Revisão Acesso aberto Revisado por pares

Everything You Always Wanted to Know About Organoid-Based Models (and Never Dared to Ask)

2022; Elsevier BV; Volume: 14; Issue: 2 Linguagem: Inglês

10.1016/j.jcmgh.2022.04.012

ISSN

2352-345X

Autores

Isabelle Hautefort, Martina Poletti, Diana Papp, Tamás Korcsmáros,

Tópico(s)

Science, Research, and Medicine

Resumo

Homeostatic functions of a living tissue, such as the gastrointestinal tract, rely on highly sophisticated and finely tuned cell-to-cell interactions. These crosstalks evolve and continuously are refined as the tissue develops and give rise to specialized cells performing general and tissue-specific functions. To study these systems, stem cell–based in vitro models, often called organoids, and non–stem cell–based primary cell aggregates (called spheroids) appeared just over a decade ago. These models still are evolving and gaining complexity, making them the state-of-the-art models for studying cellular crosstalk in the gastrointestinal tract, and to investigate digestive pathologies, such as inflammatory bowel disease, colorectal cancer, and liver diseases. However, the use of organoid- or spheroid-based models to recapitulate in vitro the highly complex structure of in vivo tissue remains challenging, and mainly restricted to expert developmental cell biologists. Here, we condense the founding knowledge and key literature information that scientists adopting the organoid technology for the first time need to consider when using these models for novel biological questions. We also include information that current organoid/spheroid users could use to add to increase the complexity to their existing models. We highlight the current and prospective evolution of these models through bridging stem cell biology with biomaterial and scaffold engineering research areas. Linking these complementary fields will increase the in vitro mimicry of in vivo tissue, and potentially lead to more successful translational biomedical applications. Deepening our understanding of the nature and dynamic fine-tuning of intercellular crosstalks will enable identifying novel signaling targets for new or repurposed therapeutics used in many multifactorial diseases. Homeostatic functions of a living tissue, such as the gastrointestinal tract, rely on highly sophisticated and finely tuned cell-to-cell interactions. These crosstalks evolve and continuously are refined as the tissue develops and give rise to specialized cells performing general and tissue-specific functions. To study these systems, stem cell–based in vitro models, often called organoids, and non–stem cell–based primary cell aggregates (called spheroids) appeared just over a decade ago. These models still are evolving and gaining complexity, making them the state-of-the-art models for studying cellular crosstalk in the gastrointestinal tract, and to investigate digestive pathologies, such as inflammatory bowel disease, colorectal cancer, and liver diseases. However, the use of organoid- or spheroid-based models to recapitulate in vitro the highly complex structure of in vivo tissue remains challenging, and mainly restricted to expert developmental cell biologists. Here, we condense the founding knowledge and key literature information that scientists adopting the organoid technology for the first time need to consider when using these models for novel biological questions. We also include information that current organoid/spheroid users could use to add to increase the complexity to their existing models. We highlight the current and prospective evolution of these models through bridging stem cell biology with biomaterial and scaffold engineering research areas. Linking these complementary fields will increase the in vitro mimicry of in vivo tissue, and potentially lead to more successful translational biomedical applications. Deepening our understanding of the nature and dynamic fine-tuning of intercellular crosstalks will enable identifying novel signaling targets for new or repurposed therapeutics used in many multifactorial diseases. SummaryAlthough revolutionary and increasingly used, organoids remain a challenging model for new users. In this review, we provide a general introduction for improving the accessibility to these models. We highlight areas for cross-disciplinary collaboration with biomaterial, tissue engineering, and nanofabrication sciences to broaden the application of organoids. Although revolutionary and increasingly used, organoids remain a challenging model for new users. In this review, we provide a general introduction for improving the accessibility to these models. We highlight areas for cross-disciplinary collaboration with biomaterial, tissue engineering, and nanofabrication sciences to broaden the application of organoids. The sophistication and functioning complexity of all different organs in human beings are fascinating and yet so challenging to accurately define and investigate. Decoding the complex molecular and cellular interactions taking place in each organ, and how they malfunction in diseases, is instrumental to the progress of biomedical research and eventually to personalized medicine. Previously established in vitro models (cell lines, primary cell cultures) were either too simplified or not translatable to human beings and had limitations in recapitulating the different cell types and their interactions. Scientists have had to re-explore embryology and tissue development to devise and develop novel stem cell–based in vitro models that allow studying the mechanism of the vast range of interactions taking place within an organ in health and disease. The complexity of a functional organ resides mainly in the fact that all its cells sense, adapt, and respond to their immediate and distant environments. In the gastrointestinal tract (GIT), this includes not only external factors (eg, diet, microbes1Freire R. Ingano L. Serena G. Cetinbas M. Anselmo A. Sapone A. Sadreyev R.I. Fasano A. Senger S. Human gut derived-organoids provide model to study gluten response and effects of microbiota-derived molecules in celiac disease.Sci Rep. 2019; 9: 7029Crossref PubMed Scopus (42) Google Scholar, 2In J. Foulke-Abel J. Zachos N.C. Hansen A.-M. Kaper J.B. Bernstein H.D. Halushka M. Blutt S. Estes M.K. Donowitz M. Kovbasnjuk O. Enterohemorrhagic Escherichia coli reduce mucus and intermicrovillar bridges in human stem cell-derived colonoids.Cell Mol Gastroenterol Hepatol. 2016; 2: 48-62.e3Abstract Full Text Full Text PDF PubMed Google Scholar, 3Tomosada Y. Villena J. Murata K. Chiba E. Shimazu T. Aso H. Iwabuchi N. Xiao J. Saito T. Kitazawa H. Immunoregulatory effect of bifidobacteria strains in porcine intestinal epithelial cells through modulation of ubiquitin-editing enzyme A20 expression.PLoS One. 2013; 8e59259Crossref PubMed Scopus (74) Google Scholar) but also neighboring cells, cells from other tissues within the same or from distant organs.1Freire R. Ingano L. Serena G. Cetinbas M. Anselmo A. Sapone A. Sadreyev R.I. Fasano A. Senger S. Human gut derived-organoids provide model to study gluten response and effects of microbiota-derived molecules in celiac disease.Sci Rep. 2019; 9: 7029Crossref PubMed Scopus (42) Google Scholar,4Kong S. Zhang Y.H. Zhang W. Regulation of intestinal epithelial cells properties and functions by amino acids.Biomed Res Int. 2018; 2018: 2819154Crossref PubMed Scopus (63) Google Scholar, 5Yan F. Cao H. Cover T.L. Whitehead R. Washington M.K. Polk D.B. Soluble proteins produced by probiotic bacteria regulate intestinal epithelial cell survival and growth.Gastroenterology. 2007; 132: 562-575Abstract Full Text Full Text PDF PubMed Scopus (545) Google Scholar, 6Isaacs-Ten A. Echeandia M. Moreno-Gonzalez M. Brion A. Goldson A. Philo M. Patterson A.M. Parker A. Galduroz M. Baker D. Rushbrook S.M. Hildebrand F. Beraza N. Intestinal microbiome-macrophage crosstalk contributes to cholestatic liver disease by promoting intestinal permeability in mice.Hepatology. 2020; 72: 2090-2108Crossref PubMed Scopus (28) Google Scholar, 7Saunders P.R. Miceli P. Vallance B.A. Wang L. Pinto S. Tougas G. Kamath M. Jacobson K. Noradrenergic and cholinergic neural pathways mediate stress-induced reactivation of colitis in the rat.Auton Neurosci. 2006; 124: 56-68Abstract Full Text Full Text PDF PubMed Scopus (28) Google Scholar, 8Han S.J. Li H. Kim M. D'Agati V. Lee H.T. Intestinal Toll-like receptor 9 deficiency leads to Paneth cell hyperplasia and exacerbates kidney, intestine, and liver injury after ischemia/reperfusion injury.Kidney Int. 2019; 95: 859-879Abstract Full Text Full Text PDF PubMed Scopus (10) Google Scholar For example, the different cell types of the intestinal epithelium (eg, enterocytes, enteroendocrine cells, goblet cells) communicate with luminal or mucosa-associated microbes from the resident gut microbiota or with pathogens during infection.9Derrien M. Alvarez A.-S. de Vos W.M. The gut microbiota in the first decade of life.Trends Microbiol. 2019; 27: 997-1010Abstract Full Text Full Text PDF PubMed Scopus (137) Google Scholar, 10Soderholm A.T. Pedicord V.A. Intestinal epithelial cells: at the interface of the microbiota and mucosal immunity.Immunology. 2019; 158: 267-280Crossref PubMed Scopus (59) Google Scholar, 11Nigro G. Hanson M. Fevre C. Lecuit M. Sansonetti P.J. Intestinal organoids as a novel tool to study microbes-epithelium interactions.Methods Mol Biol. 2019; 1576: 183-194Crossref PubMed Scopus (11) Google Scholar Intestinal epithelial cells also interact with each other and with tissue types from the intestine, such as the underlying mesenchyme,12Bahar Halpern K. Massalha H. Zwick R.K. Moor A.E. Castillo-Azofeifa D. Rozenberg M. Farack L. Egozi A. Miller D.R. Averbukh I. Harnik Y. Weinberg-Corem N. de Sauvage F.J. Amit I. Klein O.D. Shoshkes-Carmel M. Itzkovitz S. Lgr5+ telocytes are a signaling source at the intestinal villus tip.Nat Commun. 2020; 11: 1936Crossref PubMed Scopus (39) Google Scholar, 13Visco V. Bava F.A. d'Alessandro F. Cavallini M. Ziparo V. Torrisi M.R. Human colon fibroblasts induce differentiation and proliferation of intestinal epithelial cells through the direct paracrine action of keratinocyte growth factor.J Cell Physiol. 2009; 220: 204-213Crossref PubMed Scopus (0) Google Scholar, 14Kedinger M. Duluc I. Fritsch C. Lorentz O. Plateroti M. Freund J.N. Intestinal epithelial-mesenchymal cell interactions.Ann N Y Acad Sci. 1998; 859: 1-17Crossref PubMed Scopus (95) Google Scholar the gut-associated innate and adaptive immune system,15Allaire J.M. Crowley S.M. Law H.T. Chang S.-Y. Ko H.-J. Vallance B.A. The intestinal epithelium: central coordinator of mucosal immunity.Trends Immunol. 2018; 39: 677-696Abstract Full Text Full Text PDF PubMed Scopus (272) Google Scholar, 16McDole J.R. Wheeler L.W. McDonald K.G. Wang B. Konjufca V. Knoop K.A. Newberry R.D. Miller M.J. Goblet cells deliver luminal antigen to CD103+ dendritic cells in the small intestine.Nature. 2012; 483: 345-349Crossref PubMed Scopus (579) Google Scholar, 17Takahashi Y. Sato S. Kurashima Y. Lai C.-Y. Otsu M. Hayashi M. Yamaguchi T. Kiyono H. Reciprocal inflammatory signaling between intestinal epithelial cells and adipocytes in the absence of immune cells.EBioMedicine. 2017; 23: 34-45Abstract Full Text Full Text PDF PubMed Scopus (25) Google Scholar the enteric nervous system,13Visco V. Bava F.A. d'Alessandro F. Cavallini M. Ziparo V. Torrisi M.R. Human colon fibroblasts induce differentiation and proliferation of intestinal epithelial cells through the direct paracrine action of keratinocyte growth factor.J Cell Physiol. 2009; 220: 204-213Crossref PubMed Scopus (0) Google Scholar,18Neunlist M. Van Landeghem L. Mahé M.M. Derkinderen P. des Varannes S.B. Rolli-Derkinderen M. The digestive neuronal-glial-epithelial unit: a new actor in gut health and disease.Nat Rev Gastroenterol Hepatol. 2013; 10: 90-100Crossref PubMed Scopus (169) Google Scholar, 19Puzan M. Hosic S. Ghio C. Koppes A. Enteric nervous system regulation of intestinal stem cell differentiation and epithelial monolayer function.Sci Rep. 2018; 8: 6313Crossref PubMed Scopus (47) Google Scholar, 20Grenham S. Clarke G. Cryan J.F. Dinan T.G. Brain-gut-microbe communication in health and disease.Front Physiol. 2011; 2: 94Crossref PubMed Scopus (560) Google Scholar or even distant organs such as the liver, the lungs, or the brain.21Atif M. Warner S. Oo Y.H. Linking the gut and liver: crosstalk between regulatory T cells and mucosa-associated invariant T cells.Hepatol Int. 2018; 12: 305-314Crossref PubMed Scopus (13) Google Scholar, 22Ghosh S.S. Wang J. Yannie P.J. Ghosh S. Intestinal barrier function and metabolic/liver diseases.Liver Res. 2020; 4: 81-87Crossref Scopus (7) Google Scholar, 23Wittkopf N. Neurath M.F. Becker C. Immune-epithelial crosstalk at the intestinal surface.J Gastroenterol. 2014; 49: 375-387Crossref PubMed Scopus (43) Google Scholar Accumulating evidence highlights the importance of maintaining an equilibrium between the intercellular crosstalks through intricate and dynamic regulatory pathways.24Li H. Fan C. Lu H. Feng C. He P. Yang X. Xiang C. Zuo J. Tang W. Protective role of berberine on ulcerative colitis through modulating enteric glial cells-intestinal epithelial cells-immune cells interactions.Acta Pharm Sin B. 2020; 10: 447-461Crossref PubMed Scopus (43) Google Scholar, 25Talbot J, Hahn P, Kroehling L, Nguyen H, Li D, Littman DR. VIP-producing enteric neurons interact with innate lymphoid cells to regulate feeding-dependent intestinal epithelial barrier functions. Nat 579:575–580.Google Scholar, 26Peterson L.W. Artis D. Intestinal epithelial cells: regulators of barrier function and immune homeostasis.Nat Rev Immunol. 2014; 14: 141-153Crossref PubMed Scopus (1431) Google Scholar Complex mechanisms ensure such biological systems can cope with transient fluctuations in the environment. Yet, alterations of key regulatory mechanisms (including host genetics or environmental factors) dramatically impact the growth, differentiation, maturation, and functions of many cell types. Malfunction of specific or multiple epithelial cell types consequently impairs intercellular crosstalks and can lead to chronic diseases such as Inflammatory bowel disease.27Fair K.L. Colquhoun J. Hannan N.R.F. Intestinal organoids for modelling intestinal development and disease.Philos Trans R Soc Lond B Biol Sci. 2018; 373: 20170217Crossref PubMed Scopus (31) Google Scholar, 28Dutta D. Heo I. Clevers H. Disease modeling in stem cell-derived 3D organoid systems.Trends Mol Med. 2017; 23: 393-410Abstract Full Text Full Text PDF PubMed Scopus (367) Google Scholar, 29Burger E. Araujo A. López-Yglesias A. Rajala M.W. Geng L. Levine B. Hooper L.V. Burstein E. Yarovinsky F. Loss of Paneth cell autophagy causes acute susceptibility to Toxoplasma gondii-mediated inflammation.Cell Host Microbe. 2018; 23: 177-190.e4Abstract Full Text Full Text PDF PubMed Scopus (54) Google Scholar This review focuses on how recently established stem cell–based models recapitulate host cell–cell interactions. We summarize the intrinsic limitations and complementarity of the different models that scientists should bear in mind when developing novel experimental approaches. In this review, we only briefly discuss the impact microbes have on intestinal cells and how this can be studied with stem cell–based models (for detailed descriptions, see reviews published elsewhere30Poletti M. Arnauts K. Ferrante M. Korcsmaros T. Organoid-based models to study the role of host-microbiota interactions in IBD.J Crohns Colitis. 2021; 15: 1222-1235Crossref PubMed Scopus (6) Google Scholar, 31Holloway E.M. Capeling M.M. Spence J.R. Biologically inspired approaches to enhance human organoid complexity.Development. 2019; 146: dev166173Crossref PubMed Scopus (51) Google Scholar, 32George M.M. Rahman M. Connors J. Stadnyk A.W. Opinion: are organoids the end of model evolution for studying host intestinal epithelium/microbe interactions?.Microorganisms. 2019; 7: 406Crossref Scopus (6) Google Scholar). Many factors and specific cell types are responsible for the maintenance of the stem cell niche, and for the differentiation of its progeny cells. Several of these molecules and cells have been identified already, such as epidermal growth factor, Wingless and Int-1 (WNT), R-spondin, bone morphogenetic protein (BMP), as well as pericryptal myofibroblasts, mesenchymal cells, and processes such as autophagy.33Santos A.J.M. Lo Y.-H. Mah A.T. Kuo C.J. The intestinal stem cell niche: homeostasis and adaptations.Trends Cell Biol. 2018; 28: 1062-1078Abstract Full Text Full Text PDF PubMed Scopus (79) Google Scholar, 34Meran L. Baulies A. Li V.S.W. Intestinal stem cell niche: the extracellular matrix and cellular components.Stem Cells Int. 2017; 2017: 7970385Crossref PubMed Google Scholar, 35Pastuła A. Marcinkiewicz J. Cellular interactions in the intestinal stem cell niche.Arch Immunol Ther Exp (Warsz). 2019; 67: 19-26Crossref PubMed Scopus (11) Google Scholar However, their exact roles remain to be mechanistically unraveled for each cell type of the tissue of interest such as the intestinal epithelium. Filling these knowledge gaps requires improving in vitro culture systems of primary cells, particularly stem cells. Grown from stem cells and necessitating extracellular matrix-like scaffolding and specific niche factors, 3-dimensional (3D) cellular structures, termed organoids, can be created. Organoids can self-renew and generate in vitro functional structures containing the cell types present in the tissue they model (eg, mini-guts, mini-brains).36Yin Y.-B. de Jonge H.R. Wu X. Yin Y.-L. Mini-gut: a promising model for drug development.Drug Discov Today. 2019; 24: 1784-1794Crossref PubMed Scopus (13) Google Scholar, 37Developmental biology: "Mini-guts" made with nerves.Nature. 2016; 539: 471Crossref Scopus (0) Google Scholar, 38Li V.S.W. Modelling intestinal inflammation and infection using "mini-gut" organoids.Nat Rev Gastroenterol Hepatol. 2021; 18: 89-90Crossref PubMed Scopus (5) Google Scholar These organoid models have now widely revolutionized in vitro models to study health and disease. All differentiated cell types within an organ derive from progenitor cells, themselves being progenies of stem cells. Stem cells play an essential role in embryonic development and in the maintenance of most parts of an organ (eg, in the GIT they are essential for rapid renewal of the epithelium). Stem cells have been studied for decades and most recently have been used to develop in vitro cultures of organoids with cell types that to date could not be cultured in a dish.39Kim J. Koo B.-K. Knoblich J.A. Human organoids: model systems for human biology and medicine.Nat Rev Mol Cell Biol. 2020; 21: 571-584Crossref PubMed Scopus (352) Google Scholar There are 2 main routes to developing stem cell–based in vitro models, relying on 2 main classes of stem cells: adult stem cells (aSCs) that reside within certain fast renewing tissues such as the GIT epithelium, the lung alveoli cells or the skin epidermidis, and pluripotent stem cells (either embryonic pluripotent stem cells [ePSCs] or induced pluripotent stem cells [iPSCs]). Adult stem cells are undifferentiated cells naturally capable of self-regenerating asymmetrically. They renew themselves and produce progenitor cells that will proliferate and differentiate into all of the functional cell types normally residing in the tissue from which they derive.40Clevers H. Modeling development and disease with organoids.Cell. 2016; 165: 1586-1597Abstract Full Text Full Text PDF PubMed Scopus (1216) Google Scholar, 41Mummery C. Roelen B.A.J. Clevers H. Adult stem cells.Stem Cells. 2014; : 279-290Crossref Google Scholar, 42Sato T. Vries R.G. Snippert H.J. van de Wetering M. Barker N. Stange D.E. van Es J.H. Abo A. Kujala P. Peters P.J. Clevers H. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche.Nature. 2009; 459: 262-265Crossref PubMed Scopus (3741) Google Scholar aSCs can be cultured in vitro to generate heterotypic 3D organoid structures, containing all or most of the different cell types normally present in the tissue of origin. aSC-derived organoids can be generated from healthy or diseased patient tissue samples,43Yang H. Sun L. Liu M. Mao Y. Patient-derived organoids: a promising model for personalized cancer treatment.Gastroenterol Rep (Oxf). 2018; 6: 243-245Crossref PubMed Scopus (29) Google Scholar, 44Maru Y. Tanaka N. Itami M. Hippo Y. Efficient use of patient-derived organoids as a preclinical model for gynecologic tumors.Gynecol Oncol. 2019; 154: 189-198Abstract Full Text Full Text PDF PubMed Scopus (59) Google Scholar, 45Suzuki K. Murano T. Shimizu H. Ito G. Nakata T. Fujii S. Ishibashi F. Kawamoto A. Anzai S. Kuno R. Kuwabara K. Takahashi J. Hama M. Nagata S. Hiraguri Y. Takenaka K. Yui S. Tsuchiya K. Nakamura T. Ohtsuka K. Watanabe M. Okamoto R. Single cell analysis of Crohn's disease patient-derived small intestinal organoids reveals disease activity-dependent modification of stem cell properties.J Gastroenterol. 2018; 53: 1035-1047Crossref PubMed Scopus (47) Google Scholar and animal models.42Sato T. Vries R.G. Snippert H.J. van de Wetering M. Barker N. Stange D.E. van Es J.H. Abo A. Kujala P. Peters P.J. Clevers H. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche.Nature. 2009; 459: 262-265Crossref PubMed Scopus (3741) Google Scholar,46Fujii M. Matano M. Toshimitsu K. Takano A. Mikami Y. Nishikori S. Sugimoto S. Sato T. Human intestinal organoids maintain self-renewal capacity and cellular diversity in niche-inspired culture condition.Cell Stem Cell. 2018; 23: 787-793.e6Abstract Full Text Full Text PDF PubMed Scopus (162) Google Scholar Organoids can be maintained in culture for a long time through repeated passaging, during which they will maintain stable genetic and epigenetic signatures. During life, organs such as the GIT are exposed to different environmental signals (various microbes, food, antibiotics and general medications, inflammatory events, surgery), which will result in epigenetic modifications (eg, methylations, histone DNA packaging) within individual cells including stem cells.47Zhang L. Lu Q. Chang C. Epigenetics in health and disease.Adv Exp Med Biol. 2020; 1253: 3-55Crossref PubMed Scopus (59) Google Scholar Although nongenetic, these modifications will be heritable by the daughter cells during mitosis, impacting gene expression in differentiated progeny cells. Hence, organoids derived from tissue of the similar organ or genetic background but carrying different epigenetic profiles will behave differently, reflecting the differences in the original donors.47Zhang L. Lu Q. Chang C. Epigenetics in health and disease.Adv Exp Med Biol. 2020; 1253: 3-55Crossref PubMed Scopus (59) Google Scholar, 48Forsberg S.L. Ilieva M. Maria Michel T. Epigenetics and cerebral organoids: promising directions in autism spectrum disorders.Transl Psychiatry. 2018; 8: 14Crossref PubMed Scopus (27) Google Scholar, 49Cavalli G. Heard E. Advances in epigenetics link genetics to the environment and disease.Nature. 2019; 571: 489-499Crossref PubMed Scopus (358) Google Scholar, 50Sarvestani S.K. Signs S.A. Lefebvre V. Mack S. Ni Y. Morton A. Chan E.R. Li X. Fox P. Ting A. Kalady M.F. Cruise M. Ashburn J. Stiene J. Lai W. Liska D. Xiang S. Huang E.H. Cancer-predicting transcriptomic and epigenetic signatures revealed for ulcerative colitis in patient-derived epithelial organoids.Oncotarget. 2018; 9: 28717-28730Crossref PubMed Scopus (13) Google Scholar, 51Kraiczy J. Zilbauer M. Intestinal epithelial organoids as tools to study epigenetics in gut health and disease.Stem Cells Int. 2019; 2019: 7242415Crossref PubMed Scopus (13) Google Scholar As a result, organoids generated from different host backgrounds (eg, diseased vs control patient-derived) will allow interrogating the role of epigenetic signatures on cellular functions and, thus, on cell–cell interactions taking place in these multicellular structures in health and disease.50Sarvestani S.K. Signs S.A. Lefebvre V. Mack S. Ni Y. Morton A. Chan E.R. Li X. Fox P. Ting A. Kalady M.F. Cruise M. Ashburn J. Stiene J. Lai W. Liska D. Xiang S. Huang E.H. Cancer-predicting transcriptomic and epigenetic signatures revealed for ulcerative colitis in patient-derived epithelial organoids.Oncotarget. 2018; 9: 28717-28730Crossref PubMed Scopus (13) Google Scholar, 51Kraiczy J. Zilbauer M. Intestinal epithelial organoids as tools to study epigenetics in gut health and disease.Stem Cells Int. 2019; 2019: 7242415Crossref PubMed Scopus (13) Google Scholar, 52Lewis S.K. Nachun D. Martin M.G. Horvath S. Coppola G. Jones D.L. DNA methylation analysis validates organoids as a viable model for studying human intestinal aging.Cell Mol Gastroenterol Hepatol. 2020; 9: 527-541Abstract Full Text Full Text PDF PubMed Scopus (0) Google Scholar Depending on the source of stem cells used (adult, embryonic, or induced pluripotent), organoids can contain 1 or more tissue types. For instance, aSC-derived organoids established from intestinal crypt-derived stem cells will contain only epithelial cells. This type of organoid is ideal for achieving a simplified system, yet these models often lack the presence of underlying cells (eg, immune, mesenchyme, enteric nervous systems) and therefore will only reflect a limited part of the interactions staged in the whole intestinal system. To overcome this limitation, scientists have attempted growing cellular spheres derived from embedded minced tissue in an air–liquid interface culture system, successfully obtaining aSC-derived epithelial cells surrounded by a robust mesenchyme and stromal environment.53Ootani A. Li X. Sangiorgi E. Ho Q.T. Ueno H. Toda S. Sugihara H. Fujimoto K. Weissman I.L. Capecchi M.R. Kuo C.J. Sustained in vitro intestinal epithelial culture within a Wnt-dependent stem cell niche.Nat Med. 2009; 15: 701-706Crossref PubMed Scopus (578) Google Scholar,54Li X. Nadauld L. Ootani A. Corney D.C. Pai R.K. Gevaert O. Cantrell M.A. Rack P.G. Neal J.T. Chan C.W.-M. Yeung T. Gong X. Yuan J. Wilhelmy J. Robine S. Attardi L.D. Plevritis S.K. Hung K.E. Chen C.Z. Ji H.P. Kuo C.J. Oncogenic transformation of diverse gastrointestinal tissues in primary organoid culture.Nat Med. 2014; 20: 769-777Crossref PubMed Scopus (255) Google Scholar Yet, this alternative culture method of aSCs requires a solid expertise of ex vivo tissue culture methodologies and cannot be the primary choice for new users of organoid models. In addition, aSCs are a scarce cell population in some tissues/organs, which sometimes makes their harvest challenging or impossible, and often necessitates the use of an alternative source of stem cells such as PSCs. Tissues that either do not contain easily culturable stem cells, or are not easily accessible for stem cells to be collected (eg, brain), also can be cultured as organoids from pluripotent stem cells (either ePSCs or iPSCs).55Lancaster M.A. Corsini N.S. Wolfinger S. Gustafson E.H. Phillips A.W. Burkard T.R. Otani T. Livesey F.J. Knoblich J.A. Guided self-organization and cortical plate formation in human brain organoids.Nat Biotechnol. 2017; 35: 659-666Crossref PubMed Scopus (337) Google Scholar,56Renner M. Lancaster M.A. Bian S. Choi H. Ku T. Peer A. Chung K. Knoblich J.A. Self-organized developmental patterning and differentiation in cerebral organoids.EMBO J. 2017; 36: 1316-1329Crossref PubMed Scopus (171) Google Scholar ePSCs are the naturally present stem cells in an embryo, while iPSCs require first reprogramming of existing cells such as fibroblasts into PSCs. All PSCs are self-renewing cells that first derive into the 3 primary germ layers: ectoderm, endoderm, and mesoderm germ. In a second step, these germ layers will produce all cell types existing in the body.57Spence J.R. Mayhew C.N. Rankin S.A. Kuhar M.F. Vallance J.E. Tolle K. Hoskins E.E. Kalinichenko V.V. Wells S.I. Zorn A.M. Shroyer N.F. Wells J.M. Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro.Nature. 2011; 470: 105-109Crossref PubMed Scopus (1175) Google Scholar, 58Bilic J. Izpisua Belmonte J.C. Concise review: Induced pluripotent stem cells versus embryonic stem cells: close enough or yet too far apart?.Stem Cells. 2012; 30: 33-41Crossref PubMed Scopus (160) Google Scholar, 59Romito A. Cobellis G. Pluripotent stem cells: current understanding and future directions.Stem Cells Int. 2016; 2016: 9451492Crossref PubMed Scopus (64) Google Scholar As a result, PSC-derived organoids can include more than 1 tissue type and neighboring cells to the tissue of interest (eg, the mesenchyme), opening the door to in vitro reproduction of many more in vivo intercellular interactions than aSC-derived organoids would allow.57Spence J.R. Mayhew C.N. Rankin S.A. Kuhar M.F. Vallance J.E. Tolle K. Hoskins E.E. Kalinichenko V.V. Wells S.I. Zorn A.M. Shroyer N.F. Wells J.M. Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro.Nature. 2011; 470: 105-109Crossref PubMed Scopus (1175) Google Scholar,60Múnera J.O. Sundaram N. Rankin S.A. Hill D. Watson C. Mahe M. Vallance J.E. Shroyer N.F. Sinagoga K.L. Zarzoso-Lacoste A. Hudson J.R. Howell J.C. Chatuvedi P. Spence J.R. Shannon J.M. Zorn A.M. Helmrath M.A. Wells J.M. Differentiation of human pluripotent stem cells into colonic organoids via transient activation of BMP signaling.Cell Stem Cell. 2017; 21: 51-64.e6Crossref PubMed Scopus (8) Google Scholar However, mastering the right time-dependent modifications of the culture conditions of these cells to obtain the correct germ layer, and subsequently guide its evolution through all correct developmental stages to result in the required organ-modeling organoids, is extremely difficult, making these models accessible to only specialized laboratories.61Ota H. Miki N. Microtechnology-based three-dimensional spheroid formation.Front Biosci (Elite Ed). 2013; 5: 37-48Crossref PubMed Google Scholar,62Ryu N.-E. Lee S.-H. Park H. Spheroid culture system methods and applications for mesenchymal stem cells.C

Referência(s)