Generic placeholder image

Current Stem Cell Research & Therapy

Editor-in-Chief

ISSN (Print): 1574-888X
ISSN (Online): 2212-3946

Review Article

Therapeutic Potential of Amniotic Fluid Derived Mesenchymal Stem Cells Based on their Differentiation Capacity and Immunomodulatory Properties

Author(s): Carl R. Harrell, Marina Gazdic, Crissy Fellabaum, Nemanja Jovicic, Valentin Djonov, Nebojsa Arsenijevic and Vladislav Volarevic*

Volume 14, Issue 4, 2019

Page: [327 - 336] Pages: 10

DOI: 10.2174/1574888X14666190222201749

Price: $65

Abstract

Background: Amniotic Fluid Derived Mesenchymal Stem Cells (AF-MSCs) are adult, fibroblast- like, self-renewable, multipotent stem cells. During the last decade, the therapeutic potential of AF-MSCs, based on their huge differentiation capacity and immunomodulatory characteristics, has been extensively explored in animal models of degenerative and inflammatory diseases.

Objective: In order to describe molecular mechanisms responsible for the therapeutic effects of AFMSCs, we summarized current knowledge about phenotype, differentiation potential and immunosuppressive properties of AF-MSCs.

Methods: An extensive literature review was carried out in March 2018 across several databases (MEDLINE, EMBASE, Google Scholar), from 1990 to present. Keywords used in the selection were: “amniotic fluid derived mesenchymal stem cells”, “cell-therapy”, “degenerative diseases”, “inflammatory diseases”, “regeneration”, “immunosuppression”. Studies that emphasized molecular and cellular mechanisms responsible for AF-MSC-based therapy were analyzed in this review.

Results: AF-MSCs have huge differentiation and immunosuppressive potential. AF-MSCs are capable of generating cells of mesodermal origin (chondrocytes, osteocytes and adipocytes), neural cells, hepatocytes, alveolar epithelial cells, insulin-producing cells, cardiomyocytes and germ cells. AF-MSCs, in juxtacrine or paracrine manner, regulate proliferation, activation and effector function of immune cells. Due to their huge differentiation capacity and immunosuppressive characteristic, transplantation of AFMSCs showed beneficent effects in animal models of degenerative and inflammatory diseases of nervous, respiratory, urogenital, cardiovascular and gastrointestinal system.

Conclusion: Considering the fact that amniotic fluid is obtained through routine prenatal diagnosis, with minimal invasive procedure and without ethical concerns, AF-MSCs represents a valuable source for cell-based therapy of organ-specific or systemic degenerative and inflammatory diseases.

Keywords: Amniotic fluid, mesenchymal stem cells, cell-based therapy, regeneration, immunomodulation, fibroblast.

[1]
Calvin SE, Oyen ML. Microstructure and mechanics of the chorioamnion membrane with an emphasis on fracture propertiesAnn N Y Acad Sci 2007; 1101: 166e85
[2]
Pipino C, Pierdomenico L, Di Tomo P, et al. Molecular and phenotypic characterization of human amniotic fluid-derived cells: A morphological and proteomic approach. Stem Cells Dev 2015; 24(12): 1415-28.
[3]
Savickiene J, Treigyte G, Baronaite S, et al. Human amniotic fluid mesenchymal stem cells from second- and third-trimester amniocentesis: Differentiation potential, molecular signature, and proteome analysis. Stem Cells Int 2015; 2015: 319238.
[4]
Roubelakis MG, Trohatou O, Anagnou NP. Amniotic fluid and amniotic membrane stem cells: Marker discovery. Stem Cells Int 2012; 2012: 107836.
[5]
Eslaminejad MB, Jahangir S. Amniotic fluid stem cells and their application in cell-based tissue regeneration. Int J Fertil Steril 2012; 6: 147-56.
[6]
Moraghebi R, Kirkeby A, Chaves P, et al. Term amniotic fluid: An unexploited reserve of mesenchymal stromal cells for reprogramming and potential cell therapy applications. Stem Cell Res Ther 2017; 8(1): 190.
[7]
Bitsika V, Roubelakis MG, Zagoura D, et al. Human amniotic fluid-derived mesenchymal stem cells as therapeutic vehicles: A novel approach for the treatment of bladder cancer. Stem Cells Dev 2012; 21(7): 1097-111.
[8]
Tsai MS, Lee JL, Chang YJ, Hwang SM. Isolation of human multipotent mesenchymal stem cells from second-trimester amniotic fluid using a novel two-stage culture protocol. Hum Reprod 2004; 19(6): 1450-6.
[9]
Steigman SA, Fauza DO. Isolation of mesenchymal stem cells from amniotic fluid and placentaCurr Protoc Stem Cell Biol 2007; Chapter 1: Unit 1E2.
[10]
In ’t Anker PS, Scherjon SA, Kleijburg-van der Keur C, et al. Isolation of mesenchymal stem cells of fetal or maternal origin from human placenta. Stem Cells 2004; 22: 1338-45.
[11]
Schmidt D, Achermann J, Odermatt B, et al. Prenatally fabricated autologous human living heart valves based on amniotic fluid derived progenitor cells as single cell source. Circulation 2007; 116(11)(Suppl.): I64-70.
[12]
Cananzi M, De Coppi P. CD117(+) amniotic fluid stem cells: state of the art and future perspectives. Organogenesis 2012; 8: 77-88.
[13]
De Coppi P, Bartsch G Jr, Siddiqui MM, et al. Isolation of amniotic stem cell lines with potential for therapy. Nat Biotechnol 2007; 25: 100-6.
[14]
Chen Z, Chan MK, Steichenko N, et al. Heterogeneity of stem cells in human amniotic fluid. J Regen Med 2014; 3: 1-8.
[15]
Savickienė J, Matuzevičius D, Baronaitė S, et al. Histone modifications pattern associated with a state of mesenchymal stem cell cultures derived from amniotic fluid of normal and fetus-affected gestations. J Cell Biochem 2017; 118(11): 3744-55.
[16]
Spitzhorn LS, Rahman MS, Schwindt L, et al. Isolation and molecular characterization of amniotic fluid-derived mesenchymal stem cells obtained from caesarean sections. Stem Cells Int 2017; 2017: 5932706.
[17]
Moschidou D, Mukherjee S, Blundell MP, et al. Valproic acid confers functional pluripotency to human amniotic fluid stem cells in a transgene-free approach Mol Ther 2012; 20: 1953e67
[18]
You Q, Cai L, Zheng J, et al. Isolation of human mesenchymal stem cells from third-trimester amniotic fluid Int J Gynaecol Obstet 2008; 103: 149e52
[19]
Roubelakis MG, Pappa KI, Bitsika V, et al. Molecular and proteomic characterization of human mesenchymal stem cells derived from amniotic fluid: comparison to bone marrow mesenchymal stem cellsStem Cells Dev 2007; 16(6): 931e52
[20]
Zimmermann S, Voss M, Kaiser S, et al. Lack of telomerase activity in human mesenchymal stem cells. Leuk Off J Leuk Soc Am Leuk Res Fund UK 2003; 17(6): 1146-9.
[21]
Roubelakis MG, Bitsika V, Zagoura D, et al. In vitro and in vivo properties of distinct populations of amniotic fluid mesenchymal progenitor cells. J Cell Mol Med 2011; 15(9): 1896-913.
[22]
Chen Z, Jadhav A, Wang F, Perle M, Basch RK, Young B. Senescence and longevity in amniotic fluid derived cells. Stem Cell Discovery 2013; 3: 47-55.
[23]
Carraro G, Perin L, Sedrakyan S, et al. Human amniotic fluid stem cells can integrate and differentiate into epithelial lung lineages. Stem Cells 2008; 26(11): 2902-11.
[24]
Moorefield EC, McKee EE, Solchaga L, et al. Cloned, CD117 selected human amniotic fluid stem cells are capable of modulating the immune response. PLoS One 2011; 6: e26535.
[25]
Moschidou D, Mukherjee S, Blundell MP, et al. Human mid-trimester amniotic fluid stem cells cultured under embryonicstem cell conditions with valproic acid acquire pluripotent characteristicsStem Cells Dev 2013; 22: 444e58
[26]
Joerger-Messerli MS, Marx C, Oppliger B, Mueller M, Surbek DV, Schoeberlein A. Mesenchymal stem cells from wharton’s jelly and amniotic fluid. Best Pract Res Clin Obstet Gynaecol 2016; 31: 30-44.
[27]
Kim J, Lee Y, Kim H, et al. Human amniotic fluid-derived stem cells have characteristics of multipotent stem cells. Cell Prolif 2007; 40: 75-90.
[28]
Prusa AR, Marton E, Rosner M, Bernaschek G, Hengstschlager M. Oct-4-expressing cells in human amniotic fluid: A new source for stem cell research? Hum Reprod 2003; 18(7): 1489-93.
[29]
Klemmt PA, Vafaizadeh V, Groner B. The potential of amniotic fluid stem cells for cellular therapy and tissue engineering. Expert Opin Biol Ther 2011; 11: 1297-314.
[30]
Perin L, Sedrakyan S, Giuliani S, et al. Protective effect of human amniotic fluid stem cells in an immunodeficient mouse model of acute tubular necrosis. PLoS One 2010; 5: e9357.
[31]
Murphy S, Atala A. Amniotic Fluid Stem Cells. In: Cetrulo KJ, Cetrulo CL Jr, Taghizadeh RR, Ed.: Perinatal Stem Cells. New Jerseys Wiley-Blackwell 2013; pp. 1-15
[32]
Zhou J, Wang D, Liang T, Guo Q, Zhang G. Amniotic fluid-derived mesenchymal stem cells: Characteristics and therapeutic applications. Arch Gynecol Obstet 2014; 290(2): 223-31.
[33]
Kunisaki SM, Freedman DA, Fauza DO. Fetal tracheal reconstruction with cartilaginous grafts engineered from mesenchymal amniocytes. J Pediatr Surg 2006; 41(4): 675e82.
[34]
Gray FL, Turner CG, Ahmed A, et al. Prenatal tracheal reconstruction with a hybrid amniotic mesenchymal stem cellsengineered construct derived from decellularized airway. J Pediatr Surg 2012; 47(6): 1072e9.
[35]
Kunisaki SM, Jennings RW, Fauza DO. Fetal cartilage engineering from amniotic mesenchymal progenitor cellsStem Cells Dev 2006; 15(2): 245e53
[36]
Steigman SA, Ahmed A, Shanti RM, et al. Sternal repair with bone grafts engineered from amniotic mesenchymal stem cells. J Pediatr Surg 2009; 44(6): 1120e6.
[37]
Turner CG, Klein JD, Gray FL, et al. Craniofacial repair with fetal bone grafts engineered from amniotic mesenchymal stem cells. J Surg Res 2012; 178(2): 785e90
[38]
Zheng YB, Gao ZL, Xie C, et al. Characterization and hepatogenic differentiation of mesenchymal stem cells from human amniotic fluid and human bone marrow: A comparative study. Cell Biol Int 2008; 32: 1439-48.
[39]
Li Y, Xu W, Yan J, et al. Differentiation of human amniotic fluid-derived mesenchymal stem cells into type II alveolar epithelial cells in vitro. Int J Mol Med 2014; 33(6): 1507-13.
[40]
Mu XP, Ren LQ, Yan HW, et al. Enhanced differentiation of human amniotic fluid-derived stem cells into insulin-producing cells in vitro. J Diabetes Investig 2017; 8(1): 34-43.
[41]
Lai D, Wang F, Chen Y, Wang L, Wang Y, Cheng W. Human amniotic fluid stem cells have a potential to recover ovarian function in mice with chemotherapy-induced sterility. BMC Dev Biol 2013; 13: 34.
[42]
Cipriani S, Bonini D, Marchina E, et al. Mesenchymal cells from human amniotic fluid survive and migrate after transplantation into adult rat brain. Cell Biol Int 2007; 31(8): 845-50.
[43]
Deng J, Petersen BE, Steindler DA, Jorgensen ML, Laywell ED. Mesenchymal stem cells spontaneously express neural proteins in culture and are neurogenic after transplantation. Stem Cells 2006; 24(4): 1054-64.
[44]
Yan ZJ, Hu YQ, Zhang HT, et al. Comparison of the neural differentiation potential of human mesenchymal stem cells from amniotic fluid and adult bone marrow. Cell Mol Neurobiol 2013; 33(4): 465-75.
[45]
Thangnipon W, Puangmalai N, Suwanna N, et al. Potential role of N-benzylcinnamide in inducing neuronal differentiation from human amniotic fluid mesenchymal stem cells. Neurosci Lett 2016; 610: 6-12.
[46]
Soler R, Fullhase C, Hanson A, et al. Stem cell therapy ameliorates bladder dysfunction in an animal model of Parkinson disease. J Urol 2012; 187: 1491e7.
[47]
Pan HC, Yang DY, Chiu YT, et al. Enhanced regeneration in injured sciatic nerve by human amniotic mesenchymal stem cell. J Clin Neurosci 2006; 13(5): 570e5.
[48]
Yang DY, Sheu ML, Su HL, et al. Dual regeneration of muscle and nerve by intravenous administration of human amniotic fluidderived mesenchymal stem cells regulated by stromal cell-derived factor-1alpha in a sciatic nerve injury model. J Neurosurg 2012; 116(6): 1357e67.
[49]
Mohammadian F, Eatemadi A, Daraee H. Application of stem cell for the regeneration of spiral ganglion neurons. Cell Mol Biol(Noisy-le-grand) 2017; 63(1): 6-12.
[50]
Jindal H, Bhatt B, Sk S, Singh Malik J. Alzheimer disease immunotherapeutics: Then and now. Hum Vaccin Immunother 2014; 10(9): 2741-3.
[51]
Jiang P, Dickson DW. Parkinson’s disease: Experimental models and reality. Acta Neuropathol 2018; 135(1): 13-32.
[52]
Chang YJ, Ho TY, Wu ML, et al. Amniotic fluid stem cells with low gamma-interferon response showed behavioral improvement in Parkinsonism rat model. PLoS One 2013; 8: e76118.
[53]
Pan HC, Cheng FC, Chen CJ, et al. Post-injury regeneration in rat sciatic nerve facilitated by neurotrophic factors secreted by amniotic fluid mesenchymal stem cells. J Clin Neurosci 2007; 14(11): 1089e98.
[54]
Dionigi B, Ahmed A, Brazzo J 3rd, et al. Partial or complete coverage of experimental spina bifida by simple intraamniotic injection of concentrated amniotic mesenchymal stem cells. J Pediatr Surg 2015; 50: 69e73
[55]
Liu H, Liu DQ, Li BW, et al. Human amniotic fluid-derived stem cells can differentiate into hepatocyte-like cells in vitro and in vivo. In Vitro Cell Dev Biol Anim 2011; 47: 601-8.
[56]
Zagoura DS, Roubelakis MG, Bitsika V, et al. Therapeutic potential of a distinct population of human amniotic fluid mesenchymal stem cells and their secreted molecules in mice with acute hepatic failure. Gut 2012; 61(6): 894-906.
[57]
Volarevic V, Al-Qahtani A, Arsenijevic N, Pajovic S, Lukic ML. Interleukin-1 receptor antagonist (IL-1Ra) and IL-1Ra producing mesenchymal stem cells as modulators of diabetogenesis. Autoimmunity 2010; 43(4): 255-63.
[58]
Zheng YB, Zhang XH, Huang ZL, et al. Amniotic-fluid-derived mesenchymal stem cells overexpressing interleukin-1 receptor antagonist improve fulminant hepatic failure. PLoS One 2012; 7(7): e41392.
[59]
Li Y, Gu C, Xu W, et al. Therapeutic effects of amniotic fluid-derived mesenchymal stromal cells on lung injury in rats with emphysema. Respir Res 2014; 15: 120.
[60]
Vadasz S, Jensen T, Moncada C, et al. Second and third trimester amniotic fluid mesenchymal stem cells can repopulate a decellularized lung scaffold and express lung markers. J Pediatr Surg 2014; 49(11): 1554e63
[61]
Liu C, Wu H. From Beta cell replacement to beta cell regeneration: Implications for antidiabetic therapy. J Diabetes Sci Technol 2014; 8(6): 1221-6.
[62]
Mu XP, Ren LQ, Yan HW, et al. Enhanced differentiation of human amniotic fluid-derived stem cells into insulin-producing cells in vitro. J Diabetes Investig 2017; 8(1): 34-43.
[63]
Kuhn EN, Wu SM. Origin of cardiac progenitor cells in the developing and postnatal heart. J Cell Physiol 2010; 225(2): 321-5.
[64]
Zhao P, Ise H, Hongo M, Ota M, Konishi I, Nikaido T. Human amniotic mesenchymal cells have some characteristics of cardiomyocytes. Transplantation 2005; 79(5): 528-35.
[65]
Iop L, Chiavegato A, Callegari A, et al. Different cardiovascular potential of adult- and fetal-type mesenchymal stem cells in a rat model of heart cryoinjury. Cell Transplant 2008; 17(6): 679-94.
[66]
Di Trapani M, Bassi G, Ricciardi M, et al. Comparative study of immune regulatory properties of stem cells derived from different tissues Stem Cells Dev 2013; 22: 2990e3002
[67]
Kode JA, Mukherjee S, Joglekar MV, et al. Mesenchymal stem cells: immunobiology and role in immunomodulation and tissue regenerationCytotherapy 2009; 11: 377e91
[68]
Ren G, Zhao X, Zhang L, et al. Inflammatory cytokine-induced intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 in mesenchymal stem cells are critical for immunosuppression. J Immunol 2010; 184(5): 2321-8.
[69]
Mareschi K, Castiglia S, Sanavio F, et al. Immunoregulatory effects on T lymphocytes by human mesenchymal stromal cells isolated from bone marrow, amniotic fluid, and placenta. Exp Hematol 2016; 44(2): 138-50.
[70]
Bright JJ, Kerr LD, Sriram S. TGF-beta inhibits IL-2-induced tyrosine phosphorylation and activation of Jak-1 and Stat 5 in T lymphocytes. J Immunol 1997; 159(1): 175-83.
[71]
Di Nicola M, Carlo-Stella C, Magni M, et al. Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood 2002; 99(10): 3838-43.
[72]
Volarevic V, Gazdic M, Simovic Markovic B, Jovicic N, Djonov V, Arsenijevic N. Mesenchymal stem cell-derived factors: Immuno-modulatory effects and therapeutic potential. Biofactors 2017; 43(5): 633-44.
[73]
Kalinski P. Regulation of immune responses by prostaglandin E2. J Immunol 2012; 188(1): 21-8.
[74]
Kota DJ, Prabhakara KS, Toledano-Furman N, et al. Prostaglandin E2 indicates therapeutic efficacy of mesenchymal stem cells in experimental traumatic brain injury. Stem Cells 2017; 35(5): 1416-30.
[75]
Ren G, Su J, Zhang L, et al. Species variation in the mechanisms of mesenchymal stem cell-mediated immunosuppression. Stem Cells 2009; 27(8): 1954-62.
[76]
Milosavljevic N, Gazdic M, Simovic Markovic B, et al. Mesenchymal stem cells attenuate liver fibrosis by suppressing Th17 cells - an experimental study. Transpl Int 2018; 31(1): 102-15.
[77]
Milosavljevic N, Gazdic M, Simovic Markovic B, et al. Mesenchymal stem cells attenuate acute liver injury by altering ratio between interleukin 17 producing and regulatory natural killer T cells. Liver Transpl 2017; 23(8): 1040-50.
[78]
Yi T, Song SU. Immunomodulatory properties of mesenchymal stem cells and their therapeutic applications. Arch Pharm Res 2012; 35(2): 213-21.
[79]
Markovic BS, Kanjevac T, Harrell CR, et al. Molecular and cellular mechanisms involved in mesenchymal stem cell-based therapy of inflammatory bowel diseases. Stem Cell Rev 2017.
[http://dx.doi.org/10.1007/s12015-017-9789-2]
[80]
Legaki E, Roubelakis MG, Theodoropoulos GE, et al. Therapeutic potential of secreted molecules derived from human amniotic fluid mesenchymal stem/stroma cells in a mice model of colitis. Stem Cell Rev 2016; 12(5): 604-12.
[81]
Zani A, Cananzi M, Fascetti-Leon F, et al. Amniotic fluid stem cells improve survival and enhance repair of damaged intestine in necrotising enterocolitis via a COX-2 dependent mechanism. Gut 2014; 63: 300e9
[82]
Yang DY, Sheu ML, Su HL, et al. Dual regeneration of muscle and nerve by intravenous administration of human amniotic fluidderived mesenchymal stem cells regulated by stromal cell-derived factor-1alpha in a sciatic nerve injury model. J Neurosurg 2012; 116(6): 1357e67.
[83]
Li L, Wang D, Zhou J, Cheng Y, Liang T, Zhang G. Characteristics of human amniotic fluid mesenchymal stem cells and their tropism to human ovarian cancer. PLoS One 2015; 10(4): e0123350.
[84]
Bitsika V, Roubelakis MG, Zagoura D, et al. Human amniotic fluid-derived mesenchymal stem cells as therapeutic vehicles: a novel approach for the treatment of bladder cancer. Stem Cells Dev 2012; 21(7): 1097-111.
[85]
Dong Z, Greene G, Pettaway C, et al. Suppression of angiogenesis, tumorigenicity, and metastasis by human prostate cancer cells engineered to produce interferon-beta. Cancer Res 1999; 59(4): 872-9.
[86]
Qin XQ, Runkel L, Deck C, DeDios C, Barsoum J. Interferon-beta induces S phase accumulation selectively in human transformed cells. J Interferon Cytokine Res 1997; 17(6): 355-67.
[87]
Schiller JH, Storer B, Bittner G, Willson JK, Borden EC. Phase II trial of a combination of interferon-beta ser and interferon-gamma in patients with advanced malignant melanoma. J Interferon Res 1988; 8(5): 581-9.
[88]
Targeted Stem Cells Expressing TRAIL as a Therapy for Lung Cancer (TACTICAL). Available from:. https://clinicaltrials.gov/ct2/ show/NCT03298763 NLM identifier: NCT03298763. (Accessed on May 2, 2018).

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy