open access

Vol 74, No 2 (2023)
Review paper
Submitted: 2022-07-07
Accepted: 2022-09-13
Published online: 2023-04-11
Get Citation

Thyroid organoids: Advances and applications

Jinlu Zhao1, Yi Ren1, Zhenzhong Ge2, Xingyu Zhao1, Wang Li1, He Wang3, Meng Jiang4
·
Pubmed: 37039493
·
Endokrynol Pol 2023;74(2):121-127.
Affiliations
  1. Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
  2. The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Nanjing, China
  3. Department of Pathology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
  4. Faculty of Computing, Harbin Institute of Technology, Harbin, China

open access

Vol 74, No 2 (2023)
Review Article
Submitted: 2022-07-07
Accepted: 2022-09-13
Published online: 2023-04-11

Abstract

Organoids are derived from stem cells under three-dimensional culture conditions through self-assembly, and they can recapitulate the structural and functional characteristics of organs in vivo during culture. Organoids can be generated from both normal and malignant tissues. Those derived from normal tissues are widely used in the field of regenerative medicine. Meanwhile, tumour-derived organoids retain the phenotypic heterogeneity and atypia of the primary tumour, thereby providing a reliable in vitro model for the study of tumour pathogenesis and treatment. The thyroid gland is one of the most important endocrine organs regulating the body’s energy metabolism and growth; however, it is also associated with a high incidence of malignancy. Organoid is an effective tool for thyroid research.

Thyroid tumour-derived organoids can inherit the histopathological properties of primary tumours, and thyroid tissue-derived organoids can form follicular structures and secrete thyroid hormones. The above characteristics of organoids provide a reliable way to study the mechanism of thyroid genesis and tumour development in vitro. In this review, we focus on current knowledge and strategies for the establishment of thyroid organoids in thyroid regeneration and tumour research aiming to increase our understanding of the pathogenesis of thyroid tumours and the regenerative treatment of patients with hypothyroidism.

Abstract

Organoids are derived from stem cells under three-dimensional culture conditions through self-assembly, and they can recapitulate the structural and functional characteristics of organs in vivo during culture. Organoids can be generated from both normal and malignant tissues. Those derived from normal tissues are widely used in the field of regenerative medicine. Meanwhile, tumour-derived organoids retain the phenotypic heterogeneity and atypia of the primary tumour, thereby providing a reliable in vitro model for the study of tumour pathogenesis and treatment. The thyroid gland is one of the most important endocrine organs regulating the body’s energy metabolism and growth; however, it is also associated with a high incidence of malignancy. Organoid is an effective tool for thyroid research.

Thyroid tumour-derived organoids can inherit the histopathological properties of primary tumours, and thyroid tissue-derived organoids can form follicular structures and secrete thyroid hormones. The above characteristics of organoids provide a reliable way to study the mechanism of thyroid genesis and tumour development in vitro. In this review, we focus on current knowledge and strategies for the establishment of thyroid organoids in thyroid regeneration and tumour research aiming to increase our understanding of the pathogenesis of thyroid tumours and the regenerative treatment of patients with hypothyroidism.

Get Citation

Keywords

organoids; thyroid; regenerative medicine; tumour research

About this article
Title

Thyroid organoids: Advances and applications

Journal

Endokrynologia Polska

Issue

Vol 74, No 2 (2023)

Article type

Review paper

Pages

121-127

Published online

2023-04-11

Page views

2999

Article views/downloads

1106

DOI

10.5603/EP.a2023.0019

Pubmed

37039493

Bibliographic record

Endokrynol Pol 2023;74(2):121-127.

Keywords

organoids
thyroid
regenerative medicine
tumour research

Authors

Jinlu Zhao
Yi Ren
Zhenzhong Ge
Xingyu Zhao
Wang Li
He Wang
Meng Jiang

References (55)
  1. Li Mo, Izpisua Belmonte JC. Organoids - Preclinical Models of Human Disease. N Engl J Med. 2019; 380(6): 569–579.
  2. Sato T, Vries RG, Snippert HJ, et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009; 459(7244): 262–265.
  3. Antonica F, Kasprzyk DF, Opitz R, et al. Generation of functional thyroid from embryonic stem cells. Nature. 2012; 491(7422): 66–71.
  4. Mazzaferri E, Jhiang S. Long-term impact of initial surgical and medical therapy on papillary and follicular thyroid cancer. Am J Med. 1994; 97(5): 418–428.
  5. Garber JR, Cobin RH, Gharib H, et al. American Association of Clinical Endocrinologists and American Thyroid Association Taskforce on Hypothyroidism in Adults. Clinical practice guidelines for hypothyroidism in adults: cosponsored by the American Association of Clinical Endocrinologists and the American Thyroid Association. Endocr Pract. 2012; 18(6): 988–1028.
  6. Saini S, Tulla K, Maker AV, et al. Therapeutic advances in anaplastic thyroid cancer: a current perspective. Mol Cancer. 2018; 17(1): 154.
  7. Behjati S, Huch M, van Boxtel R, et al. Genome sequencing of normal cells reveals developmental lineages and mutational processes. Nature. 2014; 513(7518): 422–425.
  8. Li Z, Araoka T, Wu J, et al. 3D Culture Supports Long-Term Expansion of Mouse and Human Nephrogenic Progenitors. Cell Stem Cell. 2016; 19(4): 516–529.
  9. Bartfeld S, Bayram T, van de Wetering M, et al. In vitro expansion of human gastric epithelial stem cells and their responses to bacterial infection. Gastroenterology. 2015; 148(1): 126–136.e6.
  10. Samimi H, Atlasi R, Parichehreh-Dizaji S, et al. A systematic review on thyroid organoid models: time-trend and its achievements. Am J Physiol Endocrinol Metab. 2021; 320(3): E581–E590.
  11. Bonnier F, Keating ME, Wróbel TP, et al. Cell viability assessment using the Alamar blue assay: a comparison of 2D and 3D cell culture models. Toxicol In Vitro. 2015; 29(1): 124–131.
  12. Hartung T. Thoughts on limitations of animal models. Parkinsonism Relat Disord. 2008; 14 Suppl 2: S81–S83.
  13. Shanks N, Greek R, Greek J. Are animal models predictive for humans? Philos Ethics Humanit Med. 2009; 4(1).
  14. Dutta D, Heo I, Clevers H. Disease Modeling in Stem Cell-Derived 3D Organoid Systems. Trends Mol Med. 2017; 23(5): 393–410.
  15. Takebe T, Wells JM. Organoids by design. Science. 2019; 364(6444): 956–959.
  16. Li Y, Tang P, Cai S, et al. Organoid based personalized medicine: from bench to bedside. Cell Regen. 2020; 9(1): 21.
  17. McCracken KW, Catá EM, Crawford CM, et al. Modelling human development and disease in pluripotent stem-cell-derived gastric organoids. Nature. 2014; 516(7531): 400–404.
  18. Dutta D, Heo I, Clevers H. Disease Modeling in Stem Cell-Derived 3D Organoid Systems. Trends Mol Med. 2017; 23(5): 393–410.
  19. Hussein SM, Batada NN, Vuoristo S, et al. Copy number variation and selection during reprogramming to pluripotency. Nature. 2011; 471(7336): 58–62.
  20. Laurent LC, Ulitsky I, Slavin I, et al. Dynamic changes in the copy number of pluripotency and cell proliferation genes in human ESCs and iPSCs during reprogramming and time in culture. Cell Stem Cell. 2011; 8(1): 106–118.
  21. Korkaya H, Liu S, Wicha MS. Breast cancer stem cells, cytokine networks, and the tumor microenvironment. J Clin Invest. 2011; 121(10): 3804–3809.
  22. Plaks V, Kong N, Werb Z. The cancer stem cell niche: how essential is the niche in regulating stemness of tumor cells? Cell Stem Cell. 2015; 16(3): 225–238.
  23. Brown JW, Mills JC. Implantable synthetic organoid matrices for intestinal regeneration. Nat Cell Biol. 2017; 19(11): 1307–1308.
  24. Vazin T, Schaffer DV. Engineering strategies to emulate the stem cell niche. Trends Biotechnol. 2010; 28(3): 117–124.
  25. Saito Y, Onishi N, Takami H, et al. Development of a functional thyroid model based on an organoid culture system. Biochem Biophys Res Commun. 2018; 497(2): 783–789.
  26. de Lau WBM, Snel B, Clevers HC. The R-spondin protein family. Genome Biol. 2012; 13(3): 242.
  27. MacDonald BT, He Xi. Frizzled and LRP5/6 receptors for Wnt/β-catenin signaling. Cold Spring Harb Perspect Biol. 2012; 4(12).
  28. Mah AT, Yan KS, Kuo CJ. Wnt pathway regulation of intestinal stem cells. J Physiol. 2016; 594(17): 4837–4847.
  29. Gehart H, Clevers H. Tales from the crypt: new insights into intestinal stem cells. Nat Rev Gastroenterol Hepatol. 2019; 16(1): 19–34.
  30. Xu H, Lyu X, Yi M, et al. Organoid technology and applications in cancer research. J Hematol Oncol. 2018; 11(1): 116.
  31. Tilson SG, Haley EM, Triantafillu UL, et al. ROCK Inhibition Facilitates In Vitro Expansion of Glioblastoma Stem-Like Cells. PLoS One. 2015; 10(7): e0132823.
  32. Takebe T, Sekine K, Enomura M, et al. Vascularized and functional human liver from an iPSC-derived organ bud transplant. Nature. 2013; 499(7459): 481–484.
  33. Spence JR, Mayhew CN, Rankin SA, et al. Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro. Nature. 2011; 470(7332): 105–109.
  34. Hohwieler M, Illing A, Hermann PC, et al. Human pluripotent stem cell-derived acinar/ductal organoids generate human pancreas upon orthotopic transplantation and allow disease modelling. Gut. 2017; 66(3): 473–486.
  35. Taguchi A, Nishinakamura R. Higher-Order Kidney Organogenesis from Pluripotent Stem Cells. Cell Stem Cell. 2017; 21(6): 730–746.e6.
  36. Coraux C, Hilmi C, Rouleau M, et al. Reconstituted Skin from Murine Embryonic Stem Cells. Curr Biol. 2003; 13(10): 849–853.
  37. Longmire TA, Ikonomou L, Hawkins F, et al. Efficient derivation of purified lung and thyroid progenitors from embryonic stem cells. Cell Stem Cell. 2012; 10(4): 398–411.
  38. Antonica F, Kasprzyk DF, Opitz R, et al. Generation of functional thyroid from embryonic stem cells. Nature. 2012; 491(7422): 66–71.
  39. Kurmann AA, Serra M, Hawkins F, et al. Regeneration of Thyroid Function by Transplantation of Differentiated Pluripotent Stem Cells. Cell Stem Cell. 2015; 17(5): 527–542.
  40. Lan L, Cui D, Nowka K, et al. Stem cells derived from goiters in adults form spheres in response to intense growth stimulation and require thyrotropin for differentiation into thyrocytes. J Clin Endocrinol Metab. 2007; 92(9): 3681–3688.
  41. Tai MH, Chang CC, Kiupel M, et al. Oct4 expression in adult human stem cells: evidence in support of the stem cell theory of carcinogenesis. Carcinogenesis. 2005; 26(2): 495–502.
  42. Ogundipe VML, Groen AH, Hosper N, et al. Generation and Differentiation of Adult Tissue-Derived Human Thyroid Organoids. Stem Cell Reports. 2021; 16(4): 913–925.
  43. Grüters A, Krude H. Detection and treatment of congenital hypothyroidism. Nat Rev Endocrinol. 2011; 8(2): 104–113.
  44. Yang Y, Opara EC, Liu Y, et al. Microencapsulation of porcine thyroid cell organoids within a polymer microcapsule construct. Exp Biol Med (Maywood). 2017; 242(3): 286–296.
  45. McGranahan N, Swanton C. Biological and therapeutic impact of intratumor heterogeneity in cancer evolution. Cancer Cell. 2015; 27(1): 15–26.
  46. Tuveson D, Clevers H. Cancer modeling meets human organoid technology. Science. 2019; 364(6444): 952–955.
  47. Sondorp LHJ, Ogundipe VML, Groen AH, et al. Patient-Derived Papillary Thyroid Cancer Organoids for Radioactive Iodine Refractory Screening. Cancers (Basel). 2020; 12(11).
  48. Chen D, Tan Y, Li Z, et al. Organoid Cultures Derived From Patients With Papillary Thyroid Cancer. J Clin Endocrinol Metab. 2021; 106(5): 1410–1426.
  49. Vlachogiannis G, Hedayat S, Vatsiou A, et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science. 2018; 359(6378): 920–926.
  50. Broutier L, Mastrogiovanni G, Verstegen MMa, et al. Human primary liver cancer-derived organoid cultures for disease modeling and drug screening. Nat Med. 2017; 23(12): 1424–1435.
  51. Kim M, Mun H, Sung CO, et al. Patient-derived lung cancer organoids as in vitro cancer models for therapeutic screening. Nat Commun. 2019; 10(1): 3991.
  52. Walsh AJ, Cook RS, Sanders ME, et al. Quantitative optical imaging of primary tumor organoid metabolism predicts drug response in breast cancer. Cancer Res. 2014; 74(18): 5184–5194.
  53. Kim J, Koo BK, Knoblich JA. Human organoids: model systems for human biology and medicine. Nat Rev Mol Cell Biol. 2020; 21(10): 571–584.
  54. Pollen AA, Bhaduri A, Andrews MG, et al. Establishing Cerebral Organoids as Models of Human-Specific Brain Evolution. Cell. 2019; 176(4): 743–756.e17.
  55. Nagle PW, Plukker JT, Muijs CT, et al. Patient-derived tumor organoids for prediction of cancer treatment response. Semin Cancer Biol. 2018; 53: 258–264.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

Via MedicaWydawcą jest  VM Media Group sp. z o.o., Grupa Via Medica, ul. Świętokrzyska 73, 80–180 Gdańsk

tel.:+48 58 320 94 94, faks:+48 58 320 94 60, e-mail:  viamedica@viamedica.pl