Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 May 1.
Published in final edited form as: Gastroenterology. 2016 Jan 14;150(5):1098–1112. doi: 10.1053/j.gastro.2015.12.042

Organoid Models of Human Gastrointestinal Development and Disease

Priya H Dedhia 1,4, Nina Bertaux-Skeirik 5, Yana Zavros 5,*, Jason R Spence 2,3,4,*
PMCID: PMC4842135  NIHMSID: NIHMS751600  PMID: 26774180

Abstract

We have greatly advanced our ability to grow a diverse range of tissue-derived and pluripotent stem cell-derived gastrointestinal (GI) tissues in vitro. These systems, broadly referred to as organoids, have allowed the field to move away from the often non-physiologic, transformed cell lines that have been used for decades in GI research. Organoids are derived from primary tissues and have the capacity for long-term growth. They contain varying levels of cellular complexity and physiologic similarity to native organ systems. We review the latest discoveries from studies of tissue-derived and pluripotent stem cell-derived intestinal, gastric, esophageal, liver, and pancreatic organoids. These studies have provided important insights into GI development, tissue homeostasis, and disease and might be used to develop personalized medicines.


The human gastrointestinal (GI) tract comprises this foregut, midgut, and hindgut—each region gives rise to different tissues and organs 14. The foregut endoderm gives rise to the epithelium of the oral cavity, pharynx, esophagus, stomach, liver, pancreas, and proximal duodenum. The midgut and hindgut endoderm give rise to the epithelium of the distal duodenum, jejunum, ileum, colon, rectum and anal canal, as well as the epithelial lining of the bladder and urethra 436. In vitro organoid cultures are generated from stem or progenitor cells and have the capabilities for long-term growth, cellular diversity, function, and spatial organization specific to the organ they represent and they have been used to study endoderm-derived GI organs 3754. Although they were initially developed as a way to culture primary mouse GI tissues, organoid technologies now enable the growth of diverse primary human tissues in vitro 43, 44, 47, 50, 52, 5560.

Before the development of human GI organoids, the GI tract was modelled using cell lines and explanted tissue, both of which have significant limitations (reviewed in 61). Cell lines, for example, are often derived from malignant specimens or are immortalized by viral infection, cell fusion, or introduction of oncogenes, which limit their use in developmental, physiologic, and regenerative studies 6267. Explant cultures, which have organotypic properties such as complex 3-dimensional (3D) architecture and cellular heterogeneity, have important roles in studies of development and physiology, but are limited by their short-term nature 53, 6875. Recent advances in maintaining GI tissues in long-term in vitro cultures have allowed researchers to overcome many of these barriers, invigorating basic and translational research 3752, 54. For example, long-term organoid cultures have been used to examine the stem cell niche, cellular differentiation, interactions between cells, and physiologic functions 37, 38, 4042, 44, 4651, 54, 57, 58, 60, 7685. They have also been used to model malignancy, infection, and inflammation, as well as in toxicology studies.

Many research articles and reviews have highlighted the use of organoid systems to characterize the adult stem cell niche and gut morphogenesis, and will not be covered in detail in this review (see 1, 3, 4043, 56, 58, 59, 70, 8695). We aim to define and discuss the variety of long-term in vitro GI models that have been created, and how they have increased our understanding of development, genetic diseases of the GI tract, malignancy, host–pathogen interactions, tissue reprogramming, and regeneration.

Defining Organotypic GI Culture Models

The term organotypic has referred to tissue explanted from an organ that continues to function as it would in vivo 71, 96, 97. However, with recent advances in de novo differentiation of 3D organ-like tissues from human pluripotent stem cells, and in long-term culture of human primary tissues, the umbrella of the term organotypic has expanded 98. We use the term organotypic to define short-term explant models and long-term in vitro culture systems that recapitulate the complex cellular diversity and function of the native organ. Long-term organotypic culture systems include primary (tissue-derived) epithelium-only organoids 37, 38, 4042, 44, 45, 49, 51, 54, 60, 70, tissue-derived epithelial–mesenchymal organoids 46, 48, 70, 99, and pluripotent stem cell (PSC)-derived organoids 47, 50, 80, 100, 101 (see Figure 1).

Figure 1. Composition, Cellular Sources and Nomenclature for GI Organotypic Culture Systems.

Figure 1

GI organotypic cultures consist of short-term explant cultures and long-term organoid cultures. The 3 types of GI organoid cultures include 1) tissue-derived epithelial, 2) tissue-derived epithelial–mesenchymal, and 3) PSC-derived organoids. Tissue-derived epithelium and PSC-derived intestinal organoids are shown as examples of each type of tissue.

The ability to grow long-term intestinal organotypic cultures in vitro was made possible through identification of conditions that mimic the intestinal stem cell niche 48, 49. In 2009, Sato et al isolated intestinal crypts from mice that gave rise to epithelium-only organoids when cultured with laminin-rich Matrigel and growth factors, which reflected the intestinal niche in vivo 49. These epithelial organoids could be generated from a single intestinal stem cell, identified by LGR5 expression 49. Using another strategy, Ootani et al generated epithelium–mesenchyme organoids by culturing minced fragments of small intestine or colon in a collagen matrix exposed to an air–liquid interface. These fragments gave rise to highly proliferative epithelium, with supportive myofibroblasts located along the basolateral aspect of the epithelial cells. The resulting epithelial–mesenchymal organoids could be maintained in vitro for up to 1 year 48. For long-term growth, intestinal organoids, in both studies, required media containing growth factors that stimulate Wnt signaling; this recapitulates in vivo environment of the intestinal crypt, which contains high levels of Wnt signaling.

These achievements laid the groundwork for studies describing differentiation and growth of human embryonic and induced PSC (ESC and iPSC)-derived organoids 47, 50, 80, 100, 101. In these studies, ESCs or iPSCs were differentiated into intestine or gastric tissue using step-wise differentiation that mimicked stages of embryonic development (reviewed in 1, 3). This culture system, similar to culture of tissue-derived epithelial and epithelial-mesenchymal organoids, required a 3D extracellular matrix with media that supported high levels of Wnt signaling.

Similar 3D cultures, in environments with high levels of Wnt signaling, have more recently enabled the culture of primary human esophageal, gastric, liver, pancreatic, and colon organoids (see Figure 2) 37, 38, 44, 60. We discuss briefly advances in pancreas and liver organoids, but will focus on the implantation of PSC-derived and tissue-derived epithelial esophageal, gastric, and intestinal organoids for studies of development, disease, and regenerative medicine.

Figure 2. GI organ diversity represented in organoid systems.

Figure 2

GI organoids have been generated from esophagus, liver, biliary tree, stomach, pancreas, small intestine, and large intestine. (Note: GI tract schematic modified from reference 4).

Pancreas and Liver

The distal foregut gives rise to organ buds that form the liver and the dorsal and ventral pancreas. During development, the hepatoblast gives rise to ductal epithelium and hepatocytes (reviewed in 102). There is controversy over the process, but in adults, hepatocytes mediate regeneration after liver damage, and a bi-potent progenitor in the ductal epithelium89, 103 has been reported to give rise to hepatocytes and ducts. Interestingly, this bi-potent progenitor also generates epithelial liver organoids in mice.42, 43, 89 Culture of these cells with hepatocyte growth factor, epidermal growth factor (EGF), R-spondin 1, and fibroblast growth factor 10 produces liver organoids that express duct markers. However, inhibition of Notch and transforming growth factor-β (TGFB) promoted hepatocyte differentiation 42. Liver organoids expressing ductal markers have not demonstrated cholangiocyte function.

In addition to tissue-derived liver organoids, hPSC-derived live buds have recently been described. These were generated by combining human iPSC-derived hepatoblasts 104 with human umbilical endothelial vein cells and human mesenchymal stem cells to develop self-organizing 3D tissues 52. Transplantation of the liver buds into mice resulted in vascularization and maturation of the tissue. Furthermore, transplanted liver buds were capable of rescuing drug-induced liver failure. Similar to tissue-derived epithelial liver organoids, however, these liver buds did not generate functional ductal epithelium. More recently, ESCs and iPSCs have been used to generate functional ductal epithelium characterized by bile acid transfer, cystic fibrosis transmembrane conductance regulator (CFTR)-mediated fluid secretion, and responses to the hormones, secretin, and somatostatin 58, 59. It will be interesting to determine if inclusion of PSC-derived cholangiocyte organoids in PSC-derived liver buds results in organization of functional hepatocyte and ductal structures.

Similar to liver epithelial organoids, human pancreatic epithelial organoids can be generated from embryonic progenitors or adult progenitors that arise after injury 41. Pancreatic duct ligation results in Wnt activity and the development of LGR5+ progenitors in the pancreatic ductal epithelium, which can generate mouse pancreatic epithelial organoids. In vitro and upon transplantation after reaggregation with embryonic pancreas, pancreatic organoids primarily expressed ductal markers. However, 5% of re-aggregated transplanted organoids gave rise to monohormonal insulin-, glucagon-, and somatostatin-producing endocrine lineages. On the other hand, mouse pancreatic epithelial organoids generated from mouse embryonic tissue can give rise to acinar and endocrine structures in vitro 40

These findings provide evidence for the limited differentiation potential of organoids generated from adult pancreatic progenitors, compared to organoids derived from embryonic pancreatic progenitors. Human epithelial pancreatic organoids have also been generated from pancreatic ductal cells. Although organoids from adult progenitors are important in the study of pancreatic ductal adenocarcinoma, these organoids require ectopic expression of PDX1, MAFA, and neurogenin-3 (NEUROG3 or PMN) for differentiation into insulin-producing cells. Further studies are required to elucidate conditions for endocrine differentiation of human pancreatic organoids in the absence of genetic modification 56, 105.

Development

Although mouse models and genetic analyses have greatly increased our understanding of embryonic GI development, mouse development does not always recapitulate human development, particularly in development of the intestinal stem cell niche 106111. For example, Paneth cells, which contribute to maintenance of the adult stem cell niche 112, develop in the first trimester of human gestation. However, Paneth cells do not differentiate until post-natal day 14 in mice 106, 107. Furthermore, EGF increases intestinal epithelial maturation in mice but decreases maturation of human fetal intestine in vitro 108111. GI organoids generated from human specimens are therefore a valuable adjunct to mouse developmental studies, and can be used to study developmental events that are difficult to study in vivo.

Small intestine epithelial organoids generated from fetal mice have been used to study the fetal intestinal niche 57, 81. In contrast to adult epithelial organoids, which demonstrate budding morphology, fetal epithelial organoids frequently formed spherical organoids or spheroids. This tendency to form spheroids, however, decreased with age such that intestine from post-natal day 15 mice did not generate spheroids 81. Interestingly, mRNA expression of several important intestinal genes differed markedly between fetal spherical and budding organoids, such that fetal budding organoids more closely resembled adult budding organoids 81.

In a separate study, exogenous Wnt ligand increased maturation of fetal spheroids into budding organoids 57. Epithelial spheroids generated from human fetal small intestine and PSC-derived human intestinal organoids were more closely related to immature mouse fetal spheroids than to adult tissue 57. These studies exploited the fetal state captured by the intestinal organoid culture system to demonstrate that Wnt signaling stimulates a transition of the fetal organoids grown in vitro from spheroid to budding structure. Furthermore, mouse fetal spheroids that readily engrafted into areas of colitis, induced by dextran sodium sulfate (DSS), appeared to adopt a colonic fate, indicated by expression of the colonic enzyme carbonic anhydrase-II and lack of expression of small intestine enzymes such as lysozyme and alkaline-phosphatase 57. These data indicate that mouse fetal small intestine spheroids, in contrast to adult organoids 113, appear to retain plasticity reflecting their immature status. It is unclear if human fetal intestinal spheroids possess similar cellular plasticity.

In addition to using primary human fetal tissue to study human developmental processes, we can use human PSC-derived tissues to study these processes, as they closely resemble fetal tissue. However, human PSC-derived tissues cannot be used to study physiology and function of adult tissues. For example, hepatocytes or liver organoids generated from PSCs were more closely related to early fetal liver rather than adult liver, based on expression of important regulators of liver development 52, 104, 114. Similarly, the maturational status of early iterations of PSC-derived insulin producing cells represented fetal pancreatic β cells, based on their transcriptional profile and lack of response to glucose 115.

The transcriptome of PSC-derived gastric and lung organoids more closely resembles that of fetal compared to adult lung 47, 116. In addition, RNA sequence-based transcriptional profiling confirmed that human PSC-derived small intestine organoids are most similar to fetal tissue 82. This study demonstrated that features of human fetal to adult maturation included increased expression of the intestinal stem cell marker OLFM4, as well as increased Paneth cell differentiation and expression of brush border enzymes on enterocytes. Transplantation of PSC-derived organoids into immunocompromised mice resulted in the maturation of these fetal-like tissues into tissues resembling those of adults. Their intestinal architecture included prominent crypt-villus structures; increased expression of OLFM4; brush border enzymes; and other markers of mature small intestine such as dipeptidyl peptidase 4, glucose transporter type 2, and villin82, 117.

Given the diverse array of in vitro grown human tissues (primary or PSC-derived, fetal or adult, epithelium only or epithelium plus mesenchyme), it will be important to consider the experimental strengths and limitations of each system when designing experiments and implementing such systems. For example, tissue-derived epithelial intestinal organoids can be generated in less than 2 weeks, whereas PSC-derived intestinal organoids require 5 weeks. Fetal epithelial spheroids generated from early intestine progress into budding organoids with low efficiency, whereas fetal epithelial spheroids from perinatal tissue progress to budding organoids with high efficiency 57, 81. Furthermore, mouse epithelial organoids require induction of inflammation for transplantation, whereas human epithelial organoids have n successfully transplanted. Fetal epithelial spheroids might therefore be best suited for in vitro physiologic studies that characterize the early fetal intestine. In contrast, PSC-derived intestinal organoids undergo maturation with high efficiency when transplanted in vivo, and are therefore amenable to in vivo studies of human development.

Models of GI Disease

GI organoids have been used to model human genetic diseases, infections, inflammatory bowel diseases (IBD), and malignancies. Because of our ability to grow robust and diverse patient-derived organoids, we can use these tissues to conduct drug screens and identify personalized approaches for treating disease.

Monogenic diseases that affect the epithelium

Epithelial organoids have been used to assess function of the CFTR—mutations in this transporter can cause CF 78, 118. Although CF is most commonly associated with the pulmonary epithelium, CFTR mutations result in accumulation of thick secretions in the pulmonary and GI tracts. Moreover, simple rectal biopsies are relatively non-invasive compared to bronchoscopy or endoscopy, and can be conducted at a routine doctors visit. Dekkers et al identified forskolin-induced swelling of normal epithelial intestinal organoids as a physiologic function of non-mutant CFTR. Strikingly, epithelial organoids generated primarily from patients with CF did not undergo forskolin-induced swelling 78. However, forskolin-induced swelling was restored in patient-derived organoids upon incubation with pharmacologic agents that improved function of the CFTR. Furthermore, correction of CFTR mutations in epithelial organoids from the small intestine of patients with CF, using the CRISPR/Cas9 genome editing system, reduced forskolin-induced swelling 118. Epithelial organoid cultures can therefore model disease physiology, and genome editing can be used to repair genetic mutations in organoids.

GI epithelial organoids have also been used to study microvillus inclusion disease (MVID) and multiple intestinal atresia (MIA) 119, 120. MVID is frequently caused by mutations in myosin Vb (MYO5B) that result in inappropriate localization of apical proteins and enterocyte polarization 121, 122. Wiegerinck et al identified a mutation in syntaxin 3 (STX3) by whole-exome sequencing in patient with MVID but no mutation in MYO5B. Duodenal epithelial enteroids from these patients had partial loss of microvilli, inappropriate accumulation of vesicles, and absence of STX3 protein 120. Patients with MIA can develop atresia from the stomach to rectum 123. Using a combination of genome-wide linkage analysis and whole-exome sequencing, Bigorge et al and Avitzur et al identified previously uncharacterized mutations in the tetratricopeptide repeat domain–7A (TTC7A) of patients with MIA and combined immunodeficiency. Intestinal epithelial organoids generated from a subset of these patients demonstrated disturbed polarity and disrupted epithelial architecture 119. Epithelial organoids generated from patient specimens can therefore model and increase our understanding of numerous genetic diseases of the GI tract. Epithelial organoids could be of particular use helping us identify the GI component of complex genetic diseases that affect multiple organ systems.

Enteric anendocrinosis is an extremely rare disease caused by mutations in NEUROG3 that is characterized by severe malabsorptive diarrhea and a lack of intestinal enteroendocrine cells 124, 125. PSC-derived intestinal organoids with adenoviral knockdown of NEUROG3 have fewer enteroendocrine cells, similar to patients with anendocrinosis50. Researchers recently disrupted NEUROG3 in ESCs126, but these cells could still form mature pancreatic endocrine cells. We anticipate the PSC-derived human intestinal organoids will be devoid of enteroendocrine cells, although additional studies are required.

IBD

IBD is characterized by mucosal inflammation of the GI tract that is caused by a combination of genetic susceptibility, immune dysregulation, disruptions in the microbial communities, and environmental factors 127. Organoid cultures have not yet been used to directly model IBD, but have been used to study cell death, mucosal integrity, and the effects of inflammatory cytokines 118, 128130. For example, IBD specimens were found to contain foci of hypoxic inflammation 131 and levels of hypoxia inducible factor 2α (HIF2A or EPAS) were increased in the intestinal epithelial cells from mice with colitis and patients with IBD 129. Disruption of Hif2α in mice reduced features of colitis induced by DSS. Intestinal epithelial cells from the HIF2A-null mice expressed lower levels of inflammatory cytokines, including tumor necrosis factor (TNF), following DSS induction of colitis, compared to mice with that express HIF2A. Similarly, under hypoxic conditions, PSC-derived intestinal organoids increased expression of TNF 129.

IBD can also lead to fibrosis in the intestine, and PSC-derived organoids have been used to model a fibrosis-like response to TGFB stimulation that could be reversed by anti-fibrotic drugs 132. Abnormal Paneth cell phenotypes are associated with specific susceptibility alleles for Crohn’s disease 133. Administration of interferon-γ (IFNG), an inflammatory cytokine, to cultures of mouse small intestinal organoids caused Paneth cell degranulation, associated with apoptosis 134. In the healthy intestine, Paneth cells are quickly replaced. However, continuous exposure to IFNG causes progressive loss of Paneth cells, which could contribute to development of IBD 134. Further studies, potentially with epithelial organoids from patients with IBD, are necessary to determine if Paneth cell degranulation is involved in the pathogenesis of IBD.

IBD is associated with increased risk for colorectal cancer. Oxidative stress could be involved in the pathogenesis of IBD and/or IBD-associated cancer. Deletion of the antioxidant selenoprotein P exacerbated DSS-induced colitis and tumorigenesis 135. Small intestine epithelial organoids generated from these mice had increased baseline proliferation but decreased survival when exposed to the oxidative stressor hydrogen peroxide 135. It will be interesting to determine if this paradoxical relationship also occurs in epithelial organoids generated from patients with IBD susceptibility alleles.

Cell death has been analyzed in mouse small intestine epithelial organoids; findings might be applied studies of IBD 128. Future studies of organoid models will facilitate the exploration of individual and combinatorial contributions of Paneth cell and goblet cell dysfunction, the gut microbiome, and cytokine levels to the pathogenesis of IBD.

Interactions between GI tissues and microbes

Many different types of microbes can cause GI diseases or contribute to their development in susceptible individuals. Studies of GI organoids might increase our understanding of susceptibility to and pathogenesis of microbial infections and possible therapies. Gastric organoids are of particular importance because the recapitulate in vivo physiology and respond to infections like in vivo tissues; they might be used to study malignant transformation secondary to infection.

In the stomach, histamine stimulates parietal cells to secrete acid. Gastric epithelial organoids co-cultured with immortalized stomach mesenchymal cells reduced intra-luminal pH in response to histamine; this effect was reversed by the proton pump inhibitor omeprazole136. Helicobacter pylori cause most cases of peptic ulcer disease by inducing hypersecretion of acid and have been associated with gastric cancer. Proliferation increases in PSC-derived gastric organoids infected with H pylori but not with variants that lack its virulence factor, CagA47.

Injection of human gastric epithelial organoids with H pylori leads to activation of the transcription factor NF-κB, which is activated in gastric cancer cells. This led to increased expression of interleukin-8, an inflammatory cytokine 137. A separate study that found activation of NF-κB following injection of H pylori into mouse gastric epithelial organoids; 84 it reported upregulation of Sonic hedgehog, a protein involved in vertebrate development and the immune response to H pylori 84. In a similar study, Wroblewski et al showed that H pylori infection of mouse gastric epithelial organoids resulted in mislocalization of occludin at the tight junction, previously observed in vivo138, 139. Similar to findings from PSC-derived gastric organoids, this study demonstrated that only H pylori variants that expressed CagA increased proliferation in gastric organoids138. Together, these studies have shown that H pylori infection can be modeled gastric organoids and can be used to study gastric transformation.

Intestinal organoids have also been used to study pathogens 130, 140143. Colonization of mouse small intestine epithelial organoids with Salmonella enterica serovar Typhimurium (referred to hereafter as Salmonella) reduced their growth, disrupted tight junctions, activated inflammatory signaling via NF-κB, and reduced expression of Lgr5140. This study revealed the complexity of the response to infection, even in the absence of immune cells.

Another study capitalized on the absence of immune cells by studying the effect of α-defensins—antimicrobial peptides produced in response to Salmonella infection 141. Small intestine epithelial organoids made from mice with wild-type or mutated α-defensins were injected with Salmonella. Absence of mature α-defensin reduced intra-luminal bacterial killing, which could be partially restored by expression of human α-defensin. This study demonstrated the contribution of α-defensins to the immune response to Salmonella, which had been a challenge to establish in vivo due to the commensal microbiota and cellular immune response.

Forbester et al demonstrated that injection of Salmonella into the lumen of human PSC-derived organoids resulted in epithelial invasion, but this could be inhibited with injection of a mutant strain of Salmonella with reduced invasive capacity 142. Researchers have also studied human PSC-derived organoids infected with Clostridium difficile—the leading cause of healthcare-associated diarrhea, on intestinal epithelium 130. The obligate anaerobe was able to survive up to 12 hours after intra-luminal injection, indicating low intra-luminal oxygen concentration despite ambient oxygen culture conditions. Furthermore, purified toxin A and toxin B, determinants of C difficile virulence, also disrupted paracellular barrier function whereas a non-toxogenic C difficile strain did not. PSC-derived intestinal organoids can therefore be used to study anaerobic pathogen infections and possible treatments.

GI organoids have been an indispensable tool for studies of factors that contribute to complex and poorly understood diseases such as necrotizing enterocolitis (NEC). NEC, which affects premature, formula-fed infants, results in bowel necrosis. Sodhi et al demonstrated that lack of Toll-like receptor-4 (TLR4) protected neonatal mice from hypoxia-induced NEC 144. Interestingly, small intestine epithelial organoids generated from these mutant mice had increased in goblet cell differentiation. Administration of the Notch inhibitor, dibenzapine, protected wild-type mice from NEC.

Researchers observed an increase in p53 up-regulated modulator of apoptosis (PUMA), which regulates apoptosis, upon activation of TLR4 145. Activation of TLR4 in mouse small intestine organoids also resulted in PUMA upregulation, as well as reduced proliferation and increased apoptosis. However, apoptosis was not observed in organoids that lacked PUMA. Furthermore, PUMA was upregulated in intestinal crypts of wild-type mice and patients with NEC, but not TLR4-null mice. The use of human neonatal epithelial organoids and PSC-derived organoids to study the effects of microbes on the intestinal epithelium in the presence and absence of TLR4 signaling could increase our understanding of the pathogenesis of NEC. Finkbeiner et al demonstrated that clinical rotavirus isolates can replicate within the epithelial and mesenchymal cell compartments of PSC-derived intestinal organoids. Epithelial and PSC-derived GI organoids can therefore be used to study host–pathogen interactions and develop new therapies for GI infections.

In addition to pathogens, GI organoids have been used to understand the influence of indigenous (commensal) microbes on epithelial health. Stimulation of mouse small intestine crypts with muramyl dipeptide, a common bacterial peptidoglycan motif, increased organoid formation146. This survival advantage was lost with deletion of NOD2, a receptor that directly senses bacterial products. Mice given muramyl dipeptide before intestinal insult with doxorubicin had increased crypt survival, not seen in NOD2-knockout mice. Lukovac et al demonstrated that presence of certain commensal bacteria led to expression of genes involved in lipid metabolism in the intestinal epithelia 147.

Cancer

GI cancer organoids can model the pathophysiology of GI malignancies and their response to treatment. Colonic epithelial cells accumulate genetic changes that transform them into adenocarcinomas—intestinal organoids are powerful tools for studying this process. In epithelial organoids generated from intestinal adenomas of APC-deficient mice, R-spondin-1 was not required for constitutive Wnt signaling 60. Lentiviral knockdown of APC in mouse small intestine epithelial organoids confirmed Wnt activation and perturbed differentiation; these organoids generated subcutaneous tumors when injected into immunocompromised mice 149. Activation of Kras in these organoids further increased the tumor burden 149.

APC loss of heterozygosity was modeled using epithelial organoids generated from the small intestine of ApcMin/+mice, a model of familial adenomatous polyposis 150, 151. These organoids developed a cystic morphology and acquired DNA damage150, believed to result from increased activity of the repair protein RAD21. ApcMin/+ organoids with Rad21 haploinsufficiency had reduced loss of heterozygosity at Apc. Interestingly, when these organoids were incubated with TGFB, apoptosis increased, associated with increased levels of BCL2-like 11 (apoptosis facilitator) (BIM) 152. Furthermore, in patient-derived colon adenocarcinoma organoids, a BH3 mimetic agent caused cell death within the organoid. Strikingly mutations in Kras, in conjunction with Apc deletion, delayed apoptosis and decreased expression of Bim. Studies in intestinal organoids support the concept that loss of contributes to colorectal carcinogenesis.

CRISPR-Cas9 genome editing has been used to mutate tumor suppressor genes such as APC, SMAD4, and TP53, and the oncogenes KRAS and PI3KCA, in colon epithelial organoids grown from normal human colon tissues 153. Organoids in which all 5 genes were mutated grew independent of growth factors and had gene expression signatures similar to those of epithelial organoids generated from human colon adenomas. Organoids containing the 5 mutations formed tumors when transplanted into mice, but with lower frequency than colorectal adenocarcinoma organoids. Transplantation of engineered and patient-derived colorectal adenocarcinoma organoids under the kidney capsule of immunocompromised mice resulted in tumor formation, although patient-derived adenoma organoids did not engraft. 153. Organoids containing inactivating mutations of APC, SMAD4, and TP53 and activating mutations in KRAS also grew independently of growth factors and formed solid tumors following subcutaneous injection into immunocompromised mice 154. In contrast, patient-derived, control organoids did not engraft after subcutaneous injection. Epithelial organoids can therefore be used to assess the combinatorial effects of mutations associated with malignancy.

Consistent with studies linking mutations in the Wnt signaling pathway to human colon adenocarcinoma, organoids with high expression of the Wnt target gene, EphB2, have a transcriptional signature similar to that of intestinal stem cells and are more likely to form tumors following subcutaneous injection into immunocompromised mice 155. Similarly, mouse small intestinal cells expressing high levels of EPHB2 generated organoids more efficiently than small intestinal cells with low expression of EPHB2 155. These data indicate that epithelial organoids can be used to assess the proliferative capacity of a population of cells, and can also be used to test the efficacy of different chemotherapeutic agents 156.

Human colonic organoids generated from adenocarcinomas and normal tissues can be used to screen drugs and other molecules for their effects on tissues from different patients, with different genomes. Studies of the response of colon adenocarcinoma organoids to small molecules associated loss-of-function mutations in TP53 with resistance to the MDM2 inhibitor nutlin-3a. Organoids from colonic tumors with KRAS mutations were resistant to the anti-EGFR inhibitors cetuximab and BIBW2992 157. GI organoids can therefore be used to bridge pharmacologic studies on short-term cultures with xenotransplant studies, to develop individualized treatment regimens. Organoids might be used in high-throughput screens of drugs and pharmacologic agents for those with effects in the patients from which organoids were generated.

Similar to organoids derived from epithelial tissues, epithelial–mesenchymal organoids have been generated from tumor samples 99, 158. Li et al generated mouse colonic epithelial-mesenchymal organoids with deletion of Apc, activating mutations in Kras, and lentiviral knockdown of p53 and SMAD4. Interestingly, the epithelial–mesenchymal organoids generated from small intestine were more susceptible to transformation than organoids from the colon. The small intestine organoids required alteration of only 2 genes whereas colon organoids required all 4 alterations for transformation.

Adenocarcinoma of the stomach carries is the second most common cause of cancer worldwide and patients have poor prognoses 159. Given the poor response of gastric cancer to treatment, there is a need determine which patients are most likely to respond to specific drugs. Mouse epithelial–mesenchymal organoids from normal tissue have been transformed into gastric cancer organoids through alterations to Kras or p53 99. Gastric organoids with activating mutations of Kras or loss of p53 become dysplastic and generate adenocarcinomas in mice99.

Gastric epithelial organoids have been used to model hereditary gastric cancer and determine the role of TGFB receptor 2 (TGFBR2) in metastasis. Mutations in the cadherin 1, type 1 gene (CDH1 or ECAD) are associated with hereditary gastric cancer. Nadauld et al performed genome sequence analysis of a patient who developed gastric cancer and later developed metastases. This study identified TGFBR2 as genetic factor that increased risk for metastatic disease. Knockdown of TGFBR2 in mouse gastric epithelial-mesenchymal organoids with disruptions in Cdh1 and p53 increased tumorigeniciy when organoids were transplanted back into mice 158.

Whole-genome sequencing and epigenetic profiling of gastric cancer and adjacent normal tissue specimens identified mutations in RHOA 160. Mouse intestinal epithelial organoids with lentiviral overexpression of RHOA mutants survived in the absence of the ROCK inhibitor, which prevents anoikis, after passage. RHOA mutations therefore seem to promote survival of these cells and contribute to gastric carcinogenesis. Studies of organoids generated from patients’ tumors could provide more information about the oncogenic effects of RHOA mutations.

Human gastric cancer epithelial organoids have also been generated by culture of primary cells from resected gastric adenocarcinomas. Human gastric organoids derived from normal and tumor tissues have similar morphologies in culture. However, orthotopic tumors derived from organoids transplanted into mice engraft within the gastric epithelium and generate metaplastic phenotypes similar to those of the patients’ tumor specimens (unpublished results, Bertaux-Skeirik and Zavros). This approach might be used to develop an organoid-based platform for testing the specific responses of individual patients’ tumors to different therapeutic agents.

Epithelial organoids have also been generated from Barrett’s esophagus (BE) and gastric cancer specimens 60, 160. BE predisposes esophageal tissues to dysplasia and adenocarcinoma. BE can be identified based on the presence of intestinal goblet cells. Esophageal epithelial organoids generated from patients with BE could be induced to undergo goblet cell differentiation 60. Given recent developments in the ability to culture mouse esophageal epithelial organoids, it might be possible to develop normal human esophageal epithelial organoids, which could be compared to organoids form BE or esophageal cancer cells.

