Revisão Acesso aberto Revisado por pares

In Vitro Models for the Study of Liver Biology and Diseases: Advances and Limitations

2022; Elsevier BV; Volume: 15; Issue: 3 Linguagem: Inglês

10.1016/j.jcmgh.2022.11.008

ISSN

2352-345X

Autores

Savneet Kaur, Srivatsan Kidambi, Martí Ortega‐Ribera, Lê Thị Thanh Thủy, Natalia Nieto, Victoria C. Cogger, Wei‐Fen Xie, Frank Tacke, Jordi Gracia‐Sancho,

Tópico(s)

Pancreatic function and diabetes

Resumo

In vitro models of liver (patho)physiology, new technologies, and experimental approaches are progressing rapidly. Based on cell lines, induced pluripotent stem cells or primary cells derived from mouse or human liver as well as whole tissue (slices), such in vitro single- and multicellular models, including complex microfluidic organ-on-a-chip systems, provide tools to functionally understand mechanisms of liver health and disease. The International Society of Hepatic Sinusoidal Research (ISHSR) commissioned this working group to review the currently available in vitro liver models and describe the advantages and disadvantages of each in the context of evaluating their use for the study of liver functionality, disease modeling, therapeutic discovery, and clinical applicability. In vitro models of liver (patho)physiology, new technologies, and experimental approaches are progressing rapidly. Based on cell lines, induced pluripotent stem cells or primary cells derived from mouse or human liver as well as whole tissue (slices), such in vitro single- and multicellular models, including complex microfluidic organ-on-a-chip systems, provide tools to functionally understand mechanisms of liver health and disease. The International Society of Hepatic Sinusoidal Research (ISHSR) commissioned this working group to review the currently available in vitro liver models and describe the advantages and disadvantages of each in the context of evaluating their use for the study of liver functionality, disease modeling, therapeutic discovery, and clinical applicability. SummaryThis review provides a glimpse on the most advanced methods for studying liver sinusoidal biology in vitro to uncover liver disease pathophysiology focusing on liver-on-a-chip and microfluidic devices, liver scaffolds and matrices, mechanobiological cues, spheroids/organoids, liver slices and omics. This review provides a glimpse on the most advanced methods for studying liver sinusoidal biology in vitro to uncover liver disease pathophysiology focusing on liver-on-a-chip and microfluidic devices, liver scaffolds and matrices, mechanobiological cues, spheroids/organoids, liver slices and omics. Liver disease represents one of the leading causes of death worldwide, and the incidence of some pathologies, such as nonalcoholic fatty liver disease (NAFLD), nonalcoholic steatohepatitis (NASH), and liver cancer, continues to increase.1Karlsen T.H. Sheron N. Zelber-Sagi S. et al.The EASL–Lancet Liver Commission: protecting the next generation of Europeans against liver disease complications and premature mortality.Lancet. 2022; 399: 61-116Abstract Full Text Full Text PDF PubMed Google Scholar Despite years of research, liver diseases still have limited treatment options in the clinic. This paucity of treatments is explained partly by the limitations of traditional in vitro tools and animal models that do not accurately mimic the clinical pathophysiology of diseases and have a low accuracy for drug discovery purposes. Indeed, several studies have shown that traditional cell culture methodologies do not reflect the complexity of a human liver in vivo and thus cannot predict drug sensitivity. In contrast, animal models differ in biology compared with human pathologies, which explains why promising therapies tested in animal models often fail when tested in human beings and, unfortunately, the field of hepatology has numerous recent examples of failures in clinical phases.2Nevzorova Y.A. Boyer-Diaz Z. Cubero F.J. Gracia-Sancho J. Animal models for liver disease – a practical approach for translational research.J Hepatol. 2020; 73: 423-440Abstract Full Text Full Text PDF PubMed Scopus (87) Google Scholar With the advent of precision medicine, which offers much hope for individual patient outcomes, there is increased demand for robust and patient-specific tools to better improve our understanding and treatment of complex and multifactorial diseases such as liver diseases. Advances in vascular biology, microfluidics, and bioengineering have led to the development of sophisticated in vitro models that could fill this gap (Figure 1). In addition, omics techniques provide further insight to preclinical research in hepatology. In this review, we discuss the benefits and limitations of advanced in vitro research techniques that presently are being applied to the study of liver diseases and further critique how these tools may provide insight into the prediction of patient responses to a therapy. During the 21st century, the development of biology-inspired devices aimed at mimicking the sinusoidal niche integrating microfluidics led to the rapidly evolving liver-on-a-chip (LoC) technology.3Polidoro M.A. Ferrari E. Marzorati S. et al.Experimental liver models: from cell culture techniques to microfluidic organs-on-chip.Liver Int. 2021; 41: 1744-1761Crossref PubMed Scopus (13) Google Scholar The design of these in vitro liver-resembling tools, which have been reviewed extensively by Ortega-Ribera et al4Ortega-Ribera M. Yeste J. Villa R. Gracia-Sancho J. Nanoengineered biomaterials for the treatment of liver diseases.in: Mozafari M. Rajadas J. Kaplan D. Nanoengineered biomaterials for regenerative medicine. Elsevier, 2018: 417-441Google Scholar and is out of the scope of the present review, is inspired in sinusoidal cell biology, architecture, and hemodynamics, but materialized under each research teams’ eyes in terms of appearance, size, fabrication procedures, costs, and microfluidics integration, leading to significant variation in the finalized product. The latest advances in the field include chronic liver disease–specific devices, LoC models designed to study key pathophysiological processes in the development of liver disease, and to understand the interconnection with other organs-on-chip to better depict liver functions and systemic implications (Figure 2). Multi-organ chips, for instance, liver-, adipose-, and gut-on-a-chip connected, may be particularly suitable to understand organ-crosstalk in chronic liver disease (CLD), such as NAFLD/NASH or cholangiopathies.5Shroff T. Aina K. Maass C. et al.Studying metabolism with multi-organ chips: new tools for disease modelling, pharmacokinetics and pharmacodynamics.Open Biol. 2022; 12210333Crossref PubMed Scopus (0) Google Scholar In recent years, disease-focused LoC devices mimicking some of the landmark etiological characteristics of CLD have been developed. Fat accumulation in hepatocytes, occurring in NAFLD/NASH, has been represented in LoC under the combination of glucose and free fatty acids (usually a 2:1 ratio of oleic and palmitic acid).6Hassan S. Sebastian S. Maharjan S. et al.Liver-on-a-chip models of fatty liver disease.Hepatology. 2020; 71: 733-740Crossref PubMed Scopus (39) Google Scholar Antifibrotic compounds such as obeticholic acid, elafibranor,7Du K. Li S. Li C. et al.Modeling nonalcoholic fatty liver disease on a liver lobule chip with dual blood supply.Acta Biomater. 2021; 134: 228-239Crossref PubMed Scopus (7) Google Scholar pioglitazone, or metformin8Teng Y. Zhao Z. Tasnim F. et al.A scalable and sensitive steatosis chip with long-term perfusion of in situ differentiated HepaRG organoids.Biomaterials. 2021; 275120904Crossref Scopus (12) Google Scholar,9Wang Y. Wang H. Deng P. et al.Modeling human nonalcoholic fatty liver disease (NAFLD) with an organoids-on-a-chip system.ACS Biomater Sci Eng. 2020; 6: 5734-5743Crossref PubMed Scopus (4) Google Scholar showed promising results in reducing lipid droplets in these in vitro settings. Indeed, the anti-NASH agent lanifibranor efficiently reduced hepatocytic lipid accumulation,10Lefere S. Puengel T. Hundertmark J. et al.Differential effects of selective- and pan-PPAR agonists on experimental steatohepatitis and hepatic macrophages.J Hepatol. 2020; 73: 757-770Abstract Full Text Full Text PDF PubMed Scopus (91) Google Scholar and improved human hepatocyte and hepatic stellate cell (HSC) phenotype11Boyer-Diaz Z. Aristu-Zabalza P. Andrés-Rozas M. et al.Pan-PPAR agonist lanifibranor improves portal hypertension and hepatic fibrosis in experimental advanced chronic liver disease.J Hepatol. 2021; 74: 1188-1199Abstract Full Text Full Text PDF PubMed Scopus (0) Google Scholar in a LoC model but not in 2-dimensional (2D) cell cultures, supporting the specific value of multicellular LoC devices over traditional monocell cultures. Alcohol-associated liver disease has been addressed in several publications, focusing on its impact in sinusoidal cell biology during development12Deng J. Chen Z. Zhang X. et al.A liver-chip-based alcoholic liver disease model featuring multi-non-parenchymal cells.Biomed Microdevices. 2019; 21: 57Crossref PubMed Scopus (0) Google Scholar or recovery from alcohol (abstinence) either with perfused spheroid13Lee J. Choi B. No D.Y. et al.A 3D alcoholic liver disease model on a chip.Integr Biol (Camb). 2016; 8: 302-308Crossref PubMed Google Scholar or layered cultures.14Nawroth J.C. Petropolis D.B. Manatakis D.V. et al.Modeling alcohol-associated liver disease in a human Liver-Chip.Cell Rep. 2021; 36109393Abstract Full Text Full Text PDF PubMed Scopus (10) Google Scholar Importantly, Ortega-Prieto et al15Ortega-Prieto A.M. Skelton J.K. Wai S.N. et al.3D microfluidic liver cultures as a physiological preclinical tool for hepatitis B virus infection.Nat Commun. 2018; 9: 682Crossref PubMed Scopus (133) Google Scholar developed a model for hepatitis B virus (HBV) long-term infection in primary human hepatocytes that recapitulates virus–host interactions and its associated immune effectors. LoC devices using primary cells isolated through standardized protocols from preclinical models of CLD and from patients' liver tissue also have been developed.16Ortega-Ribera M. Fernández-Iglesias A. Illa X. et al.Resemblance of the human liver sinusoid in a fluidic device with biomedical and pharmaceutical applications.Biotechnol Bioeng. 2018; 115: 2585-2594Crossref PubMed Scopus (25) Google Scholar,17Fernandez-Igleasias A. Ortega Ribera M. Guixé-Muntet S. Gracia-Sancho J. 4 In 1: antibody-free protocol for isolating the main hepatic cells from healthy and cirrhotic single rat livers.J Cell Mol Med. 2019; 23: 877-886Crossref PubMed Scopus (8) Google Scholar These specialized LoC settings may widen the current knowledge on disease dynamics and provide potential applicability as in vitro preclinical models for drug screening. Even though LoC complexity has increased outstandingly since the initial models/prototypes, the intricacy of the whole liver still is under-represented. In this regard, several scientists brought the attention and focused their studies on specific processes or structures. For example, the essential features of the bile duct containing primary mouse cholangiocytes,18Du Y. Khandekar G. Llewellyn J. et al.A bile duct-on-a-chip with organ-level functions.Hepatology. 2020; 71: 1350-1363Crossref PubMed Scopus (31) Google Scholar the unique vasculature organization of the liver,19Ya S. Ding W.G. Li S. et al.On-chip construction of liver lobules with self-assembled perfusable hepatic sinusoid networks.ACS Appl Mater Interfaces. 2021; 13: 32640-32652Crossref PubMed Scopus (6) Google Scholar the sinusoidal zonation within LoC devices,20Kang Y.B.A. Eo J. Mert S. et al.Metabolic patterning on a chip: towards in vitro liver zonation of primary rat and human hepatocytes.Sci Rep. 2018; 8: 8951Crossref PubMed Scopus (60) Google Scholar neutrophil recruitment and interaction with liver sinusoidal endothelial cells (LSECs) after lipopolysaccharide stimulation,21Du Y. Li N. Yang H. et al.Mimicking liver sinusoidal structures and functions using a 3D-configured microfluidic chip.Lab Chip. 2017; 17: 782-794Crossref PubMed Google Scholar or drug metabolism and toxicity22Jang K.J. Otieno M.A. Ronxhi J. et al.Reproducing human and cross-species drug toxicities using a liver-chip.Sci Transl Med. 2019; 11 (eaax5516)Crossref Scopus (186) Google Scholar now are embedded in available LoC systems. Moreover, CLD has been reported extensively as a systemic syndrome with major extrahepatic implications.23Tsochatzis E.A. Bosch J. Burroughs A.K. Liver cirrhosis.Lancet. 2014; 383: 1749-1761Abstract Full Text Full Text PDF PubMed Scopus (1144) Google Scholar Therefore, the combination of LoC devices now has evolved to the extent that disease-specific models are being combined with others such as intestine, brain, kidney,24Theobald J. Ghanem A. Wallisch P. et al.Liver-kidney-on-chip to study toxicity of drug metabolites.ACS Biomater Sci Eng. 2018; 4: 78-89Crossref PubMed Scopus (68) Google Scholar, 25De Gregorio V. Telesco M. Corrado B. et al.Intestine-liver axis on-chip reveals the intestinal protective role on hepatic damage by emulating ethanol first-pass metabolism.Front Bioeng Biotechnol. 2020; 8: 163Crossref PubMed Scopus (6) Google Scholar, 26Wogram E. Svoboda D. Communal C. et al.Human physiomimetic model integrating microphysiological systems of the gut, liver, and brain for studies of neurodegenerative diseases.Sci Adv. 2021; 7eabd1707PubMed Google Scholar or even metastasis niche-on-chip models27Kim J. Lee C. Kim I. et al.Three-dimensional human liver-chip emulating premetastatic niche formation by breast cancer-derived extracellular vesicles.ACS Nano. 2020; 14: 14971-14988Crossref PubMed Scopus (37) Google Scholar to re-create body-on-chip structures to further study the gut–liver–brain axis, systemic drug clearance, or exosome communication between the liver and the tumor microenvironment. However, alongside these advances in multi-organ and etiology-centered approaches, a lack of consensus in cellular sources and mechanobiological cues within the various LoC models remain as unsolved challenges in the field. A key component for the engineering of in vitro liver models is the development of the appropriate scaffold/matrix that recapitulates the hepatic microenvironment well enough to result in realistic functional cells. Several factors affect the efficiency of a scaffold as a support for liver cell growth and function including porosity, pore size, biomechanical properties, and the scaffold design. To simulate the microenvironment of natural extracellular matrix (ECM), substrate design and biomechanical properties are of great significance; hence, bioinspired and biomimetic approaches have been explored to model the healthy or damaged liver (Figure 3). Double layers of collagen have been used for years as a well-established 2D in vitro model for sandwich cultures of hepatocytes.28Dunn J.C.Y. Tompkins R.G. Yarmush M.L. Hepatocytes in collagen sandwich: evidence for transcriptional and translational regulation.J Cell Biol. 1992; 116: 1043-1053Crossref PubMed Google Scholar Recent studies have identified, and characterized, the hepatic matrisome comprising ECM signatures beyond collagen that potentially can provide matrix for in vitro systems to study liver diseases.29Arteel G.E. Naba A. The liver matrisome – looking beyond collagens.JHEP Rep. 2020; 2100115PubMed Google Scholar,30Chen W. Desert R. Ge X. et al.The matrisome genes from hepatitis B–related hepatocellular carcinoma unveiled.Hepatol Commun. 2021; 5: 1571-1585Crossref PubMed Scopus (1) Google Scholar Scaffolds have been fabricated using natural polymers such as gelatin, elastin, silk fibroin, chitosan, chitin, fibrin, and fibrinogen, or synthetic polymers such as polylactic acid, poly(glycolic acid), polyhydroxyalkanoate, and poly (lactic-co-glycolic acid).31Kaur S. Tripathi D.M. Venugopal J.R. Ramakrishna S. Advances in biomaterials for hepatic tissue engineering.Curr Opin Biomed Eng. 2020; 13: 190-196Crossref Scopus (7) Google Scholar Modified versions of biomaterials such as collagen-incorporated poly (lactic-co-glycolic acid) have resulted in enhancement of hepatocyte survival and functions likely owing to an increase in the bioactivity of the newly developed scaffolds.32Brown J.H. Das P. DiVito M.D. et al.Nanofibrous PLGA electrospun scaffolds modified with type I collagen influence hepatocyte function and support viability in vitro.Acta Biomater. 2018; 73: 217-227Crossref PubMed Scopus (61) Google Scholar,33Das P. DiVito M.D. Wertheim J.A. Tan L.P. Collagen-I and fibronectin modified three-dimensional electrospun PLGA scaffolds for long-term in vitro maintenance of functional hepatocytes.Mater Sci Eng C. 2020; 111110723Crossref Scopus (15) Google Scholar Similarly, natural polymers such as silk fibroin have been modified with arginyl-glycyl-aspartic acid (RGD, an integrin-based cell adhesion motif), which has been reported to support the growth of functional hepatocyte clusters.34Janani G. Nandi Samit K. Mandal Biman B. Functional hepatocyte clusters on bioactive blend silk matrices towards generating bioartificial liver constructs.Acta Biomater. 2018; 67: 167-182Crossref PubMed Scopus (46) Google Scholar The modification with RGD also may support attachment of LSECs, known as endothelization of materials.35Bartneck M. Topuz F. Tag C.G. et al.Molecular response of liver sinusoidal endothelial cells on hydrogels.Mater Sci Eng C. 2015; 51: 64-72Crossref PubMed Scopus (8) Google Scholar Efficient spheroid cultures of hepatocytes have been reported on highly porous hydrogel scaffolds composed of alginate and galactosylated chitosan.36Lou R. Xie H. Zheng H. et al.Alginate-based microcapsules with galactosylated chitosan internal for primary hepatocyte applications.Int J Biol Macromol. 2016; 93: 1133-1140Crossref PubMed Scopus (20) Google Scholar In addition, synthetic polymer thin films–based scaffolds allow organized hepatocyte culture and patterned co-culture of hepatocytes with nonparenchymal cells.37Kidambi S. Yarmush R.S. Novik E.R. et al.Oxygen-mediated enhancement of primary hepatocyte metabolism, functional polarization, gene expression, and drug clearance.Proc Natl Acad Sci U S A. 2009; 106: 15714-15719Crossref PubMed Scopus (160) Google Scholar, 38Kidambi S. Sheng L. Yarmush M.L. et al.Patterned co-culture of primary hepatocytes and fibroblasts using polyelectrolyte multilayer templates.Macromol Biosci. 2007; 7: 344-353Crossref PubMed Scopus (70) Google Scholar, 39Kidambi S. Lee I. Chan C. Controlling primary hepatocyte adhesion and spreading on protein-free polyelectrolyte multilayer films.J Am Chem Soc. 2004; 126: 16286-16287Crossref PubMed Scopus (72) Google Scholar Recent interest in mechanical signaling has led to the development of scaffolds that can re-create liver stiffness in physiological and pathologic conditions. In this context, heparin hydrogel has been developed to modulate stiffness and has shown that hepatocytes cultured on a softer heparin hydrogel (10 kPa) retained 5 times higher levels of albumin production compared with those on a stiffer heparin gel (110 kPa) after 5 days.40You J. Park S.A. Shin D.S. et al.Characterizing the effects of heparin gel stiffness on function of primary hepatocytes.Tissue Eng Part A. 2013; 19: 2655-2663Crossref PubMed Scopus (0) Google Scholar Primary hepatocytes grown on modified polyacrylamide gels with cell-adhesive ligands are shown to reduce albumin production and impair hepatocyte function with increasing stiffness.41Chen A.A. Khetani S.R. Lee S. et al.Modulation of hepatocyte phenotype in vitro via chemomechanical tuning of polyelectrolyte multilayers.Biomaterials. 2009; 30: 1113-1120Crossref PubMed Scopus (74) Google Scholar,42Semler E.J. Lancin P.A. Dasgupta A. Moghe P.V. Engineering hepatocellular morphogenesis and function via ligand-presenting hydrogels with graded mechanical compliance.Biotechnol Bioeng. 2005; 89: 296-307Crossref PubMed Scopus (0) Google Scholar Desai et al43Desai S.S. Tung J.C. Zhou V.X. et al.Physiological ranges of matrix rigidity modulate primary mouse hepatocyte function in part through hepatocyte nuclear factor 4 alpha.Hepatology. 2016; 64: 261-275Crossref PubMed Scopus (99) Google Scholar used polyacrylamide gels to tune the substrate stiffness and showed that fibrotic levels of stiffness significantly inhibit hepatocyte-specific functions in part through the inhibition of the hepatocyte nuclear factor 4α transcriptional network mediated via the Rho/Rho-associated protein kinase pathway. An innovative platform called bio-engineered adhesive siloxane substrate with tunable stiffness based on a polydimethyl siloxane substrate in combination with polyelectrolyte multilayer film-coating technology was developed to engineer mechanically tunable substrates mimicking physiologic and pathologic liver stiffness.44Natarajan V. Moeun Y. Kidambi S. Exploring interactions between primary hepatocytes and non-parenchymal cells on physiological and pathological liver stiffness.Biology (Basel). 2021; 10: 408PubMed Google Scholar, 45Kidambi S. Stiffness and hepatocytes function in vitro.in: In: Mueller, S. (eds) Liver Elastography. Springer, 2020: 645-660Crossref Scopus (0) Google Scholar, 46Natarajan V. Moeller M. Casey C.A. Harris Edward N. Matrix stiffness regulates liver sinusoidal endothelial cell function mimicking responses in fatty liver disease.bioRxiv. 2020; 27Google Scholar, 47Ganesan M. Dagur R.S. Makarov E. et al.Matrix stiffness regulate apoptotic cell death in HIV-HCV co-infected hepatocytes: importance for liver fibrosis progression.Biochem Biophys Res Commun. 2018; 500: 717-722Crossref PubMed Scopus (0) Google Scholar, 48Natarajan V. Berglund E.J. Chen D.X. Kidambi S. Substrate stiffness regulates primary hepatocyte functions.RSC Adv. 2015; 5: 80956-80966Crossref PubMed Google Scholar More recently, 3-dimensional (3D) bioprinting has emerged for precise spatial positioning of both cells and biomaterials or bioinks such as alginate together in 3D complex geometries and providing mechanical support.49Kim Y. Kang K. Jeong J. et al.Three-dimensional (3D) printing of mouse primary hepatocytes to generate 3D hepatic structure.Ann Surg Treat Res. 2017; 92: 67-72Crossref PubMed Scopus (0) Google Scholar,50Yang H. Sun L. Pang Y.H. et al.Three-dimensional bioprinted hepatorganoids prolong survival of mice with liver failure.Gut. 2021; 70: 567-574Crossref PubMed Scopus (51) Google Scholar Nguyen et al51Nguyen D.G. Funk J. Robbins J.B. et al.Bioprinted 3D primary liver tissues allow assessment of organ-level response to clinical drug induced toxicity in vitro.PLoS One. 2016; 11e0158674Crossref Scopus (238) Google Scholar have bioprinted hepatocytes and nonparenchymal cells in 3D architecture and developed models of drug-induced liver injury. Recent studies have printed liver cells along with a liver decellularized ECM bioink, creating an environment for maximal cellular function.52Sharma A. Rawal P. Tripathi D.M. et al.Upgrading hepatic differentiation and functions on 3d printed silk-decellularized liver hybrid scaffolds.ACS Biomater Sci Eng. 2021; 7: 3861-3873Crossref PubMed Scopus (8) Google Scholar, 53Lee H. Chae S. Kim J.Y. et al.Cell-printed 3D liver-on-a-chip possessing a liver microenvironment and biliary system.Biofabrication. 2019; 11025001Crossref Scopus (83) Google Scholar, 54Lee H. Han W. Kim H. et al.Development of liver decellularized extracellular matrix bioink for three-dimensional cell printing-based liver tissue engineering.Biomacromolecules. 2017; 18: 1229-1237Crossref PubMed Scopus (0) Google Scholar With 3D bioprinting, vascular and biliary fluidic channels also have been created successfully in the LoC device format.53Lee H. Chae S. Kim J.Y. et al.Cell-printed 3D liver-on-a-chip possessing a liver microenvironment and biliary system.Biofabrication. 2019; 11025001Crossref Scopus (83) Google Scholar 3D bioprinting of spheroids and organoids represent the next level of technological advancement for creating the highly complex liver architecture.55Rawal P. Tripathi D.M. Ramakrishna S. Kaur S. Prospects for 3D bioprinting of organoids.Bio-Des Manuf. 2021; 4: 627-640Crossref Scopus (15) Google Scholar As described earlier, over the past century, 2D cell cultures have been used as common in vitro models to study cellular responses to stimulation and allowing the construction of low-cost, simple, and well-accepted models of liver disease. However, they do not precisely reflect the true physiological state of cells in vivo owing to the absence of structural, mechanical, and biochemical cues, as well as the interaction between cells and extracellular matrices.56Duval K. Grover H. Han L.H. et al.Modeling physiological events in 2D vs. 3D cell culture.Physiology. 2017; 32: 266-277Crossref PubMed Scopus (804) Google Scholar To overcome this limitation, novel 3D cell culture platforms including liver spheroid and organoid cultures are being created to better mimic the in vivo conditions.57Ramaiahgari S.C. Den Braver M.W. Herpers B. et al.A 3D in vitro model of differentiated HepG2 cell spheroids with improved liver-like properties for repeated dose high-throughput toxicity studies.Arch Toxicol. 2014; 88: 1083-1095PubMed Google Scholar, 58Sato K. Zhang W. Safarikia S. et al.Organoids and spheroids as models for studying cholestatic liver injury and cholangiocarcinoma.Hepatology. 2021; 74: 491-502Crossref PubMed Scopus (20) Google Scholar, 59Takebe T. Zhang R.R. Koike H. et al.Generation of a vascularized and functional human liver from an iPSC-derived organ bud transplant.Nat Protoc. 2014; 9: 396-409Crossref PubMed Scopus (261) Google Scholar, 60Sekine K. Takebe T. Taniguchi H. Liver regeneration using cultured liver bud.Methods Mol Biol. 2017; 1597: 207-216Crossref PubMed Scopus (2) Google Scholar, 61Takebe T. Sekine K. Kimura M. et al.Massive and reproducible production of liver buds entirely from human pluripotent stem cells.Cell Rep. 2017; 21: 2661-2670Abstract Full Text Full Text PDF PubMed Scopus (191) Google Scholar 3D spheroids are produced via self-assembly, in which monodispersed cells form 3D microtissues called multicellular spheroids, and mimic natural processes that occur during embryogenesis, morphogenesis, and organogenesis.62Achilli T.M. Meyer J. Morgan J.R. Advances in the formation, use and understanding of multi-cellular spheroids.Expert Opin Biol Ther. 2012; 12: 1347-1360Crossref PubMed Scopus (345) Google Scholar 3D organoids derive from either pluripotent stem cells, neonatal tissue stem cells, or adult stem cells/adult progenitors, in which cells spontaneously self-organize into properly differentiated functional cell types and progenitors, resembling their in vivo counterparts and recapitulating at least some functions of the organ.63Huch M. Koo B.K. Modeling mouse and human development using organoid cultures.Development. 2015; 142: 3113-3125Crossref PubMed Scopus (301) Google Scholar In the field of studying liver diseases, recent innovation of hepatic 3D spheroids also offer a promising application via combination of 3D printing–based techniques and HepG2 liver spheroid culture models to develop in situ quantitative evaluation and high-throughput monitoring of drug-induced hepatotoxicity.64Hong S. Song J.M. A 3D cell printing-fabricated HepG2 liver spheroid model for high-content in situ quantification of drug-induced liver toxicity.Biomater Sci. 2021; 9: 5939-5950Crossref PubMed Google Scholar HepG2 cell-laden hydrogel constructs were 3D printed in the shape of a cross on the mini-9-well plate, which showed the HepG2 liver spheroids embedded in the gelatin-alginate hydrogel. On the 6th day of culture, HepG2 liver spheroids exposed to varying concentrations of troglitazone and nefazodone were used to predict hepatotoxicity. This model provided a promising tool for screening and characterization of hepatotoxicity in a 3D spheroid-embedded hydrogel system that more closely resembles conditions in vivo. In 2013, Takebe et al59Takebe T. Zhang R.R. Koike H. et al.Generation of a vascularized and functional human liver from an iPSC-derived organ bud transplant.Nat Protoc. 2014; 9: 396-409Crossref PubMed Scopus (261) Google Scholar described the in vitro generation of 3D liver bud organoids from human induced pluripotent stem cell–derived hepatic endoderm cells co-cultured with endothelial and mesenchymal lineages. Interestingly, when these liver buds were ectopically transplanted at various sites including the cranium, subrenal capsule, and distal and proximal-mesentery in immunodeficient mice they were able to rescue the drug-induced lethal liver failure model.61Takebe T. Sekine K. Kimura M. et al.Massive and reproducible production of liver buds entirely from human pluripotent stem cells.Cell Rep. 2017; 21: 2661-2670Abstract Full Text Full Text PDF PubMed Scopus (191) Google Scholar,65Takebe T. Sekine K. Enomura M. et al.Vascularized and functional human liver from an iPSC-derived organ bud transplant.Nature. 2013; 499: 481-484Crossref PubMed Scopus (1348) Google Scholar These studies have provided a promising new approach to study regenerative medicine and to translate these techniques for treating patients with end-stage liver failure.65Takebe T. Sekine K. Enomura M. et al.Vascularized and functional human liver from an iPSC-derived organ bud transplant.Nature. 2013; 499: 481-484Crossref PubMed Scopus (1348) Google Scholar Furthermore, single-cell RNA sequencing (scRNA-seq) data from human liver bud organoids showed several aspects of heterotypic interlineage communication and organ development.66Camp J.G. Sekine K. Gerber T. et al.Multilineage communication regulates human liver bud development from pluripotency.Nature. 2017; 546: 533-538Crossref PubMed Scopus (298) Google Scholar Interestingly, Shinozawa et al67Shinozawa T. Kimura M. Cai Y. et al.High-fidelity drug-induced liver injury screen using human pluripotent stem cell-derived organoids.Gastroenterology. 2021; 160: 831-846.e10Abstract Full Text Full Text PDF PubMed Scopus (75) Google Scholar reported a simple, robust, and high-throughput human liver organoid system to measure bile transport activity by live fluor

Referência(s)