Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Gut Microbes ; 15(2): 2281012, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37992398

ABSTRACT

The intestinal epithelium is constantly exposed to microbes residing in the lumen. Traditionally, the response to microbial interactions has been studied in cell lines derived from cancerous tissues, e.g. Caco-2. It is, however, unclear how the responses in these cancer cell lines reflect the responses of a normal epithelium and whether there might be microbial strain-specific effects. To address these questions, we derived organoids from the small intestine from a cohort of healthy individuals. Culturing intestinal epithelium on a flat laminin matrix induced their differentiation, facilitating analysis of microbial responses via the apical membrane normally exposed to the luminal content. Here, it was evident that the healthy epithelium across multiple individuals (n = 9) demonstrates robust acute both common and strain-specific responses to a range of probiotic bacterial strains (BB-12Ⓡ, LGGⓇ, DSM33361, and Bif195). Importantly, parallel experiments using the Caco-2 cell line provide no acute response. Collectively, we demonstrate that primary epithelial cells maintained as organoids represent a valuable resource for assessing interactions between the epithelium and luminal microbes across individuals, and that these models are likely to contribute to a better understanding of host microbe interactions.


Subject(s)
Gastrointestinal Microbiome , Humans , Caco-2 Cells , Epithelial Cells/metabolism , Organoids , Epithelium , Intestinal Mucosa/microbiology
2.
Sci Adv ; 9(28): eadf9460, 2023 07 14.
Article in English | MEDLINE | ID: mdl-37436997

ABSTRACT

During intestinal organogenesis, equipotent epithelial progenitors mature into phenotypically distinct stem cells that are responsible for lifelong maintenance of the tissue. While the morphological changes associated with the transition are well characterized, the molecular mechanisms underpinning the maturation process are not fully understood. Here, we leverage intestinal organoid cultures to profile transcriptional, chromatin accessibility, DNA methylation, and three-dimensional (3D) chromatin conformation landscapes in fetal and adult epithelial cells. We observed prominent differences in gene expression and enhancer activity, which are accompanied by local changes in 3D organization, DNA accessibility, and methylation between the two cellular states. Using integrative analyses, we identified sustained Yes-Associated Protein (YAP) transcriptional activity as a major gatekeeper of the immature fetal state. We found the YAP-associated transcriptional network to be regulated at various levels of chromatin organization and likely to be coordinated by changes in extracellular matrix composition. Together, our work highlights the value of unbiased profiling of regulatory landscapes for the identification of key mechanisms underlying tissue maturation.


Subject(s)
Epigenomics , Intestinal Mucosa , Adult , Humans , Intestines , Epithelium , Chromatin/genetics
4.
Nat Commun ; 13(1): 715, 2022 02 07.
Article in English | MEDLINE | ID: mdl-35132078

ABSTRACT

Organs are anatomically compartmentalised to cater for specialised functions. In the small intestine (SI), regionalisation enables sequential processing of food and nutrient absorption. While several studies indicate the critical importance of non-epithelial cells during development and homeostasis, the extent to which these cells contribute to regionalisation during morphogenesis remains unexplored. Here, we identify a mesenchymal-epithelial crosstalk that shapes the developing SI during late morphogenesis. We find that subepithelial mesenchymal cells are characterised by gradients of factors supporting Wnt signalling and stimulate epithelial growth in vitro. Such a gradient impacts epithelial gene expression and regional villus formation along the anterior-posterior axis of the SI. Notably, we further provide evidence that Wnt signalling directly regulates epithelial expression of Sonic Hedgehog (SHH), which, in turn, acts on mesenchymal cells to drive villi formation. Taken together our results uncover a mechanistic link between Wnt and Hedgehog signalling across different cellular compartments that is central for anterior-posterior regionalisation and correct formation of the SI.


Subject(s)
Hedgehog Proteins/metabolism , Intestinal Mucosa/metabolism , Intestine, Small/embryology , Mesenchymal Stem Cells/metabolism , Wnt Signaling Pathway/physiology , Animals , Cell Lineage , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Intestinal Mucosa/cytology , Intestinal Mucosa/embryology , Intestine, Small/cytology , Intestine, Small/metabolism , Mesenchymal Stem Cells/cytology , Mice , Morphogenesis , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Wnt Signaling Pathway/genetics
5.
Trends Endocrinol Metab ; 31(11): 848-858, 2020 11.
Article in English | MEDLINE | ID: mdl-33086077

ABSTRACT

Dietary patterns, microbiome dysbiosis, and gut microbial metabolites (GMMs) have a pivotal role in the homeostasis of intestinal epithelial cells and in disease progression, such as that of colorectal cancer (CRC). Although GMMs and microorganisms have crucial roles in many biological activities, models for deciphering diet-microbiome-host relationships are largely limited to animal models. Thus, intestinal organoids (IOs) have provided unprecedented opportunities for the generation of in vitro platforms with the sufficient level of complexity to model physiological and pathological diet-microbiome-host conditions. Overall, IO responses to GMM metabolites and microorganisms can provide new insights into the mechanisms by which those agents may prevent or trigger diseases, significantly extending our knowledge of diet-microbiome-host interactions.


Subject(s)
Gastrointestinal Microbiome/physiology , Organoids/microbiology , Animals , Humans , Microbiota/physiology , Single-Cell Analysis
6.
Sci Rep ; 8(1): 15255, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30323305

ABSTRACT

The processes involved in renewal of the epithelium that lines the mouse stomach remain unclear. Apart from the cells in the isthmus, several other populations located deeper in the gastric glands have been suggested to contribute to the maintenance of the gastric epithelium. Here, we reveal that Lrig1 is expressed in the basal layer of the forestomach and the lower part of glands in the corpus and pylorus. In the glandular epithelium of the stomach, Lrig1 marks a heterogeneous population comprising mainly non-proliferative cells. Yet, fate-mapping experiments using a knock-in mouse line expressing Cre specifically in Lrig1+ cells demonstrate that these cells are able to contribute to the long-term maintenance of the gastric epithelium. Moreover, when cultured in vitro, cells expressing high level of Lrig1 have much higher organoid forming potential than the corresponding cellular populations expressing lower levels of Lrig1. Taken together, these observations show that Lrig1 is expressed primarily by differentiated cells, but that these cells can be recruited to contribute to the maintenance of the gastric epithelium. This confirms previous observations that cells located in the lower segments of gastric glands can participate in tissue replenishment.


Subject(s)
Biomarkers , Cell Proliferation , Epithelial Cells/metabolism , Gastric Mucosa/metabolism , Membrane Glycoproteins/physiology , Nerve Tissue Proteins/physiology , Animals , Biomarkers/metabolism , Cell Dedifferentiation/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/physiology , Gastric Mucosa/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Stomach/cytology
7.
Cell Stem Cell ; 22(1): 35-49.e7, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29249464

ABSTRACT

Tissue regeneration requires dynamic cellular adaptation to the wound environment. It is currently unclear how this is orchestrated at the cellular level and how cell fate is affected by severe tissue damage. Here we dissect cell fate transitions during colonic regeneration in a mouse dextran sulfate sodium (DSS) colitis model, and we demonstrate that the epithelium is transiently reprogrammed into a primitive state. This is characterized by de novo expression of fetal markers as well as suppression of markers for adult stem and differentiated cells. The fate change is orchestrated by remodeling the extracellular matrix (ECM), increased FAK/Src signaling, and ultimately YAP/TAZ activation. In a defined cell culture system recapitulating the extracellular matrix remodeling observed in vivo, we show that a collagen 3D matrix supplemented with Wnt ligands is sufficient to sustain endogenous YAP/TAZ and induce conversion of cell fate. This provides a simple model for tissue regeneration, implicating cellular reprogramming as an essential element.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cellular Reprogramming , Extracellular Matrix/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Phosphoproteins/metabolism , Regeneration , Animals , Biomarkers/metabolism , Cell Cycle Proteins , Fetus/metabolism , Humans , Mechanotransduction, Cellular , Mice, Inbred C57BL , Signal Transduction , Transcription, Genetic , Transcriptional Activation/genetics , YAP-Signaling Proteins
9.
Mol Oncol ; 11(11): 1616-1629, 2017 11.
Article in English | MEDLINE | ID: mdl-28881081

ABSTRACT

Transgenic porcine cancer models bring novel possibilities for research. Their physical similarities with humans enable the use of surgical procedures and treatment approaches used for patients, which facilitates clinical translation. Here, we aimed to develop an inducible oncopig model of intestinal cancer. Transgenic (TG) minipigs were generated using somatic cell nuclear transfer by handmade cloning. The pigs encode two TG cassettes: (a) an Flp recombinase-inducible oncogene cassette containing KRAS-G12D, cMYC, SV40LT - which inhibits p53 - and pRB and (b) a 4-hydroxytamoxifen (4-OHT)-inducible Flp recombinase activator cassette controlled by the intestinal epithelium-specific villin promoter. Thirteen viable transgenic minipigs were born. The ability of 4-OHT to activate the oncogene cassette was confirmed in vitro in TG colonic organoids and ex vivo in tissue biopsies obtained by colonoscopy. In order to provide proof of principle that the oncogene cassette could also successfully be activated in vivo, three pigs were perorally treated with 400 mg tamoxifen for 2 × 5 days. After two months, one pig developed a duodenal neuroendocrine carcinoma with a lymph node metastasis. Molecular analysis of the carcinoma and metastasis confirmed activation of the oncogene cassette. No tumor formation was observed in untreated TG pigs or in the remaining two treated pigs. The latter indicates that tamoxifen delivery can probably be improved. In summary, we have generated a novel inducible oncopig model of intestinal cancer, which has the ability to form metastatic disease already two months after induction. The model may be helpful in bridging the gap between basic research and clinical usage. It opens new venues for longitudinal studies of tumor development and evolution, for preclinical assessment of new anticancer regimens, for pharmacology and toxicology assessments, as well as for studies into biological mechanisms of tumor formation and metastasis.


Subject(s)
Animals, Genetically Modified/genetics , Cloning, Organism/methods , Disease Models, Animal , Intestinal Neoplasms/genetics , Nuclear Transfer Techniques , Swine, Miniature/genetics , Animals , Embryo Culture Techniques/methods , Embryo Transfer/methods , Female , Humans , Intestinal Mucosa/metabolism , Intestinal Neoplasms/pathology , Intestines/pathology , Swine
10.
Curr Opin Cell Biol ; 43: 22-29, 2016 12.
Article in English | MEDLINE | ID: mdl-27474805

ABSTRACT

Reliable disease models are needed in order to improve quality of healthcare. This includes gaining better understanding of disease mechanisms, developing new therapeutic interventions and personalizing treatment. Up-to-date, the majority of our knowledge about disease states comes from in vivo animal models and in vitro cell culture systems. However, it has been exceedingly difficult to model disease at the tissue level. Since recently, the gap between cell line studies and in vivo modeling has been narrowing thanks to progress in biomaterials and stem cell research. Development of reliable 3D culture systems has enabled a rapid expansion of sophisticated in vitro models. Here we focus on some of the latest advances and future perspectives in 3D organoids for human disease modeling.


Subject(s)
Disease , Models, Biological , Organoids/pathology , Tissue Culture Techniques/methods , Animals , Humans
11.
J Invest Dermatol ; 135(11): 2753-2763, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26121213

ABSTRACT

Activation of Wnt/ß-catenin signaling in adult mouse epidermis leads to expansion of the stem cell compartment and redirects keratinocytes in the interfollicular epidermis and sebaceous glands (SGs) to differentiate along the hair follicle (HF) lineages. Here we demonstrate that during epidermal development and homeostasis there is reciprocal activation of the androgen receptor (AR) and ß-catenin in cells of the HF bulb. AR activation reduced ß-catenin-dependent transcription, blocked ß-catenin-induced induction of HF growth, and prevented ß-catenin-mediated conversion of SGs into HFs. Conversely, AR inhibition enhanced the effects of ß-catenin activation, promoting HF proliferation and differentiation, culminating in the formation of benign HF tumors and a complete loss of SG identity. We conclude that AR signaling has a key role in epidermal stem cell fate selection by modulating responses to ß-catenin in adult mouse skin.


Subject(s)
Epithelial Cells/metabolism , Hair Follicle/growth & development , Receptors, Androgen/metabolism , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , Animals , Cell Differentiation , Cells, Cultured , Epithelial Cells/cytology , Hair Follicle/metabolism , Humans , Keratinocytes/metabolism , Male , Mice , Mice, Transgenic , Models, Animal , Real-Time Polymerase Chain Reaction , Sebaceous Glands/cytology , Sensitivity and Specificity , Stem Cells/metabolism
12.
Cell Stem Cell ; 13(6): 734-44, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24139758

ABSTRACT

Regeneration and homeostasis in the adult intestinal epithelium is driven by proliferative resident stem cells, whose functional properties during organismal development are largely unknown. Here, we show that human and mouse fetal intestine contains proliferative, immature progenitors, which can be expanded in vitro as Fetal Enterospheres (FEnS). A highly similar progenitor population can be established during intestinal differentiation of human induced pluripotent stem cells. Established cultures of mouse fetal intestinal progenitors express lower levels of Lgr5 than mature progenitors and propagate in the presence of the Wnt antagonist Dkk1, and new cultures can be induced to form mature intestinal organoids by exposure to Wnt3a. Following transplantation in a colonic injury model, FEnS contribute to regeneration of colonic epithelium by forming epithelial crypt-like structures expressing region-specific differentiation markers. This work provides insight into mechanisms underlying development of the mammalian intestine and points to future opportunities for patient-specific regeneration of the digestive tract.


Subject(s)
Colon/injuries , Colon/physiology , Fetus/cytology , Intestines/embryology , Regeneration , Stem Cell Transplantation , Stem Cells/cytology , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Cell Differentiation , Cell Proliferation , Colon/cytology , Colon/pathology , Humans , Intestinal Mucosa/pathology , Intestines/cytology , Mice , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Signal Transduction , Spheroids, Cellular/pathology , Stem Cells/metabolism
14.
Cell Rep ; 3(2): 427-41, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23403291

ABSTRACT

Although the sebaceous gland (SG) plays an important role in skin function, the mechanisms regulating SG differentiation and carcinoma formation are poorly understood. We previously reported that c-MYC overexpression stimulates SG differentiation. We now demonstrate roles for the androgen receptor (AR) and p53. MYC-induced SG differentiation was reduced in mice lacking a functional AR. High levels of MYC triggered a p53-dependent DNA damage response, leading to accumulation of proliferative SG progenitors and inhibition of AR signaling. Conversely, testosterone treatment or p53 deletion activated AR signaling and restored MYC-induced differentiation. Poorly differentiated human sebaceous carcinomas exhibited high p53 and low AR expression. Thus, the consequences of overactivating MYC in the SG depend on whether AR or p53 is activated, as they form a regulatory axis controlling proliferation and differentiation.


Subject(s)
Proto-Oncogene Proteins c-myc/metabolism , Receptors, Androgen/metabolism , Sebaceous Glands/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Differentiation/drug effects , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/metabolism , Receptors, Androgen/genetics , Sebaceous Gland Neoplasms/metabolism , Sebaceous Gland Neoplasms/pathology , Sebaceous Glands/cytology , Signal Transduction/drug effects , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Testosterone/pharmacology , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...