Cmgh Review + Engineered Human Gi Cultures to Study the Microbiome

  • Journal Listing
  • Cell Mol Gastroenterol Hepatol
  • five.v(3); 2018 Mar
  • PMC5904028

Cell Mol Gastroenterol Hepatol. 2018 Mar; five(3): 241–251.

Engineered Human Gastrointestinal Cultures to Written report the Microbiome and Infectious Diseases

Received 2017 Sep 20; Accepted 2017 Dec iv.

Abstract

New models to study the intestine are key to understanding intestinal diseases and developing novel treatments. Intestinal organ-like culture systems (organoids and enteroids) have substantially avant-garde the study of the human gastrointestinal tract. Stem cell–derived cultures produce self-organizing structures that contain the multiple differentiated abdominal epithelial cell types including enterocytes, goblet, Paneth, and enteroendocrine cells. Agreement host–microbial interactions is one expanse in which these cultures are expediting major advancements. This review discusses how organoid and enteroid cultures are biologically and physiologically relevant systems to investigate the effects of commensal organisms and study the pathogenesis of human infectious diseases. These cultures can be established from many donors and they retain the genetic and biologic backdrop of the donors, which can lead to the discovery of host-specific factors that affect susceptibility to infection and result in personalized approaches to treat individuals. The continued development of these cultures to incorporate more facets of the gastrointestinal tract, including neurons, allowed cells, and the microbiome, will unravel new mechanisms regulating host–microbial interactions with the long-term goal of translating findings into novel preventive or therapeutic treatments for gastrointestinal infections.

Keywords: Enteroids, Organoids, Infections, Microbiome, Host-Microbial Interactions

Abbreviations used in this paper: HBGA, histo-blood group antigen; IFN, interferon; IL, interleukin; 3D, three-dimensional

Summary

Enteroid and organoid cultures accept revolutionized the written report of homo small intestinal biology and are outstanding models to explore relationships between microbial organisms and the homo host. Increasing the complexity of these cultures will contribute to a meliorate understanding of gut function.

The human minor intestine is a complex organ with an epithelial surface that provides a protective bulwark confronting a diverse and hostile environment. New models to study the intestine volition be a key component in developing novel treatments for many intestinal diseases. Although tissue engineering of the homo small intestine is still in its infancy and no in vitro systems are however available to study the human intestine as a whole, intestinal organ-like culture systems with long-term chapters for expansion in vitro have given rise to elegant models of the gastrointestinal epithelium. These cultures surpass and overcome limitations of the colonic adenocarcinoma cell lines used previously that retain altered cellular pathways of transformed cells and practice not reflect cellular or host diversity. 2 pivotal technologies essentially advanced production of novel human minor intestinal cultures derived from different sources (Figure 1). Tissue-derived stem cells isolated from human abdominal biopsies or surgical specimens can exist differentiated into epithelial only cultures.i, 2 Human pluripotent stem cells (either embryonic in origin or reprogrammed somatic cells) can be directed to give rise to intestinal epithelial cultures associated with mesenchyme.three In both systems, the stem cells produce self-organizing cultures that incorporate the multiple differentiated intestinal epithelial jail cell types including enterocytes, goblet, Paneth, and enteroendocrine cells. In 2012, the intestinal stem prison cell consortium proposed nomenclature to distinguish epithelial only (enteroids) from epithelial/mesenchymal (organoids) cultures, and nosotros use this terminology from here on.iv These cultures take launched a new era in the report of homo gastrointestinal tract biology, physiology, and pathophysiology.

An external file that holds a picture, illustration, etc.  Object name is gr1.jpg

Derivation of organoids and enteroids and modes of exposure to microbial organisms in three-dimensional or 2-dimensional format. Modified from Mills and Estes.67

This review discusses how enteroid and organoid cultures are biologically and physiologically relevant systems to investigate the effects of commensal organisms and written report the pathogenesis of human being infectious diseases. The pocket-size intestinal epithelium is the main site of replication for many microbial communities and gastrointestinal pathogens, yet fundamental noesis of the molecular mechanisms of human abdominal epithelial prison cell–microbial interactions remains limited. Agreement host-microbial interactions is ane surface area where these cultures are expediting major advancements. The unique interactions between human host cells and benign and pathogenic organisms are not reproduced in animal systems and unicellular transformed jail cell lines. Primary prison cell cultures and tissue explants can be used to pattern the human intestine, but their utilize is limited to short-term culture and continued access to the master textile. Past contrast, enteroid and organoid cultures are revealing answers to longstanding questions concerning how microbial organisms collaborate with and induce responses in the human being intestinal epithelium. Human-specific susceptibility factors including genetic risk factors, age, gender, and ethnicity that touch infection and pathogenesis can be elucidated. Both enteroid and organoid cultures grown from many donors resulting in "banks" volition lead the way to personalized approaches to care for individuals who develop disease. The connected development of these cultures to include more facets of the gastrointestinal tract will facilitate farther discoveries into host-microbial interactions with the long-term goal of translating findings into novel preventive or therapeutic treatments for gastrointestinal infections.

Organization of the Pocket-size Intestine

The human modest intestine consists of different prison cell types and regional specialization that contribute to its biology and physiology. The apical side of the polarized epithelium faces the lumen of the gastrointestinal tract. The epithelium not but functions equally a barrier to the outside surround but also has secretory and absorptive capabilities. Cellular heterogeneity is the basis for these different functions every bit goblet, enteroendocrine, and Paneth cells secrete mucous, neuroendocrine, and antimicrobial factors, respectively, whereas the enterocytes are primarily responsible for arresting and secreting luminal components basolaterally into the portal circulation. The epithelium has 2 distinct zones, the crypt that houses the stem cells and Paneth cells and the villus that is equanimous of the differentiated secretory and absorptive cells and protrudes into the lumen of the intestine. The crypt has enormous capacity for cocky-renewal, which arises from multipotent stem cell populations that proliferate, differentiate, and migrate to become the differentiated villus cells. Adding to this complexity is regional specificity that provides unique functional backdrop in the 3 sections of small-scale intestine (duodenum, jejunum, and ileum). Underlying the epithelium is the stroma consisting of mesenchymal, immune, neuronal, polish musculus, and endothelial cells, which provide back up, both physically and biologically, for the epithelium. The circuitous nature of the modest intestine presents specific challenges for tissue engineering that range from supporting the diverse cell types that crave vastly unlike factors to survive in vitro and communication pathways between the intestine and other organs that influence cellular office, to deriving an in vitro culture platform in which the biology of the homo organ is fully recapitulated. Although not all the same fully achieved, remarkable progress is beingness made.

Engineered Man Gastrointestinal Tissue

Both organoid and enteroid cultures are equanimous of heterogeneous cell populations that recapitulate the in vivo pocket-size intestinal epithelium. They self-organize into three-dimensional (3D) structures termed mini-guts that are maintained indefinitely past using in vitro culture techniques with propagation in extracellular support matrices such equally Matrigel, a basement membrane analog that helps direct the polarity of the epithelium.3, 5, 6 The medium roofing the Matrigel-embedded organoids and enteroids contains growth factors that promote stem cell proliferation including loftier concentrations of Wnt-3a and the WNT point amplifier R-spondin, epidermal growth factor, and noggin, a secreted bone morphogenetic protein inhibitor. Withdrawal of some of these growth factors redirects the stem cells toward a differentiated jail cell state resulting in cultures that are composed of all the major differentiated jail cell types, in the appropriate frequencies, that comprise the human pocket-sized intestinal epithelium in vivo. The epithelium exhibits polarization with defined apical and basolateral surfaces, and the cultures tin generate several cell types including Thousand cells,seven, 8 Tuft cells,9 and Paneth cells5 that previously could not be generated in vitro. Furthermore, these epithelial cultures retain like physiological functions of the original intestinal tissue.10, 11, 12 For example, they exhibit functional peptide and ion transport and secrete chloride in response to known secretagogues.3, thirteen, fourteen Most important, the stem cells showroom a low frequency of mutation,5 and these cultures can be expanded indefinitely and cryopreserved, which allows long-term storage and sharing between research groups to enable biological validation. Organoid and enteroid cultures accept several advantages over transformed jail cell lines and animal models. They are able to be generated from man cells, simulate man diseases, tin can be used to study genetic effects on abdominal biology, are models that will theoretically lead to the establishment of personalized medicine approaches for clinical handling, and potentially will exist used in regenerative medicine. In addition, they can be genetically manipulated by using transfection, electroporation, and lentivirus transduction.13, 15, sixteen These new experimental tools are applicable for both basic and translational enquiry.

Small Intestinal Organoids

Intestinal organoids have several advantages as an experimental system. Considering of their origin from induced pluripotent stem cells, the institution of organoid cultures does not rely on the availability of intestinal tissue. In add-on, organoid cultures accept the advantage of also containing mesenchymal cells that interact closely with the epithelium.3, 17 This mixture of mesenchymal-epithelial cells mimics what is seen in vivo where the mesenchyme is a key factor in controlling the growth, differentiation, and functionality of the epithelium. Although the architectural complexity of the organoid cultures approaches what is seen in vivo, full maturation of the epithelium is non seen, and heterogeneous differentiation between organoids is often observed.three, 18, nineteen Transcriptional profiling analyses indicates that homo organoids exhibit a more fetal phenotype compared with mature adult pocket-size intestine,20, 21, 22 and the establishment of definitive regions has been limited to broadly proximal and distal pocket-sized intestine.23 Yet, further maturation is achieved when organoids are implanted under the kidney capsule of a mouse where they recruit mouse stromal factors that help the organoids develop into a more adult-like phenotype.17, 21

Small Intestinal Enteroids

Enteroid cultures have different advantages equally an experimental organisation. These cultures exhibit cellular differentiation and regional stem prison cell specificity like to what is found in vivo.2 Villus-like cystic structures bud off from a single primal lumen, with stem cells and Paneth cells at the lesser of the structure with the more mature enterocytes, goblet, and enteroendocrine cells found closer to the lumen. Enteroids retain regional specificity based on the origin of the tissue (duodenum, jejunum, ileum) and are physiologically active, exhibiting absorptive and secretory part, and the apical surface is coated with mucin secreted past the goblet cells.24 Withal, these cultures lack mesenchymal cells, and thus defined exogenously added factors are necessary to support their growth. Enteroids can be used to study a number of different somatic disorders, and many are able to replicate disease phenotypes such as cystic fibrosis25 and cancer2 in vitro. One of the challenges in working with both the 3D organoids and enteroids is admission to the apical/luminal side of the cells that faces inward in the 3D structure. Several methods tin can be used to gain admission to the interior compartment of the enclosed 3D construction including microinjection into the lumen directly or mechanical disruption of the enteroids.26, 27 Enteroids besides tin be plated as two-dimensional cultures in monolayers on culture dishes or on Transwells in which epithelial barrier function can be assessed28, 29 (Effigy 1). In addition, monolayer cultures allow ease of access to both the apical and basolateral sides of the epithelium.29, thirty, 31

Using Gastrointestinal Organoids and Enteroids to Written report Host-Microbial Interactions

Microbiome

The microbiome lies on the upmost side of the modest intestinal epithelium and has a significant bear on on gastrointestinal physiology and host health. Several diseases have been linked to alterations in the pocket-sized abdominal microbiome including inflammatory bowel disease32 and obesity,33 suggesting there are of import molecular and cellular mechanisms that are regulated by commensal organisms. In addition to directly interacting with receptors on the apical surface of the epithelium,34 the commensal organisms produce metabolic products that regulate host physiology35 and provide a means of communication to other organs in the body.36, 37 The microbiome, called a "new organ," provides a variety of functions such every bit maintaining intestinal barrier integrity through regulation of tight junction proteins,38 modulating the host immune system,39 affecting host lipid metabolism,xl and inhibiting colonization with pathogenic organisms.40 The localization and density of microbial communities vary within regions of the minor intestine (duodenum, jejunum, ileum) as well as from the lumen to the apical surface of the epithelium.41, 42 Although high-throughput Deoxyribonucleic acid sequencing has made strides in determining the quantity and identity of the microbial communities, the molecular effects of specific organisms on the epithelium still remain undefined.

Enteroids and organoids offering many advantages when studying the microbiota. Although systems such as humanized and gnotobiotic mice accept provided the foundation for understanding the bear upon of the microbiome in the context of healthy and affliction states, there are pregnant differences in physiology, anatomy, and microbial limerick between these animals and the human intestine.43 Better approaches are needed to brand advances in understanding how human physiology is modulated past microbial communities. Host-specific microbiota grown from a single individual, cultivated classically or using novel bioreactors,44 and paired with epithelial cultures from the same individual's intestine enables the study of patient-specific interventions for many gastrointestinal diseases. Despite these advantages, at that place have been few to no studies utilizing these civilization systems to explore the mechanisms past which individual bacteria impact human epithelial physiology. One barrier is the anaerobic requirement of many bacteria. Anaerobic organisms injected into the lumen of 3D organoids have survived for 12 hours, suggesting the oxygen weather condition in 3D cultures may facilitate interactions between commensal bacteria and the small intestine epithelium.45 Commensal Escherichia coli replicated, were completely contained within the lumen, and did not cause harm after microinjection into the lumen of organoids.46 Mucin was proposed as a defense force mechanism to contain the non-pathogenic bacteria. Transcriptional analysis of the leaner-exposed enteroids found upregulation of genes associated with gastrointestinal maturation.

Pathogens

In contrast to the organisms in the lumen of the intestine that maintain a homeostatic relationship with an intact intestinal epithelium and provide beneficial functions, pathogenic organisms employ the epithelium equally a site of attachment, invasion, and replication, leading to significant pathology and disease in the small intestine. Gastrointestinal infections (viral, bacterial, and parasitic) touch on betwixt 76 million (foodborne disease) and 135 million people each yr.47 The development of treatments for gastrointestinal infections has been limited by a lack of understanding of the pathogenesis of many human intestinal organisms. Significant numbers of human small intestinal infections are non replicated well in vitro or in animals, limiting the identification and characterization of pathogen receptors, cellular entry pathways, mechanisms of epithelial barrier disruption, and the epithelial response to infection. Other areas that could benefit from increased research include studies on the induction of antimicrobial factors within the epithelium that prevent infection from occurring and the role of homo genetic factors that determine susceptibility to infection and affliction. New systems will advance the understanding of biological interactions betwixt the epithelium, commensal leaner, and pathogenic organisms and their relevance to man health.

Small abdominal organoids and enteroids are new approaches to accost some of the outstanding questions for many human small-scale intestinal pathogens. Because of the sheer numbers of cultures that can be established from individuals of dissimilar genetic backgrounds, organoids and enteroids can accost genetically controlled susceptibility of human cells to infection with homo pathogens. Factors influencing entry of the pathogen into the intestinal epithelium and cell types important for replication can be determined and evaluated. A better understanding of signaling pathways that comprise the human innate immune response of the epithelium, which is the first immune response triggered by pathogenic infection, is attainable in these systems. These pathways are important targets in the development of therapeutics to treat infected individuals.

Both organoids and enteroids have been used to study the pathogenesis of several enteric viruses (Table 1) including human being rotavirus, a mutual pocket-sized intestinal viral pathogen that causes dehydrating vomiting and diarrhea in young children.48 Initially, organoid cultures and later enteroid cultures were shown to back up direct growth of human being rotavirus strains.xi, 49, 50, 51 As expected, enterocytes are the predominant prison cell type targeted past the virus in either culture organization. Unexpectedly, rotavirus as well infected mesenchymal cells in the organoids, demonstrating a previously unknown prison cell tropism. Enteroendocrine cells also are infected in the enteroid cultures, and rotavirus-infected or viral enterotoxin-treated enteroid cultures showroom swelling indicative of a pathophysiological response likely reflecting chloride secretion that is associated with the illness symptom of diarrhea.11 Recent studies demonstrate that susceptibility to rotavirus infection and response to rotavirus vaccines may be mediated by host genetic factors such equally the expression of histo-blood group antigens (HBGAs).52 Enteroids from different patients support varying levels of rotavirus replication mimicking differences in susceptibility between different individuals in the population. Exposure to one licensed rotavirus vaccine resulted in lower vaccine replication in specific enteroids when compared with other enteroids, indicating putative host factors that influence vaccine immunogenicity and efficacy.11 Organoid and enteroid cultures provide a new tool in which epidemiologic data on host susceptibility factors can exist tested and validated. I explanation for the variation in host response may be the innate allowed response mounted by the epithelium. Enteroids answer to human rotavirus infection with a robust innate immune response, with the predominant transcriptional pathway induced by human rotavirus infection beingness a type III interferon (IFN) response that activates IFN-stimulated genes.51 Enteroids have been used to model antiviral effects of both IFNs and ribavirin against patient-derived rotavirus strains.50 Sensitivity to handling with these antivirals varied substantially on the basis of enteroid line.50 The importance of this response in limiting epithelial infection remains to exist elucidated. Enteroids also can be used to evaluate antivirals for the treatment of rotavirus-infected children and chronically infected transplant patients,53, 54 and may provide new preclinical assays for understanding vaccine attenuation.

Tabular array 1

Insights Into Man Viral Infections of Homo Abdominal Enteroids

Virus Enteroid type Strains Cells infected Responses Innate responses Reference
Rotavirus HIOs Man rotavirus replication Enterocytes, mesenchymal cells 49
Rotavirus Differentiated HIEs from adults, all small-scale intestinal segments in 3D cultures Human rotavirus replicates more efficiently than animate being rotavirus Enterocytes, enteroendocrine cells; not stem cells Swelling induced by viral infection and enterotoxin handling Predominant type III IFN response 6, 11, 15
Rotavirus HIEs from adults Human being rotavirus replication Antiviral testing Selected innate response genes induced 50, 53
Human norovirus Differentiated HIEs from adults, all pocket-size intestinal segments on monolayers Multiple human norovirus strains replicate; strain-specific requirements for replication; some require bile Enterocytes; not stem cells Inactivation and neutralization tested Host-specific susceptibility to infection based on host HBGA expression 6, xxx
Enteroviruses HIEs from human fetal small intestine in 3D cultures Strain-specific differences in relative replication efficiency with EV11 and CBV high and EV71 low Strain-specific responses 56
Echovirus 11 Good infections; cpe; cell death; mislocalization of occludin; infectious virus produced in levels similar to Caco-2 cells Enteroendocrine cells; non goblet cells Differential induction of 350 transcripts; cytokines, chemokines; IFN-stimulated genes 56
Coxsackievirus B Good infections past immunofluorescence and viral RNA Differential consecration of 13 transcripts 56
Enterovirus 71 Lower levels of replication No significant induction of transcripts detected 56

Human being noroviruses crusade 699 1000000 episodes of gastroenteritis and 219,000 deaths globally every year.55 For decades petty progress had been made into preventive or therapeutic modalities for norovirus disease. In a landmark discovery, homo noroviruses, previously uncultivatable in transformed epithelial cell cultures and small fauna models, were demonstrated to replicate in human enteroid cultures.thirty Robust and reproducible in vitro cultivation of human noroviruses in enterocytes was seen for multiple virus strains in differentiated cultures from all segments of the small intestine. In addition, norovirus replication in enteroids mimics epidemiologic information on differences in host susceptibility to infection; enteroids derived from individuals who lack a functional fucosyltransferase 2 enzyme to limited specific HBGA receptors associated with susceptibility to infection did non support virus replication.30 Unexpectedly, strain-specific requirements were discovered, with the addition of bile being required to support replication in enteroids for some norovirus strains. This organisation can exist used to evaluate and develop antivirals, inactivating agents, and to sympathize virus infectivity and replication. These studies demonstrate the potential of human being enteroid cultures for studying and understanding the host factors and pathogenesis of previously not-cultivatable pathogens.

Other enteric viruses classified as enteroviruses, such equally echovirus xi, coxsackievirus B, enterovirus 71, and poliovirus, infect and replicate to different levels in human being enteroid cultures produced from human being fetal small intestine.56 Transcriptional analysis of echovirus 11–infected enteroids showed differential induction of 350 transcripts, whereas only 13 transcripts were induced after coxsackievirus B infection. The transcripts induced by echovirus included immune factors such equally cytokines, chemokines, and IFN-stimulated genes, possibly explaining lower levels of replication compared with coxsackievirus B. Like rotavirus, in addition to infecting enterocytes, echovirus eleven infected enteroendocrine cells. These studies contrast previous work in transformed cell lines that suggested enterovirus did not infect the intestinal epithelium but rather used it as a conduit to reach other target cells.57 Taken together, these studies of viral infections demonstrate human enteroid cultures provide important insights into viral pathogenesis that are not replicated in transformed cell lines.

The pathogenesis of human bacterial infections likewise has been studied in organoid and enteroid systems. Bacterial pathogens cause meaning foodborne gastroenteritis, and many cause their effects in the small intestine. The transcriptional response of organoid cultures was examined later microinjection with salmonella strains58and included cytokine responses such equally interleukin (IL)-23A, which is linked to protection of the abdominal barrier,59 and IL8, IL1B, tumor necrosis factor, and CXCL2, which are proinflammatory. Goblet and enteroendocrine cell-associated genes were besides upregulated. Imaging approaches documented the attachment of salmonella on the apical surface of the organoid epithelium and classic vacuoles containing the pathogen. Enteroids were used to demonstrate adherence patterns when exposed to different strains of pathogenic E. coli.29 Similar to rotavirus, 3D human enteroids responded to luminal microinjection of enterotoxigenic E coli by swelling representing secretion of fluid.60 The Shiga toxins produced by enterohemorrhagic E coli stimulate toxin micropinocytosis and host actin remodeling.31 Studies using non-foodborne bacteria such as Clostridium difficile that cause infectious diarrhea demonstrated disruption of the epithelial bulwark, alterations in transporter expression related to diarrhea, loss of polarity, and reduction of mucous production when organoid cultures were exposed to either the pathogen or its toxins.45, 61, 62 Vibrio cholera toxin elicited a faster increase in cyclic adenosine monophosphate in enteroids expressing blood grouping O antigens than those that expressed A antigen.63 These studies had an almost perfect parallel to clinical and epidemiologic data showing correlation betwixt O blood grouping and risk for cholera. Overall, the employ of these engineered gastrointestinal culture systems to study human bacterial infections demonstrates the potential for developing systems that permit host factors that contribute to affliction pathogenesis caused by a microbial amanuensis to exist elucidated.

Approaches using organoid or enteroid cultures to study helminth and protozoa infections accept lagged behind those used to report viruses and bacteria. Parasites are a worldwide health trouble and cause meaning morbidity and mortality affecting close to three.5 billion people. There are many difficulties in propagating these organisms in vitro including host brake and complicated life cycles. Major questions still remain for many gastrointestinal parasites including mechanisms of disease, loss of bulwark part, and identification of virulence factors. In addition, lilliputian is known nigh host factors that affect infection. Human enteroid and organoid cultures accept the potential to brand major inroads into addressing some of these questions as they pertain to parasite infections. Once the systems are more established, these models could be used to screen new anti-parasitic drugs.

Challenges to Existing Engineered Gastrointestinal Tissues

Organoid and enteroid cultures are simple to maintain, offer the unique adequacy of examining susceptibility to infections within certain genetic populations, and let comparative analysis betwixt these cultures and man tissue. Most importantly, these systems offering pathways to personalized medicine. Several areas to better these systems for studies involving beneficial and pathogenic organisms include adding physical system, allowed cells, and neuronal signaling. Moving forward, the development of organoids and enteroid cultures will simply serve to enhance these methods for use in studying the microbiome and infectious pathogens (Figure two).

An external file that holds a picture, illustration, etc.  Object name is gr2.jpg

Future platform development of enteroids and organoids to facilitate studies with microbial organisms.

Advances in culturing suggest that proper temporal organization in a crypt-villus–similar unit of measurement in a two-dimensional format is an attainable goal12 and one that will be necessary to consider when examining the interactions of many microbial organisms with the epithelium. Currently, organoids and enteroids lack the definitive catacomb-villus structure in monolayer format that facilitates the natural route of exposure to the upmost surface. Of importance as well will be incorporation of the mesenchyme (enteroids) and the mucosal muscular layer, the muscularis mucosa (enteroids and organoids), both of which participate in bidirectional signaling with the epithelium. Another hurdle in culturing these tissues is controlling the variability of reagents. Matrigel and non-commercially produced growth factors are expensive, have lot-to-lot variation, and are not fully defined. As culture weather condition become more sophisticated, the development of defined reagents will ensure reproducibility of circuitous cellular systems.

Organoids and enteroids also lack components of the immune system. The complex interaction between the epithelium, the immune system, and microbes is key to agreement small-scale intestinal physiology. Co-cultures of enteroids with macrophages take been successfully established,64 resulting in enhancement of epithelial barrier function and enteroid maturation. A coordinated response of these co-cultures to exposure with enterotoxigenic and enteropathogenic East coli infections was documented by observing intraepithelial macrophage projections, efficient phagocytosis, and stabilized barrier function. Organoids cultured with neutrophils restate innate cellular responses when exposed to Shiga toxin–producing Eastward coli including cytokine response, loss of epithelial integrity, and activation of stress responses that involve reactive oxygen species.46 These studies provide critical testify that illustrates the importance of incorporating the immune system into these cultures in the context of studying microbial organisms.

The enteric nervous system also plays a cardinal function in the physiology of infections through its regulation of movement, secretion, fluid period across the epithelium, and epithelial bulwark integrity.65 The myenteric and submucosal plexi have networks of neurons and glia that extend throughout the various intestinal compartments including where they play a disquisitional function in modulating epithelial function. Because of its location and function, the enteric nervous system is thought to attune responses to insults by triggering pathways that result in diarrhea and vomiting. Withal, the dissection of these contributions has been difficult, with no current way to study the enteric nervous arrangement in vitro. Recent work by Workman et al66 utilizing a tissue-engineering science arroyo resulted in the generation of normal abdominal enteric nervous arrangement within an organoid civilisation. Importantly, these neurons functionally regulated waves of propagating contractions. This system shows tremendous promise to dissect the molecular aspects of enteric nervous organisation–epithelial interactions with both microbiome and pathogenic organisms.

Every bit advances are fabricated in organoid and enteroid cultures, validation will exist an important consideration. Variability is an inevitable result of using human tissue every bit the source of these cultures. Transcriptional analyses of multiple jejunal enteroid cultures revealed that the transcriptional profile clusters first by individual. However, infection of these cultures with human rotavirus revealed a singled-out and mutual transcriptional response,51 indicating that amid the man variability, common biological responses exist that are about likely relevant to agreement host-microbial interactions. Thus research directed toward determining host responses to microbial pathogens or commensals will benefit from validation across multiple organoid or enteroid lines. Analyses in cultures representing biologic replicates will be paramount to discovering and validating global host factors that can and then be farther tested in patients. Well-characterized biobanks of homo intestinal organoids and enteroids generated from multiple individuals and shared among researchers volition exist required and volition provide unique tools to define common host genes and responses involved in host-microbial interactions.

Summary

It is evident that the use of engineered human gastrointestinal organoid and enteroid models that recapitulate the complication and cellularity of the small intestine is facilitating discoveries and providing new, cardinal cognition on normal homo abdominal physiology and pathophysiology and how the host responds to microbial pathogens and commensals. Application of these models ranges from patient-specific personalized medicine studies to pre-clinical studies for vaccines and therapeutics where the bear on of differences in host susceptibility and response is a critical parameter for response cess. Pregnant efforts continue to exist made in the development of complex intestinal culture systems and volition contribute to the long-term goal of developing integrated engineered human tissue models and human organs on a chip to understand gut function in wellness and disease.

Footnotes

Conflicts of interest The authors disclose the following: M.K. Estes is named as an inventor on patents related to cloning of the Norwalk virus genome and has received consultant'southward fees from Takeda Vaccines, Inc. The remaining authors disclose no conflicts.

Funding Supported by grants U18-TR000552, UH3-TR00003, U19-AI116497, RO1-AI080656, U01-DK103168, and P30-DK56338 from the National Institutes of Health and the Nutrient Research Initiative Competitive grant 2011-68003-30395 from the U.Southward. Department of Agriculture, National Plant of Food and Agriculture.

References

ane. Sato T., Vries R.G., Snippert H.J., van de Wetering M., Barker N., Stange D.Due east., van Es J.H., Abo A., Kujala P., Peters P.J., Clevers H. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009;459:262–265. [PubMed] [Google Scholar]

2. Sato T., Stange D.E., Ferrante M., Vries R.G., van Es J.H., van den Brink S., Van Houdt W.J., Pronk A., Van Chiliad.J., Siersema P.D., Clevers H. Long-term expansion of epithelial organoids from homo colon, adenoma, adenocarcinoma, and Barrett's epithelium. Gastroenterology. 2011;141:1762–1772. [PubMed] [Google Scholar]

3. Spence J.R., Mayhew C.N., Rankin S.A., Kuhar Thou.F., Vallance J.Due east., Tolle K., Hoskins E.Due east., Kalinichenko V.V., Wells Southward.I., Zorn A.K., Shroyer N.F., Wells J.K. Directed differentiation of human pluripotent stalk cells into intestinal tissue in vitro. Nature. 2011;470:105–109. [PMC free article] [PubMed] [Google Scholar]

4. Stelzner 1000., Helmrath M., Dunn J.C., Henning S.J., Houchen C.West., Kuo C., Lynch J., Li L., Magness S.T., Martin M.G., Wong M.H., Yu J., NIH Intestinal Stalk Jail cell Consortium A nomenclature for intestinal in vitro cultures. Am J Physiol Gastrointest Liver Physiol. 2012;302:G1359–G1363. [PMC gratuitous commodity] [PubMed] [Google Scholar]

5. Sato T., van Es J.H., Snippert H.J., Stange D.Due east., Vries R.G., van den Born M., Barker N., Shroyer N.F., van de Wetering Chiliad., Clevers H. Paneth cells constitute the niche for Lgr5 stem cells in intestinal crypts. Nature. 2011;469:415–418. [PMC free article] [PubMed] [Google Scholar]

6. Zou W.Y., Blutt S.E., Crawford S.Eastward., Ettayebi 1000., Zeng X.Fifty., Saxena K., Ramani S., Karandikar U.C., Zachos N.C., Estes One thousand.K. Human intestinal enteroids: new models to written report gastrointestinal virus infections. Methods Mol Biol. 2017;15:ane–xix. [PMC gratis article] [PubMed] [Google Scholar]

7. de Lau W., Kujala P., Schneeberger Thou., Middendorp South., Li 5.S., Barker Northward., Martens A., Hofhuis F., DeKoter R.P., Peters P.J., Nieuwenhuis E., Clevers H. Peyer'due south patch M cells derived from Lgr5(+) stem cells require SpiB and are induced by RankL in cultured "miniguts" Mol Cell Biol. 2012;32:3639–3647. [PMC gratis article] [PubMed] [Google Scholar]

8. Rouch J.D., Scott A., Lei Due north.Y., Solorzano-Vargas R.S., Wang J., Hanson E.M., Kobayashi Thousand., Lewis Grand., Stelzner Yard.G., Dunn J.C., Eckmann Fifty., Martin M.One thousand. Evolution of functional microfold (M) cells from abdominal stem cells in master human enteroids. PLoS 1. 2016;11:e0148216. [PMC complimentary article] [PubMed] [Google Scholar]

9. Basak O., Beumer J., Wiebrands Yard., Seno H., van O.A., Clevers H. Induced quiescence of Lgr5+ stem cells in intestinal organoids enables differentiation of hormone-producing enteroendocrine cells. Cell Stalk Cell. 2017;xx:177–190. [PubMed] [Google Scholar]

10. Liu J., Walker North.M., Melt M.T., Ootani A., Clarke Fifty.50. Functional Cftr in catacomb epithelium of organotypic enteroid cultures from murine pocket-size intestine. Am J Physiol Prison cell Physiol. 2012;302:C1492–C1503. [PMC free article] [PubMed] [Google Scholar]

11. Saxena Yard., Blutt Southward.E., Ettayebi Yard., Zeng X.Fifty., Broughman J.R., Crawford S.E., Karandikar U.C., Sastri N.P., Conner M.Due east., Opekun A.R., Graham D.Y., Qureshi West., Sherman V., Foulke-Abel J., In J., Kovbasnjuk O., Zachos N.C., Donowitz M., Estes M.K. Man intestinal enteroids: a new model to written report human rotavirus infection, host restriction, and pathophysiology. J Virol. 2015;xc:43–56. [PMC free commodity] [PubMed] [Google Scholar]

12. Wang Y., Gunasekara D.B., Reed M.I., DiSalvo Grand., Bultman S.J., Sims C.Due east., Magness S.T., Allbritton Northward.L. A microengineered collagen scaffold for generating a polarized crypt-villus architecture of human being small intestinal epithelium. Biomaterials. 2017;128:44–55. [PMC free article] [PubMed] [Google Scholar]

13. Schwank G., Koo B.K., Sasselli V., Dekkers J.F., Heo I., Demircan T., Sasaki N., Boymans S., Cuppen Eastward., van der Ent C.K., Nieuwenhuis E.Due east., Beekman J.K., Clevers H. Functional repair of CFTR past CRISPR/Cas9 in intestinal stem prison cell organoids of cystic fibrosis patients. Jail cell Stem Prison cell. 2013;13:653–658. [PubMed] [Google Scholar]

14. Foulke-Abel J., In J., Yin J., Zachos North.C., Kovbasnjuk O., Estes M.K., de J.H., Donowitz M. Human being enteroids as a model of upper pocket-size intestinal ion transport physiology and pathophysiology. Gastroenterology. 2016;150:638–649. [PMC gratis article] [PubMed] [Google Scholar]

15. Koo B.K., Stange D.E., Sato T., Karthaus West., Farin H.F., Huch G., van Es J.H., Clevers H. Controlled factor expression in primary Lgr5 organoid cultures. Nat Methods. 2011;nine:81–83. [PubMed] [Google Scholar]

16. Yilmaz O.H., Katajisto P., Lamming D.West., Gultekin Y., Bauer-Rowe K.Eastward., Sengupta S., Birsoy Yard., Dursun A., Yilmaz V.O., Selig K., Nielsen G.P., Mino-Kenudson M., Zukerberg Fifty.R., Bhan A.K., Deshpande 5., Sabatini D.M. mTORC1 in the Paneth cell niche couples intestinal stem-cell function to calorie intake. Nature. 2012;486:490–495. [PMC free article] [PubMed] [Google Scholar]

17. Watson C.L., Mahe Grand.Grand., Munera J., Howell J.C., Sundaram N., Poling H.M., Schweitzer J.I., Vallance J.Eastward., Mayhew C.N., Sun Y., Grabowski G., Finkbeiner S.R., Spence J.R., Shroyer North.F., Wells J.M., Helmrath M.A. An in vivo model of human small intestine using pluripotent stem cells. Nat Med. 2014;20:1310–1314. [PMC free commodity] [PubMed] [Google Scholar]

eighteen. McCracken K.W., Cata E.One thousand., Crawford C.M., Sinagoga Yard.50., Schumacher M., Rockich B.East., Tsai Y.H., Mayhew C.N., Spence J.R., Zavros Y., Wells J.M. Modelling homo evolution and disease in pluripotent stem-prison cell-derived gastric organoids. Nature. 2014;516:400–404. [PMC free article] [PubMed] [Google Scholar]

19. Takasato K., Er P.X., Chiu H.S., Maier B., Baillie G.J., Ferguson C., Parton R.Thou., Wolvetang E.J., Roost M.S., Chuva de Sousa Lopes S.M., Piddling M.H. Kidney organoids from human iPS cells contain multiple lineages and model human nephrogenesis. Nature. 2015;526:564–568. [PubMed] [Google Scholar]

20. McCracken Chiliad.W., Howell J.C., Wells J.Thousand., Spence J.R. Generating human intestinal tissue from pluripotent stem cells in vitro. Nat Protoc. 2011;six:1920–1928. [PMC free article] [PubMed] [Google Scholar]

21. Finkbeiner S.R., Hill D.R., Altheim C.H., Dedhia P.H., Taylor Thousand.J., Tsai Y.H., Chin A.M., Mahe Chiliad.M., Watson C.50., Freeman J.J., Nattiv R., Thomson Thousand., Klein O.D., Shroyer N.F., Helmrath M.A., Teitelbaum D.H., Dempsey P.J., Spence J.R. Transcriptome-broad analysis reveals hallmarks of human intestine development and maturation in vitro and in vivo. Stem Prison cell Reports. 2015;4:1140–1155. [PMC free commodity] [PubMed] [Google Scholar]

22. Aurora M., Spence J.R. hPSC-derived lung and intestinal organoids every bit models of human fetal tissue. Dev Biol. 2016;420:230–238. [PMC free article] [PubMed] [Google Scholar]

23. Tsai Y.H., Nattiv R., Dedhia P.H., Nagy M.S., Chin A.M., Thomson M., Klein O.D., Spence J.R. In vitro patterning of pluripotent stem jail cell-derived intestine recapitulates in vivo human evolution. Development. 2017;144:1045–1055. [PMC free article] [PubMed] [Google Scholar]

24. Middendorp Southward., Schneeberger K., Wiegerinck C.L., Mokry M., Akkerman R.D., van Westward.S., Clevers H., Nieuwenhuis E.Eastward. Adult stalk cells in the small intestine are intrinsically programmed with their location-specific function. Stalk Cells. 2014;32:1083–1091. [PubMed] [Google Scholar]

25. Dekkers J.F., Wiegerinck C.50., de Jonge H.R., Bronsveld I., Janssens H.M., de Winter-de Groot Thousand.1000., Brandsma A.Thou., de Jong N.West., Bijvelds One thousand.J., Scholte B.J., Nieuwenhuis Due east.Due east., van den Brink S., Clevers H., van der Ent C.K., Middendorp S., Beekman J.M. A functional CFTR assay using primary cystic fibrosis intestinal organoids. Nat Med. 2013;xix:939–945. [PubMed] [Google Scholar]

26. Dutta D., Heo I., Clevers H. Disease modeling in stem cell-derived 3D organoid systems. Trends Mol Med. 2017;23:393–410. [PubMed] [Google Scholar]

27. Bartfeld S., Clevers H. Organoids as model for infectious diseases: culture of human and murine stomach organoids and microinjection of Helicobacter pylori. J Vis Exp. 2015;105:53359. [PMC costless commodity] [PubMed] [Google Scholar]

28. Moon C., VanDussen K.L., Miyoshi H., Stappenbeck T.S. Development of a primary mouse intestinal epithelial cell monolayer civilization organization to evaluate factors that modulate IgA transcytosis. Mucosal Immunol. 2014;seven:818–828. [PMC gratis article] [PubMed] [Google Scholar]

29. VanDussen One thousand.L., Marinshaw J.Thou., Shaikh North., Miyoshi H., Moon C., Tarr P.I., Ciorba Thousand.A., Stappenbeck T.South. Development of an enhanced human gastrointestinal epithelial culture system to facilitate patient-based assays. Gut. 2015;64:911–920. [PMC free article] [PubMed] [Google Scholar]

30. Ettayebi 1000., Crawford Due south.East., Murakami Thou., Broughman J.R., Karandikar U., Tenge V.R., Neill F.H., Blutt S.Eastward., Zeng X.50., Qu L., Kou B., Opekun A.R., Burrin D., Graham D.Y., Ramani S., Atmar R.L., Estes M.K. Replication of human noroviruses in stem prison cell-derived man enteroids. Science. 2016;353:1387–1393. [PMC gratuitous commodity] [PubMed] [Google Scholar]

31. In J., Foulke-Abel J., Zachos Due north.C., Hansen A.One thousand., Kaper J.B., Bernstein H.D., Halushka Thousand., Blutt S., Estes M.K., Donowitz M., Kovbasnjuk O. Enterohemorrhagic Escherichia coli reduce mucus and intermicrovillar bridges in human stem cell-derived colonoids. Cell Mol Gastroenterol Hepatol. 2016;2:48–62. [PMC free article] [PubMed] [Google Scholar]

32. Forbes J.D., Van D.G., Bernstein C.N. The gut microbiota in immune-mediated inflammatory diseases. Front Microbiol. 2016;7:1081. [PMC free article] [PubMed] [Google Scholar]

33. Turnbaugh P.J., Ley R.East., Mahowald Thou.A., Magrini 5., Mardis E.R., Gordon J.I. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444:1027–1031. [PubMed] [Google Scholar]

34. Earle G.A., Billings Thousand., Sigal Grand., Lichtman J.Southward., Hansson 1000.C., Elias J.E., Amieva M.R., Huang K.C., Sonnenburg J.50. Quantitative imaging of gut microbiota spatial organization. Cell Host Microbe. 2015;18:478–488. [PMC free article] [PubMed] [Google Scholar]

35. Geva-Zatorsky N., Alvarez D., Hudak J.Eastward., Reading North.C., Erturk-Hasdemir D., Dasgupta S., von Andrian U.H., Kasper D.L. In vivo imaging and tracking of host-microbiota interactions via metabolic labeling of gut anaerobic bacteria. Nat Med. 2015;21:1091–1100. [PMC free article] [PubMed] [Google Scholar]

36. Nicholson J.Grand., Holmes East., Kinross J., Burcelin R., Gibson G., Jia W., Pettersson S. Host-gut microbiota metabolic interactions. Science. 2012;336:1262–1267. [PubMed] [Google Scholar]

37. Vogt Southward.L., Pena-Diaz J., Finlay B.B. Chemic communication in the gut: effects of microbiota-generated metabolites on gastrointestinal bacterial pathogens. Anaerobe. 2015;34:106–115. [PubMed] [Google Scholar]

38. Bansal T., Alaniz R.C., Forest T.K., Jayaraman A. The bacterial signal indole increases epithelial-cell tight-junction resistance and attenuates indicators of inflammation. Proc Natl Acad Sci. 2010;107:228–233. [PMC free article] [PubMed] [Google Scholar]

39. Zelante T., Iannitti R.G., Cunha C., De Luca A., Giovannini M., Pieraccini G., Zecchi R., D'Angelo C., Massi-Benedetti C., Fallarino F., Carvalho A., Puccetti P., Romani 50. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and rest mucosal reactivity via interleukin-22. Immunity. 2013;39:372–385. [PubMed] [Google Scholar]

xl. Buffie C.G., Bucci V., Stein R.R., McKenney P.T., Ling L., Gobourne A., No D., Liu H., Kinnebrew G., Viale A., Littmann E., van den Brink M.R., Jenq R.R., Taur Y., Sander C., Cantankerous J.R., Toussaint Due north.C., Xavier J.B., Pamer Due east.G. Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile. Nature. 2015;517:205–208. [PMC gratuitous commodity] [PubMed] [Google Scholar]

41. Albenberg L., Esipova T.V., Approximate C.P., Bittinger K., Chen J., Laughlin A., Grunberg S., Baldassano R.North., Lewis J.D., Li H., Thom South.R., Bushman F.D., Vinogradov South.A., Wu G.D. Correlation between intraluminal oxygen gradient and radial sectionalisation of intestinal microbiota. Gastroenterology. 2014;147:1055–1063. [PMC free article] [PubMed] [Google Scholar]

42. Vaishnava South., Yamamoto Yard., Severson K.K., Ruhn K.A., Yu X., Koren O., Ley R., Wakeland East.K., Hooper L.V. The antibacterial lectin RegIIIgamma promotes the spatial segregation of microbiota and host in the intestine. Science. 2011;334:255–258. [PMC costless article] [PubMed] [Google Scholar]

43. Treuting P.M., Clifford C.B., Sellers R.Due south., Brayton C.F. Of mice and microflora: considerations for genetically engineered mice. Vet Pathol. 2012;49:44–63. [PubMed] [Google Scholar]

44. Auchtung J.M., Robinson C.D., Britton R.A. Cultivation of stable, reproducible microbial communities from different fecal donors using minibioreactor arrays (MBRAs) Microbiome. 2015;iii:42. [PMC complimentary article] [PubMed] [Google Scholar]

45. Leslie J.Fifty., Huang Southward., Opp J.South., Nagy M.Southward., Kobayashi M., Immature V.B., Spence J.R. Persistence and toxin production by Clostridium difficile inside human abdominal organoids result in disruption of epithelial paracellular bulwark role. Infect Immun. 2015;83:138–145. [PMC free article] [PubMed] [Google Scholar]

46. Karve Southward.South., Pradhan S., Ward D.5., Weiss A.A. Intestinal organoids model human responses to infection past commensal and Shiga toxin producing Escherichia coli. PLoS One. 2017;12:e0178966. [PMC free article] [PubMed] [Google Scholar]

47. Sandler R.Due south., Everhart J.E., Donowitz One thousand., Adams E., Cronin One thousand., Goodman C., Gemmen Due east., Shah S., Avdic A., Rubin R. The burden of selected digestive diseases in the Usa. Gastroenterology. 2002;122:1500–1511. [PubMed] [Google Scholar]

48. Greenberg H.B., Estes M.K. Rotaviruses: from pathogenesis to vaccination. Gastroenterology. 2009;136:1939–1951. [PMC gratis commodity] [PubMed] [Google Scholar]

49. Finkbeiner S.R., Zeng X.L., Utama B., Atmar R.L., Shroyer N.F., Estes Thousand.Thou. Stalk cell-derived human intestinal organoidsas an infection model for rotaviruses. MBio. 2012;3 e00159-12. [PMC free commodity] [PubMed] [Google Scholar]

50. Yin Y., Bijvelds Yard., Dang W., Xu L., van der Eijk A.A., Knipping Yard., Tuysuz Northward., Dekkers J.F., Wang Y., de J.J., Sprengers D., van der Laan L.J., Beekman J.M., Ten B.D., Metselaar H.J., de J.H., Koopmans M.P., Peppelenbosch M.P., Pan Q. Modeling rotavirus infection and antiviral therapy using main intestinal organoids. Antiviral Res. 2015;123:120–131. [PubMed] [Google Scholar]

51. Saxena K., Simon L.K., Zeng X.L., Blutt South.E., Crawford S.E., Sastri N.P., Karandikar U.C., Ajami N.J., Zachos Due north.C., Kovbasnjuk O., Donowitz Yard., Conner M.East., Shaw C.A., Estes M.K. A paradox of transcriptional and functional innate interferon responses of human intestinal enteroids to enteric virus infection. Proc Natl Acad Sci. 2017;114:E570–E579. [PMC free article] [PubMed] [Google Scholar]

52. Hu L., Crawford S.E., Czako R., Cortes-Penfield North.West., Smith D.F., Le P.J., Estes 1000.K., Prasad B.Five. Cell zipper protein VP8* of a human rotavirus specifically interacts with A-type histo-blood grouping antigen. Nature. 2012;485:256–259. [PMC gratuitous commodity] [PubMed] [Google Scholar]

53. Yin Y., Wang Y., Dang West., Xu L., Su J., Zhou X., Wang W., Felczak K., van der Laan L.J., Pankiewicz K.W., van der Eijk A.A., Bijvelds M., Sprengers D., de Jonge H., Koopmans M.P., Metselaar H.J., Peppelenbosch Yard.P., Pan Q. Mycophenolic acrid potently inhibits rotavirus infection with a loftier barrier to resistance development. Antiviral Res. 2016;133:41–49. [PubMed] [Google Scholar]

54. Yin Y., Metselaar H.J., Sprengers D., Peppelenbosch One thousand.P., Pan Q. Rotavirus in organ transplantation: drug-virus-host interactions. Am J Transplant. 2015;xv:585–593. [PubMed] [Google Scholar]

55. Cortes-Penfield Northward.W., Ramani S., Estes M.Yard., Atmar R.L. Prospects and challenges in the evolution of a norovirus vaccine. Clin Ther. 2017;39:1537–1549. [PMC costless article] [PubMed] [Google Scholar]

56. Drummond C.Grand., Bolock A.Grand., Ma C., Luke C.J., Good M., Coyne C.B. Enteroviruses infect homo enteroids and induce antiviral signaling in a cell lineage-specific manner. Proc Natl Acad Sci. 2017;114:1672–1677. [PMC free article] [PubMed] [Google Scholar]

57. Sicinski P., Rowinski J., Warchol J.B., Jarzabek Z., Gut W., Szczygiel B., Bielecki K., Koch Grand. Poliovirus type 1 enters the human being host through intestinal Thousand cells. Gastroenterology. 1990;98:56–58. [PubMed] [Google Scholar]

58. Forbester J.L., Goulding D., Vallier 50., Hannan N., Hale C., Pickard D., Mukhopadhyay Due south., Dougan G. Interaction of Salmonella enterica Serovar Typhimurium with intestinal organoids derived from human induced pluripotent stem cells. Infect Immun. 2015;83:2926–2934. [PMC complimentary article] [PubMed] [Google Scholar]

59. Macho-Fernandez E., Koroleva E.P., Spencer C.M., Tighe G., Torrado Eastward., Cooper A.M., Fu Y.X., Tumanov A.5. Lymphotoxin beta receptor signaling limits mucosal damage through driving IL-23 product by epithelial cells. Mucosal Immunol. 2015;8:403–413. [PMC costless commodity] [PubMed] [Google Scholar]

lx. Pattison A.Thou., Blomain Due east.S., Merlino D.J., Wang F., Crissey G.A., Kraft C.L., Rappaport J.A., Snook A.E., Lynch J.P., Waldman S.A. Abdominal enteroids model guanylate cyclase C-dependent secretion induced by heat-stable enterotoxins. Infect Immun. 2016;84:3083–3091. [PMC free article] [PubMed] [Google Scholar]

61. Engevik M.A., Yacyshyn M.B., Engevik K.A., Wang J., Darien B., Hassett D.J., Yacyshyn B.R., Worrell R.T. Homo Clostridium difficile infection: altered mucus production and limerick. Am J Physiol Gastrointest Liver Physiol. 2015;308:G510–G524. [PMC free article] [PubMed] [Google Scholar]

62. Wang X., Yamamoto Y., Wilson 50.H., Zhang T., Howitt B.Due east., Farrow K.A., Kern F., Ning G., Hong Y., Khor C.C., Chevalier B., Bertrand D., Wu L., Nagarajan N., Sylvester F.A., Hyams J.South., Devers T., Bronson R., Lacy D.B., Ho K.Y., Crum C.P., McKeon F., Xian W. Cloning and variation of ground land intestinal stem cells. Nature. 2015;522:173–178. [PMC costless commodity] [PubMed] [Google Scholar]

63. Kuhlmann F.M., Santhanam Southward., Kumar P., Luo Q., Ciorba M.A., Fleckenstein J.Thou. Blood group O-dependent cellular responses to cholera toxin: parallel clinical and epidemiological links to severe cholera. Am J Trop Med Hyg. 2016;95:440–443. [PMC free commodity] [PubMed] [Google Scholar]

64. Noel G., Baetz N.W., Staab J.F., Donowitz M., Kovbasnjuk O., Pasetti G.F., Zachos N.C. A primary human macrophage-enteroid co-culture model to investigate mucosal gut physiology and host-pathogen interactions. Sci Rep. 2017;7:45270. [PMC free article] [PubMed] [Google Scholar]

65. Furness J.B. The enteric nervous arrangement and neurogastroenterology. Nat Rev Gastroenterol Hepatol. 2012;9:286–294. [PubMed] [Google Scholar]

66. Workman M.J., Mahe M.One thousand., Trisno Southward., Poling H.One thousand., Watson C.L., Sundaram N., Chang C.F., Schiesser J., Aubert P., Stanley E.G., Elefanty A.Yard., Miyaoka Y., Mandegar Thou.A., Conklin B.R., Neunlist Grand., Brugmann S.A., Helmrath M.A., Wells J.M. Engineered human being pluripotent-stem-cell-derived abdominal tissues with a functional enteric nervous system. Nat Med. 2017;23:49–59. [PMC complimentary article] [PubMed] [Google Scholar]

67. Mills 1000., Estes M.M. Physiologically relevant human being tissue models for infectious diseases. Drug Discov Today. 2016;21:1540–1552. [PMC free commodity] [PubMed] [Google Scholar]


Articles from Cellular and Molecular Gastroenterology and Hepatology are provided here courtesy of Elsevier


barefootallashom.blogspot.com

Source: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5904028/

0 Response to "Cmgh Review + Engineered Human Gi Cultures to Study the Microbiome"

Post a Comment

Iklan Atas Artikel

Iklan Tengah Artikel 1

Iklan Tengah Artikel 2

Iklan Bawah Artikel