Abstract
Organoids, as 3D in vitro models derived from stem cells, have unparalleled advantages over traditional cell and animal models for studying organogenesis, disease mechanisms, drug screening, and personalized diagnosis and treatment. Despite the tremendous progress made in organoid technology, the translational application of organoids still presents enormous challenges due to the complex structure and function of human organs. In this review, the limitations of the translational application of traditional organoid technologies are first described. Next, we explore ways to address many of the limitations of traditional organoid cultures by engineering various dimensions of organoid systems. Finally, we discuss future directions in the field, including potential roles in drug screening, simulated microphysiology system and personalized diagnosis and treatment. We hope that this review inspires future research into organoids and microphysiology system.
Keywords: Organoid, organoids-on-chips, bioengineering, microenvironment, Microphysiological system
Introduction
For a long time, human beings have been plagued with questions about the development of the human body from the time of the embryo, how diseases affect human organs, the impact of external environmental factors on the human body, and the development of new therapies and new drugs. Most research on these issues is currently limited to traditional two-dimensional cellular and animal models, but due to species differences and research costs,1,2 there is an urgent need for a model that can more closely mimic the real environment of the human body and be cost-effective. In response to this need, organoid and organoids-on-chips technologies have emerged.
Organoids are self-organizing 3D tissues, usually derived from stem cells (pluripotent, embryonic, or adult stem cells), that mimic the key functions, structure, and biological complexity of organs.3,4 Organoid research was fast-tracked in 2009 when Sato’ team 5 cultured adult stem cells derived from the mouse intestine to produce the first miniature intestinal organoid with small intestinal crypt and villus structures. Although organoids show great potential to a certain extent, there is still a large gap between organoids and real human organs, which is due to the complex composition and many functions of real human organs. To date, a number of engineering strategies to support organoid culture and growth, proliferation, differentiation and maturation have been reported. For example, by applying engineering principles and design concepts to traditional organoid technologies, such as organoids-on-chips, 6 and 3D printing, 7 these engineering tools can be used to optimize organoid culture methods to ensure reproducibility as well as reliability, and these technologies enable precise control of the culture microenvironment so that organoids can more closely resemble real physiological environments. In addition to basic culture, precise analysis of organoids plays a crucial role, and engineering enables visualization and dynamic analysis of organoid microenvironments as well as biochemical metrics. On the basis of these insights, developing engineering strategies to precisely control organoid growth and development is important. In this review, we aim to highlight engineering strategies to alleviate the current organoid dilemma. First, some of the limitations of organoid systems in general are outlined. Next, strategies to overcome these limitations are detailed. Finally, we present the challenges and perspectives of using organoids in biomedical applications.
Limitations of current organoid systems
Limited survival time
Inadequate vascularization within organoids results in a limited nutrient and oxygen supply, which affects their long-term growth and long-term maintenance of their functional activities, and the shorter survival time of organoids is often a direct cause of their limited use for biomedical research. For example, blood vessels facilitate both nutrient supply and neuronal cell differentiation in brain tissues. 8 As the size of brain organoids increases, the lack of internal nutrients and the difficulty in removing metabolic waste lead to necrosis of the center of the organoid. This necrosis further interferes with its normal development and neuron migration routes, a problem that can be addressed, at least in part, through oscillating cultures.9,10
Although the above problems can also be solved by mechanically fragmenting the organoid, the phenotype of the organoid is disrupted by the constant fragmentation of the already-formed cellular tissue structure, all of which can affect the results of the study. Moreover, pluripotent stem cell-derived organoids are not fragmented or passaged.
New strategies to address nutritional accessibility are currently being developed, such as through brain slice culture. 11 The supply of nutrients is not the only problem; similarly, experiments often require the delivery of certain substances to specific areas of organs, but owing to the lack of a functioning vasculature system, these substances do not accurately reach the desired location.
Limited maturity
Many organoids have been successfully constructed, such as mammary organoids that secrete milk, 12 gastric organoids that secrete gastric acid, 13 and liver-islet axis organoids that respond to blood glucose stimulation. 14 However, no organoid has yet been successfully built that can completely replicate the full function of the corresponding organ. The vast majority of organoid models lack key specific cell types, and fail to fully recapitulate the complexity of human organs due to the lack of mesenchymal regions, immune cells, innervation, vascularization, corresponding flora microorganisms, lymphatic vessels, etc.
In addition to the influence of biological factors, the physical microenvironment also appears to be crucial for revealing the process of organogenesis and development in the human body, and the maturation of organs in vitro can be improved through the modulation of external environmental factors such as mechanical force stimulation, light stimulation, or electrical stimulation.15–17 However, integrating these features remains a major challenge for current technology. 6 The long-term culture of organoids in vitro is also a major concern; the limitation of organoid lifespan does not mimic the development of natural organs very well; for example, epithelial organoids have a lifespan of approximately 1 week, which is far from the time it takes for stem cells to fully differentiate into the full range of cell types in vivo.18,19 Brain organoids simulate only the phenotype of the fetal brain, and their mature development needs to be further facilitated if a model of the mature adult brain is to be obtained.20,21
Heterogeneity
Organoid cultures exhibit striking heterogeneity and variable complexity in terms of their cellular composition and are prone to poorly controlled morphogenesis during self-assembly, for example, morphological and structural differences between batches, functional differences, 22 however, these differences are usually due to the stochastic nature of in vitro self-assembly and organ developmental selection. Therefore, there is a great need for engineering tools, such as precise regulation of the medium composition and innovation of the extracellular matrix, to reduce the variability of organoids during development and increase the homogeneity of organoids, which in turn can lead to consistent conclusions in the study of disease mechanisms.
In addition, organoid construction methods are mostly manual, and some key factors, such as the number of cells, the proportion of types, and the proportion of extracellular matrix, can lead to differences in organoids when they are at the initial stage of organoid development. By increasing the means of automation, the use of robotic liquid handling systems during organoid culture can independently perform a series of precisely controlled tasks, including complex operations such as initial stem cell allocation, media addition and replacement, drug testing, and real-time analysis,23,24 which are expected to increase the homogeneity and reduce the heterogeneity of organoids.
Insufficient function monitoring
Organoid reconstructions of various systems have replicated the development of different human systems and a portion of their functional and structural phenotypes, providing important insights into organogenesis and development as well as diseases of various systems. Therefore, it is particularly important for the monitoring of the structural-functional activity of organoids. Most of the common monitoring devices are traditional optical monitoring devices, but traditional optical microscopy monitoring provides relatively limited information. The use of conventional optical microscopes provides limited data due to situations such as individual three-dimensionally growing organoids being in different focal planes. 22 Similarly, traditional electrophysiological methods make accurate monitoring of the physiological properties of nerve organoids difficult because of their highly complex three-dimensional structure.
Recently, several studies have subdivided the physiological and pathological states of neural organoid by using techniques such as high-content imaging, 25 multi-electrode arrays, 26 optogenetics, 27 and biosensors. 28 Other organoids, such as liver organoids, can be used to assess the physiological or pathological function of the liver in vitro by analyzing the metabolites of exogenous compounds as well as endogenous substrates or the metabolic synthesis of bile and proteins. 29 However, the extremely low levels of some metabolites or compounds make it extremely difficult to monitor them. By incorporating miniature biochemical sensor technology, it is not only possible to achieve a high level of sensitivity, enabling the monitoring of concentrations up to the micromolar or nanomolar level, but also to minimize the impact on cellular activity. 30 Nevertheless, typical organoids are mostly cultured in Matrigel, which makes it difficulty in monitoring biochemical sensors. Therefore, to monitor the target with homogenized information, relatively precise control of the position and the shape of the organoids is necessary. High-throughput testing can be performed with increased automation, both when basic organoid cultures are performed and when high-throughput screening of organoids is performed. 31 These issues limit the further application of organoids in biomedical research. Hence, the modulation of some of the controllable factors of organoids (e.g. batch differences in Matrigel and growth factor content) may be beneficial in mitigating these limitations. 22 In response to some of these shortcomings, the use of engineering tools can help address some of the limitations of organoid platforms.
Engineered organoid formation
Organoid
Organoids are three-dimensional multicellular in vitro cultures formed by the self-organization of stem cells,32,33 which have significant potential in research areas such as growth and development, disease modeling, and drug discovery and have become an important development direction in tissue engineering and regenerative medicine34,35 (Figure 1).
Figure 1.
Schematic of the different organoids that can be derived from PSCs or tissue.
Compared with traditional animal models, organoid cultures are more amenable to intervention and in-depth biomedical research. Organoids can be generated from pluripotent stem cells (PSCs), including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), as well as from tissue samples. Currently, organoid technology is used in areas such as drug discovery, 36 personalized diagnosis, 37 and cell therapy. 19 In recent decades, remarkable progress has been made in the development of various organoids (e.g. brain, 9 heart, 38 kidney, 39 lung, 40 stomach, 41 prostate gland, 42 pancreas, 43 mammary gland, 12 liver, 44 and others). Notably, organoids derived from tissues of tumor patients have also been successfully established. 45 These organoids offer a broad spectrum of applications, spanning from organogenesis and disease modeling to personalized medical interventions and drug screening strategies. Hereditary disorders are diseases caused by alterations in genetic material; these disorders are often accompanied by a wide range of birth defects, and owing to the complexity of their disorders, many genetic disorders lack effective treatment. Organoids have a wide range of applications for modeling such diseases. To date, numerous genetic diseases have been reproduced by organoids, such as human microcephaly, 9 polycystic kidney disease, 46 autism spectrum disorders 47 and other disorders, and these models provide new tools for further exploration of disease pathogenesis as well as therapeutic strategies. Additionally, various types of tumor-patient tissue-derived organoids have been used for research, such as gastroesophageal cancer organoids, 48 lung cancer organoids, 36 and pancreatic cancer organoids, 49 and these models provide abundant data for the treatment of the corresponding diseases. Similarly, substantial progress has been made in the study of microbial-host interactions, for example, gut flora-on-oncogene interactions, 50 gut microbial-host interactions in preterm infants. 51 Despite great progress in organoid technology, several limitations remain, including low maturity and high heterogeneity.
To minimize the impact of these problems as much as possible, more reliable organoids are being cultivated through the use of organoids-on-chips technology, biomaterials, microfluidics, biochemical sensors, and other technologies. As an alternative to the extracellular matrix (ECM), Matrigel was prepared from soluble basement membranes extracted from the tumors of Engelbreth-Holm-Swarm (EHS) mice enriched with extracellular matrix proteins. Nonetheless, the uncertainty of its composition and batch variability limit its application in biomedical research. Consequently, alternatives to Matrigel have been used, for example, for the spinal cord, 52 brain, 53 and alveoli. 54 These biomaterials can reduce the batch variability of organoid generation and increase homogeneity.
Similarly, the maturation of organoids is very limited because of the lack of a complex physiological microenvironment in vitro. Therefore, engineering the culture environment to mimic fluid shear forces in vivo, for example, is more conducive to organoid development and maturation and improves the reliability of organoids.
Organoids-on-chips
Organoids-on-chips refers to the organoid system based on microfluidic technology, which replaces of two-dimensional cells in organoids-on-chips with three-dimensional cells or organoids, and is an extension of traditional organs-on-chips methods in biotechnology, 6 which focus on the microfluidic chips technology by combining it with a variety of methods, such as cell biology, biomaterials, and engineering. We summarized the key components of the organoids-on-chips model, including the organoid type, cell source, extracellular matrix, chip design, and application (Table 1).
Table 1.
Summary of existing human organoids-on-chips models used for biomedicine.
| Organoid | Cell sources | Extracellular matrix | Chip design | Application | Ref. |
|---|---|---|---|---|---|
| Brain | hiPSCs | Matrigel | A chip Consists of bottom layer micropillars and top layer ring structure | Simplifying brain organoid formation protocols and overcoming the potential limitations | Zhu et al. 55 |
| A chip consists of patterned micropillars and ring structure | Modeling prenatal brain exposure to valproic acid | Cui et al. 56 | |||
| Fibrinogen, collagen type Ⅰ | A chip consists of five parallel channels separated by micropillars, a removable polyester membrane | Modeling brain organoids vascularization and deciphering potential vascularization factors | Shaji et al. 57 | ||
| Matrigel | A chip that integrates perfusable vascular chambers and organoid chambers | Modeling the interaction between brain organoid and vascular system | Salmon et al. 58 | ||
| hESCs | A tubular organoid perfusion device (upper) and an organoid holder device (bottom) | Revealing a new method for inducing aging phenotype within brain organoids | Ao et al. 59 | ||
| Retinal | mESC | PEG hydrogels | A chip consists of U-shaped hydrogel microporous array and PDMS ring | Standardizing retinal organoids and reducing heterogeneity | Decembrini et al. 60 |
| Heart | hiPSCs | decellularized ECM hydrogels | A chip consists of three culture chambers and two medium chambers | Modeling Long QT syndrome and cardiac fibrosis | Min et al. 61 |
| Colon | Patient | Matrigel | A chip capable of perfusing vascularized colonic organoids | Modeling vascularization and inflammation within colon organoids | Rajasekar et al. 62 |
| Kidney | hPSCs | Matrigel | A chip capable of perfusing vascular networks | Modeling kidney organoids vascularization | Homan et al. 63 |
| pancreatic tumor | Patient | Matrigel | A chip that can support various cells to self assemble into 3D tissues in substantive space | Reproducing the microenvironment of pancreatic tumor | Lai Benjamin et al. 64 |
| Liver | hESCs | 4-arm-PEG-maleimide | A perfusable chip composed of channels of different shapes and depths | Established human vascularized hepatobiliary organoids | Abbasalizadeh et al. 65 |
hiPSCs: human induced pluripotent stem cells; PSCs: human pluripotent stem cells; hESCs: human embryonic stem cells; PEG: polyethylene glycol; PDMS: polydimethylsiloxane; mESC: mouse embryonic stem cells.
Furthermore, we also present a conceptual diagram of some organoids-on-chips (Figure 2). In microfluidic devices, cells and tissues can be cultured in miniature volumes and controlled microenvironments designed to mimic specific in vivo niche.66–70 Owing to the characteristics of certain shear forces in three-dimensional culture environments, these cells mimic the physiological state of humans more closely and can be simulated in vitro to reconstruct a tissue and organ microenvironment that encompasses a variety of living cells, functional tissue interfaces, biofluids, mechanical force stimulation, and other complex factors, all of which respond to the primary structural and functional characteristics of the human body’s tissues and organs.
Figure 2.

Conceptual schematic of a human organoids-on-a-chips.
To address the high heterogeneity in conventional organoid cultures, which is predominantly caused by complex manual manipulation, microfluidics can offer a solution by utilizing microstructures. Numerous microfluidic devices capable of controlling morphology have been employed to generate organoids. Additionally, microcolumns of a certain size can control the size of the organoid and improve the homogeneity of the organoid. 55 Similarly, retinal organoids can be standardized in a controlled manner in microporous arrays. 60 Inter tissue fluid in brain tissue plays an important role in nutrient transfer, metabolic waste removal, and intercellular communication; however, most current in vitro studies of neurological disorders suffer from the drawbacks of two-dimensional cell cultures and the neglect of inter tissue fluid flow. Park et al. 71 mimicked Alzheimer’s disease (AD) through the use of a three-dimensional sphere-based microfluidic chip. In this study, the toxic effects of β-amyloid were tested via neurospheres cultured under dynamic and static conditions, and surprisingly, dynamic cultures appeared to have greater neurodestructiveness and deeper synaptic dysfunction than static cultures, characteristics that are highly consistent with the pathology of AD. Thus, brain organs-on-chips provide structural and functional support for reproducing Alzheimer’s disease. Cui’s research team 56 cultured cortical organoids exposed to valproic acid (VPA) on perfusable organoids-on-chips. An in vitro model outlined the injury features of prenatal exposure to valproic acid, including increased neuronal progenitor cells, suppressed neurons, and altered forebrain regionalization, and analysis of transcriptomic data revealed a high degree of similarity between VPA-exposed organoids and postmortem autistic brains and organoids originating from autistic patients. Thus, this brain organoids-on-chips microarray model enables researchers to explore neurodevelopmental disorders under exposure to a variety of environmental factors and can be extended to applications in disease modeling and drug screening.
Apart from the nervous system, structurally and functionally mature cardiac tissue offers new tools for use in drug development and cardiotoxicity testing. By combining microfluidic microarray technology with decellularized heart extracellular matrix (HEM), Min’s team 61 constructed a perfusable culture system based on pluripotent stem cells (hiPSCs)-derived cardiac tissues, HEM enhances the upregulation of genes involved in cardiac collagen deposition, fibrosis, and cardiomyocyte maturation in vitro. By stacking ductless chips on an axial shaker, large-scale culture of mature cardiac tissues can be achieved. Compared with conventional dynamic culture systems, more complex media flow was provided to cardiac tissue and facilitated electrical synchronization, and an improved ability to synchronize contractions of cardiac tissue was found. By transplanting cardiac tissues into rats, the generated cardiac tissues promoted functional and structural regeneration of the heart and achieved functional integration of the transplanted myocardium with the host myocardium. These cardiac tissues can also be used to assess contraction patterns and electrophysiological changes in drugs, as well as to model diseases such as long QT syndrome and cardiac fibrosis. In brief, these dynamic organoids-on-a-chips systems72,73 can provide more reliable tools for basic medical research.
Furthermore, current organoid systems typically lack blood circulation, make it difficult for nutrients and oxygen to reach the core, have a continuously increase metabolic waste, cause extensive cell death and premature cell differentiation in the later stages of culture, resemble embryonic or fetal organs rather than adult organs in their developmental stages and functions, and lack relevant developmental signals,9,74–76 which greatly limits the technological applications of organoids. Therefore, Shaji et al. 57 developed a three-layer, five-channel microfluidic device based on polydimethylsiloxane (PDMS) and directly cocultured brain organoids derived from human-induced pluripotent stem cells (hiPSCs) in a perfusable vascular bed generated by the self-assembly of human umbilical vein endothelial cells (HUVECs) to confirm the differentiation of the hiPSCs into brain organoids and to evaluate the stability and perfusability of the vascular bed during coculture. This study used brain organoids cultured on microarrays to identify genes involved in angiogenesis, and key members of the vascular endothelial growth factor (VEGF) signaling pathway were consistent with the angiogenic process observed in brain organoids cultured on microarrays.
Recently, in a study of the interaction of colonic organoids with HUVECs and fibroblasts, Rajasekar et al. 62 independently cultured up to perfusable vascularized colonic organoids in vitro and modeled colonic inflammation with innate immune function. This technique is useful for high-throughput drug screening, but the use of this customized well plate makes the user’s use in terms of spatial control inflexible. Salmon’s team 58 overcomes this limitation by printing microfluidic chips via 3D printing technology to realize interactions between organoids and the vascular system on a chip, and this 3D printing-based organoid chips were able to more accurately recreate the true physiological roles of vascular endothelial cells and brain organoids. Similarly, vascularized heart, pancreatic and liver cancer tissues can be fabricated, providing a new platform for organogenesis development, disease modeling and drug development.
Most organs, with the exception of the vascular system, possess tissue-resident immune cells. However, human organoids lack these immune cells, which restricts their applicability in modeling numerous normal physiological and disease mechanisms. Ranga, by developing a microphysiological analysis platform (MAP) for human brain organoids, reproduced the physiological interactions between monocytes and brain organoids derived from hiPSCs. By co-culturing young (20–30 years old) and old (>60 years old) primary monocytes with cultured human cortical organoids, it was found that primary monocytes from older (>60 years old) donors have a greater capacity to infiltrate cortical organoids and promote the expression of proteins associated with senescence (e.g. p16). This platform provides a new potential method for inducing aging phenotypes within brain organoids without external genetic manipulation and offers new research avenues for studying immune-driven brain aging as well as restoring homeostasis in the aging brain. In addition to immune co-culture of normal organoids, immune co-culture of tumor organoids for immunotherapy of tumors is also a bottleneck that urgently needs to be overcome. Xu et al. 77 developed a novel culture of breast cancer organoids with a biomimetic matrix and immune environment. Initially, an alginate cryogel was prepared to serve as a biomimetic tumor matrix. Subsequently, tumor-associated macrophages (TAMs) were cultured within this matrix and induced to polarize in the M2 direction. Finally breast cancer cells were added to form an organoid model that was cocultured with immune cells. Direct co-culture significantly enhanced breast cancer organoid growth and cancer invasiveness phenotypes, a model that helps researchers understand gene expression, protein interactions, and other information pertinent to breast cancer progression. The same coculture idea can be applied to other tumor organoids (e.g. colorectal cancer, non-small cell lung cancer, and gastric cancer).
Currently, it is very difficult to coculture human organoids and immune cells in the long term. The coculture conditions of organoids are usually the best combination of the ratio of each cell type; however, each cell has its own specific culture conditions, and the long-term supply of nutrients to the organoids is a major problem, especially from the tumor tissues, to generate organoids in a relatively inefficient manner. It is hoped that future cross fertilization of technologies from various disciplines will help to bring organoid immune cell coculture technology closer to the real environment in the body as well as to realize long-term culture, thus broadening our understanding of the complex interactions between various organs in the human body.
3D printing
3D printing has shown very significant advantages in in vitro organoid modeling, including being able to explore the effects of a large number of variables on a system using a small number of samples in a highly efficient way and enabling organoids to maintain their original phenotype. It is faster, less costly, and easier to perform than traditional methods. For example, 3D-printed small rotating bioreactors reduce the media volume and incubator space compared with rotating bioreactors used for long-term maintenance of organoids.10,78 In recent years, suboptimal culture conditions for most organoids have led to the overuse of plastic.
To address the above problems, Rezaei’s team 79 developed a 3D-printed pipeline for the preparation of a personalized culture platform that spatially cultures individual brain organoids independently while ensuring fluid flow, an integrated platform that enables all culture steps such as cell aggregation, balloon growth, hydrogel embedding, and maturation of the organoids to take place in individual cell culture wells without the need for the organoids to be transferred. In the long run, this microprinted device could efficiently and dynamically culture brain organoids, provide sufficient nutrients and oxygen, reduce the risk of contamination, and be suitable for large-scale culture.
Similarly, to reduce the heterogeneity of retinal organoids (ROs) and efficiently generate homogeneous ROs, Sun et al. 80 developed a novel polydimethylsiloxane (PDMS) microwell platform in which ROs can be assembled in one step and remain homogeneous and maturationally differentiated for a long period of time (more than 25 weeks) without the use of BMP4 and Matrigel. The platform can be used for subsequent retinal disease modeling, drug screening, and the production of clinically translatable ROs. To construct a vascularized bionic chip, the central culture chamber for brain organoids is designed with “open holes” to support vascular network formation and vascular invasion. 58
In addition to common hydrogels, decellularized extracellular matrix (dECM) is important because of its ability to have tissue-specific or organ-specific extracellular matrix properties. The dECM is able to support cell adhesion, migration, and proliferation due to its unique structural proteins, cytokines, signaling molecules, and other physical properties, such as mechanical properties similar to those of natural tissue.81–83 In one study, human colonic organoids were derived by modifying colonic dECM for photopolymerization, which served as a bioink. These organoids were then encapsulated into hollow tubular structures also derived from colonic dECM. When compared to those cultured in traditional Matrigel, these organoids exhibited similar high cell viability and colon-specific morphology. Over time, they were found to possess a more complex cellular composition and the ability to promote self-renewal and differentiation, features that are not comparable to traditional Matrigel cultures. 84
In addition, 3D printing is uniquely advantageous for reproducing the tumor microenvironment (TME), enabling precise control of biological, physical, and chemical cues in the TME and providing new possibilities for in vitro modeling of tumors. Using dual nozzles for 3D printing, glioblastoma cells and macrophages were integrated onto the gel after the hydrogel was printed, and the model reconstructed the role of tumor immunity, including the polarization of macrophages to glioblastoma cell-associated phenotypes and the invasiveness of glioblastoma cells to normal brain tissue. This integrated 3D printing system provides an advantageous tool for constructing other organs and tissues for vascularization or immune co-culture. 85 However, the reality is that monodisperse tumor cells rarely exist, and most current uses of monodisperse cells do not rebuild the tumor process very well. 86
By integrating bioprinted nozzles to allow individual tumor spheres to be placed in bioprinted scaffolds, this technique allows for the integration of other elements of the microenvironment with the tumor organoids and ensures high fidelity of printing. 87 The synergistic use of 3D printing, organoid, and organoid microarray technologies has an incalculable impact on tissue engineering and holds promise for solving future medical problems.
ECM for organoids
Typically, an organoid culture system is established by adding a single cell suspension of prepared stem cells to Matrigel. However, owing to the complex and ill-defined composition of Matrigel,88,89 batch stabilization is difficult to achieve and may lead to organoid heterogeneity as well as unreliable reproducibility.
Furthermore, because Matrigel is derived from animal sources, it greatly limits its clinical use and human organoid transplantation. Therefore, the search for specific hydrogels that can overcome these problems has become particularly necessary. Currently, attention is being given to hydrogels composed of natural materials as well as synthetic ones, with fibrin, 90 hyaluronic acid, 91 or polyethylene glycol hydrogels88,92 being able to reduce batch-to-batch variation as well as ethical issues compared with Matrigel of animal origin. The characteristics and types of different extracellular matrices and their application in organoids culture are summarized in Table 2. Giobbe et al. 93 reported a greater percentage of stem cells than crypts in small intestinal organoids by using an intestinal decellularized extracellular matrix for intestinal organoid cultures due to its favorable physiological and mechanical properties, demonstrating that organoid ontogeny and development are closely related to the surrounding environment.
Table 2.
Materials systems for organoids.
| Type of ECM | Characteristic | Application | Ref. |
|---|---|---|---|
| Matrigel | High biocompatibility and stability, batch variability | Brain, kidney, etc. | Lancaster et al. 9 , Huang et al. 94 |
| Decellularized ECM hydrogels | Good stability, high biocompatibility and high cost, widely used | Intestine | Giobbe et al. 93 |
| Hyaluronic acid | High biocompatibility and good stability | Kidney | Astashkina et al. 95 |
| Alginate | Low cost and high stability and reproducibility, low mechanical stiffness | Brain | Cassel de Camps et al. 96 |
| Collagen | Widely used and good biocompatibility, easy to degradation | Intestine | DiMarco et al. 97 |
| silk fibroin | Low immunogenicity and suitable mechanical stiffness | Breast cancer | Shi et al. 98 |
| Fibrin | High biocompatibility and easy to degrade | Intestine | Broguiere et al. 90 |
| Plasma-derived extracellular matrix | Optimum growth factors, chemicals, necessary physical cues and high repeatability | Hepatocellular carcinoma | El-Derby et al. 99 |
| PEG | Controllable physical environment and low immunogenicity | Intestine | Gjorevski et al. 100 |
| PCL | Good mechanical properties and stability | Brain | Rothenbücher et al. 101 |
| PLGA | High biocompatibility and undesirable degradation products | Intestine | Shaffiey et al. 102 |
| Synthetic peptide hydrogels | High biocompatibility, replenish regularly | Kidney | Treacy et al. 103 |
| PGA | High biocompatibility, suitable mechanical stiffness | cholangiocyte | Tysoe et al. 104 |
PEG: polyethylene glycol; PCL: polycaprolactone; PLGA: polylactic-glycolic acid; PGA: polyglycolic acid.
Like other hydrogels, decellularized extra-tissue matrices alone have drawbacks such as batch-to-batch variability and a lack of bio-hardness, and to address these issues, De Santis et al. 105 introduced a natural extract, sodium alginate, into the dECM, which was shown to support the growth and differentiation of the human trachea, and the stabilization of tracheal structures for up to 28 days.
Conventional engineered hydrogels are difficult to characterize according to the different stages of organoid culture. Surprisingly, Urciuolo’s team 106 developed a hydrogel-based method for the dynamic fabrication of hydrogels in organoids that can be personalized according to specific culture environments and incubation times, matching the conditions required for organoids at different stages. The method was validated for the characterization of multiple organoids and was found to direct the orientation of neural axons in spinal cord organoids, as well as to control differential cell migration in tumor organoids. It was also able to promote the ontogeny and development of liver organoids, small intestinal organoids, and lung organoids. In addition to some natural materials, synthetic biomaterials such as polyethylene glycol (PEG), 107 hyaluronic acid, 91 alginate, 108 can be less susceptible to batch-to-batch variations than Matrigel, and PEG can be customized to mimic the natural ECM on the basis of the respective culture characteristics of different types of organoids. For example, Tomaszewski’s team 109 modified PEG with a number of different kinds of peptides with basement membrane binders as an ECM for follicular organoids, and demonstrated that the modified PEG promoted follicular organoid maturation. Similarly, four-armed maleimide PEG (PEG-4-MAL) was used to design an ECM for organoid culture, and this ECM supports the developmental maturation of human intestinal organoids.110,111
To reduce the heterogeneity of intestinal organoids, Gjorevski’s team 100 embedded intestinal stem cells in a hydrogel containing arginine-glycine-aspartic acid and photosensitive polyethylene glycol, which degraded and softened the matrix under 405 nm ultraviolet light, thereby controlling symmetry disruption and crypt formation in the organoids and achieving homogenization of the intestinal organoids. It is also expected that the heterogeneity of other types of organoids will be reduced through the development of a wider variety of biomaterials and the combination of different materials for different organoids. Other synthetic materials, such as hyaluronic acid and self-assembling peptides (PeptiGel Alpha4/5), have also been used to generate organoids.98,103
Artificial intelligence
To minimize the limitations of organoids in culture and application, many other modern technologies, including automation and artificial intelligence (AI), have been developed that can increase the reproducibility and reliability of organoids in culture as well as application. Traditional organoid culture is usually performed manually; however, frequent manual operations may introduce human error, which may affect the stability of the culture system.5,22,112
AI provides a new framework for organoid construction, multi-scale morphological and functional analysis of organoids, preclinical evaluation, and application, which can be used to explore the potential of AI-generated organoids in detail. In the construction strategy, the matrix gel is designed to more closely mimic the natural structure and functional properties of human organs. Machine learning provides a powerful solution to personalize the design of the chemical, structural, and biomechanical properties of synthetic Matrigel. The culture conditions including temperature, oxygen content, pH, and other factors are precisely controlled. The Parameters of the culture system were programed to improve the accuracy of organoids culture under various conditions; for example, Jiang et al. 31 developed an automated organoid culture analysis system, a platform that homogenizes organoid precursors by spiking the culture wells with a microfluidic droplet printer after mixing the stem cells with Matrigel. Similarly, the types of growth factors, which are crucial induction components, are optimally combined and concentrated in the most efficient and economical manner.
In addition to automation and improved homogeneity at the organoid culture stage, a number of advances have been made in the automation of complex operations in applications such as drug testing and real-time analysis of results.23,24 By combining organoids with artificial intelligence, Renner’s research colleagues 113 have established a process for executing organoid systems that is scalable, reproducible, and capable of high-throughput screening compared with traditional methods. Furthermore, the microfluidic system enables the integration of a nanosensing system to automate the monitoring of important parameters in culture such as oxygen and metabolites. 114 Moreover to improving the efficiency and accuracy of morphological analysis, machine learning can reduce the impact of human intervention.
Although the industry has not yet been able to reach a consistent standard, it is expected that an industry consensus will be reached in the near future.
Construction of physical and chemical microenvironments
Mechanical force
It is well known that physical microenvironmental factors in the body, such as mechanical forces, light stimulation, electrical stimulation, and other complex factors are essential for the development of human organs and tissues.52,115,116 A variety of engineered materials are being explored for modeling the microenvironment of organoid cultures (Table 3). For example, by constructing a methacryloyl gelatin hydrogel system with tunable mechanical properties for use in the culture of cochlear organoids, Xia et al. 115 found that moderately stiff ECM stimulated the proliferation of cochlear progenitor cells (CPCs) through the mechanism of ITGA3/F-actin/YAP signaling, whereas an increase in stiffness inhibited the proliferation of CPCs and stimulated the differentiation toward inner ear hair cells (HCs). These findings suggest that ECM mechanical forces are the driving force that triggers intracellular signaling cascades that direct sensory epithelial generation.
Table 3.
