Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Nat Cell Biol ; 26(2): 250-262, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38321203

ABSTRACT

A key aspect of nutrient absorption is the exquisite division of labour across the length of the small intestine, with individual nutrients taken up at different proximal:distal positions. For millennia, the small intestine was thought to comprise three segments with indefinite borders: the duodenum, jejunum and ileum. By examining the fine-scale longitudinal transcriptional patterns that span the mouse and human small intestine, we instead identified five domains of nutrient absorption that mount distinct responses to dietary changes, and three regional stem cell populations. Molecular domain identity can be detected with machine learning, which provides a systematic method to computationally identify intestinal domains in mice. We generated a predictive model of transcriptional control of domain identity and validated the roles of Ppar-δ and Cdx1 in patterning lipid metabolism-associated genes. These findings represent a foundational framework for the zonation of absorption across the mammalian small intestine.


Subject(s)
Duodenum , Intestine, Small , Humans , Mice , Animals , Intestine, Small/metabolism , Duodenum/metabolism , Intestines , Jejunum/metabolism , Ileum/metabolism , Mammals
2.
bioRxiv ; 2023 Sep 22.
Article in English | MEDLINE | ID: mdl-37790430

ABSTRACT

A key aspect of nutrient absorption is the exquisite division of labor across the length of the small intestine, with individual classes of micronutrients taken up at different positions. For millennia, the small intestine was thought to comprise three segments with indefinite borders: the duodenum, jejunum, and ileum. By examining fine-scale longitudinal segmentation of the mouse and human small intestines, we identified transcriptional signatures and upstream regulatory factors that define five domains of nutrient absorption, distinct from the three traditional sections. Spatially restricted expression programs were most prominent in nutrient-absorbing enterocytes but initially arose in intestinal stem cells residing in three regional populations. While a core signature was maintained across mice and humans with different diets and environments, domain properties were influenced by dietary changes. We established the functions of Ppar-ẟ and Cdx1 in patterning lipid metabolism in distal domains and generated a predictive model of additional transcription factors that direct domain identity. Molecular domain identity can be detected with machine learning, representing the first systematic method to computationally identify specific intestinal regions in mice. These findings provide a foundational framework for the identity and control of longitudinal zonation of absorption along the proximal:distal small intestinal axis.

3.
JCI Insight ; 8(6)2023 03 22.
Article in English | MEDLINE | ID: mdl-36821371

ABSTRACT

Epithelial organoids derived from intestinal tissue, called enteroids, recapitulate many aspects of the organ in vitro and can be used for biological discovery, personalized medicine, and drug development. Here, we interrogated the cell signaling environment within the developing human intestine to identify niche cues that may be important for epithelial development and homeostasis. We identified an EGF family member, EPIREGULIN (EREG), which is robustly expressed in the developing human crypt. Enteroids generated from the developing human intestine grown in standard culture conditions, which contain EGF, are dominated by stem and progenitor cells and feature little differentiation and no spatial organization. Our results demonstrate that EREG can replace EGF in vitro, and EREG leads to spatially resolved enteroids that feature budded and proliferative crypt domains and a differentiated villus-like central lumen. Multiomic (transcriptome plus epigenome) profiling of native crypts, EGF-grown enteroids, and EREG-grown enteroids showed that EGF enteroids have an altered chromatin landscape that is dependent on EGF concentration, downregulate the master intestinal transcription factor CDX2, and ectopically express stomach genes, a phenomenon that is reversible. This is in contrast to EREG-grown enteroids, which remain intestine like in culture. Thus, EREG creates a homeostatic intestinal niche in vitro, enabling interrogation of stem cell function, cellular differentiation, and disease modeling.


Subject(s)
Epidermal Growth Factor , Intestines , Humans , Epiregulin , Intestinal Mucosa , Cell Differentiation
4.
Cell Stem Cell ; 30(1): 10-19, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36608674

ABSTRACT

The development of an organism depends on intrinsic genetic programs of progenitor cells and their spatiotemporally complex extrinsic environment. Ex vivo generation of organoids from progenitor cells provides a platform for recapitulating and exploring development. Current approaches rely largely on soluble morphogens or engineered biomaterials to manipulate the physical environment, but the emerging field of synthetic biology provides a powerful toolbox to genetically manipulate cell communication, adhesion, and even cell fate. Applying these modular tools to organoids should lead to a deeper understanding of developmental principles, improved organoid models, and an enhanced capability to design tissues for regenerative purposes.


Subject(s)
Organoids , Synthetic Biology , Biocompatible Materials , Stem Cells , Cell Differentiation
5.
Cell Mol Gastroenterol Hepatol ; 15(3): 689-715, 2023.
Article in English | MEDLINE | ID: mdl-36356835

ABSTRACT

BACKGROUND & AIMS: Axin1 is a negative regulator of wingless-type MMTV integration site family, member 1 (Wnt)/ß-catenin signaling with tumor-suppressor function. The Wnt pathway has a critical role in the intestine, both during homeostasis and cancer, but the role of Axin1 remains elusive. METHODS: We assessed the role of Axin1 in normal intestinal homeostasis, with control, epithelial-specific, Axin1-knockout mice (Axin1ΔIEC) and Axin2-knockout mice. We evaluated the tumor-suppressor function of Axin1 during chemically induced colorectal tumorigenesis and dextran sulfate sodium-induced colitis, and performed comparative gene expression profiling by whole-genome RNA sequencing. The clinical relevance of the Axin1-dependent gene expression signature then was tested in a database of 2239 clinical colorectal cancer (CRC) samples. RESULTS: We found that Axin1 was dispensable for normal intestinal homeostasis and redundant with Axin2 for Wnt pathway down-regulation. Axin1 deficiency in intestinal epithelial cells rendered mice more susceptible to chemically induced colon carcinogenesis, but reduced dextran sulfate sodium-induced colitis by attenuating the induction of a proinflammatory program. RNA-seq analyses identified an interferon γ/T-helper1 immune program controlled by Axin1 that enhances the inflammatory response and protects against CRC. The Axin1-dependent gene expression signature was applied to human CRC samples and identified a group of patients with potential vulnerability to immune checkpoint blockade therapies. CONCLUSIONS: Our study establishes, in vivo, that Axin1 has redundant function with Axin2 for Wnt down-regulation and infers a new role for Axin1. Physiologically, Axin1 stimulates gut inflammation via an interferon γ/Th1 program that prevents tumor growth. Linked to its T-cell-mediated effect, the colonic Axin1 signature offers therapeutic perspectives for CRC.


Subject(s)
Colitis , Interferon-gamma , Mice , Animals , Humans , Dextran Sulfate/toxicity , Carcinogenesis/genetics , Colitis/chemically induced , Wnt Signaling Pathway/genetics , Mice, Knockout , Axin Protein/genetics , Axin Protein/metabolism
6.
Nature ; 614(7946): 144-152, 2023 02.
Article in English | MEDLINE | ID: mdl-36509107

ABSTRACT

Cell adhesion molecules are ubiquitous in multicellular organisms, specifying precise cell-cell interactions in processes as diverse as tissue development, immune cell trafficking and the wiring of the nervous system1-4. Here we show that a wide array of synthetic cell adhesion molecules can be generated by combining orthogonal extracellular interactions with intracellular domains from native adhesion molecules, such as cadherins and integrins. The resulting molecules yield customized cell-cell interactions with adhesion properties that are similar to native interactions. The identity of the intracellular domain of the synthetic cell adhesion molecules specifies interface morphology and mechanics, whereas diverse homotypic or heterotypic extracellular interaction domains independently specify the connectivity between cells. This toolkit of orthogonal adhesion molecules enables the rationally programmed assembly of multicellular architectures, as well as systematic remodelling of native tissues. The modularity of synthetic cell adhesion molecules provides fundamental insights into how distinct classes of cell-cell interfaces may have evolved. Overall, these tools offer powerful abilities for cell and tissue engineering and for systematically studying multicellular organization.


Subject(s)
Cell Adhesion Molecules , Cell Communication , Synthetic Biology , Cadherins/chemistry , Cell Adhesion , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/metabolism , Integrins/chemistry , Synthetic Biology/methods , Protein Domains , Binding Sites , Cell Engineering
7.
Dev Cell ; 56(3): 356-365.e9, 2021 02 08.
Article in English | MEDLINE | ID: mdl-33484640

ABSTRACT

Renewing tissues have the remarkable ability to continually produce both proliferative progenitor and specialized differentiated cell types. How are complex milieus of microenvironmental signals interpreted to coordinate tissue-cell-type composition? Here, we investigate the responses of intestinal epithelium to individual and paired perturbations across eight epithelial signaling pathways. Using a high-throughput approach that combines enteroid monolayers and quantitative imaging, we identified conditions that enrich for specific cell types as well as interactions between pathways. Importantly, we found that modulation of transit-amplifying cell proliferation changes the ratio of differentiated secretory to absorptive cell types. These observations highlight an underappreciated role for transit-amplifying cells in the tuning of differentiated cell-type composition.


Subject(s)
Epithelial Cells/cytology , Intestines/cytology , Animals , Cell Proliferation , Epithelial Cells/metabolism , ErbB Receptors/metabolism , Humans , Interleukin-4/metabolism , Intestinal Absorption , Male , Mice, Inbred C57BL , Models, Biological , Organoids/cytology , Protein Interaction Mapping , Signal Transduction
8.
Proc Natl Acad Sci U S A ; 117(20): 11136-11146, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32371487

ABSTRACT

The intestinal epithelium acts as a barrier between the organism and its microenvironment, including the gut microbiota. It is the most rapidly regenerating tissue in the human body thanks to a pool of intestinal stem cells (ISCs) expressing Lgr5 The intestinal epithelium has to cope with continuous stress linked to its digestive and barrier functions. Epithelial repair is crucial to maintain its integrity, and Lgr5-positive intestinal stem cell (Lgr5+ISC) resilience following cytotoxic stresses is central to this repair stage. We show here that autophagy, a pathway allowing the lysosomal degradation of intracellular components, plays a crucial role in the maintenance and genetic integrity of Lgr5+ISC under physiological and stress conditions. Using conditional mice models lacking the autophagy gene Atg7 specifically in all intestinal epithelial cells or in Lgr5+ISC, we show that loss of Atg7 induces the p53-mediated apoptosis of Lgr5+ISC. Mechanistically, this is due to increasing oxidative stress, alterations to interactions with the microbiota, and defective DNA repair. Following irradiation, we show that Lgr5+ISC repair DNA damage more efficiently than their progenitors and that this protection is Atg7 dependent. Accordingly, we found that the stimulation of autophagy on fasting protects Lgr5+ISC against DNA damage and cell death mediated by oxaliplatin and doxorubicin treatments. Finally, p53 deletion prevents the death of Atg7-deficient Lgr5+ISC but promotes genetic instability and tumor formation. Altogether, our findings provide insights into the mechanisms underlying maintenance and integrity of ISC and highlight the key functions of Atg7 and p53.


Subject(s)
Autophagy-Related Protein 7/metabolism , Autophagy/physiology , Intestines/physiology , Stem Cells/metabolism , Animals , Apoptosis , Autophagy-Related Protein 7/genetics , DNA Damage , DNA Repair , Disease Models, Animal , Epithelial Cells/metabolism , Female , Genes, p53/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestines/pathology , Male , Mice , Mice, Knockout , Receptors, G-Protein-Coupled/metabolism , Stem Cells/cytology
9.
J Hepatol ; 73(3): 628-639, 2020 09.
Article in English | MEDLINE | ID: mdl-32205193

ABSTRACT

BACKGROUND & AIMS: Paneth cells (PCs) synthesize and secrete antimicrobial peptides that are key mediators of host-microbe interactions, establishing a balance between intestinal microflora and enteric pathogens. We observed that their number increases in experimental portal hypertension and aimed to investigate the mechanisms by which these cells can contribute to the regulation of portal pressure. METHODS: We first treated Math1Lox/LoxVilcreERT2 mice with tamoxifen to induce the complete depletion of intestinal PCs. Subsequently, we performed partial portal vein or bile duct ligation. We then studied the effects of these interventions on hemodynamic parameters, proliferation of blood vessels and the expression of genes regulating angiogenesis. Intestinal organoids were cultured and exposed to different microbial products to study the composition of their secreted products (by proteomics) and their effects on the proliferation and tube formation of endothelial cells (ECs). In vivo confocal laser endomicroscopy was used to confirm the findings on blood vessel proliferation. RESULTS: Portal hypertension was significantly attenuated in PC-depleted mice compared to control mice and was associated with a decrease in portosystemic shunts. Depletion of PCs also resulted in a significantly decreased density of blood vessels in the intestinal wall and mesentery. Furthermore, we observed reduced expression of intestinal genes regulating angiogenesis in Paneth cell depleted mice using arrays and next generation sequencing. Tube formation and wound healing responses were significantly decreased in ECs treated with conditioned media from PC-depleted intestinal organoids exposed to intestinal microbiota-derived products. Proteomic analysis of conditioned media in the presence of PCs revealed an increase in factors regulating angiogenesis and additional metabolic processes. In vivo endomicroscopy showed decreased vascular proliferation in the absence of PCs. CONCLUSIONS: These results suggest that in response to intestinal flora and microbiota-derived factors, PCs secrete not only antimicrobial peptides, but also pro-angiogenic signaling molecules, thereby promoting intestinal and mesenteric angiogenesis and regulating portal hypertension. LAY SUMMARY: Paneth cells are present in the lining of the small intestine. They prevent the passage of bacteria from the intestine into the blood circulation by secreting substances to fight bacteria. In this paper, we discovered that these substances not only act against bacteria, but also increase the quantity of blood vessels in the intestine and blood pressure in the portal vein. This is important, because high blood pressure in the portal vein may result in several complications which could be targeted with novel approaches.


Subject(s)
Escherichia coli Infections/metabolism , Escherichia coli/metabolism , Gastrointestinal Microbiome/genetics , Hypertension, Portal/metabolism , Hypertension, Portal/microbiology , Neovascularization, Pathologic/metabolism , Paneth Cells/metabolism , Animals , Culture Media, Conditioned , Disease Models, Animal , Escherichia coli Infections/microbiology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intestine, Small/metabolism , Intestine, Small/microbiology , Male , Mice , Mice, Transgenic , Organoids/metabolism , Organoids/microbiology , Paneth Cells/drug effects , Pore Forming Cytotoxic Proteins/metabolism , Proteome , Proteomics/methods , Tamoxifen/pharmacology
10.
Curr Opin Cell Biol ; 63: 88-101, 2020 04.
Article in English | MEDLINE | ID: mdl-32036295

ABSTRACT

Stem cell behavior is tightly regulated by spatiotemporal signaling from the niche, which is a four-dimensional microenvironment that can instruct stem cells to remain quiescent, self-renew, proliferate, or differentiate. In this review, we discuss recent advances in understanding the signaling cues provided by the stem cell niche in two contrasting adult tissues, the rapidly cycling intestinal epithelium and the slowly renewing skeletal muscle. Drawing comparisons between these two systems, we discuss the effects of niche-derived growth factors and signaling molecules, metabolic cues, the extracellular matrix and biomechanical cues, and immune signals on stem cells. We also discuss the influence of the niche in defining stem cell identity and function in both normal and pathophysiologic states.


Subject(s)
Adult Stem Cells/metabolism , Gastrointestinal Microbiome/genetics , Stem Cell Niche/physiology , Cell Differentiation , Humans , Signal Transduction
11.
Med Sci (Paris) ; 33(3): 290-296, 2017 Mar.
Article in French | MEDLINE | ID: mdl-28367816

ABSTRACT

The intestinal epithelial cells are crucial mediators of intestinal homeostasis. The intestinal epithelium is the largest of the body's mucosal surfaces exposed to the environment. Intestinal homeostasis is essentially based on the maintenance of intestinal epithelial cell integrity, a complex process involving a balance between the intestinal flora, the immune system and the energy expenses linked to metabolism. Autophagy appears to be central to these functions and allows the epithelium to adapt to its environment and different stress situations by participating in antibacterial defense, by controlling the composition of the intestinal flora and the immune response, and by participating in energy homeostasis. Alterations of this protective mechanism are involved in inflammatory bowel diseases and colorectal cancer.


Subject(s)
Autophagy/physiology , Homeostasis/physiology , Intestines/physiology , Animals , Colorectal Neoplasms/pathology , Colorectal Neoplasms/physiopathology , Humans , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/physiopathology , Intestinal Diseases/pathology , Intestinal Diseases/physiopathology , Intestinal Mucosa/pathology , Intestinal Mucosa/physiology , Intestines/pathology
12.
Nat Cell Biol ; 17(8): 1062-73, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26214133

ABSTRACT

Here, we show that autophagy is activated in the intestinal epithelium in murine and human colorectal cancer and that the conditional inactivation of Atg7 in intestinal epithelial cells inhibits the formation of pre-cancerous lesions in Apc(+/-) mice by enhancing anti-tumour responses. The antibody-mediated depletion of CD8(+) T cells showed that these cells are essential for the anti-tumoral responses mediated by the inhibition of autophagy. We show that Atg7 deficiency leads to intestinal dysbiosis and that the microbiota is required for anticancer responses. In addition, Atg7 deficiency resulted in a stress response accompanied by metabolic defects, AMPK activation and p53-mediated cell-cycle arrest in tumour cells but not in normal tissue. This study reveals that the inhibition of autophagy within the epithelium may prevent the development and progression of colorectal cancer in genetically predisposed patients.


Subject(s)
Adenocarcinoma/prevention & control , Adenoma/prevention & control , Cell Transformation, Neoplastic/metabolism , Colon/metabolism , Colorectal Neoplasms/prevention & control , Immunity, Mucosal , Microbiota/immunology , Microtubule-Associated Proteins/deficiency , Ubiquitin-Activating Enzymes/metabolism , AMP-Activated Protein Kinases/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/microbiology , Adenocarcinoma/pathology , Adenoma/genetics , Adenoma/immunology , Adenoma/microbiology , Adenoma/pathology , Animals , Autophagy , Autophagy-Related Protein 7 , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/microbiology , Cell Cycle Checkpoints , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/pathology , Colon/immunology , Colon/microbiology , Colon/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/pathology , Disease Models, Animal , Dysbiosis , Enzyme Activation , Female , Genes, APC , Host-Pathogen Interactions , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/genetics , Time Factors , Tumor Burden , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL