Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
1.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38732126

ABSTRACT

Enterohemorrhagic Escherichia coli (EHEC) is a critical public health concern due to its role in severe gastrointestinal illnesses in humans, including hemorrhagic colitis and the life-threatening hemolytic uremic syndrome. While highly pathogenic to humans, cattle, the main reservoir for EHEC, often remain asymptomatic carriers, complicating efforts to control its spread. Our study introduces a novel method to investigate EHEC using organoid-derived monolayers from adult bovine ileum and rectum. These polarized epithelial monolayers were exposed to EHEC for four hours, allowing us to perform comparative analyses between the ileal and rectal tissues. Our findings mirrored in vivo observations, showing a higher colonization rate in the rectum compared with the ileum (44.0% vs. 16.5%, p < 0.05). Both tissues exhibited an inflammatory response with increased expression levels of TNF-a (p < 0.05) and a more pronounced increase of IL-8 in the rectum (p < 0.01). Additionally, the impact of EHEC on the mucus barrier varied across these gastrointestinal regions. Innovative visualization techniques helped us study the ultrastructure of mucus, revealing a net-like mucin glycoprotein organization. While further cellular differentiation could enhance model accuracy, our research significantly deepens understanding of EHEC pathogenesis in cattle and informs strategies for the preventative measures and therapeutic interventions.


Subject(s)
Enterohemorrhagic Escherichia coli , Ileum , Organoids , Rectum , Animals , Cattle , Ileum/microbiology , Ileum/metabolism , Ileum/ultrastructure , Rectum/microbiology , Enterohemorrhagic Escherichia coli/pathogenicity , Organoids/metabolism , Organoids/microbiology , Mucus/metabolism , Escherichia coli Infections/microbiology , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/ultrastructure
2.
Vet Res ; 54(1): 63, 2023 Jul 31.
Article in English | MEDLINE | ID: mdl-37525204

ABSTRACT

Chicken infection with Salmonella Typhimurium is an important source of foodborne human diseases. Salmonella colonizes the avian intestinal tract and more particularly the caecum, without causing symptoms. This thus poses a challenge for the prevention of foodborne transmission. Until now, studies on the interaction of Salmonella with the avian gut intestine have been limited by the absence of in vitro intestinal culture models. Here, we established intestinal crypt-derived chicken organoids to better decipher the impact of Salmonella intracellular replication on avian intestinal epithelium. Using a 3D organoid model, we observed a significantly higher replication rate of the intracellular bacteria in caecal organoids than in ileal organoids. Our model thus recreates intracellular environment, allowing Salmonella replication of avian epithelium according to the intestinal segment. Moreover, an inhibition of the cellular proliferation was observed in infected ileal and caecal organoids compared to uninfected organoids. This appears with a higher effect in ileal organoids, as well as a higher cytokine and signaling molecule response in infected ileal organoids at 3 h post-infection (hpi) than in caecal organoids that could explain the lower replication rate of Salmonella observed later at 24 hpi. To conclude, this study demonstrates that the 3D organoid is a model allowing to decipher the intracellular impact of Salmonella on the intestinal epithelium cell response and illustrates the importance of the gut segment used to purify stem cells and derive organoids to specifically study epithelial cell -Salmonella interaction.


Subject(s)
Chickens , Salmonella typhimurium , Humans , Animals , Salmonella typhimurium/physiology , Intestines , Intestinal Mucosa/microbiology , Cecum , Organoids/microbiology
3.
Cell Death Dis ; 14(1): 15, 2023 01 11.
Article in English | MEDLINE | ID: mdl-36631452

ABSTRACT

Skin infections caused by drug-resistant Staphylococcus aureus occur at high rates nationwide. Mouse primary epidermal organoids (mPEOs) possess stratified histological and morphological characteristics of epidermis and are highly similar to their derived tissue at the transcriptomic and proteomic levels. Herein, the susceptibility of mPEOs to methicillin-resistant S. aureus USA300 infection was investigated. The results show that mPEOs support USA300 colonization and invasion, exhibiting swollen epithelial squamous cells with nuclear necrosis and secreting inflammatory factors such as IL-1ß. Meanwhile mPEOs beneficial to observe the process of USA300 colonization with increasing infection time, and USA300 induces mPEOs to undergo pyroptosis and autophagy. In addition, we performed a drug screen for the mPEO infection model and showed that vancomycin restores cell viability and inhibits bacterial internalization in a concentration-dependent manner. In conclusion, we establish an in vitro skin infection model that contributes to the examination of drug screening strategies and antimicrobial drug mechanisms.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Organoids , Staphylococcal Infections , Animals , Mice , Drug Evaluation, Preclinical/methods , Epidermis/metabolism , Epidermis/microbiology , Epidermis/pathology , Proteomics , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Organoids/metabolism , Organoids/microbiology
4.
Drug Discov Today ; 27(1): 223-233, 2022 01.
Article in English | MEDLINE | ID: mdl-34418577

ABSTRACT

Approaches based on animal and two-dimensional (2D) cell culture models cannot ensure reliable results in modeling novel pathogens or in drug testing in the short term; therefore, there is rising interest in platforms such as organoids. To develop a toolbox that can be used successfully to overcome current issues in modeling various infections, it is essential to provide a framework of recent achievements in applying organoids. Organoids have been used to study viruses, bacteria, and protists that cause, for example, respiratory, gastrointestinal, and liver diseases. Their future as models of infection will be associated with improvements in system complexity, including abilities to model tissue structure, a dynamic microenvironment, and coinfection. Teaser. Organoids are a flexible tool for modelling viral, bacterial and protist infections. They can provide fast and reliable information on the biology of pathogens and in drug screening, and thus have become essential in combatting emerging infectious diseases.


Subject(s)
Drug Evaluation, Preclinical , Infections , Organoids , Animals , Cells, Cultured , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/trends , Infections/drug therapy , Infections/microbiology , Models, Animal , Organoids/drug effects , Organoids/microbiology , Reproducibility of Results
6.
Am J Physiol Gastrointest Liver Physiol ; 321(6): G693-G704, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34643092

ABSTRACT

All-time preservation of an intact mucosal barrier is crucial to ensuring intestinal homeostasis and, hence, the organism's overall health maintenance. This complex process relies on an equilibrated signaling system between the intestinal epithelium and numerous cell populations inhabiting the gut mucosa. Any perturbations of this delicate cross talk, particularly regarding the immune cell compartment and microbiota, may sustainably debilitate the intestinal barrier function. As a final joint event, a critical rise in epithelial permeability facilitates the exposure of submucosal immunity to microbial antigens, resulting in uncontrolled inflammation, collateral tissue destruction, and dysbiosis. Organoid-derived intestinal coculture models have established themselves as convenient tools to reenact such pathophysiological events, explore interactions between selected cell populations, and assess their roles with a central focus on intestinal barrier recovery and stabilization.


Subject(s)
Intestinal Mucosa/cytology , Organoids/cytology , Primary Cell Culture/methods , Animals , Coculture Techniques/methods , Gastrointestinal Microbiome , Humans , Intestinal Mucosa/microbiology , Lymphocytes/cytology , Macrophages/cytology , Organoids/microbiology
7.
APMIS ; 129(11): 653-662, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34580927

ABSTRACT

Infective endocarditis (IE) is a heart valve infection with high mortality rates. IE results from epithelial lesions, inducing sterile healing vegetations consisting of platelets, leucocytes, and fibrin that are susceptible for colonization by temporary bacteremia. Clinical testing of new treatments for IE is difficult and fast models sparse. The present study aimed at establishing an in vitro vegetation simulation IE model for fast screening of novel treatment strategies. A healing promoting platelet and leucocyte-rich fibrin patch was used to establish an IE organoid-like model by colonization with IE-associated bacterial isolates Staphylococcus aureus, Streptococcus spp (S. mitis group), and Enterococcus faecalis. The patch was subsequently exposed to tobramycin, ciprofloxacin, or penicillin. Bacterial colonization was evaluated by microscopy and quantitative bacteriology. We achieved stable bacterial colonization on the patch, comparable to clinical IE vegetations. Microscopy revealed uneven, biofilm-like colonization of the patch. The surface-associated bacteria displayed increased tolerance to antibiotics compared to planktonic bacteria. The present study succeeded in establishing an IE simulation model with the relevant pathogens S. aureus, S. mitis group, and E. faecalis. The findings indicate that the IE model mirrors the natural IE process and has the potential for fast screening of treatment candidates.


Subject(s)
Endocarditis, Bacterial/microbiology , Models, Biological , Anti-Bacterial Agents/pharmacology , Bacteremia/microbiology , Bacteria/drug effects , Bacteria/growth & development , Biofilms/drug effects , Biofilms/growth & development , Colony Count, Microbial , Drug Tolerance , Endocarditis, Bacterial/drug therapy , Endocarditis, Bacterial/pathology , Humans , Organoids/cytology , Organoids/drug effects , Organoids/microbiology
8.
Cell Rep ; 36(3): 109351, 2021 07 20.
Article in English | MEDLINE | ID: mdl-34289360

ABSTRACT

Recurrence of uropathogenic Escherichia coli (UPEC) infections has been attributed to reactivation of quiescent intracellular reservoirs (QIRs) in deep layers of the bladder wall. QIRs are thought to arise late during infection following dispersal of bacteria from intracellular bacterial communities (IBCs) in superficial umbrella cells. Here, we track the formation of QIR-like bacteria in a bladder organoid model that recapitulates the stratified uroepithelium within a volume suitable for high-resolution live-cell imaging. Bacteria injected into the organoid lumen enter umbrella-like cells and proliferate to form IBC-like bodies. In parallel, single bacteria penetrate deeper layers of the organoid wall, where they localize within or between uroepithelial cells. These "solitary" bacteria evade killing by antibiotics and neutrophils and are morphologically distinct from bacteria in IBCs. We conclude that bacteria with QIR-like properties may arise at early stages of infection, independent of IBC formation and rupture.


Subject(s)
Anti-Bacterial Agents/pharmacology , Models, Biological , Neutrophils/pathology , Organoids/microbiology , Urinary Bladder/microbiology , Uropathogenic Escherichia coli/physiology , Animals , Cell Differentiation/drug effects , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Female , Humans , Imaging, Three-Dimensional , Mice, Inbred C57BL , Microbial Viability/drug effects , Movement , Neutrophils/drug effects , Organoids/drug effects , Organoids/ultrastructure , Urinary Bladder/pathology , Uropathogenic Escherichia coli/drug effects , Uropathogenic Escherichia coli/growth & development , Uropathogenic Escherichia coli/ultrastructure
9.
Vet Res ; 52(1): 94, 2021 Jun 26.
Article in English | MEDLINE | ID: mdl-34174960

ABSTRACT

Small intestinal organoids, or enteroids, represent a valuable model to study host-pathogen interactions at the intestinal epithelial surface. Much research has been done on murine and human enteroids, however only a handful studies evaluated the development of enteroids in other species. Porcine enteroid cultures have been described, but little is known about their functional responses to specific pathogens or their associated virulence factors. Here, we report that porcine enteroids respond in a similar manner as in vivo gut tissues to enterotoxins derived from enterotoxigenic Escherichia coli, an enteric pathogen causing postweaning diarrhoea in piglets. Upon enterotoxin stimulation, these enteroids not only display a dysregulated electrolyte and water balance as shown by their swelling, but also secrete inflammation markers. Porcine enteroids grown as a 2D-monolayer supported the adhesion of an F4+ ETEC strain. Hence, these enteroids closely mimic in vivo intestinal epithelial responses to gut pathogens and are a promising model to study host-pathogen interactions in the pig gut. Insights obtained with this model might accelerate the design of veterinary therapeutics aimed at improving gut health.


Subject(s)
Enterotoxigenic Escherichia coli/physiology , Enterotoxins/toxicity , Escherichia coli Infections/veterinary , Intestine, Small/physiopathology , Organoids/physiopathology , Swine Diseases/physiopathology , Animals , Escherichia coli Infections/microbiology , Escherichia coli Infections/physiopathology , Host-Pathogen Interactions , Intestine, Small/microbiology , Organoids/microbiology , Sus scrofa , Swine , Swine Diseases/microbiology
10.
Microbiol Spectr ; 9(1): e0000321, 2021 09 03.
Article in English | MEDLINE | ID: mdl-34106568

ABSTRACT

Gastrointestinal infections cause significant morbidity and mortality worldwide. The complexity of human biology and limited insights into host-specific infection mechanisms are key barriers to current therapeutic development. Here, we demonstrate that two-dimensional epithelial monolayers derived from human intestinal organoids, combined with in vivo-like bacterial culturing conditions, provide significant advancements for the study of enteropathogens. Monolayers from the terminal ileum, cecum, and ascending colon recapitulated the composition of the gastrointestinal epithelium, in which several techniques were used to detect the presence of enterocytes, mucus-producing goblet cells, and other cell types following differentiation. Importantly, the addition of receptor activator of nuclear factor kappa-B ligand (RANKL) increased the presence of M cells, critical antigen-sampling cells often exploited by enteric pathogens. For infections, bacteria were grown under in vivo-like conditions known to induce virulence. Overall, interesting patterns of tissue tropism and clinical manifestations were observed. Shigella flexneri adhered efficiently to the cecum and colon; however, invasion in the colon was best following RANKL treatment. Both Salmonella enterica serovars Typhi and Typhimurium displayed different infection patterns, with S. Typhimurium causing more destruction of the terminal ileum and S. Typhi infecting the cecum more efficiently than the ileum, particularly with regard to adherence. Finally, various pathovars of Escherichia coli validated the model by confirming only adherence was observed with these strains. This work demonstrates that the combination of human-derived tissue with targeted bacterial growth conditions enables powerful analyses of human-specific infections that could lead to important insights into pathogenesis and accelerate future vaccine development. IMPORTANCE While traditional laboratory techniques and animal models have provided valuable knowledge in discerning virulence mechanisms of enteric pathogens, the complexity of the human gastrointestinal tract has hindered our understanding of physiologically relevant, human-specific interactions; and thus, has significantly delayed successful vaccine development. The human intestinal organoid-derived epithelial monolayer (HIODEM) model closely recapitulates the diverse cell populations of the intestine, allowing for the study of human-specific infections. Differentiation conditions permit the expansion of various cell populations, including M cells that are vital to immune recognition and the establishment of infection by some bacteria. We provide details of reproducible culture methods and infection conditions for the analyses of Shigella, Salmonella, and pathogenic Escherichia coli in which tissue tropism and pathogen-specific infection patterns were detected. This system will be vital for future studies that explore infection conditions, health status, or epigenetic differences and will serve as a novel screening platform for therapeutic development.


Subject(s)
Cell Culture Techniques/methods , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae/physiology , Gastrointestinal Tract/microbiology , Organoids/microbiology , Enterobacteriaceae/genetics , Enterobacteriaceae/pathogenicity , Enterocytes/microbiology , Epithelial Cells/cytology , Epithelial Cells/microbiology , Epithelium/microbiology , Gastrointestinal Tract/cytology , Humans , Organoids/cytology , Virulence
11.
Cell Host Microbe ; 29(6): 867-878, 2021 Jun 09.
Article in English | MEDLINE | ID: mdl-34111395

ABSTRACT

The important and diverse roles of the gut microbiota in human health and disease are increasingly recognized. The difficulty of inferring causation from metagenomic microbiome sequencing studies and from mouse-human interspecies differences has prompted the development of sophisticated in vitro models of human gut-microbe interactions. Here, we review recent advances in the co-culture of microbes with intestinal and colonic epithelia, comparing the rapidly developing fields of organoids and organs-on-chips with other standard models. We describe how specific individual processes by which microbes and epithelia interact can be recapitulated in vitro. Using examples of bacterial, viral, and parasitic infections, we highlight the advantages of each culture model and discuss current trends and future possibilities to build more complex co-cultures.


Subject(s)
Gastrointestinal Microbiome , Host Microbial Interactions , Microchip Analytical Procedures/methods , Organoids/microbiology , Precision Medicine/methods , Animal Testing Alternatives , Animals , Coculture Techniques/methods , Humans , Intestinal Mucosa , Mice , Microbial Interactions , Microfluidics/methods , Models, Animal
12.
mSphere ; 6(3)2021 05 12.
Article in English | MEDLINE | ID: mdl-33980679

ABSTRACT

Human respiratory syncytial virus (HRSV) is the leading cause of bronchiolitis in infants. Two subgroups of HRSV (A and B) routinely cocirculate. Most research has been performed with HRSV-A strains because these are easier to culture than HRSV-B strains. In this study, we aimed to compare the replicative fitness and HRSV-induced innate cytokine responses of HRSV-A and HRSV-B strains in disease-relevant cell culture models. We used two recombinant (r) clinical isolate-based HRSV strains (A11 and B05) and one recombinant laboratory-adapted HRSV strain (A2) to infect commercially available nasal, bronchial, and small-airway cultures. Epithelial cells from all anatomical locations were susceptible to HRSV infection despite the induction of a dominant type III interferon response. Subgroup A viruses disseminated and replicated faster than the subgroup B virus. Additionally, we studied HRSV infection and innate responses in airway organoids (AOs) cultured at air-liquid interface (ALI). Results were similar to the commercially obtained bronchial cells. In summary, we show that HRSV replicates well in cells from both the upper and the lower airways, with a slight replicative advantage for subgroup A viruses. Lastly, we showed that AOs cultured at ALI are a valuable model for studying HRSV ex vivo and that they can be used in the future to study factors that influence HRSV disease severity.IMPORTANCE Human respiratory syncytial virus (HRSV) is the major cause of bronchiolitis and pneumonia in young infants and causes almost 200,000 deaths per year. Currently, there is no vaccine or treatment available, only a prophylactic monoclonal antibody (palivizumab). An important question in HRSV pathogenesis research is why only a fraction (1 to 3%) of infants develop severe disease. Model systems comprising disease-relevant HRSV isolates and accurate and reproducible cell culture models are indispensable to study infection, replication, and innate immune responses. Here, we used differentiated AOs cultured at ALI to model the human airways. Subgroup A viruses replicated better than subgroup B viruses, which we speculate fits with epidemiological findings that subgroup A viruses cause more severe disease in infants. By using AOs cultured at ALI, we present a highly relevant, robust, and reproducible model that allows for future studies into what drives severe HRSV disease.


Subject(s)
Bronchi/virology , Nose/virology , Organoids/microbiology , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/physiology , Virus Replication , Bronchi/cytology , Cytokines/analysis , Cytokines/immunology , Epithelial Cells/virology , Humans , Immunity, Innate , In Vitro Techniques , Kinetics , Nose/cytology , Respiratory Syncytial Virus, Human/classification , Respiratory Syncytial Virus, Human/immunology
13.
mBio ; 12(3)2021 05 18.
Article in English | MEDLINE | ID: mdl-34006652

ABSTRACT

The intestinal epithelium is a primary interface for engagement of the host response by foodborne pathogens, like Salmonella enterica Typhimurium. While the interaction of S Typhimurium with the mammalian host has been well studied in transformed epithelial cell lines or in the complex intestinal environment in vivo, few tractable models recapitulate key features of the intestine. Human intestinal organoids (HIOs) contain a polarized epithelium with functionally differentiated cell subtypes, including enterocytes and goblet cells and a supporting mesenchymal cell layer. HIOs contain luminal space that supports bacterial replication, are more amenable to experimental manipulation than animals and are more reflective of physiological host responses. Here, we use the HIO model to define host transcriptional responses to S Typhimurium infection, also determining host pathways dependent on Salmonella pathogenicity island-1 (SPI-1)- and -2 (SPI-2)-encoded type 3 secretion systems (T3SS). Consistent with prior findings, we find that S Typhimurium strongly stimulates proinflammatory gene expression. Infection-induced cytokine gene expression was rapid, transient, and largely independent of SPI-1 T3SS-mediated invasion, likely due to continued luminal stimulation. Notably, S Typhimurium infection led to significant downregulation of host genes associated with cell cycle and DNA repair, leading to a reduction in cellular proliferation, dependent on SPI-1 and SPI-2 T3SS. The transcriptional profile of cell cycle-associated target genes implicates multiple miRNAs as mediators of S Typhimurium-dependent cell cycle suppression. These findings from Salmonella-infected HIOs delineate common and distinct contributions of SPI-1 and SPI-2 T3SSs in inducing early host responses during enteric infection and reinforce host cell proliferation as a process targeted by SalmonellaIMPORTANCESalmonella enterica serovar Typhimurium (S Typhimurium) causes a significant health burden worldwide, yet host responses to initial stages of intestinal infection remain poorly understood. Due to differences in infection outcome between mice and humans, physiological human host responses driven by major virulence determinants of Salmonella have been more challenging to evaluate. Here, we use the three-dimensional human intestinal organoid model to define early responses to infection with wild-type S Typhimurium and mutants defective in the SPI-1 or SPI-2 type-3 secretion systems. While both secretion system mutants show defects in mouse models of oral Salmonella infection, the specific contributions of each secretion system are less well understood. We show that S Typhimurium upregulates proinflammatory pathways independently of either secretion system, while the downregulation of the host cell cycle pathways relies on both SPI-1 and SPI-2. These findings lay the groundwork for future studies investigating how SPI-1- and SPI-2-driven host responses affect infection outcome and show the potential of this model to study host-pathogen interactions with other serovars to understand how initial interactions with the intestinal epithelium may affect pathogenesis.


Subject(s)
Bacterial Proteins/genetics , Enterocytes/microbiology , Gene Expression Profiling , Host-Pathogen Interactions/genetics , Membrane Proteins/genetics , Organoids/microbiology , Salmonella typhimurium/genetics , Cell Line , Gene Expression Regulation, Bacterial , Humans , Intestinal Mucosa/microbiology , Intestines/cytology , Intestines/microbiology , Salmonella typhimurium/pathogenicity , Serogroup , Virulence Factors
14.
Cell Mol Gastroenterol Hepatol ; 12(2): 769-782, 2021.
Article in English | MEDLINE | ID: mdl-33895425

ABSTRACT

Intestinal organoids have become indispensable tools for many gastrointestinal researchers, advancing their studies of nontransformed intestinal epithelial cells, and their roles in an array of diseases, including inflammatory bowel disease and colon cancer. In many cases. these diseases, as well as many enteric infections, reflect pathogenic interactions between bacteria and the gut epithelium. The complexity of studying this microbe-epithelial interface in vivo has led to significant focus on modeling this cross-talk using organoid models. Considering how quickly the organoid field is advancing, it can be difficult to keep up to date with the latest techniques, as well as their respective strengths and weaknesses. This review addresses the advantages of using organoids derived from adult stem cells and the recently identified differences that biopsy location and patient age can have on organoids and their interactions with microbes. Several approaches to introducing bacteria in a relevant (apical) manner (ie, microinjecting 3-dimensional spheroids, polarity-reversed organoids, and 2-dimensional monolayers) also are addressed, as are the key readouts that can be obtained using these models. Lastly, the potential for new approaches, such as air-liquid interface, to facilitate studying bacterial interactions with important but understudied epithelial subsets such as goblet cells and their products, is evaluated.


Subject(s)
Bacteria/metabolism , Epithelial Cells/microbiology , Host-Pathogen Interactions , Intestines/pathology , Organoids/microbiology , Animals , Humans , Stem Cells/metabolism
15.
Front Immunol ; 12: 646906, 2021.
Article in English | MEDLINE | ID: mdl-33854511

ABSTRACT

Over the last decades, Adherent-Invasive Escherichia coli (AIEC) has been linked to the pathogenesis of Crohn's Disease. AIEC's characteristics, as well as its interaction with the gut immune system and its role in intestinal epithelial barrier dysfunction, have been extensively studied. Nevertheless, the currently available techniques to investigate the cross-talk between this pathogen and intestinal epithelial cells (IECs) are based on the infection of immortalized cell lines. Despite their many advantages, cell lines cannot reproduce the conditions in tissues, nor do they reflect interindividual variability or gut location-specific traits. In that sense, the use of human primary cultures, either healthy or diseased, offers a system that can overcome all of these limitations. Here, we developed a new infection model by using freshly isolated human IECs. For the first time, we generated and infected monolayer cultures derived from human colonic organoids to study the mechanisms and effects of AIEC adherence and invasion on primary human epithelial cells. To establish the optimal conditions for AIEC invasion studies in human primary organoid-derived epithelial monolayers, we designed an infection-kinetics study to assess the infection dynamics at different time points, as well as with two multiplicities of infection (MOI). Overall, this method provides a model for the study of host response to AIEC infections, as well as for the understanding of the molecular mechanisms involved in adhesion, invasion and intracellular replication. Therefore, it represents a promising tool for elucidating the cross-talk between AIEC and the intestinal epithelium in healthy and diseased tissues.


Subject(s)
Epithelial Cells/metabolism , Escherichia coli Infections/metabolism , Intestinal Mucosa/metabolism , Organoids/metabolism , Algorithms , Bacterial Adhesion , Cell Culture Techniques/methods , Crohn Disease/metabolism , Crohn Disease/microbiology , Epithelial Cells/microbiology , Escherichia coli/physiology , Escherichia coli Infections/microbiology , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/microbiology , Models, Biological , Organoids/cytology , Organoids/microbiology , Reproducibility of Results
16.
Br J Cancer ; 124(11): 1751-1753, 2021 05.
Article in English | MEDLINE | ID: mdl-33742142

ABSTRACT

Changes in the microbiome are associated with the development of colorectal cancer, but causal explanations have been lacking. We recently demonstrated that pks+ Escherichia coli induce a specific mutational pattern using intestinal organoids and these mutations are present in the genomes of colorectal cancer. This finding warrants further studies on the microbial role in oncogenic mutation induction, cancer development and future preventive strategies.


Subject(s)
Colorectal Neoplasms/genetics , Gastrointestinal Microbiome/genetics , Transcriptome , Cells, Cultured , Colon/metabolism , Colon/microbiology , Colorectal Neoplasms/microbiology , DNA Damage/genetics , Escherichia coli/genetics , Host Microbial Interactions/genetics , Humans , INDEL Mutation , Mutation , Organoids/metabolism , Organoids/microbiology , Organoids/pathology , Peptides/genetics , Peptides/physiology , Polyketides , Polymorphism, Single Nucleotide , Transcriptome/physiology
17.
NPJ Biofilms Microbiomes ; 7(1): 8, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33495449

ABSTRACT

Bacterial biofilms cause 65% of all human infections and are highly resistant to antibiotic therapy but lack specific treatments. To provide a human organoid model for studying host-microbe interplay and enabling screening for novel antibiofilm agents, a human epidermis organoid model with robust methicillin-resistant Staphylococcus aureus (MRSA) USA300 and Pseudomonas aeruginosa PAO1 biofilm was developed. Treatment of 1-day and 3-day MRSA and PAO1 biofilms with antibiofilm peptide DJK-5 significantly and substantially reduced the bacterial burden. This model enabled the screening of synthetic host defense peptides, revealing their superior antibiofilm activity against MRSA compared to the antibiotic mupirocin. The model was extended to evaluate thermally wounded skin infected with MRSA biofilms resulting in increased bacterial load, cytotoxicity, and pro-inflammatory cytokine levels that were all reduced upon treatment with DJK-5. Combination treatment of DJK-5 with an anti-inflammatory peptide, 1002, further reduced cytotoxicity and skin inflammation.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Models, Biological , Organoids/microbiology , Anti-Bacterial Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Bacterial Load/drug effects , Biofilms/growth & development , Burns/drug therapy , Burns/immunology , Burns/microbiology , Drug Evaluation, Preclinical , Drug Therapy, Combination , Humans , Methicillin-Resistant Staphylococcus aureus/drug effects , Oligopeptides/pharmacology , Oligopeptides/therapeutic use , Organoids/drug effects , Organoids/immunology , Organoids/injuries , Pseudomonas aeruginosa/drug effects , Skin/drug effects , Skin/immunology , Skin/injuries , Skin/microbiology
18.
Cell Death Dis ; 12(1): 35, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33414472

ABSTRACT

Technology of generating human epidermal derivatives with physiological relevance to in vivo epidermis is continuously investigated for improving their effects on modeling of human natural dermatological status in basic and clinical studies. Here, we report a method of robust establishment and expansion of human primary epidermal organoids (hPEOs) under a chemically defined condition. hPEOs reconstruct morphological, molecular, and functional features of human epidermis and can expand for 6 weeks. Remarkably, hPEOs are permissive for dermatophyte infections caused by Trichophyton Rubrum (T. rubrum). The T. rubrum infections on hPEOs reflect many aspects of known clinical pathological reactions and reveal that the repression on IL-1 signaling may contribute to chronic and recurrent infections with the slight inflammation caused by T. rubrum in human skin. Thus, our present study provides a new insight into the pathogenesis of T. rubrum infections and indicates that hPEOs are a potential ex vivo model for both basic studies of skin diseases and clinical studies of testing potential antifungal drugs.


Subject(s)
Arthrodermataceae/immunology , Dermatomycoses/immunology , Epidermal Cells , Organoids , Adolescent , Adult , Cells, Cultured , Child , Epidermal Cells/immunology , Epidermal Cells/microbiology , Epidermis , Humans , Middle Aged , Organoids/immunology , Organoids/microbiology , Young Adult
19.
Ann Biomed Eng ; 49(4): 1233-1244, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33409849

ABSTRACT

To generate physiologically-relevant experimental models, the study of enteric diarrheal diseases is turning increasingly to advanced in vitro models that combine ex vivo, stem cell-derived "organoid" cell lines with bioengineered culture environments that expose them to mechanical stimuli, such as fluid flow. However, such approaches require considerable technical expertise with both microfabrication and organoid culture, and are, therefore, inaccessible to many researchers. For this reason, we have developed a perfusion system that is simple to fabricate, operate, and maintain. Its dimensions approximate the volume and cell culture area of traditional 96-well plates and allow the incorporation of fastidious primary, stem cell-derived cell lines with only minimal adaptation of their established culture techniques. We show that infections with enteroaggregative E. coli and norovirus, common causes of infectious diarrhea, in the system display important differences from static models, and in some ways better recreate the pathophysiology of in vivo infections. Furthermore, commensal strains of bacteria can be added alongside the pathogens to simulate the effects of a host microbiome on the infectious process. For these reasons, we believe that this perfusion system is a powerful, yet easily accessible tool for studying host-pathogen interactions in the human intestine.


Subject(s)
Caliciviridae Infections , Escherichia coli Infections , Escherichia coli , Gastrointestinal Diseases , Norovirus , Organ Culture Techniques , Organoids/microbiology , Adult , Biofilms , Cells, Cultured , Escherichia coli/physiology , Escherichia coli Proteins/metabolism , Fimbriae Proteins/metabolism , Host-Pathogen Interactions , Humans , Intestine, Small/cytology , Intestine, Small/metabolism , Intestine, Small/microbiology , Mucins/metabolism , Norovirus/physiology , Organoids/metabolism , Perfusion , Stem Cells , Virulence Factors/metabolism , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...