6
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Organoids and Engineered Organ Systems

      editorial
      , PhD , MD
      Cellular and Molecular Gastroenterology and Hepatology
      Elsevier

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          This introduction to the special Cellular and Molecular Gastroenterology and Hepatology series Organoids and Engineered Organ Systems summarizes original research articles, reviews, and commentaries published within the journal over the past year. A few studies remain to be published, but the series is nearly complete and can be accessed at cmghjournal.org/content/collection_organoids. The revolution initiated by the availability of organoids and engineered organ systems has transformed the research landscape for gastrointestinal, liver, and pancreatobiliary scientists. These powerful new in vitro cellular models to study mechanisms of human gastrointestinal (GI) physiology and pathophysiology have advanced rapidly as a result of collaborative interactions between bioengineers and cell biologists; some of these astonishing advances are included in the special article series. Other contributions highlight the breadth of the topic and show the broad impact that this technology is having on scientific discovery and therapeutic developments in our field. Organoid cell culture technologies take advantage of basic discoveries relevant to stem cell biology and GI tissue development to design culture strategies to sustain the growth of human organ-specific stem cells or to instruct pluripotent stem cells to follow developmental cascades and generate organotypic structures. In parallel, bioengineers and materials scientists have explored novel microfabrication and microfluidic strategies that, when combined with these primary human stem cell cultures, have led to innovative culture models of digestive organs. These dynamic culture systems, commonly containing both organ-specific stem/progenitor and differentiated cells, have been established for human intestine, colon, stomach, esophagus, liver, biliary tissue, and pancreas, providing a major improvement over traditional transformed cell lines derived from GI cancers that have served as the dominant in vitro cell model for many decades. Cultured GI organ models are being used to advance our understanding of a wide array of physiological functions and diseases, providing insights into stem cell biology, mechanisms of cellular differentiation, transport physiology, disease pathogenesis, drug discovery, cancer genetics, inflammatory mechanisms, and more. It is particularly encouraging that bioengineers, biologists, and clinicians are working together to advance this emerging area, each bringing their specialized perspectives and expertise to work toward engineering physiologically relevant human GI organ cell culture systems to model aspects of human GI biology and disease. We are especially pleased that this special series of Cellular and Molecular Gastroenterology and Hepatology captures this diversity in thought and approach, including articles from different expert viewpoints studying a variety of GI organ systems and describing new technological platforms and materials, which together have contributed to several basic scientific discoveries. This topical collection succeeds at multiple levels by providing both in-depth subject reviews for those seeking to learn more about this quickly developing area as well as cutting edge original research articles from leading investigators. Two review articles in particular provide key summaries of GI organoid technology from the bioengineering and developmental biology perspectives. Gural et al 1 provide a comprehensive view of 2-dimensional (2D) and 3-dimensional culture platforms used to study human liver-dependent infectious diseases in their article entitled “Engineered Livers for Infectious Diseases.” This review discusses how these various human liver culture platforms serve as essential tools for biological and therapeutic discovery. The investigators highlight advances in the study of hepatitis and malarial parasites, using these novel culture systems to model human liver cell responses to these important human pathogens. Wells’ laboratory has used insights from developmental biology and pluripotent stem cells to develop novel culture systems of human luminal organs (stomach, intestine, and colon). With co-authors, Wells discusses mechanisms of stomach development and examines how developmental signaling gradients can be manipulated in culture to induce human pluripotent stem cells to differentiate into human gastric tissue in culture. 2 The review, entitled “Translating Developmental Principles to Generate Human Gastric Organoids,” highlights the importance that knowledge of basic developmental mechanisms and cell biology plays in developing strategies to generate complex organotypic cultures. Importantly, organoids generated from human pluripotent stem cells include organ-specific stromal and epithelial cells and thereby provides a potential system for exploration of epithelial-mesenchymal cross-talk during organogenesis and in disease. From the primary research publications it was difficult to select which ones to highlight for this commentary. These primary research studies show the exciting breadth of this maturing and critically important field. Various contributions explore tissue-engineering advances as well as basic and translational discoveries made possible through the use of cultured GI organ systems representing human colon, liver, intestine, and esophagus. Microfabricated organoid culture systems often are designed to allow high-throughput analysis for small-molecule screens, toxicology studies, or drug development. Thus, these in vitro systems show great promise for in vitro reproduction of in vivo human cellular responses, which will be an important component of future drug discovery pipelines. One of these studies that best represents the intersection of basic cell biology with tissue microfabrication and bioengineering was provided by the Magness and Allbritton laboratories. The work, entitled “Formation of Human Colonic Crypt Array by Application of Chemical Gradients Across a Shaped Epithelial Monolayer,” describes a new bioengineered model of the human colonic epithelium. 3 Together these 2 groups, with strengths in stem cell biology and microfabricated platforms, engineered a human colon culture system designed to mimic fundamental aspects of tissue structure. Collagen scaffolds, molded to the shape of human crypts, were seeded with human colonic epithelial stem cells. Microfluidic methodologies were used to establish opposing Wnt and bone morphogenetic protein (BMP) signaling gradients that supported stem/progenitor cells at the base and differentiated cells at the top, thereby replicating in vivo morphology in vitro. To further validate this in vitro system, the investigators assessed responses to short-chain fatty acids and inflammatory mediators and found that they were similar to those observed in vivo. Equally exciting is the adaptability of the primary tissue organoids into diverse research platforms, including 3-dimensional, 2D, and tissue chip approaches. An advance in human intestinal tissue chip technology is reported in an article from Barrett’s laboratory entitled, “Enhanced Utilization of Induced Pluripotent Stem Cell-Derived Human Intestinal Organoids Using Microengineered Chips.” 4 To improve the accessibility of human intestinal cells for high-throughput drug and toxicology screens, Barrett’s group seeded chips with intestinal epithelial cells isolated from human pluripotent stem cell–derived intestinal organoids. This would be a reliable and uniform cell source for the mass production of human intestinal chips. In contrast, the report from Donowitz’s group entitled, “Molecular Basis and Differentiation-Associated Alterations of Anion Secretion in Human Duodenal Enteroid Monolayers,” used a different approach and plated human intestinal organoids in 2D monolayers (which are more convenient for electrophysiological analysis) to define key mechanisms of anion secretion associated with cellular differentiation. 5 Both of these studies afford new opportunities not available with transformed human colon cancer cell lines. Another cell biological advance is reported by the Khetani group in “A Cell Culture Platform to Maintain Long-Term Phenotype of Primary Human Hepatocytes and Endothelial Cells.” 6 This study highlighted a current theme to develop better organoid models by including other relevant cell types in co-culture with GI epithelial cells. Here, they created cultures that included primary human hepatocytes, primary human liver sinusoidal endothelial cells, and mouse 3T3-J2 fibroblast cells to build a more physiologically relevant organotypic system. The tricultures were stable for weeks, showing greater differentiated cellular function than prior cultures. The complexity of cell–cell interactions in these cultures may provide a platform to explore how these contacts support hepatocyte survival and differentiated cell function. Co-culture approaches such as these are currently being used widely in different cell and organ contexts to uncover mechanisms of cell–cell or microbe–cell signaling. Last, but by no means least, the Spence group reported on a novel method of organoid production that greatly impacts clinical and translational research. In “A Method for Cryogenic Preservation of Human Biopsy Specimens and Subsequent Organ Culture,” 7 they described a practical method to cryopreserve live human biopsy tissue, which is simple enough to be adopted in the clinics. These frozen human specimens then can be stored or shipped frozen and later thawed to generate new cultures of gastrointestinal epithelial organoids (stomach, intestine, colon). They show that this methodology is robust and efficient for biopsy specimens from human beings aged 2 to 70 years, yields organoids no different from those grown from fresh nonfrozen biopsy specimens, and show that frozen biopsy specimens can be shipped on dry ice with no loss in culture efficiencies. We anticipate this approach will be rapidly assimilated and widely used in clinical centers, further driving interest in human organoid research and allowing study of patients around the world. Together, this collection highlights some of the important contributions that GI organoids and engineered organ systems have had on our field, and their great potential for highly impactful contributions in the future. Organotypic GI culture models are fueling breakthroughs in the study of human GI development, physiology, pathophysiology, therapeutics, and toxicology. These technologies establish experimentally tractable and physiologically relevant systems to understand human cellular responses, model human diseases, and provide important alternatives to approaches using human cancer cell lines and animal models. It is exciting to see bioengineers and GI scientists working in close collaboration to advance these model systems. Through this shared effort, the future of GI research is bright and made even more powerful when combined with Clustered Regulatory Interspersed Short Palindromic Repeat (CRISPR)/CRISPR-associated protein 9 gene editing approaches to engineer organoid models of human genetic diseases and to tease out the molecular and cellular mechanisms of GI function by manipulation of specific genes and signaling pathways. The next generation of novel human GI organoids along with enhanced microfabrication techniques will yield new insights and provide innovative laboratory platforms to continue the advancement of our understanding of human disease pathogenesis and therapeutics.

          Related collections

          Most cited references7

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          Enhanced Utilization of Induced Pluripotent Stem Cell–Derived Human Intestinal Organoids Using Microengineered Chips

          Background and Aims Human intestinal organoids derived from induced pluripotent stem cells have tremendous potential to elucidate the intestinal epithelium’s role in health and disease, but it is difficult to directly assay these complex structures. This study sought to make this technology more amenable for study by obtaining epithelial cells from induced pluripotent stem cell–derived human intestinal organoids and incorporating them into small microengineered Chips. We then investigated if these cells within the Chip were polarized, had the 4 major intestinal epithelial subtypes, and were biologically responsive to exogenous stimuli. Methods Epithelial cells were positively selected from human intestinal organoids and were incorporated into the Chip. The effect of continuous media flow was examined. Immunocytochemistry and in situ hybridization were used to demonstrate that the epithelial cells were polarized and possessed the major intestinal epithelial subtypes. To assess if the incorporated cells were biologically responsive, Western blot analysis and quantitative polymerase chain reaction were used to assess the effects of interferon (IFN)-γ, and fluorescein isothiocyanate–dextran 4 kDa permeation was used to assess the effects of IFN-γ and tumor necrosis factor-α on barrier function. Results The optimal cell seeding density and flow rate were established. The continuous administration of flow resulted in the formation of polarized intestinal folds that contained Paneth cells, goblet cells, enterocytes, and enteroendocrine cells along with transit-amplifying and LGR5 + stem cells. Administration of IFN-γ for 1 hour resulted in the phosphorylation of STAT1, whereas exposure for 3 days resulted in a significant upregulation of IFN-γ related genes. Administration of IFN-γ and tumor necrosis factor-α for 3 days resulted in an increase in intestinal permeability. Conclusions We demonstrate that the Intestine-Chip is polarized, contains all the intestinal epithelial subtypes, and is biologically responsive to exogenous stimuli. This represents a more amenable platform to use organoid technology and will be highly applicable to personalized medicine and a wide range of gastrointestinal conditions.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Formation of Human Colonic Crypt Array by Application of Chemical Gradients Across a Shaped Epithelial Monolayer

            Background & Aims The successful culture of intestinal organoids has greatly enhanced our understanding of intestinal stem cell physiology and enabled the generation of novel intestinal disease models. Although of tremendous value, intestinal organoid culture systems have not yet fully recapitulated the anatomy or physiology of the in vivo intestinal epithelium. The aim of this work was to re-create an intestinal epithelium with a high density of polarized crypts that respond in a physiologic manner to addition of growth factors, metabolites, or cytokines to the basal or luminal tissue surface as occurs in vivo. Methods A self-renewing monolayer of human intestinal epithelium was cultured on a collagen scaffold microfabricated with an array of crypt-like invaginations. Placement of chemical factors in either the fluid reservoir below or above the cell-covered scaffolding created a gradient of that chemical across the growing epithelial tissue possessing the in vitro crypt structures. Crypt polarization (size of the stem/proliferative and differentiated cell zones) was assessed in response to gradients of growth factors, cytokines, and bacterial metabolites. Results Chemical gradients applied to the shaped human epithelium re-created the stem/proliferative and differentiated cell zones of the in vivo intestine. Short-chain fatty acids applied as a gradient from the luminal side confirmed long-standing hypotheses that butyrate diminished stem/progenitor cell proliferation and promoted differentiation into absorptive colonocytes. A gradient of interferon-γ and tumor necrosis factor-α significantly suppressed the stem/progenitor cell proliferation, altering crypt formation. Conclusions The in vitro human colon crypt array accurately mimicked the architecture, luminal accessibility, tissue polarity, cell migration, and cellular responses of in vivo intestinal crypts.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              A Method for Cryogenic Preservation of Human Biopsy Specimens and Subsequent Organoid Culture

              Human tissue–derived gastrointestinal (GI) organoids have revolutionized the study of human biology,1, 2, 3 and are powerful tools for studying human physiology and disease; however, generation of organoids is limited by access to human tissue and a short window of viability for human samples, putting a hard limit on the time and place in which a patient sample can be used for research. 4 These restraints mean that a laboratory must be relatively geographically close to the source of collection to use the sample within the window of viability. Patient-derived organoids also are being used for drug development, stem cell therapies, and personalized medicine; however, it is not always feasible to prospectively develop organoid line efforts given the time and labor involved.5, 6 To overcome these limitations, we sought to develop a practical method to cryopreserve live human biopsy tissue, which then could be stored or shipped frozen and later thawed to generate new cultures of GI epithelium-only organoids (also referred to as enteroids/colonoids). Here, we describe a simple and robust method to cryopreserve human biopsy specimens that subsequently could be thawed and used to generate epithelium-only organoids. Results/Discussion Endoscopic biopsy specimens were collected with 8-mm2 biopsy forceps (average biopsy size, 5 mm2), transported to the laboratory, divided further into 2- to 3-mm2 fragments, then transferred into a cryopreservation vial containing freezing medium. We tested 2 freezing media formulations: a complex medium (LWRN, fetal bovine serum, dimethyl sulfoxide, Y27632, CHIR99021; see Supplementary Materials and Methods section)7, 9; and a simple freezing medium (Dulbecco's modified Eagle medium/F12, 10% fetal bovine serum, and 10% dimethyl sulfoxide). By using a Mr. Frosty (ThermoFisher Scientific, Waltham, MA) cell-freezing container, the biopsy fragments were frozen to -80°C overnight. Both freezing media formulations performed equivalently (data not shown), thus we focused on the simple formulation. Upon thawing the cryopreserved biopsy specimens, 4 methods were developed for thawing and subsequent establishment of organoid cultures (Figure 1). Frozen vials were thawed at 37°C, the tissue was washed in recovery media (see Supplementary Materials and Methods section, Figure 1). For techniques 1 and 3, tissue was partially digested with dispase or EDTA followed by mechanical perturbation to loosen adhesion between cell layers (Figure 1, Supplementary Figure 1, Supplementary Figure 2, Supplementary Figure 3). 3 The epithelium was pelleted by centrifugation and then resuspended in recovery medium. For techniques 1 and 3, isolated epithelium was embedded in Matrigel (Corning, Bedford, MA) where it was cultured for 3 days in recovery media. 8 Figure 1 Schematic of the methods developed that allow successful generation of patient biopsy-derived epithelial organoids after cryopreservation. (A) The process of endoscopic biopsy collection followed by cryopreservation in a simple freezing medium can be accomplished in typical clinical settings with readily available equipment. After cryopreservation, organoid cultures can be established using 3 different techniques. Technique 1 (left) used a dispase digestion to isolate crypts from freshly thawed tissue. Technique 2 (middle–left) adds a step relative to technique 1 in which the entire biopsy specimen is embedded in Matrigel and allowed to repair from the freezing process before tissue digestion and crypt isolation. Technique 3 (middle–right) is very similar to technique 1 but uses a gentle EDTA treatment to separate the epithelium from the mesenchyme. Technique 4 (right) involves isolation of epithelial crypts before cyropreservation so that, upon thawing, cultures can be seeded immediately without additional tissue manipulations. All 4 techniques result in pure organoid cultures 2 weeks after initially thawing the biopsy specimen. (B) Organoids lines were generated using techniques 1 and 2. (C) Organoid lines were generated using technique 3. (D) Organoid lines were generated using technique 4. We also tested a second thawing technique that omitted enzymatic digestion (technique 2) (Figure 1, Supplementary Figure 2). In this case we thawed the biopsy specimen, washed with recovery media, and embedded whole tissue fragments in Matrigel. These cultures were grown in recovery media for 1 week, where we observed epithelial cysts growing from the biopsy fragment (Supplementary Figure 2 A). Tissue was removed from Matrigel and the epithelium was dissociated enzymatically using dispase, similar to technique 1, and cultured further (Figure 1). We also found that whole fresh crypts could be isolated, cryopreserved, and recovered to generate organoids (technique 4) (Figure 1, Supplementary Figure 2 C). By using techniques 1 and 2, we established 20 frozen biopsy-derived organoid lines from 4 different regions of the GI tract (stomach, duodenum, ileum, and colon) from healthy patients, and 1 organoid line from adenomatous tissue (Figure 1 B). We generated 3 patient-specific organoid lines using technique 3 (Figure 1 C), and 7 patient-specific organoid lines using technique 4 (Figure 1 D). All techniques had a 100% success rate, showing the robustness of the method. The initial growth of organoids from frozen samples is delayed when compared with freshly isolated epithelium (Figure 2 A), but frozen organoids eventually catch up with fresh tissue organoids and are indistinguishable after the first passage (Figure 2 A and B). Figure 2 Organoids derived from fresh and frozen tissue appear morphologically indistinguishable after passage, and transcriptional profiles of organoids derived from fresh and frozen biopsy specimens are nearly identical. (A) The size of organoids derived from different sections of intestine were measured at 4, 7, and 10 days of growth and after the first split. (B) Organoids derived from duodenum (representative biopsy specimen shown at top left, organoids columns A and B), ileum (biopsy top middle, organoids columns C and D), and colon (representative biopsy specimen shown at top right, organoids columns E and F) have different morphologic characteristics when derived from fresh tissue (A1–A4, C1–C4, and E1–E4). Duodenum-derived organoids grow as smooth cystic structures regardless of whether they originate from fresh or frozen tissue (A1–A4 and B1–B4). Organoids from fresh ileum and colon grow with distinct budded morphology whereas those derived from patient-matched frozen tissue (D1–D4 and F1–F4) lack these morphologic distinctions during the early phases of culture establishment. After the first passage and for the remainder of the observed lifetime of the culture, fresh and frozen organoid morphologies cannot be distinguished (A4–F4). (C and D) Clustering organoid RNA sequencing samples by (C) source of variance with principal component analysis or by (D) sample similarity using Pearson correlation. Both analyses generate groups from the same gastrointestinal region regardless of derivation from fresh or frozen tissue biopsy specimens. (E) Differential expression analysis comparing fresh-derived duodenum organoids with frozen-derived duodenum organoids (left), or fresh-derived colon organoids with frozen-derived colon organoids (middle), show a near lack of significant differences in gene expression. In marked contrast, comparing organoids from different regions of the small intestine, such as duodenum and ileum (right), yields more than 9% of genes showing significantly different expression. *Significant difference denotes an adjusted P value of ≤ .01 and fold difference ≥2 (log2 fold difference ≥1 or ≤-1). †Biopsy images are representative and not necessarily tissue-matched to the subsequent organoid images. To determine if cryopreservation caused any molecular differences within the organoids, we performed RNA sequencing on a cohort of fresh tissue organoids (n = 10) and cryopreserved tissue-derived organoids (n = 5). Principal component analysis showed that the strongest driver of variability was the GI region from which organoids were derived (Figure 2 C). Similarly, using Pearson correlation, fresh and frozen organoids from the same region of the intestine clustered together, and there was no clustering based on fresh vs frozen status (Figure 2 D). Finally, we performed differential expression analysis to compare organoids derived from fresh vs frozen biopsy specimens from the same region of the intestine (ie, duodenum or colon). 9 Strikingly, frozen and fresh organoids were nearly identical, with only 46 (0.24% of all genes expressed) and 72 genes (0.35% of all genes expressed) showing significant expression differences in the duodenum and colon, respectively (log2 fold change ≥ 1 or log2 fold change ≤ -1; P ≤ .01) (Figure 2 E). By contrast, comparing duodenum vs ileum using the same method showed 1777 (9.32% of all genes expressed) differentially expressed genes (Figure 2 E). Organoid generation from cryopreserved biopsy specimens was robust and patients ranged in age from 2 to 70 years and included both males and females. We kept a subset of samples frozen for times ranging from 3 days to 10 months with successful organoid cultures derived in each case (Figure 1 B–D). To test the idea that cryopreserved biopsy specimens can be shipped long distances, 2 patient samples were cryopreserved at the University of Michigan and shipped on dry ice to Baylor College of Medicine, where organoid cultures were established successfully from both patients (Figure 1, samples 104 and 105). Biopsy specimens from 3 patients were shipped on dry ice from Ankara, Turkey, to the University of Michigan, where organoids were established successfully from these samples (Figure 1 B, samples 132, 133, and 134). These results indicate that GI tissue can be cryopreserved, shipped long distances, and cultured from a diverse human demographic. In summary, we have shown human biopsy specimens from multiple regions of the GI tract can be cryopreserved and, upon thawing, used to establish long-term organoid cultures. We speculate that this technically simple process will be adaptable to biopsy specimens from other organs/tissues, and that the major obstacle will be the identification of tissue-specific recovery conditions after thawing. Frozen tissue samples now can be shipped across the globe, effectively freeing patients, hospitals, clinics, researchers, and diagnostics laboratories from the necessity of geographic proximity.
                Bookmark

                Author and article information

                Contributors
                Journal
                Cell Mol Gastroenterol Hepatol
                Cell Mol Gastroenterol Hepatol
                Cellular and Molecular Gastroenterology and Hepatology
                Elsevier
                2352-345X
                2019
                02 March 2019
                : 7
                : 3
                : 679-681
                Affiliations
                [1]Department of Molecular and Integrative Physiology, The University of Michigan, Ann Arbor, Michigan
                [2]Janssen Research and Development, Spring House, Pennsylvania
                Author notes
                [] Correspondence Address correspondence to: Linda C. Samuelson, PhD, Department of Molecular and Integrative Physiology, The University of Michigan, Ann Arbor, Michigan 48104. lcsam@ 123456umich.edu
                Article
                S2352-345X(19)30021-9
                10.1016/j.jcmgh.2019.02.005
                6477475
                30831074
                a1966c00-7cd8-4f3c-91a8-561f9b87c5c8
                © 2019 The Authors

                This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

                History
                Categories
                Commentary

                Comments

                Comment on this article