We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Research progress in seed cells for cartilage tissue engineering

    Baoshuai Bai‡

    Research Institute of Plastic Surgery, Wei Fang Medical University, Wei Fang, Shandong, 261053, China

    Shanghai Key Laboratory of Tissue Engineering, Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China

    National Tissue Engineering Center of China, Shanghai, 200240, China

    ‡The author contributed equally

    Search for more papers by this author

    ,
    Mengjie Hou‡

    Shanghai Key Laboratory of Tissue Engineering, Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China

    National Tissue Engineering Center of China, Shanghai, 200240, China

    ‡The author contributed equally

    Search for more papers by this author

    ,
    Junxiang Hao‡

    Research Institute of Plastic Surgery, Wei Fang Medical University, Wei Fang, Shandong, 261053, China

    Shanghai Key Laboratory of Tissue Engineering, Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China

    National Tissue Engineering Center of China, Shanghai, 200240, China

    ‡The author contributed equally

    Search for more papers by this author

    ,
    Yanhan Liu

    Shanghai JiaoTong University School of Medicine, Shanghai, 200240, China

    ,
    Guangyu Ji

    *Author for correspondence: Tel.: +86 212 327 1699;

    E-mail Address: jigy_dor@163.com

    Department of Thoracic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200240, China

    &
    Guangdong Zhou

    **Author for correspondence: Tel.: +86 212 327 1699;

    E-mail Address: guangdongzhou@126.com

    Research Institute of Plastic Surgery, Wei Fang Medical University, Wei Fang, Shandong, 261053, China

    Shanghai Key Laboratory of Tissue Engineering, Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China

    National Tissue Engineering Center of China, Shanghai, 200240, China

    Published Online:https://doi.org/10.2217/rme-2022-0023

    Cartilage defects trouble millions of patients worldwide and their repair via conventional treatment is difficult. Excitingly, tissue engineering technology provides a promising strategy for efficient cartilage regeneration with structural regeneration and functional reconstruction. Seed cells, as biological prerequisites for cartilage regeneration, determine the quality of regenerated cartilage. The proliferation, differentiation and chondrogenesis of seed cells are greatly affected by their type, origin and generation. Thus, a systematic description of the characteristics of seed cells is necessary. This article reviews in detail the cellular characteristics, research progress, clinical translation challenges and future research directions of seed cells while providing guidelines for selecting appropriate seed cells for cartilage regeneration.

    Graphical abstract

    Plain language summary

    Cartilage defects affect millions of patients worldwide and their repair via conventional treatment is quite difficult. Excitingly, tissue engineering technology provides a promising strategy for efficient cartilage regeneration. The seed cell, as a biological prerequisite for cartilage regeneration, determines the quality of regenerated cartilage. This article reviews in detail the cellular characteristics, research progress, clinical translation challenges and future research directions of various chondrocytes, chondroprogenitor cells and stem cells. Chondrocytes, especially elastic chondrocytes, could complete subcutaneous cartilage regeneration, whereas stem cells are superior for composite defects, allografts and cartilage defects caused by inflammation. In brief, this article provides a guide for selecting appropriate seed cells for cartilage regeneration.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Krishnan Y, Grodzinsky AJ. Cartilage diseases. Matrix Biol. 71–72, 51–69 (2018).
    • 2. Rotter N, Brenner RE. Cartilage repair across germ layer origins. Lancet (London) 388(10055), 1957–1958 (2016).
    • 3. Wang L, Huang J, Huang C et al. Adult stem cells and hydrogels for cartilage regeneration. Curr. Stem Cell Res. Ther. 13(7), 533–546 (2018).
    • 4. Daly AC, Freeman FE, Gonzalez-Fernandez T, Critchley SE, Nulty J, Kelly DJ. 3D bioprinting for cartilage and osteochondral tissue engineering. Adv. Healthc. Mater. 6(22), 1700298 (2017).
    • 5. Sharma P, Kumar P, Sharma R, Bhatt VD, Dhot PS. Tissue engineering; current status & futuristic scope. J. Med. Life 12(3), 225–229 (2019).
    • 6. Shi R, Huang Y, Ma C, Wu C, Tian W. Current advances for bone regeneration based on tissue engineering strategies. Front. Med. 13(2), 160–188 (2019).
    • 7. Varela-Eirin M, Loureiro J, Fonseca E et al. Cartilage regeneration and ageing: targeting cellular plasticity in osteoarthritis. Ageing Res. Rev. 42, 56–71 (2018).
    • 8. Le H, Xu W, Zhuang X, Chang F, Wang Y, Ding J. Mesenchymal stem cells for cartilage regeneration. J. Tissue Eng. 11, 2041731420943839 (2020).
    • 9. Lee DH, Kim SJ, Kim SA, Ju GI. Past, present, and future of cartilage restoration: from localized defect to arthritis. Knee Surg. Relat. Res. 34(1), 1 (2022).
    • 10. Lorda-Diez CI, Duarte-Olivenza C, Hurle JM, Montero JA. Transforming growth factor beta signaling: the master sculptor of fingers. Dev. Dyn. 251(1), 125–136 (2022).
    • 11. Trompeter N, Gardinier JD, Debarros V et al. Insulin-like growth factor-1 regulates the mechanosensitivity of chondrocytes by modulating TRPV4. Cell Calcium. 99, 102467 (2021).
    • 12. Huang K, Li Q, Li Y et al. Cartilage tissue regeneration: the roles of cells, stimulating factors and scaffolds. Curr. Stem Cell Res. Ther. 13(7), 547–567 (2018).
    • 13. Lin TH, Wang HC, Cheng WH, Hsu HC, Yeh ML. Osteochondral tissue regeneration using a tyramine-modified bilayered PLGA scaffold combined with articular chondrocytes in a porcine model. Int. J. Mol.Sci. 20(2), (2019).
    • 14. Huang X, Das R, Patel A, Nguyen TD. Physical stimulations for bone and cartilage regeneration. Regen. Eng. Transl. Med. 4(4), 216–237 (2018).
    • 15. Okubo R, Asawa Y, Watanabe M et al. Proliferation medium in three-dimensional culture of auricular chondrocytes promotes effective cartilage regeneration in vivo. Regen. Ther. 11, 306–315 (2019).
    • 16. He A, Ye A, Song N et al. Phenotypic redifferentiation of dedifferentiated microtia chondrocytes through a three-dimensional chondrogenic culture system. Am. J. Transl. Res. 12(6), 2903–2915 (2020).
    • 17. Patel JM, Saleh KS, Burdick JA, Mauck RL. Bioactive factors for cartilage repair and regeneration: improving delivery, retention, and activity. Acta Biomater. 93, 222–238 (2019).
    • 18. Miyanaga T, Ueda Y, Miyanaga A, Yagishita M, Hama N. Angiogenesis after administration of basic fibroblast growth factor induces proliferation and differentiation of mesenchymal stem cells in elastic perichondrium in an in vivo model: mini review of three sequential republication-abridged reports. Cell Mol. Biol. Lett. 23, 49 (2018).
    • 19. Messaoudi O, Henrionnet C, Bourge K, Loeuille D, Gillet P, Pinzano A. Stem cells and extrusion 3D printing for hyaline cartilage engineering. Cells 10(1), (2020).
    • 20. He A, Xia H, Xiao K et al. Cell yield, chondrogenic potential, and regenerated cartilage type of chondrocytes derived from ear, nasoseptal, and costal cartilage. J. Tissue Eng. Regen. Med. 12(4), 1123–1132 (2018).
    • 21. Xu Y, Xu Y, Bi B et al. A moldable thermosensitive hydroxypropyl chitin hydrogel for 3D cartilage regeneration in vitro and in vivo. Acta Biomater. 108, 87–96 (2020).
    • 22. Childs RD, Nakao H, Isogai N, Murthy A, Landis WJ. An analytical study of neocartilage from microtia and otoplasty surgical remnants: a possible application for BMP7 in microtia development and regeneration. PloS One 15(6), e0234650 (2020).
    • 23. Li XS, Sun JJ. Regenerative medicine of tissue engineering: auricular cartilage regeneration and functional reconstruction. Lin Chung Er Bi Yan Hou Tou Jing Wai Ke Za Zhi 33(6), 567–571 (2019).
    • 24. Bernstein JL, Cohen BP, Lin A, Harper A, Bonassar LJ, Spector JA. Tissue engineering auricular cartilage using late passage human auricular chondrocytes. Ann. Plast. Surg. 80(Suppl. 4), S168–S173 (2018).
    • 25. Alkaya D, Gurcan C, Kilic P, Yilmazer A, Gurman G. Where is human-based cellular pharmaceutical R&D taking us in cartilage regeneration? 3 Biotech 10(4), 161 (2020).
    • 26. Humphries S, Joshi A, Webb WR, Kanegaonkar R. Auricular reconstruction: where are we now? A critical literature review. Eur. Arch. Otorhinolaryngol. doi:10.1007/s06903-5 (2021).
    • 27. Hou M, Tian B, Bai B et al. Dominant role of in situ native cartilage niche for determining the cartilage type regenerated by BMSCs. Bioactive Mater. doi:10.1016/j.bioactmat.2021.11.007 (2021).
    • 28. Liu CF, Samsa WE, Zhou G, Lefebvre V. Transcriptional control of chondrocyte specification and differentiation. Semin. Cell Dev. Biol. 62, 34–49 (2017).
    • 29. Gao Y, Gao J, Li H et al. Autologous costal chondral transplantation and costa-derived chondrocyte implantation: emerging surgical techniques. Ther. Adv. Musculoskelet. Disease 11, 1759720x19877131 (2019).
    • 30. Bonilla A. Pediatric microtia reconstruction with autologous rib: personal experience and technique with 1000 pediatric patients with microtia. Facial Plast. Surg. Clin. North Am. 26(1), 57–68 (2018).
    • 31. Jessop ZM, Javed M, Otto IA et al. Combining regenerative medicine strategies to provide durable reconstructive options: auricular cartilage tissue engineering. Stem Cell Res. Ther. 7, 19 (2016).
    • 32. Carballo CB, Nakagawa Y, Sekiya I, Rodeo SA. Basic science of articular cartilage. Clin. Sports Med. 36(3), 413–425 (2017).
    • 33. Meng W, Gao L, Venkatesan JK, Wang G, Madry H, Cucchiarini M. Translational applications of photopolymerizable hydrogels for cartilage repair. J. Exp. Orthop. 6(1), 47 (2019).
    • 34. Li T, Chen S, Pei M. Contribution of neural crest-derived stem cells and nasal chondrocytes to articular cartilage regeneration. CMLS 77(23), 4847–4859 (2020).
    • 35. Buckley CD, Ospelt C, Gay S, Midwood KS. Location, location, location: how the tissue microenvironment affects inflammation in RA. Nat. Rev. Rheumatol. 17(4), 195–212 (2021).
    • 36. Hu W, Chen Y, Dou C, Dong S. Microenvironment in subchondral bone: predominant regulator for the treatment of osteoarthritis. Ann. Rheum. Dis. 80(4), 413–422 (2020).
    • 37. Bilgen B, Jayasuriya CT, Owens BD. Current concepts in meniscus tissue engineering and repair. Adv. Healthc. Mater. 7(11), e1701407 (2018).
    • 38. Hadidi P, Paschos NK, Huang BJ, Aryaei A, Hu JC, Athanasiou KA. Tendon and ligament as novel cell sources for engineering the knee meniscus. Osteoarthritis Cartilage 24(12), 2126–2134 (2016).
    • 39. Andia I, Maffulli N. Biological therapies in regenerative sports medicine. Sports Med. (Auckland) 47(5), 807–828 (2017).
    • 40. Ural IH, Alptekin K, Ketenci A, Solakoglu S, Alpak H, Özyalçın S. Fibroblast transplantation results to the degenerated rabbit lumbar intervertebral discs. Open Orthop. J. 11, 404–416 (2017).
    • 41. Zhou G, Jiang H, Yin Z et al. In vitro regeneration of patient-specific ear-shaped cartilage and its first clinical application for auricular reconstruction. EBioMedicine 28, 287–302 (2018). •• The clinical application of ear reconstruction is undoubtedly a milestone achievement.
    • 42. Tliba O, Panettieri RA Jr. Paucigranulocytic asthma: uncoupling of airway obstruction from inflammation. J. Allergy Clin. Immunol. 143(4), 1287–1294 (2019).
    • 43. Jia L, Zhang Y, Yao L et al. Regeneration of human-ear-shaped cartilage with acellular cartilage matrix-based biomimetic scaffolds. Appl. Mater. Today 20, (2020). •• The use of acellular cartilage matrix to regenerate cartilage is the most likely scaffold to achieve stable cartilage regeneration.
    • 44. Yin Z, Li D, Liu Y et al. Regeneration of elastic cartilage with accurate human-ear shape based on PCL strengthened biodegradable scaffold and expanded microtia chondrocytes. Appl. Mater. Today 20, (2020).
    • 45. Xu Y, Li D, Yin Z et al. Tissue-engineered trachea regeneration using decellularized trachea matrix treated with laser micropore technique. Acta Biomater. 58, 113–121 (2017).
    • 46. Zhang Y, Xu Y, Liu Y et al. Porous decellularized trachea scaffold prepared by a laser micropore technique. J. Mech. Behav. Biomed. Mater. 90, 96–103 (2019).
    • 47. Sueyoshi S, Chitose SI, Sato K, Fukahori M, Kurita T, Umeno H. Stable tracheal regeneration using organotypically cultured tissue composed of autologous chondrocytes and epithelial cells in beagles. Ann. Otol. Rhinol. Laryngol. 128(7), 585–594 (2019).
    • 48. Li D, Yin Z, Liu Y et al. Regeneration of trachea graft with cartilage support, vascularization, and epithelization. Acta Biomater. 89, 206–216 (2019).
    • 49. Xu Y, Guo Y, Li Y et al. Biomimetic trachea regeneration using a modular ring strategy based on poly(sebacoyl diglyceride)/polycaprolactone for segmental trachea defect repair. Adv. Funct. Mater. 30(42), (2020). •• To simulate the physiological structure of the tracheal cartilage ring and connective tissue between the rings alternately, a breakthrough has been made in the simulation of the alternate mode of loaded and unloaded cells.
    • 50. Etienne H, Fabre D, Gomez Caro A et al. Tracheal replacement. Eur. Resp. J. 51(2), (2018).
    • 51. Delaere PR, Van Raemdonck D. Commentary: the sobering truth about tracheal regeneration. J. Thorac. Cardiovasc. Surg. 159(6), 2537–2539 (2020).
    • 52. Shin YS, Lee BH, Choi JW et al. Tissue-engineered tracheal reconstruction using chondrocyte seeded on a porcine cartilage-derived substance scaffold. Int. J. Pediatric Otorhinolaryngol. 78(1), 32–38 (2014).
    • 53. Jiang Y, Tuan RS. Role of NGF-TrkA signaling in calcification of articular chondrocytes. FASEB J. 33(9), 10231–10239 (2019).
    • 54. Jacob G, Shimomura K, Krych AJ, Nakamura N. The meniscus tear: a review of stem cell therapies. Cells 9(1), (2019).
    • 55. Lyu FJ, Cheung KM, Zheng Z, Wang H, Sakai D, Leung VY. IVD progenitor cells: a new horizon for understanding disc homeostasis and repair. Nat. Rev. Rheumatol. 15(2), 102–112 (2019).
    • 56. Kim DH, Lim JY, Kim SW et al. Characteristics of nasal septal cartilage-derived progenitor cells during prolonged cultivation. Otolaryngol. Head Neck Surg. 159(4), 774–782 (2018).
    • 57. Guo Q, Zhou P, Li B. Identification and characterizations of annulus fibrosus-derived stem cells. Methods Mol. Biol.(Clifton) 1842, 207–216 (2018).
    • 58. Hou M, Bai B, Tian B et al. Cartilage Regeneration Characteristics of Human and Goat Auricular Chondrocytes Front. Bioengin. Biotech. 29(9), (2021).
    • 59. Qasim M, Chae DS, Lee NY. Bioengineering strategies for bone and cartilage tissue regeneration using growth factors and stem cells. J. Biomed. Mater. Res. A 108(3), 394–411 (2020).
    • 60. Cho H, Kim J, Kim S et al. Dual delivery of stem cells and insulin-like growth factor-1 in coacervate-embedded composite hydrogels for enhanced cartilage regeneration in osteochondral defects. J. Control Release 327, 284–295 (2020).
    • 61. Ma Q, Liao J, Cai X. Different sources of stem cells and their application in cartilage tissue engineering. Curr. Stem Cell Res. Ther. 13(7), 568–575 (2018).
    • 62. Wang T, Xu W, Zhao X et al. Repair of osteochondral defects mediated by double-layer scaffolds with natural osteochondral-biomimetic microenvironment and interface. Mater. Today Bio. 14, 100234 (2022).
    • 63. Zha K, Li X, Yang Z et al. Heterogeneity of mesenchymal stem cells in cartilage regeneration: from characterization to application. NPJ Regen. Med. 6(1), 14 (2021).
    • 64. Ansboro S, Roelofs AJ, De Bari C. Mesenchymal stem cells for the management of rheumatoid arthritis: immune modulation, repair or both? Curr. Opin. Rheumatol. 29(2), 201–207 (2017).
    • 65. Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat. Rev. Immunol. 8(9), 726–736 (2008).
    • 66. Fuchs E, Blau HM. Tissue stem cells: architects of their niches. Cell Stem Cell 27(4), 532–556 (2020).
    • 67. Wei F, Li Z, Crawford R, Xiao Y, Zhou Y. Immunoregulatory role of exosomes derived from differentiating mesenchymal stromal cells on inflammation and osteogenesis. J. Tissue Eng. Regen. Med. 13(11), 1978–1991 (2019).
    • 68. Barczewska M, Jezierska-Wozniak K, Habich A et al. Evaluation of regenerative processes in the pig model of intervertebral disc degeneration after transplantation of bone marrow-derived mesenchymal stem cells. Folia Neuropathol. 56(2), 124–132 (2018).
    • 69. Varma DM, Dinicolas MS, Nicoll SB. Injectable, redox-polymerized carboxymethylcellulose hydrogels promote nucleus pulposus-like extracellular matrix elaboration by human MSCs in a cell density-dependent manner. J. Biomater. Appl. 33(4), 576–589 (2018).
    • 70. Takagi T, Kabata T, Hayashi K et al. Periodic injections of adipose-derived stem cell sheets attenuate osteoarthritis progression in an experimental rabbit model. BMC Musculoskelet. Disord. 21(1), 691 (2020).
    • 71. Lopez-Santalla M, Mancheño-Corvo P, Menta R et al. Human adipose-derived mesenchymal stem cells modulate experimental autoimmune arthritis by modifying early adaptive T cell responses. Stem Cells (Dayton) 33(12), 3493–3503 (2015).
    • 72. Southworth TM, Naveen NB, Nwachukwu BU, Cole BJ, Frank RM. Orthobiologics for focal articular cartilage defects. Clin. Sports Med. 38(1), 109–122 (2019). •• The allogeneic application of adipose-derived mesenchymal stem cells makes it a seed cell with great potential in the field of cartilage regeneration.
    • 73. Mariñas-Pardo L, García-Castro J, Rodríguez-Hurtado I, Rodríguez-García MI, Núñez-Naveira L, Hermida-Prieto M. Allogeneic adipose-derived mesenchymal stem cells (horse allo 20) for the treatment of osteoarthritis-associated lameness in horses: characterization, safety, and efficacy of intra-articular treatment. Stem Cells Dev. 27(17), 1147–1160 (2018).
    • 74. Liu J, Zhou P, Long Y, Huang C, Chen D. Repair of bone defects in rat radii with a composite of allogeneic adipose-derived stem cells and heterogeneous deproteinized bone. Stem Cell Res. Ther. 9(1), 79 (2018).
    • 75. Zhou X, Wang J, Huang X et al. Injectable decellularized nucleus pulposus-based cell delivery system for differentiation of adipose-derived stem cells and nucleus pulposus regeneration. Acta Biomater. 81, 115–128 (2018).
