Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.700
Filter
Add more filters

Publication year range
1.
Cell ; 182(6): 1606-1622.e23, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32888429

ABSTRACT

The enteric nervous system (ENS) coordinates diverse functions in the intestine but has eluded comprehensive molecular characterization because of the rarity and diversity of cells. Here we develop two methods to profile the ENS of adult mice and humans at single-cell resolution: RAISIN RNA-seq for profiling intact nuclei with ribosome-bound mRNA and MIRACL-seq for label-free enrichment of rare cell types by droplet-based profiling. The 1,187,535 nuclei in our mouse atlas include 5,068 neurons from the ileum and colon, revealing extraordinary neuron diversity. We highlight circadian expression changes in enteric neurons, show that disease-related genes are dysregulated with aging, and identify differences between the ileum and proximal/distal colon. In humans, we profile 436,202 nuclei, recovering 1,445 neurons, and identify conserved and species-specific transcriptional programs and putative neuro-epithelial, neuro-stromal, and neuro-immune interactions. The human ENS expresses risk genes for neuropathic, inflammatory, and extra-intestinal diseases, suggesting neuronal contributions to disease.


Subject(s)
Enteric Nervous System/cytology , Enteric Nervous System/metabolism , Gene Expression Regulation, Developmental/genetics , Neurons/metabolism , Nissl Bodies/metabolism , RNA, Messenger/metabolism , Single-Cell Analysis/methods , Aging/genetics , Aging/metabolism , Animals , Circadian Clocks/genetics , Colon/cytology , Colon/metabolism , Endoplasmic Reticulum, Rough/genetics , Endoplasmic Reticulum, Rough/metabolism , Endoplasmic Reticulum, Rough/ultrastructure , Epithelial Cells/metabolism , Female , Genetic Predisposition to Disease/genetics , Humans , Ileum/cytology , Ileum/metabolism , Inflammation/genetics , Inflammation/metabolism , Intestinal Diseases/genetics , Intestinal Diseases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Neurons/cytology , Nissl Bodies/genetics , Nissl Bodies/ultrastructure , RNA, Messenger/genetics , RNA-Seq , Ribosomes/metabolism , Ribosomes/ultrastructure , Stromal Cells/metabolism
2.
Nat Immunol ; 22(6): 699-710, 2021 06.
Article in English | MEDLINE | ID: mdl-34040226

ABSTRACT

It is increasingly recognized that immune development within mucosal tissues is under the control of environmental factors during early life. However, the cellular mechanisms that underlie such temporally and regionally restrictive governance of these processes are unclear. Here, we uncover an extrathymic pathway of immune development within the colon that is controlled by embryonic but not bone marrow-derived macrophages, which determines the ability of these organs to receive invariant natural killer T (iNKT) cells and allow them to establish local residency. Consequently, early-life perturbations of fetal-derived macrophages result in persistent decreases of mucosal iNKT cells and is associated with later-life susceptibility or resistance to iNKT cell-associated mucosal disorders. These studies uncover a host developmental program orchestrated by ontogenically distinct macrophages that is regulated by microbiota, and they reveal an important postnatal function of macrophages that emerge in fetal life.


Subject(s)
Colitis/immunology , Intestinal Mucosa/immunology , Listeriosis/immunology , Macrophages/immunology , Mucosal-Associated Invariant T Cells/immunology , Animals , Cell Proliferation/genetics , Colitis/microbiology , Colitis/pathology , Colon/cytology , Colon/embryology , Colon/immunology , Colon/pathology , Cytokines/metabolism , Diphtheria Toxin/administration & dosage , Diphtheria Toxin/immunology , Disease Models, Animal , Embryo, Mammalian , Female , Gastrointestinal Microbiome/immunology , Gene Expression Regulation, Developmental/immunology , Germ-Free Life , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/embryology , Intestinal Mucosa/pathology , Listeriosis/microbiology , Listeriosis/pathology , Macrophages/metabolism , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , RNA-Seq , Signal Transduction/genetics , Signal Transduction/immunology
3.
Nat Immunol ; 21(3): 343-353, 2020 03.
Article in English | MEDLINE | ID: mdl-32066951

ABSTRACT

Gastrointestinal microbiota and immune cells interact closely and display regional specificity; however, little is known about how these communities differ with location. Here, we simultaneously assess microbiota and single immune cells across the healthy, adult human colon, with paired characterization of immune cells in the mesenteric lymph nodes, to delineate colonic immune niches at steady state. We describe distinct helper T cell activation and migration profiles along the colon and characterize the transcriptional adaptation trajectory of regulatory T cells between lymphoid tissue and colon. Finally, we show increasing B cell accumulation, clonal expansion and mutational frequency from the cecum to the sigmoid colon and link this to the increasing number of reactive bacterial species.


Subject(s)
Colon/immunology , Colon/microbiology , Gastrointestinal Microbiome/immunology , Adult , B-Lymphocytes/immunology , Colon/cytology , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocyte Activation , Organ Specificity , RNA-Seq , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Transcriptome
4.
Cell ; 165(7): 1708-1720, 2016 Jun 16.
Article in English | MEDLINE | ID: mdl-27264604

ABSTRACT

In the mammalian intestine, crypts of Leiberkühn house intestinal epithelial stem/progenitor cells at their base. The mammalian intestine also harbors a diverse array of microbial metabolite compounds that potentially modulate stem/progenitor cell activity. Unbiased screening identified butyrate, a prominent bacterial metabolite, as a potent inhibitor of intestinal stem/progenitor proliferation at physiologic concentrations. During homeostasis, differentiated colonocytes metabolized butyrate likely preventing it from reaching proliferating epithelial stem/progenitor cells within the crypt. Exposure of stem/progenitor cells in vivo to butyrate through either mucosal injury or application to a naturally crypt-less host organism led to inhibition of proliferation and delayed wound repair. The mechanism of butyrate action depended on the transcription factor Foxo3. Our findings indicate that mammalian crypt architecture protects stem/progenitor cell proliferation in part through a metabolic barrier formed by differentiated colonocytes that consume butyrate and stimulate future studies on the interplay of host anatomy and microbiome metabolism.


Subject(s)
Bacteria/metabolism , Butyrates/metabolism , Colon/cytology , Colon/microbiology , Gastrointestinal Microbiome , Stem Cells/metabolism , Acyl-CoA Dehydrogenase/deficiency , Acyl-CoA Dehydrogenase/genetics , Animals , Cell Proliferation , Intestine, Small/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Oxidation-Reduction , Pathogen-Associated Molecular Pattern Molecules/metabolism , Stem Cells/cytology , Zebrafish
5.
Nature ; 629(8012): 669-678, 2024 May.
Article in English | MEDLINE | ID: mdl-38600382

ABSTRACT

Interleukin 22 (IL-22) has a non-redundant role in immune defence of the intestinal barrier1-3. T cells, but not innate lymphoid cells, have an indispensable role in sustaining the IL-22 signalling that is required for the protection of colonic crypts against invasion during infection by the enteropathogen Citrobacter rodentium4 (Cr). However, the intestinal epithelial cell (IEC) subsets targeted by T cell-derived IL-22, and how T cell-derived IL-22 sustains activation in IECs, remain undefined. Here we identify a subset of absorptive IECs in the mid-distal colon that are specifically targeted by Cr and are differentially responsive to IL-22 signalling. Major histocompatibility complex class II (MHCII) expression by these colonocytes was required to elicit sustained IL-22 signalling from Cr-specific T cells, which was required to restrain Cr invasion. Our findings explain the basis for the regionalization of the host response to Cr and demonstrate that epithelial cells must elicit MHCII-dependent help from IL-22-producing T cells to orchestrate immune protection in the intestine.


Subject(s)
Citrobacter rodentium , Colon , Epithelial Cells , Intestinal Mucosa , T-Lymphocytes , Animals , Female , Male , Mice , Citrobacter rodentium/immunology , Colon/cytology , Colon/immunology , Colon/microbiology , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/microbiology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Epithelial Cells/metabolism , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Interleukin-22/immunology , Interleukin-22/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/cytology , Mice, Inbred C3H , Mice, Inbred C57BL , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
6.
Nature ; 623(7989): 1044-1052, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37993709

ABSTRACT

All nucleated cells express major histocompatibility complex I and interferon-γ (IFNγ) receptor1, but an epithelial cell-specific function of IFNγ signalling or antigen presentation by means of major histocompatibility complex I has not been explored. We show here that on sensing IFNγ, colonic epithelial cells productively present pathogen and self-derived antigens to cognate intra-epithelial T cells, which are critically located at the epithelial barrier. Antigen presentation by the epithelial cells confers extracellular ATPase expression in cognate intra-epithelial T cells, which limits the accumulation of extracellular adenosine triphosphate and consequent activation of the NLRP3 inflammasome in tissue macrophages. By contrast, antigen presentation by the tissue macrophages alongside inflammasome-associated interleukin-1α and interleukin-1ß production promotes a pathogenic transformation of CD4+ T cells into granulocyte-macrophage colony-stimulating-factor (GM-CSF)-producing T cells in vivo, which promotes colitis and colorectal cancer. Taken together, our study unravels critical checkpoints requiring IFNγ sensing and antigen presentation by epithelial cells that control the development of pathogenic CD4+ T cell responses in vivo.


Subject(s)
Antigen Presentation , Colon , Epithelial Cells , Interferon-gamma , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Colitis/immunology , Colitis/pathology , Colitis/prevention & control , Colon/cytology , Colon/immunology , Colon/pathology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/prevention & control , Epithelial Cells/immunology , Epithelial Cells/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Inflammasomes/immunology , Inflammasomes/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-1alpha/immunology , Interleukin-1beta/immunology , Macrophages/immunology , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
7.
Nat Immunol ; 17(4): 441-50, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26855029

ABSTRACT

Epithelial tissues continually undergo apoptosis. Commensal organisms that inhabit the epithelium influence tissue homeostasis, in which regulatory T cells (Treg cells) have a central role. However, the physiological importance of epithelial cell apoptosis and how the number of Treg cells is regulated are both incompletely understood. Here we found that apoptotic epithelial cells negatively regulated the commensal-stimulated proliferation of Treg cells. Gut commensals stimulated CX3CR1(+)CD103(-)CD11b(+) dendritic cells (DCs) to produce interferon-ß (IFN-ß), which augmented the proliferation of Treg cells in the intestine. Conversely, phosphatidylserine exposed on apoptotic epithelial cells suppressed IFN-ß production by the DCs via inhibitory signaling mediated by the cell-surface glycoprotein CD300a and thus suppressed Treg cell proliferation. Our findings reveal a regulatory role for apoptotic epithelial cells in maintaining the number of Treg cell and tissue homeostasis.


Subject(s)
Apoptosis/immunology , Epidermis/immunology , Epithelial Cells/immunology , Gastrointestinal Microbiome/immunology , Interferon-beta/immunology , Intestinal Mucosa/immunology , Respiratory Mucosa/immunology , T-Lymphocytes, Regulatory/immunology , Allergens/toxicity , Animals , Colitis/chemically induced , Colitis/immunology , Colitis/pathology , Colon/cytology , Colon/immunology , Dendritic Cells/immunology , Dermatitis, Allergic Contact/etiology , Dermatitis, Allergic Contact/immunology , Dermatitis, Allergic Contact/pathology , Dextran Sulfate/toxicity , Epidermal Cells , Flow Cytometry , Immunohistochemistry , Intestinal Mucosa/cytology , Langerhans Cells/immunology , Lung/cytology , Lung/immunology , Mice , Mice, Knockout , Ovalbumin/toxicity , Real-Time Polymerase Chain Reaction , Receptors, Immunologic/genetics , Respiratory Mucosa/cytology , Salmonella Infections/immunology , Salmonella typhimurium
8.
Cell ; 149(1): 146-58, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22464327

ABSTRACT

Lineage mapping has identified both proliferative and quiescent intestinal stem cells, but the molecular circuitry controlling stem cell quiescence is incompletely understood. By lineage mapping, we show Lrig1, a pan-ErbB inhibitor, marks predominately noncycling, long-lived stem cells that are located at the crypt base and that, upon injury, proliferate and divide to replenish damaged crypts. Transcriptome profiling of Lrig1(+) colonic stem cells differs markedly from the profiling of highly proliferative, Lgr5(+) colonic stem cells; genes upregulated in the Lrig1(+) population include those involved in cell cycle repression and response to oxidative damage. Loss of Apc in Lrig1(+) cells leads to intestinal adenomas, and genetic ablation of Lrig1 results in heightened ErbB1-3 expression and duodenal adenomas. These results shed light on the relationship between proliferative and quiescent intestinal stem cells and support a model in which intestinal stem cell quiescence is maintained by calibrated ErbB signaling with loss of a negative regulator predisposing to neoplasia.


