Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 155
Filter
1.
Life Sci Alliance ; 5(4)2022 04.
Article in English | MEDLINE | ID: mdl-35064075

ABSTRACT

Differentiation and lineage specification are controlled by cooperation of growth factor signalling. The involvement of epigenetic regulators in lineage specification remains largely elusive. Here, we show that the histone methyltransferase Mll1 prevents intestinal progenitor cells from differentiation, whereas it is also involved in secretory lineage specification of Paneth and goblet cells. Using conditional mutagenesis in mice and intestinal organoids, we demonstrate that loss of Mll1 renders intestinal progenitor cells permissive for Wnt-driven secretory differentiation. However, Mll1-deficient crypt cells fail to segregate Paneth and goblet cell fates. Mll1 deficiency causes Paneth cell-determined crypt progenitors to exhibit goblet cell features by unleashing Mapk signalling, resulting in increased numbers of mixed Paneth/goblet cells. We show that loss of Mll1 abolishes the pro-proliferative effect of Mapk signalling in intestinal progenitor cells and promotes Mapk-induced goblet cell differentiation. Our data uncover Mll1 and its downstream targets Gata4/6 as a regulatory hub of Wnt and Mapk signalling in the control of lineage specification of intestinal secretory Paneth and goblet cells.


Subject(s)
MAP Kinase Signaling System/genetics , Wnt Signaling Pathway/genetics , Animals , Cell Differentiation/genetics , Epigenesis, Genetic/genetics , Epigenomics/methods , Female , Goblet Cells/cytology , Goblet Cells/metabolism , Humans , Intestinal Mucosa/metabolism , Intestines , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Transgenic , Organoids/metabolism , Paneth Cells/cytology , Paneth Cells/metabolism , Stem Cells/metabolism , Wnt Signaling Pathway/physiology
2.
Science ; 375(6576): eaaw9021, 2022 Jan 07.
Article in English | MEDLINE | ID: mdl-34990240

ABSTRACT

Epithelial organoids are stem cell­derived tissues that approximate aspects of real organs, and thus they have potential as powerful tools in basic and translational research. By definition, they self-organize, but the structures formed are often heterogeneous and irreproducible, which limits their use in the lab and clinic. We describe methodologies for spatially and temporally controlling organoid formation, thereby rendering a stochastic process more deterministic. Bioengineered stem cell microenvironments are used to specify the initial geometry of intestinal organoids, which in turn controls their patterning and crypt formation. We leveraged the reproducibility and predictability of the culture to identify the underlying mechanisms of epithelial patterning, which may contribute to reinforcing intestinal regionalization in vivo. By controlling organoid culture, we demonstrate how these structures can be used to answer questions not readily addressable with the standard, more variable, organoid models.


Subject(s)
Intestinal Mucosa/growth & development , Organogenesis , Organoids/growth & development , Tissue Engineering , Animals , Cell Differentiation , Cell Shape , Epithelial Cells/cytology , Hydrogels , Intestinal Mucosa/anatomy & histology , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Mice , Organoids/anatomy & histology , Organoids/cytology , Organoids/metabolism , Paneth Cells/cytology , Receptors, Notch/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/physiology , Tissue Culture Techniques , YAP-Signaling Proteins/metabolism
3.
Development ; 148(21)2021 11 01.
Article in English | MEDLINE | ID: mdl-34751748

ABSTRACT

Although the role of the transcription factor NF-κB in intestinal inflammation and tumor formation has been investigated extensively, a physiological function of NF-κB in sustaining intestinal epithelial homeostasis beyond inflammation has not been demonstrated. Using NF-κB reporter mice, we detected strong NF-κB activity in Paneth cells, in '+4/+5' secretory progenitors and in scattered Lgr5+ crypt base columnar stem cells of small intestinal (SI) crypts. To examine NF-κB functions in SI epithelial self-renewal, mice or SI crypt organoids ('mini-guts') with ubiquitously suppressed NF-κB activity were used. We show that NF-κB activity is dispensable for maintaining SI epithelial proliferation, but is essential for ex vivo organoid growth. Furthermore, we demonstrate a dramatic reduction of Paneth cells in the absence of NF-κB activity, concomitant with a significant increase in goblet cells and immature intermediate cells. This indicates that NF-κB is required for proper Paneth versus goblet cell differentiation and for SI epithelial homeostasis, which occurs via regulation of Wnt signaling and Sox9 expression downstream of NF-κB. The current study thus presents evidence for an important role for NF-κB in intestinal epithelial self-renewal.


Subject(s)
Goblet Cells/cytology , Intestine, Small/cytology , NF-kappa B/metabolism , Paneth Cells/cytology , Animals , Cell Differentiation , Cell Self Renewal , Goblet Cells/metabolism , Homeostasis , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Intestine, Small/pathology , Mice , NF-kappa B/genetics , Organoids/cytology , Organoids/growth & development , Organoids/metabolism , Paneth Cells/metabolism , SOX9 Transcription Factor/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway
4.
Nutrients ; 13(6)2021 Jun 12.
Article in English | MEDLINE | ID: mdl-34204790

ABSTRACT

The beneficial effects of human milk suppressing the development of intestinal pathologies such as necrotizing enterocolitis in preterm infants are widely known. Human milk (HM) is rich in a multitude of bioactive factors that play major roles in promoting postnatal maturation, differentiation, and the development of the microbiome. Previous studies showed that HM is rich in hyaluronan (HA) especially in colostrum and early milk. This study aims to determine the role of HA 35 KDa, a HM HA mimic, on intestinal proliferation, differentiation, and the development of the intestinal microbiome. We show that oral HA 35 KDa supplementation for 7 days in mouse pups leads to increased villus length and crypt depth, and increased goblet and Paneth cells, compared to controls. We also show that HA 35 KDa leads to an increased predominance of Clostridiales Ruminococcaceae, Lactobacillales Lactobacillaceae, and Clostridiales Lachnospiraceae. In seeking the mechanisms involved in the changes, bulk RNA seq was performed on samples from the terminal ileum and identified upregulation in several genes essential for cellular growth, proliferation, and survival. Taken together, this study shows that HA 35 KDa supplemented to mouse pups promotes intestinal epithelial cell proliferation, as well as the development of Paneth cells and goblet cell subsets. HA 35 KDa also impacted the intestinal microbiota; the implications of these responses need to be determined.


Subject(s)
Dietary Supplements , Gastrointestinal Microbiome/drug effects , Hyaluronic Acid/pharmacology , Intestine, Small/growth & development , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Goblet Cells/cytology , Intestinal Mucosa/drug effects , Intestine, Small/cytology , Intestines/cytology , Intestines/growth & development , Mice , Paneth Cells/cytology
5.
Cell Death Dis ; 12(1): 131, 2021 01 27.
Article in English | MEDLINE | ID: mdl-33504792

ABSTRACT

Intestinal Paneth cells are professional exocrine cells that play crucial roles in maintenance of homeostatic microbiome, modulation of mucosal immunity, and support for stem cell self-renewal. Dysfunction of these cells may lead to the pathogenesis of human diseases such as inflammatory bowel disease (IBD). Cdk5 activator binding protein Cdk5rap3 (also known as C53 and LZAP) was originally identified as a binding protein of Cdk5 activator p35. Although previous studies have indicated its involvement in a wide range of signaling pathways, the physiological function of Cdk5rap3 remains largely undefined. In this study, we found that Cdk5rap3 deficiency resulted in very early embryonic lethality, indicating its indispensable role in embryogenesis. To further investigate its function in the adult tissues and organs, we generated intestinal epithelial cell (IEC)-specific knockout mouse model to examine its role in intestinal development and tissue homeostasis. IEC-specific deletion of Cdk5rap3 led to nearly complete loss of Paneth cells and increased susceptibility to experimentally induced colitis. Interestingly, Cdk5rap3 deficiency resulted in downregulation of key transcription factors Gfi1 and Sox9, indicating its crucial role in Paneth cell fate specification. Furthermore, Cdk5rap3 is highly expressed in mature Paneth cells. Paneth cell-specific knockout of Cdk5rap3 caused partial loss of Paneth cells, while inducible acute deletion of Cdk5rap3 resulted in disassembly of the rough endoplasmic reticulum (RER) and abnormal zymogen granules in the mature Paneth cells, as well as loss of Paneth cells. Together, our results provide definitive evidence for the essential role of Cdk5rap3 in Paneth cell development and maintenance.


Subject(s)
Cell Cycle Proteins/metabolism , Paneth Cells/cytology , Paneth Cells/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , Cell Differentiation/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
6.
Nat Cell Biol ; 23(1): 23-31, 2021 01.
Article in English | MEDLINE | ID: mdl-33398177

ABSTRACT

A detailed understanding of intestinal stem cell (ISC) self-renewal and differentiation is required to treat chronic intestinal diseases. However, the different models of ISC lineage hierarchy1-6 and segregation7-12 are subject to debate. Here, we have discovered non-canonical Wnt/planar cell polarity (PCP)-activated ISCs that are primed towards the enteroendocrine or Paneth cell lineage. Strikingly, integration of time-resolved lineage labelling with single-cell gene expression analysis revealed that both lineages are directly recruited from ISCs via unipotent transition states, challenging the existence of formerly predicted bi- or multipotent secretory progenitors7-12. Transitory cells that mature into Paneth cells are quiescent and express both stem cell and secretory lineage genes, indicating that these cells are the previously described Lgr5+ label-retaining cells7. Finally, Wnt/PCP-activated Lgr5+ ISCs are molecularly indistinguishable from Wnt/ß-catenin-activated Lgr5+ ISCs, suggesting that lineage priming and cell-cycle exit is triggered at the post-transcriptional level by polarity cues and a switch from canonical to non-canonical Wnt/PCP signalling. Taken together, we redefine the mechanisms underlying ISC lineage hierarchy and identify the Wnt/PCP pathway as a new niche signal preceding lateral inhibition in ISC lineage priming and segregation.


Subject(s)
Cell Lineage , Cell Polarity , Enteroendocrine Cells/cytology , Intestinal Mucosa/cytology , Paneth Cells/cytology , Stem Cells/cytology , Wnt Proteins/metabolism , Animals , Cell Self Renewal , Enteroendocrine Cells/metabolism , Female , Gene Expression Profiling , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Paneth Cells/metabolism , Receptors, G-Protein-Coupled/physiology , Single-Cell Analysis , Stem Cells/metabolism , beta Catenin/metabolism
7.
Cell Metab ; 32(5): 889-900.e7, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33147486

ABSTRACT

Differential WNT and Notch signaling regulates differentiation of Lgr5+ crypt-based columnar cells (CBCs) into intestinal cell lineages. Recently we showed that mitochondrial activity supports CBCs, while adjacent Paneth cells (PCs) show reduced mitochondrial activity. This implies that CBC differentiation into PCs involves a metabolic transition toward downregulation of mitochondrial dependency. Here we show that Forkhead box O (FoxO) transcription factors and Notch signaling interact in determining CBC fate. In agreement with the organoid data, Foxo1/3/4 deletion in mouse intestine induces secretory cell differentiation. Importantly, we show that FOXO and Notch signaling converge on regulation of mitochondrial fission, which in turn provokes stem cell differentiation into goblet cells and PCs. Finally, scRNA-seq-based reconstruction of CBC differentiation trajectories supports the role of FOXO, Notch, and mitochondria in secretory differentiation. Together, this points at a new signaling-metabolic axis in CBC differentiation and highlights the importance of mitochondria in determining stem cell fate.


Subject(s)
Goblet Cells , Intestines/cytology , Mitochondria/metabolism , Paneth Cells , Stem Cells , Animals , Cell Differentiation , Cell Line , Forkhead Transcription Factors/metabolism , Goblet Cells/cytology , Goblet Cells/metabolism , Mice , Mitochondrial Dynamics , Paneth Cells/cytology , Paneth Cells/metabolism , Receptors, Notch/metabolism , Stem Cells/cytology , Stem Cells/metabolism
8.
Int J Mol Sci ; 21(14)2020 Jul 14.
Article in English | MEDLINE | ID: mdl-32674311

ABSTRACT

Intestinal organoids have emerged as the new paradigm for modelling the healthy and diseased intestine with patient-relevant properties. In this study, we show directed differentiation of induced pluripotent stem cells towards intestinal-like phenotype within a microfluidic device. iPSCs are cultured against a gel in microfluidic chips of the OrganoPlate, in which they undergo stepwise differentiation. Cells form a tubular structure, lose their stem cell markers and start expressing mature intestinal markers, including markers for Paneth cells, enterocytes and neuroendocrine cells. Tubes develop barrier properties as confirmed by transepithelial electrical resistance (TEER). Lastly, we show that tubules respond to pro-inflammatory cytokine triggers. The whole procedure for differentiation lasts 14 days, making it an efficient process to make patient-specific organoid tubules. We anticipate the usage of the platform for disease modelling and drug candidate screening.


Subject(s)
Cell Differentiation/physiology , Induced Pluripotent Stem Cells/cytology , Intestines/cytology , Biomarkers/metabolism , Caco-2 Cells , Cell Line , Cell Line, Tumor , Cytokines/metabolism , Enterocytes/cytology , Enterocytes/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Inflammation/metabolism , Lab-On-A-Chip Devices , Neuroendocrine Cells/cytology , Neuroendocrine Cells/metabolism , Organoids/cytology , Organoids/metabolism , Paneth Cells/cytology , Paneth Cells/metabolism
9.
Sci Rep ; 10(1): 12164, 2020 07 22.
Article in English | MEDLINE | ID: mdl-32699335

ABSTRACT

Cryptdins are disulfide-rich cationic antimicrobial peptides secreted by mouse Paneth cells and are known to exhibit potent antimicrobial activity against various deadly pathogens. Keeping in view the extremely low yield obtained from mouse Paneth cells and high cost of synthetic peptide(s), herein, we have attempted to produce cryptdin-2 in Escherichia coli using recombinant technology. To avoid lethal effects of peptide on the host cells, cryptdin-2 was expressed as a fusion protein with thioredoxin as fusion partner which yielded 40 mg/L protein in the soluble fraction. Subsequently, mature cryptdin-2 was cleaved from the fusion partner and purified by cation exchange chromatography. Since conjugation of poly(ethylene) glycol (PEG) has been known to improve the biological properties of biomolecules, therefore, we further attempted to prepare PEG-conjugated variant of cryptdin-2 using thiol specific PEGylation. Though the antimicrobial activity of PEGylated cryptdin-2 was compromised to some extent, but it was found to have enhanced serum stability for longer duration as compared to its un-modified forms. Also, it was found to exhibit reduced toxicity to the host cells. Further, its synergism with gentamicin suggests that PEGylated cryptdin-2 can be used with conventional antibiotics, thereby indicating its possibility to be used as an adjunct therapy.


Subject(s)
Defensins/pharmacology , Polyethylene Glycols/chemistry , Staphylococcus aureus/drug effects , Amino Acid Sequence , Animals , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Cell Survival/drug effects , Defensins/chemistry , Defensins/genetics , Defensins/metabolism , Mice , Microbial Sensitivity Tests , Mutagenesis, Site-Directed , Paneth Cells/cytology , Paneth Cells/metabolism , RAW 264.7 Cells , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/pharmacology
10.
PLoS One ; 15(4): e0230231, 2020.
Article in English | MEDLINE | ID: mdl-32240190

ABSTRACT

Enteroids are cultured primary intestinal epithelial cells that recapitulate epithelial lineage development allowing for a more complex and physiologically relevant model for scientific study. The large presence of intestinal stem cells (ISC) in these enteroids allows for the study of metabolite effects on cellular processes and resulting progeny cells. Short-chain fatty acids (SCFA) such as butyrate (BUT) are bacterial metabolites produced in the gastrointestinal tract that are considered to be beneficial to host cells. Therefore, the objective was to study the effects of SCFAs on biomarkers of ISC activity, differentiation, barrier function and epithelial defense in the intestine using mouse and human enteroid models. Enteroids were treated with two concentrations of acetate (ACET), propionate (PROP), or BUT for 24 h. Enteroids treated with BUT or PROP showed a decrease in proliferation via EdU uptake relative to the controls in both mouse and human models. Gene expression of Lgr5 was shown to decrease with BUT and PROP treatments, but increased with ACET. As a result of BUT and PROP treatments, there was an increase in differentiation markers for enterocyte, Paneth, goblet, and enteroendocrine cells. Gene expression of antimicrobial proteins Reg3ß, Reg3γ, and Defb1 were stimulated by BUT and PROP, but not by ACET which had a greater effect on expression of tight junction genes Cldn3 and Ocln in 3D enteroids. Similar results were obtained with human enteroids treated with 10 mM SCFAs and grown in either 3D or Transwell™ model cultures, although tight junctions were influenced by BUT and PROP, but not ACET in monolayer format. Furthermore, BUT and PROP treatments increased transepithelial electrical resistance after 24 h compared to ACET or control. Overall, individual SCFAs are potent stimulators of cellular gene expression, however, PROP and especially BUT show great efficacy for driving cell differentiation and gene expression.


Subject(s)
Acetic Acid/pharmacology , Butyric Acid/pharmacology , Gene Expression Regulation/drug effects , Propionates/pharmacology , Spheroids, Cellular/drug effects , Animals , Cell Culture Techniques , Cell Differentiation/drug effects , Claudin-3/genetics , Claudin-3/metabolism , Enterocytes/cytology , Enterocytes/drug effects , Enterocytes/metabolism , Enteroendocrine Cells/cytology , Enteroendocrine Cells/drug effects , Enteroendocrine Cells/metabolism , Goblet Cells/cytology , Goblet Cells/drug effects , Goblet Cells/metabolism , Humans , Mice , Occludin/genetics , Occludin/metabolism , Pancreatitis-Associated Proteins/genetics , Pancreatitis-Associated Proteins/metabolism , Paneth Cells/cytology , Paneth Cells/drug effects , Paneth Cells/metabolism , Primary Cell Culture , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Tight Junctions/drug effects , beta-Defensins/genetics , beta-Defensins/metabolism
11.
Gut Microbes ; 11(4): 997-1014, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32138622

ABSTRACT

Little is known about the regulatory effect of microbiota on the proliferation and regeneration of ISCs. Here, we found that L. reuteri stimulated the proliferation of intestinal epithelia by increasing the expression of R-spondins and thus activating the Wnt/ß-catenin pathway. The proliferation-stimulating effect of Lactobacillus on repair is further enhanced under TNF -induced intestinal mucosal damage, and the number of Lgr5+ cells is maintained. Moreover, compared to the effects of C. rodentium on the induction of intestinal inflammation and crypt hyperplasia in mice, L. reuteri protected the intestinal mucosal barrier integrity by moderately modulating the Wnt/ß-catenin signaling pathway to avoid overactivation. L. reuteri had the ability to maintain the number of Lgr5+ cells and stimulate intestinal epithelial proliferation to repair epithelial damage and reduce proinflammatory cytokine secretion in the intestine and the LPS concentration in serum. Moreover, activation of the Wnt/ß-catenin pathway also induced differentiation toward Paneth cells and increased antimicrobial peptide expression to inhibit C. rodentium colonization. The protective effect of Lactobacillus against C. rodentium infection disappeared upon application of the Wnt antagonist Wnt-C59 in both mice and intestinal organoids. This study demonstrates that Lactobacillus is effective at maintaining intestinal epithelial regeneration and homeostasis as well as at repairing intestinal damage after pathological injury and is thus a promising alternative therapeutic method for intestinal inflammation.


Subject(s)
Intestinal Mucosa/pathology , Intestinal Mucosa/physiology , Limosilactobacillus reuteri/physiology , Animals , Cell Differentiation , Cell Proliferation , Citrobacter rodentium/growth & development , Enteritis/microbiology , Enteritis/prevention & control , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/prevention & control , Epithelial Cells/cytology , Intestinal Mucosa/cytology , Intestine, Small/microbiology , Mice , Mice, Inbred C57BL , Organoids , Paneth Cells/cytology , Probiotics , Regeneration , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology , Wnt Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism
12.
Nat Methods ; 17(3): 335-342, 2020 03.
Article in English | MEDLINE | ID: mdl-32066960

ABSTRACT

Despite the widespread adoption of organoids as biomimetic tissue models, methods to comprehensively analyze cell-type-specific post-translational modification (PTM) signaling networks in organoids are absent. Here, we report multivariate single-cell analysis of such networks in organoids and organoid cocultures. Simultaneous analysis by mass cytometry of 28 PTMs in >1 million single cells derived from small intestinal organoids reveals cell-type- and cell-state-specific signaling networks in stem, Paneth, enteroendocrine, tuft and goblet cells, as well as enterocytes. Integrating single-cell PTM analysis with thiol-reactive organoid barcoding in situ (TOBis) enables high-throughput comparison of signaling networks between organoid cultures. Cell-type-specific PTM analysis of colorectal cancer organoid cocultures reveals that shApc, KrasG12D and Trp53R172H cell-autonomously mimic signaling states normally induced by stromal fibroblasts and macrophages. These results demonstrate how standard mass cytometry workflows can be modified to perform high-throughput multivariate cell-type-specific signaling analysis of healthy and cancerous organoids.


Subject(s)
Biomimetics , Colorectal Neoplasms/pathology , Gene Expression Regulation , Intestine, Small/cytology , Organoids/metabolism , Signal Transduction , Animals , Cell Differentiation , Coculture Techniques/methods , Colorectal Neoplasms/metabolism , Cytophotometry/methods , Enterocytes/cytology , Enteroendocrine Cells/cytology , Female , Fibroblasts/cytology , Goblet Cells/cytology , Humans , Macrophages/cytology , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Paneth Cells/cytology , Single-Cell Analysis/methods , Sulfhydryl Compounds/chemistry , Tumor Suppressor Protein p53/metabolism
13.
PLoS One ; 14(9): e0221618, 2019.
Article in English | MEDLINE | ID: mdl-31509557

ABSTRACT

Paneth cells (PCs) are specialized epithelial cells of the small bowel that contain multiple secretory granules filled with antimicrobial peptides and trophic factors, which are essential for the control of the microorganisms growth and maintaining intestinal integrity. Alterations in their function are associated with an imbalance of the normal microbiota, gastrointestinal infections and inflammatory processes, such as Crohn's disease (CD). One of the most common murine models for studying CD is IL-10-/- mouse. IL-10-/- mice when housed in conventional conditions and take contact with commensal microorganisms develop an acute enterocolitis mediated by a Th1 immune response. Even though, alterations in PCs function are related to CD, they had not been characterized yet in this mouse model. Here we show that in specific pathogen free conditions IL-10-/- mice have aberrant granules and a large number of immature PCs at the bottom of the crypt in the ileum of IL-10-/- mice before developing intestinal inflammation, along with a reduced expression of Indian Hedgehog. In addition, IL-10-/- Paneth cells presented a reduced expression of cryptidin-4, and a heterogeneous distribution of lysozyme+ granules. The alterations in the maturation of the PCs at the bottom of the crypt were not modified after the colonization by the conventional microbiota. On the other hand, depletion of microbiota altered the phenotype, but did not normalize PCs. Our results suggest that IL-10 could be necessary for the integrity of PCs. Moreover, our results help to explain why IL-10-/- mice develop enterocolitis in response to microorganisms.


Subject(s)
Interleukin-10/genetics , Paneth Cells/cytology , Secretory Vesicles/metabolism , alpha-Defensins/genetics , Animals , Cell Differentiation , Cells, Cultured , Disease Models, Animal , Gene Knockdown Techniques , Hedgehog Proteins/metabolism , Male , Mice , Microbiota , Paneth Cells/immunology , Paneth Cells/metabolism , Phenotype , Specific Pathogen-Free Organisms , Th1 Cells/immunology
14.
Sci Rep ; 9(1): 13115, 2019 09 11.
Article in English | MEDLINE | ID: mdl-31511628

ABSTRACT

Abundant evidence from many laboratories supports the premise that α-defensin peptides secreted from Paneth cells are key mediators of host-microbe interactions in the small intestine that contribute to host defense and homeostasis. α-defensins are among the most highly expressed antimicrobial peptides at this mucosal surface in many mammals, including humans and mice; however, there is striking variation among species in the number and primary structure of α-defensin paralogs. Studies of these biomolecules in vivo are further complicated by striking variations between laboratory mouse strains. Herein, we report an experimental approach to determine with precision and specificity expression levels of α-defensin (Defa) mRNA in the small intestine of C57BL/6 mice through an optimized set of oligonucleotide primers for qRT-PCR assays and cloned cDNA plasmids corresponding to the Defa paralogs. This approach demonstrated marked differences in α-defensin expression in C57BL/6 mice with respect to proximal/distal anatomical location and developmental stage, which have not been described previously. These data underscore the importance of careful attention to method (primer choice, proximal vs. distal location, and developmental stage) in analysis of antimicrobial peptide expression and their impact.


Subject(s)
Gene Expression Regulation, Developmental , Intestine, Small/metabolism , Paneth Cells/metabolism , RNA, Messenger/metabolism , alpha-Defensins/metabolism , Animals , Female , Male , Mice , Mice, Inbred C57BL , Paneth Cells/cytology , RNA, Messenger/genetics , alpha-Defensins/genetics
15.
Am J Physiol Gastrointest Liver Physiol ; 317(5): G580-G591, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31433211

ABSTRACT

Congenital tufting enteropathy (CTE) is an autosomal recessive disease characterized by severe intestinal failure in infancy and mutations in the epithelial cell adhesion molecule (EPCAM) gene. Previous studies of CTE in mice expressing mutant EpCAM show neonatal lethality. Hence, to study the cellular, molecular, and physiological alterations that result from EpCAM mutation, a tamoxifen-inducible mutant EpCAM enteroid model has been generated. The presence of mutant EpCAM in the model was confirmed at both mRNA and protein levels. Immunofluorescence microscopy demonstrated the reduced expression of mutant EpCAM. Mutant enteroids had reduced budding potential as well as significantly decreased mRNA expression for epithelial lineage markers (Mucin 2, lysozyme, sucrase-isomaltase), proliferation marker Ki67, and secretory pathway transcription factors (Atoh1, Hnf1b). Significantly decreased numbers of Paneth and goblet cells were confirmed by staining. These findings were correlated with intestinal tissue from CTE patients and the mutant mice model that had significantly fewer Paneth and goblet cells than in healthy counterparts. FITC-dextran studies demonstrated significantly impaired barrier function in monolayers derived from mutant enteroids compared with control monolayers. In conclusion, we have established an ex vivo CTE model. The role of EpCAM in the budding potential, differentiation, and barrier function of enteroids is noted. Our study establishes new facets of EpCAM biology that will aid in understanding the pathophysiology of CTE and role of EpCAM in health and disease.NEW & NOTEWORTHY Here, we develop a novel ex vivo enteroid model for congenital tufting enteropathy (CTE) based on epithelial cell adhesion molecule (EPCAM) gene mutations found in patients. With this model we demonstrate the role of EpCAM in maintaining the functional homeostasis of the intestinal epithelium, including differentiation, proliferation, and barrier integrity. This study further establishes a new direction in EpCAM biology that will help in understanding the detailed pathophysiology of CTE and role of EpCAM.


Subject(s)
Diarrhea, Infantile/genetics , Epithelial Cell Adhesion Molecule/genetics , Intestinal Mucosa/cytology , Malabsorption Syndromes/genetics , Tissue Culture Techniques/methods , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Diarrhea, Infantile/pathology , Epithelial Cell Adhesion Molecule/metabolism , Female , Goblet Cells/cytology , Goblet Cells/metabolism , Goblet Cells/physiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Malabsorption Syndromes/pathology , Male , Mice , Mice, Inbred C57BL , Paneth Cells/cytology , Paneth Cells/metabolism , Paneth Cells/physiology
16.
Nature ; 569(7754): 66-72, 2019 05.
Article in English | MEDLINE | ID: mdl-31019299

ABSTRACT

Intestinal organoids are complex three-dimensional structures that mimic the cell-type composition and tissue organization of the intestine by recapitulating the self-organizing ability of cell populations derived from a single intestinal stem cell. Crucial in this process is a first symmetry-breaking event, in which only a fraction of identical cells in a symmetrical sphere differentiate into Paneth cells, which generate the stem-cell niche and lead to asymmetric structures such as the crypts and villi. Here we combine single-cell quantitative genomic and imaging approaches to characterize the development of intestinal organoids from single cells. We show that their development follows a regeneration process that is driven by transient activation of the transcriptional regulator YAP1. Cell-to-cell variability in YAP1, emerging in symmetrical spheres, initiates Notch and DLL1 activation, and drives the symmetry-breaking event and formation of the first Paneth cell. Our findings reveal how single cells exposed to a uniform growth-promoting environment have the intrinsic ability to generate emergent, self-organized behaviour that results in the formation of complex multicellular asymmetric structures.


Subject(s)
Intestines/cytology , Organoids/cytology , Organoids/growth & development , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Calcium-Binding Proteins , Cell Cycle Proteins , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Organoids/metabolism , Paneth Cells/cytology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Single-Cell Analysis , YAP-Signaling Proteins
17.
Cell Rep ; 26(9): 2266-2273.e4, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30811977

ABSTRACT

Wnt signals at the base of mammalian crypts play a pivotal role in intestinal stem cell (ISC) homeostasis, whereas aberrant Wnt activation causes colon cancer. Precise control of Wnt signal strength is governed by a number of negative inhibitory mechanisms acting at distinct levels of the cascade. Here, we identify the Wnt negative regulatory role of Sh3bp4 in the intestinal crypt. We show that the loss of Sh3bp4 increases ISC and Paneth cell numbers in murine intestine and accelerates adenoma development in Apcmin mice. Mechanistically, human SH3BP4 inhibits Wnt signaling downstream of ß-catenin phosphorylation and ubiquitination. This Wnt inhibitory role is dependent on the ZU5 domain of SH3BP4. We further demonstrate that SH3BP4 is expressed at the perinuclear region to restrict nuclear localization of ß-catenin. Our data uncover the tumor-suppressive role of SH3BP4 that functions as a negative feedback regulator of Wnt signaling through modulating ß-catenin's subcellular localization.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Carcinogenesis , Intestinal Mucosa/metabolism , Stem Cells/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line, Tumor , Cell Nucleus , HEK293 Cells , Humans , Intestinal Mucosa/cytology , Mice , Mice, Knockout , Paneth Cells/cytology , Protein Domains , Wnt Proteins/metabolism
18.
Gut Microbes ; 10(1): 45-58, 2019.
Article in English | MEDLINE | ID: mdl-29883265

ABSTRACT

Paneth cells (PCs) are epithelial cells found in the small intestine, next to intestinal stem cells (ISCs) at the base of the crypts. PCs secrete antimicrobial peptides (AMPs) that regulate the commensal gut microbiota. In contrast, little is known regarding how the enteric microbiota reciprocally influences PC function. In this study, we sought to characterize the impact of the enteric microbiota on PC biology in the mouse small intestine. This was done by first enumerating jejunal PCs in germ-free (GF) versus conventionally raised (CR) mice. We next evaluated the possible functional consequences of altered PC biology in these experimental groups by assessing epithelial proliferation, ISC numbers, and the production of AMPs. We found that PC numbers were significantly increased in CR versus GF mice; however, there were no differences in ISC numbers or cycling activity between groups. Of the AMPs assessed, only Reg3γ transcript expression was significantly increased in CR mice. Intriguingly, this increase was abrogated in cultured CR versus GF enteroids, and could not be re-induced with various bacterial ligands. Our findings demonstrate the enteric microbiota regulates PC function by increasing PC numbers and inducing Reg3γ expression, though the latter effect may not involve direct interactions between bacteria and the intestinal epithelium. In contrast, the enteric microbiota does not appear to regulate jejunal ISC census and proliferation. These are critical findings for investigators using GF mice and the enteroid system to study PC and ISC biology.


Subject(s)
Gastrointestinal Microbiome , Intestine, Small/cytology , Intestine, Small/microbiology , Multipotent Stem Cells/cytology , Paneth Cells/cytology , Paneth Cells/metabolism , Animals , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , Cell Count , Cell Proliferation , Female , Germ-Free Life , Intestinal Mucosa/cytology , Mice, Inbred C57BL , Pancreatitis-Associated Proteins/genetics , Transcription, Genetic
19.
Cells ; 8(1)2018 12 22.
Article in English | MEDLINE | ID: mdl-30583538

ABSTRACT

Autophagy, an intracellular degradation mechanism, has many immunological functions and is a constitutive process necessary for maintaining cellular homeostasis and organ structure. One of the functions of autophagy is to control the innate immune response. Many studies conducted in recent years have revealed the contribution of autophagy to the innate immune response, and relationships between this process and various diseases have been reported. Inflammatory bowel disease is an intractable disorder with unknown etiology; however, immunological abnormalities in the intestines are known to be involved in the pathology of inflammatory bowel disease, as is dysfunction of autophagy. In Crohn's disease, many associations with autophagy-related genes, such as ATG16L1, IRGM, NOD2, and others, have been reported. Abnormalities in the ATG16L1 gene, in particular, have been reported to cause autophagic dysfunction, resulting in enhanced production of inflammatory cytokines by macrophages as well as abnormal function of Paneth cells, which are important in intestinal innate immunity. In this review, we provide an overview of the autophagy mechanism in innate immune cells in inflammatory bowel disease.


Subject(s)
Autophagy-Related Proteins , Autophagy , Crohn Disease , Immunity, Innate/genetics , Paneth Cells/pathology , Animals , Autophagy/genetics , Autophagy/immunology , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/immunology , Crohn Disease/genetics , Crohn Disease/immunology , Cytokines/immunology , Disease Models, Animal , GTP-Binding Proteins/genetics , GTP-Binding Proteins/immunology , Genetic Predisposition to Disease , Humans , Inflammasomes/immunology , Mice , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/immunology , Paneth Cells/cytology
20.
Cell Host Microbe ; 24(6): 833-846.e6, 2018 12 12.
Article in English | MEDLINE | ID: mdl-30543778

ABSTRACT

Symbionts play an indispensable role in gut homeostasis, but underlying mechanisms remain elusive. To clarify the role of lactic-acid-producing bacteria (LAB) on intestinal stem-cell (ISC)-mediated epithelial development, we fed mice with LAB-type symbionts such as Bifidobacterium and Lactobacillus spp. Here we show that administration of LAB-type symbionts significantly increased expansion of ISCs, Paneth cells, and goblet cells. Lactate stimulated ISC proliferation through Wnt/ß-catenin signals of Paneth cells and intestinal stromal cells. Moreover, Lactobacillus plantarum strains lacking lactate dehydrogenase activity, which are deficient in lactate production, elicited less ISC proliferation. Pre-treatment with LAB-type symbionts or lactate protected mice in response to gut injury provoked by combined treatments with radiation and a chemotherapy drug. Impaired ISC-mediated epithelial development was found in mice deficient of the lactate G-protein-coupled receptor, Gpr81. Our results demonstrate that LAB-type symbiont-derived lactate plays a pivotal role in promoting ISC-mediated epithelial development in a Gpr81-dependent manner.


Subject(s)
Goblet Cells/cytology , Lactic Acid/metabolism , Lactobacillus plantarum/metabolism , Paneth Cells/cytology , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Goblet Cells/drug effects , Goblet Cells/radiation effects , HEK293 Cells , Humans , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Lactobacillus plantarum/genetics , Methotrexate/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Paneth Cells/drug effects , Paneth Cells/radiation effects , Receptors, G-Protein-Coupled/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...