Pluripotent Nontumorigenic Adipose Tissue-Derived Muse Cells have Immunomodulatory Capacity Mediated by Transforming Growth Factor-β1

María L Gimeno, Florencia Fuertes, Andres E Barcala Tabarrozzi, Alejandra I Attorressi, Rodolfo Cucchiani, Luis Corrales, Talita C Oliveira, Mari C Sogayar, Leticia Labriola, Ricardo A Dewey, Marcelo J Perone, María L Gimeno, Florencia Fuertes, Andres E Barcala Tabarrozzi, Alejandra I Attorressi, Rodolfo Cucchiani, Luis Corrales, Talita C Oliveira, Mari C Sogayar, Leticia Labriola, Ricardo A Dewey, Marcelo J Perone

Abstract

Adult mesenchymal stromal cell-based interventions have shown promising results in a broad range of diseases. However, their use has faced limited effectiveness owing to the low survival rates and susceptibility to environmental stress on transplantation. We describe the cellular and molecular characteristics of multilineage-differentiating stress-enduring (Muse) cells derived from adipose tissue (AT), a subpopulation of pluripotent stem cells isolated from human lipoaspirates. Muse-AT cells were efficiently obtained using a simple, fast, and affordable procedure, avoiding cell sorting and genetic manipulation methods. Muse-AT cells isolated under severe cellular stress, expressed pluripotency stem cell markers and spontaneously differentiated into the three germ lineages. Muse-AT cells grown as spheroids have a limited proliferation rate, a diameter of ∼15 µm, and ultrastructural organization similar to that of embryonic stem cells. Muse-AT cells evidenced high stage-specific embryonic antigen-3 (SSEA-3) expression (∼60% of cells) after 7-10 days growing in suspension and did not form teratomas when injected into immunodeficient mice. SSEA-3+ -Muse-AT cells expressed CD105, CD29, CD73, human leukocyte antigen (HLA) class I, CD44, and CD90 and low levels of HLA class II, CD45, and CD34. Using lipopolysaccharide-stimulated macrophages and antigen-challenged T-cell assays, we have shown that Muse-AT cells have anti-inflammatory activities downregulating the secretion of proinflammatory cytokines, such as interferon-γ and tumor necrosis factor-α. Muse-AT cells spontaneously gained transforming growth factor-β1 expression that, in a phosphorylated SMAD2-dependent manner, might prove pivotal in their observed immunoregulatory activity through decreased expression of T-box transcription factor in T cells. Collectively, the present study has demonstrated the feasibility and efficiency of obtaining Muse-AT cells that can potentially be harnessed as immunoregulators to treat immune-related disorders. Stem Cells Translational Medicine 2017;6:161-173.

Keywords: Antigen-specific response; Immunomodulation; Spheroids/clusters; Stem cells; T lymphocytes.

© 2016 The Authors Stem Cells Translational Medicine published by Wiley Periodicals, Inc. on behalf of AlphaMed Press.

Figures

Figure 1
Figure 1
Generation and culture of liposuction‐derived Muse‐AT cells. (A): Scheme of Muse‐AT cell preparation protocol. Muse‐AT cells were obtained after digestion with collagenase and severe stress conditions. Clusters were generated when Muse‐AT cells were seeded on nonadherent plastic. (B): Muse‐AT cells formed clusters of 50–150 µm in diameter that grew in suspension culture. Scale bar = 50 µm. Left inset, black arrow indicates a single Muse‐AT cell at the edge of a cluster. Scale bar = 20 µm. (C): Transmission electron microscopy of a representative Muse‐AT cell after 5–7 days in culture showing a high nucleus to cytoplasm ratio. Scale bar = 500 nm. (D): Muse‐AT cells were seeded immediately on nonadhesive dishes after isolation (day 0) and formed clusters after 3 days of culture (day 3, cycle 1). After mechanical disaggregation, spheroids formed again, reaching 50–150 µm in diameter during the second and third growth cycle. Abbreviations: FBS, fetal bovine serum; Muse‐AT, multilineage‐differentiating stress‐enduring cells derived from adipose tissue; O/N, overnight; w/o, without.
Figure 2
Figure 2
Expression of pluripotency stem cell markers of Muse‐AT cell clusters. (A): Representative immunostaining of the stem cell markers Nanong, OCT4, TRA1‐60, Sox‐2, and SSEA‐4 were observed in almost all Muse‐AT cells in the clusters. Scale bar = 100 µm. White arrowheads indicate single cells shown in upper right insets. Scale bar = 5 µm. (B): Immunofluorescence quantification of stem cell markers. (C): SSEA‐3 was most highly expressed by Muse‐AT cells compared with ASCs, as assessed by fluorescence activated cell sorter (FACS) staining. (D): Immunofluorescence staining confirmed SSEA‐3 expression by Muse‐AT cell clusters. Scale bar = 100 µm. (E): FACS staining of several CDs revealed the immunophenotype of Muse‐AT cells. *, p < .05; **, p < .005; n = 3 samples analyzed. Abbreviations: ASCs, adipose‐derived stromal cells; d, day; Muse‐AT, multilineage‐differentiating stress‐enduring cells derived from adipose tissue; SSEA, stage‐specific embryonic antigen.
Figure 3
Figure 3
Muse‐AT cells differentiate into the three germ cell lineages. (A): Muse‐AT cells cultured in adherent plastic have the ability to spontaneously differentiate into mesodermal, endodermal, and ectodermal lineages, as demonstrated by the expression of mRNAs for Nkx2.5, α‐fetoprotein, and MAP‐2 respectively, determined by qualitative real‐time polymerase chain reaction. (B): In the presence of specific differentiation medium, immunofluorescence staining revealed positivity for hepatocyte (α‐fetoprotein, red), myocyte (SMA, green), and neuron (MAP‐2, red) markers. Nuclei were stained with 4′,6‐diamidino‐2‐phenylindole (blue). Original magnification ×200. Abbreviations: GAPDH, glyceraldehyde‐3‐phosphate dehydrogenase; MAP‐2, microtubule‐associated protein 2; Muse‐AT, multilineage‐differentiating stress‐enduring cells derived from adipose tissue; Nkx2.5, NK2 homeobox 5; SMA, smooth muscle actin.
Figure 4
Figure 4
Lack of teratoma formation after transplantation and stability of Muse‐AT cells. (A): Muse‐AT cells were injected (106) i.t. into NODscid mice. The transplanted mice were monitored weekly for the appearance of tumors. The P19 embryonic carcinoma cell line was injected (106) as the control. The mice were sacrificed when the tumors became outwardly apparent. NODscid mice injected with Muse‐AT cells did not develop teratoma during the observed period (up to 6 months). (B): H&E staining of testes showed normal tissue structure in NODscid Muse‐AT cells of the injected mice. (C): Representative normal karyotype of Muse‐AT cells that showed expression of stem cell markers. Abbreviations: d, day; i.t., intratesticular; Muse‐AT, multilineage‐differentiating stress‐enduring cells derived from adipose tissue.
Figure 5
Figure 5
Spontaneous expression of TGF‐β1 by Muse‐AT clusters. (A): Immunofluorescence staining of TGF‐β1 at different time points in culture. (B): Immunofluorescence quantification showed maximal expression of TGF‐β1 after 10 days in culture. (C): Expression levels of TGF‐β1 mRNA of Muse‐AT cell clusters at 0, 5, and 10 days in culture were determined by real‐time polymerase chain reaction. Ct values were normalized to the expression of the HPRT gene, and data are expressed relative to the values obtained for day 0. *, p < .05; **, p < .005; n = 5. Abbreviations: ASC, adipose‐derived stromal cell; d, day; Muse‐AT, multilineage‐differentiating stress‐enduring cells derived from adipose tissue; TGF‐β1, transforming growth factor‐β1.
Figure 6
Figure 6
Muse‐AT cell activity on LPS‐stimulated macrophages and splenocytes. (A): Muse‐AT cells in coculture with the RAW macrophage cell line. RAW cells were seeded onto the lower chamber of a Transwell plate (Corning) and stimulated with LPS for 20 minutes. LPS was removed, the cells washed, and Muse‐AT cells were then cultured (RAW/Muse‐AT in a 1:10; 1:1, and 10:1 ratio) in the upper chamber. (B): Muse‐AT cell CM effects on LPS‐stimulated RAW cells with and without the presence of SB 431542 (SB; 10 ng/ml). (C, D): Freshly isolated murine peritoneal MΦ stimulated with LPS. The effect of Muse‐AT cell CM on the secretion of TNF‐α (C) and IL‐10 (D) was evaluated with and without SB in the culture medium. (E): Antigen‐specific T‐cell response of NOD BDC2.5 splenocytes in culture. Muse‐AT cell CM was assayed at different dilutions (1 to 5 to 1 to 5,000; n = 3). (F): Muse‐AT cell CM downregulated antigen‐stimulated T‐bet expression assessed by Western blot (n = 3). (G): Inhibitory effect on antigen‐challenged T cells on TNF‐α secretion by Muse‐AT cell CM (n = 3). (H): Muse‐AT cell CM enhanced anti‐inflammatory cytokine IL‐10 secretion in antigen‐stimulated T cells (n = 6). The expression levels of mRNA IL‐10 relative to mRNA HPRT are indicated as an inset. Cytokine levels were quantified in 72‐hour culture supernatants by enzyme‐linked immunosorbent assays. OVA was used as a control. Mi concentration = 5 nM. *, p < .05; **, p < .005; ***, p < .0005. Abbreviations: ASCsCM, adipose stem cell conditioned media; CM, conditioned media; GAPDH, glyceraldehyde‐3‐phosphate dehydrogenase; IL, interleukin; LPS, lipopolysaccharide; MΦ, macrophages; Mi, mimotope; Muse‐AT, multilineage‐differentiating stress‐enduring cells derived from adipose tissue; NOD, nonobese diabetic; OVA, ovalbumin; Spl, splenocytes; T‐bet, T‐box transcription factor (TBX21); TNF‐α, tumor necrosis factor‐α.
Figure 7
Figure 7
TGF‐β1 signaling blockade on cytokine secretion. (A): Exogenously added TGF‐β1 to antigen‐stimulated splenocytes showed a concentration response dependence on IFN‐γ secretion. (B): The effect of TGF‐β1 signaling blockade was assessed using SB at different concentrations (1.25–10 ng/ml) on antigen‐challenged T cells followed IL‐10 quantification. (C): Inhibition of TGF‐β1 signaling with 10 ng/ml of SB restored IFN‐γ secretion levels. (D): SB diminished pSMAD2 levels assessed by Western blot (WB) in antigen‐stimulated T cells. (E): Quantification of WB analysis shown in (D). Cytokine measured by enzyme‐linked immunosorbent assays in 72‐hour supernatants. *, p < .05; **, p < .005; ***, p < .0005; n = 3. Abbreviations: CM, conditioned media; GAPDH, glyceraldehyde‐3‐phosphate dehydrogenase; IFN‐γ, interferon‐γ; Mi, mimotope; Muse‐AT, multilineage‐differentiating stress‐enduring cells derived from adipose tissue; p, phosphorylated; SB, SB 431542; Spl, splenocytes; TGF‐β1, transforming growth factor‐β1.

References

    1. Takahashi K, Tanabe K, Ohnuki M et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007;131:861–872.
    1. Sánchez Alvarado A, Yamanaka S. Rethinking differentiation: Stem cells, regeneration, and plasticity. Cell 2014;157:110–119.
    1. Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat Rev Immunol 2008;8:726–736.
    1. Thomson JA, Itskovitz‐Eldor J, Shapiro SS et al. Embryonic stem cell lines derived from human blastocysts. Science 1998;282:1145–1147.
    1. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006;126:663–676.
    1. Connick P, Kolappan M, Crawley C et al. Autologous mesenchymal stem cells for the treatment of secondary progressive multiple sclerosis: An open‐label phase 2a proof‐of‐concept study. Lancet Neurol 2012;11:150–156.
    1. Hare JM, Fishman JE, Gerstenblith G et al. Comparison of allogeneic vs autologous bone marrow–derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: The POSEIDON randomized trial. JAMA 2012;308:2369–2379.
    1. Cashman TJ, Gouon‐Evans V, Costa KD. Mesenchymal stem cells for cardiac therapy: Practical challenges and potential mechanisms. Stem Cell Rev 2013;9:254–265.
    1. Kuroda Y, Kitada M, Wakao S et al. Unique multipotent cells in adult human mesenchymal cell populations. Proc Natl Acad Sci USA 2010;107:8639–8643.
    1. Wakao S, Kitada M, Kuroda Y et al. Multilineage‐differentiating stress‐enduring (Muse) cells are a primary source of induced pluripotent stem cells in human fibroblasts. Proc Natl Acad Sci USA 2011;108:9875–9880.
    1. Kinoshita K, Kuno S, Ishimine H et al. Therapeutic potential of adipose‐derived SSEA‐3‐positive muse cells for treating diabetic skin ulcers. STEM CELLS TRANSLATIONAL MEDICINE 2015;4:146–155.
    1. Uchida H, Morita T, Niizuma K et al. Transplantation of unique subpopulation of fibroblasts, Muse cells, ameliorates experimental stroke possibly via robust neuronal differentiation. STEM CELLS 2016;34:160–173.
    1. Heneidi S, Simerman AA, Keller E et al. Awakened by cellular stress: Isolation and characterization of a novel population of pluripotent stem cells derived from human adipose tissue. PLoS One 2013;8:e64752.
    1. Gimble JM, Katz AJ, Bunnell BA. Adipose‐derived stem cells for regenerative medicine. Circ Res 2007;100:1249–1260.
    1. Li L, Baroja ML, Majumdar A et al. Human embryonic stem cells possess immune‐privileged properties. STEM CELLS 2004;22:448–456.
    1. Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood 2005;105:1815–1822.
    1. Jarvinen L, Badri L, Wettlaufer S et al. Lung resident mesenchymal stem cells isolated from human lung allografts inhibit T cell proliferation via a soluble mediator. J Immunol 2008;181:4389–4396.
    1. Kim J, Hematti P. Mesenchymal stem cell‐educated macrophages: A novel type of alternatively activated macrophages. Exp Hematol 2009;37:1445–1453.
    1. Mougiakakos D, Jitschin R, Johansson CC et al. The impact of inflammatory licensing on heme oxygenase‐1‐mediated induction of regulatory T cells by human mesenchymal stem cells. Blood 2011;117:4826–4835.
    1. Haskins K, McDuffie M. Acceleration of diabetes in young NOD mice with a CD4+ islet‐specific T cell clone. Science 1990;249:1433–1436.
    1. Stadinski BD, Delong T, Reisdorph N et al. Chromogranin A is an autoantigen in type 1 diabetes. Nat Immunol 2010;11:225–231.
    1. Judkowski V, Pinilla C, Schroder K et al. Identification of MHC class II‐restricted peptide ligands, including a glutamic acid decarboxylase 65 sequence, that stimulate diabetogenic T cells from transgenic BDC2.5 nonobese diabetic mice. J Immunol 2001;166:908–917.
    1. Castro CN, Barcala Tabarrozi AE, Noguerol MA et al. Disease‐modifying immunotherapy for the management of autoimmune diabetes. Neuroimmunomodulation 2010;17:173–176.
    1. Perone MJ, Bertera S, Tawadrous ZS et al. Dendritic cells expressing transgenic galectin‐1 delay onset of autoimmune diabetes in mice. J Immunol 2006;177:5278–5289.
    1. Hong HS, Lee J, Lee E et al. A new role of substance P as an injury‐inducible messenger for mobilization of CD29(+) stromal‐like cells. Nat Med 2009;15:425–435.
    1. Chase LG, Yang S, Zachar V et al. Development and characterization of a clinically compliant xeno‐free culture medium in good manufacturing practice for human multipotent mesenchymal stem cells. STEM CELLS TRANSLATIONAL MEDICINE 2012;1:750–758.
    1. Melief SM, Geutskens SB, Fibbe WE et al. Multipotent stromal cells skew monocytes towards an anti‐inflammatory function: The link with key immunoregulatory molecules. Haematologica 2013;98:e121–e122.
    1. Lichtman SN, Wang J, Lemasters JJ. LPS receptor CD14 participates in release of TNF‐alpha in RAW 264.7 and peritoneal cells but not in Kupffer cells. Am J Physiol 1998;275 1 Pt 1:G39–G46.
    1. Castro CN, Barcala Tabarrozzi AE, Winnewisser J et al. Curcumin ameliorates autoimmune diabetes. Evidence in accelerated murine models of type 1 diabetes. Clin Exp Immunol 2014;177:149–160.
    1. Szabo SJ, Kim ST, Costa GL et al. A novel transcription factor, T‐bet, directs Th1 lineage commitment. Cell 2000;100:655–669.
    1. Yang X, Letterio JJ, Lechleider RJ et al. Targeted disruption of SMAD3 results in impaired mucosal immunity and diminished T cell responsiveness to TGF‐beta. EMBO J 1999;18:1280–1291.
    1. Caplan AI, Correa D. The MSC: An injury drugstore. Cell Stem Cell 2011;9:11–15.
    1. Prockop DJ, Oh JY. Mesenchymal stem/stromal cells (MSCs): Role as guardians of inflammation. Mol Ther 2012;20:14–20.
    1. Hodgetts SI, Beilharz MW, Scalzo AA et al. 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 2000;9:489–502.
    1. Mingliang R, Bo Z, Zhengguo W. Stem cells for cardiac repair: Status, mechanisms, and new strategies. Stem Cells Int 2011;2011:310928.
    1. Bartosh TJ, Ylöstalo JH, Mohammadipoor A et al. Aggregation of human mesenchymal stromal cells (MSCs) into 3D spheroids enhances their antiinflammatory properties. Proc Natl Acad Sci USA 2010;107:13724–13729.
    1. Frith JE, Thomson B, Genever PG. Dynamic three‐dimensional culture methods enhance mesenchymal stem cell properties and increase therapeutic potential. Tissue Eng Part C Methods 2010;16:735–749.
    1. Yeh HY, Liu BH, Hsu SH. The calcium‐dependent regulation of spheroid formation and cardiomyogenic differentiation for MSCs on chitosan membranes. Biomaterials 2012;33:8943–8954.
    1. Stiehler M, Bünger C, Baatrup A et al. Effect of dynamic 3‐D culture on proliferation, distribution, and osteogenic differentiation of human mesenchymal stem cells. J Biomed Mater Res A 2009;89:96–107.
    1. Miao Z, Jin J, Chen L et al. Isolation of mesenchymal stem cells from human placenta: Comparison with human bone marrow mesenchymal stem cells. Cell Biol Int 2006;30:681–687.
    1. Ogura F, Wakao S, Kuroda Y et al. Human adipose tissue possesses a unique population of pluripotent stem cells with nontumorigenic and low telomerase activities: potential implications in regenerative medicine. Stem Cells Dev 2014;23:717–728.
    1. Yamazaki S, Iwama A, Takayanagi S et al. Cytokine signals modulated via lipid rafts mimic niche signals and induce hibernation in hematopoietic stem cells. EMBO J 2006;25:3515–3523.
    1. Ylöstalo JH, Bartosh TJ, Coble K et al. Human mesenchymal stem/stromal cells cultured as spheroids are self‐activated to produce prostaglandin E2 that directs stimulated macrophages into an anti‐inflammatory phenotype. STEM CELLS 2012;30:2283–2296.
    1. Nemeth K, Leelahavanichkul A, Yuen PS et al. Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)‐dependent reprogramming of host macrophages to increase their interleukin‐10 production. Nat Med 2009;15:42–49.
    1. Shi Y, Su JAI, Roberts AI et al. How mesenchymal stem cells interact with tissue immune responses. Trends Immunol 2012;33:136–143.
    1. Sun L, Akiyama K, Zhang H et al. Mesenchymal stem cell transplantation reverses multiorgan dysfunction in systemic lupus erythematosus mice and humans. STEMCELLS 2009;27:1421–1432.
    1. Popp FC, Renner P, Eggenhofer E et al. Mesenchymal stem cells as immunomodulators after liver transplantation. Liver Transpl 2009;15:1192–1198.
    1. Zhou B, Yuan J, Zhou Y et al. Administering human adipose‐derived mesenchymal stem cells to prevent and treat experimental arthritis. Clin Immunol 2011;141:328–337.
    1. Dalal J, Gandy K, Domen J. Role of mesenchymal stem cell therapy in Crohn's disease. Pediatr Res 2012;71:445–451.
    1. Kim H‐S, Yun J‐W, Shin T‐H et al. Human umbilical cord blood mesenchymal stem cell‐derived PGE2 and TGF‐β1 alleviate atopic dermatitis by reducing mast cell degranulation. STEM CELLS 2015;33:1254–1266.
    1. Maxson S, Lopez EA, Yoo D et al. Concise review: Role of mesenchymal stem cells in wound repair. STEM CELLS TRANSLATIONAL MEDICINE 2012;1:142–149.
    1. Rodríguez TM, Saldías A, Irigo M et al. Effect of TGF‐β1 stimulation on the secretome of human adipose‐derived mesenchymal stromal cells. STEM CELLS TRANSLATIONAL MEDICINE 2015;4:894–898.
    1. Patel SA, Meyer JR, Greco SJ et al. Mesenchymal stem cells protect breast cancer cells through regulatory T cells: Role of mesenchymal stem cell‐derived TGF‐β. J Immunol 2010;184:5885–5894.
    1. Kulkarni AB, Huh CG, Becker D et al. Transforming growth factor β1 null mutation in mice causes excessive inflammatory response and early death. Proc Natl Acad Sci USA 1993;90:770–774.
    1. Di Sabatino A, Pickard KM, Rampton D et al. Blockade of transforming growth factor β upregulates T‐box transcription factor T‐bet, and increases T helper cell type 1 cytokine and matrix metalloproteinase‐3 production in the human gut mucosa. Gut 2008;57:605–612.

Source: PubMed

3
Iratkozz fel