Building physical microenvironment for organoids.
| Stimulus type | Material | Stimulus form | Organoid response | Organoid | Ref. |
|---|---|---|---|---|---|
| Mechanical force | |||||
| gelatin methacryloyl (GelMA)-HA-Arg-Gly-Asp (RGD) hydrogel | Controlling the stiffness of medium (1.5 kPa) | Stimulated CPC proliferation | Cochlear | Xia et al. 115 | |
| Alginate | Manipulating the percentage of the alginate gels | Mirroring the development sequence of the spinal cord in vivo | Spinal cord | Koser et al. 117 | |
| PEG | Transferring strains from silicone membrane to growing organoids | Intermediate matrix stiffness improved the efficiency of neural tube organoids | neural tube organoids | Abdel Fattah et al. 118 | |
| A two-dimensional compartment | Forcing to expand in only x and y dimension | Revealing the biomechanics that cortical folding during development | Brain | Karzbrun et al. 119 | |
| SpinΩ | Continuous low-shear | Enhance cell viability and promote maintenance of the stem cell niche and reduce cost | Brain | Qian et al. 10 | |
| Millifuidic chip | Fluidic shear stress | More mature vascular network | Kidney | Homan et al. 63 | |
| PEG-RGD hydrogel | Matrix stiffness | Modeling fibrotic liver mechanics | Liver | Sorrentino et al. 120 | |
| Light | |||||
| allyl sulfide hydrogels | Varying the duration of irradiation | More crypt formation | Intestine | Hushka et al. 121 | |
| strain-promoted azide-alkyne cycloaddition hydrogel | a photoinduced allyl sulfide exchange reaction | deforming intestinal organoid epithelial shape | Intestine | Yavitt et al. 122 | |
| multivalent click ligand | Mild Light stimulation | Prompting cell proliferation and steering macrophages toward an M2-like phenotype | Oral cancer | Xu et al. 123 | |
| Sulfur- and Nitrogen-Doped Carbon Nanodots | near-infrared light | Affecting organoids viability | Breast cancer | Roscigno et al. 124 | |
| Optogenetic regulation | Blue light-emitting | Inhibiting the release of the parathyroid hormone | Parathyroid gland | Liu et al. 125 | |
| Upconversion nanoparticles | Near-infrared light | Quantifying and imaging proteins | Hepatic ductal | Liu et al. 126 | |
| HD-Br | Near-infrared light | Tracking lysosomal and mitochondrial interactions dynamically | Liver | Fang et al. 127 | |
| CBPH | Near-infrared light | Inhibiting the migration, invasion, and regenerative capacity | Breast cancer | Li et al. 128 | |
| BPQDs@EXO nanospheres | Near-infrared light | Damaging genetic substance and ablating | Bladder cancer | Liu et al. 129 | |
| Electricity | |||||
| PEDOT:PSS, SEBS | Varying currents and pulse train frequency | Evoking calcium signals | Cortex | Li et al. 130 | |
| A modified SCHEEPDOG bioreactor | Varying the electric field strength | Increasing in volume | Intestine | Shim et al. 131 | |
| Pressure-sensitive transistor | Applying an electrical stimulation pulse | Changing in ECG, pressure and calcium flux | Heart | Kim et al. 132 | |
| Chips consisting of a porous inner well and a solid outer well | Applying an chronic electrical stimulation | Increasing in maturity | iMB-VOs | Dailamy et al. 133 | |
| 3D MMF | Varying voltage pulses | Evoking corresponding field potentials | Brain | Park et al. 134 | |
PEDOT:PSS: poly(3,4-ethylenedioxythiophene) polystyrene sulfonate; SEBS: poly(styrene-ethylene-butylene-styrene); C: cisplatin; BP: black phosphorus; HA: hyaluronic acid; BPQDs: black phosphorus quantum dots; EXO: exosome vector; iMB-VOs: myo-vascular organoids.
In addition to cochlear tissue, Koser et al. 117 have studied the mechanical properties of spinal cord tissue. Similarly, not only is the ontogeny of normal brain tissue affected by the external environment during development, but the mechanical traits of brain tissue are altered in a number of common degenerative neurological disorders. For example, Alzheimer’s disease (AD), the most common progressive neurodegenerative brain disease as well as the most common cause of dementia worldwide, 135 and β-amyloid deposition, a typical pathologic marker in AD patients, may affect the stiffness of brain tissue. 136 With age, the composition and physical properties of the extracellular matrix will alter the mechanosensitivity of neurons and glial cells, which in turn accelerates the progression of neurodegenerative diseases. This includes changes in the biomechanics of brain tissue associated with conditions such as Parkinson’s disease (PD), stroke, and AD.137,138 Similarly, the developmental maturation as well as proliferation of cells can be facilitated by utilizing these subtle physical microenvironments. For example, the role of the ECM in neural development was investigated by Ranga et al., 139 who reported that different stiffnesses of the matrix modulate the development of the neural tube.
In another study, Abdel Fattah et al. 118 developed a device that can apply mechanical external forces to promote the growth and development of neural tube organoid. Meanwhile, neural tube organoid was embedded in hydrogels with different hardnesses (soft 0.7 kPa, medium 2 kPa, and hard 8 kPa), respectively, and it was found that the developmental efficiency of neural tube organoid was increased in medium hydrogels compared with soft and hard hydrogels. This phenomenon was not found in hydrogels of other hardnesses. This suggests that the mechanical forces of the extracellular matrix play a regulatory role in the development of organoid organogenesis. In addition to the effects of mechanical forces on neural development, Karzbrun et al. 119 generated brain organoids in a microprocessed culture chamber and simulated the physical process of brain folding by forcing the organoids to expand in the x and y dimensions. Organoid culture combined with microfluidics has also been used to increase the viability of organoids, for example, SpinΩ. 10 In addition to brain organoids, kidney organoids, 63 and liver organoids 120 are also affected by shear stress.
Optical stimulation
In addition to the effects of mechanical stimulation, light factors and electrical stimulation 140 affect on the growth and development of organoids. Gabriel et al. 141 generated brain organoids containing optic vesicles through a staged cell culture protocol, and the structures could sense light while sending signals to other areas of the brain.
Jgamadze’s team 142 transplanted brain organoids into the brains of rats with a damaged visual cortices; the grafts established synaptic connections with their hosts, and the hosts responded to visual stimuli such as flashing lights. Given the important role of light stimulation in the regulation of neurogenesis and development, further exploration of the mechanisms underlying the effects of light stimulation is urgently needed. Although stem cell-derived organoids currently provide a foundation for the study of organs in vivo, existing recording techniques have primarily focused on measuring localized and acute neural activity in a limited number of cells within the organoids. To address this technology gap, Wilson’s team 143 combined electrophysiological recordings with optical imaging of the xenografted cortical organoids and the surrounding host cortex through the use of transparent graphene microelectrode arrays. By applying a visual stimulus and recording the subsequent electrical response, they demonstrated that the cortical organoids, after being implanted in the mouse brain, were able to establish synaptic connections with the surrounding host cortical tissues. These organoids received visual stimulus inputs from the mouse brain and generated corresponding electrophysiological responses.
Similarly, Osaki et al. 27 interconnected two brain organoids cultured in vitro via axon bundles to simulate the interconnections of different regions in the brain and used optogenetics to control specific neuronal populations. In addition to the direct effect of light stimulation on organoids, the fate of stem cells is regulated by manipulating scaffold properties through the incorporation of special biomaterials with photoactivity into biocompatible scaffolds.144–146 For example, Hushka’s team 121 optimized allyl sulfide hydrogels for use in intestinal organoid cultures and modulated the formation and structure of intestinal organoid crypts through optical stimulation. Similarly, Yavitt et al. 122 utilized light-induced hydrogel cross-linking exchange reactions to alter the curvature of epithelial cells for morphogenetic control of intestinal organoids and found that the effect of light-induced viscoelasticity on mechanotransduction pathways is regulated by time, which provides new ideas and directions for in-depth study of the pathogenesis of intestinal diseases as well as for the development of new therapeutics.
In addition to exploring the effects on organoids, Liu et al. 126 developed a photosensitive nanoprobe to detect and quantify protein levels in liver organoids. Fang ’s team 127 also dynamically tracked lysosomes and mitochondria by using light-sensitive probes. Similarly, by incorporating light-sensitive materials into the culture of tumor organoids, researchers have developed many new therapies to treat various tumors, including oral cancer, 123 breast cancer,124,128 and bladder cancer. 129 In addition to culturing organoids by photostimulation, Broguiere et al. 147 also innovated the two-photon pattern technique by using photopatterned nerve growth factor to guide nerve axons. This light-responsive platform could provide a valuable in vitro model for in-depth investigation of the mechanisms of early histomorphogenesis and the development of organoids, as well as for the study of the pathology and therapeutic measures of various systemic types of diseases.
Electrophysiology system
Electrical stimulation of stem cell differentiation and developmental maturation produces considerable effects. 148 In addition to having a similar cellular structure and organ function, the neural organoids were able to mimic the electrophysiology of the early human brain. Thus, electrophysiological recording of organoid activity plays a key role in the development of the organoid field.
Traditional fluorescence imaging methods such as calcium indicators are limited owing to phototoxicity and temporal resolution, and traditional electrophysiological recording studies using membrane clamps or multi-electrode arrays to monitor electrical activity are often short-term or even require slicing of organoids and functional measurements in a planar, 2D format, which greatly limits functional monitoring of 3D cultures and greatly affects electrophysiological recording efficiency and reliability of electrophysiological recordings. 149 Park et al. 134 achieved the matching of electrode arrays to other 3D cultures, such as destination organoids, by utilizing reversible engineering control over the geometry of the cultures. Although breakthroughs have been made in the electrophysiological monitoring of 3D cultures, they can bring organoids into contact with the substrate, which in turn affects the normal structure and function of the organoids. On the basis of this, Yang et al. 26 developed an electronic recording platform called “kirigami,” inspired by paper cutting and 3D folding. This platform can spontaneously transform into various 3D geometries in the suspended state, seamlessly integrate complete neurological organoids and assemblies, and support long-term recordings without interfering with the organoid’s differentiation and development.
In addition to the monitoring of electrophysiological information in organoids, exogenous electrical stimulation also promotes the proliferation and differentiation of stem cells. 148 Tai et al. 150 have devised a technique that utilizes hydroacoustic excitation to induce the piezoelectric effect in electrospun poly-nanofiber scaffolds. This technique drives the generation of electrical charge, which in turn confirms the multidirectional differentiation of neural stem cells and controls their fate through exogenous electrical signals. Li et al. 130 developed a stretchable electrode system that can be tightly integrated with neural organoids to trigger calcium signals by applying electrical stimulation. The electrical properties of the organoid can be flexibly conditioned at different current amplitudes and frequencies. In addition to studying the normal nervous system, the 3D neural technology platform developed by Park et al. 134 has made significant contributions to exploring neurodevelopmental disorders. Other organoids also respond to electrical stimulation, Shim’s team 131 also applied external electric field stimulation to 3D kidney and intestinal organoid models and observed that the electric field was able to cause significant tissue expansion.
Recently, electrophysiological tools with electronic devices have been used to study the electrical function of cardiac organoids during cardiac development. For example, Kim’s team 132 embedded three-dimensional liquid metal electrodes with a low modulus and low impedance into the heart organoids to achieve ECG signal acquisition and electrical stimulation without restricting the development of the organoids. By applying chronic electrical stimulation to myovascular organoids, Dailamy et al. 133 demonstrated that electrical stimulation is a potential pathway for organoids to mature and vascularize skeletal muscle tissue.
Response mechanisms combining other physicochemical stimuli (e.g. temperature, magnetic fields, metal ions, enzymes, ultrasound, pH, etc.) can be engineered to promote physical cues for organoid development.151–153
Information acquisition and processing
Organoid imaging analysis
Due to the unique 3D structure of organoids, the application of conventional imaging techniques faces great challenges in maximizing the advantages of organoid systems. Thus, there is a need to utilize advanced imaging techniques to dig deeper into their complex structures and functions. With the aid of advanced microscopy techniques such as confocal microscopy, 154 light sheet microscopy, 155 and two-photon microscopy, 156 it is possible to acquire images with higher resolution than conventional microscopes.
3D imaging offers more detailed and comprehensive information about organoid structures, including cell types and shapes. Given the unique opacity of organoids, the tissue transparency technique is particularly suited for their study. This technique allows researchers to clearly observe the internal structures of tissues in three-dimensional space without damaging them. It involves removing or replacing light-scattering elements with special solutions to render the tissues transparent, and then using microscopes capable of capturing the structure of these transparent tissues. Currently, transparent solutions such as SeeDB 157 and ScaleA2 158 are more widely used. By introducing fructose glycerol into tissue transparency techniques, Rios et al.154,159 obtained high-resolution volumetric images of multiple organ types, such as the intestines, mammary glands, and breast tumors, and the fructose glycerol reagent had greater transparency and quench resistance effects than conventional transparency protocols.
In addition to tissue transparency techniques, Khan et al. 160 present a positron emission microscopy approach that enables visualization as well as quantification of glycolytic activity in tumors by imaging a radiotracer (18F-fluorodeoxyglucose used here) in organoids. It can be used for personalized diagnosis and treatment with translational capabilities.
Biosensor
Electrochemical biosensors have also been used to some extent in organoid analysis, where they are immune to ambient light interference, are not dependent on microscopy, avoid the use of complex data analysis software, and are able to dynamically monitor the functional activity of organoids in real time, enabling the monitoring of multiple biomarkers through the use of small sample sizes, unlike microscopy image analysis.22,32 Zhang et al. 161 achieved continuous monitoring of cardiac and hepatic organs by integrating multiple biosensors into an organs-on-a-chips platform, which enables continuous and automated monitoring of a range of microenvironmental parameters (e.g. pH, oxygen, temperature, and several biomarkers) such as drug toxicity through the administration of adriamycin, acetaminophen, and biomarkers such as albumin, glutathione S-transferase alpha, and creatine kinase. Creatine kinases and other biomarkers were used to assess their toxicity. In addition to monitoring a single organoid, Lee et al. 162 constructed a way to culture breast cancer and cardiac organoids on the same platform, where they can communicate with each other, and by using electrochemical immunoadaptor sensors, they can noninvasively monitor biomarkers in the response of both to chemotherapeutic agents for the purpose of early monitoring and prediction of chemotherapy-induced cardiotoxicity. Similarly, Nguyen et al. 163 developed a dual-sensing nanoreporter capable of feeding back the activities of two key enzymes involved in T-cell-mediated target cell death during and subsequent to immune checkpoint antibody treatment, achieving efficient monitoring of dynamic immune processes in the tumor microenvironment and promising early prediction of treatment outcomes.
Although a wide variety of biosensors have been used in biomedical research, monitoring the different locations of organoids is still somewhat deficient because of their unique three-dimensional structure. To address this problem, Kalmykov’s research team 164 developed a 3D self-recoiling biosensor array (3D-SR-BA) triggered by electricity, pH, magnetism, or other physicochemical factors that transforms 2D electrodes into 3D shapes. By evaluating the electrode arrays via cardiac analogs, phase coordination between calcium transients and potentials was observed, revealing a great deal of potential for monitoring the electrophysiological activity of the analogs. The accuracy of physiological signals from organoids still needs to be optimized due to differences in cell source and culture conditions as well as sensor design. Converting the physicochemical signals of organoids into generalized information still urgently needs further research. Additionally, the complex metabolites of organoids require more strict materials for sensors.
Conclusions and perspectives
This review provides an overview of engineering strategies to overcome the limitations of traditional organoids. Although these technologies have essentially optimized the structure, and function, there is still potential for improvement.
The development of organs in vivo requires a certain growth environment, in particular, various mechanical parameters such as stress-strain and forces of interaction between cellular tissues and between cells and body fluids. There is no unanimous consensus on the growth environment in which organoid development occurs. The external environment needs to be accurately regulated to better approximate the real in vivo physiological environment of the human body, which in turn guides the differentiation and developmental maturation of the organoid. By combining microfluidics and organs-on-a-chips technologies, the perfusion of organoids simulates the real fluid shear force, mechanical stress, and other factors in vivo and restores the environment in the human body, which can promote the differentiation and developmental maturity of organoids.
In addition to nutrient delivery, the design of the organoids-on-chips also considers that the simulated fluid must not generate excessive fluid force, as high pressure can damage tissue cells. Furthermore, measures are taken to ensure that the flow within the channels is not affected by bubbles or other factors and that the material of the chip does not absorb drugs or other chemicals. Currently, organoids still have certain limitations in replicating the complex in vivo environment. For example, the liver exhibits metabolic heterogeneity across different regions of hepatocytes, impacting its executive function. Similarly, establishing a gas-liquid interface culture for the respiratory tract has significant potential.
The complex and changeable external environment leads to a high degree of variability in the phenotype of organoids, which also restricts the clinical transformation of organoids to a certain degree. Additionally, simulating real organs with organoids suffers from a lack of vascularization and immune function, the complexity of Matrigel in mimicking the ECM, and heterogeneity. In addition to the limitations of the culture microenvironment, the blood flow and immune function of the organoids are also a formidable challenge. Consequently, the results obtained from biomedical experiments conducted solely on organoids often differ from those observed in the real in vivo environment. Therefore, the integration of multiple cell types and spatial control using 3D printing and microfluidics can maximize the reproduction of the real ECM environment in vivo. The use of 3D printed microarrays to enable precise manipulation of individual organoids has led to a significant reduction in the batch variation of organoids and enhanced their homogeneity. The use of organoid culture media and Matrigel also limits their further clinical translation due to their unspecified composition, which may cause structural-functional variability of organs as well as adverse toxic reactions. In the future, new alternatives that are closer to the condition of real organs in the human body, for example, for culturing brain organoids using a medium that is closer to the composition of cerebrospinal fluid, are expected to be developed.
Apart from issues in culture and application, real-time monitoring various functional indices of organoids, which requires an in-depth exploration of the complex structure of organoids and their fine cellular behaviors with the help of engineering tools to reproduce the interactions between the organs and the real cellular ecological behaviors, is challenging. Smirnova et al. 165 introduced the term “organoid intelligence” (OI), which aims to expand the definition of biocomputing to include brain-directed OI computations, that is, to utilize the self-assembly mechanisms of brain organoids to memorize and compute inputs. The human brain is incomparable to computers in terms of information processing efficiency as well as energy efficiency. In the future, it is expected that the combination of the human brain and machines will be realized through the engineering and intellectualization of brain organoids into the currently emerging ChatGPT to mutually integrate with artificial intelligence and thus complement each other’s strengths. The expandability, feasibility, and durability of the organoids are supported through the use of microfluidic devices that provide various types of external information, such as electrical, chemical, and machine-programed signals with purposeful information to the organoids, and high-resolution outputs of electrophysiological signals obtained from microelectrode arrays and implantable probes that are analyzed, can be used directly for computation, and can be used as biofeedback to facilitate learning, to facilitate organoid learning.
The convergence of organoids and AI provides new opportunities to elucidate not only the biological mechanisms of human cognition, learning, and memory, but also new tools for a deeper understanding of the pathogenesis of diseases in various systems and for the development of new therapeutic regimens to address the numerous clinical needs of the future.
While organoid models have been validated with some known drug groups, such as cardiac organoids, and routinely incorporate cardiac fibroblasts, challenges remain in achieving long-term stable, perfusable blood vessels and incorporating immune cells to better mimic inflammatory (myocarditis), autoimmune (lupus), and regenerative (treatment of myocardial infarction) conditions. More work is needed to explore the interactions between neurons and the myocardium to better model the effects of the autonomic nervous system, capture circadian rhythms and heart rate variability, and understand metabolic effects. In addition, further development of sex-specific models of hormone-treated and senescent organ models is needed. 166 Any patient’s tissue can be used to create personalized organoids, although there are some differences in growth rates between patient tissue samples. Cells can be stored in biobanks and passaged in culture for long periods of time while retaining patient-specific phenotypes. This robustness is critical for standardization and clinical application. 167
Microphysiological systems (MPS), including organoid and organ-on-a-chip technologies, can overcome the limitations of the two major experimental platforms, cell culture and model animals, which are widely used for drug efficacy and safety. Patel et al. 168 compared MPS cultures to static cultures and found that the model predicted a reduction in hypoxia and that the MPS platform maintained organoid viability and function. In addition to improving organoid survival, it also provides the ability to evaluate in situ parameters. Similarly, the MPS platform has revealed the gastric mucosal defense mechanism of H. pylori. 169 Other organ classes, such as kidney, 63 liver, 170 and renal cancer, 171 have also been explored for disease and drug screening through integration with MPS.
Engineered organoids represent a critical step toward high-resolution modeling of quantitative assays of complex biological processes. Bioengineered organoids are therefore a valuable addition to the library of in vitro models and open exciting prospects for translational research, especially in the field of drug discovery. The focus of the organoid should be on the purpose of the model, namely the biological process it is designed to capture and the question we want to answer. This purpose should guide our choices in terms of model complexity and throughput.
In conclusion, organoid research is still in its infancy, and addressing these challenges requires a multidisciplinary effort. We believe that the engineering of organoids represents a new era for realizing the translation of organoids from the laboratory to the clinic.
Footnotes
ORCID iD: Gaofeng Liang
https://orcid.org/0000-0003-0097-0296
Statements and Declarations
Funding: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This work was financially supported by the Special Foundation for Basic Research Program of Higher Education Institutions of Henan Province (22ZX005), Key R&D project of Henan Province (221111310600), Joint Fund of Henan Province Science and Technology R&D Program (225200810020) and Postdoctoral Initiation Fund of Henan Academy of Sciences (231823049).
The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
References
- 1. Sebastião MJ, Gomes-Alves P, Reis I, et al. Bioreactor-based 3D human myocardial ischemia/reperfusion in vitro model: a novel tool to unveil key paracrine factors upon acute myocardial infarction. Transl Res 2020; 215: 57–74. [DOI] [PubMed] [Google Scholar]
- 2. Davies M, Peramuhendige P, King L, et al. Evaluation of in vitro models for assessment of human intestinal metabolism in drug discovery. Drug Metab Dispos 2020; 48(11): 1169–1182. [DOI] [PubMed] [Google Scholar]
- 3. Lancaster MA, Knoblich JA. Generation of cerebral organoids from human pluripotent stem cells. Nat Protoc 2014; 9(10): 2329–2340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Simian M, Bissell MJ. Organoids: a historical perspective of thinking in three dimensions. J Cell Biol 2017; 216(1): 31–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Sato T, Vries RG, Snippert HJ, et al. Single lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 2009; 459(7244): 262–265. [DOI] [PubMed] [Google Scholar]
- 6. Park SE, Georgescu A, Huh D. Organoids-on-a-chip. Science 2019; 364(6444): 960–965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Ren Y, Yang X, Ma Z, et al. Developments and opportunities for 3D bioprinted organoids. Int J Bioprinting 2021; 7(3): 364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Shen Q, Goderie SK, Jin L, et al. Endothelial cells stimulate self-renewal and expand neurogenesis of neural stem cells. Science 2004; 304(5675): 1338–1340. [DOI] [PubMed] [Google Scholar]
- 9. Lancaster MA, Renner M, Martin CA, et al. Cerebral organoids model human brain development and microcephaly. Nature 2013; 501(7467): 373–379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Qian X, Nguyen HN, Song MM, et al. Brain-region-specific organoids using mini-bioreactors for modeling ZIKV exposure. Cell 2016; 165(5): 1238–1254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Szebényi K, Wenger LMD, Sun Y, et al. Human ALS/FTD brain organoid slice cultures display distinct early astrocyte and targetable neuronal pathology. Nat Neurosci 2021; 24(11): 1542–1554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Sahu S, Sahoo S, Sullivan T, et al. Spatiotemporal modulation of growth factors directs the generation of multilineage mouse embryonic stem cell-derived mammary organoids. Dev Cell 2024; 59(2): 175–186.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Eicher AK, Kechele DO, Sundaram N, et al. Functional human gastrointestinal organoids can be engineered from three primary germ layers derived separately from pluripotent stem cells. Cell Stem Cell 2022; 29(1): 36–51.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Tao T, Deng P, Wang Y, et al. Microengineered multi-organoid system from hiPSCs to recapitulate human liver-islet axis in normal and type 2 diabetes. Adv Sci 2022; 9(5): e2103495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Park JS, Chu JS, Tsou AD, et al. The effect of matrix stiffness on the differentiation of mesenchymal stem cells in response to TGF-β. Biomaterials 2011; 32(16): 3921–3930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Hu J, Shi Y, Zhang J, et al. Melanopsin retinal ganglion cells mediate light-promoted brain development. Cell 2022; 185(17): 3124–3137.e15. [DOI] [PubMed] [Google Scholar]
- 17. Kawai T, Morioka S, Miyata H, et al. The significance of electrical signals in maturing spermatozoa for phosphoinositide regulation through voltage-sensing phosphatase. Nat Commun 2024; 15(1): 7289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Fatehullah A, Tan SH, Barker N. Organoids as an in vitro model of human development and disease. Nat Cell Biol 2016; 18(3): 246–254. [DOI] [PubMed] [Google Scholar]
- 19. Clevers H. Modeling development and disease with organoids. Cell 2016; 165(7): 1586–1597. [DOI] [PubMed] [Google Scholar]
- 20. Camp JG, Badsha F, Florio M, et al. Human cerebral organoids recapitulate gene expression programs of fetal neocortex development. Proc Natl Acad Sci USA 2015; 112(51): 15672–15677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Velasco S, Kedaigle AJ, Simmons SK, et al. Individual brain organoids reproducibly form cell diversity of the human cerebral cortex. Nature 2019; 570(7762): 523–527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Brandenberg N, Hoehnel S, Kuttler F, et al. High-throughput automated organoid culture via stem-cell aggregation in microcavity arrays. Nat Biomed Eng 2020; 4(9): 863–874. [DOI] [PubMed] [Google Scholar]
- 23. Renner H, Grabos M, Becker KJ, et al. A fully automated high-throughput workflow for 3D-based chemical screening in human midbrain organoids. eLife 2020; 9: 1–19. DOI: 10.7554/eLife.52904 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Wang Y, Jeon H. 3D cell cultures toward quantitative high-throughput drug screening. Trends Pharmacol Sci 2022; 43(7): 569–581. [DOI] [PubMed] [Google Scholar]
- 25. Darville H, Poulet A, Rodet-Amsellem F, et al. Human pluripotent stem cell-derived cortical neurons for high throughput medication screening in autism: a proof of concept study in SHANK3 haploinsufficiency syndrome. EBioMedicine 2016; 9: 293–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Yang X, Forró C, Li TL, et al. Kirigami electronics for long-term electrophysiological recording of human neural organoids and assembloids. Nat Biotechnol 2024; 42(12): 1836–1843. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Osaki T, Duenki T, Chow SYA, et al. Complex activity and short-term plasticity of human cerebral organoids reciprocally connected with axons. Nat Commun 2024; 15(1): 2945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Samarasinghe RA, Miranda OA, Buth JE, et al. Identification of neural oscillations and epileptiform changes in human brain organoids. Nat Neurosci 2021; 24(10): 1488–1500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Collins S, Yuen G, Tu T, et al. in vitro models of the liver: disease modeling, drug discovery and clinical applications. In: JE Tirnitz-Parker. (ed.) Hepatocellular carcinoma. Brisbane: Codon Publications, 2019, pp.47–67. [PubMed] [Google Scholar]
- 30. Caluori G, Pribyl J, Pesl M, et al. Non-invasive electromechanical cell-based biosensors for improved investigation of 3D cardiac models. Biosens Bioelectron 2019; 124-125: 129–135. [DOI] [PubMed] [Google Scholar]
- 31. Jiang S, Zhao H, Zhang W, et al. An automated organoid platform with inter-organoid homogeneity and inter-patient heterogeneity. Cell Rep Med 2020; 1(9): 100161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Hofer M, Lutolf MP. Engineering organoids. Nat Rev Mater 2021; 6(5): 402–420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Zhao Z, Chen X, Dowbaj AM, et al. Organoids. Nat Rev Methods Primers 2022; 2: 402–420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Yin X, Mead BE, Safaee H, et al. Engineering stem cell organoids. Cell Stem Cell 2016; 18(1): 25–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Clevers H, Lancaster M, Takebe T, Advances in organoid technology: Hans Clevers, Madeline Lancaster, and Takanori Takebe. Cell Stem Cell 2017; 20(6): 759–762. [Google Scholar]
- 36. Kim M, Mun H, Sung CO, et al. Patient-derived lung cancer organoids as in vitro cancer models for therapeutic screening. Nat Commun 2019; 10(1): 3991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Al Shihabi A, Davarifar A, Nguyen HTL, et al. Personalized chordoma organoids for drug discovery studies. Sci Adv 2022; 8(7): eabl3674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Schmidt C, Deyett A, Ilmer T, et al. Multi-chamber cardioids unravel human heart development and cardiac defects. Cell 2023; 186(25): 5587–5605.e27. [DOI] [PubMed] [Google Scholar]
- 39. Zeng Z, Huang B, Parvez RK, et al. Generation of patterned kidney organoids that recapitulate the adult kidney collecting duct system from expandable ureteric bud progenitors. Nat Commun 2021; 12(1): 3641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Ren B, Chiaravalloti TR, Belony NL, et al. Design and realization of lung organoid cultures for COVID-19 applications. Biodes Manuf 2023; 6(6): 646–660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Barker N, Huch M, Kujala P, et al. Lgr5(+ve) stem cells drive self-renewal in the stomach and build long-lived gastric units in vitro. Cell Stem Cell 2010; 6(1): 25–36. [DOI] [PubMed] [Google Scholar]
- 42. Karthaus W, Iaquinta P, Drost J, et al. Identification of multipotent luminal progenitor cells in human prostate organoid cultures. Cell 2014; 159(1): 163–175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Hohwieler M, Illing A, Hermann PC, et al. Human pluripotent stem cell-derived acinar/ductal organoids generate human pancreas upon orthotopic transplantation and allow disease modelling. Gut 2017; 66(3): 473–486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Takebe T, Sekine K, Enomura M, et al. Vascularized and functional human liver from an iPSC-derived organ bud transplant. Nature 2013; 499(7459): 481–484. [DOI] [PubMed] [Google Scholar]
- 45. Yan Z, Ohuchida K, Fei S, et al. Inhibition of ERK1/2 in cancer-associated pancreatic stellate cells suppresses cancer-stromal interaction and metastasis. J Exp Clin Cancer Res 2019; 38(1): 221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Vishy CE, Thomas C, Vincent T, et al. Genetics of cystogenesis in base-edited human organoids reveal therapeutic strategies for polycystic kidney disease. Cell Stem Cell 2024; 31(4): 537–553.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Kondo T, Ebinuma I, Tanaka H, et al. Rapid and robust multi-phenotypic assay system for ALS using human iPS cells with mutations in causative genes. Int J Mol Sci 2023; 24(8): 1–2. 20417314251319475 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Zhao H, Cheng Y, Kalra A, et al. Generation and multiomic profiling of a TP53/CDKN2A double-knockout gastroesophageal junction organoid model. Sci Transl Med 2022; 14(673): eabq6146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Duan X, Zhang T, Feng L, et al. A pancreatic cancer organoid platform identifies an inhibitor specific to mutant KRAS. Cell Stem Cell 2024; 31(1): 71–88.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Kadosh E, Snir-Alkalay I, Venkatachalam A, et al. The gut microbiome switches mutant p53 from tumour-suppressive to oncogenic. Nature 2020; 586(7827): 133–138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Masi AC, Fofanova TY, Lamb CA, et al. Distinct gene expression profiles between human preterm-derived and adult-derived intestinal organoids exposed to Enterococcus faecalis: a pilot study. Gut 2021; 71(10): 2141–2143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Chooi WH, Ng CY, Ow V, et al. Defined alginate hydrogels support spinal cord organoid derivation, maturation, and modeling of spinal cord diseases. Adv Healthc Mater 2023; 12(9): e2202342. [DOI] [PubMed] [Google Scholar]
- 53. Chen C, Rengarajan V, Kjar A, et al. A matrigel-free method to generate matured human cerebral organoids using 3D-Printed microwell arrays. Bioact Mater 2021; 6(4): 1130–1139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Loebel C, Weiner AI, Eiken MK, et al. Microstructured hydrogels to guide self-assembly and function of lung alveolospheres. Adv Mater 2022; 34(28): e2202992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Zhu Y, Wang L, Yu H, et al. In situ generation of human brain organoids on a micropillar array. Lab Chip 2017; 17(17): 2941–2950. [DOI] [PubMed] [Google Scholar]
- 56. Cui K, Wang Y, Zhu Y, et al. Neurodevelopmental impairment induced by prenatal valproic acid exposure shown with the human cortical organoid-on-a-chip model. Microsyst Nanoeng 2020; 6: 49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Shaji M, Tamada A, Fujimoto K, et al. Deciphering potential vascularization factors of on-chip co-cultured hiPSC-derived cerebral organoids. Lab Chip 2024; 24(4): 680–696. [DOI] [PubMed] [Google Scholar]
- 58. Salmon I, Grebenyuk S, Abdel Fattah AR, et al. Engineering neurovascular organoids with 3D printed microfluidic chips. Lab Chip 2022; 22(8): 1615–1629. [DOI] [PubMed] [Google Scholar]
- 59. Ao Z, Song S, Tian C, et al. Understanding immune-driven brain aging by human brain organoid microphysiological analysis platform. Adv Sci 2022; 9(27): e2200475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Decembrini S, Hoehnel S, Brandenberg N, et al. Hydrogel-based milliwell arrays for standardized and scalable retinal organoid cultures. Sci Rep 2020; 10(1): 10275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Min S, Kim S, Sim WS, et al. Versatile human cardiac tissues engineered with perfusable heart extracellular microenvironment for biomedical applications. Nat Commun 2024; 15(1): 2564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Rajasekar S, Lin DSY, Abdul L, et al. IFlowPlate-A customized 384-well plate for the culture of perfusable vascularized colon organoids. Adv Mater 2020; 32(46): e2002974. [DOI] [PubMed] [Google Scholar]
- 63. Homan KA, Gupta N, Kroll KT, et al. Flow-enhanced vascularization and maturation of kidney organoids in vitro. Nat Methods 2019; 16(3): 255–262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Lai Benjamin FL, Lu Rick X, Hu Y, et al. Recapitulating pancreatic tumor microenvironment through synergistic use of patient organoids and organ-on-a-chip vasculature. Adv Funct Mater 2020; 30(48): 1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Abbasalizadeh S, Babaee S, Kowsari-Esfahan R, et al. Continuous production of highly functional vascularized hepatobiliary organoids from human pluripotent stem cells using a scalable microfluidic platform. Adv Funct Mater 2023; 33(49): 2210233. [Google Scholar]
- 66. Meyvantsson I, Beebe DJ. Cell culture models in microfluidic systems. Annu Rev Anal Chem 2008; 1: 423–449. [DOI] [PubMed] [Google Scholar]
- 67. MacKerron C, Robertson G, Zagnoni M, et al. A microfluidic platform for the characterisation of CNS active compounds. Sci Rep 2017; 7(1): 15692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Halldorsson S, Lucumi E, Gómez-Sjöberg R, et al. Advantages and challenges of microfluidic cell culture in polydimethylsiloxane devices. Biosens Bioelectron 2015; 63: 218–231. [DOI] [PubMed] [Google Scholar]
- 69. Osaki T, Uzel SGM, Kamm RD. Microphysiological 3D model of amyotrophic lateral sclerosis (ALS) from human iPS-derived muscle cells and optogenetic motor neurons. Sci Adv 2018; 4(10): eaat5847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Oddo A, Peng B, Tong Z, et al. Advances in microfluidic blood-brain barrier (BBB) models. Trends Biotechnol 2019; 37(12): 1295–1314. [DOI] [PubMed] [Google Scholar]
- 71. Park J, Lee BK, Jeong GS, et al. Three-dimensional brain-on-a-chip with an interstitial level of flow and its application as an in vitro model of Alzheimer’s disease. Lab Chip 2015; 15(1): 141–150. [DOI] [PubMed] [Google Scholar]
- 72. Wang 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(10): 5734–5743. [DOI] [PubMed] [Google Scholar]
- 73. Nguyen VVT, Ye S, Gkouzioti V, et al. A human kidney and liver organoid-based multi-organ-on-a-chip model to study the therapeutic effects and biodistribution of mesenchymal stromal cell-derived extracellular vesicles. J Extracell Vesicles 2022; 11(11): e12280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Vargas-Valderrama A, Messina A, Mitjavila-Garcia MT, et al. The endothelium, a key actor in organ development and hPSC-derived organoid vascularization. J Biomed Sci 2020; 27(1): 67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Rauth S, Karmakar S, Batra SK, et al. Recent advances in organoid development and applications in disease modeling. Biochim Biophys Acta Rev Cancer 2021; 1875(2): 188527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Shi Y, Sun L, Wang M, et al. Vascularized human cortical organoids (vOrganoids) model cortical development in vivo. PLoS Biol 2020; 18(5): e3000705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Xu NY, Li J, Wang ML, et al. Fabrication of a coculture organoid model in the biomimetic matrix of alginate to investigate breast cancer progression in a TAMs-Leading immune microenvironment. ACS Appl Mater Interfaces 2024; 16(9): 11275–11288. [DOI] [PubMed] [Google Scholar]
- 78. Qian X, Jacob F, Song MM, et al. Generation of human brain region-specific organoids using a miniaturized spinning bioreactor. Nat Protoc 2018; 13(3): 565–580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Rezaei B, Giacomoni J, Nilsson F, et al. Modular 3D printed platform for fluidically connected human brain organoid culture. Biofabrication 2023; 16(1): 1–9. 20417314251319475 [DOI] [PubMed] [Google Scholar]
- 80. Sun X, Cui Z, Liang Y, et al. One-stop assembly of adherent 3D retinal organoids from hiPSCs based on 3D-printed derived PDMS microwell platform. Biofabrication 2023; 15(3): 1–17. 20417314251319475 [DOI] [PubMed] [Google Scholar]
- 81. Bonnans C, Chou J, Werb Z. Remodelling the extracellular matrix in development and disease. Nat Rev Mol Cell Biol 2014; 15(12): 786–801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Caralt M, Uzarski JS, Iacob S, et al. Optimization and critical evaluation of decellularization strategies to develop renal extracellular matrix scaffolds as biological templates for organ engineering and transplantation. Am J Transplant 2015; 15(1): 64–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Ferreira LP, Jorge C, Lagarto MR, et al. Photoacoustic processing of decellularized extracellular matrix for biofabricating living constructs. Acta Biomater 2024; 183: 74–88. [DOI] [PubMed] [Google Scholar]
- 84. Chen HJ, Wei Z, Sun J, et al. A recellularized human colon model identifies cancer driver genes. Nat Biotechnol 2016; 34(8): 845–851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Liu TK, Pang Y, Zhou ZZ, et al. An integrated cell printing system for the construction of heterogeneous tissue models. Acta Biomater 2019; 95: 245–257. [DOI] [PubMed] [Google Scholar]
- 86. Ding S, Feng L, Wu J, et al. Bioprinting of stem cells: Interplay of bioprinting process, bioinks, and stem cell properties. ACS Biomater Sci Eng 2018; 4(9): 3108–3124. [DOI] [PubMed] [Google Scholar]
- 87. Mekhileri NV, Lim KS, Brown GCJ, et al. Automated 3D bioassembly of micro-tissues for biofabrication of hybrid tissue engineered constructs. Biofabrication 2018; 10(2): 024103. [DOI] [PubMed] [Google Scholar]
- 88. Aisenbrey EA, Murphy WL. Synthetic alternatives to Matrigel. Nat Rev Mater 2020; 5(7): 539–551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Kleinman HK, Martin GR. Matrigel: basement membrane matrix with biological activity. Semin Cancer Biol 2005; 15(5): 378–386. [DOI] [PubMed] [Google Scholar]
- 90. Broguiere N, Isenmann L, Hirt C, et al. Growth of epithelial organoids in a defined hydrogel. Adv Mater 2018; 30(43): e1801621. [DOI] [PubMed] [Google Scholar]
- 91. Lindborg BA, Brekke JH, Vegoe AL, et al. Rapid induction of cerebral organoids from human induced pluripotent stem cells using a chemically defined hydrogel and defined Cell Culture Medium. Stem Cells Transl Med 2016; 5(7): 970–979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Agarwal T, Celikkin N, Costantini M, et al. Recent advances in chemically defined and tunable hydrogel platforms for organoid culture. Des Manuf 2021; 4(3): 641–674. [Google Scholar]
- 93. Giobbe GG, Crowley C, Luni C, et al. Extracellular matrix hydrogel derived from decellularized tissues enables endodermal organoid culture. Nat Commun 2019; 10(1): 5658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Huang B, Zeng Z, Kim S, et al. Long-term expandable mouse and human-induced nephron progenitor cells enable kidney organoid maturation and modeling of plasticity and disease. Cell Stem Cell 2024; 31(6): 921–939.e17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Astashkina AI, Mann BK, Prestwich GD, et al. A 3-D organoid kidney culture model engineered for high-throughput nephrotoxicity assays. Biomaterials 2012; 33(18): 4700–4711. [DOI] [PubMed] [Google Scholar]
- 96. Cassel de Camps C, Aslani S, Stylianesis N, et al. Hydrogel mechanics influence the growth and development of embedded brain organoids. ACS Appl Bio Mater 2022; 5(1): 214–224. [DOI] [PubMed] [Google Scholar]
- 97. DiMarco RL, Su J, Yan KS, et al. Engineering of three-dimensional microenvironments to promote contractile behavior in primary intestinal organoids. Integr Biol 2014; 6(2): 127–142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Shi W, Mirza S, Kuss M, et al. Embedded bioprinting of breast tumor cells and organoids using Low-Concentration collagen-based bioinks. Adv Healthc Mater 2023; 12(26): e2300905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. El-Derby AM, Khedr MA, Ghoneim NI, et al. Plasma-derived extracellular matrix for xenofree and cost-effective organoid modeling for hepatocellular carcinoma. J Transl Med 2024; 22(1): 487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Gjorevski N, Nikolaev M, Brown TE, et al. Tissue geometry drives deterministic organoid patterning. Science 2022; 375(6576): eaaw9021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Rothenbücher TSP, Gürbüz H, Pereira MP, et al. Next generation human brain models: engineered flat brain organoids featuring gyrification. Biofabrication 2021; 13(1): 011001. [DOI] [PubMed] [Google Scholar]
- 102. Shaffiey SA, Jia H, Keane T, et al. Intestinal stem cell growth and differentiation on a tubular scaffold with evaluation in small and large animals. Regen Med 2016; 11(1): 45–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Treacy NJ, Clerkin S, Davis JL, et al. Growth and differentiation of human induced pluripotent stem cell (hiPSC)-derived kidney organoids using fully synthetic peptide hydrogels. Bioact Mater 2023; 21: 142–156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Tysoe OC, Justin AW, Brevini T, et al. Isolation and propagation of primary human cholangiocyte organoids for the generation of bioengineered biliary tissue. Nat Protoc 2019; 14(6): 1884–1925. [DOI] [PubMed] [Google Scholar]
- 105. De Santis MM, Alsafadi HN, Tas S, et al. Extracellular-matrix-reinforced bioinks for 3D bioprinting human tissue. Adv Mater 2021; 33(3): e2005476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Urciuolo A, Giobbe GG, Dong Y, et al. Hydrogel-in-hydrogel live bioprinting for guidance and control of organoids and organotypic cultures. Nat Commun 2023; 14(1): 3128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Kloxin AM, Kasko AM, Salinas CN, et al. Photodegradable hydrogels for dynamic tuning of physical and chemical properties. Science 2009; 324(5923): 59–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Wilkinson DC, Alva-Ornelas JA, Sucre JM, et al. Development of a three-dimensional bioengineering technology to generate lung tissue for personalized disease modeling. Stem Cells Transl Med 2017; 6(2): 622–633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Tomaszewski CE, DiLillo KM, Baker BM, et al. Sequestered cell-secreted extracellular matrix proteins improve murine folliculogenesis and oocyte maturation for fertility preservation. Acta Biomater 2021; 132: 313–324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Cruz-Acuña R, Quirós M, Farkas AE, et al. Synthetic hydrogels for human intestinal organoid generation and colonic wound repair. Nat Cell Biol 2017; 19(11): 1326–1335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Enemchukwu NO, Cruz-Acuña R, Bongiorno T, et al. Synthetic matrices reveal contributions of ECM biophysical and biochemical properties to epithelial morphogenesis. J Cell Biol 2016; 212(1): 113–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Sato T, Stange DE, Ferrante M, et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett’s epithelium. Gastroenterology 2011; 141(5): 1762–1772. [DOI] [PubMed] [Google Scholar]
- 113. Renner H, Schöler HR, Bruder JM. Combining automated organoid workflows with artificial intelligence-based analyses: opportunities to build a new generation of interdisciplinary high-throughput screens for Parkinson’s disease and beyond. Mov Disord 2021; 36(12): 2745–2762. [DOI] [PubMed] [Google Scholar]
- 114. Saorin G, Caligiuri I, Rizzolio F. Microfluidic organoids-on-a-chip: the future of human models. Semin Cell Dev Biol 2023; 144: 41–54. [DOI] [PubMed] [Google Scholar]
- 115. Xia M, Wu M, Li Y, et al. Varying mechanical forces drive sensory epithelium formation. Sci Adv 2023; 9(44): eadf2664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Ruiter FAA, Morgan FLC, Roumans N, et al. Soft, dynamic hydrogel confinement improves kidney organoid lumen morphology and reduces epithelial-mesenchymal transition in culture. Adv Sci 2022; 9(20): e2200543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Koser DE, Moeendarbary E, Hanne J, et al. CNS cell distribution and axon orientation determine local spinal cord mechanical properties. Biophys J 2015; 108(9): 2137–2147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Abdel Fattah AR, Daza B, Rustandi G, et al. Actuation enhances patterning in human neural tube organoids. Nat Commun 2021; 12(1): 3192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Karzbrun E, Kshirsagar A, Cohen SR, et al. Human Brain organoids on a chip reveal the physics of folding. Nat Phys 2018; 14(5): 515–522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Sorrentino G, Rezakhani S, Yildiz E, et al. Mechano-modulatory synthetic niches for liver organoid derivation. Nat Commun 2020; 11(1): 3416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Hushka EA, Yavitt FM, Brown TE, et al. Relaxation of extracellular matrix forces directs crypt formation and architecture in intestinal organoids. Adv Healthc Mater 2020; 9(8): e1901214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Yavitt FM, Kirkpatrick BE, Blatchley MR, et al. In situ modulation of intestinal organoid epithelial curvature through photoinduced viscoelasticity directs crypt morphogenesis. Sci Adv 2023; 9(3): eadd5668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Xu Y, Zhou A, Chen W, et al. An integrative bioorthogonal nanoengineering strategy for dynamically constructing heterogenous tumor spheroids. Adv Mater 2023; 35(49): e2304172. [DOI] [PubMed] [Google Scholar]
- 124. Roscigno G, Affinito A, Quintavalle C, et al. Ultrasmall carbon nanodots as theranostic nanoheaters for precision breast cancer phototherapy: establishing the translational potential in tumor-in-a-dish models. ACS Biomater Sci Eng 2024; 10(7): 4269–4278. [DOI] [PubMed] [Google Scholar]
- 125. Liu Y, Zhang L, Hu N, et al. An optogenetic approach for regulating human parathyroid hormone secretion. Nat Commun 2022; 13(1): 771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Liu G, Wei J, Li X, et al. Near-infrared-responded high sensitivity nanoprobe for steady and visualized detection of albumin in hepatic organoids and mouse liver. Adv Sci 2022; 9(26): e2202505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Fang H, Yao S, Chen Q, et al. De novo-designed near-infrared nanoaggregates for super-resolution monitoring of lysosomes in cells, in whole organoids, and in vivo. ACS Nano 2019; 13(12): 14426–14436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Li Y, Xiong J, Guo W, et al. Decomposable black phosphorus nano-assembly for controlled delivery of cisplatin and inhibition of breast cancer metastasis. J Control Release 2021; 335: 59–74. [DOI] [PubMed] [Google Scholar]
- 129. Liu J, Yi K, Zhang Q, et al. Strong penetration-induced effective photothermal therapy by exosome-mediated black phosphorus quantum dots. Small 2021; 17(49): e2104585. [DOI] [PubMed] [Google Scholar]
- 130. Li TL, Liu Y, Forro C, et al. Stretchable mesh microelectronics for the biointegration and stimulation of human neural organoids. Biomaterials 2022; 290: 121825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Shim G, Breinyn IB, Martínez-Calvo A, et al. Bioelectric stimulation controls tissue shape and size. Nat Commun 2024; 15(1): 2938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Kim M, Hwang JC, Min S, et al. Multimodal characterization of cardiac organoids using integrations of pressure-sensitive transistor arrays with three-dimensional liquid metal electrodes. Nano Lett 2022; 22(19): 7892–7901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Dailamy A, Parekh U, Katrekar D, et al. Programmatic introduction of parenchymal cell types into blood vessel organoids. Stem Cell Reports 2021; 16(10): 2432–2441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Park Y, Franz CK, Ryu H, et al. Three-dimensional, multifunctional neural interfaces for cortical spheroids and engineered assembloids. Sci Adv 2021; 7(12): 1–11. 20417314251319475 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Knopman DS, Amieva H, Petersen RC, et al. Alzheimer disease. Nat Rev Dis Primers 2021; 7(1): 33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Hu J, Chen Q, Zhu H, et al. Microglial piezo1 senses aβ fibril stiffness to restrict Alzheimer’s disease. Neuron 2023; 111(1): 15–29.e8. [DOI] [PubMed] [Google Scholar]
- 137. Lipp A, Trbojevic R, Paul F, et al. Cerebral magnetic resonance elastography in supranuclear palsy and idiopathic Parkinson’s disease. NeuroImage Clin 2013; 3: 381–387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Freimann FB, Müller S, Streitberger K, et al. MR elastography in a murine stroke model reveals correlation of macroscopic viscoelastic properties of the brain with neuronal density. NMR Biomed 2013; 26(11): 1534–1539. [DOI] [PubMed] [Google Scholar]
- 139. Ranga A, Girgin M, Meinhardt A, et al. Neural tube morphogenesis in synthetic 3D microenvironments. Proc Natl Acad Sci USA 2016; 113(44): E6831–E6839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Zhu R, Sun Z, Li C, et al. Electrical stimulation affects neural stem cell fate and function in vitro. Exp Neurol 2019; 319: 112963. [DOI] [PubMed] [Google Scholar]
- 141. Gabriel E, Albanna W, Pasquini G, et al. Human brain organoids assemble functionally integrated bilateral optic vesicles. Cell Stem Cell 2021; 28(10): 1740–1757.e1748. [DOI] [PubMed] [Google Scholar]
- 142. Jgamadze D, Lim JT, Zhang Z, et al. Structural and functional integration of human forebrain organoids with the injured adult rat visual system. Cell Stem Cell 2023; 30(2): 137–152.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Wilson MN, Thunemann M, Liu X, et al. Multimodal monitoring of human cortical organoids implanted in mice reveal functional connection with visual cortex. Nat Commun 2022; 13(1): 7945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Brown TE, Anseth KS. Spatiotemporal hydrogel biomaterials for regenerative medicine. Chem Soc Rev 2017; 46(21): 6532–6552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Crowder SW, Leonardo V, Whittaker T, et al. Material cues as potent regulators of epigenetics and stem cell function. Cell Stem Cell 2016; 18(1): 39–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Wells RG. The role of matrix stiffness in regulating cell behavior. Hepatology 2008; 47(4): 1394–1400. [DOI] [PubMed] [Google Scholar]
- 147. Broguiere N, Lüchtefeld I, Trachsel L, et al. Morphogenesis guided by 3D patterning of growth factors in biological matrices. Adv Mater 2020; 32(25): e1908299. [DOI] [PubMed] [Google Scholar]
- 148. Sandquist EJ, Uz M, Sharma AD, et al. Stem cells, bioengineering, and 3-D scaffolds for nervous system repair and regeneration. In: LG Zhang, DL Kaplan. (eds) Neural engineering: from advanced biomaterials to 3D fabrication techniques. Cham: Springer International Publishing, 2016, pp.25–81. [Google Scholar]
- 149. Trujillo CA, Gao R, Negraes PD, et al. Complex oscillatory waves emerging from cortical organoids model early human brain network development. Cell Stem Cell 2019; 25(4): 558–569.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Tai Y, Ico G, Low K, et al. Formation of 3D self-organized neuron-glial interface derived from neural stem cells via Mechano-electrical stimulation. Adv Healthc Mater 2021; 10(19): e2100806. [DOI] [PubMed] [Google Scholar]
- 151. Blache U, Ford EM, Ha B, et al. Engineered hydrogels for mechanobiology. Nat Rev Methods Primers 2022; 2: 98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Cao H, Duan L, Zhang Y, et al. Current hydrogel advances in physicochemical and biological response-driven biomedical application diversity. Signal Transduct Target Ther 2021; 6(1): 426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Tanaka M, Nakahata M, Linke P, et al. Stimuli-responsive hydrogels as a model of the dynamic cellular microenvironment. Polym J 2020; 52(8): 861–870. [Google Scholar]
- 154. Dekkers JF, Alieva M, Wellens LM, et al. High-resolution 3D imaging of fixed and cleared organoids. Nat Protoc 2019; 14(6): 1756–1771. [DOI] [PubMed] [Google Scholar]
- 155. de Medeiros G, Ortiz R, Strnad P, et al. Multiscale light-sheet organoid imaging framework. Nat Commun 2022; 13(1): 4864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Kim S, Lowe A, Dharmat R, et al. Generation, transcriptome profiling, and functional validation of cone-rich human retinal organoids. Proc Natl Acad Sci USA 2019; 116(22): 10824–10833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Ke MT, Fujimoto S, Imai T. SeeDB: a simple and morphology-preserving optical clearing agent for neuronal circuit reconstruction. Nat Neurosci 2013; 16(8): 1154–1161. [DOI] [PubMed] [Google Scholar]
- 158. Hama H, Kurokawa H, Kawano H, et al. Scale: a chemical approach for fluorescence imaging and reconstruction of transparent mouse brain. Nat Neurosci 2011; 14(11): 1481–1488. [DOI] [PubMed] [Google Scholar]
- 159. Rios AC, Capaldo BD, Vaillant F, et al. Intraclonal plasticity in mammary tumors revealed through large-scale single-cell resolution 3D imaging. Cancer Cell 2019; 35(6): 618–632.e6. [DOI] [PubMed] [Google Scholar]
- 160. Khan S, Shin JH, Ferri V, et al. High-resolution positron emission microscopy of patient-derived tumor organoids. Nat Commun 2021; 12(1): 5883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Zhang YS, Aleman J, Shin SR, et al. Multisensor-integrated organs-on-chips platform for automated and continual in situ monitoring of organoid behaviors. Proc Natl Acad Sci USA 2017; 114(12): E2293–E2302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Lee J, Mehrotra S, Zare-Eelanjegh E, et al. A heart-breast cancer-on-a-chip platform for disease modeling and monitoring of cardiotoxicity induced by cancer chemotherapy. Small 2021; 17(15): 2004258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Nguyen A, Ramesh A, Fish A, et al. Dual-sensing nanoreporter for dynamic and high-throughput monitoring of immune checkpoint inhibitor responses in tumor-derived organoids. Adv Funct Mater 2024; 34(33): 2400393. [Google Scholar]
- 164. Kalmykov A, Huang C, Bliley J, et al. Organ-on-e-chip: three-dimensional self-rolled biosensor array for electrical interrogations of human electrogenic spheroids. Sci Adv 2019; 5(8): eaax0729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Smirnova L, Caffo BS, Gracias DH, et al. Organoid intelligence (OI): the new frontier in biocomputing and intelligence-in-a-dish. Front Sci 2023; 1: 1. [Google Scholar]
- 166. Wu Q, Rafatian N, Wagner KT, et al. SARS-CoV-2 pathogenesis in an angiotensin II-induced heart-on-a-chip disease model and extracellular vesicle screening. Proc Natl Acad Sci USA 2024; 121(28): e2403581121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Spelier S, de Winter-de Groot K, Keijzer-Nieuwenhuijze N, et al. Organoid-guided synergistic treatment of minimal function CFTR mutations with CFTR modulators, roflumilast and simvastatin: a personalised approach. Eur Respir J 2024; 63(1): 1–5. 20417314251319475 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Patel SN, Ishahak M, Chaimov D, et al. Organoid microphysiological system preserves pancreatic islet function within 3D matrix. Sci Adv 2021; 7(7): eaba5515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. Jeong HJ, Park JH, Kang JH, et al. Organoid-based human stomach micro-physiological system to recapitulate the dynamic mucosal defense mechanism. Adv Sci 2023; 10(27): e2300164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Bircsak KM, DeBiasio R, Miedel M, et al. A 3D microfluidic liver model for high throughput compound toxicity screening in the OrganoPlate®. Toxicology 2021; 450: 152667. [DOI] [PubMed] [Google Scholar]
- 171. Miller CP, Tsuchida C, Zheng Y, et al. A 3D human renal cell carcinoma-on-a-chip for the study of tumor angiogenesis. Neoplasia 2018; 20(6): 610–620. [DOI] [PMC free article] [PubMed] [Google Scholar]