Epithelial organoids generated from colon cancer tissues have many features of the tumors from which they were derived, are easily expandable, and can be used in screens for drug efficacy and toxicity. Tissue epithelial–mesenchymal organoids and PSC-derived organoids might also be used in screens of pharmacologic agents for activity against gut tumors.

Regenerative Medicine

Factors such as ease of culture, expandability, capacity to model heterogeneous cell fates, and suitability to genetic manipulation make GI organoids a useful tool. Long-term organotypic cultures have been used to study the stem cell niche, and recently as guides for normal development and sources of replacement tissues. Generation of autologous tissue derived from biopsy specimens or iPSCs offers advantages over conventional organ transplants, which are limited in availability and require immunosuppression.

Endocrine differentiation of gut organoids

GI endocrine cells secrete hormones; disruption of to process can cause disease and death. For example, autoimmune destruction of insulin-producing pancreatic β cells results in type I diabetes, and loss of enteroendocrine cells leads to anendocrinosis and life-threatening diarrhea 124, 125. Dysregulation of gut hormones have also been implicated in obesity and metabolic syndrome. Gut organoids can be used to study the pathogenesis of these diseases, and perhaps more importantly, as an alternative tissue source for therapeutics 161.

Ectopic expression of NGN3 or NEUROG3 in mouse epithelial mesenchymal organoids, human PSC-derived intestinal organoids, and human PSC-derived gastric organoids increased numbers of enteroendocrine cells, as it does in mice47, 48, 50, 162, 163, 164. Furthermore, lentiviral knockdown of ARX in PSC-derived intestinal organoids led to reduction of CCK and secretin, similar to mice with intestine-specific disruption of Arx 77.

PSC-derived intestinal organoids have also been used to study trans-differentiation, or conversion from 1 cell lineage into another, of insulin-producing cells165, 166. Bouchi et al identified the transcription factor FOXO1 as a marker of enteroendocrine cell progenitors and a regulator of cell fate decision. Overexpression of a dominant-negative form of FOXO1 increased insulin expression, indicating transdifferentiation into insulin-producing cells. Ectopic expression of the dominant-negative form of FOXO1 also resulted in upregulation of transcription factors important for the development of insulin-producing pancreatic β cells, such as NEUROG3, MAFA, NEUROD1, and NKX6.1. Remarkably, expression of the dominant-negative form of FOXO1 also resulted in secretion of c-peptide, a byproduct that forms when proinsulin is cleaved into insulin in response to insulin secretagogues such as glucose, arginine, and potassium chloride.

Insulin-producing cells in PSC-derived organoids can detect insulin secretagogues and secrete c-peptide. When organoids that express the dominant-negative form of FOXO1 were transplanted subcutaneously into humanized mice, however, human c-peptide from could not be detected in serum. This result is likely due to low yield of insulin-producing cells after FOXO1 inhibition (approximately 0.05% of cells in the organoid). Genes can be expressed from adenoviral vectors in organoids for short-term experiments, but expression is transient. Bouchi et al used lentiviral vectors to knockdown FOXO1 in PSC-derived organoids. Similar to their results with adenoviral inhibition of FOXO1, lentiviral knockdown resulted in an increase in insulin-producing cells.

In another study showing that PSC-derived intestinal organoids can be transdifferentiated into insulin-producing cells, lentivirus was used to ectopically and constitutively express transcription factors necessary for pancreatic β-cell development, including PMN 166. PMN-expressing organoids produced insulin mRNA and protein. Moreover, 22% of cells produced c-peptide, indicating that PMN overexpression results in more efficient generation of insulin-producing cells than FOXO1 inhibition. It is not clear whether insulin-producing cells generated by PMN overexpression can reduce features of diabetes in animal models.

Bouchi et al also confirmed the presence of enteroendocrine cells in PSC-derived organoids that produce glucagon-like peptide-1, gastric inhibitory polypeptide, somatostatin, ghrelin, and cholecystokinin. As such, PSC-derived organoids can be used to study regulators of hormone secretion and the effects of hormone secretion on other tissue types, in vitro and in vivo. PSC-derived organoids might also be used in bariatric surgery research. Weight loss following bariatric surgery does not occur via reduction of enteric surface area or carrying capacity, but rather via biochemical changes 167. PSC-derived organoids, which are in transplanted in discontinuity with the gut, permit delineation of enteroendocrine hormone production due to enteral content vs changes due to biochemical modulators. They might be used to identify biochemical that obviate the need for bariatric surgery.

Gut regeneration

Short-bowel syndrome is a devastating disease that affects children and adults. Many patients must have had more than 50% of their small bowel resected due to congenital anomalies, infection or inflammation, or intestinal ischemia. As a result, they do not maintain fluid, electrolyte, or nutrient homeostasis. Supportive therapy, including parenteral nutrition, and curative therapy, such as small-bowel transplantation, cause complications with significant morbidity and mortality. Human intestinal tissue was successfully integrated into the intestines of mice. However, given that patients with short-bowel syndrome lack sufficient intestinal length, integration of tissue into existing bowel is not feasible, and therefore, alternative sources to replace missing tissue are needed168.

Each of the 3 types of gut organoids have been successfully transplanted into mice, so these tissues might be used to engineer small intestine 82, 117, 99, 57, 113, 54. Mice with DSS-induced colitis given enemas containing epithelial colon organoids had higher body weights 2 weeks after transplantation than mice that did not receive the organoids. The organoids engrafted and were functional 4 weeks after transplant 54. Similarly, mouse fetal and adult small intestine epithelial organoids engrafted into the distal colon after induction of inflammation 57, 113.

Orthotopic transplant of mouse intestinal organoid tissue has required induction of inflammation. Human epithelial organoids transplanted under the kidney capsule or subcutaneously do not engraft in the absence of transforming mutations 153, 154. In contrast, human PSC-derived intestinal organoids have been transplanted under the kidney capsule or in the omentum without the use of inflammatory agents such as DSS 117, 82. Additional studies are necessary to determine if human epithelial organoids can engraft in the colon in the presence of inflammation; this might be a new therapeutic approach for IBD 127.

Intestinal organoids have not yet been used to lengthen small intestine. Most bowel-lengthening studies use organoid units, generated from fragments of full-thickness jejunum or ileum. These contain a mesenchymal core surrounded by a polarized intestinal epithelium. After siting onto polymer scaffolds, organoid units were first successfully transplanted into the omentum of syngeneic Lewis rats to create tissue-engineered small intestine (TESI) 169. TESI have been appended to the anastomosis in rat models of small bowel resection 170. Interestingly, inclusion of TESI in the anastomosis increased bowel length 9 months after bowel reconstruction and caused faster weight gain after reconstruction 170, 171.

TESI has also been generated from human small intestinal tissue and successfully transplanted into immunocompromised mice 168. The major limitation of this approach, however, is the inability to expand TESI in vitro. In contrast, reconstituted epithelial–mesenchymal organoids, epithelial organoids, and PSC-derived organoids can all be expanded in vitro. More studies are needed to determine whether these approaches can be used to increase bowel length. Reconstituted epithelial–mesenchymal organoids and PSC-derived organoids are likely to be effective because of their mesenchymal support structure. However, synthetic scaffolds could provide the necessary support for transplanted epithelial organoids.

LIMITATIONS

Despite the strengths that organoids offer over traditional cell lines, their use is restricted. For example, although organoids are complex and often retain multiple cell lineages, they lack many of the cellular inputs present in an in vivo system (eg, neural, endothelial, or immune cells). We can increase the complexity of these systems through co-culture with additional cell types, but organoids should still be viewed as a reductionist system.

Organoids are also limited by significant variations among laboratories in methods and tissue sources. This issue is less of a concern with PSC-derived organoids, because laboratories generally access the same cell lines and follow well-established methods to differentiate organoids. Conversely, tissue-derived organoids are generated from patients using growth methods that have been optimized in each laboratory. This heterogeneity affects reproducibility if the derivation and characterization of the lines are not well described. For example, changing growth conditions for tissue-derived intestinal epithelial organoids can skew the differentiation state from stem cell-enriched to a balance of stem cells and differentiated cell types.

A practical limitation of organoid models is their cost of maintenance. Many commercial growth factors that are required for culture—these can be expensive and have poor bioactivity. These limitations also restrict assays that require significant scale-up, such as drug screens, providing barriers to their use in personalized medicine approaches. Many groups have turned to cell lines that generate growth factors in conditioned media, to reduce costs and improve bioactivity, but this introduces additional experimental variation.

Future Directions

Organoid cultures have been developed for multiple GI organs. These systems are multicellular, similar to in vivo models, but have the flexibility of in vitro systems. As such, gut organoids have become a powerful tool with steadily increasing applications.

Intestinal organoid cultures continue to offer insights into the stem cell niche. Researchers have recently begun to focus on generating patient-specific tissues. Individualized organoids can be used to identify intestine-specific pathophysiologies in complex disease processes, such as lack of forskolin-induced swelling in organoids with mutated CFTR. Patient-specific organoids can be used to study mechanisms of diseases such as MVID and MIA. Organoids generated from normal and tumor specimens can be used to characterize the events that occur during transformation, growth, and progression of malignancies such as colorectal or gastric cancers. Finally, organoids generated from patient biopsies could be used to regenerate intestinal tissues for patients with disorders such as short bowel syndrome or IBD and avoid the adverse effects of immune rejection or immune suppression. Finally, organoids can be used in screens of pharmacologic agents that have effects on cells and tissues from individual patients, to advance personalized medicine.

Acknowledgments

We would like to thank Aaron Zorn and James Wells for kindly providing the GI tract schematic shown in Figure 2, which is modified from their outstanding 2009 review on endoderm development (see reference 4). We also would like to thank Fraser Marshall for his assistance with construction of the Figures.

JRS is supported by the Intestinal Stem Cell Consortium (U01DK103141), a collaborative research project funded by the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) and the National Institute of Allergy and Infectious Diseases (NIAID). JRS is also supported by NIAID U19AI116482 and NIDDK K01DK091415. PHD is supported by an NIH T32 postdoctoral fellowship from the University of Michigan Center for Organogenesis (T32-HD007505), and by a fellowship from the American College of Surgeons.

List of abbreviations

Bim

BH3-only protein BimEL isoform

DSS

Dextran sodium sulfate

ECAD

Epithelial cadherin

EGF

Epithelial growth factor

ESC

Embryonic stem cell

GI

Gastrointestinal

iPSC

Induced pluripotent stem cells

IFNG

Interferon-γ

NEC

Necrotizing enterocolitis

PSC

Pluripotent stem cell

PUMA

p53 up-regulated modulator of apoptosis

TESI

Tissue engineered small intestine

TGFB

Transforming growth factor β

TLR4

Toll-like receptor

TGFBR2

Transforming growth factor-beta receptor 2

TNF

Tumor necrosis factor

Footnotes

Author contributions:

All authors contributed to writing and editing this review article.

PHD generated the figures and wrote the figure legends.

Conflicts of Interest: The authors declare that no conflicts exist.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Sinagoga KL, MKW, Wells JM. Multiple factors affecting the differentiation of human enteroendocrine cells. Endoderm Lineages in Development and Disease Keystone Symposium; 2015; Keystone, CO. [Google Scholar]
  • 2.Spence JR, Wells JM. Translational embryology: using embryonic principles to generate pancreatic endocrine cells from embryonic stem cells. Dev Dyn. 2007;236(12):3218–27. doi: 10.1002/dvdy.21366. [DOI] [PubMed] [Google Scholar]
  • 3.Wells JM, Spence JR. How to make an intestine. Development. 2014;141(4):752–60. doi: 10.1242/dev.097386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Zorn AM, Wells JM. Vertebrate endoderm development and organ formation. Annu Rev Cell Dev Biol. 2009;25:221–51. doi: 10.1146/annurev.cellbio.042308.113344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Chawengsaksophak K, et al. Cdx2 is essential for axial elongation in mouse development. Proc Natl Acad Sci U S A. 2004;101(20):7641–5. doi: 10.1073/pnas.0401654101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Dufort D, et al. The transcription factor HNF3beta is required in visceral endoderm for normal primitive streak morphogenesis. Development. 1998;125(16):3015–25. doi: 10.1242/dev.125.16.3015. [DOI] [PubMed] [Google Scholar]
  • 7.Chalmers AD, Slack JM. The Xenopus tadpole gut: fate maps and morphogenetic movements. Development. 2000;127(2):381–92. doi: 10.1242/dev.127.2.381. [DOI] [PubMed] [Google Scholar]
  • 8.Kinkel MD, et al. Cdx4 is required in the endoderm to localize the pancreas and limit beta-cell number. Development. 2008;135(5):919–29. doi: 10.1242/dev.010660. [DOI] [PubMed] [Google Scholar]
  • 9.Martinez Barbera JP, et al. The homeobox gene Hex is required in definitive endodermal tissues for normal forebrain, liver and thyroid formation. Development. 2000;127(11):2433–45. doi: 10.1242/dev.127.11.2433. [DOI] [PubMed] [Google Scholar]
  • 10.Tremblay KD, Zaret KS. Distinct populations of endoderm cells converge to generate the embryonic liver bud and ventral foregut tissues. Dev Biol. 2005;280(1):87–99. doi: 10.1016/j.ydbio.2005.01.003. [DOI] [PubMed] [Google Scholar]
  • 11.Dessimoz J, et al. FGF signaling is necessary for establishing gut tube domains along the anterior-posterior axis in vivo. Mech Dev. 2006;123(1):42–55. doi: 10.1016/j.mod.2005.10.001. [DOI] [PubMed] [Google Scholar]
  • 12.Goessling W, et al. APC mutant zebrafish uncover a changing temporal requirement for wnt signaling in liver development. Dev Biol. 2008;320(1):161–74. doi: 10.1016/j.ydbio.2008.05.526. [DOI] [PubMed] [Google Scholar]
  • 13.Haremaki T, et al. Integration of multiple signal transducing pathways on Fgf response elements of the Xenopus caudal homologue Xcad3. Development. 2003;130(20):4907–17. doi: 10.1242/dev.00718. [DOI] [PubMed] [Google Scholar]
  • 14.Li Y, et al. Sfrp5 coordinates foregut specification and morphogenesis by antagonizing both canonical and noncanonical Wnt11 signaling. Genes Dev. 2008;22(21):3050–63. doi: 10.1101/gad.1687308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.McLin VA, Rankin SA, Zorn AM. Repression of Wnt/beta-catenin signaling in the anterior endoderm is essential for liver and pancreas development. Development. 2007;134(12):2207–17. doi: 10.1242/dev.001230. [DOI] [PubMed] [Google Scholar]
  • 16.Tiso N, et al. BMP signalling regulates anteroposterior endoderm patterning in zebrafish. Mech Dev. 2002;118(1–2):29–37. doi: 10.1016/s0925-4773(02)00252-6. [DOI] [PubMed] [Google Scholar]
  • 17.Wells JM, Melton DA. Early mouse endoderm is patterned by soluble factors from adjacent germ layers. Development. 2000;127(8):1563–72. doi: 10.1242/dev.127.8.1563. [DOI] [PubMed] [Google Scholar]
  • 18.Wills A, et al. Bmp signaling is necessary and sufficient for ventrolateral endoderm specification in Xenopus. Dev Dyn. 2008;237(8):2177–86. doi: 10.1002/dvdy.21631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Chen Y, et al. Retinoic acid signaling is essential for pancreas development and promotes endocrine at the expense of exocrine cell differentiation in Xenopus. Dev Biol. 2004;271(1):144–60. doi: 10.1016/j.ydbio.2004.03.030. [DOI] [PubMed] [Google Scholar]
  • 20.Franklin V, et al. Regionalisation of the endoderm progenitors and morphogenesis of the gut portals of the mouse embryo. Mech Dev. 2008;125(7):587–600. doi: 10.1016/j.mod.2008.04.001. [DOI] [PubMed] [Google Scholar]
  • 21.Kim BM, et al. The stomach mesenchymal transcription factor Barx1 specifies gastric epithelial identity through inhibition of transient Wnt signaling. Dev Cell. 2005;8(4):611–22. doi: 10.1016/j.devcel.2005.01.015. [DOI] [PubMed] [Google Scholar]
  • 22.Kumar M, et al. Signals from lateral plate mesoderm instruct endoderm toward a pancreatic fate. Dev Biol. 2003;259(1):109–22. doi: 10.1016/s0012-1606(03)00183-0. [DOI] [PubMed] [Google Scholar]
  • 23.Stafford D, V, Prince E. Retinoic acid signaling is required for a critical early step in zebrafish pancreatic development. Curr Biol. 2002;12(14):1215–20. doi: 10.1016/s0960-9822(02)00929-6. [DOI] [PubMed] [Google Scholar]
  • 24.Rankin SA, et al. A gene regulatory network controlling hhex transcription in the anterior endoderm of the organizer. Dev Biol. 2011;351(2):297–310. doi: 10.1016/j.ydbio.2010.11.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Roberts DJ, et al. Sonic hedgehog is an endodermal signal inducing Bmp-4 and Hox genes during induction and regionalization of the chick hindgut. Development. 1995;121(10):3163–74. doi: 10.1242/dev.121.10.3163. [DOI] [PubMed] [Google Scholar]
  • 26.Bayha E, et al. Retinoic acid signaling organizes endodermal organ specification along the entire antero-posterior axis. PLoS One. 2009;4(6):e5845. doi: 10.1371/journal.pone.0005845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Huang D, et al. A conserved retinoic acid responsive element in the murine Hoxb-1 gene is required for expression in the developing gut. Development. 1998;125(16):3235–46. doi: 10.1242/dev.125.16.3235. [DOI] [PubMed] [Google Scholar]
  • 28.Dale L, et al. Bone morphogenetic protein 4: a ventralizing factor in early Xenopus development. Development. 1992;115(2):573–85. doi: 10.1242/dev.115.2.573. [DOI] [PubMed] [Google Scholar]
  • 29.Grapin-Botton A. Antero-posterior patterning of the vertebrate digestive tract: 40 years after Nicole Le Douarin’s PhD thesis. Int J Dev Biol. 2005;49(2–3):335–47. doi: 10.1387/ijdb.041946ag. [DOI] [PubMed] [Google Scholar]
  • 30.Keenan ID, Sharrard RM, Isaacs HV. FGF signal transduction and the regulation of Cdx gene expression. Dev Biol. 2006;299(2):478–88. doi: 10.1016/j.ydbio.2006.08.040. [DOI] [PubMed] [Google Scholar]
  • 31.Lickert H, Kemler R. Functional analysis of cis-regulatory elements controlling initiation and maintenance of early Cdx1 gene expression in the mouse. Dev Dyn. 2002;225(2):216–20. doi: 10.1002/dvdy.10149. [DOI] [PubMed] [Google Scholar]
  • 32.Niederreither K, et al. The regional pattern of retinoic acid synthesis by RALDH2 is essential for the development of posterior pharyngeal arches and the enteric nervous system. Development. 2003;130(11):2525–34. doi: 10.1242/dev.00463. [DOI] [PubMed] [Google Scholar]
  • 33.Northrop JL, Kimelman D. Dorsal-ventral differences in Xcad-3 expression in response to FGF-mediated induction in Xenopus. Dev Biol. 1994;161(2):490–503. doi: 10.1006/dbio.1994.1047. [DOI] [PubMed] [Google Scholar]
  • 34.Seifert AW, Harfe BD, Cohn MJ. Cell lineage analysis demonstrates an endodermal origin of the distal urethra and perineum. Dev Biol. 2008;318(1):143–52. doi: 10.1016/j.ydbio.2008.03.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Wang Z, et al. Retinoic acid regulates morphogenesis and patterning of posterior foregut derivatives. Dev Biol. 2006;297(2):433–45. doi: 10.1016/j.ydbio.2006.05.019. [DOI] [PubMed] [Google Scholar]
  • 36.Tam PP, et al. Sequential allocation and global pattern of movement of the definitive endoderm in the mouse embryo during gastrulation. Development. 2007;134(2):251–60. doi: 10.1242/dev.02724. [DOI] [PubMed] [Google Scholar]
  • 37.Barker N, et al. Lgr5(+ve) stem cells drive self-renewal in the stomach and build long-lived gastric units in vitro. Cell Stem Cell. 2010;6(1):25–36. doi: 10.1016/j.stem.2009.11.013. [DOI] [PubMed] [Google Scholar]
  • 38.DeWard AD, Cramer J, Lagasse E. Cellular heterogeneity in the mouse esophagus implicates the presence of a nonquiescent epithelial stem cell population. Cell Rep. 2014;9(2):701–11. doi: 10.1016/j.celrep.2014.09.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Gonzalez LM, et al. Cell lineage identification and stem cell culture in a porcine model for the study of intestinal epithelial regeneration. PLoS One. 2013;8(6):e66465. doi: 10.1371/journal.pone.0066465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Greggio C, et al. Artificial three-dimensional niches deconstruct pancreas development in vitro. Development. 2013;140(21):4452–62. doi: 10.1242/dev.096628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Huch M, et al. Unlimited in vitro expansion of adult bi-potent pancreas progenitors through the Lgr5/R-spondin axis. EMBO J. 2013;32(20):2708–21. doi: 10.1038/emboj.2013.204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Huch M, et al. In vitro expansion of single Lgr5+ liver stem cells induced by Wnt-driven regeneration. Nature. 2013;494(7436):247–50. doi: 10.1038/nature11826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Huch M, et al. Long-term culture of genome-stable bipotent stem cells from adult human liver. Cell. 2015;160(1–2):299–312. doi: 10.1016/j.cell.2014.11.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Jung P, et al. Isolation and in vitro expansion of human colonic stem cells. Nat Med. 2011;17(10):1225–7. doi: 10.1038/nm.2470. [DOI] [PubMed] [Google Scholar]
  • 45.Kalabis J, et al. Isolation and characterization of mouse and human esophageal epithelial cells in 3D organotypic culture. Nat Protoc. 2012;7(2):235–46. doi: 10.1038/nprot.2011.437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Katano T, et al. Establishment of a long-term three-dimensional primary culture of mouse glandular stomach epithelial cells within the stem cell niche. Biochem Biophys Res Commun. 2013;432(4):55–63. doi: 10.1016/j.bbrc.2013.02.051. [DOI] [PubMed] [Google Scholar]
  • 47.McCracken KW, et al. Modelling human development and disease in pluripotent stem-cell-derived gastric organoids. Nature. 2014;516(7531):400–4. doi: 10.1038/nature13863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Ootani A, et al. Sustained in vitro intestinal epithelial culture within a Wnt-dependent stem cell niche. Nat Med. 2009;15(6):701–6. doi: 10.1038/nm.1951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Sato T, et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009;459(7244):262–5. doi: 10.1038/nature07935. [DOI] [PubMed] [Google Scholar]
  • 50.Spence JR, et al. Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro. Nature. 2011;470(7332):105–9. doi: 10.1038/nature09691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Stange DE, et al. Differentiated Troy+ chief cells act as reserve stem cells to generate all lineages of the stomach epithelium. Cell. 2013;155(2):357–68. doi: 10.1016/j.cell.2013.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Takebe T, et al. Vascularized and functional human liver from an iPSC-derived organ bud transplant. Nature. 2013;499(7459):481–4. doi: 10.1038/nature12271. [DOI] [PubMed] [Google Scholar]
  • 53.Tham DM, et al. Expression of extracellular glutathione peroxidase in human and mouse gastrointestinal tract. Am J Physiol. 1998;275(6 Pt 1):G1463–71. doi: 10.1152/ajpgi.1998.275.6.G1463. [DOI] [PubMed] [Google Scholar]
  • 54.Yui S, et al. Functional engraftment of colon epithelium expanded in vitro from a single adult Lgr5(+) stem cell. Nat Med. 2012;18(4):618–23. doi: 10.1038/nm.2695. [DOI] [PubMed] [Google Scholar]
  • 55.Lahar N, et al. Intestinal subepithelial myofibroblasts support in vitro and in vivo growth of human small intestinal epithelium. PLoS One. 2011;6(11):e26898. doi: 10.1371/journal.pone.0026898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Boj SF, et al. Organoid models of human and mouse ductal pancreatic cancer. Cell. 2015;160(1–2):324–38. doi: 10.1016/j.cell.2014.12.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Fordham RP, et al. Transplantation of expanded fetal intestinal progenitors contributes to colon regeneration after injury. Cell Stem Cell. 2013;13(6):734–44. doi: 10.1016/j.stem.2013.09.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ogawa M, et al. Directed differentiation of cholangiocytes from human pluripotent stem cells. Nat Biotechnol. 2015;33(8):853–61. doi: 10.1038/nbt.3294. [DOI] [PubMed] [Google Scholar]
  • 59.Sampaziotis F, et al. Cholangiocytes derived from human induced pluripotent stem cells for disease modeling and drug validation. Nat Biotechnol. 2015;33(8):845–52. doi: 10.1038/nbt.3275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Sato T, et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett’s epithelium. Gastroenterology. 2011;141(5):1762–72. doi: 10.1053/j.gastro.2011.07.050. [DOI] [PubMed] [Google Scholar]
  • 61.Pageot LP, et al. Human cell models to study small intestinal functions: recapitulation of the crypt-villus axis. Microsc Res Tech. 2000;49(4):394–406. doi: 10.1002/(SICI)1097-0029(20000515)49:4<394::AID-JEMT8>3.0.CO;2-K. [DOI] [PubMed] [Google Scholar]
  • 62.Paul EC, Hochman J, Quaroni A. Conditionally immortalized intestinal epithelial cells: novel approach for study of differentiated enterocytes. Am J Physiol. 1993;265(1 Pt 1):C266–78. doi: 10.1152/ajpcell.1993.265.1.C266. [DOI] [PubMed] [Google Scholar]
  • 63.Sambuy Y, et al. The Caco-2 cell line as a model of the intestinal barrier: influence of cell and culture-related factors on Caco-2 cell functional characteristics. Cell Biol Toxicol. 2005;21(1):1–26. doi: 10.1007/s10565-005-0085-6. [DOI] [PubMed] [Google Scholar]
  • 64.Rousset M, et al. Reversible forskolin-induced impairment of sucrase-isomaltase mRNA levels, biosynthesis, and transport to the brush border membrane in Caco-2 cells. J Cell Physiol. 1989;141(3):627–35. doi: 10.1002/jcp.1041410322. [DOI] [PubMed] [Google Scholar]
  • 65.Pinto M, SR-L, Appay MD, Kedinger M, Triadou N, Dussaulx E, Lacroix B, Simon-Assman P, Haffen K, Fogh J, Zweibaum A. Enterocyte-like differentiation and polarization of the human colon carcinoma cell line Caco-2 in culture. Biol Cell. 1983;47:323–30. [Google Scholar]
  • 66.Menard DBJ. Human intestinal brush border membrane hydrolases. In: BG, editor. Membrane Physiopathology. Kluwer Academic Publisher; Norwell: 1994. pp. 319–341. [Google Scholar]
  • 67.Zweibaum A, Chantret I. Human colon carcinoma cell lines as in vitro models for the study of intestinal cell differentiation. In: Smith FSMW, editor. Adaptation and development of gastrointestinal function. Manchester University Press; Manchester, England: 1989. pp. 103–112. [Google Scholar]
  • 68.Evans GS, et al. The development of a method for the preparation of rat intestinal epithelial cell primary cultures. J Cell Sci. 1992;101(Pt 1):219–31. doi: 10.1242/jcs.101.1.219. [DOI] [PubMed] [Google Scholar]
  • 69.Fukamachi H. Proliferation and differentiation of fetal rat intestinal epithelial cells in primary serum-free culture. J Cell Sci. 1992;103(Pt 2):511–9. doi: 10.1242/jcs.103.2.511. [DOI] [PubMed] [Google Scholar]
  • 70.Aaltonen J, et al. An autosomal locus causing autoimmune disease: autoimmune polyglandular disease type I assigned to chromosome 21. Nat Genet. 1994;8(1):83–7. doi: 10.1038/ng0994-83. [DOI] [PubMed] [Google Scholar]
  • 71.Randall KJ, Turton J, Foster JR. Explant culture of gastrointestinal tissue: a review of methods and applications. Cell Biol Toxicol. 2011;27(4):267–84. doi: 10.1007/s10565-011-9187-5. [DOI] [PubMed] [Google Scholar]
  • 72.Fletcher G, et al. A role for GATA factors in Xenopus gastrulation movements. Mech Dev. 2006;123(10):730–45. doi: 10.1016/j.mod.2006.07.007. [DOI] [PubMed] [Google Scholar]
  • 73.Browning TH, Trier JS. Organ culture of mucosal biopsies of human small intestine. J Clin Invest. 1969;48(8):1423–32. doi: 10.1172/JCI106108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Danielsen EM. Biosynthesis of intestinal microvillar proteins. Pulse-chase labelling studies on aminopeptidase N and sucrase-isomaltase. Biochem J. 1982;204(3):639–45. doi: 10.1042/bj2040639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Montgomery RK, Zinman HM, Smith BT. Organotypic differentiation of trypsin-dissociated fetal rat intestine. Dev Biol. 1983;100(1):181–9. doi: 10.1016/0012-1606(83)90209-9. [DOI] [PubMed] [Google Scholar]
  • 76.de Lau W, et al. Lgr5 homologues associate with Wnt receptors and mediate R-spondin signalling. Nature. 2011;476(7360):293–7. doi: 10.1038/nature10337. [DOI] [PubMed] [Google Scholar]
  • 77.Du A, et al. Arx is required for normal enteroendocrine cell development in mice and humans. Dev Biol. 2012;365(1):175–88. doi: 10.1016/j.ydbio.2012.02.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Dekkers JF, et al. A functional CFTR assay using primary cystic fibrosis intestinal organoids. Nat Med. 2013;19(7):939–45. doi: 10.1038/nm.3201. [DOI] [PubMed] [Google Scholar]
  • 79.Mizutani T, et al. Real-time analysis of P-glycoprotein-mediated drug transport across primary intestinal epithelium three-dimensionally cultured in vitro. Biochem Biophys Res Commun. 2012;419(2):238–43. doi: 10.1016/j.bbrc.2012.01.155. [DOI] [PubMed] [Google Scholar]
  • 80.Forster R, et al. Human intestinal tissue with adult stem cell properties derived from pluripotent stem cells. Stem Cell Reports. 2014;2(6):838–52. doi: 10.1016/j.stemcr.2014.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Mustata RC, et al. Identification of Lgr5-independent spheroid-generating progenitors of the mouse fetal intestinal epithelium. Cell Rep. 2013;5(2):421–32. doi: 10.1016/j.celrep.2013.09.005. [DOI] [PubMed] [Google Scholar]
  • 82.Finkbeiner SR, et al. Transcriptome-wide Analysis Reveals Hallmarks of Human Intestine Development and Maturation In Vitro and In Vivo. Stem Cell Reports. 2015 doi: 10.1016/j.stemcr.2015.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Grun D, et al. Single-cell messenger RNA sequencing reveals rare intestinal cell types. Nature. 2015;525(7568):251–5. doi: 10.1038/nature14966. [DOI] [PubMed] [Google Scholar]
  • 84.Schumacher MA, et al. Helicobacter pylori-induced Sonic Hedgehog expression is regulated by NFkappaB pathway activation: the use of a novel in vitro model to study epithelial response to infection. Helicobacter. 2015;20(1):19–28. doi: 10.1111/hel.12152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Koo BK, et al. Controlled gene expression in primary Lgr5 organoid cultures. Nat Methods. 2012;9(1):81–3. doi: 10.1038/nmeth.1802. [DOI] [PubMed] [Google Scholar]
  • 86.Rookmaaker MB, et al. Development and application of human adult stem or progenitor cell organoids. Nat Rev Nephrol. 2015;11(9):546–54. doi: 10.1038/nrneph.2015.118. [DOI] [PubMed] [Google Scholar]
  • 87.Sterneckert JL, Reinhardt P, Scholer HR. Investigating human disease using stem cell models. Nat Rev Genet. 2014;15(9):625–39. doi: 10.1038/nrg3764. [DOI] [PubMed] [Google Scholar]
  • 88.Ader M, Tanaka EM. Modeling human development in 3D culture. Curr Opin Cell Biol. 2014;31:23–8. doi: 10.1016/j.ceb.2014.06.013. [DOI] [PubMed] [Google Scholar]
  • 89.Dorrell C, et al. The organoid-initiating cells in mouse pancreas and liver are phenotypically and functionally similar. Stem Cell Res. 2014;13(2):275–83. doi: 10.1016/j.scr.2014.07.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Forbes SJ. Organoid cultures boost human liver cell expansion. Hepatology. 2015 doi: 10.1002/hep.27970. [DOI] [PubMed] [Google Scholar]
  • 91.Leushacke M, Barker N. Ex vivo culture of the intestinal epithelium: strategies and applications. Gut. 2014;63(8):1345–54. doi: 10.1136/gutjnl-2014-307204. [DOI] [PubMed] [Google Scholar]
  • 92.Sato T, Clevers H. SnapShot: Growing Organoids from Stem Cells. Cell. 2015;161(7):1700–1700 e1. doi: 10.1016/j.cell.2015.06.028. [DOI] [PubMed] [Google Scholar]
  • 93.Sato T, Clevers H. Growing self-organizing mini-guts from a single intestinal stem cell: mechanism and applications. Science. 2013;340(6137):1190–4. doi: 10.1126/science.1234852. [DOI] [PubMed] [Google Scholar]
  • 94.Greggio C, De Franceschi F, Grapin-Botton A. Concise reviews: In vitro-produced pancreas organogenesis models in three dimensions: self-organization from few stem cells or progenitors. Stem Cells. 2015;33(1):8–14. doi: 10.1002/stem.1828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Lancaster MA, Knoblich JA. Organogenesis in a dish: modeling development and disease using organoid technologies. Science. 2014;345(6194):1247125. doi: 10.1126/science.1247125. [DOI] [PubMed] [Google Scholar]
  • 96.Maximov A. Contributions to Embryology. Vol. 16. Carnegie Institute; Washington: 1925. Tissue cultures of young mammalian embryos; pp. 47–113. [Google Scholar]
  • 97.Strangeways TSP, HBF Experimental Studies on the Differentiation of Embryonic Tissues Growing in vivo and in vitro.--I. The Development of the Undifferentiated Limb-Bud (a) when Subcutaneously Grafted into the Post-Embryonic Chick and (b) when Cultivated in vitro. I Proc Roy Soc. 100(703):273–282. [Google Scholar]
  • 98.Shamir ER, Ewald AJ. Three-dimensional organotypic culture: experimental models of mammalian biology and disease. Nat Rev Mol Cell Biol. 2014;15(10):647–64. doi: 10.1038/nrm3873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Li X, et al. Oncogenic transformation of diverse gastrointestinal tissues in primary organoid culture. Nat Med. 2014;20(7):769–77. doi: 10.1038/nm.3585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Hannan NR, et al. Generation of multipotent foregut stem cells from human pluripotent stem cells. Stem Cell Reports. 2013;1(4):293–306. doi: 10.1016/j.stemcr.2013.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.McCracken KW, et al. Generating human intestinal tissue from pluripotent stem cells in vitro. Nat Protoc. 2011;6(12):1920–8. doi: 10.1038/nprot.2011.410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Miyajima A, Tanaka M, Itoh T. Stem/progenitor cells in liver development, homeostasis, regeneration, and reprogramming. Cell Stem Cell. 2014;14(5):561–74. doi: 10.1016/j.stem.2014.04.010. [DOI] [PubMed] [Google Scholar]
  • 103.Yanger K, et al. Adult hepatocytes are generated by self-duplication rather than stem cell differentiation. Cell Stem Cell. 2014;15(3):340–9. doi: 10.1016/j.stem.2014.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Si-Tayeb K, et al. Highly efficient generation of human hepatocyte-like cells from induced pluripotent stem cells. Hepatology. 2010;51(1):297–305. doi: 10.1002/hep.23354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Lee J, et al. Expansion and conversion of human pancreatic ductal cells into insulin-secreting endocrine cells. Elife. 2013;2:e00940. doi: 10.7554/eLife.00940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Bry L, et al. Paneth cell differentiation in the developing intestine of normal and transgenic mice. Proc Natl Acad Sci U S A. 1994;91(22):10335–9. doi: 10.1073/pnas.91.22.10335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Mallow EB, et al. Human enteric defensins. Gene structure and developmental expression. J Biol Chem. 1996;271(8):4038–45. doi: 10.1074/jbc.271.8.4038. [DOI] [PubMed] [Google Scholar]
  • 108.Beaulieu JF, Menard D, Calvert R. Influence of epidermal growth factor on the maturation of the fetal mouse duodenum in organ culture. J Pediatr Gastroenterol Nutr. 1985;4(3):476–81. doi: 10.1097/00005176-198506000-00026. [DOI] [PubMed] [Google Scholar]
  • 109.Calvert R, Beaulieu JF, Menard D. Epidermal growth factor (EGF) accelerates the maturation of fetal mouse intestinal mucosa in utero. Experientia. 1982;38(9):1096–7. doi: 10.1007/BF01955387. [DOI] [PubMed] [Google Scholar]
  • 110.Menard D, Arsenault P, Pothier P. Biologic effects of epidermal growth factor in human fetal jejunum. Gastroenterology. 1988;94(3):656–63. doi: 10.1016/0016-5085(88)90236-3. [DOI] [PubMed] [Google Scholar]
  • 111.Menard D, Corriveau L, Arsenault P. Differential effects of epidermal growth factor and hydrocortisone in human fetal colon. J Pediatr Gastroenterol Nutr. 1990;10(1):13–20. doi: 10.1097/00005176-199001000-00003. [DOI] [PubMed] [Google Scholar]
  • 112.Sato T, et al. Paneth cells constitute the niche for Lgr5 stem cells in intestinal crypts. Nature. 2011;469(7330):415–8. doi: 10.1038/nature09637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Fukuda M, et al. Small intestinal stem cell identity is maintained with functional Paneth cells in heterotopically grafted epithelium onto the colon. Genes Dev. 2014;28(16):1752–7. doi: 10.1101/gad.245233.114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Shan J, et al. Identification of small molecules for human hepatocyte expansion and iPS differentiation. Nat Chem Biol. 2013;9(8):514–20. doi: 10.1038/nchembio.1270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Hrvatin S, et al. Differentiated human stem cells resemble fetal, not adult, beta cells. Proc Natl Acad Sci U S A. 2014;111(8):3038–43. doi: 10.1073/pnas.1400709111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Dye BR, et al. In vitro generation of human pluripotent stem cell derived lung organoids. Elife. 2015;4 doi: 10.7554/eLife.05098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Watson CL, et al. An in vivo model of human small intestine using pluripotent stem cells. Nat Med. 2014;20(11):1310–4. doi: 10.1038/nm.3737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Schwank G, et al. Functional repair of CFTR by CRISPR/Cas9 in intestinal stem cell organoids of cystic fibrosis patients. Cell Stem Cell. 2013;13(6):653–8. doi: 10.1016/j.stem.2013.11.002. [DOI] [PubMed] [Google Scholar]
  • 119.Bigorgne AE, et al. TTC7A mutations disrupt intestinal epithelial apicobasal polarity. J Clin Invest. 2014;124(1):328–37. doi: 10.1172/JCI71471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Wiegerinck CL, et al. Loss of syntaxin 3 causes variant microvillus inclusion disease. Gastroenterology. 2014;147(1):65–68 e10. doi: 10.1053/j.gastro.2014.04.002. [DOI] [PubMed] [Google Scholar]
  • 121.Muller T, et al. MYO5B mutations cause microvillus inclusion disease and disrupt epithelial cell polarity. Nat Genet. 2008;40(10):1163–5. doi: 10.1038/ng.225. [DOI] [PubMed] [Google Scholar]
  • 122.Szperl AM, et al. Functional characterization of mutations in the myosin Vb gene associated with microvillus inclusion disease. J Pediatr Gastroenterol Nutr. 2011;52(3):307–13. doi: 10.1097/MPG.0b013e3181eea177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Cole C, et al. Hereditary multiple intestinal atresias: 2 new cases and review of the literature. J Pediatr Surg. 2010;45(4):E21–4. doi: 10.1016/j.jpedsurg.2010.01.017. [DOI] [PubMed] [Google Scholar]
  • 124.Sayar E, et al. Extremely rare cause of congenital diarrhea: enteric anendocrinosis. Pediatr Int. 2013;55(5):661–3. doi: 10.1111/ped.12169. [DOI] [PubMed] [Google Scholar]
  • 125.Jensen JN, et al. Mutant neurogenin-3 in congenital malabsorptive diarrhea. N Engl J Med. 2007;356(17):1781–2. doi: 10.1056/NEJMc063247. author reply 1782. [DOI] [PubMed] [Google Scholar]
  • 126.McGrath PS, et al. The Basic Helix-Loop-Helix Transcription Factor NEUROG3 Is Required for Development of the Human Endocrine Pancreas. Diabetes. 2015;64(7):2497–505. doi: 10.2337/db14-1412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Okamoto R, Watanabe M. Perspectives for Regenerative Medicine in the Treatment of Inflammatory Bowel Diseases. Digestion. 2015;92(2):73–77. doi: 10.1159/000438663. [DOI] [PubMed] [Google Scholar]
  • 128.Grabinger T, et al. Ex vivo culture of intestinal crypt organoids as a model system for assessing cell death induction in intestinal epithelial cells and enteropathy. Cell Death Dis. 2014;5:e1228. doi: 10.1038/cddis.2014.183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Xue X, et al. Endothelial PAS domain protein 1 activates the inflammatory response in the intestinal epithelium to promote colitis in mice. Gastroenterology. 2013;145(4):831–41. doi: 10.1053/j.gastro.2013.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Leslie JL, et al. Persistence and toxin production by Clostridium difficile within human intestinal organoids result in disruption of epithelial paracellular barrier function. Infect Immun. 2015;83(1):138–45. doi: 10.1128/IAI.02561-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Giatromanolaki A, et al. Hypoxia inducible factor 1alpha and 2alpha overexpression in inflammatory bowel disease. J Clin Pathol. 2003;56(3):209–13. doi: 10.1136/jcp.56.3.209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Rodansky ES, et al. Intestinal organoids: a model of intestinal fibrosis for evaluating anti-fibrotic drugs. Exp Mol Pathol. 2015;98(3):346–51. doi: 10.1016/j.yexmp.2015.03.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.VanDussen KL, et al. Genetic variants synthesize to produce paneth cell phenotypes that define subtypes of Crohn’s disease. Gastroenterology. 2014;146(1):200–9. doi: 10.1053/j.gastro.2013.09.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Farin HF, et al. Paneth cell extrusion and release of antimicrobial products is directly controlled by immune cell-derived IFN-gamma. J Exp Med. 2014;211(7):1393–405. doi: 10.1084/jem.20130753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Barrett CW, et al. Selenoprotein P influences colitis-induced tumorigenesis by mediating stemness and oxidative damage. J Clin Invest. 2015;125(7):2646–60. doi: 10.1172/JCI76099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Schumacher MA, et al. The use of murine-derived fundic organoids in studies of gastric physiology. J Physiol. 2015;593(8):1809–27. doi: 10.1113/jphysiol.2014.283028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Bartfeld S, et al. In vitro expansion of human gastric epithelial stem cells and their responses to bacterial infection. Gastroenterology. 2015;148(1):126–136 e6. doi: 10.1053/j.gastro.2014.09.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Wroblewski LE, et al. Helicobacter pylori targets cancer-associated apical-junctional constituents in gastroids and gastric epithelial cells. Gut. 2015;64(5):720–30. doi: 10.1136/gutjnl-2014-307650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Wroblewski LE, et al. Helicobacter pylori dysregulation of gastric epithelial tight junctions by urease-mediated myosin II activation. Gastroenterology. 2009;136(1):236–46. doi: 10.1053/j.gastro.2008.10.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Zhang YG, et al. Salmonella-infected crypt-derived intestinal organoid culture system for host-bacterial interactions. Physiol Rep. 2014;2(9) doi: 10.14814/phy2.12147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Wilson SS, et al. A small intestinal organoid model of non-invasive enteric pathogen-epithelial cell interactions. Mucosal Immunol. 2015;8(2):352–61. doi: 10.1038/mi.2014.72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Forbester JL, et al. Interaction of Salmonella enterica Serovar Typhimurium with Intestinal Organoids Derived from Human Induced Pluripotent Stem Cells. Infect Immun. 2015;83(7):2926–34. doi: 10.1128/IAI.00161-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Engevik MA, et al. Human Clostridium difficile infection: altered mucus production and composition. Am J Physiol Gastrointest Liver Physiol. 2015;308(6):G510–24. doi: 10.1152/ajpgi.00091.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Sodhi CP, et al. Intestinal epithelial Toll-like receptor 4 regulates goblet cell development and is required for necrotizing enterocolitis in mice. Gastroenterology. 2012;143(3):708–18 e1–5. doi: 10.1053/j.gastro.2012.05.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Neal MD, et al. Toll-like receptor 4 is expressed on intestinal stem cells and regulates their proliferation and apoptosis via the p53 up-regulated modulator of apoptosis. J Biol Chem. 2012;287(44):37296–308. doi: 10.1074/jbc.M112.375881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Nigro G, et al. The cytosolic bacterial peptidoglycan sensor Nod2 affords stem cell protection and links microbes to gut epithelial regeneration. Cell Host Microbe. 2014;15(6):792–8. doi: 10.1016/j.chom.2014.05.003. [DOI] [PubMed] [Google Scholar]
  • 147.Lukovac S, et al. Differential modulation by Akkermansia muciniphila and Faecalibacterium prausnitzii of host peripheral lipid metabolism and histone acetylation in mouse gut organoids. MBio. 2014;5(4) doi: 10.1128/mBio.01438-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Siegel R, Desantis C, Jemal A. Colorectal cancer statistics, 2014. CA Cancer J Clin. 2014;64(2):104–17. doi: 10.3322/caac.21220. [DOI] [PubMed] [Google Scholar]
  • 149.Onuma K, et al. Genetic reconstitution of tumorigenesis in primary intestinal cells. Proc Natl Acad Sci U S A. 2013;110(27):11127–32. doi: 10.1073/pnas.1221926110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Xu H, et al. Cohesin Rad21 mediates loss of heterozygosity and is upregulated via Wnt promoting transcriptional dysregulation in gastrointestinal tumors. Cell Rep. 2014;9(5):1781–97. doi: 10.1016/j.celrep.2014.10.059. [DOI] [PubMed] [Google Scholar]
  • 151.Moser AR, et al. The Min (multiple intestinal neoplasia) mutation: its effect on gut epithelial cell differentiation and interaction with a modifier system. J Cell Biol. 1992;116(6):1517–26. doi: 10.1083/jcb.116.6.1517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Wiener Z, et al. Oncogenic mutations in intestinal adenomas regulate Bim-mediated apoptosis induced by TGF-beta. Proc Natl Acad Sci U S A. 2014;111(21):E2229–36. doi: 10.1073/pnas.1406444111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Matano M, et al. Modeling colorectal cancer using CRISPR-Cas9-mediated engineering of human intestinal organoids. Nat Med. 2015;21(3):256–62. doi: 10.1038/nm.3802. [DOI] [PubMed] [Google Scholar]
  • 154.Drost J, et al. Sequential cancer mutations in cultured human intestinal stem cells. Nature. 2015;521(7550):43–7. doi: 10.1038/nature14415. [DOI] [PubMed] [Google Scholar]
  • 155.Merlos-Suarez A, et al. The intestinal stem cell signature identifies colorectal cancer stem cells and predicts disease relapse. Cell Stem Cell. 2011;8(5):511–24. doi: 10.1016/j.stem.2011.02.020. [DOI] [PubMed] [Google Scholar]
  • 156.Emmink BL, et al. Differentiated human colorectal cancer cells protect tumor-initiating cells from irinotecan. Gastroenterology. 2011;141(1):269–78. doi: 10.1053/j.gastro.2011.03.052. [DOI] [PubMed] [Google Scholar]
  • 157.van de Wetering M, et al. Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell. 2015;161(4):933–45. doi: 10.1016/j.cell.2015.03.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Nadauld LD, et al. Metastatic tumor evolution and organoid modeling implicate TGFBR2 as a cancer driver in diffuse gastric cancer. Genome Biol. 2014;15(8):428. doi: 10.1186/s13059-014-0428-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Gomceli I, Demiriz B, Tez M. Gastric carcinogenesis. World J Gastroenterol. 2012;18(37):5164–70. doi: 10.3748/wjg.v18.i37.5164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Wang K, et al. Whole-genome sequencing and comprehensive molecular profiling identify new driver mutations in gastric cancer. Nat Genet. 2014;46(6):573–82. doi: 10.1038/ng.2983. [DOI] [PubMed] [Google Scholar]
  • 161.Troke RC, Tan TM, Bloom SR. The future role of gut hormones in the treatment of obesity. Ther Adv Chronic Dis. 2014;5(1):4–14. doi: 10.1177/2040622313506730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Lopez-Diaz L, et al. Intestinal Neurogenin 3 directs differentiation of a bipotential secretory progenitor to endocrine cell rather than goblet cell fate. Dev Biol. 2007;309(2):298–305. doi: 10.1016/j.ydbio.2007.07.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Jenny M, et al. Neurogenin3 is differentially required for endocrine cell fate specification in the intestinal and gastric epithelium. EMBO J. 2002;21(23):6338–47. doi: 10.1093/emboj/cdf649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Lee CS, et al. Neurogenin 3 is essential for the proper specification of gastric enteroendocrine cells and the maintenance of gastric epithelial cell identity. Genes Dev. 2002;16(12):1488–97. doi: 10.1101/gad.985002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Bouchi R, et al. FOXO1 inhibition yields functional insulin-producing cells in human gut organoid cultures. Nat Commun. 2014;5:4242. doi: 10.1038/ncomms5242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Chen YJ, et al. De novo formation of insulin-producing “neo-beta cell islets” from intestinal crypts. Cell Rep. 2014;6(6):1046–58. doi: 10.1016/j.celrep.2014.02.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Ryan KK, et al. FXR is a molecular target for the effects of vertical sleeve gastrectomy. Nature. 2014;509(7499):183–8. doi: 10.1038/nature13135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Levin DE, et al. Human tissue-engineered small intestine forms from postnatal progenitor cells. J Pediatr Surg. 2013;48(1):129–37. doi: 10.1016/j.jpedsurg.2012.10.029. [DOI] [PubMed] [Google Scholar]
  • 169.Choi RS, Vacanti JP. Preliminary studies of tissue-engineered intestine using isolated epithelial organoid units on tubular synthetic biodegradable scaffolds. Transplant Proc. 1997;29(1–2):848–51. doi: 10.1016/s0041-1345(96)00164-9. [DOI] [PubMed] [Google Scholar]
  • 170.Kaihara S, et al. Long-term follow-up of tissue-engineered intestine after anastomosis to native small bowel. Transplantation. 2000;69(9):1927–32. doi: 10.1097/00007890-200005150-00031. [DOI] [PubMed] [Google Scholar]
  • 171.Grikscheit TC, et al. Tissue-engineered small intestine improves recovery after massive small bowel resection. Ann Surg. 2004;240(5):748–54. doi: 10.1097/01.sla.0000143246.07277.73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Behjati S, et al. Genome sequencing of normal cells reveals developmental lineages and mutational processes. Nature. 2014;513(7518):422–5. doi: 10.1038/nature13448. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES