Generic placeholder image

Current Stem Cell Research & Therapy

Editor-in-Chief

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

Review Article

Paracrine Interactions Involved in Human Induced Pluripotent Stem Cells Differentiation into Chondrocytes

Author(s): Yunchang Zhao, Honghao Liu, Chunjie Zhao, Peng Dang, Haijian Li* and Maryam Farzaneh*

Volume 15, Issue 3, 2020

Page: [233 - 242] Pages: 10

DOI: 10.2174/1574888X15666191224122058

Price: $65

Abstract

Osteoarthritis (OA), as a degenerative joint disease, is the most common form of joint disorder that causes pain, stiffness, and other symptoms associated with OA. Various genetic, biomechanical, and environmental factors have a relevant role in the development of OA. To date, extensive efforts are currently being made to overcome the poor self-healing capacity of articular cartilage. Despite the pivotal role of chondrocytes, their proliferation and repair capacity after tissue injury are limited. Therefore, the development of new strategies to overcome these constraints is urgently needed. Recent advances in regenerative medicine suggest that pluripotent stem cells are promising stem cell sources for cartilage repair. Pluripotent stem cells are undifferentiated cells that have the capacity to differentiate into different types of cells and can self-renew indefinitely. In the past few decades, numerous attempts have been made to regenerate articular cartilage by using induced pluripotent stem cells (iPSCs). The potential applications of patient-specific iPSCs hold great promise for regenerative medicine and OA treatment. However, there are different culture conditions for the preparation and characterization of human iPSCs-derived chondrocytes (hiChondrocytes). Recent biochemical analyses reported that several paracrine factors such as TGFb, BMPs, WNT, Ihh, and Runx have been shown to be involved in cartilage cell proliferation and differentiation from human iPSCs. In this review, we summarize and discuss the paracrine interactions involved in human iPSCs differentiation into chondrocytes in different cell culture media.

Keywords: Osteoarthritis, pluripotent stem cells, induced pluripotent stem cells, chondrocytes, paracrine mechanisms, germ layers.

[1]
Luyten F, Bierma-Zeinstra S, Dell’Accio F, Kraus V, Nakata K, Sekiya I, Eds. Toward classification criteria for early osteoarthritis of the knee Seminars in arthritis and rheumatism. Elsevier 2018.
[2]
Hart HF, Gross KD, Crossley KM, Barton CJ, Felson DT, Guermazi A, et al. Is step rate associated with worsening of patellofemoral and tibiofemoral joint osteoarthritis in women and men? The multicenter osteoarthritis study. Arthritis Care Res 2019.
[3]
Lee WY, Wang B. Cartilage repair by mesenchymal stem cells: Clinical trial update and perspectives. J Orthop Translat 2017; 9: 76-88.
[http://dx.doi.org/10.1016/j.jot.2017.03.005] [PMID: 29662802]
[4]
Man GS, Mologhianu G. Osteoarthritis pathogenesis - a complex process that involves the entire joint. J Med Life 2014; 7(1): 37-41.
[PMID: 24653755]
[5]
Kohn MD, Sassoon AA, Fernando ND. Classifications in brief: Kellgren-Lawrence classification of osteoarthritis. Springer 2016.
[6]
Blanco FJ. Clinical features and diagnosis of osteoarthritis Atlas of Osteoarthritis 2018; 52.
[7]
Sandler RD, Dunkley L. Osteoarthritis and the inflammatory arthritides. Surgery 2018; 36(1): 21-6.
[PMID: 29636101]
[8]
Hellevik AI, Johnsen MB, Langhammer A, et al. Metabolic syndrome as a risk factor for total hip or knee replacement due to primary osteoarthritis: a prospective cohort study (the HUNT study and the Norwegian Arthroplasty Register). Clin Epidemiol 2018; 10: 83-96.
[http://dx.doi.org/10.2147/CLEP.S145823] [PMID: 29391831]
[9]
Johnson VL, Hunter DJ. The epidemiology of osteoarthritis. Best Pract Res Clin Rheumatol 2014; 28(1): 5-15.
[http://dx.doi.org/10.1016/j.berh.2014.01.004] [PMID: 24792942]
[10]
Mckevitt S, Healey E, Jinks C, Rathod-Mistry T, Quicke J. The association between comorbidity and physical activity levels in people with osteoarthritis: secondary analysis from two randomised controlled trials. Osteoarthritis Cartilage 2019; 27: S494.
[http://dx.doi.org/10.1016/j.joca.2019.02.551]
[11]
Vincent HK, Heywood K, Connelly J, Hurley RW. Obesity and weight loss in the treatment and prevention of osteoarthritis. PM R 2012; 4(5)(Suppl.): S59-67.
[http://dx.doi.org/10.1016/j.pmrj.2012.01.005] [PMID: 22632704]
[12]
Duarte Campos DF, Drescher W, Rath B, Tingart M, Fischer H. Supporting Biomaterials for Articular Cartilage Repair. Cartilage 2012; 3(3): 205-21.
[http://dx.doi.org/10.1177/1947603512444722] [PMID: 26069634]
[13]
Musumeci G, Castrogiovanni P, Leonardi R, et al. New perspectives for articular cartilage repair treatment through tissue engineering: A contemporary review. World J Orthop 2014; 5(2): 80-8.
[http://dx.doi.org/10.5312/wjo.v5.i2.80] [PMID: 24829869]
[14]
Akkiraju H, Nohe A. Role of Chondrocytes in Cartilage Formation, Progression of Osteoarthritis and Cartilage Regeneration. J Dev Biol 2015; 3(4): 177-92.
[http://dx.doi.org/10.3390/jdb3040177] [PMID: 27347486]
[15]
Caldwell KL, Wang J. Cell-based articular cartilage repair: the link between development and regeneration. Osteoarthritis Cartilage 2015; 23(3): 351-62.
[http://dx.doi.org/10.1016/j.joca.2014.11.004] [PMID: 25450846]
[16]
Simon TM, Jackson DW. Articular cartilage: injury pathways and treatment options. Sports Med Arthrosc Rev 2018; 26(1): 31-9.
[http://dx.doi.org/10.1097/JSA.0000000000000182] [PMID: 29300225]
[17]
Martín AR, Patel JM, Zlotnick HM, Carey JL, Mauck RL. Emerging therapies for cartilage regeneration in currently excluded ‘red knee’ populations. NPJ Regen Med 2019; 4: 12.
[http://dx.doi.org/10.1038/s41536-019-0074-7] [PMID: 31231546]
[18]
Gregori D, Giacovelli G, Minto C, et al. Association of pharmacological treatments with long-term pain control in patients with knee osteoarthritis: a systematic review and meta-analysis. JAMA 2018; 320(24): 2564-79.
[http://dx.doi.org/10.1001/jama.2018.19319] [PMID: 30575881]
[19]
Majeed MH, Sherazi SAA, Bacon D, Bajwa ZH. Pharmacological treatment of pain in osteoarthritis: a descriptive review. Curr Rheumatol Rep 2018; 20(12): 88.
[http://dx.doi.org/10.1007/s11926-018-0794-5] [PMID: 30465131]
[20]
Nakayama N, Lee JY, Matthias N, Umeda K, Yan Q, Huard J. Cartilage regeneration using pluripotent stem cell-derived chondroprogenitors: promise and challenges Pluripotent Stem Cells. INTECH 2016; pp. 385-425.
[21]
Guzzo RM, O’Sullivan MB. Human pluripotent stem cells: advances in chondrogenic differentiation and articular cartilage regeneration. Curr Mol Biol Rep 2016; 2(3): 113-22.
[http://dx.doi.org/10.1007/s40610-016-0041-7]
[22]
Gibson JD, O’Sullivan MB, Alaee F, et al. Regeneration of articular cartilage by human esc‐derived mesenchymal progenitors treated sequentially with BMP‐2 and Wnt5a. Stem Cells Transl Med 2017; 6(1): 40-50.
[http://dx.doi.org/10.5966/sctm.2016-0020] [PMID: 28170184]
[23]
Wei Y, Zeng W, Wan R, et al. Chondrogenic differentiation of induced pluripotent stem cells from osteoarthritic chondrocytes in alginate matrix. Eur Cell Mater 2012; 23(1): 1-12.
[http://dx.doi.org/10.22203/eCM.v023a01] [PMID: 22241609]
[24]
Ko J-Y, Kim K-I, Park S, Im G-I. In vitro chondrogenesis and in vivo repair of osteochondral defect with human induced pluripotent stem cells. Biomaterials 2014; 35(11): 3571-81.
[http://dx.doi.org/10.1016/j.biomaterials.2014.01.009] [PMID: 24462354]
[25]
Kafienah W, Owaidah A, Al-Obaidi A. Induced pluripotent stem cells in cartilage repair Rad Hrvatske akademije znanosti i umjetnosti: Medicinske znanosti 2016; (526= 43): 56.
[26]
Guzzo RM, Drissi H. Differentiation of human induced pluripotent stem cells to chondrocytes Cartilage Tissue Engineering. Springer 2015; pp. 79-95.
[27]
Tsumaki N, Okada M, Yamashita A. iPS cell technologies and cartilage regeneration. Bone 2015; 70: 48-54.
[http://dx.doi.org/10.1016/j.bone.2014.07.011] [PMID: 25026496]
[28]
Abdal Dayem A, Lee SB, Kim K, et al. Production of mesenchymal stem cells through stem cell reprogramming. Int J Mol Sci 2019; 20(8): 1922.
[http://dx.doi.org/10.3390/ijms20081922] [PMID: 31003536]
[29]
Zhu Y, Wang Y, Zhao B, et al. Comparison of exosomes secreted by induced pluripotent stem cell-derived mesenchymal stem cells and synovial membrane-derived mesenchymal stem cells for the treatment of osteoarthritis. Stem Cell Res Ther 2017; 8(1): 64.
[http://dx.doi.org/10.1186/s13287-017-0510-9] [PMID: 28279188]
[30]
Huang Y-Z, Xie H-Q, Silini A, et al. Mesenchymal stem/progenitor cells derived from articular cartilage, synovial membrane and synovial fluid for cartilage regeneration: current status and future perspectives. Stem Cell Rev Rep 2017; 13(5): 575-86.
[http://dx.doi.org/10.1007/s12015-017-9753-1] [PMID: 28721683]
[31]
Farzaneh M, Alishahi M, Derakhshan Z, Sarani NH, Attari F, Khoshnam SE. The expression and functional roles of miRNAs in embryonic and lineage-specific stem cells. Curr Stem Cell Res Ther 2019; 14(3): 278-89.
[http://dx.doi.org/10.2174/1574888X14666190123162402] [PMID: 30674265]
[32]
Wang G, Farzaneh M. Mini review; differentiation of human pluripotent stem cells into oocytes. Curr Stem Cell Res Ther 2020.
[http://dx.doi.org/10.2174/1574888X15666200116100121] [PMID: 31951188]
[33]
Farzaneh M, Attari F, Khoshnam SE. Concise review: LIN28/let-7 signaling, a critical double-negative feedback loop during pluripotency, reprogramming, and Tumorigenicity. Cell Reprogram 2017; 19(5): 289-93.
[http://dx.doi.org/10.1089/cell.2017.0015] [PMID: 28846452]
[34]
Ha CW, Park YB, Kim SH, Lee HJ. Intra-articular Mesenchymal Stem Cells in Osteoarthritis of the Knee: A Systematic Review of Clinical Outcomes and Ev-idence of Cartilage Repair Arthroscopy: The journal of arthroscopic & related surgery : official publication of the Arthroscopy Association of North America and the International Arthroscopy Association 2019 35(1): 277-88.
[http://dx.doi.org/10.1016/j.arthro.2018.07.028]
[35]
Chahla J, Piuzzi NS, Mitchell JJ, et al. Intra-Articular Cellular Therapy for Osteoarthritis and Focal Cartilage Defects of the Knee: A Systematic Review of the Literature and Study Quality Analysis. J Bone Joint Surg Am 2016; 98(18): 1511-21.
[http://dx.doi.org/10.2106/JBJS.15.01495] [PMID: 27655978]
[36]
Ha C-W, Park Y-B, Kim SH, Lee H-J. Intra-articular mesenchymal stem cells in osteoarthritis of the knee: a systematic review of clinical outcomes and evidence of cartilage repair. Arthroscopy 2018.
[PMID: 30455086]
[37]
Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007; 131(5): 861-72.
[http://dx.doi.org/10.1016/j.cell.2007.11.019] [PMID: 18035408]
[38]
Schmidt R, Plath K. The roles of the reprogramming factors Oct4, Sox2 and Klf4 in resetting the somatic cell epigenome during induced pluripotent stem cell generation. Genome Biol 2012; 13(10): 251.
[http://dx.doi.org/10.1186/gb-2012-13-10-251] [PMID: 23088445]
[39]
Fu K, Chronis C, Soufi A, et al. Comparison of reprogramming factor targets reveals both species-specific and conserved mechanisms in early iPSC reprogramming. BMC Genomics 2018; 19(1): 956.
[http://dx.doi.org/10.1186/s12864-018-5326-1] [PMID: 30577748]
[40]
Takahashi K, Yamanaka S. A decade of transcription factor-mediated reprogramming to pluripotency. Nat Rev Mol Cell Biol 2016; 17(3): 183-93.
[http://dx.doi.org/10.1038/nrm.2016.8] [PMID: 26883003]
[41]
Castro-Viñuelas R, Sanjurjo-Rodríguez C, Piñeiro-Ramil M, et al. Induced pluripotent stem cells for cartilage repair: current status and future perspectives. Eur Cell Mater 2018; 36: 96-109.
[http://dx.doi.org/10.22203/eCM.v036a08] [PMID: 30204229]
[42]
Lewandowski J, Kurpisz M. Techniques of Human Embryonic Stem Cell and Induced Pluripotent Stem Cell Derivation. Arch Immunol Ther Exp (Warsz) 2016; 64(5): 349-70.
[http://dx.doi.org/10.1007/s00005-016-0385-y] [PMID: 26939778]
[43]
Calloni R, Cordero EAA, Henriques JAP, Bonatto D. Reviewing and updating the major molecular markers for stem cells. Stem Cells Dev 2013; 22(9): 1455-76.
[http://dx.doi.org/10.1089/scd.2012.0637] [PMID: 23336433]
[44]
Muchkaeva IA, Dashinimaev EB, Terskikh VV, Sukhanov YV, Vasiliev AV. Molecular mechanisms of induced pluripotency. Acta Naturae 2012; 4(1): 12-22.
[http://dx.doi.org/10.32607/20758251-2012-4-1-12-22] [PMID: 22708059]
[45]
Farzaneh M, Derakhshan Z, Hallajzadeh J, Sarani NH, Nejabatdoust A, Khoshnam SE. Suppression of TGF-β and ERK Signaling Pathways as a New Strategy to Provide Rodent and Non-Rodent Pluripotent Stem Cells. Curr Stem Cell Res Ther 2019; 14(6): 466-73.
[http://dx.doi.org/10.2174/1871527318666190314110529] [PMID: 30868962]
[46]
Weissenberger M, Weissenberger MH, Gilbert F, Groll J, Evans CH, Steinert AF. Reduced hypertrophy in vitro after chondrogenic differentiation of adult human mesenchymal stem cells following adenoviral SOX9 gene delivery. BMC Musculoskelet Disord 2020; 21(1): 109.
[http://dx.doi.org/10.1186/s12891-020-3137-4] [PMID: 32066427]
[47]
Sheyn D, Ben-David S, Shapiro G, et al. Human Induced Pluripotent Stem Cells Differentiate Into Functional Mesenchymal Stem Cells and Repair Bone Defects. Stem Cells Transl Med 2016; 5(11): 1447-60.
[http://dx.doi.org/10.5966/sctm.2015-0311] [PMID: 27400789]
[48]
Bezenah JR, Kong YP, Putnam AJ. Evaluating the potential of endothelial cells derived from human induced pluripotent stem cells to form microvascular networks in 3D cultures. Sci Rep 2018; 8(1): 2671.
[http://dx.doi.org/10.1038/s41598-018-20966-1] [PMID: 29422650]
[49]
Smith SS, Lila T, Trautman J, Blatz A. A Fast Optical Ion Channel Assay for Assessing Action Potentials in Human Induced Pluripotent Stem Cell Cardiomyocytes. Biophys J 2018; 114(3): 625a.
[http://dx.doi.org/10.1016/j.bpj.2017.11.3376]
[50]
Nelakanti RV, Kooreman NG, Wu JC. Teratoma formation: A tool for monitoring pluripotency in stem cell research Curr Protoc Stem Cell Biol 2015; 32: 4A.8.1-4A.8.17.
[http://dx.doi.org/10.1002/9780470151808.sc04a08s32]
[51]
Gutierrez-Aranda I, Ramos-Mejia V, Bueno C, et al. Human induced pluripotent stem cells develop teratoma more efficiently and faster than human embryonic stem cells regardless the site of injection. Stem Cells 2010; 28(9): 1568-70.
[http://dx.doi.org/10.1002/stem.471] [PMID: 20641038]
[52]
Lo B, Parham L. Ethical issues in stem cell research. Endocr Rev 2009; 30(3): 204-13.
[http://dx.doi.org/10.1210/er.2008-0031] [PMID: 19366754]
[53]
de Wert G, Mummery C. Human embryonic stem cells: research, ethics and policy. Hum Reprod 2003; 18(4): 672-82.
[http://dx.doi.org/10.1093/humrep/deg143] [PMID: 12660256]
[54]
Pearl JI, Kean LS, Davis MM, Wu JC. Pluripotent stem cells: Immune to the immune system?. Science translational medicine 2012; 4(164): 164ps25-ps25.
[http://dx.doi.org/10.1126/scitranslmed.3005090]
[55]
C Watt J. R Kobayashi N. The bioethics of human pluripotent stem cells: will induced pluripotent stem cells end the debate? Open Stem Cell J 2010; 2(1)
[56]
Volarevic V, Markovic BS, Gazdic M, et al. Ethical and Safety Issues of Stem Cell-Based Therapy. Int J Med Sci 2018; 15(1): 36-45.
[http://dx.doi.org/10.7150/ijms.21666] [PMID: 29333086]
[57]
Kolagar TA, Farzaneh M, Nikkar N, Anbiyaiee A, Heydari E, Khoshnam SE. Human Pluripotent Stem Cells in Neurodegenerative Diseases: Potentials, Advances, and Limitations. Curr Stem Cell Res Ther 2019.
[http://dx.doi.org/10.2174/1574888X14666190823142911] [PMID: 31441732]
[58]
Marks R. Articular Cartilage Regeneration: An Update of Possible Treatment Approaches. Int J Orthod 2017; 4(4): 770-8.
[http://dx.doi.org/10.17554/j.issn.2311-5106.2017.04.221]
[59]
Iturriaga L, Hernáez-Moya R, Erezuma I, Dolatshahi-Pirouz A, Orive G. Advances in stem cell therapy for cartilage regeneration in osteoarthritis. Expert Opin Biol Ther 2018; 18(8): 883-96.
[http://dx.doi.org/10.1080/14712598.2018.1502266] [PMID: 30020816]
[60]
Murphy C, Mobasheri A, Táncos Z, Kobolák J, Dinnyés A. The potency of induced pluripotent stem cells in cartilage regeneration and osteoarthritis treatment Cell Biology and Translational Medicine. Springer 2017; Vol. 1: pp. 55-68.
[61]
Toh WS. Pluripotent stem cells: differentiation potential and therapeutic efficacy for cartilage repair. Pluripotent Stem Cells-From the Bench to the Clinic 2016.
[62]
Wang R, Xu B, Xu H. TGF-β1 promoted chondrocyte proliferation by regulating Sp1 through MSC-exosomes derived miR-135b. Cell Cycle 2018; 17(24)
[http://dx.doi.org/10.1080/15384101.2018.1556063] [PMID: 30526325]
[63]
Li J, Zhao Z, Liu J, et al. MEK/ERK and p38 MAPK regulate chondrogenesis of rat bone marrow mesenchymal stem cells through delicate interaction with TGF-β1/Smads pathway. Cell Prolif 2010; 43(4): 333-43.
[http://dx.doi.org/10.1111/j.1365-2184.2010.00682.x] [PMID: 20590658]
[64]
Xu R, Li J, Wei B, Huo W, Wang L. MicroRNA-483-5p modulates the expression of cartilage-related genes in human chondrocytes through down-regulating TGF-β1 expression. Tohoku J Exp Med 2017; 243(1): 41-8.
[http://dx.doi.org/10.1620/tjem.243.41] [PMID: 28924102]
[65]
Wu C, Jiao H, Lai Y, et al. Kindlin-2 controls TGF-β signalling and Sox9 expression to regulate chondrogenesis. Nat Commun 2015; 6: 7531.
[http://dx.doi.org/10.1038/ncomms8531] [PMID: 26151572]
[66]
Guo L, Cai T, Chen K, et al. Kindlin-2 regulates mesenchymal stem cell differentiation through control of YAP1/TAZ. J Cell Biol 2018; 217(4): 1431-51.
[http://dx.doi.org/10.1083/jcb.201612177] [PMID: 29496737]
[67]
Zhou N, Li Q, Lin X, et al. BMP2 induces chondrogenic differentiation, osteogenic differentiation and endochondral ossification in stem cells. Cell Tissue Res 2016; 366(1): 101-11.
[http://dx.doi.org/10.1007/s00441-016-2403-0] [PMID: 27083447]
[68]
Karolak MR, Yang X, Elefteriou F. FGFR1 signaling in hypertrophic chondrocytes is attenuated by the Ras-GAP neurofibromin during endochondral bone formation. Hum Mol Genet 2015; 24(9): 2552-64.
[http://dx.doi.org/10.1093/hmg/ddv019] [PMID: 25616962]
[69]
Horita M, Nishida K, Hasei J, et al. Involvement of ADAM12 in Chondrocyte Differentiation by Regulation of TGF-β1-Induced IGF-1 and RUNX-2 Expressions. Calcif Tissue Int 2019; 105(1): 97-106.
[http://dx.doi.org/10.1007/s00223-019-00549-6] [PMID: 30993375]
[70]
Zhang Y, Yang TL, Li X, Guo Y. Functional analyses reveal the essential role of SOX6 and RUNX2 in the communication of chondrocyte and osteoblast Osteoporosis international : A journal established as result of cooperation between the European Foundation for Osteopo-rosis and the National Osteoporosis Foundation of the USA 2015 26(2): 553-61.
[http://dx.doi.org/10.1007/s00198-014-2882-3]
[71]
Fujita T, Azuma Y, Fukuyama R, et al. Runx2 induces osteoblast and chondrocyte differentiation and enhances their migration by coupling with PI3K-Akt signaling. J Cell Biol 2004; 166(1): 85-95.
[http://dx.doi.org/10.1083/jcb.200401138] [PMID: 15226309]
[72]
Kim E-J, Cho S-W, Shin J-O, Lee M-J, Kim K-S, Jung H-S. Ihh and Runx2/Runx3 signaling interact to coordinate early chondrogenesis: a mouse model. PLoS One 2013; 8(2) e55296
[http://dx.doi.org/10.1371/journal.pone.0055296] [PMID: 23383321]
[73]
Narcisi R, Cleary MA, Brama PA, et al. Long-term expansion, enhanced chondrogenic potential, and suppression of endochondral ossification of adult human MSCs via WNT signaling modulation. Stem Cell Reports 2015; 4(3): 459-72.
[http://dx.doi.org/10.1016/j.stemcr.2015.01.017] [PMID: 25733021]
[74]
Rodrigues M, Griffith LG, Wells A. Growth factor regulation of proliferation and survival of multipotential stromal cells. Stem Cell Res Ther 2010; 1(4): 32.
[http://dx.doi.org/10.1186/scrt32] [PMID: 20977782]
[75]
Suchorska WM, Augustyniak E, Richter M, Trzeciak T. Gene expression profile in human induced pluripotent stem cells: Chondrogenic differentiation in vitro, part A. Mol Med Rep 2017; 15(5): 2387-401.
[http://dx.doi.org/10.3892/mmr.2017.6334] [PMID: 28447755]
[76]
Kakar S, Einhorn TA, Vora S, et al. Enhanced chondrogenesis and Wnt signaling in PTH-treated fractures. J Bone Miner Res 2007; 22(12): 1903-12.
[http://dx.doi.org/10.1359/jbmr.070724] [PMID: 17680724]
[77]
Kovermann NJ, Basoli V, Della Bella E, et al. BMP2 and TGF-β Cooperate Differently during Synovial-Derived Stem-Cell Chondrogenesis in a Dexamethasone-Dependent Manner. Cells 2019; 8(6): 636.
[http://dx.doi.org/10.3390/cells8060636] [PMID: 31242641]
[78]
Huang H, Xu H, Zhang J. Current Tissue Engineering Approaches for Cartilage Regen-eration Cartilage Tissue Engineering and Regeneration Techniques: IntechOpen 2019.
[http://dx.doi.org/10.5772/intechopen.84429]
[79]
Yamashita A, Tamamura Y, Morioka M, Karagiannis P, Shima N, Tsumaki N. Considerations in hiPSC-derived cartilage for articular cartilage repair. Inflamm Regen 2018; 38: 17.
[http://dx.doi.org/10.1186/s41232-018-0075-8] [PMID: 30305854]
[80]
Suchorska WM, Augustyniak E, Richter M, Trzeciak T. Comparison of Four Protocols to Generate Chondrocyte-Like Cells from Human Induced Pluripotent Stem Cells (hiPSCs). Stem Cell Rev Rep 2017; 13(2): 299-308.
[http://dx.doi.org/10.1007/s12015-016-9708-y] [PMID: 27987073]
[81]
Adkar SS, Wu C-L, Willard VP, et al. Highly efficient chondrogenic differentiation of human iPSCs and purification via a reporter allele generated by CRISPR-Cas9 genome editing. bioRxiv 2018; 37(1) 252767
[82]
Nakayama N, Pothiawala A, Lee JY, et al. Human pluripotent stem cell-derived chondroprogenitors for cartilage tissue engineering. Cell Mol Life Sci 2020. Epub ahead of print
[http://dx.doi.org/10.1007/s00018-019-03445-2] [PMID: 31915836]
[83]
Suchorska WM, Augustyniak E, Richter M, Trzeciak T. Comparison of Four Protocols to Generate Chondrocyte-Like Cells from Human Induced Pluripotent Stem Cells (hiPSCs). Stem cell reviews and reports 2017; 13(2): 299-308.
[84]
Stelcer E, Kulcenty K, Rucinski M, et al. Chondrogenic differentiation in vitro of hiPSCs activates pathways engaged in limb development. Stem Cell Res (Amst) 2018; 30: 53-60.
[http://dx.doi.org/10.1016/j.scr.2018.05.006] [PMID: 29783101]
[85]
Medvedev SP, Grigor’eva EV, Shevchenko AI, et al. Human induced pluripotent stem cells derived from fetal neural stem cells successfully undergo directed differentiation into cartilage. Stem Cells Dev 2011; 20(6): 1099-112.
[http://dx.doi.org/10.1089/scd.2010.0249] [PMID: 20846027]
[86]
Koyama N, Miura M, Nakao K, et al. Human induced pluripotent stem cells differentiated into chondrogenic lineage via generation of mesenchymal progenitor cells. Stem Cells Dev 2013; 22(1): 102-13.
[http://dx.doi.org/10.1089/scd.2012.0127] [PMID: 22817676]
[87]
Nejadnik H, Diecke S, Lenkov OD, et al. Improved approach for chondrogenic differentiation of human induced pluripotent stem cells. Stem Cell Rev Rep 2015; 11(2): 242-53.
[http://dx.doi.org/10.1007/s12015-014-9581-5] [PMID: 25578634]
[88]
Yamashita A, Morioka M, Yahara Y, et al. Generation of scaffoldless hyaline cartilaginous tissue from human iPSCs. Stem Cell Reports 2015; 4(3): 404-18.
[http://dx.doi.org/10.1016/j.stemcr.2015.01.016] [PMID: 25733017]
[89]
Zhu Y, Wu X, Liang Y, et al. Repair of cartilage defects in osteoarthritis rats with induced pluripotent stem cell derived chondrocytes. BMC Biotechnol 2016; 16(1): 78.
[http://dx.doi.org/10.1186/s12896-016-0306-5] [PMID: 27829414]
[90]
Li J, Dong S. The signaling pathways involved in chondrocyte differentia-tion and hypertrophic differentiation. Stem Cells Int 2016.
[http://dx.doi.org/10.1155/2016/2470351]
[91]
Steinert AF, Proffen B, Kunz M, et al. Hypertrophy is induced during the in vitro chondrogenic differentiation of human mesenchymal stem cells by bone morphogenetic protein-2 and bone morphogenetic protein-4 gene transfer. Arthritis Res Ther 2009; 11(5): R148.
[http://dx.doi.org/10.1186/ar2822] [PMID: 19799789]
[92]
Driessen BJH, Logie C, Vonk LA. Cellular reprogramming for clinical cartilage repair. Cell Biol Toxicol 2017; 33(4): 329-49.
[http://dx.doi.org/10.1007/s10565-017-9382-0] [PMID: 28144824]
[93]
Lee J, Smeriglio P, Chu CR, Bhutani N. Human iPSC-derived chondrocytes mimic juvenile chondrocyte function for the dual advantage of increased proliferation and resistance to IL-1β. Stem Cell Res Ther 2017; 8(1): 244.
[http://dx.doi.org/10.1186/s13287-017-0696-x] [PMID: 29096706]
[94]
Dehne T, Karlsson C, Ringe J, Sittinger M, Lindahl A. Chondrogenic differentiation potential of osteoarthritic chondrocytes and their possible use in matrix-associated autologous chondrocyte transplantation. Arthritis Res Ther 2009; 11(5): R133.
[http://dx.doi.org/10.1186/ar2800] [PMID: 19723327]
[95]
Augustyniak E, Trzeciak T, Richter M, Kaczmarczyk J, Suchorska W. The role of growth factors in stem cell-directed chondrogenesis: a real hope for damaged cartilage regeneration. Int Orthop 2015; 39(5): 995-1003.
[http://dx.doi.org/10.1007/s00264-014-2619-0] [PMID: 25512139]
[96]
Mueller MB, Tuan RS. Functional characterization of hypertrophy in chondrogenesis of human mesenchymal stem cells. Arthritis Rheum 2008; 58(5): 1377-88.
[http://dx.doi.org/10.1002/art.23370] [PMID: 18438858]
[97]
Lach MS, Wroblewska J, Kulcenty K, Richter M, Trzeciak T, Suchorska WM. Chondrogenic Differentiation of Pluripotent Stem Cells under Controllable Serum-Free Conditions. Int J Mol Sci 2019; 20(11): 2711.
[http://dx.doi.org/10.3390/ijms20112711] [PMID: 31159483]
[98]
Rana D, Kumar S, Webster TJ, Ramalingam M. Impact of Induced Pluripotent Stem Cells in Bone Repair and Regeneration. Curr Osteoporos Rep 2019; 17(4): 226-34.
[http://dx.doi.org/10.1007/s11914-019-00519-9] [PMID: 31256323]
[99]
Dicks A, Wu C-L, Steward N, Adkar SS, Gersbach CA, Guilak F. Prospective isolation of chondroprogenitors from human iPSCs based on cell surface markers identified using a CRISPR-Cas9-generated. Reporter bioRxiv 2019; 11(1): 675983.
[100]
Joseph JS, Malindisa ST, Ntwasa M. Two-Dimensional (2D) and Three-Dimensional (3D) Cell Culturing in Drug Discovery. Cell Culture 2018; 2: 1-22.
[101]
Centeno EGZ, Cimarosti H, Bithell A. 2D versus 3D human induced pluripotent stem cell-derived cultures for neurodegenerative disease modelling. Mol Neurodegener 2018; 13(1): 27.
[http://dx.doi.org/10.1186/s13024-018-0258-4] [PMID: 29788997]
[102]
Kim MJ, Son MJ, Son MY, et al. Generation of human induced pluripotent stem cells from osteoarthritis patient-derived synovial cells. Arthritis Rheum 2011; 63(10): 3010-21.
[http://dx.doi.org/10.1002/art.30488] [PMID: 21953087]
[103]
Limraksasin P, Kondo T, Zhang M, et al. In vitro fabrication of hybrid bone/cartilage complex using mouse induced pluripotent stem cells. Int J Mol Sci 2020; 21(2): 581.
[http://dx.doi.org/10.3390/ijms21020581] [PMID: 31963264]
[104]
Liu J, Nie H, Xu Z, et al. The effect of 3D nanofibrous scaffolds on the chondrogenesis of induced pluripotent stem cells and their application in restoration of cartilage defects. PLoS One 2014; 9(11)e111566
[http://dx.doi.org/10.1371/journal.pone.0111566] [PMID: 25389965]
[105]
Zhao P, Gu H, Mi H, Rao C, Fu J, Turng L-s. Fabrication of scaffolds in tissue engineering: A review. Front Mech Eng 2018; 13(1): 107-19.
[http://dx.doi.org/10.1007/s11465-018-0496-8]
[106]
Guo B, Ma PX. Conducting polymers for tissue engineering. Biomacromolecules 2018; 19(6): 1764-82.
[http://dx.doi.org/10.1021/acs.biomac.8b00276] [PMID: 29684268]
[107]
Choi JR, Yong KW, Choi JY. Effects of mechanical loading on human mesenchymal stem cells for cartilage tissue engineering. J Cell Physiol 2018; 233(3): 1913-28.
[http://dx.doi.org/10.1002/jcp.26018] [PMID: 28542924]
[108]
Nath S, Devi GR. Three-dimensional culture systems in cancer research: Focus on tumor spheroid model. Pharmacol Ther 2016; 163: 94-108.
[http://dx.doi.org/10.1016/j.pharmthera.2016.03.013] [PMID: 27063403]
[109]
Liu Z, Tang M, Zhao J, Chai R, Kang J. Looking into the Future: Toward Advanced 3D Biomaterials for Stem-Cell-Based Regenerative Medicine. Adv Mater 2018; 30(17)e1705388
[http://dx.doi.org/10.1002/adma.201705388] [PMID: 29450919]
[110]
Mahboudi H, Soleimani M, Enderami SE, et al. The effect of nanofibre-based polyethersulfone (PES) scaffold on the chondrogenesis of human induced pluripotent stem cells. Artif Cells Nanomed Biotechnol 2018; 46(8): 1948-56.
[PMID: 29103309]
[111]
Arora A, Bhattacharjee A, Mahajan A, Katti DS. Cartilage Tissue Engineering: Scaffold, Cell, and Growth Factor-Based Strategies Regenerative Medicine: Laboratory to Clinic. Springer 2017; pp. 233-57.
[112]
Park S, Im G-I. Embryonic stem cells and induced pluripotent stem cells for skeletal regeneration. Tissue Eng Part B Rev 2014; 20(5): 381-91.
[http://dx.doi.org/10.1089/ten.teb.2013.0530] [PMID: 24206162]
[113]
Nam Y, Rim YA, Lee J, Ju JH. Current therapeutic strategies for stem cell-based cartilage re-generation. Stem Cells Int 2018.
[http://dx.doi.org/10.1155/2018/8490489]
[114]
Dash BC, Xu Z, Lin L, et al. Stem Cells and Engineered Scaffolds for Regenerative Wound Healing. Bioengineering (Basel) 2018; 5(1): 23.
[http://dx.doi.org/10.3390/bioengineering5010023] [PMID: 29522497]
[115]
Riley RS, June CH, Langer R, Mitchell MJ. Delivery technologies for cancer immunotherapy. Nat Rev Drug Discov 2019; 18(3): 175-96.
[http://dx.doi.org/10.1038/s41573-018-0006-z] [PMID: 30622344]
[116]
Leberfinger AN, Moncal KK, Ravnic DJ, Ozbolat IT. 3D printing for cell therapy applications Cell Therapy. Springer 2017; pp. 227-48.
[117]
Correa D, Lietman SA, Eds. Articular cartilage repair: Current needs, methods and research directions Seminars in Cell & Developmental Biology. Elsevier 2017.
[118]
Armiento AR, Alini M, Stoddart MJ. Articular fibrocartilage-Why does hyaline cartilage fail to repair? Adv Drug Deliv Rev 2018.
[PMID: 30605736]
[119]
Borrelli J Jr, Olson SA, Godbout C, Schemitsch EH, Stannard JP, Giannoudis PV. Understanding Articular Cartilage Injury and Potential Treatments. J Orthop Trauma 2019; 33(Suppl. 6): S6-S12.
[http://dx.doi.org/10.1097/BOT.0000000000001472] [PMID: 31083142]
[120]
Medvedeva EV, Grebenik EA, Gornostaeva SN, et al. Repair of damaged articular cartilage: current approaches and future directions. Int J Mol Sci 2018; 19(8): 2366.
[http://dx.doi.org/10.3390/ijms19082366] [PMID: 30103493]
[121]
Zhu H, Gong H, Liu Q, Chen HH. Three-dimensional Bioprinting for Cartilage Regeneration. Technology (Singap) 2018; 5(04)
[122]
Canadas RF, Pirraco RP, Oliveira JM, Reis RL, Marques AP. Stem cells for osteochondral regeneration Osteochondral Tissue Engineering. Springer 2018; pp. 219-40.
[123]
Anson DS. The use of retroviral vectors for gene therapy-what are the risks? A review of retroviral pathogenesis and its relevance to retroviral vector-mediated gene delivery. Genet Vaccines Ther 2004; 2(1): 9.
[http://dx.doi.org/10.1186/1479-0556-2-9] [PMID: 15310406]
[124]
Edelstein ML, Abedi MR, Wixon J. Gene therapy clinical trials worldwide to 2007—an update. The Journal of Gene Medicine: A cross‐disciplinary journal for research on the science of gene transfer and its clinical applications 2007; 9(10): 833-42.
[125]
Akki R, Ramya MG, Vardhani K. A Review Article on Gene Therapy. J Drug Deliv Ther 2019; 9(4): 688-92.
[126]
Ginn SL, Amaya AK, Alexander IE, Edelstein M, Abedi MR. Gene therapy clinical trials worldwide to 2017: An update. J Gene Med 2018; 20(5)e3015
[http://dx.doi.org/10.1002/jgm.3015] [PMID: 29575374]
[127]
Schlaeger TM, Daheron L, Brickler TR, et al. A comparison of non-integrating reprogramming methods. Nat Biotechnol 2015; 33(1): 58-63.
[http://dx.doi.org/10.1038/nbt.3070] [PMID: 25437882]
[128]
Hu W, He Y, Xiong Y, et al. Derivation, expansion, and motor neuron differentiation of human-induced pluripotent stem cells with non-integrating episomal vectors and a defined xenogeneic-free culture system. Mol Neurobiol 2016; 53(3): 1589-600.
[http://dx.doi.org/10.1007/s12035-014-9084-z] [PMID: 25663198]
[129]
Jenkins MJ. Decisional tools for cost-effective bioprocess design for cell therapies and patient-specific drug discovery tools: UCL. University College London 2018.
[130]
Kawata M, Mori D, Kanke K, et al. Simple and Robust Differentiation of Human Pluripotent Stem Cells toward Chondrocytes by Two Small-Molecule Compounds. Stem Cell Reports 2019; 13(3): 530-44.
[http://dx.doi.org/10.1016/j.stemcr.2019.07.012] [PMID: 31402337]
[131]
Rim YA, Nam Y, Park N, et al. Different Chondrogenic Potential among Human Induced Pluripotent Stem Cells from Diverse Origin Primary Cells. Stem Cells Int 2018; 20189432616
[http://dx.doi.org/10.1155/2018/9432616] [PMID: 29535785]
[132]
Diederichs S, Gabler J, Autenrieth J, et al. Differential regulation of SOX9 protein during chondrogenesis of induced pluripotent stem cells versus mesenchymal stromal cells: a shortcoming for cartilage formation. Stem Cells Dev 2016; 25(8): 598-609.
[http://dx.doi.org/10.1089/scd.2015.0312] [PMID: 26906619]
[133]
Liu Y, Goldberg AJ, Dennis JE, Gronowicz GA, Kuhn LT. One-step derivation of mesenchymal stem cell (MSC)-like cells from human pluripotent stem cells on a fibrillar collagen coating. PLoS One 2012; 7(3)e33225
[http://dx.doi.org/10.1371/journal.pone.0033225] [PMID: 22457746]
[134]
Qu C, Puttonen KA, Lindeberg H, et al. Chondrogenic differentiation of human pluripotent stem cells in chondrocyte co-culture. Int J Biochem Cell Biol 2013; 45(8): 1802-12.
[http://dx.doi.org/10.1016/j.biocel.2013.05.029] [PMID: 23735325]
[135]
Umeda K, Zhao J, Simmons P, Stanley E, Elefanty A, Nakayama N. Human chondrogenic paraxial mesoderm, directed specification and prospective isolation from pluripotent stem cells. Sci Rep 2012; 2: 455.
[http://dx.doi.org/10.1038/srep00455] [PMID: 22701159]
[136]
Lee J, Taylor SE, Smeriglio P, et al. Early induction of a prechondrogenic population allows efficient generation of stable chondrocytes from human induced pluripotent stem cells. FASEB J 2015; 29(8): 3399-410.
[http://dx.doi.org/10.1096/fj.14-269720] [PMID: 25911615]
[137]
Chijimatsu R, Ikeya M, Yasui Y, et al. Characterization of mesenchymal stem cell-like cells derived from human iPSCs via neural crest development and their ap-plication for osteochondral repair. Stem Cells international 2017.
[http://dx.doi.org/10.1155/2017/1960965]
[138]
Oldershaw RA, Baxter MA, Lowe ET, et al. Directed differentiation of human embryonic stem cells toward chondrocytes. Nat Biotechnol 2010; 28(11): 1187-94.
[http://dx.doi.org/10.1038/nbt.1683] [PMID: 20967028]
[139]
Stelcer E, Kulcenty K, Suchorska WM. Chondrocytes differentiated from human induced pluripotent stem cells: Response to ionizing radiation. PLoS One 2018; 13(10) e0205691
[http://dx.doi.org/10.1371/journal.pone.0205691] [PMID: 30352062]
[140]
Uto S, Nishizawa S, Takasawa Y, et al. Bone and cartilage repair by transplantation of induced pluripotent stem cells in murine joint defect model. Biomed Res 2013; 34(6): 281-8.
[http://dx.doi.org/10.2220/biomedres.34.281] [PMID: 24389404]
[141]
Saito T, Yano F, Mori D, Kawata M, Hoshi K, Takato T, et al. Hyaline cartilage formation and tumorigenesis of implanted tissues derived from human induced pluripotent stem cells Biomedical research (Tokyo, Japan) 2015; 36(3): 179-86
[http://dx.doi.org/10.2220/biomedres.36.179]
[142]
Cheng A, Kapacee Z, Peng J, et al. Cartilage repair using human embryonic stem cell-derived chondroprogenitors. Stem Cells Transl Med 2014; 3(11): 1287-94.
[http://dx.doi.org/10.5966/sctm.2014-0101] [PMID: 25273540]
[143]
Villa-Diaz LG, Brown SE, Liu Y, et al. Derivation of mesenchymal stem cells from human induced pluripotent stem cells cultured on synthetic substrates. Stem Cells 2012; 30(6): 1174-81.
[http://dx.doi.org/10.1002/stem.1084] [PMID: 22415987]
[144]
Nguyen D, Hägg DA, Forsman A, et al. Cartilage tissue engineering by the 3D bioprinting of iPS cells in a nanocellulose/alginate bioink. Sci Rep 2017; 7(1): 658.
[http://dx.doi.org/10.1038/s41598-017-00690-y] [PMID: 28386058]

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