Subject(s)
Colon/metabolism , Genes, Tumor Suppressor , Intestine, Small/metabolism , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Adenoma/pathology , Adenomatous Polyposis Coli Protein/metabolism , Animals , Colon/cytology , ErbB Receptors/metabolism , Gene Expression Profiling , Humans , Intestinal Neoplasms/pathology , Intestine, Small/cytology , Mice , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism
9.
Nature ; 592(7852): 99-104, 2021 04.
Article in English | MEDLINE | ID: mdl-33627870

ABSTRACT

The small intestine is the main organ for nutrient absorption, and its extensive resection leads to malabsorption and wasting conditions referred to as short bowel syndrome (SBS). Organoid technology enables an efficient expansion of intestinal epithelium tissue in vitro1, but reconstruction of the whole small intestine, including the complex lymphovascular system, has remained challenging2. Here we generate a functional small intestinalized colon (SIC) by replacing the native colonic epithelium with ileum-derived organoids. We first find that xenotransplanted human ileum organoids maintain their regional identity and form nascent villus structures in the mouse colon. In vitro culture of an organoid monolayer further reveals an essential role for luminal mechanistic flow in the formation of villi. We then develop a rat SIC model by repositioning the SIC at the ileocaecal junction, where the epithelium is exposed to a constant luminal stream of intestinal juice. This anatomical relocation provides the SIC with organ structures of the small intestine, including intact vasculature and innervation, villous structures, and the lacteal (a fat-absorbing lymphatic structure specific to the small intestine). The SIC has absorptive functions and markedly ameliorates intestinal failure in a rat model of SBS, whereas transplantation of colon organoids instead of ileum organoids invariably leads to mortality. These data provide a proof of principle for the use of intestinal organoids for regenerative purposes, and offer a feasible strategy for SBS treatment.


Subject(s)
Colon/cytology , Ileum/transplantation , Intestinal Mucosa/cytology , Organoids/transplantation , Regeneration , Regenerative Medicine/methods , Short Bowel Syndrome/therapy , Animals , Colon/blood supply , Colon/innervation , Colon/surgery , Disease Models, Animal , Heterografts , Humans , Ileum/cytology , Intestinal Mucosa/blood supply , Intestinal Mucosa/innervation , Intestinal Mucosa/surgery , Male , Organ Culture Techniques , Organoids/cytology , Rats , Rats, Inbred Lew , Short Bowel Syndrome/pathology , Short Bowel Syndrome/surgery
10.
Nature ; 593(7859): 405-410, 2021 05.
Article in English | MEDLINE | ID: mdl-33911282

ABSTRACT

Somatic mutations drive the development of cancer and may contribute to ageing and other diseases1,2. Despite their importance, the difficulty of detecting mutations that are only present in single cells or small clones has limited our knowledge of somatic mutagenesis to a minority of tissues. Here, to overcome these limitations, we developed nanorate sequencing (NanoSeq), a duplex sequencing protocol with error rates of less than five errors per billion base pairs in single DNA molecules from cell populations. This rate is two orders of magnitude lower than typical somatic mutation loads, enabling the study of somatic mutations in any tissue independently of clonality. We used this single-molecule sensitivity to study somatic mutations in non-dividing cells across several tissues, comparing stem cells to differentiated cells and studying mutagenesis in the absence of cell division. Differentiated cells in blood and colon displayed remarkably similar mutation loads and signatures to their corresponding stem cells, despite mature blood cells having undergone considerably more divisions. We then characterized the mutational landscape of post-mitotic neurons and polyclonal smooth muscle, confirming that neurons accumulate somatic mutations at a constant rate throughout life without cell division, with similar rates to mitotically active tissues. Together, our results suggest that mutational processes that are independent of cell division are important contributors to somatic mutagenesis. We anticipate that the ability to reliably detect mutations in single DNA molecules could transform our understanding of somatic mutagenesis and enable non-invasive studies on large-scale cohorts.


Subject(s)
Blood Cells/metabolism , Cell Differentiation/genetics , DNA Mutational Analysis/methods , Muscle, Smooth/metabolism , Mutation , Neurons/metabolism , Single Molecule Imaging/methods , Stem Cells/metabolism , Alzheimer Disease/genetics , Blood Cells/cytology , Cell Division , Cohort Studies , Colon/cytology , Epithelium/metabolism , Granulocytes/cytology , Granulocytes/metabolism , Healthy Volunteers , Humans , Male , Middle Aged , Muscle, Smooth/cytology , Mutagenesis , Mutation Rate , Neurons/cytology , Stem Cells/cytology
11.
Nature ; 589(7841): 270-275, 2021 01.
Article in English | MEDLINE | ID: mdl-33116299

ABSTRACT

There is an urgent need to create novel models using human disease-relevant cells to study severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) biology and to facilitate drug screening. Here, as SARS-CoV-2 primarily infects the respiratory tract, we developed a lung organoid model using human pluripotent stem cells (hPSC-LOs). The hPSC-LOs (particularly alveolar type-II-like cells) are permissive to SARS-CoV-2 infection, and showed robust induction of chemokines following SARS-CoV-2 infection, similar to what is seen in patients with COVID-19. Nearly 25% of these patients also have gastrointestinal manifestations, which are associated with worse COVID-19 outcomes1. We therefore also generated complementary hPSC-derived colonic organoids (hPSC-COs) to explore the response of colonic cells to SARS-CoV-2 infection. We found that multiple colonic cell types, especially enterocytes, express ACE2 and are permissive to SARS-CoV-2 infection. Using hPSC-LOs, we performed a high-throughput screen of drugs approved by the FDA (US Food and Drug Administration) and identified entry inhibitors of SARS-CoV-2, including imatinib, mycophenolic acid and quinacrine dihydrochloride. Treatment at physiologically relevant levels of these drugs significantly inhibited SARS-CoV-2 infection of both hPSC-LOs and hPSC-COs. Together, these data demonstrate that hPSC-LOs and hPSC-COs infected by SARS-CoV-2 can serve as disease models to study SARS-CoV-2 infection and provide a valuable resource for drug screening to identify candidate COVID-19 therapeutics.


Subject(s)
Antiviral Agents/pharmacology , COVID-19/virology , Colon/cytology , Drug Evaluation, Preclinical/methods , Lung/cytology , Organoids/drug effects , Organoids/virology , SARS-CoV-2/drug effects , Animals , COVID-19/prevention & control , Colon/drug effects , Colon/virology , Drug Approval , Female , Heterografts/drug effects , Humans , In Vitro Techniques , Lung/drug effects , Lung/virology , Male , Mice , Organoids/cytology , Organoids/metabolism , SARS-CoV-2/genetics , United States , United States Food and Drug Administration , Viral Tropism , Virus Internalization/drug effects , COVID-19 Drug Treatment
12.
Development ; 149(1)2022 01 01.
Article in English | MEDLINE | ID: mdl-34910127

ABSTRACT

Although Wnt signaling is clearly important for the intestinal epithelial homeostasis, the relevance of various sources of Wnt ligands themselves remains incompletely understood. Blocking the release of Wnt in distinct stromal cell types suggests obligatory functions of several stromal cell sources and yields different observations. The physiological contribution of epithelial Wnt to tissue homeostasis remains unclear. We show here that blocking epithelial Wnts affects colonic Reg4+ epithelial cell differentiation and impairs colonic epithelial regeneration after injury in mice. Single-cell RNA analysis of intestinal stroma showed that the majority of Wnt-producing cells were contained in transgelin (Tagln+) and smooth muscle actin α2 (Acta2+) expressing populations. We genetically attenuated Wnt production from these stromal cells using Tagln-Cre and Acta2-CreER drivers, and found that blockage of Wnt release from either epithelium or Tagln+ and Acta2+ stromal cells impaired colonic epithelial healing after chemical-induced injury. Aggregated blockage of Wnt release from both epithelium and Tagln+ or Acta2+ stromal cells drastically diminished epithelial repair, increasing morbidity and mortality. These results from two uncharacterized stromal populations suggested that colonic recovery from colitis-like injury depends on multiple Wnt-producing sources.


Subject(s)
Actins/metabolism , Colitis, Ulcerative/metabolism , Intestinal Mucosa/metabolism , Microfilament Proteins/metabolism , Muscle Proteins/metabolism , Wnt3A Protein/metabolism , Wound Healing , Actins/genetics , Animals , Cells, Cultured , Colon/cytology , Colon/metabolism , Colon/physiology , Intestinal Mucosa/cytology , Mice , Mice, Inbred C57BL , Microfilament Proteins/genetics , Muscle Proteins/genetics , Pancreatitis-Associated Proteins/genetics , Pancreatitis-Associated Proteins/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Stem Cells/metabolism , Wnt3A Protein/genetics
13.
PLoS Biol ; 20(1): e3001527, 2022 01.
Article in English | MEDLINE | ID: mdl-35089911

ABSTRACT

CRISPR-associated nucleases are powerful tools for precise genome editing of model systems, including human organoids. Current methods describing fluorescent gene tagging in organoids rely on the generation of DNA double-strand breaks (DSBs) to stimulate homology-directed repair (HDR) or non-homologous end joining (NHEJ)-mediated integration of the desired knock-in. A major downside associated with DSB-mediated genome editing is the required clonal selection and expansion of candidate organoids to verify the genomic integrity of the targeted locus and to confirm the absence of off-target indels. By contrast, concurrent nicking of the genomic locus and targeting vector, known as in-trans paired nicking (ITPN), stimulates efficient HDR-mediated genome editing to generate large knock-ins without introducing DSBs. Here, we show that ITPN allows for fast, highly efficient, and indel-free fluorescent gene tagging in human normal and cancer organoids. Highlighting the ease and efficiency of ITPN, we generate triple fluorescent knock-in organoids where 3 genomic loci were simultaneously modified in a single round of targeting. In addition, we generated model systems with allele-specific readouts by differentially modifying maternal and paternal alleles in one step. ITPN using our palette of targeting vectors, publicly available from Addgene, is ideally suited for generating error-free heterozygous knock-ins in human organoids.


Subject(s)
DNA/genetics , Deoxyribonuclease I/metabolism , Genetic Loci , Organoids/metabolism , Recombinational DNA Repair , Staining and Labeling/methods , Alleles , Base Sequence , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , Colon/cytology , Colon/metabolism , DNA/metabolism , DNA End-Joining Repair , Deoxyribonuclease I/genetics , Electroporation/methods , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Gene Knock-In Techniques , Genetic Vectors , Genome, Human , Heterozygote , Humans , Organoids/cytology
14.
Nature ; 567(7746): 49-55, 2019 03.
Article in English | MEDLINE | ID: mdl-30814735

ABSTRACT

The colonic epithelium facilitates host-microorganism interactions to control mucosal immunity, coordinate nutrient recycling and form a mucus barrier. Breakdown of the epithelial barrier underpins inflammatory bowel disease (IBD). However, the specific contributions of each epithelial-cell subtype to this process are unknown. Here we profile single colonic epithelial cells from patients with IBD and unaffected controls. We identify previously unknown cellular subtypes, including gradients of progenitor cells, colonocytes and goblet cells within intestinal crypts. At the top of the crypts, we find a previously unknown absorptive cell, expressing the proton channel OTOP2 and the satiety peptide uroguanylin, that senses pH and is dysregulated in inflammation and cancer. In IBD, we observe a positional remodelling of goblet cells that coincides with downregulation of WFDC2-an antiprotease molecule that we find to be expressed by goblet cells and that inhibits bacterial growth. In vivo, WFDC2 preserves the integrity of tight junctions between epithelial cells and prevents invasion by commensal bacteria and mucosal inflammation. We delineate markers and transcriptional states, identify a colonic epithelial cell and uncover fundamental determinants of barrier breakdown in IBD.


Subject(s)
Colon/cytology , Colon/pathology , Epithelial Cells/classification , Epithelial Cells/cytology , Health , Inflammatory Bowel Diseases/pathology , Ion Channels/metabolism , Animals , Biomarkers/analysis , Colitis, Ulcerative/genetics , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/pathology , Colon/microbiology , Epithelial Cells/microbiology , Epithelial Cells/pathology , Genetic Predisposition to Disease/genetics , Goblet Cells/cytology , Goblet Cells/metabolism , Goblet Cells/pathology , Humans , Hydrogen-Ion Concentration , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/microbiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Mice , Natriuretic Peptides/metabolism , Proteins/metabolism , Single-Cell Analysis , Stem Cells/cytology , Stem Cells/metabolism , Stem Cells/pathology , Tight Junctions/metabolism , Transcription, Genetic , WAP Four-Disulfide Core Domain Protein 2
15.
Nature ; 574(7779): 532-537, 2019 10.
Article in English | MEDLINE | ID: mdl-31645730

ABSTRACT

The colorectal adenoma-carcinoma sequence has provided a paradigmatic framework for understanding the successive somatic genetic changes and consequent clonal expansions that lead to cancer1. However, our understanding of the earliest phases of colorectal neoplastic changes-which may occur in morphologically normal tissue-is comparatively limited, as for most cancer types. Here we use whole-genome sequencing to analyse hundreds of normal crypts from 42 individuals. Signatures of multiple mutational processes were revealed; some of these were ubiquitous and continuous, whereas others were only found in some individuals, in some crypts or during certain periods of life. Probable driver mutations were present in around 1% of normal colorectal crypts in middle-aged individuals, indicating that adenomas and carcinomas are rare outcomes of a pervasive process of neoplastic change across morphologically normal colorectal epithelium. Colorectal cancers exhibit substantially increased mutational burdens relative to normal cells. Sequencing normal colorectal cells provides quantitative insights into the genomic and clonal evolution of cancer.


Subject(s)
Colon/cytology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Mutation , Prodromal Symptoms , Rectum/cytology , Adenoma/genetics , Adenoma/pathology , Aged , Axin Protein/genetics , Carcinoma/genetics , Carcinoma/pathology , Cell Transformation, Neoplastic , Clone Cells/cytology , Clone Cells/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Copy Number Variations , DNA Mutational Analysis , Female , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Middle Aged , Stem Cells/cytology , Stem Cells/metabolism
16.
Nature ; 566(7743): 249-253, 2019 02.
Article in English | MEDLINE | ID: mdl-30700914

ABSTRACT

Environmental genotoxic factors pose a challenge to the genomic integrity of epithelial cells at barrier surfaces that separate host organisms from the environment. They can induce mutations that, if they occur in epithelial stem cells, contribute to malignant transformation and cancer development1-3. Genome integrity in epithelial stem cells is maintained by an evolutionarily conserved cellular response pathway, the DNA damage response (DDR). The DDR culminates in either transient cell-cycle arrest and DNA repair or elimination of damaged cells by apoptosis4,5. Here we show that the cytokine interleukin-22 (IL-22), produced by group 3 innate lymphoid cells (ILC3) and γδ T cells, is an important regulator of the DDR machinery in intestinal epithelial stem cells. Using a new mouse model that enables sporadic inactivation of the IL-22 receptor in colon epithelial stem cells, we demonstrate that IL-22 is required for effective initiation of the DDR following DNA damage. Stem cells deprived of IL-22 signals and exposed to carcinogens escaped DDR-controlled apoptosis, contained more mutations and were more likely to give rise to colon cancer. We identified metabolites of glucosinolates, a group of phytochemicals contained in cruciferous vegetables, to be a widespread source of genotoxic stress in intestinal epithelial cells. These metabolites are ligands of the aryl hydrocarbon receptor (AhR)6, and AhR-mediated signalling in ILC3 and γδ T cells controlled their production of IL-22. Mice fed with diets depleted of glucosinolates produced only very low levels of IL-22 and, consequently, the DDR in epithelial cells of mice on a glucosinolate-free diet was impaired. This work identifies a homeostatic network protecting stem cells against challenge to their genome integrity by AhR-mediated 'sensing' of genotoxic compounds from the diet. AhR signalling, in turn, ensures on-demand production of IL-22 by innate lymphocytes directly regulating components of the DDR in epithelial stem cells.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Colon/cytology , Interleukins/pharmacology , Mutagens/pharmacology , Stem Cells/drug effects , Stem Cells/metabolism , Animals , Apoptosis/drug effects , Cell Transformation, Neoplastic/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/prevention & control , DNA Damage , Diet/adverse effects , Glucosinolates/administration & dosage , Glucosinolates/pharmacology , Immunity, Innate , Interleukins/biosynthesis , Intestinal Mucosa/cytology , Ligands , Mice , Mutagens/administration & dosage , Mutation/genetics , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Interleukin/metabolism , Stem Cells/cytology , T-Lymphocytes/metabolism , Interleukin-22
17.
J Biol Chem ; 299(8): 105068, 2023 08.
Article in English | MEDLINE | ID: mdl-37468102

ABSTRACT

Although it was described previously for estrogen (E2) regulation of intestinal epithelial Cl- and HCO3- secretion in sex difference, almost nothing is known about the roles of estrogen receptor (ER) subtypes in regulating E2-modulated epithelial ion transports and epithelial restitution. Here, we aimed to investigate ERα and ERß subtypes in the regulation of E2-modulated colonic epithelial HCO3- and Cl- secretion and epithelial restitution. Through physiological and biochemical studies, in combination of genetic knockdown, we showed that ERα attenuated female colonic Cl- secretion but promoted Ca2+-dependent HCO3- secretion via store-operated calcium entry (SOCE) mechanism in mice. However, ERß attenuated HCO3- secretion by inhibiting Ca2+via the SOCE and inhibiting cAMP via protein kinases. Moreover, ERα but not ERß promoted epithelial cell restitution via SOCE/Ca2+ signaling. ERα also enhanced cyclin D1, proliferating cell nuclear antigen, and ß-catenin expression in normal human colonic epithelial cells. All ERα-mediated biological effects could be attenuated by its selective antagonist and genetic knockdown. Finally, both ERα and ERß were expressed in human colonic epithelial cells and mouse colonic tissues. We therefore conclude that E2 modulates complex colonic epithelial HCO3- and Cl- secretion via ER subtype-dependent mechanisms and that ERα is specifically responsible for colonic epithelial regeneration. This study provides novel insights into the molecular mechanisms of how ERα and ERß subtypes orchestrate functional homeostasis of normal colonic epithelial cells.


Subject(s)
Colon , Epithelial Cells , Estrogen Receptor alpha , Ion Transport , Receptors, Estrogen , Animals , Female , Humans , Mice , Epithelial Cells/metabolism , Estradiol/metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Estrogens/metabolism , Receptors, Estrogen/metabolism , Colon/cytology
18.
Nat Methods ; 18(7): 829-834, 2021 07.
Article in English | MEDLINE | ID: mdl-34183831

ABSTRACT

We introduce a cost-effective and easily implementable scan unit that converts any camera-based microscope with optical sectioning capability into a multi-angle projection imaging system. Projection imaging reduces data overhead and accelerates imaging by a factor of >100, while also allowing users to readily view biological phenomena of interest from multiple perspectives on the fly. By rapidly interrogating the sample from just two perspectives, our method also enables real-time stereoscopic imaging and three-dimensional particle localization. We demonstrate projection imaging with spinning disk confocal, lattice light-sheet, multidirectional illumination light-sheet and oblique plane microscopes on specimens that range from organelles in single cells to the vasculature of a zebrafish embryo. Furthermore, we leverage our projection method to rapidly image cancer cell morphodynamics and calcium signaling in cultured neurons at rates up to 119 Hz as well as to simultaneously image orthogonal views of a beating embryonic zebrafish heart.


Subject(s)
Image Processing, Computer-Assisted/methods , Microscopy, Confocal/instrumentation , Microscopy, Confocal/methods , Animals , Colon/cytology , Embryo, Nonmammalian/cytology , Female , Heart/diagnostic imaging , Heart/embryology , Humans , Imaging, Three-Dimensional , Male , Mice , Mice, Transgenic , Neurons/cytology , Rats, Sprague-Dawley , Spheroids, Cellular/pathology , Zebrafish/embryology
19.
EMBO Rep ; 23(3): e53246, 2022 02 03.
Article in English | MEDLINE | ID: mdl-34939731

ABSTRACT

Regulatory T lymphocyte (Treg) homing reactions mediated by G protein-coupled receptor (GPCR)-ligand interactions play a central role in maintaining intestinal immune homeostasis by restraining inappropriate immune responses in the gastrointestinal tract. However, the origin of Treg homing to the colon remains mysterious. Here, we report that the C10ORF99 peptide (also known as CPR15L and AP57), a cognate ligand of GPR15 that controls Treg homing to the colon, originates from a duplication of the flanking CDHR1 gene and is functionally paired with GPR15 in amniotes. Evolutionary analysis and experimental data indicate that the GPR15-C10ORF99 pair is functionally conserved to mediate colonic Treg homing in amniotes and their expression patterns are positively correlated with herbivore diet in the colon. With the first herbivorous diet in early amniotes, a new biological process (herbivorous diet short-chain fatty acid-C10ORF99/GPR15-induced Treg homing colon immune homeostasis) emerged, and we propose an evolutionary model whereby GPR15-C10ORF99 functional pairing has initiated the first colonic Treg homing reaction in amniotes. Our findings also highlight that GPCR-ligand pairing leads to physiological adaptation during vertebrate evolution.


Subject(s)
Antimicrobial Cationic Peptides , Colon/cytology , DNA-Binding Proteins , Receptors, G-Protein-Coupled , T-Lymphocytes, Regulatory , Animals , Colon/immunology , Ligands , Protein Binding , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , T-Lymphocytes, Regulatory/cytology
20.
Cell ; 139(4): 802-13, 2009 Nov 13.
Article in English | MEDLINE | ID: mdl-19914172

ABSTRACT

The kinesin superfamily proteins (KIFs) are motor proteins that transport organelles and protein complexes in a microtubule- and ATP-dependent manner. We identified KIF26A as a new member of the murine KIFs. KIF26A is a rather atypical member as it lacks ATPase activity. Mice with a homozygous deletion of Kif26a developed a megacolon with enteric nerve hyperplasia. Kif26a-/- enteric neurons showed hypersensitivity for GDNF-Ret signaling, and we find that KIF26A suppressed GDNF-Ret signaling by direct binding and inhibition of Grb2, an essential component of GDNF/Akt/ERK signaling. We therefore propose that the unconventional kinesin KIF26A plays a key role in enteric nervous system development by repressing a cell growth signaling pathway.


Subject(s)
Enteric Nervous System/embryology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Hirschsprung Disease/metabolism , Kinesins/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Signal Transduction , Animals , Cell Growth Processes , Cell Line , Colon/cytology , Colon/embryology , Colon/innervation , GRB2 Adaptor Protein/metabolism , Kinesins/genetics , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neurons/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL