, Cho S (2010) Role of Oxidate Stress in Stem, Cancer, and Cancer Stem Cells. Cancers 2: 859–884.
    1. Fulda S, Gorman AM, Hori O, Samali A (2009) Cellular Stress Responses: Cell Survival and Cell Death. International Journal of Cell Biology 2010.
    1. Hodgetts SI, Beilharz MW, Scalzo AA, Grounds MD (2000) Why do cultured transplanted myoblasts die in vivo? DNA quantification shows enhanced survival of donor male myoblasts in host mice depleted of CD4+ and CD8+ cells or Nk1.1+ cells. Cell Transplant 9: 489–502.
    1. Oh JS, Kim KN, An SS, Pennant WA, Kim HJ, et al. (2010) Cotransplantation of mouse neural stem cells (mNSCs) with adipose tissue-derived mesenchymal stem cells improves mNSC survival in a rat spinal cord injury model. Cell Transplant 20: 837–849.
    1. Mingliang R, Bo Z, Zhengguo W (2011) Stem cells for cardiac repair: status, mechanisms, and new strategies. Stem Cells Int 2011: 310928.
    1. Hofmann M, Wollert KC, Meyer GP, Menke A, Arseniev L, et al. (2005) Monitoring of bone marrow cell homing into the infarcted human myocardium. Circulation 111: 2198–2202.
    1. Kultz D (2005) Molecular and evolutionary basis of the cellular stress response. Annu Rev Physiol 67: 225–257.
    1. Chacko SM, Ahmed S, Selvendiran K, Kuppusamy ML, Khan M, et al. (2010) Hypoxic preconditioning induces the expression of prosurvival and proangiogenic markers in mesenchymal stem cells. Am J Physiol Cell Physiol 299: C1562–1570.
    1. Csete M (2005) Oxygen in the cultivation of stem cells. Ann N Y Acad Sci 1049: 1–8.
    1. Grayson WL, Zhao F, Bunnell B, Ma T (2007) Hypoxia enhances proliferation and tissue formation of human mesenchymal stem cells. Biochem Biophys Res Commun 358: 948–953.
    1. Abdollahi H, Harris LJ, Zhang P, McIlhenny S, Srinivas V, et al. (2009) The role of hypoxia in stem cell differentiation and therapeutics. J Surg Res 165: 112–117.
    1. Eliasson P, Rehn M, Hammar P, Larsson P, Sirenko O, et al... (2010) Hypoxia mediates low cell-cycle activity and increases the proportion of long-term-reconstituting hematopoietic stem cells during in vitro culture. Exp Hematol 38: 301–310 e302.
    1. Kuroda Y, Kitada M, Wakao S, Nishikawa K, Tanimura Y, et al. (2010) Unique multipotent cells in adult human mesenchymal cell populations. Proc Natl Acad Sci U S A 107: 8639–8643.
    1. Wakao S, Kitada M, Kuroda Y, Shigemoto T, Matsuse D, et al. (2011) Multilineage-differentiating stress-enduring (Muse) cells are a primary source of induced pluripotent stem cells in human fibroblasts. Proc Natl Acad Sci U S A 108: 9875–9880.
    1. Gimble JM, Katz AJ, Bunnell BA (2007) Adipose-derived stem cells for regenerative medicine. Circ Res 100: 1249–1260.
    1. Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, et al. (2002) Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell 13: 4279–4295.
    1. Hristov M, Zernecke A, Bidzhekov K, Liehn EA, Shagdarsuren E, et al. (2007) Importance of CXC chemokine receptor 2 in the homing of human peripheral blood endothelial progenitor cells to sites of arterial injury. Circ Res 100: 590–597.
    1. Oyagi S, Hirose M, Kojima M, Okuyama M, Kawase M, et al. (2006) Therapeutic effect of transplanting HGF-treated bone marrow mesenchymal cells into CCl4-injured rats. J Hepatol 44: 742–748.
    1. Hermann A, Gastl R, Liebau S, Popa MO, Fiedler J, et al. (2004) Efficient generation of neural stem cell-like cells from adult human bone marrow stromal cells. J Cell Sci 117: 4411–4422.
    1. Di Rocco G, Iachininoto MG, Tritarelli A, Straino S, Zacheo A, et al. (2006) Myogenic potential of adipose-tissue-derived cells. J Cell Sci 119: 2945–2952.
    1. Chazenbalk G, Bertolotto C, Heneidi S, Jumabay M, Trivax B, et al. (2011) Novel pathway of adipogenesis through cross-talk between adipose tissue macrophages, adipose stem cells and adipocytes: evidence of cell plasticity. PLoS One 6: e17834.
    1. Listenberger LL, Brown DA (2007) Fluorescent detection of lipid droplets and associated proteins. Curr Protoc Cell Biol Chapter 24: Unit 24 22.
    1. Sarugaser R, Hanoun L, Keating A, Stanford WL, Davies JE (2009) Human mesenchymal stem cells self-renew and differentiate according to a deterministic hierarchy. PLoS One 4: e6498.
    1. Beier JP, Bitto FF, Lange C, Klumpp D, Arkudas A, et al. (2011) Myogenic differentiation of mesenchymal stem cells co-cultured with primary myoblasts. Cell Biol Int 35: 397–406.
    1. Fink T, Zachar V (2011) Adipogenic differentiation of human mesenchymal stem cells. Methods Mol Biol 698: 243–251.
    1. Bunnell BA, Flaat M, Gagliardi C, Patel B, Ripoll C (2008) Adipose-derived stem cells: isolation, expansion and differentiation Methods. 45: 115–120.
    1. Kuhlmann WD, Peschke P (2006) Hepatic progenitor cells, stem cells, and AFP expression in models of liver injury. Int J Exp Pathol 87: 343–359.
    1. Carlini P, Ferranti P, Polizio F, Ciriolo MR, Rotilio G (2007) Purification and characterization of Alpha-Fetoprotein from the human hepatoblastoma HepG2 cell line in serum-free medium. Biometals 20: 869–878.
    1. Si-Tayeb K, Noto FK, Nagaoka M, Li J, Battle MA, et al. (2010) Highly efficient generation of human hepatocyte-like cells from induced pluripotent stem cells. Hepatology 51: 297–305.
    1. Teramoto K, Asahina K, Kumashiro Y, Kakinuma S, Chinzei R, et al. (2005) Hepatocyte differentiation from embryonic stem cells and umbilical cord blood cells. J Hepatobiliary Pancreat Surg 12: 196–202.
    1. Levy YS, Merims D, Panet H, Barhum Y, Melamed E, et al. (2003) Induction of neuron-specific enolase promoter and neuronal markers in differentiated mouse bone marrow stromal cells. J Mol Neurosci 21: 121–132.
    1. Yuan SH, Martin J, Elia J, Flippin J, Paramban RI, et al. (2011) Cell-surface marker signatures for the isolation of neural stem cells, glia and neurons derived from human pluripotent stem cells. PLoS One 6: e17540.
    1. MacPherson PA, Jones S, Pawson PA, Marshall KC, McBurney MW (1997) P19 cells differentiate into glutamatergic and glutamate-responsive neurons in vitro. Neuroscience 80: 487–499.
    1. Molnar E, Baude A, Richmond SA, Patel PB, Somogyi P, et al. (1993) Biochemical and immunocytochemical characterization of antipeptide antibodies to a cloned GluR1 glutamate receptor subunit: cellular and subcellular distribution in the rat forebrain. Neuroscience 53: 307–326.
    1. Woodbury D, Reynolds K, Black IB (2002) Adult bone marrow stromal stem cells express germline, ectodermal, endodermal, and mesodermal genes prior to neurogenesis. J Neurosci Res 69: 908–917.
    1. Kuwabara T, Hsieh J, Muotri A, Yeo G, Warashina M, et al. (2009) Wnt-mediated activation of NeuroD1 and retro-elements during adult neurogenesis. Nat Neurosci 12: 1097–1105.
    1. Gottlieb DI, Huettner JE (1999) An in vitro pathway from embryonic stem cells to neurons and glia. Cells Tissues Organs 165: 165–172.
    1. Karumbayaram S, Novitch BG, Patterson M, Umbach JA, Richter L, et al. (2009) Directed differentiation of human-induced pluripotent stem cells generates active motor neurons. Stem Cells 27: 806–811.
    1. Li M, Pevny L, Lovell-Badge R, Smith A (1998) Generation of purified neural precursors from embryonic stem cells by lineage selection. Curr Biol 8: 971–974.
    1. Reubinoff BE, Itsykson P, Turetsky T, Pera MF, Reinhartz E, et al. (2001) Neural progenitors from human embryonic stem cells. Nat Biotechnol 19: 1134–1140.
    1. Zhang SC, Wernig M, Duncan ID, Brustle O, Thomson JA (2001) In vitro differentiation of transplantable neural precursors from human embryonic stem cells. Nat Biotechnol 19: 1129–1133.
    1. Suzuki S, Namiki J, Shibata S, Mastuzaki Y, Okano H (2010) The neural stem/progenitor cell marker nestin is expressed in proliferative endothelial cells, but not in mature vasculature. J Histochem Cytochem 58: 721–730.
    1. Kuznetsov SA, Rodionov VI, Gelfand VI, Rosenblat VA (1981) Purification of high-Mr microtubule proteins MAP1 and MAP2. FEBS Lett 135: 237–240.
    1. del Castillo FJ, Cohen-Salmon M, Charollais A, Caille D, Lampe PD, et al. (2010) Consortin, a trans-Golgi network cargo receptor for the plasma membrane targeting and recycling of connexins. Hum Mol Genet 19: 262–275.
    1. Liu L, Wise DR, Diehl JA, Simon MC (2008) Hypoxic reactive oxygen species regulate the integrated stress response and cell survival. J Biol Chem 283: 31153–31162.
    1. Hollenbeck ST, Senghaas A, Komatsu I, Zhang Y, Erdmann D, et al. (2012) Tissue engraftment of hypoxic-preconditioned adipose-derived stem cells improves flap viability. Wound Repair Regen 20: 872–878.
    1. Blanpain C, Lowry WE, Geoghegan A, Polak L, Fuchs E (2004) Self-renewal, multipotency, and the existence of two cell populations within an epithelial stem cell niche. Cell 118: 635–648.
    1. Li L, Bhatia R (2011) Stem cell quiescence. Clin Cancer Res 17: 4936–4941.
    1. Medici D, Shore EM, Lounev VY, Kaplan FS, Kalluri R, et al. (2010) Conversion of vascular endothelial cells into multipotent stem-like cells. Nat Med 16: 1400–1406.
    1. Xiao N, Jani K, Morgan K, Okabe R, Cullen DE, et al. (2012) Hematopoietic stem cells lacking Ott1 display aspects associated with aging and are unable to maintain quiescence during proliferative stress. Blood 119: 4898–4907.
    1. Acharyya S, Oskarsson T, Vanharanta S, Malladi S, Kim J, et al. (2012) A CXCL1 paracrine network links cancer chemoresistance and metastasis. Cell 150: 165–178.
    1. Huang J, Zhang Z, Guo J, Ni A, Deb A, et al. (2010) Genetic modification of mesenchymal stem cells overexpressing CCR1 increases cell viability, migration, engraftment, and capillary density in the injured myocardium. Circ Res 106: 1753–1762.
    1. Plath K, Lowry WE (2011) Progress in understanding reprogramming to the induced pluripotent state. Nat Rev Genet 12: 253–265.
    1. Couture JP, Daviau A, Fradette J, Blouin R (2009) The mixed-lineage kinase DLK is a key regulator of 3T3-L1 adipocyte differentiation. PLoS One 4: e4743.
    1. Thompson SA, Copeland CR, Reich DH, Tung L (2011) Mechanical coupling between myofibroblasts and cardiomyocytes slows electric conduction in fibrotic cell monolayers. Circulation 123: 2083–2093.
    1. Banas A, Teratani T, Yamamoto Y, Tokuhara M, Takeshita F, et al. (2007) Adipose tissue-derived mesenchymal stem cells as a source of human hepatocytes. Hepatology 46: 219–228.
    1. Behbahan IS, Duan Y, Lam A, Khoobyari S, Ma X, et al. (2011) New approaches in the differentiation of human embryonic stem cells and induced pluripotent stem cells toward hepatocytes. Stem Cell Rev 7: 748–759.
    1. Hamazaki T, Iiboshi Y, Oka M, Papst PJ, Meacham AM, et al. (2001) Hepatic maturation in differentiating embryonic stem cells in vitro. FEBS Lett 497: 15–19.
    1. Hu AB, Cai JY, Zheng QC, He XQ, Shan Y, et al. (2004) High-ratio differentiation of embryonic stem cells into hepatocytes in vitro. Liver Int 24: 237–245.
    1. Zamule SM, Coslo DM, Chen F, Omiecinski CJ (2011) Differentiation of human embryonic stem cells along a hepatic lineage. Chem Biol Interact 190: 62–72.
    1. Mizuno H, Tobita M, Uysal AC (2012) Concise review: Adipose-derived stem cells as a novel tool for future regenerative medicine. Stem Cells 30: 804–810.
    1. Mignone JL, Kukekov V, Chiang AS, Steindler D, Enikolopov G (2004) Neural stem and progenitor cells in nestin-GFP transgenic mice. J Comp Neurol 469: 311–324.
    1. Kang KS, Trosko JE (2011) Stem cells in toxicology: fundamental biology and practical considerations. Toxicol Sci 120 Suppl 1S269–289.
    1. Vasiliou V, Nebert DW (2005) Analysis and update of the human aldehyde dehydrogenase (ALDH) gene family. Hum Genomics 2: 138–143.
    1. Trosko JE, Chang CC, Wilson MR, Upham B, Hayashi T, et al. (2000) Gap junctions and the regulation of cellular functions of stem cells during development and differentiation. Methods 20: 245–264.
    1. Loven J, Orlando DA, Sigova AA, Lin CY, Rahl PB, et al. (2012) Revisiting global gene expression analysis. Cell 151: 476–482.
    1. Meier P, Finch A, Evan G (2000) Apoptosis in development. Nature 407: 796–801.

Source: PubMed

3
Iratkozz fel