    • 76. Ying J, Han Z, Zeng Y et al. Evaluation of intervertebral disc regeneration with injection of mesenchymal stem cells encapsulated in PEGDA-microcryogel delivery system using quantitative T2 mapping: a study in canines. Am. J. Transl. Res. 11(4), 2028–2041 (2019).
    • 77. Ishiguro H, Kaito T, Yarimitsu S et al. Intervertebral disc regeneration with an adipose mesenchymal stem cell-derived tissue-engineered construct in a rat nucleotomy model. Acta Biomater. 87, 118–129 (2019).
    • 78. Tao SC, Yuan T, Zhang YL, Yin WJ, Guo SC, Zhang CQ. Exosomes derived from miR00405-021-p-overexpressing human synovial mesenchymal stem cells enhance cartilage tissue regeneration and prevent osteoarthritis of the knee in a rat model. Theranostics 7(1), 180–195 (2017).
    • 79. Yao Y, Li ZY, Zhang H et al. Synovial fluid-derived synovial fragments represent an improved source of synovial mesenchymal stem cells in the temporomandibular joint. Int. J. Mol. Med. 41(1), 173–183 (2018).
    • 80. Zupan J, Drobnič M, Stražar K. Synovium-derived mesenchymal stem/stromal cells and their promise for cartilage regeneration. Adv. Exp. Med. Biol. 1212, 87–106 (2020).
    • 81. Beeravolu N, Brougham J, Khan I, Mckee C, Perez-Cruet M, Chaudhry GR. Human umbilical cord derivatives regenerate intervertebral disc. J. Tissue Eng. Regen. Med. 12(1), e579–e591 (2018).
    • 82. Topoluk N, Hawkins R, Tokish J, Mercuri J. Amniotic mesenchymal stromal cells exhibit preferential osteogenic and chondrogenic differentiation and enhanced matrix production compared with adipose mesenchymal stromal cells. Am. J. Sports Med. 45(11), 2637–2646 (2017).
    • 83. Romani R, Manni G, Donati C et al. S1P promotes migration, differentiation and immune regulatory activity in amniotic-fluid-derived stem cells. Eur. J. Pharmacol. 833, 173–182 (2018).
    • 84. Torre P, Flores AI. Current status and future prospects of perinatal stem cells. Genes 12(1), (2020).
    • 85. Tempest N, Maclean A, Hapangama DK. Endometrial stem cell markers: current concepts and unresolved questions. Int. J. Mol. Sci. 19(10), (2018).
    • 86. Ma J, Wu J, Han L et al. Comparative analysis of mesenchymal stem cells derived from amniotic membrane, umbilical cord, and chorionic plate under serum-free condition. Stem Cell Res. Ther. 10(1), 19 (2019).
    • 87. Chen JY, Mou XZ, Du XC, Xiang C. Comparative analysis of biological characteristics of adult mesenchymal stem cells with different tissue origins. Asian Pac. J. Trop. Med. 8(9), 739–746 (2015).
    • 88. Uzieliene I, Urbonaite G, Tachtamisevaite Z, Mobasheri A, Bernotiene E. The potential of menstrual blood-derived mesenchymal stem cells for cartilage repair and regeneration: novel aspects. Stem Cells Int. 2018, 5748126 (2018).
    • 89. Li H, Johnson NR, Usas A et al. Sustained release of bone morphogenetic protein 2 via coacervate improves the osteogenic potential of muscle-derived stem cells. Stem Cells Transl. Med. 2(9), 667–677 (2013).
    • 90. Chen YR, Yan X, Yuan FZ et al. The use of peripheral blood-derived stem cells for cartilage repair and regeneration in vivo: a review. Front. Pharmacol. 11, 404 (2020).
    • 91. Yang X, Ku T, Sun Z et al. Assessment of the carcinogenic effect of 2,3,7,8-tetrachlorodibenzo-p-dioxin using mouse embryonic stem cells to form teratoma in vivo. Toxicol. Lett. 312, 139–147 (2019).
    • 92. Yang YH, Zhang RZ, Cheng S et al. Generation of induced pluripotent stem cells from human epidermal keratinocytes. Cell Reprogram. 20(6), 356–364 (2018).
    • 93. Zhang W, Ling C, Zhang A et al. An all-silk-derived functional nanosphere matrix for sequential biomolecule delivery and in situ osteochondral regeneration. Bioactive Mater. 5(4), 832–843 (2020).
    • 94. Li X, Wang M, Jing X et al. Bone marrow- and adipose tissue-derived mesenchymal stem cells: characterization, differentiation, and applications in cartilage tissue engineering. Crit. Rev. Eukaryot. Gene Expr. 28(4), 285–310 (2018).
    • 95. Hwang ES. Senescence suppressors: their practical importance in replicative lifespan extension in stem cells. Cell. Mol. Life Sci. 71(21), 4207–4219 (2014).
    • 96. Chen L, Carlton M, Chen X et al. Effect of fibronectin, FGF-2, and BMP4 in the stemness maintenance of BMSCs and the metabolic and proteomic cues involved. Stem Cell Res. Ther. 12(1), 165 (2021).
    • 97. Zhou Q, Cai Y, Jiang Y, Lin X. Exosomes in osteoarthritis and cartilage injury: advanced development and potential therapeutic strategies. Int. J. Biol. Sci. 16(11), 1811–1820 (2020).
    • 98. Wei W, Ma Y, Yao X et al. Advanced hydrogels for the repair of cartilage defects and regeneration. Bioact. Mater. 6(4), 998–1011 (2021).
    • 99. Zhang FX, Liu P, Ding W et al. Injectable Mussel-Inspired highly adhesive hydrogel with exosomes for endogenous cell recruitment and cartilage defect regeneration. Biomaterials 278, 121169 (2021).
    • 100. Liu B, Yang F, Wei X et al. An exploratory study of articular cartilage and subchondral bone reconstruction with bone marrow mesenchymal stem cells combined with porous tantalum/Bio-Gide collagen membrane in osteonecrosis of the femoral head. Mater. Sci. Eng. C Mater. Biol. Appl. 99, 1123–1132 (2019). •• The multidifferentiation potential of bone marrow-derived stem cells gives it a huge advantage in coping with articular cartilage-subchondral bone defects.
    • 101. Li H, Shen S, Fu H et al. Immunomodulatory functions of mesenchymal stem cells in tissue engineering. Stem Cells Int. 2019, 9671206 (2019).
    • 102. Richardson SM, Kalamegam G, Pushparaj PN et al. Mesenchymal stem cells in regenerative medicine: focus on articular cartilage and intervertebral disc regeneration. Methods (San Diego) 99, 69–80 (2016).
    • 103. Feng C, Liu H, Yang Y, Huang B, Zhou Y. Growth and differentiation factor-5 contributes to the structural and functional maintenance of the intervertebral disc. Cell Physiol. Biochem. 35(1), 1–16 (2015).
    • 104. Khalilijafarabad N, Behnamghader A, Khorasani MT, Mozafari M. Platelet-rich plasma-hyaluronic acid/chondroitin sulfate/carboxymethyl chitosan hydrogel for cartilage regeneration. Biotechnol. Appl. Biochem. doi:10.1002/bab.2130 (2021).
    • 105. Gao L, Xu W, Li T et al. Stem cell therapy: a promising therapeutic method for intracerebral hemorrhage. Cell Transplant. 27(12), 1809–1824 (2018).
    • 106. Jiang T, Liu W, Lv X et al. Potent in vitro chondrogenesis of CD105 enriched human adipose-derived stem cells. Biomaterials 31(13), 3564–3571 (2010).
    • 107. Pak J, Lee JH, Pak N et al. Cartilage regeneration in humans with adipose tissue-derived stem cells and adipose stromal vascular fraction cells: updated status. Int. J. Mol. Sci. 19(7), (2018).
    • 108. Megaloikonomos PD, Panagopoulos GN, Bami M et al. Harvesting, isolation and differentiation of rat adipose-derived stem cells. Curr. Pharm. Biotechnol. 19(1), 19–29 (2018).
    • 109. Gu X, Li C, Yin F, Yang G. Adipose-derived stem cells in articular cartilage regeneration: current concepts and optimization strategies. Histol. Histopathol. 33(7), 639–653 (2018).
    • 110. Toratani T, Nakase J, Numata H et al. Scaffold-free tissue-engineered allogenic adipose-derived stem cells promote meniscus healing. Arthroscopy 33(2), 346–354 (2017).
    • 111. Vahedi P, Jarolmasjed S, Shafaei H, Roshangar L, Soleimani Rad J, Ahmadian E. In vivo articular cartilage regeneration through infrapatellar adipose tissue derived stem cell in nanofiber polycaprolactone scaffold. Tissue Cell 57, 49–56 (2019).
    • 112. Xu L, Shunmei E, Lin S et al. Sox11-modified mesenchymal stem cells accelerate cartilage defect repair in SD rats. Cell Tissue Res. 376(2), 247–255 (2019).
    • 113. Zou J, Bai B, Yao Y. Progress of co-culture systems in cartilage regeneration. Expert Opin. Biol. Ther. 18(11), 1151–1158 (2018).