Organoid > Volume 4; 2024 > Article
Yoon, Yoon, and Jang: Transformative potential of three-dimensional bioprinting technology for advanced organoid research

Abstract

Organoid research has emerged as a transformative field in biomedicine, focusing on the in vitro development of 3-dimensional (3D) structures that mimic human organs. Derived from various types of stem cells, organoids closely replicate human organ structures and functions, offering significant advantages over 2-dimensional cell cultures and animal models, particularly for drug development, tissue engineering, and precision medicine. Recent innovations, including the integration of biofabrication technologies, have significantly increased the structural complexity and maturity of organoids, expanding their biomedical applications. A critical factor in organoid culture is the utilization of the extracellular matrix (ECM), particularly decellularized ECM hydrogels. These hydrogels are instrumental in organoid growth and development, effectively simulating in vivo environments and supporting organoid functionality across various organ systems. The integration of 3D bioprinting technology into organoid research marks a transformative shift that has enabled the creation of intricate and customized structures. This review demonstrates that these technological innovations have not only revolutionized tissue engineering and regenerative medicine, but also hold immense potential for pharmacology, disease modeling, and personalized medical interventions. The synergistic integration of these technologies presents a promising future for medical research, paving the way for significant advancements in disease modeling, drug discovery, and the development of patient-specific treatments, and marking our entry into a new era of precision medicine and personalized healthcare solutions.

Introduction

Organoids, defined as 3-dimensional (3D) organ-like structures grown in vitro, represent a significant breakthrough in both biological and clinical research. These structures offer a near-physiological reproduction of native tissues and have emerged as transformative alternatives to traditional monolayer cell cultures and animal models [1,2]. The limitations of 2-dimensional (2D) cell monolayers—namely, the lack of complexity of tissue interactions and the often-inconsistent human relevance of animal models—have highlighted the need for more accurate in vitro 3D models that can replicate human organ structures and functions [3]. Organoids are primarily constructed from cells capable of self-renewal and self-organization, including embryonic stem cells, induced pluripotent stem cells, primary tissues, adult stem cells, and cancer stem cells [4]. These cells undergo proliferation, differentiation, and self-organization to form structures that closely resemble in vivo organ systems [5]. With advancements in stem cell biology and biomaterials, a variety of human organoids have been developed that effectively simulate the complex structures and functionalities of organs, such as the brain, intestine, liver, kidney, lung, and various tumors [6-11].
Organoid research has been significantly influenced by two key advancements: the exploration of decellularized extracellular matrix (dECM) hydrogels and their integration with 3D bioprinting technology [12]. dECM hydrogels, which are vital for organ development and function, provide a platform for cultivating organoids that replicate physiological conditions more closely and facilitate appropriate cell behavior, replication, and disease modeling [12,13]. Furthermore, 3D bioprinting technology marks a transformative phase in organoid research by enhancing the structural and functional complexity of organoid models and expanding their application in pharmacology and regenerative medicine [14].
Our review explores the transformative potential of dECM hydrogels for advanced organoid culture and the integration of 3D bioprinting technology into organoid research (Fig. 1). We delve into the diverse applications of dECM hydrogels in various organ systems, highlighting their crucial role in maintaining the structural integrity and enhancing the functional attributes of tissues and organs. Moreover, 3D bioprinting technology has been shown to mimic human tissue architecture, augmenting the biological relevance and functionality of organoids. These advancements not only represent a paradigm shift in tissue engineering and regenerative medicine, but also open new avenues for precise disease modeling, drug testing, and personalized therapeutic strategies [15].
At the forefront of this revolution has been the synergistic application of tissue-specific dECM hydrogels and 3D bioprinting technology, which play a crucial role in creating more physiologically accurate organoids [16]. The convergence of these technologies not only advances our understanding of complex medical conditions, but also opens doors to new possibilities for effective and personalized healthcare solutions [17]. Hence, this integration signifies a promising future in medical research with significant implications for disease modeling, drug discovery, and the development of patient-specific treatments [14].

Transformative potential of decellularized extracellular matrix hydrogel for advanced organoid culture

Ethics statement: This study was a literature review of previously published studies and was therefore exempt from institutional review board approval.
Previous methodologies for organoid culture primarily relied on the influence of biochemical signals from the composition of the culture medium and intercellular interactions to regulate growth [18]. Recent research has underscored the crucial role of the chemical composition of the extracellular matrix (ECM), as well as mechanical signals related to the expansion and differentiation of organoids [19]. During organoid formation, the ECM provides niches for stem cells through cell-matrix interactions. The chemical and mechanical characteristics of the ECM significantly affect the cellular phenotype. However, in certain circumstances, the ECM can inadvertently induce aberrant cellular states, such as those observed in diseases or tumors, illustrating the complex interplay between the ECM and cellular behavior [20]. Therefore, the stem cell signaling pathways involved in organoid formation require a highly specific ECM environment [5]. The ECM encloses cells within a complex fibrous network composed of various macromolecules such as glycoproteins, proteoglycans, and glycosaminoglycans. Advancements in our understanding of the extracellular milieu have facilitated the development of sophisticated 3D culture systems for organoids supported by ECM scaffolds. Providing proper ECM cues to cells for organoid formation and culture is crucial [21]. Diverse scaffold systems, including Matrigel, ECM, natural polymers, and synthetic polymers, have been tailored to meet the requirements of organoid cultures for application in clinical settings [22].
Each system has its advantages and disadvantages for organoid culture. Matrigel is one of the most common substrates for organoid culture because it can be readily obtained as a commercialized product and is easy to handle [23]. Matrigel is composed of various elements, including laminin, collagen type IV, and growth factors, which are essential for growth of organoids [24]. However, Matrigel is derived from a mouse tumor, raising concerns about the potential transmission of animal pathogens that could impact the immune system [25]. Additionally, Matrigel has ECM components associated with tumors, such as laminin, and is considerably different from matrisomes in normal tissues. Therefore, it could provide improper tissue-specific cues for targeting human organs in organoid culturing systems [26]. Thus, numerous researchers have attempted to find and develop alternatives to Matrigel [27]. Natural polymers, including collagen, fibrin, gelatin, and alginate, are preferred as alternatives to Matrigel because of their bioactivity and similarity to the human ECM. Their microarchitectures, such as microfibers and microspheres, promote the attachment and proliferation of organoids [28]. However, natural polymers may display variable gelation kinetics, complicating the attainment and preservation of the intended scaffold architecture. This inconsistency can affect the uniformity and reproducibility of organoid cultures, which often fail to replicate precise tissue-specific ECM compositions [29]. Synthetic hydrogels have also emerged to support organoid cultures, offering a solution for the complexity and variability of natural matrices [30]. Synthetic polymers, such as polyethylene glycol (PEG), polyacrylamide (PAAM), and polyvinyl alcohol (PVA) are usually chemically defined, and their mechanical properties and chemical composition can be accurately modulated to enhance organoid differentiation [31]. However, synthetic polymers still exhibit low bioactivity and insufficient tissue-specific ECM composition. Therefore, the incorporation of a dECM hydrogel in organoid culture increases the physiological relevance of these models by providing a biomimetic microenvironment and facilitates the investigation of cell behavior in a context that closely resembles native tissue, enabling more accurate and insightful research outcomes.
dECM, which contains tissue-specific compositions and molecules, plays a vital role in organoid culture by providing appropriate biochemical and biophysical signals that stand out from other biomaterials used in organoid cultures, such as Matrigel and synthetic polymers [32]. The tissue-specific nature of the dECM delivers unique biochemical, biophysical, and biomechanical cues to organoids akin to in vivo cell-matrix communication systems [29]. Considering the diverse composition and organization of ECM across different organs, the use of tissue-specific dECM hydrogels offers a customized approach for organoid culture. This method provides tissue-specific dECM content, creating niche environments for stem cells and a biomimetic microstructure that closely mimics the natural organ context [21]. Furthermore, dECM hydrogels have a native tissue-oriented architecture with topographical cues and elastic properties [29]. These cues activate or deactivate the cellular mechanisms underlying cell adhesion, migration, proliferation, and differentiation. Furthermore, Saldin et al. [33] investigated the cellular responses of survival, proliferation, differentiation, and tissue-specific functions using a dECM hydrogel as an organoid culture scaffold. Prior studies have investigated the efficacy of dECM hydrogels as scaffolds for organoid culture across various organoid culture systems. As detailed in Table 1, liver organoids [34], brain organoids [35], endoderm-derived human organoids [16], and islet organoids [36], when combined with a dECM, exhibit improved functions resembling those of native tissues.
Saheli et al. [34] investigated whether a sheep liver dECM hydrogel could promote the physiological function of liver organoids formed with human hepatocarcinoma cells, human endothelial cells, and mesenchymal cells. Self-organized liver organoids expressed albumin and cytochrome P450 3A4 (CYP3A4), key markers of liver function, when cultured in the liver dECM hydrogel (Fig. 2A). The 3D liver dECM gel significantly enhanced the functional activity of these organoids compared to traditional 2D culture and collagen gel-based culture at both the gene (e.g., albumin) and protein levels (albumin secretion and urea production). Similarly, Simsa et al. [35] demonstrated that human embryonic stem cells (hESCs) cultured within brain-derived dECM hydrogels exhibited gene expression patterns and differentiation profiles remarkably similar to those observed in hESCs grown in Matrigel. This finding suggests that brain dECM hydrogels possess the necessary biochemical and structural properties to effectively support hESC culture.
Consequently, these dECM hydrogels have emerged as promising alternatives to Matrigel, particularly for applications requiring a more physiologically relevant ECM environment. This equivalence in performance highlights the potential of brain dECM hydrogels in stem cell research and opens new avenues for exploring tissue-specific interactions in neurodevelopmental studies [35]. Giobbe et al. [16] explored the potential of a small intestine (SI) mucosa/submucosa dECM hydrogel as a culture environment for endoderm-derived human organoids, including gastric, hepatic, pancreatic, and SI organoids. Gastric organoids cultured in this hydrogel expressed epithelial markers, such as zonula occludens-1, epithelial cadherin, and actin, along with gastric-specific markers such as ezrin and mucin-5AC (Fig. 2B). SI organoids were successfully maintained for eight consecutive passages over two months, as shown by morphological analyses (Fig. 2C). Additionally, Giobbe et al. [16] demonstrated the angiogenic potential of endoderm-derived human organoids cultured in SI dECM gel using a chick chorioallantoic membrane assay. Bi et al. [36] investigated the ability of pancreatic dECM hydrogels to induce the self-assembly of human islet organoids via pluripotent stem cell differentiation. Islet organoids cultured on Matrigel-coated substrates supplemented with pancreatic dECM expressed all hormone-secreting pancreatic islet cell types, detected using C-peptide, glucagon, somatostatin, and pancreatic polypeptide expression (Fig. 2D). Therefore, tissue-specific dECM hydrogels hold significant potential as culture scaffolds for organoids. They not only maintain the structure of these organoids but also promote crucial functions specific to each tissue and organ.

Integration of 3D bioprinting technology in organoid research for its biomedical applications

The integration of 3D bioprinting technology into organoid development represents a groundbreaking advance in biomedical research. This technology offers a unique combination of meticulous precision and control over cell and biomaterial placement, along with the ability to produce complex large-scale structures [37]. As highlighted by Murphy and Atala, this precise control enables the creation of highly organized and structured organoid models that surpass the traditional methods in accurately mimicking the human tissue architecture [12]. The capabilities of 3D bioprinting extend beyond the micrometer and millimeter scales. Kang et al. [38] demonstrated the ability to bioprint complex structures at the centimeter scale, including lumens, branched vasculature, and tubular intestinal epithelia with in vivo-like features. The scalability of 3D bioprinting is crucial for high-throughput screening and clinical applications. Furthermore, 3D bioprinting enables the customization of organoids to cater to specific patient needs or disease characteristics, thereby advancing personalized medical applications, as discussed by Zhang et al. [39]. This aspect of customization is a key feature of bioprinting-assisted tissue emergence, a concept introduced by Brassard et al. [40] that utilizes a syringe-based extrusion system paired with a manually controlled stage. The integration of these capabilities, ranging from micro-to macroscales, coupled with customization potential, highlights the revolutionary role of 3D bioprinting technology in organoid research, which broadens our knowledge and abilities in fields such as development, regeneration, and disease modeling [41].
These advancements are aligned with significant progress in organoid research. For example, in pharmacology, significant strides have been made in bioprinting liver organoids that mimic the critical functions of human liver tissue, primarily for drug toxicity assessments [42]. This achievement exemplifies the potential of 3D bioprinting to create complex organ structures. Additionally, skin organoids with integrated vascular structures have been developed to improve wound healing in animal models [43]. Similarly, previous cancer research demonstrated the use of bioprinted breast cancer organoids to study tumor growth and drug resistance, marking a significant advancement in the understanding of cancer biology [44,45]. Moreover, the development of a hydrogel-in-hydrogel live bioprinting system that guides neural axon directionality in organotypic spinal cords demonstrates the potential of 3D bioprinting in neural applications, particularly in spinal cord regeneration [41]. Collectively, these diverse applications showcase the broad spectrum of 3D bioprinting in organoid research, impacting fields ranging from pharmacology to personalized medicine. This technology opens new avenues for treatment development and offers a deeper understanding of complex biological processes.
The integration of 3D bioprinting into organoid research has led to a series of innovative advancements across various medical fields, reflecting the vast potential and impact of this technology [46]. This was exemplified by a groundbreaking vascularized lung cancer (LC) model by Choi et al. [47]. This model integrated patient-derived LC organoids (LCOs), lung fibroblasts, and perfusable vessels using 3D bioprinting (Fig. 3A). Notably, the use of a lung-derived dECM hydrogel in this model played a critical role in creating an authentic LC microenvironment, particularly when incorporating lung fibroblasts derived from idiopathic pulmonary fibrosis (Fig. 3B). This approach boosted cell proliferation and increased the expression of drug resistance genes in LCOs, offering insights into developing targeted therapies and identifying biomarkers for fibrosis in LC patients [47].
Simultaneously, a breakthrough in gastric cancer research was achieved with the development of a vascularized organoid model (VOM) (Fig. 4A). Kim et al. integrated patient-derived gastric cancer organoids, functional endothelium, and stomach-derived dECM (Fig. 4B) [48]. This innovative VOM exhibited the potential to contribute to personalized medicine efforts by accurately predicting individual patient responses to therapies targeting the vascular endothelial growth factor receptor 2 (VEGFR2). A key example is the response to ramucirumab, a U.S. Food and Drug Administration-approved therapy that inhibits VEGFR2. This treatment has been effective in curbing cancer cell growth and improving survival rates in gastric cancer patients, reflecting real-world treatment outcomes (Fig. 4C).
Table 2 presents the use of 3D bioprinting technology, which has substantially advanced the biological accuracy and functional capacity of organoids, leading to more realistic in vitro models that reflect the complex microenvironments of living human tissues [41,47-50]. These advancements have enabled the more accurate replication of tissue microenvironments, leading to more precise disease modeling and effective drug testing, as described by Wang et al. [51]. Furthermore, these 3D bioprinted organoids have demonstrated improved longevity and stability, facilitating longer-term studies and experiments, and marking a notable advancement over traditional organoid cultures [5]. The integration of 3D bioprinting with innovative technologies like microfluidics and biosensors has further increased the sophistication and functional relevance of these 3D organoid models [52-54].

Conclusion and future perspectives

The integration of 3D bioprinting technologies and dECM hydrogels in the field of organoid research represents a significant leap toward revolutionizing our understanding of human biology and advancing medical practices. The synergy between these advanced techniques dramatically enhances the precision, complexity, and fidelity of bioprinted structures, thereby expanding the potential applications of organoids in biomedical research, regenerative medicine, and development of personalized therapeutic strategies. Although dECM hydrogels provide a biomimetic scaffold that facilitates organoid growth, their inherent limitations in mechanical strength and printability pose challenges. These challenges can be addressed by incorporating innovative materials such as gelatin methacrylate, hyaluronic acid, alginate, and gelatin [55]. Such modifications not only increase the structural integrity and longevity of organoid models but also facilitate the creation of more complex and faithful representations of native tissue architecture and microenvironments [5,56].
However, advancing these technologies entails navigating the intricate tradeoffs between the precision of 3D bioprinting technology and the complex microenvironments that organoid cultures aim to mimic. As previously mentioned, the integration of vascular networks and functional components within a bioprinted organoid is essential for mimicking dynamic tissue interactions, whereas preserving natural organoid development within 3D bioprinting constraints poses significant challenges [57-60]. Furthermore, the introduction of new materials (e.g., other natural and synthetic polymers) into dECM hydrogels can alter their tissue-specific properties, requiring careful adjustments to balance their benefits and potential drawbacks [61]. The scalability of bioprinting for high-throughput applications such as drug discovery and personalized medicine presents significant opportunities and challenges [12,62,63]. Overcoming limitations related to the resolution, scalability, and slow maturation processes of current 3D bioprinting technologies is essential [64,65]. Moreover, as the field matures, the development of standardized protocols and regulatory guidelines is crucial to ensure the safety, efficacy, and ethical deployment of bioprinted products [66,67].
Blatchley and Anseth have emphasized the importance of middle-out strategies in organoid development. These strategies integrate the macroscale control of top-down approaches with the precise cellular and extracellular assembly characteristics of bottom-up methods. This ensures precise guidance for organoids to develop with specific geometries, complex structures, and functions. [68]. This 3D bioprinting-technology-based approach to spatiotemporal tissue engineering achieves an unparalleled level of biomimicry, increasing the clinical relevance of organoids by ensuring precise control over engineered cellular environments and providing a balanced framework for cellular self-organization [68]. This combination of structural advantages and detailed modular construction advances the field toward addressing the challenges and maximizing the potential of 3D bioprinting technology and organoid research to revolutionize healthcare. Continued investment in translational research is essential to validate the therapeutic potential of bioprinted organoids in clinical applications, paving the way for a new era in precision medicine and personalized healthcare solutions that offer profound insights into human biology and innovative therapeutic strategies.

NOTES

Conflict of interest

No potential conflict of interest relevant to this article was reported.

Funding

This research was supported by the Korean Fund for Regenerative Medicine, funded by the Ministry of Science and ICT and Ministry of Health and Welfare (No. 21A0104L1, Republic of Korea), and supported by a National Research Foundation of Korea (NRF) grant funded by the Korean government (MSIT) (No. 2021R1A2C2004981). This work was also supported by the Alchemist Project 1415180884 (20012378, Development of Meta Soft Organ Module Manufacturing Technology without Immunity Rejection and Module Assembly Robot System), funded By the Ministry of Trade, Industry & Energy (MOTIE, Korea).

Data availability

Not applicable. This study did not generate any new data.

Fig. 1.
Overview of the application of dECM hydrogels and 3-dimensional (3D) bioprinting technology in advanced organoid research. Illustration by the authors. dECM, decellularized extracellular matrix.
organoid-2024-4-e8f1.jpg
Fig. 2.
Utilization of dECM hydrogels in various organoid cultures. (A) Albumin and CYP3A4 gene expression in liver organoids. This panel shows the expression levels of albumin and CYP3A4 in self-organized liver organoids cultured in liver dECM hydrogel. Reproduced from Saheli et al. J Cell Biochem 2018;119:4320-33, with permission from John Wiley & Sons Inc. [34] (B) Gastric organoid development in dECM hydrogel. Displaying 7-day-old gastric organoids cultivated in dECM hydrogel, demonstrating the expression of both epithelial and gastric-specific markers. Reproduced from Giobbe et al. Nat Commun 2019;10:5658, according to the Creative Commons license [16]. (C) Morphological assessment of human pediatric SI organoids. This image depicts the morphology of human pediatric SI organoids over 8 consecutive passages in dECM hydrogels. Reproduced from Giobbe et al. Nat Commun 2019; 10:5658, according to the Creative Commons license [16]. (D) Pancreatic islet cell type detection in islet organoids. This panel highlights the identification of all hormone-secreting cell types within pancreatic islet organoids, achieved through the addition of pancreatic dECM (pdECM) to Matrigel. Reproduced from Bi et al. ACS Biomater Sci Eng 2020;6:4155-65, with permission from ACS Publications [36]. CYP3A4, cytochrome P450 3A4; dECM, decellularized extracellular matrix; pdECM, pancreatic dECM; SI, small intestine.
organoid-2024-4-e8f2.jpg
Fig. 3.
Development of a vascularized 3-dimensional bioprinted lung cancer organoids (LCOs) model. (A) Illustrated steps for constructing the vascularized LCOs, detailing the integration of cellular components and vascular network development. (B) Visualization of the functional organoid model showing the perfusable vascular network. Red fluorescence signifies RFP-HUVECs, illustrating the vascular structures, while green fluorescence identifies the fibroblasts within the matrix. White asterisks highlight the lung cancer organoids. The image confirms the successful formation of a perfusable vasculature and the intimate interaction among the LCOs, HUVECs, and fibroblasts. All images were reproduced from Choi et al. Biofabrication 2023;15:034104, with permission from IOP Publishing [47]. RFP, red fluorescent protein; HUVEC, human umbilical vein endothelial cell.
organoid-2024-4-e8f3.jpg
Fig. 4.
Establishment of a bioprinted vascularized organoid model (VOM). (A) Illustration of the VOM assembly, highlighting a functional endothelium alongside patient-derived organoids (PDOs) showing specific sprouting patterns. (B) Confocal image of the bioprinted construct with immunofluorescence staining depicting F-actin, CD31, and DAPI across three different PDOs (GA052, GA265, and GA232) incorporated within the VOM. (C) Staining images showcase pVEGFR2, F-actin in green representing PDOs, CD31 in red illustrating the vascular network, and DAPI in blue marking nuclei within representative PDOs. This panel also indicates a decrease in VEGFR2 expression of PDOs in response to the therapeutic agent Ramucirumab. All images were reproduced from Kim et al. Adv Funct Mater 2024;34:2306676, with permission from John Wiley & Sons Inc. [48].
organoid-2024-4-e8f4.jpg
Table 1.
Comprehensive application of tissue-specific dECM hydrogels in organoid culture
Organoid type Tissue source for dECM hydrogel Key features Reference
Liver organoids Liver Exhibit higher expression of key markers for liver function than 2-dimensional culture and collagen gel. [34]
Brain organoids Brain Show gene expression patterns and differentiation profiles similar to hESCs grown in Matrigel. [35]
Endoderm-derived human organoids Small intestine mucosa/submucosa Express epithelial markers and gastric markers. [16]
Islet organoids Pancreas Induce the self-assembly of human islet organoids from pluripotent stem cell differentiation. [36]

dECM, decellularized extracellular matrix; hESC, human embryonic stem cell.

Table 2.
Overview of 3D bioprinting technology and its applications in organoid research
Organoid type Bioprinting techniques Aims Key features Reference
Liver organoids Droplet‐based bioprinting Fabricated 3D liver tissue for drug toxicity assessments 3D bioprinting technology refines liver models to mimic natural lobule structures, ensuring the organoids maintain structural integrity and a diverse cellular arrangement within these printed formations. [49]
Breast cancer organoids Extrusion printing Fabricated 3D breast tumor models for facilitating drug discovery 1. 3D bioprinted mouse breast tumors and organoids more accurately replicate in vivo conditions. [50]
2. This technique offered an improved vessel-like structure to monitor oxygen conditions.
Liver, SI, and lung cancer organoids Hydrogel-in-hydrogel live bioprinting Investigated the geometry and mechanical properties of hydrogel to enhance neural cell migration around 3D bioprinted cancer organoids 3D bioprinting technology offers precision in sculpting organoid microenvironments, enabling the directed growth and organization of tissues, from guiding neural axons in spinal cords to controlling cell behavior in cancer, liver, and lung organoids. [41]
Patient-derived lung cancer organoids Extrusion printing Fabricated vascularized 3D lung cancer organoid model for developing targeted cancer therapies 1. 3D bioprinting technology advances organoid research by producing detailed, vascularized cancer models that closely resemble actual tumors, increasing the accuracy of in vitro studies. [47]
2. It enables the integration of patient-specific elements and disease characteristics like fibrosis, thus enhancing drug response evaluations.
Patient-derived gastric cancer organoids Extrusion printing Constructed vascularized 3D gastric cancer organoid model for developing personalized medicine 1. 3D bioprinting technology enables the creation of vascularized organoid models that incorporate patient-derived gastric cancer organoids, perfusable endothelium, and dECMs, offering a highly predictive platform for assessing individual responses to targeted therapies. [48]
2. This approach mirrors the complex tumor microenvironment and genetic diversity of cancers, significantly advancing personalized medicine by facilitating precise treatment predictions.

3D, 3-dimensional; SI, small intestine; dECM, decellularized extracellular matrix.

REFERENCES

1. Gjorevski N, Sachs N, Manfrin A, Giger S, Bragina ME, Ordóñez-Morán P, et al. Designer matrices for intestinal stem cell and organoid culture. Nature 2016;539:560-4.
crossref pmid pdf
2. Lancaster MA, Knoblich JA. Organogenesis in a dish: modeling development and disease using organoid technologies. Science 2014;345:1247125.
crossref pmid
3. Jensen C, Teng Y. Is it time to start transitioning from 2D to 3D cell culture? Front Mol Biosci 2020;7:33.
crossref pmid pmc
4. Fatehullah A, Tan SH, Barker N. Organoids as an in vitro model of human development and disease. Nat Cell Biol 2016;18:246-54.
crossref pmid pdf
5. Hofer M, Lutolf MP. Engineering organoids. Nat Rev Mater 2021;6:402-20.
crossref pmid pmc pdf
6. Lancaster MA, Renner M, Martin CA, Wenzel D, Bicknell LS, Hurles ME, et al. Cerebral organoids model human brain development and microcephaly. Nature 2013;501:373-9.
crossref pmid pdf
7. Sato T, Vries RG, Snippert HJ, van de Wetering M, Barker N, Stange DE, et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 2009;459:262-5.
pmid
8. Guan Y, Enejder A, Wang M, Fang Z, Cui L, Chen SY, et al. A human multi-lineage hepatic organoid model for liver fibrosis. Nat Commun 2021;12:6138.
crossref pmid pmc pdf
9. Takasato M, Er PX, Chiu HS, Maier B, Baillie GJ, Ferguson C, et al. Kidney organoids from human iPS cells contain multiple lineages and model human nephrogenesis. Nature 2015;526:564-8.
crossref pmid pdf
10. Miller AJ, Dye BR, Ferrer-Torres D, Hill DR, Overeem AW, Shea LD, et al. Generation of lung organoids from human pluripotent stem cells in vitro. Nat Protoc 2019;14:518-40.
crossref pmid pmc pdf
11. Drost J, Clevers H. Organoids in cancer research. Nat Rev Cancer 2018;18:407-18.
crossref pmid pdf
12. Murphy SV, Atala A. 3D bioprinting of tissues and organs. Nat Biotechnol 2014;32:773-85.
crossref pmid pdf
13. Yoon J, Singh NK, Jang J, Cho DW. 3D bioprinted in vitro secondary hyperoxaluria model by mimicking intestinal-oxalate-malabsorption-related kidney stone disease. Appl Phys Rev 2022;9:041408.
crossref pdf
14. Zhu Y, Sun L, Wu X, Gu Z, Zhao Y. Engineered human organoids for biomedical applications. Adv Funct Mater 2024;34:2310961.
crossref
15. Yaneva A, Shopova D, Bakova D, Mihaylova A, Kasnakova P, Hristozova M, et al. The progress in bioprinting and its potential impact on health-related quality of life. Bioengineering (Basel) 2023;10:910.
crossref pmid pmc
16. Giobbe GG, Crowley C, Luni C, Campinoti S, Khedr M, Kretzschmar K, et al. Extracellular matrix hydrogel derived from decellularized tissues enables endodermal organoid culture. Nat Commun 2019;10:5658.
crossref pmid pmc pdf
17. Haleem A, Javaid M, Singh RP, Suman R. Medical 4.0 technologies for healthcare: features, capabilities, and applications. Internet Things Cyber Phys Syst 2022;2:12-30.
crossref
18. Zhao Z, Chen X, Dowbaj AM, Sljukic A, Bratlie K, Lin L, et al. Organoids. Nat Rev Methods Primers 2022;2:94.
pmid pmc
19. Kratochvil MJ, Seymour AJ, Li TL, Paşca SP, Kuo CJ, Heilshorn SC. Engineered materials for organoid systems. Nat Rev Mater 2019;4:606-22.
crossref pmid pmc pdf
20. Levental KR, Yu H, Kass L, Lakins JN, Egeblad M, Erler JT, et al. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell 2009;139:891-906.
crossref pmid pmc
21. Tortorella I, Argentati C, Emiliani C, Martino S, Morena F. The role of physical cues in the development of stem cell-derived organoids. Eur Biophys J 2022;51:105-17.
crossref pmid pdf
22. Gattazzo F, Urciuolo A, Bonaldo P. Extracellular matrix: a dynamic microenvironment for stem cell niche. Biochim Biophys Acta 2014;1840:2506-19.
crossref pmid pmc
23. Fang Y, Eglen RM. Three-dimensional cell cultures in drug discovery and development. SLAS Discov 2017;22:456-72.
crossref pmid pmc pdf
24. Kleinman HK, Martin GR. Matrigel: basement membrane matrix with biological activity. Semin Cancer Biol 2005;15:378-86.
crossref pmid
25. Liu H, Bockhorn J, Dalton R, Chang YF, Qian D, Zitzow LA, et al. Removal of lactate dehydrogenase-elevating virus from human-in-mouse breast tumor xenografts by cell-sorting. J Virol Methods 2011;173:266-70.
crossref pmid pmc
26. Hughes CS, Postovit LM, Lajoie GA. Matrigel: a complex protein mixture required for optimal growth of cell culture. Proteomics 2010;10:1886-90.
crossref pmid pdf
27. Kim S, Min S, Choi YS, Jo SH, Jung JH, Han K, et al. Tissue extracellular matrix hydrogels as alternatives to Matrigel for culturing gastrointestinal organoids. Nat Commun 2022;13:1692.
crossref pmid pmc pdf
28. Aisenbrey EA, Murphy WL. Synthetic alternatives to Matrigel. Nat Rev Mater 2020;5:539-51.
crossref pmid pmc pdf
29. Heo JH, Kang D, Seo SJ, Jin Y. Engineering the extracellular matrix for organoid culture. Int J Stem Cells 2022;15:60-9.
crossref pmid pmc
30. Kaur S, Kaur I, Rawal P, Tripathi DM, Vasudevan A. Non-matrigel scaffolds for organoid cultures. Cancer Lett 2021;504:58-66.
crossref pmid
31. Gan Z, Qin X, Liu H, Liu J, Qin J. Recent advances in defined hydrogels in organoid research. Bioact Mater 2023;28:386-401.
crossref pmid pmc
32. Jin Y, Kim J, Lee JS, Min S, Kim S, Ahn DH, et al. Vascularized liver organoids generated using induced hepatic tissue and dynamic liver‐specific microenvironment as a drug testing platform. Adv Funct Mater 2018;28:1801954.
crossref pdf
33. Saldin LT, Cramer MC, Velankar SS, White LJ, Badylak SF. Extracellular matrix hydrogels from decellularized tissues: structure and function. Acta Biomater 2017;49:1-15.
crossref pmid
34. Saheli M, Sepantafar M, Pournasr B, Farzaneh Z, Vosough M, Piryaei A, et al. Three-dimensional liver-derived extracellular matrix hydrogel promotes liver organoids function. J Cell Biochem 2018;119:4320-33.
crossref pmid pdf
35. Simsa R, Rothenbücher T, Gürbüz H, Ghosheh N, Emneus J, Jenndahl L, et al. Brain organoid formation on decellularized porcine brain ECM hydrogels. PLoS One 2021;16:e0245685.
crossref pmid pmc
36. Bi H, Karanth SS, Ye K, Stein R, Jin S. Decellularized tissue matrix enhances self-assembly of islet organoids from pluripotent stem cell differentiation. ACS Biomater Sci Eng 2020;6:4155-65.
crossref pmid
37. Ren Y, Yang X, Ma Z, Sun X, Zhang Y, Li W, et al. Developments and opportunities for 3D bioprinted organoids. Int J Bioprint 2021;7:364.
crossref pmid pmc pdf
38. Kang HW, Lee SJ, Ko IK, Kengla C, Yoo JJ, Atala A. A 3D bioprinting system to produce human-scale tissue constructs with structural integrity. Nat Biotechnol 2016;34:312-9.
crossref pmid pdf
39. Zhang YS, Duchamp M, Oklu R, Ellisen LW, Langer R, Khademhosseini A. Bioprinting the cancer microenvironment. ACS Biomater Sci Eng 2016;2:1710-21.
crossref pmid pmc
40. Brassard JA, Nikolaev M, Hübscher T, Hofer M, Lutolf MP. Recapitulating macro-scale tissue self-organization through organoid bioprinting. Nat Mater 2021;20:22-9.
crossref pmid pdf
41. Urciuolo A, Giobbe GG, Dong Y, Michielin F, Brandolino L, Magnussen M, et al. Hydrogel-in-hydrogel live bioprinting for guidance and control of organoids and organotypic cultures. Nat Commun 2023;14:3128.
crossref pmid pmc pdf
42. Brooks A, Liang X, Zhang Y, Zhao CX, Roberts MS, Wang H, et al. Liver organoid as a 3D in vitro model for drug validation and toxicity assessment. Pharmacol Res 2021;169:105608.
crossref pmid
43. Lee J, Koehler KR. Skin organoids: a new human model for developmental and translational research. Exp Dermatol 2021;30:613-20.
crossref pmid pmc pdf
44. Wang Y, Shi W, Kuss M, Mirza S, Qi D, Krasnoslobodtsev A, et al. 3D bioprinting of breast cancer models for drug resistance study. ACS Biomater Sci Eng 2018;4:4401-11.
crossref pmid
45. Blanco-Fernandez B, Rey-Vinolas S, Bağcı G, Rubi-Sans G, Otero J, Navajas D, et al. Bioprinting decellularized breast tissue for the development of three-dimensional breast cancer models. ACS Appl Mater Interfaces 2022;14:29467-82.
crossref pmid pmc pdf
46. Ricci G, Gibelli F, Sirignano A. Three-dimensional bioprinting of human organs and tissues: bioethical and medico-legal implications examined through a scoping review. Bioengineering (Basel) 2023;10:1052.
crossref pmid pmc
47. Choi YM, Lee H, Ann M, Song M, Rheey J, Jang J. 3D bioprinted vascularized lung cancer organoid models with underlying disease capable of more precise drug evaluation. Biofabrication 2023;15:034104.
crossref pdf
48. Kim J, Kim J, Gao G, Choi YM, Kim J, Cho DW, et al. Bioprinted organoids platform with tumor vasculature for implementing precision personalized medicine targeted towards gastric cancer. Adv Funct Mater 2024;34:2306676.
crossref
49. Jian H, Li X, Dong Q, Tian S, Bai S. In vitro construction of liver organoids with biomimetic lobule structure by a multicellular 3D bioprinting strategy. Cell Prolif 2023;56:e13465.
pmid pmc
50. Shi W, Mirza S, Kuss M, Liu B, Hartin A, Wan S, et al. Embedded bioprinting of breast tumor cells and organoids using low-concentration collagen-based bioinks. Adv Healthc Mater 2023;12:e2300905.
crossref pmid
51. Wang X, Luo Y, Ma Y, Wang P, Yao R. Converging bioprinting and organoids to better recapitulate the tumor microenvironment. Trends Biotechnol 2024;42:648-63.
crossref pmid
52. Parihar A, Pandita V, Kumar A, Parihar DS, Puranik N, Bajpai T, et al. 3D printing: advancement in biogenerative engineering to combat shortage of organs and bioapplicable materials. Regen Eng Transl Med 2022;8:173-99.
crossref pmid pdf
53. Homan KA, Gupta N, Kroll KT, Kolesky DB, Skylar-Scott M, Miyoshi T, et al. Flow-enhanced vascularization and maturation of kidney organoids in vitro. Nat Methods 2019;16:255-62.
crossref pmid pmc pdf
54. Zhang YS, Khademhosseini A. Advances in engineering hydrogels. Science 2017;356:eaaf3627.
crossref pmid pmc
55. Xu ZY, Huang JJ, Liu Y, Chen CW, Qu GW, Wang GF, et al. Extracellular matrix bioink boosts stemness and facilitates transplantation of intestinal organoids as a biosafe Matrigel alternative. Bioeng Transl Med 2022;8:e10327.
pmid pmc
56. Kort-Mascort J, Bao G, Elkashty O, Flores-Torres S, Munguia-Lopez JG, Jiang T, et al. Decellularized extracellular matrix composite hydrogel bioinks for the development of 3D bioprinted head and neck in vitro tumor models. ACS Biomater Sci Eng 2021;7:5288-300.
crossref pmid
57. Mota C, Camarero-Espinosa S, Baker MB, Wieringa P, Moroni L. Bioprinting: from tissue and organ development to in vitro models. Chem Rev 2020;120:10547-607.
crossref pmid pdf
58. Chen EP, Toksoy Z, Davis BA, Geibel JP. 3D bioprinting of vascularized tissues for in vitro and in vivo applications. Front Bioeng Biotechnol 2021;9:664188.
crossref pmid pmc
59. Juraski AC, Sharma S, Sparanese S, da Silva VA, Wong J, Laksman Z, et al. 3D bioprinting for organ and organoid models and disease modeling. Expert Opin Drug Discov 2023;18:1043-59.
crossref pmid
60. Fang W, Yang M, Wang L, Li W, Liu M, Jin Y, et al. Hydrogels for 3D bioprinting in tissue engineering and regenerative medicine: current progress and challenges. Int J Bioprint 2023;9:759.
crossref pmid pmc pdf
61. Yoon J, Han H, Jang J. Nanomaterials-incorporated hydrogels for 3D bioprinting technology. Nano Converg 2023;10:52.
crossref pmid pmc pdf
62. Mazzocchi A, Soker S, Skardal A. 3D bioprinting for high-throughput screening: drug screening, disease modeling, and precision medicine applications. Appl Phys Rev 2019;6:011302.
crossref pmid pmc pdf
63. Peng W, Datta P, Ayan B, Ozbolat V, Sosnoski D, Ozbolat IT. 3D bioprinting for drug discovery and development in pharmaceutics. Acta Biomater 2017;57:26-46.
crossref pmid
64. Ozbolat IT. Bioprinting scale-up tissue and organ constructs for transplantation. Trends Biotechnol 2015;33:395-400.
crossref pmid
65. Zandrini T, Florczak S, Levato R, Ovsianikov A. Breaking the resolution limits of 3D bioprinting: future opportunities and present challenges. Trends Biotechnol 2023;41:604-14.
crossref pmid
66. Sanicola HW, Stewart CE, Mueller M, Ahmadi F, Wang D, Powell SK, et al. Guidelines for establishing a 3-D printing biofabrication laboratory. Biotechnol Adv 2020;45:107652.
crossref pmid
67. Skardal A, Atala A. Biomaterials for integration with 3-D bioprinting. Ann Biomed Eng 2015;43:730-46.
crossref pmid pdf
68. Blatchley MR, Anseth KS. Middle-out methods for spatiotemporal tissue engineering of organoids. Nat Rev Bioeng 2023;1:329-45.
crossref pmid pmc pdf
TOOLS
METRICS Graph View
  • 0 Crossref
  •  0 Scopus
  • 1,142 View
  • 28 Download
ORCID iDs

Jungbin Yoon
https://orcid.org/0000-0003-2867-2100

Goeun Yoon
https://orcid.org/0009-0005-2780-963X

Jinah Jang
https://orcid.org/0000-0001-9046-3495

Related articles


ABOUT
BROWSE ARTICLES
EDITORIAL POLICY
FOR CONTRIBUTORS
Editorial Office
Room 319, Hall 1 of Chonbuk National University Dental College, 20, Geonji-ro, Deokjin-gu, Jeonju 54907, Korea
Tel: +82-63-270-4024    E-mail: editor@j-organoid.org                

Copyright © 2024 by The Organoid Society.

Developed in M2PI

Close layer
prev next