Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Mar 1.
Published in final edited form as: Adv Mater. 2023 Dec 6;36(9):e2307678. doi: 10.1002/adma.202307678

ENGINEERED SYNTHETIC MATRICES FOR HUMAN INTESTINAL ORGANOID CULTURE AND THERAPEUTIC DELIVERY

Adriana Mulero-Russe a,b, Andrés J García b,c
PMCID: PMC10922691  NIHMSID: NIHMS1952761  PMID: 37987171

Abstract

Human intestinal organoids (HIOs) derived from pluripotent stem cells or adult stem cell biopsies represent a powerful platform to study human development, drug testing, and disease modeling in vitro, and serve as a cell source for tissue regeneration and therapeutic advances in vivo. Synthetic hydrogels can be engineered to serve as analogs of the extracellular matrix to support HIO growth and differentiation. These hydrogels allow for tuning the mechanical and biochemical properties of the matrix, offering an advantage over biologically derived hydrogels such as Matrigel. Human intestinal organoids have been used for repopulating transplantable intestinal grafts and for in vivo delivery to an injured intestinal site. The use of synthetic hydrogels for in vitro culture and for in vivo delivery is expected to significantly increase the relevance of human intestinal organoids for drug screening, disease modeling, and therapeutic applications.

Graphical Abstract

graphic file with name nihms-1952761-f0001.jpg

Human intestinal organoids (HIOs) derived from pluripotent stem cells or adult stem cell biopsies represent a powerful platform to study human development, drug testing, and disease modeling in vitro, and serve as a cell source for tissue regeneration and therapeutic advances in vivo. Synthetic hydrogels can be engineered to serve as analogs of the extracellular matrix to support HIO growth and differentiation as well as delivery vehicles in therapeutic applications.

Introduction

Human intestinal organoids (HIOs) are defined as in vitro, three dimensional cellular structures that recapitulate the architecture, structure, cellular composition, and function of human intestinal tissue14. Based on their remarkable ability to mimic human intestinal tissue, HIOs provide powerful platforms for regenerative medicine58, personalized medicine9, drug testing10,11, studies of intestinal development12,13, and disease modeling1416 (Fig. 1). These organoids can be derived from intestinal tissue biopsies17 (also referred to as patient-derived intestinal organoid [PDO], adult stem cell-derived [ASC], intestinal stem cells [ISCs], or enteroids) or from human pluripotent stem cells (PSC)12 (induced pluripotent stem cells [iPSCs], or embryonic stem cells [ESC]).

Figure 1.

Figure 1.

Applications of human intestinal organoids.

HIO growth and maintenance require a 3D matrix. Most commonly used is Matrigel, a murine sarcoma cell-derived reconstituted basement membrane extract that transitions from liquid to gel around 37°C. Matrigel exhibits high lot-lot compositional and structural variability, and it is composed of >1000 matrix proteins, proteoglycans, and growth factors18. The major components found in Matrigel are laminin, collagen IV, heparan sulfate proteoglycans, and nidogen. The matrix elastic modulus values depend on the total protein concentration ranging from 9.1 Pa to 288 Pa for 3 to 19 mg/mL, respectively19. Although organoid structures thrive and develop in Matrigel, concerns over its tumor-derived nature, potential immunogenicity, and lot-to-lot variability and composition limit the clinical translatability of this material. Alternatives to Matrigel have been actively researched for the past several years2022, however, Matrigel remains as the gold standard for growing HIOs.

Efforts to translate HIO therapies from the lab into the clinic can be grouped into two broad areas, one focused on synthetic hydrogel matrices sustaining in vitro expansion and differentiation, and the other one centered on transplantable grafts or delivering the organoids in vivo. In this review, we discuss in vitro and in vivo synthetic hydrogel applications for HIOs. The first section includes an overview of the biology in human intestinal organoids from PSC and ASC. Section 2 includes different design considerations when engineering a synthetic matrix. Section 3 discusses different synthetic hydrogels used for intestinal organoid culture and differentiation in vitro. In the last section, current tissue engineering approaches for intestinal regeneration are summarized.

1. Human intestinal organoid culture and growth

Human intestinal organoids can be derived from pluripotent stem cells or resident tissue adult stem cells. In this section we discuss in vitro differentiation methods for both cell types, compare among different cell sources, and discuss cell-ECM interactions relevant to intestinal organoids.

1.1. Human pluripotent stem cell-derived intestinal organoids

For PSC-derived intestinal organoids, iPSC or ESC lines can be used. Alternatively, patient-specific somatic cells23 (e.g., skin fibroblasts or blood mononuclear cells) can be reprogrammed to iPSCs. Various differentiation protocols to mature HIOs from iPSCs have been reported12,2428, but most of them converge on three stages (Fig. 2). Stage 1 encompasses iPSCs expansion and definitive endoderm (DE) differentiation on planar culture. Stage 2 involves hindgut specification, where the resulting DE monolayer self-assembles into 3D structures that spontaneously bud off from the monolayer into the culture media; these structures are referred to as intestinal spheroids. In Stage 3, the spheroids are collected and embedded within Matrigel or an alternative 3D matrix and cultured to mature into HIOs after ~28 days.

Figure 2.

Figure 2.

Human pluripotent stem cell (PSC)-derived intestinal organoid differentiation process.

Differentiation is driven by growth factor cocktails introduced to the culture in a stepwise manner to mimic the early intestinal development in vitro29. The most common molecule used during this stage is activin-a to drive definitive endoderm. WNT3A or CHIR99201 in combination with fibroblast growth factor 4 (FGF-4) are used to direct the hindgut and intestinal specification process12. To promote intestinal maturation in 3D, a combination of epithelial growth factor (EGF), R-spondin 1 and Noggin are used17. We refer the reader to this excellent review29 highlighting specific signaling pathways activated by the growth factors and small molecules used during the in vitro HIO differentiation and how this process relates to embryonic patterning.

After 28 days of in vitro culture, HIOs form luminized multicellular structures with columnar intestinal epithelium which self-assembles into crypt-like domains and is surrounded by a mesenchymal cell layer30. Although specialized intestinal epithelial cells are present in the HIO structures, including Paneth cells, goblet cells, enterocytes and enteroendocrine cells, various studies have revealed that these HIOs most closely resemble immature fetal tissue12,3133. Importantly, HIO maturity is observed after in vivo transplantation with longer microvilli and Paneth cell localization at the bottom of the intestinal crypt-like domains, resembling adult tissue architecture30,31. Different approaches have been studied to increase tissue maturity including in vivo transplantation under mechanical manipulation34 and evaluating new intestinal niche growth factors35 in vitro. Other groups have explored co-culturing with endothelial36 and neural37 cells to achieve a mature HIO model. Host-microbe interactions have also been studied as an approach to achieve tissue complexity38,39. Furthermore, Helmrath and colleagues40 reported an approach to obtain HIOs structures with human immune cells exhibiting GALT-like structures and M cells after in vivo transplantation.

1.2. Human adult stem cell- (ASC) or patient-derived intestinal organoids

For patient-derived organoids (PDOs), HIOs can be established from small intestine or colon tissue biopsies obtained through endoscopic procedures, or from organoid biobanks at commercial or academic institutions41. PDOs have been shown to maintain tissue donor identity, including regional intestinal tissue characteristics42, donor’s age43, and donor’s sex44. Additionally, the donor’s gastrointestinal disease state is also maintained, thus different disease organoids models can be developed1,16,45.

Depending on the state of the initial tissue source (e.g., fresh biopsy, established PDO lines), either freshly isolated intestinal crypt fragments or single cells are embedded with a 3D basement membrane (e.g., Matrigel). These cells will then form epithelial cyst-like structures termed enteroids which can develop intestinal crypt-like structures under appropriate culture conditions. Sato et al17 demonstrated that LGR5+ crypt stem cells are imperative for the self-renewal capacity of the formed epithelial structures in vitro. A specific combination of growth factors has been optimized to resemble intestinal stem cell niche conditions in vitro and drive the differentiation of epithelial intestinal cysts to villus-crypt like containing structures. Among these are R-spondin, EGF, and Noggin which are associated with WNT signaling activation, intestinal proliferation, and intestinal crypt expansion, respectively46. However, to maintain undifferentiated expansion of epithelial cyst structures, the maintenance/expansion culture media often contains WNT3A and R-spondin growth factors42,47. The addition of the small molecules A83–01 and SB202190 to the culture media significantly improves long term expansion of these intestinal structures46. Other groups have studied alternative growth factors to stimulate self-renewal and in vitro expansion of patient-derived intestinal organoids including the use of insulin growth factor 1 and fibroblast growth factor 2 (IGF-1 and FGF-2)48. Intestinal epithelial cell types are found on intestinal epithelial organoids containing crypt-like structures including Paneth cells, goblet cells, enterocytes and enteroendocrine cells, while the cellular composition of epithelial organoids with cyst morphology is mostly intestinal stem cells and transit amplifying cells47,48. PDOs do not contain a mesenchyme layer surrounding the structure4,17.

1.3. PSC- vs ASC-derived human intestinal organoids

The use of PSC- or ASC-derived human intestinal organoid ultimately depends on the intended application or scientific question of interest. PSC-derived organoids exhibit an immature fetal-like state and rely on in vivo transplantation for full maturation. However, they present a valuable tool to study intestinal development and gastrointestinal disease mechanisms. ASC-derived HIOs are composed primarily of epithelial structures isolated from adult intestinal crypts, while PSC-derived HIOs contain mesenchymal layer, and can be engineered to present immune, neural, and endothelial structures. Patient-derived organoids maintain donor identity, making these excellent in vitro models for drug testing and personalized medicine. HIOs can also be derived using induced pluripotent stem cells derived from human donors, however, this process is laborious and costly when compared to biopsy-derived intestinal tissue. Nevertheless, iPSC-derived HIOs present an alternative for patients who lack healthy intestinal tissue for available for biopsy. When considered for in vivo intestinal regenerative therapy and clinical translation, both iPSC- and ASC-derived are promising candidates, however, manufacturing practices, time and costs, autologous versus allogeneic tissue source, and availability of tissues should be considered49.

1.4. HIO cell-matrix interactions

The extracellular matrix (ECM) is a network of structural proteins, such as laminin, collagen and fibronectin, and proteoglycans that serves as a reservoir of growth factors and cytokines50. The protein composition of the ECM varies from tissue to tissue, and it influences cell behaviors through mechanical and biochemical signaling51,52. The main mechanism for cells to interact with their surrounding ECM is through integrin transmembrane receptors which drive mechanotransduction through various signaling pathways52,53. Integrins exist as dimers, consisting of an alpha and a beta subunit and bind to specific peptide sequences found in the different ECM proteins. Additionally, cells actively remodel their ECM, a process crucial to organogenesis, wound healing, and tissue regeneration54. This process is guided by the cleavage of different ECM components through proteolysis, mainly by matrix metalloproteinases and cathepsins55,56.

For epithelial tissues, the basement membrane is a specialized type of ECM serving as physical support for the epithelial tissue layer and separating the epithelium from the mesenchymal tissue layer57. Within the intestinal stem cell niche, the basement membrane is composed of laminins, collagen IV, and fibronectin and plays essential roles regulating epithelial cell homeostasis58. Integrins regulating the stem cell niche are spatially distributed through the crypt-villi axis highlighting the importance of ECM composition dictating intestinal epithelial cell signaling59. An important mechanosensing pathway is Hippo-YAP/TAZ signaling60. Intestinal organoids have been used to study in vitro the activation of the transcription factors and their implications in tissue repair61. Additionally, the nuclear translocation of yes associated protein (YAP) has been showed be a regulator of intestinal spheroid survival8 and intestinal stem cell expansion62. We refer the reader to this detailed review63 discussing experimental strategies to study the mechanobiology of the intestinal epithelium, including intestinal organoids.

2. Rational design of synthetic hydrogel matrices for in vitro and in vivo applications

Synthetic hydrogels are water-swollen crosslinked polymer networks which can be functionalized with biological motifs/signals depending on the intended application64. Synthetic hydrogels represent a highly tunable alternative to natural matrices to support in vitro growth for 2D and 3D single and multi-cellular structures65, and as cell delivery vehicles for in vivo applications66. Synthetic hydrogel systems have been also used as in vitro platforms to study key developmental and pathophysiological processes such as cell differentiation67,68, cell migration69,70, and cell attachment71,72. Common synthetic polymers that have been used in the tissue engineering field are listed in Table 1. Notably, poly(ethylene glycol) (PEG)-based hydrogels represent the most widely used system. Engineered hydrogel matrices offer the advantage of tuning the biochemical and biophysical properties of the matrix. This modular approach allows for manufacturing designer matrices for particular applications (e.g., in vitro or in vivo). In this section we will discuss the different design considerations for synthetic hydrogel systems, with a special focus on PEG-based hydrogels.

Table 1.

Common synthetic hydrogel polymer backbones in tissue engineering applications7377

Polymer Abbreviation
Poly(ethylene glycol) PEG
Poly(acrylamide) PAAm
Poly(vinyl alcohol) PVA
Poly(acrylic acid) PAA
poly (lactic acid) PLA
poly(lactic-co-glycolic acid) PLGA
poly(caprolactone) PCL

2.1. Crosslinking chemistries for synthetic hydrogels

An important consideration when engineering synthetic hydrogels is the crosslinking reaction for the particular hydrogel system. Hydrogels, natural or synthetic, can be prepared using one or several crosslinking mechanisms involving physical, covalent, or ionic crosslinks73. For synthetic hydrogels, covalent crosslinking generally offers a higher control over the mechanical properties of the matrix when compared to other crosslinking mechanisms78. A critical requirement is that the crosslinking reaction must be cytocompatible and without toxicities.

One attractive covalent crosslinking reaction for biomedical hydrogels is the Michael-type addition reaction (Fig. 3, i). This type of ‘click’ chemistry involves a Michael donor, typically a thiol group, reacting with a Michael acceptor, to produce a Michael adduct by creating a carbon-carbon bond at the acceptor’s β-carbon78. A commonly used reactive end group in PEG hydrogels is maleimide79, which acts a Michael acceptor, and dithiol crosslinker molecules as the Michael donor. This reaction involves neutrophilic attack of the reactive thiolate ion on the crosslinker to the double C-C bond on the maleimide group forming a thioether covalent bond80. This reaction occurs at physiological pH and temperature making it widely used conjugation for in vitro and in vivo applications. A common strategy is to react macromer units with crosslinker molecules to yield a crosslinked network via step-growth polymerization78,81.

Figure 3.

Figure 3.

Common crosslinking reactions for hydrogels in tissue engineering applications (i) Michael type addition reaction (adapted from Phelps et al)79, (ii) photo-activated thiol-ene radical mediated step-growth reaction (adapted from Lin et al)83, and (iii) strain-promoted azide-alkyne cycloaddition (SPAAC) (adapted from Agard et al86 and Macdougall et al90).

Another common type of covalent crosslinking reaction is photo-activated thiol-ene radical mediated step-growth reaction such as PEG-norbornene82 (Fig. 3, ii). This type of click reaction occurs between the norbornene groups and a dithiol crosslinker molecule in the presence of a photoinitiator (e.g., lithium phenyl-2,4,6-trimehtylbenzoylphosphinate, LAP). After exposure to ultraviolet (UV) light, radicals from the photoinitiator deprotonate the sulfhydryl group on the crosslinker forming an activated thyl radical. The thyl radical attacks the C-C double bond on the norbornene group forming a thioether covalent bond. As a result, a carbon center radical is formed on the norborne group which regenerates another thyl radical. The reaction will terminate once the limited moiety is depleted (either thiol or norbornene)82,83. This step-growth reaction provides a higher spatiotemporal control of the crosslinking reaction compared to radical-initiated chain growth polymerization as seen in PEG diacrylate hydrogels (PEGDA).

Finally, another “click” chemistry crosslinking reaction used in the tissue engineering field is strain-promoted azide alkyne cycloaddition known as SPAAC (Fig. 3, iii). In this reaction, a covalent bond is formed between a crosslinking molecule containing an azide and the macromer conjugated with a cyclooctyne containing-functional group84. A commonly used PEG macromer to conduct this reaction is PEG-dibenzocycloocytne (PEG-DBCO)85. This covalent reaction occurs as a [3+2] cycloaddition of azides and cyclooctyne under physiological conditions in the absence of a catalyzing agent86. A specific subset of hydrogels formed through SPAAC crosslinking are used in photodegradation strategies, conducted after UV exposure of nitrobenzyl ether moiety87 introduced into the covalent crosslinks or using an allyl sulfide bis(azide) crosslinker in the presence of a photoinitiator such as LAP88.

For in-depth discussion of other functional groups involved in Michael-type addition reactions, step-growth reactions, SPAAC reactions and other types of crosslinking reactions, the reader is referred elsewhere73,78,88,89.

2.2. Mechanical properties of synthetic hydrogels

Single cells and multicellular structures respond to diverse cues provided by their microenvironment. An important property of the microenvironment is matrix stiffness (more generally, viscoelasticity). The stiffness of biological extracellular matrices (ECM) is controlled by the composition, architecture, and crosslinking of ECM proteins present in the specific tissue. Matrix stiffness controls how cells migrate and regulate signaling pathways important in tissue repair, proliferation, and survival50.

For hydrogel systems, the bulk mechanical properties are often measured by rheology91 and described by the complex modulus consisting of the storage (elastic) modulus (G’) and loss (viscous) modulus (G”). Alternatively, the elastic or Young’s modulus (E) can be measured using other testing modalities such as atomic force microscopy79,92 and microrheology93,94. For synthetic hydrogels, the mechanical properties are heavily determined by the crosslink density, often described as the mesh size defined as the distance between crosslinks95. For a synthetic hydrogel exhibiting rubber-like elastic behavior, the mathematical relationship between mesh size (ξ) and storage modulus (G’) is provided by the following equation96,97, where G’ is the storage modulus, A is Avogadro’s number, R is the gas constant, and T is temperature. For synthetic hydrogels mesh size is the nanometer range97.

ξ=(GART)1/3

At the hydrogel synthesis level, different approaches are available to change the mechanical properties of the resulting crosslinked gel (Fig. 4). For hydrogels based on the crosslinking of pre-synthesized macromer (e.g., multi-armed or ‘star’ building blocks) units, these include (1) polymer macromer amount in solution (polymer density, w/v%), (2) macromer size, and (3) the number of reactive arms in the macromer (e.g., 4-arm or 8-arm). The higher the polymer density, the stiffer the matrix. Conversely, the smaller the macromer, the stiffer the matrix will be. Finally, a higher number of arms on the macromer will result in a stiffer matrix by increasing the crosslink density. It is important to note that these increase the stiffness of the matrix by decreasing the mesh size of the network. Changes in mesh size can impact other properties of the matrix, such as the diffusion of molecules through the network.

Figure 4.

Figure 4.

Methods to control the mechanical properties of synthetic hydrogel by tuning: (i) the polymer density, (ii) macromer size, and (iii) number of reactive arms.

Another factor related to the mechanical properties is how the hydrogel responds to applied stress. Most PEG hydrogels are considered to exhibit elastic behavior, observed when there is almost instantaneous and reversible deformation when stress is applied. In contrast, viscoelastic materials exhibit time-dependent stress-strain responses22,96,98. The relationship between elastic modulus (E), stress (σ) and strain (ε) for linearly-elastic material and viscoelastic material can be described with the following equations99. For a linearly elastic material, there is a linear relationship to stress described below.

σ=Eε

On the other hand, for viscoelastic materials the modulus values depends on time and is reflected by this equation.

σ=E(t)ε

Two common viscoelastic behaviors are stress relaxation and stress stiffening. During stress relaxation, the stress on the material decreases over time when exposed to a constant strain100. In the case of stress stiffening, the material will become stiffer when exposed to a constant stress greater than the critical stress point for the particular material22. Physiological ECMs typically exhibit viscoelastic behavior, thus efforts to modify PEG hydrogels or engineer viscoelastic materials have been investigated100,101.

2.3. Matrix degradability for synthetic hydrogels

The degradability of the matrix is another important design consideration when engineering a synthetic matrix. Matrix remodeling through matrix metalloproteinases (MMP) and cathepsins is essential for cell survival, proliferation, growth, and differentiation102. Natural ECMs are mostly degraded by cell-secreted proteases, targeting specific amino acid sequences present on the ECM proteins such as collagen, laminin, fibronectin, and vitronectin103. For synthetic hydrogels, the degree of degradability can be controlled by enzymatic digestion, hydrolytic degradation, or photoreactivity. For thiol-based crosslinking of synthetic hydrogels (e.g., PEG-maleimide and PEG-norbornene), a chemically stable crosslinker consisting of short di-thiol molecules is used such as dithiothreitol (DTT) or PEG-dithiol (PEGDT) to generate non-degradable networks. To target protease degradability, a short peptide crosslinker can be designed to the specific protease secretome of the desired cell or tissue of interest. A list of commonly used protease-degradable crosslinkers used in PEG hydrogels is included in Table 2. Other approaches to degrade synthetic hydrogel matrix include photocleavable crosslinkers104,105 and crosslinkers or macromers incorporating groups susceptible to hydrolytic degradation106,107. Controlling the degradation reaction kinetics and studying the degradation by-products are important factors to consider, especially when designing for in vivo applications. The rate at which the matrix degrades not only depends on the type of crosslinker used, but also on the density of crosslinks in the matrix. For example, a hydrogel with higher polymer density or higher number of arms will take longer to degrade than a hydrogel with less polymer and fewer arms, respectively. The degradation rate for degradable hydrogels can vary between hours to weeks depending on the specific design considerations107109. Combining different degradation mechanisms in the same matrix may provide additional control over the degradation rate.

Table 2.

Degradable peptide crosslinkers used in PEG hydrogels

Protease sequence Degradation mechanism References
GCRDGPQGIWGQDRCG MMP cleavable 8,110,111
GCRDVPMSMRGGDRCG MMP cleavable 112,113
GCRDIPESLRAGDRCG MMP cleavable 67
GPQGIAGQ MMP cleavable 69,111
LPRTG Sortase 111,114
GGLGPAGGK Collagenase 115
AAAAAAAAAK Elastase 115
AAPVR Elastase 115

MMP – Matrix metalloproteinases

2.4. Functionalizing synthetic matrices with bioactive ligands

A key design parameter is functionalizing the synthetic hydrogel to present bioactive signaling molecules. Cells interact with the ECM mainly through integrins, transmembrane proteins dictating essential cellular processes through cell signaling pathways52,53. Synthetic polymers lack biological motifs that interact with cells, thus including short peptide sequences to promote integrin-mediated cell adhesion is a strategy to overcome this challenge73,116,117. These short peptide motifs mimic sites in ECM proteins that are recognized by integrin and other adhesion receptors. The most commonly used cell-adhesive peptide contains the arginine-glycine-aspartic acid (RGD) motif found in fibronectin and other ECM proteins116,118. We provide a list of commonly used bioadhesive peptides on Table 3. While most naturally derived matrices contain bioactive signals, a synthetic hydrogel system offers the opportunity to evaluate the effect of a particular peptide sequence or density, or to explore the synergy between a combination of peptides in the same matrix. It is recommended to design the matrix in accordance to the integrin profile for the targeted application or cell type. In addition to integrins, cells communicate with neighboring cells through different membrane receptors such as cadherins119. The hydrogel can be designed to incorporate peptide sequences targeting these receptors to activate cell-cell mediated adhesion120,121.

Table 3.

Bioactive peptides with integrin-binding sequences used in PEG hydrogels

Peptide Name Peptide sequence ECM mimic References
RGD GRGDSPC Fibronectin 8,62,118,126
GFOGER GYGGGP(GPP)5GFOGER(GPP)5GPC Collagen- type I 111,127,128
IKVAV CGGAASIKVAVSADR Laminin α1 8
AG73 CGGRKRLQVQLSIRT Laminin α1 129
RKG GKKQRFRHRNRKG Vitronectin 130
YIGSR CGGEGYGEGYIGSR Laminin β1 131
A5G81 AGQWHRVSVRWG Laminin 132
A5G84 TWSQKALHHRVP Laminin 133
PHSRN PHSRN Fibronectin 111,118

Other biological signals of interest to incorporate in synthetic hydrogels include growth factors and cytokines (Fig. 5). In naturally occurring ECMs, growth factor sequestration and presentation are important functions of the matrix controlling important cellular processes. The matrix can be engineered to present matrix bound growth factors112,122,123, but also can be engineered to incorporate growth factor binding domains into the synthetic matrix124,125.

Figure 5.

Figure 5.

PEG hydrogel presenting bioactive ligands, growth factors, and growth factor-binding sequences.

2.5. Biological tissue-derived hydrogels

In addition to synthetic matrices, hydrogels engineered from naturally derived materials are also used in tissue engineering applications. These matrices are derived from purified naturally occurring ECM proteins (collagen, fibrin, laminin, gelatin, elastin) or polysaccharides (alginate, hyaluronic acid)134. Hydrogels derived from decellularized tissue are another example of naturally derived materials. In this case, a particular tissue or organ of interest undergoes a series of decellularization steps to remove the cellular contents and isolate the native ECM and structure for the particular tissue or organ135. An advantage to these natural material-based hydrogels is that they contain innate biochemical and mechanical properties, including integrin adhesion sites and protease degradable sequences65. Despite these advantages, several limitations hinder their use for fundamental studies, drug screening, and clinical application. These materials exhibit lot-to-lot compositional and structural variability, limited processability and manufacturing, and potential for contaminants and pathogen transfer, especially for animal tissue-derived matrices18,127. For these reasons, synthetic hydrogels offer promising alternatives as in vitro culture platforms and in vivo delivery vehicles.

3. Engineered matrices for HIO culture

HIOs offer a disruptive tool to study cell development, disease progression, and identify drug targets. Engineering a synthetic matrix to support in vitro human intestinal organoid development is a strategy to overcome dependence on Matrigel, by increasing reproducibility in scale-up, manufacturing, and screening. Table 4 summarizes the advancements in synthetic and natural derived hydrogels used for human intestinal organoid culture.

Table 4.

Synthetic and natural derived hydrogels for human intestinal organoid culture

Engineered Matrix Polymer content (stiffness) Ligands /concentration Degradation mechanism Cell source Pros and cons Refs
Synthetic hydrogels
8arm PEG-vinyl sulfone + 8arm PEG-acrylate 13.33% w/v PEG precursor (G’=1.3kPa to 190Pa) RGD [1 mM] and laminin-111 [0.1 mg/mL] Hydrolytic degradation Mouse ISC Pros: Tunable matrix properties, low batch to batch variability, user designed degradation mechanisms
Cons: Needs adhesive ligand functionalization
62
LDTM 8 arm PEG-vinyl sulfone LDTM 4 arm PEG-acrylate 2.5% w/v (shear modulus =1kPa) 3% w/v (shear modulus=400Pa to 100Pa) RGD [1 mM] RGD [1 mM] MMP degradation Hydrolytic degradation Mouse ISC Human intestinal PDO 129
4 arm PEG-maleimide 4% w/v (G’=100Pa) RGD [2 mM] MMP degradation Human PSC-HIOs 8
8 arm PEG-vinyl sulfone 5% w/v (G’=926Pa) GFOGER [1.5 mM] ECM binding peptides MMP degradation Human intestinal PDO 111
Natural derived hydrogels
HELP matrix (G’=1kPa) RGD [1 mM] hyaluronic acid elastic like protein Enzymatic degradation Human intestinal PDO Pros: Control over protein sequences dictating the hydrogel mechanical and chemical properties
Cons: Complex ELP induction and purification process from E. coli cultures
131
Alginate 1% w/v (G’=100Pa) N/A Non degradable Human PSC-HIOs Pros: naturally found material, no further chemical modifications needed, low cost
Cons: lot to lot variability, limited long term stability due to ionic crosslinking
132
Porcine intestine derived ECM 2 mg/mL (G’=40–50Pa for 5 mg/mL) Collagen proteoglycans glycoproteins Enzymatic degradation Mouse ISC, PSC-HIOs Pros: ECM protein composition matched to the tissue of interest
Cons: donor to donor variability, multi-step preparation process
133
Type I collagen 3 mg/mL (G’ not reported) Collagen Collagenase degradation Mouse ISC Pros: innate bioactive adhesion sites, no further modifications needed
Cons: lot to lot variability, structural variability
170

LDTM – low defect thiol Michael addition, MMP-matrix metalloproteinases, G’-storage modulus, PDO- patient derived organoid, PSC-HIO –pluripotent stem cell human intestinal organoid, ECM – extracellular matrix.

3.1. PEG hydrogels

Progress in engineering PEG hydrogels for intestinal organoid culture has focused on identifying matrix mechanical and chemical properties that support viability and development of murine, human patient-derived, or PSC-derived intestinal organoids post-encapsulation. The first group to demonstrate that mouse-derived intestinal stem cell (ISC) colonies can grow within engineered PEG hydrogels was Gjorevski et al62. They determined the optimal matrix stiffness (1.3 kPa) supporting ISC survival and expansion when the ISCs were embedded in an 8-arm PEG hydrogel functionalized with RGD peptide. However, when evaluating the organoid differentiation and budding formation, the authors observed a softening matrix (G’ = 190 Pa) in the presence of RGD and laminin-111 was needed for differentiation. The matrix softening, driven by hydrolysis, was achieved by combining 8-arm PEG conjugated with vinyl sulfone (PEG-8VS) and 8-arm PEG conjugated with acrylate (PEG-8Acr) to act as a degradable macromer. Hushka et al85 observed softening the matrix during murine ISC colony formation was conducive to longer crypts, suggesting higher levels of differentiation. They designed a 4-arm PEG functionalized with dibenzocyclooctyne (PEG-4DBCO) crosslinked with allyl sulfide bis(azide) molecule. This strain-promoted azide alkyne cycloaddition (SPAAC) crosslinking reaction results in a covalent bond between the cyclooctynes and the azide group. In the presence of soluble glutathione and photoinitiator LAP, after exposure to 365 nm light, the crosslinking bis(azide) containing molecules will form a thiyl radical species that crosslinks in an user-controlled time dependent manner. They observed the highest ISC colony viability with a 5 wt% PEG-4DBCO hydrogel (G’ = 1.3 kPa) functionalized with 0.8 mM RGD and murine full length laminin protein entrapped in the matrix.

For primary human intestinal enteroids, Hernandez-Gordillo et al111 identified a synthetic hydrogel composition conducive to the highest enteroid formation efficiency using a 8-arm vinyl sulfone-terminated PEG macromer (PEG-8VS, 20 kDa, 926 Pa) functionalized with a collagen I-mimicking adhesive peptide (GFOGER) at 1.5 mM and crosslinked with an MMP-degradable peptide. In this work, the authors also incorporated peptides with affinity to ECM molecules to promote synthetic matrix interaction with the expected secreted ECM from the encapsulated cells. Rezakhani et al126 engineered a low-defect thiol-Michael addition (LDTM) PEG hydrogel to grow human patient-derived organoids. The synthetic matrix formulation that supported colony formation and stemness of human intestinal stem cells was an 8-arm PEG-VS hydrogel (2.5% w/v, shear modulus = 1 kPa) containing 1 mM RGD peptide with MMP degradable crosslinker. Additionally, they observed that a hydrolytically degradable hydrogel which softens overtime prepared with 3% (w/v) 4-arm PEG acrylate macromer presenting 1 mM RGD yielded structures of similar organoid phenotype when compared to Matrigel. The hydrolytic degradation resulted in a change in shear modulus of the synthetic hydrogel from 400 Pa to 100 Pa over 4 days. These LDTM hydrogels were synthesized by crosslinking PEG macromers conjugated with bifunctional peptides to unconjugated PEG-VS macromers to avoid potential intramolecular crosslinking between functional groups. Interestingly, there are discrepancies among adhesive ligand requirements (e.g., GFOGER vs. RGD) driving human patient-derived intestinal organoid growth and maturation. This effect may may be due to cell line variability as it known PDOs retain the donor’s identity4244. When compared to the mouse intestinal organoids studies discussed in this section, the patient-derived organoids exhibited a cyst-like morphology instead of crypt-like containing structures as observed with the murine organoids. Differences in cell source species may contribute to this observation136.

To support the development of PSC-derived HIOs, Cruz-Acuña et al8 developed a 4-arm PEG-maleimide terminated (PEG-4MAL) hydrogel system. This group demonstrated that iPSC- and ESC-derived HIOs survive and undergo differentiation from spheroids to mature HIOs for 28 days in the synthetic matrix. The optimal synthetic hydrogel formulation promoting growth was 4% PEG-4MAL (20 kDa) functionalized with 2 mM RGD peptide and GPQW protease degradable crosslinker. This study showed that iPSC-derived HIO survival depends on adhesion to RGD peptide compared to other cell adhesive peptides (laminin derived AG73 and IKVAV, and collagen I-derived GFOGER), and mechanical properties of G’ = 100 Pa. Reduced viability was observed in the organoids grown at the same ligand density for AG73, IKVAV and GFOGER, when compared to RGD, showing adhesion peptide type is an important matrix design factor. Additionally, protease-dependent degradability of the hydrogel matrix was required for survival of these structures. The HIOs grown on this matrix demonstrated organoid growth kinetics, lumen formation, epithelial polarization, and expression of differentiation markers equivalent to Matrigel grown organoids.

3.2. Other engineered hydrogels for HIO culture

In addition to PEG gels, other engineered matrices that support human patient-derived and iPSC-derived HIOs have been reported. Hunt et al137 engineered a hyaluronan-elastin like protein (ELP) matrix to grow human patient-derived intestinal organoids. The recombinant ELP molecule is covalently tethered to hyaluronic acid to generate the hydrogel matrix. The recombinant ELP contains RGD integrin-binding motifs and a hydrazine moiety which reacts with benzaldehyde group incorporated in the HA protein. These investigators observed that enteroids with cyst formation and polarized epithelium develop from single cells after 3 days in culture. After treating the cultures with organoid differentiation media, they observed a change in phenotype and differentiation gene expression at similar levels to Matrigel. They determined that HA was an important signaling component in tandem with RGD to promote enteroid formation. Additionally, a stiff matrix (G’ = 1 kPa) resulted in larger spheroids when compared to a softer matrix (G’ = 400 Pa).

Capeling et al138 showed that unmodified food grade alginate supports the growth and development of iPSC-derived HIOs. This polysaccharide hydrogel is crosslinked through ionic interactions when exposed to calcium chloride. In this work, they varied the weight percent of alginate in solution to study the effects of stiffness in the viability of immature HIOs. They found that a stiffness of G’ = 100 Pa, similarly to the PEG-4MAL hydrogel8, supports high spheroid viability and HIO development. The authors concluded that under the lack of biochemical signals in the matrix, iPSC-derived HIOs can deposit their own ECM niche thus only relied on mechanical support to undergo differentiation when grown in alginate hydrogels.

Another approach to engineer a growth matrix for intestinal organoids is using gastrointestinal (GI) tissue derived hydrogels139. In this work, Kim et al described a decellularization protocol to preserve major ECM components from porcine intestinal tissue including glycosaminoglycans (GAGs). After a proteomic analysis of the obtained porcine intestinal ECM hydrogel, they observed collagen to be the principal protein component followed by proteoglycans and glycoproteins. The group identified a 2 mg/mL ECM hydrogel to support mouse ISC-derived intestinal colony formation and promote stemness. They reported the storage modulus of the tissue ECM hydrogel corresponding to 5 mg/mL ECM concentration varied between 40 Pa to 50 Pa depending on the decellularization protocol used. They also reported that their matrix supported the expression of differentiation markers of hPSC-derived intestinal organoids.

Groups have also evaluated the growth of intestinal organoids under viscoelastic hydrogel conditions. Elosegui-Artola et al140 observed that viscoelastic behavior in alginate-Matrigel gels led to increased growth and symmetry breaking of murine intestinal organoids when compared to intestinal organoids grown in elastic matrices. Additionally, they reported rapid organoid growth and crypt like structure formation in stiffer viscoelastic matrices (G’ = 1500 Pa, relaxation time (τ) = 5 s) when compared to softer viscoelastic matrices (G’ = 500 Pa, τ = 5 s). Similarly, Chrisnandy et al141 reported a synthetic hybrid PEG hydrogel formulation containing both physical and chemical crosslinks exhibiting stress relaxation properties with storage modulus values ranging from 300 Pa to 1000 Pa (τ = 24 s). When compared to only covalent crosslinked gels, the mouse intestinal organoids grown in hybrid PEG formulation showed higher gene expression of symmetry-breaking markers and YAP1 target genes.

3.3. Discussion

When evaluating different engineered matrix approaches for HIO growth and culture, one of the differentiating factors among mouse ISC, patient-derived, and PSC-derived HIOs is the matrix mechanical properties. In terms of matrix stiffness, a stiffer matrix (G’ ~ 1000 Pa) supports patient-derived HIOs and mouse ISC, while for PSC-derived HIOs, a softer matrix (G’ ~ 100 Pa) was preferred. Another key matrix property discussed was matrix degradability, either through UV or hydrolytic matrix softening or protease-driven degradation, which guided HIO survival and differentiation. Cell adhesive ligand presentation is an important property with RGD, full murine laminin, and GFOGER representing the most effective. Hernandez-Gordillo et al111 discussed the importance of taking into consideration donor to donor variability when engineering synthetic matrices for patient-derived organoids since they observed changes across donors in enteroid size and growth within the engineered formulations.

For PSC-derived HIOs, a main difference across engineered matrices is the need for solely providing mechanical support (alginate hydrogels) versus the combination of adhesive peptide presentation and mechanical support (PEG-4MAL hydrogels). When grown in an alginate matrix138, the investigators observed that the surrounding HIO mesenchyme did not invade the surrounding matrix, whereas in Matrigel or RGD presenting PEG-4MAL gels8, the mesenchymal cell layer infiltrated the matrix. A recent study142 showed that PSC-derived HIOs could grow in suspension culture suggesting that a 3D ECM matrix might not be necessary for growth. These authors hypothesized that in the absence of a 3D matrix, the mesenchymal layer self-organized and gave rise to a serosa mesothelial tissue layer surrounding the HIOs similar to human fetal intestine. One limitation, however, was the low spheroid to HIO maturation yield when grown in suspension when compared to Matrigel. Although recent efforts have focused on characterizing the mesenchymal cell population31,32,143,144, it remains unclear how the different culture approaches could influence the mesenchymal and epithelial cell components in the final HIO entity. Overall differences in matrix requirements (stiffness, degradation, and adhesive ligands) across cell source (PDO vs. PSC), and species (human vs. mouse) were observed in the discussed studies. Cell line variability as well as interspecies variability must be taken into account while designing an engineered matrix.

Engineer matrices can be expanded from growth substrate to other in vitro applications, such as drug screening and drug testing applications145. These also serve as a tool to study how human intestinal organoids respond to spatiotemporal manipulation. Researchers have studied how curvature drives intestinal crypt formation94, and the importance of tissue geometry in organoid patterning145. Other approaches in this space are discussed elsewhere146.

4. HIOs in regenerative medicine

Intestinal failure (IF) occurs when patients do not receive appropriate nutrient and fluid absorption required either by anatomical limitations or loss of intestinal function147. IF is a serious complication resulting from underlying GI diseases such as inflammatory bowel disease (IBD) or short bowel syndrome (SBS). Intestinal transplants are the most common treatment for severe cases, however, graft failure and low availability of transplantable organs remain significant challenges148,149. In IBD patients, encompassing Crohn’s disease and ulcerative colitis, patients present with ulcers or wounds in the lining of the intestine. Although current treatments such as tumor necrosis factor alpha (TNF-α) inhibitors reduce inflammation levels, in most cases full closure of the intestinal wounds is not achieved150. Advances in regenerative medicine utilizing HIOs, either iPSC-derived6,8,151,152 or patient-derived7,153, have showed promise in developing therapies to overcome challenging GI diseases. Two main approaches using intestinal organoids in tissue engineering have been pursued: (1) engineering a transplantable graft, and (2) repairing the injured intestine in vivo154 (Fig. 6). For intestinal organoid therapy, it is important to consider scale-up processes and regulatory aspects to increase the relevance and clinical translation49,155. In this section, we discuss current work in engineering new intestinal organoid therapies employing regenerative medicine principles.

Figure 6.

Figure 6.

Tissue engineering approaches using HIOs: (i) therapeutic delivery of HIOs in vivo, and (ii) engineering a transplantable graft in vitro using decellularized tissue or degradable polymer scaffolds.

4.1. Engineering tissue grafts with HIOs

One repair approach, termed de novo, is when a transplantable graft is engineered in vitro and then transplanted to treat the injured or diseases tissue. In past years, efforts have focused two starting materials. One is engineering a tubular polymeric degradable scaffold populated with human intestinal cells, often termed tissue-engineered small intestine (TESI)156158. Another approach centers on using decellularized intestinal tissue repopulated with intestinal cells. The main challenge with engineering a transplantable graft is achieving a full thickness intestinal graft capable of adsorption, secretion, and food bolus movement159. In addition, vascularization and nerve integration of the graft post-transplant are major hurdles that need to be overcome.

Finkbeiner et al152 compared both approaches to engineer a transplantable intestinal graft with (1) decellularized porcine small intestine and (2) a synthetic polyglycolic/poly-L-lactic acid (PGA/PLLA) hydrolytically degradable tubular construct, both seeded PSC-derived HIOs. For the first approach, they placed HIOs cut in half inside the luminal side of 3 mm × 3 mm squares of decellularized intestine and maintained them in culture for up to 4 weeks. The HIOs attached to the tissue with both epithelial and mesenchymal cells. However, after transplanting the grafts into the omentum or under the kidney capsule of immunocompromised mice, the areas with human cells were relatively small and expressing low levels of epithelial differentiation markers, suggesting poor graft survival. For the second approach, the authors observed that HIOs were capable of populating the PGA/PLLA tubular graft. After implantation in the omentum, the HIO PGA/PLLA grafts had high survival and intestinal epithelial architecture cells. However, these constructs lacked neuronal function important for complete integration. Another group160, compared synthetic and natural polymer materials to engineer a TESI construct using mouse ISC organoids. The scaffolds they used were polyglycolic/poly-L-lactic acid (PGA/PLLA), polycaprolactone (PCL), and commercially available collagen based wound dressing material (CollaTape) coated with PLLA. All scaffolds supported in vitro organoid growth and in vivo organoid maturation after transplantation to the peritoneal cavity of immunodeficient rats. They observed PGA/PLLA and PCL scaffolds generated neomucosa with a maintained central lumen and comparable intestinal tissue architecture to the native intestine.

Other work has focused on repopulating segments of decellularized intestine maintained under perfusion in vitro before transplantation. Meran et al7 engineered human autologous jejunal mucosal grafts using decellularized human small intestine and colon segments, which were reseeded with intestinal enteroids from pediatric patients. After reseeding with these patient-derived organoids, tissue-engineered constructs were maintained in a combination of static and perfusion culture for 7 days before transplantation either under the kidney capsule or subcutaneously in immunodeficient mice. After 1-week post-implantation, the investigators observed initial signs of vascularization and presence of epithelial cells. The researchers proposed that grafts from cadaveric donors can be reseeded with autologous cell materials to overcome current challenges in the field. Kitano et al6 used iPSC-derived midgut spheroids, resulting from the initial stages of HIO differentiation, to engineer a humanized intestinal graft with a segment of decellularized rat intestine. After seeding the spheroids inside the lumen of the decellularized rat intestine, the intestinal segment was maintained in a whole organ bioreactor with media perfusion. They observed formation of a continuous epithelial monolayer and a mesenchyme layer under the regenerated epithelium in vitro. They transplanted this bioengineered tissue construct in a heterotopic position anastomosed to the carotid artery or jugular vein in immunocompromised rats where the graft survived over a period of 4 weeks.

4.2. In vivo delivery of intestinal organoids to injury models

The second approach in intestinal organoid tissue engineering centers on delivering intestinal cells directly to the injured area in vivo. One major hurdle with this approach lies in identifying a delivery method that supports cell survival, engraftment, and retention at the site of administration49.

A proof-of-concept study demonstrating that organoids engraft into injured tissue was reported by Yui et al5. They transplanted LGR5+ mouse colonic organoids to the colon in a DSS-induced colitis murine model. To test the regenerative capacity of the intestinal organoids, they prepared a single cell suspension with ~500 organoids and delivered these via an intracolonic infusion. To prevent leakage from the single cell suspension, they glued the anal verge of each animal for at least 6 hours. The authors reported that the delivered cells engrafted to the epithelium of the injured colon as observed after 16 days post-delivery. This work motivated future studies using human intestinal organoids for tissue repair.

Sugimoto et al161 generated GFP-expressing human patient-derived colonic organoids and transplanted them to the injured colon of immunodeficient mice. The injury model consisted of an EDTA chemical injury combined with an electric toothbrush to mechanically remove the epithelium of a selected area of the mouse colon. Similarly to Yui et al5, the authors glued the anal verge to prevent the contents from spilling out after delivering the organoid suspension in a diluted Matrigel solution. They observed that organoids successfully engrafted in the injured areas and noted that the resulting human crypts were larger than the host murine crypts. They continued monitoring the animals endoscopically for up to 6 months with positive GFP signal indicative of human engrafted cells.

Using a similar injury approach, Khalil et al153 transplanted patient-derived intestinal organoids into a small intestine segment of immunodeficient mice following denuding of the epithelium via chemical and mechanical damage. Using clamps, they were able to target only a segment of the small intestine while it was still in continuity. They delivered the organoids in the injured region and used a fibrin-based sealant inside the lumen to maintain the organoids in place. After 1-week post-transplant, they observed cells positive for human markers in the delivered regions, however, they concluded that alternative transplantation techniques need to be developed since the sealant provided insufficient support for the engraftment of the donor cells.

The first demonstration of a synthetic biomaterial for in vivo delivery and engraftment of HIOs was provided by Cruz-Acuña et al8,151. Using the same engineered hydrogel engineered for in vitro PSC-derived HIO development (PEG-4MAL), they showed effective endoscopic delivery of 10 HIOs per 50 μL hydrogel into focal mechanical injuries in the colonic mucosa of immunodeficient mice. The PEG-4MAL hydrogel crosslinking chemistry made it possible to inject a liquid hydrogel precursor into the wound that polymerized in situ. HIOs delivered with the PEG-4MAL hydrogel significantly increased the wound closure rate at the injury site, whereas HIOs delivered in saline did not improve wound healing. Remarkably, localized delivery of HIOs with PEG-4MAL hydrogel resulted in complete re-epithelialization of the intestinal wounds with human-derived tissue. This result highlights the importance of a biomaterial carrier for successful HIO therapies. This study represents a proof-of-concept study for an injectable synthetic hydrogel for therapeutic delivery and engraftment of HIOs to heal intestinal wounds.

4.3. Discussion

Collectively, these studies support the ability of either PSC-derived or patient-derived HIOs to populate decellularized tissue, polymeric biomaterial grafts, and in vivo injured tissue. As observed throughout the discussed work, cell adhesion to the material and cell engraftment to the intestinal tissue are important cellular events leading to a successful outcome of implanted grafts or in vivo hydrogel localized delivery. This process is mediated by cell-ECM receptors and other cell-cell mediated communication on the engraftment site162. Soluble biochemical signals, including growth factors and inflammatory cytokines and proteins, present in the host tissue also play a role dictating the cell engraftment process163,164.

Decellularized grafts maintained in vitro under perfusion may have an advantage over those maintained at static culture since poor graft survival was observed with the later152. For transplantable grafts, one limitation in the discovery and optimization stage is the size of the animal model since this limits the engineered tissue size and the potential of orthoptic transplantation. Cell sourcing, whether patient-derived or PSC-derived HIOs, is a major consideration for these regenerative medicine approaches. Healthy tissue availability, batch-to-batch variability, and biomanufacturing costs have to be considered. Selecting between patient-derived and PSC-derived HIOs is important for in vivo delivery approach. Whereas patient-derived HIOs could promote epithelial regeneration, the mesenchymal layer in PSC-derived HIOs potentially offers additional tissue regeneration such as smooth muscle serving as a base for epithelial healing49,153. The use of a biomaterial to retain the delivered HIOs to injured site in vivo is crucial for translating this technology to the clinic. An engineered synthetic biomaterial, such as injectable PEG hydrogels, is attractive since it has been demonstrated to be non-toxic, minimal lot-to-lot variability, and affording direct control over the mechanical and biochemical properties of the matrix. The PEG-4MAL hydrogel127,151 approach is likely limited to small wounds based on the rapid gelation kinetics of the maleimide-thiol reaction. Delivery systems must be engineered to address clinically relevant wounds covering extensive areas of intestinal tissue.

Hydrogels have also been used in other intestinal applications as drug delivery vehicles165,166, wound repair adhesives167,168, and imaging agents169. Hydrogel adhesion to the intestinal tissue, understanding degradation profile and mechanisms, and non-toxic byproducts are material properties that can be translated to engineering an in vivo intestinal organoid hydrogel vehicle.

5. Conclusion and future directions

PSC-derived and patient-derived HIOs hold great promise in the field of tissue engineering and regenerative medicine. Dependence on Matrigel for HIO growth and delivery limits clinical translatability of the organoid technology due to translational/regulatory barriers, immunogenicity, and lot-to-lot variability. Synthetic hydrogels offer significant advantages in control over the matrix parameters (e.g., mechanical properties, bioactive ligand presentation, and degradability) and have been successfully deployed to generate both patient-derived and PSC-derived HIOs. For PSC-derived HIOs, Matrigel is still used in the culture and differentiation of PSCs to spheroids in the planar culture. Adaptation of completely synthetic substrates to support the entire growth process will significantly increase the translation and manufacturing of the organoid technology. Additionally, increasing HIO culture complexity and evaluating the interplay of mesenchymal, endothelial, immune, and neural components grown within a synthetic hydrogel would provide a platform for robust in vitro HIO models. Recent advancements in tissue engineering with HIOs to treat intestinal failure focus on transplantable tissue grafts and organoid in vivo delivery. The use of a biomaterial is imperative in assessing current cell delivery challenges such as promoting localized cell engraftment. Future studies in the organoid therapy space should evaluate the immune system in the tissue-engineered constructs or in vivo cell engraftment using humanized animal models and relevant functional models Incorporation of synthetic hydrogels and HIOs into microfluidic gut-on-chip will provide additional insights to intestinal physiology and pathologies and serve as an in vitro platform for drug delivery and disease modeling.

Acknowledgments

Authors acknowledge funding from the National Institutes of Health (R01 DK128840 [AJG], R01 DK133702 [AJG], F31 DK130581 [AMR]) and the National Science Foundation Cell Manufacturing Technologies Engineering Research Center (EEC 1648035). Figures were created with BioRender.com.

Biographies

graphic file with name nihms-1952761-b0002.gif

Andrés J. García is the Executive Director of the Petit Institute for Bioengineering and Bioscience and Regents’ Professor at the Georgia Institute of Technology. Dr. García’s research program integrates innovative engineering, materials science, and cell biology concepts and technologies to create cell-instructive biomaterials for regenerative medicine and generate new knowledge in mechanobiology. This cross-disciplinary effort has resulted in new biomaterial platforms that elicit targeted cellular responses and tissue repair in various biomedical applications, innovative technologies to study and exploit cell adhesive interactions, and new mechanistic insights into the interplay of mechanics and cell biology. In addition, his research has generated intellectual property and licensing agreements with start-up and multi-national companies. He is a co-founder of 3 start-up companies (CellectCell, CorAmi Therapeutics, iTolerance). He has received several distinctions, including the NSF CAREER Award; the Young Investigator Award, the Clemson Award for Basic Science, and the Founders Award from the Society for Biomaterials; the International Award from the European Society for Biomaterials; and Georgia Tech’s Outstanding Interdisciplinary Activities Award and the Class of 1934 Distinguished Professor Award. He is an elected Fellow of Biomaterials Science and Engineering (by the International Union of Societies of Biomaterials Science and Engineering), Fellow of the American Association for the Advancement of Science, Fellow of the American Society of Mechanical Engineers, and Fellow of the American Institute for Medical and Biological Engineering. He served as President for the Society for Biomaterials in 2018–2019. He is an elected member of the National Academy of Engineering, the National Academy of Medicine, and the National Academy of Inventors.

graphic file with name nihms-1952761-b0003.gif

Adriana Mulero-Russe is currently pursuing her doctoral degree in bioengineering at the Georgia Institute of Technology where her research interests focus on engineering biomaterials, tissue engineering, and regenerative medicine. Adriana received the National Institutes of Health (NIH) Ruth L. Kirschstein Predoctoral Fellowship to support her PhD thesis work in Dr. Andrés García’s Lab. She obtained her bachelor of science degree in chemical engineering from the University of Puerto Rico at Mayagüez in 2018.

References

  • 1.Gunther C, Winner B, Neurath MF & Stappenbeck TS Organoids in gastrointestinal diseases: from experimental models to clinical translation. Gut 71, 1892–1908, doi: 10.1136/gutjnl-2021-326560 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Taelman J, Diaz M & Guiu J Human Intestinal Organoids: Promise and Challenge. Front Cell Dev Biol 10, 854740, doi: 10.3389/fcell.2022.854740 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Spence JR Taming the Wild West of Organoids, Enteroids, and Mini-Guts. Cell Mol Gastroenterol Hepatol 5, 159–160, doi: 10.1016/j.jcmgh.2017.11.003 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Hautefort I, Poletti M, Papp D & Korcsmaros T Everything You Always Wanted to Know About Organoid-Based Models (and Never Dared to Ask). Cell Mol Gastroenterol Hepatol 14, 311–331, doi: 10.1016/j.jcmgh.2022.04.012 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Yui S et al. Functional engraftment of colon epithelium expanded in vitro from a single adult Lgr5(+) stem cell. Nat Med 18, 618–623, doi: 10.1038/nm.2695 (2012). [DOI] [PubMed] [Google Scholar]
  • 6.Kitano K et al. Bioengineering of functional human induced pluripotent stem cell-derived intestinal grafts. Nat Commun 8, 765, doi: 10.1038/s41467-017-00779-y (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Meran L et al. Engineering transplantable jejunal mucosal grafts using patient-derived organoids from children with intestinal failure. Nat Med 26, 1593–1601, doi: 10.1038/s41591-020-1024-z (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Cruz-Acuna R et al. Synthetic hydrogels for human intestinal organoid generation and colonic wound repair. Nat Cell Biol 19, 1326–1335, doi: 10.1038/ncb3632 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Ganesh K et al. A rectal cancer organoid platform to study individual responses to chemoradiation. Nat Med 25, 1607–1614, doi: 10.1038/s41591-019-0584-2 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Takahashi Y et al. Drug cytotoxicity screening using human intestinal organoids propagated with extensive cost-reduction strategies. Sci Rep 13, 5407, doi: 10.1038/s41598-023-32438-2 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Yoshida S, Miwa H, Kawachi T, Kume S & Takahashi K Generation of intestinal organoids derived from human pluripotent stem cells for drug testing. Sci Rep 10, 5989, doi: 10.1038/s41598-020-63151-z (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Spence JR et al. Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro. Nature 470, 105–109, doi: 10.1038/nature09691 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Sprangers J, Zaalberg IC & Maurice MM Organoid-based modeling of intestinal development, regeneration, and repair. Cell Death Differ 28, 95–107, doi: 10.1038/s41418-020-00665-z (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Clevers H Modeling Development and Disease with Organoids. Cell 165, 1586–1597, doi: 10.1016/j.cell.2016.05.082 (2016). [DOI] [PubMed] [Google Scholar]
  • 15.Zhou J et al. Infection of bat and human intestinal organoids by SARS-CoV-2. Nat Med 26, 1077–1083, doi: 10.1038/s41591-020-0912-6 (2020). [DOI] [PubMed] [Google Scholar]
  • 16.Lau HCH, Kranenburg O, Xiao H & Yu J Organoid models of gastrointestinal cancers in basic and translational research. Nat Rev Gastroenterol Hepatol 17, 203–222, doi: 10.1038/s41575-019-0255-2 (2020). [DOI] [PubMed] [Google Scholar]
  • 17.Sato T et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 459, 262–265, doi: 10.1038/nature07935 (2009). [DOI] [PubMed] [Google Scholar]
  • 18.Hughes CS, Postovit LM & Lajoie GA Matrigel: a complex protein mixture required for optimal growth of cell culture. Proteomics 10, 1886–1890, doi: 10.1002/pmic.200900758 (2010). [DOI] [PubMed] [Google Scholar]
  • 19.Slater K, Partridge J, Nandivada H Tuning the Elastic Moduli of Corning® Matrigel® and Collagen I 3D Matrices by Varying the Protein Concentration: Application Note. https://www.corning.com/catalog/cls/documents/application-notes/CLS-AC-AN-449.pdf (2018).
  • 20.Aisenbrey EA & Murphy WL Synthetic alternatives to Matrigel. Nat Rev Mater 5, 539–551, doi: 10.1038/s41578-020-0199-8 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Kozlowski MT, Crook CJ & Ku HT Towards organoid culture without Matrigel. Commun Biol 4, 1387, doi: 10.1038/s42003-021-02910-8 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Kratochvil MJ et al. Engineered materials for organoid systems. Nat Rev Mater 4, 606–622, doi: 10.1038/s41578-019-0129-9 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Al Abbar A, Ngai SC, Nograles N, Alhaji SY & Abdullah S Induced Pluripotent Stem Cells: Reprogramming Platforms and Applications in Cell Replacement Therapy. Biores Open Access 9, 121–136, doi: 10.1089/biores.2019.0046 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.McCracken KW, Howell JC, Wells JM & Spence JR Generating human intestinal tissue from pluripotent stem cells in vitro. Nat Protoc 6, 1920–1928, doi: 10.1038/nprot.2011.410 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Takahashi Y et al. A Refined Culture System for Human Induced Pluripotent Stem Cell-Derived Intestinal Epithelial Organoids. Stem Cell Reports 10, 314–328, doi: 10.1016/j.stemcr.2017.11.004 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Tamminen K et al. Intestinal Commitment and Maturation of Human Pluripotent Stem Cells Is Independent of Exogenous FGF4 and R-spondin1. PLoS One 10, e0134551, doi: 10.1371/journal.pone.0134551 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Ogawa I et al. Suspension culture of human induced pluripotent stem cell-derived intestinal organoids using natural polysaccharides. Biomaterials 288, 121696, doi: 10.1016/j.biomaterials.2022.121696 (2022). [DOI] [PubMed] [Google Scholar]
  • 28.Capeling M et al. Generation of small intestinal organoids for experimental intestinal physiology. Methods Cell Biol 159, 143–174, doi: 10.1016/bs.mcb.2020.03.007 (2020). [DOI] [PubMed] [Google Scholar]
  • 29.Wells JM & Spence JR How to make an intestine. Development 141, 752–760, doi: 10.1242/dev.097386 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Watson CL et al. An in vivo model of human small intestine using pluripotent stem cells. Nat Med 20, 1310–1314, doi: 10.1038/nm.3737 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Finkbeiner SR et al. Transcriptome-wide Analysis Reveals Hallmarks of Human Intestine Development and Maturation In Vitro and In Vivo. Stem Cell Reports 4, 1140–1155, doi: 10.1016/j.stemcr.2015.04.010 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Singh A et al. Transplanted human intestinal organoids: a resource for modeling human intestinal development. Development 150, doi: 10.1242/dev.201416 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Yu Q et al. Charting human development using a multi-endodermal organ atlas and organoid models. Cell 184, 3281–3298 e3222, doi: 10.1016/j.cell.2021.04.028 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Poling HM et al. Mechanically induced development and maturation of human intestinal organoids in vivo. Nat Biomed Eng 2, 429–442, doi: 10.1038/s41551-018-0243-9 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kilik U et al. Maturation of human intestinal epithelium from pluripotency in vitro. bioRxiv, 2021.2009.2024.460132, doi: 10.1101/2021.09.24.460132 (2021). [DOI] [Google Scholar]
  • 36.Holloway EM et al. Differentiation of Human Intestinal Organoids with Endogenous Vascular Endothelial Cells. Dev Cell 54, 516–528 e517, doi: 10.1016/j.devcel.2020.07.023 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Workman MJ et al. Engineered human pluripotent-stem-cell-derived intestinal tissues with a functional enteric nervous system. Nat Med 23, 49–59, doi: 10.1038/nm.4233 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Holloway EM, Capeling MM & Spence JR Biologically inspired approaches to enhance human organoid complexity. Development 146, doi: 10.1242/dev.166173 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Hill DR et al. Bacterial colonization stimulates a complex physiological response in the immature human intestinal epithelium. Elife 6, doi: 10.7554/eLife.29132 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Bouffi C et al. In vivo development of immune tissue in human intestinal organoids transplanted into humanized mice. Nat Biotechnol 41, 824–831, doi: 10.1038/s41587-022-01558-x (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Perrone F & Zilbauer M Biobanking of human gut organoids for translational research. Exp Mol Med 53, 1451–1458, doi: 10.1038/s12276-021-00606-x (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Urbano PCM, Angus HCK, Gadeock S, Schultz M & Kemp RA Assessment of source material for human intestinal organoid culture for research and clinical use. BMC Res Notes 15, 35, doi: 10.1186/s13104-022-05925-4 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Lewis SK et al. DNA Methylation Analysis Validates Organoids as a Viable Model for Studying Human Intestinal Aging. Cell Mol Gastroenterol Hepatol 9, 527–541, doi: 10.1016/j.jcmgh.2019.11.013 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Beanland BT et al. Investigation of murine host sex as a biological variable in epithelial barrier function and muscle contractility in human intestinal organoids. FASEB J 36, e22613, doi: 10.1096/fj.202101740RR (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Yan HHN et al. A Comprehensive Human Gastric Cancer Organoid Biobank Captures Tumor Subtype Heterogeneity and Enables Therapeutic Screening. Cell Stem Cell 23, 882–897 e811, doi: 10.1016/j.stem.2018.09.016 (2018). [DOI] [PubMed] [Google Scholar]
  • 46.Sato T et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett’s epithelium. Gastroenterology 141, 1762–1772, doi: 10.1053/j.gastro.2011.07.050 (2011). [DOI] [PubMed] [Google Scholar]
  • 47.Pleguezuelos-Manzano C et al. Establishment and Culture of Human Intestinal Organoids Derived from Adult Stem Cells. Curr Protoc Immunol 130, e106, doi: 10.1002/cpim.106 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Fujii M et al. Human Intestinal Organoids Maintain Self-Renewal Capacity and Cellular Diversity in Niche-Inspired Culture Condition. Cell Stem Cell 23, 787–793 e786, doi: 10.1016/j.stem.2018.11.016 (2018). [DOI] [PubMed] [Google Scholar]
  • 49.Kasendra M et al. Intestinal organoids: roadmap to the clinic. Am J Physiol Gastrointest Liver Physiol 321, G1–G10, doi: 10.1152/ajpgi.00425.2020 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Hynes RO The extracellular matrix: not just pretty fibrils. Science 326, 1216–1219, doi: 10.1126/science.1176009 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Vining KH & Mooney DJ Mechanical forces direct stem cell behaviour in development and regeneration. Nat Rev Mol Cell Biol 18, 728–742, doi: 10.1038/nrm.2017.108 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Streuli CH Integrins and cell-fate determination. J Cell Sci 122, 171–177, doi: 10.1242/jcs.018945 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Garcia AJ & Boettiger D Integrin-fibronectin interactions at the cell-material interface: initial integrin binding and signaling. Biomaterials 20, 2427–2433, doi: 10.1016/s0142-9612(99)00170-2 (1999). [DOI] [PubMed] [Google Scholar]
  • 54.Diller RB & Tabor AJ The Role of the Extracellular Matrix (ECM) in Wound Healing: A Review. Biomimetics (Basel) 7, doi: 10.3390/biomimetics7030087 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Bonnans C, Chou J & Werb Z Remodelling the extracellular matrix in development and disease. Nat Rev Mol Cell Biol 15, 786–801, doi: 10.1038/nrm3904 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Lu P, Takai K, Weaver VM & Werb Z Extracellular matrix degradation and remodeling in development and disease. Cold Spring Harb Perspect Biol 3, doi: 10.1101/cshperspect.a005058 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Li AC & Thompson RP Basement membrane components. J Clin Pathol 56, 885–887, doi: 10.1136/jcp.56.12.885 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Meran L, Baulies A & Li VSW Intestinal Stem Cell Niche: The Extracellular Matrix and Cellular Components. Stem Cells Int 2017, 7970385, doi: 10.1155/2017/7970385 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Rezakhani S, Gjorevski N & Lutolf MP Extracellular matrix requirements for gastrointestinal organoid cultures. Biomaterials 276, 121020, doi: 10.1016/j.biomaterials.2021.121020 (2021). [DOI] [PubMed] [Google Scholar]
  • 60.Deng F, Wu Z, Zou F, Wang S & Wang X The Hippo-YAP/TAZ Signaling Pathway in Intestinal Self-Renewal and Regeneration After Injury. Front Cell Dev Biol 10, 894737, doi: 10.3389/fcell.2022.894737 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Yui S et al. YAP/TAZ-Dependent Reprogramming of Colonic Epithelium Links ECM Remodeling to Tissue Regeneration. Cell Stem Cell 22, 35–49 e37, doi: 10.1016/j.stem.2017.11.001 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Gjorevski N et al. Designer matrices for intestinal stem cell and organoid culture. Nature 539, 560–564, doi: 10.1038/nature20168 (2016). [DOI] [PubMed] [Google Scholar]
  • 63.Perez-Gonzalez C, Ceada G, Matejcic M & Trepat X Digesting the mechanobiology of the intestinal epithelium. Curr Opin Genet Dev 72, 82–90, doi: 10.1016/j.gde.2021.10.005 (2022). [DOI] [PubMed] [Google Scholar]
  • 64.Ahmed EM Hydrogel: Preparation, characterization, and applications: A review. J Adv Res 6, 105–121, doi: 10.1016/j.jare.2013.07.006 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Caliari SR & Burdick JA A practical guide to hydrogels for cell culture. Nat Methods 13, 405–414, doi: 10.1038/nmeth.3839 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Sivaraj D et al. Hydrogel Scaffolds to Deliver Cell Therapies for Wound Healing. Front Bioeng Biotechnol 9, 660145, doi: 10.3389/fbioe.2021.660145 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Cruz-Acuna R, Mulero-Russe A, Clark AY, Zent R & Garcia AJ Identification of matrix physicochemical properties required for renal epithelial cell tubulogenesis by using synthetic hydrogels. J Cell Sci 132, doi: 10.1242/jcs.226639 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Guvendiren M & Burdick JA Engineering synthetic hydrogel microenvironments to instruct stem cells. Curr Opin Biotechnol 24, 841–846, doi: 10.1016/j.copbio.2013.03.009 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Lutolf MP et al. Synthetic matrix metalloproteinase-sensitive hydrogels for the conduction of tissue regeneration: engineering cell-invasion characteristics. Proc Natl Acad Sci U S A 100, 5413–5418, doi: 10.1073/pnas.0737381100 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Raeber GP, Lutolf MP & Hubbell JA Molecularly engineered PEG hydrogels: a novel model system for proteolytically mediated cell migration. Biophys J 89, 1374–1388, doi: 10.1529/biophysj.104.050682 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Musah S et al. Glycosaminoglycan-binding hydrogels enable mechanical control of human pluripotent stem cell self-renewal. ACS Nano 6, 10168–10177, doi: 10.1021/nn3039148 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Yang B et al. Enhanced mechanosensing of cells in synthetic 3D matrix with controlled biophysical dynamics. Nat Commun 12, 3514, doi: 10.1038/s41467-021-23120-0 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Zhu J Bioactive modification of poly(ethylene glycol) hydrogels for tissue engineering. Biomaterials 31, 4639–4656, doi: 10.1016/j.biomaterials.2010.02.044 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Bashir S et al. Fundamental Concepts of Hydrogels: Synthesis, Properties, and Their Applications. Polymers (Basel) 12, doi: 10.3390/polym12112702 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Lee KY & Mooney DJ Hydrogels for tissue engineering. Chem Rev 101, 1869–1879, doi: 10.1021/cr000108x (2001). [DOI] [PubMed] [Google Scholar]
  • 76.Cruz-Acuña R & García AJ Engineered materials to model human intestinal development and cancer using organoids. Exp Cell Res 377, 109–114, doi: 10.1016/j.yexcr.2019.02.017 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Unal AZ & West JL Synthetic ECM: Bioactive Synthetic Hydrogels for 3D Tissue Engineering. Bioconjug Chem 31, 2253–2271, doi: 10.1021/acs.bioconjchem.0c00270 (2020). [DOI] [PubMed] [Google Scholar]
  • 78.Azagarsamy MA & Anseth KS Bioorthogonal Click Chemistry: An Indispensable Tool to Create Multifaceted Cell Culture Scaffolds. ACS Macro Lett 2, 5–9, doi: 10.1021/mz300585q (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Phelps EA et al. Maleimide cross-linked bioactive PEG hydrogel exhibits improved reaction kinetics and cross-linking for cell encapsulation and in situ delivery. Adv Mater 24, 64–70, 62, doi: 10.1002/adma.201103574 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Hermanson GT in Bioconjugate Techniques (Third Edition) (ed Hermanson Greg T.) 229–258 (Academic Press, 2013). [Google Scholar]
  • 81.Vats K et al. Nanoscale physicochemical properties of chain- and step-growth polymerized PEG hydrogels affect cell-material interactions. J Biomed Mater Res A 105, 1112–1122, doi: 10.1002/jbm.a.36007 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Fairbanks BD et al. A Versatile Synthetic Extracellular Matrix Mimic via Thiol-Norbornene Photopolymerization. Adv Mater 21, 5005–5010, doi: 10.1002/adma.200901808 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Lin CC, Ki CS & Shih H Thiol-norbornene photo-click hydrogels for tissue engineering applications. J Appl Polym Sci 132, doi: 10.1002/app.41563 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Baskin JM et al. Copper-free click chemistry for dynamic in vivo imaging. Proc Natl Acad Sci U S A 104, 16793–16797, doi: 10.1073/pnas.0707090104 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Hushka EA, Yavitt FM, Brown TE, Dempsey PJ & Anseth KS Relaxation of Extracellular Matrix Forces Directs Crypt Formation and Architecture in Intestinal Organoids. Adv Healthc Mater 9, e1901214, doi: 10.1002/adhm.201901214 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Agard NJ, Prescher JA & Bertozzi CR A strain-promoted [3 + 2] azide-alkyne cycloaddition for covalent modification of biomolecules in living systems. J Am Chem Soc 126, 15046–15047, doi: 10.1021/ja044996f (2004). [DOI] [PubMed] [Google Scholar]
  • 87.McKinnon DD, Brown TE, Kyburz KA, Kiyotake E & Anseth KS Design and characterization of a synthetically accessible, photodegradable hydrogel for user-directed formation of neural networks. Biomacromolecules 15, 2808–2816, doi: 10.1021/bm500731b (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Brown TE & Anseth KS Spatiotemporal hydrogel biomaterials for regenerative medicine. Chem Soc Rev 46, 6532–6552, doi: 10.1039/c7cs00445a (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Kim J et al. Characterization of the crosslinking kinetics of multi-arm poly(ethylene glycol) hydrogels formed via Michael-type addition. Soft Matter 12, 2076–2085, doi: 10.1039/c5sm02668g (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Macdougall LJ et al. Intracellular Crowding by Bio-Orthogonal Hydrogel Formation Induces Reversible Molecular Stasis. Adv Mater 34, e2202882, doi: 10.1002/adma.202202882 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Zuidema JM, Rivet CJ, Gilbert RJ & Morrison FA A protocol for rheological characterization of hydrogels for tissue engineering strategies. J Biomed Mater Res B Appl Biomater 102, 1063–1073, doi: 10.1002/jbm.b.33088 (2014). [DOI] [PubMed] [Google Scholar]
  • 92.Drira Z & Yadavalli VK Nanomechanical measurements of polyethylene glycol hydrogels using atomic force microscopy. J Mech Behav Biomed Mater 18, 20–28, doi: 10.1016/j.jmbbm.2012.09.015 (2013). [DOI] [PubMed] [Google Scholar]
  • 93.Pokki J, Zisi I, Schulman E, Indana D & Chaudhuri O Magnetic probe-based microrheology reveals local softening and stiffening of 3D collagen matrices by fibroblasts. Biomed Microdevices 23, 27, doi: 10.1007/s10544-021-00547-2 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Yavitt FM et al. In situ modulation of intestinal organoid epithelial curvature through photoinduced viscoelasticity directs crypt morphogenesis. Sci Adv 9, eadd5668, doi: 10.1126/sciadv.add5668 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Peppas NA, Huang Y, Torres-Lugo M, Ward JH & Zhang J Physicochemical foundations and structural design of hydrogels in medicine and biology. Annu Rev Biomed Eng 2, 9–29, doi: 10.1146/annurev.bioeng.2.1.9 (2000). [DOI] [PubMed] [Google Scholar]
  • 96.Oyen ML Mechanical characterisation of hydrogel materials. International Materials Reviews 59, 44–59, doi: 10.1179/1743280413Y.0000000022 (2014). [DOI] [Google Scholar]
  • 97.Han WM et al. Synthetic matrix enhances transplanted satellite cell engraftment in dystrophic and aged skeletal muscle with comorbid trauma. Sci Adv 4, eaar4008, doi: 10.1126/sciadv.aar4008 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Anseth KS, Bowman CN & Brannon-Peppas L Mechanical properties of hydrogels and their experimental determination. Biomaterials 17, 1647–1657, doi: 10.1016/0142-9612(96)87644-7 (1996). [DOI] [PubMed] [Google Scholar]
  • 99.Chaudhuri O, Cooper-White J, Janmey PA, Mooney DJ & Shenoy VB Effects of extracellular matrix viscoelasticity on cellular behaviour. Nature 584, 535–546, doi: 10.1038/s41586-020-2612-2 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Chaudhuri O Viscoelastic hydrogels for 3D cell culture. Biomater Sci 5, 1480–1490, doi: 10.1039/c7bm00261k (2017). [DOI] [PubMed] [Google Scholar]
  • 101.Cacopardo L, Guazzelli N, Nossa R, Mattei G & Ahluwalia A Engineering hydrogel viscoelasticity. J Mech Behav Biomed Mater 89, 162–167, doi: 10.1016/j.jmbbm.2018.09.031 (2019). [DOI] [PubMed] [Google Scholar]
  • 102.Sternlicht MD & Werb Z How matrix metalloproteinases regulate cell behavior. Annu Rev Cell Dev Biol 17, 463–516, doi: 10.1146/annurev.cellbio.17.1.463 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Lopez-Otin C & Bond JS Proteases: multifunctional enzymes in life and disease. J Biol Chem 283, 30433–30437, doi: 10.1074/jbc.R800035200 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Yan F, Chen L, Tang Q & Wang R Synthesis and characterization of a photocleavable cross-linker and its application on tunable surface modification and protein photodelivery. Bioconjug Chem 15, 1030–1036, doi: 10.1021/bc049901d (2004). [DOI] [PubMed] [Google Scholar]
  • 105.Brown TE, Marozas IA & Anseth KS Amplified Photodegradation of Cell-Laden Hydrogels via an Addition-Fragmentation Chain Transfer Reaction. Adv Mater 29, doi: 10.1002/adma.201605001 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Zustiak SP & Leach JB Hydrolytically degradable poly(ethylene glycol) hydrogel scaffolds with tunable degradation and mechanical properties. Biomacromolecules 11, 1348–1357, doi: 10.1021/bm100137q (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Hunckler MD et al. Linkage Groups within Thiol-Ene Photoclickable PEG Hydrogels Control In Vivo Stability. Adv Healthc Mater 8, e1900371, doi: 10.1002/adhm.201900371 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Madl CM, Katz LM & Heilshorn SC Tuning Bulk Hydrogel Degradation by Simultaneous Control of Proteolytic Cleavage Kinetics and Hydrogel Network Architecture. ACS Macro Lett 7, 1302–1307, doi: 10.1021/acsmacrolett.8b00664 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Martin KE et al. Hydrolytic hydrogels tune mesenchymal stem cell persistence and immunomodulation for enhanced diabetic cutaneous wound healing. Biomaterials 301, 122256, doi: 10.1016/j.biomaterials.2023.122256 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Enemchukwu NO et al. Synthetic matrices reveal contributions of ECM biophysical and biochemical properties to epithelial morphogenesis. J Cell Biol 212, 113–124, doi: 10.1083/jcb.201506055 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Hernandez-Gordillo V et al. Fully synthetic matrices for in vitro culture of primary human intestinal enteroids and endometrial organoids. Biomaterials 254, 120125, doi: 10.1016/j.biomaterials.2020.120125 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Phelps EA, Templeman KL, Thule PM & Garcia AJ Engineered VEGF-releasing PEG-MAL hydrogel for pancreatic islet vascularization. Drug Deliv Transl Res 5, 125–136, doi: 10.1007/s13346-013-0142-2 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Han WM, Mohiuddin M, Anderson SE, Garcia AJ & Jang YC Co-delivery of Wnt7a and muscle stem cells using synthetic bioadhesive hydrogel enhances murine muscle regeneration and cell migration during engraftment. Acta Biomater 94, 243–252, doi: 10.1016/j.actbio.2019.06.025 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Cambria E et al. Covalent Modification of Synthetic Hydrogels with Bioactive Proteins via Sortase-Mediated Ligation. Biomacromolecules 16, 2316–2326, doi: 10.1021/acs.biomac.5b00549 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Kloxin AM, Kloxin CJ, Bowman CN & Anseth KS Mechanical properties of cellularly responsive hydrogels and their experimental determination. Adv Mater 22, 3484–3494, doi: 10.1002/adma.200904179 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Hersel U, Dahmen C & Kessler H RGD modified polymers: biomaterials for stimulated cell adhesion and beyond. Biomaterials 24, 4385–4415, doi: 10.1016/s0142-9612(03)00343-0 (2003). [DOI] [PubMed] [Google Scholar]
  • 117.Huettner N, Dargaville TR & Forget A Discovering Cell-Adhesion Peptides in Tissue Engineering: Beyond RGD. Trends Biotechnol 36, 372–383, doi: 10.1016/j.tibtech.2018.01.008 (2018). [DOI] [PubMed] [Google Scholar]
  • 118.Bellis SL Advantages of RGD peptides for directing cell association with biomaterials. Biomaterials 32, 4205–4210, doi: 10.1016/j.biomaterials.2011.02.029 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Maitre JL & Heisenberg CP Three functions of cadherins in cell adhesion. Curr Biol 23, R626–633, doi: 10.1016/j.cub.2013.06.019 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Bian L, Guvendiren M, Mauck RL & Burdick JA Hydrogels that mimic developmentally relevant matrix and N-cadherin interactions enhance MSC chondrogenesis. Proc Natl Acad Sci U S A 110, 10117–10122, doi: 10.1073/pnas.1214100110 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Zhang Z et al. Programmable integrin and N-cadherin adhesive interactions modulate mechanosensing of mesenchymal stem cells by cofilin phosphorylation. Nat Commun 13, 6854, doi: 10.1038/s41467-022-34424-0 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Seliktar D, Zisch AH, Lutolf MP, Wrana JL & Hubbell JA MMP-2 sensitive, VEGF-bearing bioactive hydrogels for promotion of vascular healing. J Biomed Mater Res A 68, 704–716, doi: 10.1002/jbm.a.20091 (2004). [DOI] [PubMed] [Google Scholar]
  • 123.Saik JE, Gould DJ, Watkins EM, Dickinson ME & West JL Covalently immobilized platelet-derived growth factor-BB promotes angiogenesis in biomimetic poly(ethylene glycol) hydrogels. Acta Biomater 7, 133–143, doi: 10.1016/j.actbio.2010.08.018 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Martino MM, Briquez PS, Ranga A, Lutolf MP & Hubbell JA Heparin-binding domain of fibrin(ogen) binds growth factors and promotes tissue repair when incorporated within a synthetic matrix. Proc Natl Acad Sci U S A 110, 4563–4568, doi: 10.1073/pnas.1221602110 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Abune L et al. Macroporous Hydrogels for Stable Sequestration and Sustained Release of Vascular Endothelial Growth Factor and Basic Fibroblast Growth Factor Using Nucleic Acid Aptamers. ACS Biomater Sci Eng 5, 2382–2390, doi: 10.1021/acsbiomaterials.9b00423 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Rezakhani S, Gjorevski N & Lutolf M Low‐Defect Thiol‐Michael Addition Hydrogels as Matrigel Substitutes for Epithelial Organoid Derivation. Advanced Functional Materials 30, doi: 10.1002/adfm.202000761 (2020). [DOI] [Google Scholar]
  • 127.Cruz-Acuna R & Garcia AJ Synthetic hydrogels mimicking basement membrane matrices to promote cell-matrix interactions. Matrix Biol 57–58, 324–333, doi: 10.1016/j.matbio.2016.06.002 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Clark AY et al. Integrin-specific hydrogels modulate transplanted human bone marrow-derived mesenchymal stem cell survival, engraftment, and reparative activities. Nat Commun 11, 114, doi: 10.1038/s41467-019-14000-9 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Wilson RL et al. Protein-Functionalized Poly(ethylene glycol) Hydrogels as Scaffolds for Monolayer Organoid Culture. Tissue Eng Part C Methods 27, 12–23, doi: 10.1089/ten.TEC.2020.0306 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Klim JR, Li L, Wrighton PJ, Piekarczyk MS & Kiessling LL A defined glycosaminoglycan-binding substratum for human pluripotent stem cells. Nat Methods 7, 989–994, doi: 10.1038/nmeth.1532 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Su J, Satchell SC, Wertheim JA & Shah RN Poly(ethylene glycol)-crosslinked gelatin hydrogel substrates with conjugated bioactive peptides influence endothelial cell behavior. Biomaterials 201, 99–112, doi: 10.1016/j.biomaterials.2019.02.001 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Zhu Y et al. Potent laminin-inspired antioxidant regenerative dressing accelerates wound healing in diabetes. Proc Natl Acad Sci U S A 115, 6816–6821, doi: 10.1073/pnas.1804262115 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Katagiri F et al. Screening of integrin-binding peptides from the laminin alpha4 and alpha5 chain G domain peptide library. Arch Biochem Biophys 521, 32–42, doi: 10.1016/j.abb.2012.02.017 (2012). [DOI] [PubMed] [Google Scholar]
  • 134.Gomez-Florit M et al. Natural-Based Hydrogels for Tissue Engineering Applications. Molecules 25, doi: 10.3390/molecules25245858 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Saldin LT, Cramer MC, Velankar SS, White LJ & Badylak SF Extracellular matrix hydrogels from decellularized tissues: Structure and function. Acta Biomater 49, 1–15, doi: 10.1016/j.actbio.2016.11.068 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Kim J, Koo BK & Knoblich JA Human organoids: model systems for human biology and medicine. Nat Rev Mol Cell Biol 21, 571–584, doi: 10.1038/s41580-020-0259-3 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Hunt DR et al. Engineered Matrices Enable the Culture of Human Patient-Derived Intestinal Organoids. Adv Sci (Weinh) 8, 2004705, doi: 10.1002/advs.202004705 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Capeling MM et al. Nonadhesive Alginate Hydrogels Support Growth of Pluripotent Stem Cell-Derived Intestinal Organoids. Stem Cell Reports 12, 381–394, doi: 10.1016/j.stemcr.2018.12.001 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Kim S et al. Tissue extracellular matrix hydrogels as alternatives to Matrigel for culturing gastrointestinal organoids. Nat Commun 13, 1692, doi: 10.1038/s41467-022-29279-4 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Elosegui-Artola A et al. Matrix viscoelasticity controls spatiotemporal tissue organization. Nat Mater 22, 117–127, doi: 10.1038/s41563-022-01400-4 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Chrisnandy A, Blondel D, Rezakhani S, Broguiere N & Lutolf MP Synthetic dynamic hydrogels promote degradation-independent in vitro organogenesis. Nat Mater 21, 479–487, doi: 10.1038/s41563-021-01136-7 (2022). [DOI] [PubMed] [Google Scholar]
  • 142.Capeling MM et al. Suspension culture promotes serosal mesothelial development in human intestinal organoids. Cell Rep 38, 110379, doi: 10.1016/j.celrep.2022.110379 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Kedinger M et al. Intestinal epithelial-mesenchymal cell interactions. Ann N Y Acad Sci 859, 1–17, doi: 10.1111/j.1749-6632.1998.tb11107.x (1998). [DOI] [PubMed] [Google Scholar]
  • 144.Walton KD et al. Hedgehog-responsive mesenchymal clusters direct patterning and emergence of intestinal villi. Proc Natl Acad Sci U S A 109, 15817–15822, doi: 10.1073/pnas.1205669109 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Brandenberg N et al. High-throughput automated organoid culture via stem-cell aggregation in microcavity arrays. Nat Biomed Eng 4, 863–874, doi: 10.1038/s41551-020-0565-2 (2020). [DOI] [PubMed] [Google Scholar]
  • 146.Blatchley MR & Anseth KS Middle-out methods for spatiotemporal tissue engineering of organoids. Nat Rev Bioeng 1, 329–345, doi: 10.1038/s44222-023-00039-3 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Sudan D et al. A multidisciplinary approach to the treatment of intestinal failure. J Gastrointest Surg 9, 165–176; discussion 176–167, doi: 10.1016/j.gassur.2004.10.014 (2005). [DOI] [PubMed] [Google Scholar]
  • 148.Martinez Rivera A & Wales PW Intestinal transplantation in children: current status. Pediatr Surg Int 32, 529–540, doi: 10.1007/s00383-016-3885-2 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Tzakis AG et al. Clinical intestinal transplantation: focus on complications. Transplant Proc 24, 1238–1240 (1992). [PMC free article] [PubMed] [Google Scholar]
  • 150.Lichtenstein GR & Rutgeerts P Importance of mucosal healing in ulcerative colitis. Inflamm Bowel Dis 16, 338–346, doi: 10.1002/ibd.20997 (2010). [DOI] [PubMed] [Google Scholar]
  • 151.Cruz-Acuna R et al. PEG-4MAL hydrogels for human organoid generation, culture, and in vivo delivery. Nat Protoc 13, 2102–2119, doi: 10.1038/s41596-018-0036-3 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Finkbeiner SR et al. Generation of tissue-engineered small intestine using embryonic stem cell-derived human intestinal organoids. Biol Open 4, 1462–1472, doi: 10.1242/bio.013235 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Khalil HA et al. Intestinal epithelial replacement by transplantation of cultured murine and human cells into the small intestine. PLOS ONE 14, e0216326, doi: 10.1371/journal.pone.0216326 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.O’Neill JD, Pinezich MR, Guenthart BA & Vunjak-Novakovic G Gut bioengineering strategies for regenerative medicine. Am J Physiol Gastrointest Liver Physiol 320, G1–G11, doi: 10.1152/ajpgi.00206.2020 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Beetler DJ et al. The evolving regulatory landscape in regenerative medicine. Molecular Aspects of Medicine 91, 101138, doi: 10.1016/j.mam.2022.101138 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Costello CM et al. Synthetic small intestinal scaffolds for improved studies of intestinal differentiation. Biotechnol Bioeng 111, 1222–1232, doi: 10.1002/bit.25180 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Sala FG, Kunisaki SM, Ochoa ER, Vacanti J & Grikscheit TC Tissue-engineered small intestine and stomach form from autologous tissue in a preclinical large animal model. J Surg Res 156, 205–212, doi: 10.1016/j.jss.2009.03.062 (2009). [DOI] [PubMed] [Google Scholar]
  • 158.Sala FG et al. A multicellular approach forms a significant amount of tissue-engineered small intestine in the mouse. Tissue Eng Part A 17, 1841–1850, doi: 10.1089/ten.TEA.2010.0564 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Clevers H et al. Tissue-Engineering the Intestine: The Trials before the Trials. Cell Stem Cell 24, 855–859, doi: 10.1016/j.stem.2019.04.018 (2019). [DOI] [PubMed] [Google Scholar]
  • 160.Liu Y et al. Comparison of polyglycolic acid, polycaprolactone, and collagen as scaffolds for the production of tissue engineered intestine. J Biomed Mater Res B Appl Biomater 107, 750–760, doi: 10.1002/jbm.b.34169 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Sugimoto S et al. Reconstruction of the Human Colon Epithelium In Vivo. Cell Stem Cell 22, 171–176 e175, doi: 10.1016/j.stem.2017.11.012 (2018). [DOI] [PubMed] [Google Scholar]
  • 162.Khalili AA & Ahmad MR A Review of Cell Adhesion Studies for Biomedical and Biological Applications. Int J Mol Sci 16, 18149–18184, doi: 10.3390/ijms160818149 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Mooney DJ & Vandenburgh H Cell delivery mechanisms for tissue repair. Cell Stem Cell 2, 205–213, doi: 10.1016/j.stem.2008.02.005 (2008). [DOI] [PubMed] [Google Scholar]
  • 164.Pompili S, Latella G, Gaudio E, Sferra R & Vetuschi A The Charming World of the Extracellular Matrix: A Dynamic and Protective Network of the Intestinal Wall. Front Med (Lausanne) 8, 610189, doi: 10.3389/fmed.2021.610189 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Goto T, Morishita M, Kavimandan NJ, Takayama K & Peppas NA Gastrointestinal transit and mucoadhesive characteristics of complexation hydrogels in rats. J Pharm Sci 95, 462–469, doi: 10.1002/jps.20566 (2006). [DOI] [PubMed] [Google Scholar]
  • 166.Ouyang J et al. Oral Hydrogel Microbeads-Mediated In Situ Synthesis of Selenoproteins for Regulating Intestinal Immunity and Microbiota. J Am Chem Soc 145, 12193–12205, doi: 10.1021/jacs.3c02179 (2023). [DOI] [PubMed] [Google Scholar]
  • 167.Hu X & Grinstaff MW Advances in Hydrogel Adhesives for Gastrointestinal Wound Closure and Repair. Gels 9, doi: 10.3390/gels9040282 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Bovone G, Dudaryeva OY, Marco-Dufort B & Tibbitt MW Engineering Hydrogel Adhesion for Biomedical Applications via Chemical Design of the Junction. ACS Biomater Sci Eng 7, 4048–4076, doi: 10.1021/acsbiomaterials.0c01677 (2021). [DOI] [PubMed] [Google Scholar]
  • 169.Tian M et al. A Transformable Mucoadhesive Microgel Network for Noninvasive Multimodal Imaging And Radioprotection of a Large Area of the Gastrointestinal Tract. Adv Mater 35, e2303436, doi: 10.1002/adma.202303436 (2023). [DOI] [PubMed] [Google Scholar]

RESOURCES