Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Antiviral Res ; 224: 105837, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38387750

ABSTRACT

The COVID-19 pandemic has shown the need to develop effective therapeutics in preparedness for further epidemics of virus infections that pose a significant threat to human health. As a natural compound antiviral candidate, we focused on α-dystroglycan, a highly glycosylated basement membrane protein that links the extracellular matrix to the intracellular cytoskeleton. Here we show that the N-terminal fragment of α-dystroglycan (α-DGN), as produced in E. coli in the absence of post-translational modifications, blocks infection of SARS-CoV-2 in cell culture, human primary gut organoids and the lungs of transgenic mice expressing the human receptor angiotensin I-converting enzyme 2 (hACE2). Prophylactic and therapeutic administration of α-DGN reduced SARS-CoV-2 lung titres and protected the mice from respiratory symptoms and death. Recombinant α-DGN also blocked infection of a wide range of enveloped viruses including the four Dengue virus serotypes, influenza A virus, respiratory syncytial virus, tick-borne encephalitis virus, but not human adenovirus, a non-enveloped virus in vitro. This study establishes soluble recombinant α-DGN as a broad-band, natural compound candidate therapeutic against enveloped viruses.


Subject(s)
COVID-19 , SARS-CoV-2 , Mice , Animals , Humans , Dystroglycans , Pandemics , Escherichia coli , Mice, Transgenic , Antiviral Agents/pharmacology
2.
Cell Rep ; 42(8): 112970, 2023 Aug 29.
Article in English | MEDLINE | ID: mdl-37556323

ABSTRACT

Pancreatic islets regulate blood glucose homeostasis through the controlled release of insulin; however, current metabolic models of glucose-sensitive insulin secretion are incomplete. A comprehensive understanding of islet metabolism is integral to studies of endocrine cell development as well as diabetic islet dysfunction. Human pluripotent stem cell-derived islets (SC-islets) are a developmentally relevant model of human islet function that have great potential in providing a cure for type 1 diabetes. Using multiple 13C-labeled metabolic fuels, we demonstrate that SC-islets show numerous divergent patterns of metabolite trafficking in proposed insulin release pathways compared with primary human islets but are still reliant on mitochondrial aerobic metabolism to derive function. Furthermore, reductive tricarboxylic acid cycle activity and glycolytic metabolite cycling occur in SC-islets, suggesting that non-canonical coupling factors are also present. In aggregate, we show that many facets of SC-islet metabolism overlap with those of primary islets, albeit with a retained immature signature.

3.
J Cell Biol ; 222(8)2023 08 07.
Article in English | MEDLINE | ID: mdl-37233325

ABSTRACT

Reticular adhesions (RAs) consist of integrin αvß5 and harbor flat clathrin lattices (FCLs), long-lasting structures with similar molecular composition as clathrin-mediated endocytosis (CME) carriers. Why FCLs and RAs colocalize is not known. Here, we show that RAs are assembled at FCLs in a process controlled by fibronectin (FN) and its receptor, integrin α5ß1. We observed that cells on FN-rich matrices displayed fewer FCLs and RAs. CME machinery inhibition abolished RAs and live-cell imaging showed that RA establishment requires FCL coassembly. The inhibitory activity of FN was mediated by the activation of integrin α5ß1 at Tensin1-positive fibrillar adhesions. Conventionally, endocytosis disassembles cellular adhesions by internalizing their components. Our results present a novel paradigm in the relationship between these two processes by showing that endocytic proteins can actively function in the assembly of cell adhesions. Furthermore, we show this novel adhesion assembly mechanism is coupled to cell migration via unique crosstalk between cell-matrix adhesions.


Subject(s)
Clathrin , Integrin alpha5beta1 , Integrin alpha5beta1/genetics , Integrin alpha5beta1/metabolism , Clathrin/genetics , Clathrin/metabolism , Cell Adhesion/physiology , Cell Movement , Endocytosis , Fibronectins/genetics , Fibronectins/metabolism , Focal Adhesions/metabolism
4.
Dis Model Mech ; 16(4)2023 04 01.
Article in English | MEDLINE | ID: mdl-36912192

ABSTRACT

Growth factors secreted by stromal fibroblasts regulate the intestinal epithelium. Stroma-derived epidermal growth factor (EGF) family ligands are implicated in epithelial regeneration and tumorigenesis, but their specific contributions and associated mechanisms remain unclear. Here, we use primary intestinal organoids modeling homeostatic, injured and tumorigenic epithelia to assess how the fibroblast-derived EGF family ligands neuregulin 1 (NRG1) and epiregulin (EREG) regulate the intestinal epithelium. NRG1 was expressed exclusively in the stroma, robustly increased crypt budding and protected intestinal epithelial organoids from radiation-induced damage. NRG1 also induced regenerative features in the epithelium, including a fetal-like transcriptome, suppression of the Lgr5+ stem cell pool and remodeling of the epithelial actin cytoskeleton. Intriguingly, unlike EGF and EREG, NRG1 failed to support the growth of pre-tumorigenic intestinal organoids lacking the tumor suppressor Apc, commonly mutated in human colorectal cancer (CRC). Interestingly, high expression of stromal NRG1 was associated with improved survival in CRC cohorts, suggesting a tumor-suppressive function. Our results highlight the power of stromal NRG1 in transcriptional reprogramming and protection of the intestinal epithelium from radiation injury without promoting tumorigenesis.


Subject(s)
Epidermal Growth Factor , Intestinal Mucosa , Neuregulin-1 , Humans , Carcinogenesis/metabolism , Epidermal Growth Factor/metabolism , Fibroblasts/metabolism , Intestinal Mucosa/metabolism , Ligands , Neuregulin-1/metabolism , Cellular Reprogramming
5.
bioRxiv ; 2023 Mar 24.
Article in English | MEDLINE | ID: mdl-36993731

ABSTRACT

Cell-to-cell signalling between niche and stem cells regulates tissue regeneration. While the identity of many mediating factors is known, it is largely unknown whether stem cells optimize their receptiveness to niche signals according to the niche organization. Here, we show that Lgr5+ small intestinal stem cells (ISCs) regulate the morphology and orientation of their secretory apparatus to match the niche architecture, and to increase transport efficiency of niche signal receptors. Unlike the progenitor cells lacking lateral niche contacts, ISCs orient Golgi apparatus laterally towards Paneth cells of the epithelial niche, and divide Golgi into multiple stacks reflecting the number of Paneth cell contacts. Stem cells with a higher number of lateral Golgi transported Epidermal growth factor receptor (Egfr) with a higher efficiency than cells with one Golgi. The lateral Golgi orientation and enhanced Egfr transport required A-kinase anchor protein 9 (Akap9), and was necessary for normal regenerative capacity in vitro . Moreover, reduced Akap9 in aged ISCs renders ISCs insensitive to niche-dependent modulation of Golgi stack number and transport efficiency. Our results reveal stem cell-specific Golgi complex configuration that facilitates efficient niche signal reception and tissue regeneration, which is compromised in the aged epithelium.

6.
Sci Adv ; 9(2): eadd5163, 2023 01 13.
Article in English | MEDLINE | ID: mdl-36638183

ABSTRACT

Nicotinamide adenine dinucleotide (NAD+) precursor nicotinamide riboside (NR) has emerged as a promising compound to improve obesity-associated mitochondrial dysfunction and metabolic syndrome in mice. However, most short-term clinical trials conducted so far have not reported positive outcomes. Therefore, we aimed to determine whether long-term NR supplementation boosts mitochondrial biogenesis and metabolic health in humans. Twenty body mass index (BMI)-discordant monozygotic twin pairs were supplemented with an escalating dose of NR (250 to 1000 mg/day) for 5 months. NR improved systemic NAD+ metabolism, muscle mitochondrial number, myoblast differentiation, and gut microbiota composition in both cotwins. NR also showed a capacity to modulate epigenetic control of gene expression in muscle and adipose tissue in both cotwins. However, NR did not ameliorate adiposity or metabolic health. Overall, our results suggest that NR acts as a potent modifier of NAD+ metabolism, muscle mitochondrial biogenesis and stem cell function, gut microbiota, and DNA methylation in humans irrespective of BMI.


Subject(s)
Gastrointestinal Microbiome , NAD , Humans , Mice , Animals , NAD/metabolism , Organelle Biogenesis , Obesity/metabolism , Muscle, Skeletal/metabolism , Cell Differentiation
7.
J Cell Sci ; 135(23)2022 12 01.
Article in English | MEDLINE | ID: mdl-36468336

ABSTRACT

Mammary epithelium is a bilayered ductal network composed of luminal and basal epithelial cells, which together drive the growth and functional differentiation of the gland. Basal mammary epithelial cells (MECs) exhibit remarkable plasticity and progenitor activity that facilitate epithelial expansion. However, their activity must be tightly regulated to restrict excess basal cell activity. Here, we show that adhesion of basal cells to laminin α5-containing basement membrane matrix, which is produced by luminal cells, presents such a control mechanism. Adhesion to laminin α5 directs basal cells towards a luminal cell fate, and thereby results in a marked decrease of basal MEC progenitor activity in vitro and in vivo. Mechanistically, these effects are mediated through ß4-integrin and activation of p21 (encoded by CDKN1A). Thus, we demonstrate that laminin matrix adhesion is a key determinant of basal identity and essential to building and maintaining a functional multicellular epithelium.


Subject(s)
Epithelial Cells , Laminin , Epithelium , Basement Membrane , Integrin beta4
8.
Sci Adv ; 8(41): eabm1847, 2022 Oct 14.
Article in English | MEDLINE | ID: mdl-36240269

ABSTRACT

Niche-derived factors regulate tissue stem cells, but apart from the mechanosensory pathways, the effect of niche geometry is not well understood. We used organoids and bioengineered tissue culture platforms to demonstrate that the conical shape of Lgr5+ small intestinal stem cells (ISCs) facilitate their self-renewal and function. Inhibition of non-muscle myosin II (NM II)-driven apical constriction altered ISC shape and reduced niche curvature and stem cell capacity. Niche curvature is decreased in aged mice, suggesting that suboptimal interactions between old ISCs and their niche develop with age. We show that activation of NM IIC or physical restriction to young topology improves in vitro regeneration by old epithelium. We propose that the increase in lateral surface area of ISCs induced by apical constriction promotes interactions between neighboring cells, and the curved topology of the intestinal niche has evolved to maximize signaling between ISCs and neighboring cells.

9.
Nature ; 607(7919): 548-554, 2022 07.
Article in English | MEDLINE | ID: mdl-35831497

ABSTRACT

The morphology and functionality of the epithelial lining differ along the intestinal tract, but tissue renewal at all sites is driven by stem cells at the base of crypts1-3. Whether stem cell numbers and behaviour vary at different sites is unknown. Here we show using intravital microscopy that, despite similarities in the number and distribution of proliferative cells with an Lgr5 signature in mice, small intestinal crypts contain twice as many effective stem cells as large intestinal crypts. We find that, although passively displaced by a conveyor-belt-like upward movement, small intestinal cells positioned away from the crypt base can function as long-term effective stem cells owing to Wnt-dependent retrograde cellular movement. By contrast, the near absence of retrograde movement in the large intestine restricts cell repositioning, leading to a reduction in effective stem cell number. Moreover, after suppression of the retrograde movement in the small intestine, the number of effective stem cells is reduced, and the rate of monoclonal conversion of crypts is accelerated. Together, these results show that the number of effective stem cells is determined by active retrograde movement, revealing a new channel of stem cell regulation that can be experimentally and pharmacologically manipulated.


Subject(s)
Cell Count , Cell Movement , Intestines , Stem Cells , Animals , Intestinal Mucosa/cytology , Intestine, Small/cytology , Intestines/cytology , Mice , Receptors, G-Protein-Coupled , Stem Cells/cytology , Wnt Proteins
10.
Nat Biotechnol ; 40(7): 1042-1055, 2022 07.
Article in English | MEDLINE | ID: mdl-35241836

ABSTRACT

Transplantation of pancreatic islet cells derived from human pluripotent stem cells is a promising treatment for diabetes. Despite progress in the generation of stem-cell-derived islets (SC-islets), no detailed characterization of their functional properties has been conducted. Here, we generated functionally mature SC-islets using an optimized protocol and benchmarked them comprehensively against primary adult islets. Biphasic glucose-stimulated insulin secretion developed during in vitro maturation, associated with cytoarchitectural reorganization and the increasing presence of alpha cells. Electrophysiology, signaling and exocytosis of SC-islets were similar to those of adult islets. Glucose-responsive insulin secretion was achieved despite differences in glycolytic and mitochondrial glucose metabolism. Single-cell transcriptomics of SC-islets in vitro and throughout 6 months of engraftment in mice revealed a continuous maturation trajectory culminating in a transcriptional landscape closely resembling that of primary islets. Our thorough evaluation of SC-islet maturation highlights their advanced degree of functionality and supports their use in further efforts to understand and combat diabetes.


Subject(s)
Islets of Langerhans Transplantation , Islets of Langerhans , Pluripotent Stem Cells , Animals , Glucose/metabolism , Humans , Insulin/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans Transplantation/methods , Mice , Pluripotent Stem Cells/metabolism
11.
Nat Cell Biol ; 24(2): 148-154, 2022 02.
Article in English | MEDLINE | ID: mdl-35165416

ABSTRACT

Metabolic characteristics of adult stem cells are distinct from their differentiated progeny, and cellular metabolism is emerging as a potential driver of cell fate conversions1-4. How these metabolic features are established remains unclear. Here we identified inherited metabolism imposed by functionally distinct mitochondrial age-classes as a fate determinant in asymmetric division of epithelial stem-like cells. While chronologically old mitochondria support oxidative respiration, the electron transport chain of new organelles is proteomically immature and they respire less. After cell division, selectively segregated mitochondrial age-classes elicit a metabolic bias in progeny cells, with oxidative energy metabolism promoting differentiation in cells that inherit old mitochondria. Cells that inherit newly synthesized mitochondria with low levels of Rieske iron-sulfur polypeptide 1 have a higher pentose phosphate pathway activity, which promotes de novo purine biosynthesis and redox balance, and is required to maintain stemness during early fate determination after division. Our results demonstrate that fate decisions are susceptible to intrinsic metabolic bias imposed by selectively inherited mitochondria.


Subject(s)
Adult Stem Cells/metabolism , Cell Differentiation , Cell Lineage , DNA, Mitochondrial/genetics , Energy Metabolism , Genes, Mitochondrial , Mammary Glands, Human/metabolism , Mitochondria/metabolism , Animals , Cell Line , Cell Proliferation , Cellular Senescence , Female , Humans , Mammary Glands, Human/cytology , Metabolome , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/genetics , Phenotype , Proteome
12.
Nat Commun ; 12(1): 6741, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34795242

ABSTRACT

Smooth muscle is an essential component of the intestine, both to maintain its structure and produce peristaltic and segmentation movements. However, very little is known about other putative roles that smooth muscle cells may have. Here, we show that smooth muscle cells may be the dominant suppliers of BMP antagonists, which are niche factors essential for intestinal stem cell maintenance. Furthermore, muscle-derived factors render epithelium reparative and fetal-like, which includes heightened YAP activity. Mechanistically, we find that the membrane-bound matrix metalloproteinase MMP17, which is exclusively expressed by smooth muscle cells, is required for intestinal epithelial repair after inflammation- or irradiation-induced injury. Furthermore, we propose that MMP17 affects intestinal epithelial reprogramming after damage indirectly by cleaving diffusible factor(s) such as the matricellular protein PERIOSTIN. Together, we identify an important signaling axis that establishes a role for smooth muscle cells as modulators of intestinal epithelial regeneration and the intestinal stem cell niche.


Subject(s)
Matrix Metalloproteinase 17/metabolism , Muscle, Smooth/metabolism , Regeneration/physiology , Stem Cell Niche/physiology , Animals , Humans , Intestinal Mucosa/metabolism , Intestines/cytology , Intestines/pathology , Signal Transduction/physiology , Stem Cells/metabolism
13.
Hum Mol Genet ; 30(24): 2429-2440, 2021 11 30.
Article in English | MEDLINE | ID: mdl-34274970

ABSTRACT

Many hereditary cancer syndromes are associated with an increased risk of small and large intestinal adenocarcinomas. However, conditions bearing a high risk to both adenocarcinomas and neuroendocrine tumors are yet to be described. We studied a family with 16 individuals in four generations affected by a wide spectrum of intestinal tumors, including hyperplastic polyps, adenomas, small intestinal neuroendocrine tumors, and colorectal and small intestinal adenocarcinomas. To assess the genetic susceptibility and understand the novel phenotype, we utilized multiple molecular methods, including whole genome sequencing, RNA sequencing, single cell sequencing, RNA in situ hybridization and organoid culture. We detected a heterozygous deletion at the cystic fibrosis locus (7q31.2) perfectly segregating with the intestinal tumor predisposition in the family. The deletion removes a topologically associating domain border between CFTR and WNT2, aberrantly activating WNT2 in the intestinal epithelium. These consequences suggest that the deletion predisposes to small intestinal neuroendocrine tumors and small and large intestinal adenocarcinomas, and reveals the broad tumorigenic effects of aberrant WNT activation in the human intestine.


Subject(s)
Adenocarcinoma , Adenoma , Colorectal Neoplasms , Neuroendocrine Tumors , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenoma/genetics , Adenoma/pathology , Colorectal Neoplasms/genetics , Humans , Intestinal Mucosa/pathology , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/pathology , Wnt2 Protein
14.
Development ; 148(12)2021 06 15.
Article in English | MEDLINE | ID: mdl-34128985

ABSTRACT

Epithelial attachment to the basement membrane (BM) is essential for mammary gland development, yet the exact roles of specific BM components remain unclear. Here, we show that Laminin α5 (Lama5) expression specifically in the luminal epithelial cells is necessary for normal mammary gland growth during puberty, and for alveologenesis during pregnancy. Lama5 loss in the keratin 8-expressing cells results in reduced frequency and differentiation of hormone receptor expressing (HR+) luminal cells. Consequently, Wnt4-mediated crosstalk between HR+ luminal cells and basal epithelial cells is compromised during gland remodeling, and results in defective epithelial growth. The effects of Lama5 deletion on gland growth and branching can be rescued by Wnt4 supplementation in the in vitro model of branching morphogenesis. Our results reveal a surprising role for BM-protein expression in the luminal mammary epithelial cells, and highlight the function of Lama5 in mammary gland remodeling and luminal differentiation.


Subject(s)
Cell Differentiation/genetics , Epithelium/metabolism , Laminin/genetics , Mammary Glands, Animal/metabolism , Signal Transduction , Wnt4 Protein/genetics , Animals , Biomarkers , Epithelial Cells , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental , Immunohistochemistry , Laminin/metabolism , Mammary Glands, Animal/embryology , Mice , Models, Biological , Morphogenesis/genetics , Organogenesis/genetics , Wnt4 Protein/metabolism
15.
Nature ; 594(7863): 430-435, 2021 06.
Article in English | MEDLINE | ID: mdl-34079124

ABSTRACT

The tumour suppressor APC is the most commonly mutated gene in colorectal cancer. Loss of Apc in intestinal stem cells drives the formation of adenomas in mice via increased WNT signalling1, but reduced secretion of WNT ligands increases the ability of Apc-mutant intestinal stem cells to colonize a crypt (known as fixation)2. Here we investigated how Apc-mutant cells gain a clonal advantage over wild-type counterparts to achieve fixation. We found that Apc-mutant cells are enriched for transcripts that encode several secreted WNT antagonists, with Notum being the most highly expressed. Conditioned medium from Apc-mutant cells suppressed the growth of wild-type organoids in a NOTUM-dependent manner. Furthermore, NOTUM-secreting Apc-mutant clones actively inhibited the proliferation of surrounding wild-type crypt cells and drove their differentiation, thereby outcompeting crypt cells from the niche. Genetic or pharmacological inhibition of NOTUM abrogated the ability of Apc-mutant cells to expand and form intestinal adenomas. We identify NOTUM as a key mediator during the early stages of mutation fixation that can be targeted to restore wild-type cell competitiveness and provide preventative strategies for people at a high risk of developing colorectal cancer.


Subject(s)
Cell Competition , Cell Transformation, Neoplastic , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Esterases/metabolism , Genes, APC , Mutation , Adenoma/genetics , Adenoma/pathology , Adenomatous Polyposis Coli Protein/genetics , Animals , Cell Competition/genetics , Cell Differentiation , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Culture Media, Conditioned , Disease Progression , Esterases/antagonists & inhibitors , Esterases/genetics , Female , Humans , Ligands , Male , Mice , Mice, Inbred C57BL , Organoids/cytology , Organoids/metabolism , Organoids/pathology , Stem Cells/cytology , Stem Cells/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway
16.
Cell Mol Gastroenterol Hepatol ; 12(3): 873-889, 2021.
Article in English | MEDLINE | ID: mdl-34058415

ABSTRACT

BACKGROUND & AIMS: Microfold cells (M cells) are immunosurveillance epithelial cells located in the Peyer's patches (PPs) in the intestine and are responsible for monitoring and transcytosis of antigens, microorganisms, and pathogens. Mature M cells use the receptor glycoprotein 2 (GP2) to aid in transcytosis. Recent studies have shown transcription factors, Spi-B and SRY-Box Transcription Factor 8 (Sox8). are necessary for M-cell differentiation, but not sufficient. An exhaustive set of factors sufficient for differentiation and development of a mature GP2+ M cell remains elusive. Our aim was to understand the role of polycomb repressive complex 2 (PRC2) as an epigenetic regulator of M-cell development. Estrogen-related-receptor γ (Esrrg), identified as a PRC2-regulated gene, was studied in depth, in addition to its relationship with Spi-B and Sox8. METHODS: Comparative chromatin immunoprecipitation and global run-on sequencing analysis of mouse intestinal organoids were performed in stem condition, enterocyte conditions, and receptor activator of nuclear factor κ B ligand-induced M-cell condition. Esrrg, which was identified as one of the PRC2-regulated transcription factors, was studied in wild-type mice and knocked out in intestinal organoids using guide RNA's. Sox8 null mice were used to study Esrrg and its relation to Sox8. RESULTS: chromatin immunoprecipitation and global run-on sequencing analysis showed 12 novel PRC2 regulated transcription factors, PRC2-regulated Esrrg is a novel M-cell-specific transcription factor acting on a receptor activator of nuclear factor κB ligand-receptor activator of nuclear factor κB-induced nuclear factor-κB pathway, upstream of Sox8, and necessary but not sufficient for a mature M-cell marker of Gp2 expression. CONCLUSIONS: PRC2 regulates a significant set of genes in M cells including Esrrg, which is critical for M-cell development and differentiation. Loss of Esrrg led to an immature M-cell phenotype lacking in Sox8 and Gp2 expression. Transcript profiling: the data have been deposited in the NCBI Gene Expression Omnibus database (GSE157629).


Subject(s)
Epithelial Cells/metabolism , Gene Expression Regulation , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Peyer's Patches/cytology , Peyer's Patches/metabolism , Polycomb Repressive Complex 2/metabolism , Animals , Biomarkers , Cell Differentiation/genetics , Gene Expression Profiling , Intestinal Mucosa/immunology , Mice , NF-kappa B/metabolism , Peyer's Patches/immunology , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , Signal Transduction
17.
Cell Rep Methods ; 1(5): 100059, 2021 09 27.
Article in English | MEDLINE | ID: mdl-35474669

ABSTRACT

The intestine is divided into functionally distinct regions along the anteroposterior (A/P) axis. How the regional identity influences the function of intestinal stem cells (ISCs) and their offspring remain largely unresolved. We introduce an imaging-based method, "Linear Analysis of Midgut" (LAM), which allows quantitative, regionally defined cellular phenotyping of the whole Drosophila midgut. LAM transforms image-derived cellular data from three-dimensional midguts into a linearized representation, binning it into segments along the A/P axis. Through automated multivariate determination of regional borders, LAM allows mapping and comparison of cellular features and frequencies with subregional resolution. Through the use of LAM, we quantify the distributions of ISCs, enteroblasts, and enteroendocrine cells in a steady-state midgut, and reveal unprecedented regional heterogeneity in the ISC response to a Drosophila model of colitis. Altogether, LAM is a powerful tool for organ-wide quantitative analysis of the regional heterogeneity of midgut cells.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Intestines , Enteroendocrine Cells , Stem Cells
18.
Mech Ageing Dev ; 191: 111330, 2020 10.
Article in English | MEDLINE | ID: mdl-32805262

ABSTRACT

Like most tissues, intestine shows multiple alterations during aging. While the main function of nutrient absorption is relatively well maintained, capacity of the intestine to respond to abrupt changes or damage declines with age. The reduction in renewal and regeneration capacity results from alterations in the stem cells that renew the epithelium, and in the complex interactions stem cells have with their microenvironment, or the Niche. This review highlights recent evidence on age-associated changes in the intestinal stem cell function, and focuses on stem cell extrinsic mechanisms. Strategies targeting niche interactions have already shown promise in alleviating problems associated with intestinal aging in animal models, and may provide means to protect the elderly for example from chemotherapy induced gastrointestinal side-effects.


Subject(s)
Aging/metabolism , Intestinal Mucosa/metabolism , Stem Cell Niche , Stem Cells/metabolism , Animals , Humans
19.
Cancer Lett ; 492: 54-62, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32711097

ABSTRACT

Estrogen hormones protect against colorectal cancer (CRC) and a preventative role of estrogen receptor beta (ERß) on CRC has been supported using full knockout animals. However, it is unclear through which cells or organ ERß mediates this effect. To investigate the functional role of intestinal ERß during colitis-associated CRC we used intestine-specific ERß knockout mice treated with azoxymethane and dextran sodium sulfate, followed by ex vivo organoid culture to corroborate intrinsic effects. We explored genome-wide impact on TNFα signaling using human CRC cell lines and chromatin immunoprecipitation assay to mechanistically characterize the regulation of ERß. Increased tumor formation in males and tumor size in females was noted upon intestine-specific ERß knockout, accompanied by enhanced local expression of TNFα, deregulation of key NFκB targets, and increased colon ulceration. Unexpectedly, we noted especially strong effects in males. We corroborated that intestinal ERß protects against TNFα-induced damage intrinsically, and characterized an underlying genome-wide signaling mechanism in CRC cell lines whereby ERß binds to cis-regulatory chromatin areas of key NFκB regulators. Our results support a protective role of intestinal ERß against colitis-associated CRC, proposing new therapeutic strategies.


Subject(s)
Colitis/prevention & control , Colorectal Neoplasms/prevention & control , Estrogen Receptor beta/physiology , Animals , Cell Line, Tumor , Cell Proliferation , Female , Humans , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/physiology , Sex Characteristics , Tumor Necrosis Factor-alpha/pharmacology
20.
Gastroenterology ; 158(5): 1389-1401.e10, 2020 04.
Article in English | MEDLINE | ID: mdl-31930988

ABSTRACT

BACKGROUND & AIMS: In addition to the Notch and Wnt signaling pathways, energy metabolism also regulates intestinal stem cell (ISC) function. Tumor suppressor and kinase STK11 (also called LKB1) regulates stem cells and cell metabolism. We investigated whether loss of LKB1 alters ISC homeostasis in mice. METHODS: We deleted LKB1 from ISCs in mice using Lgr5-regulated CRE-ERT2 (Lkb1Lgr5-KO mice) and the traced lineages by using a CRE-dependent TdTomato reporter. Intestinal tissues were collected and analyzed by immunohistochemical and immunofluorescence analyses. We purified ISCs and intestinal progenitors using flow cytometry and performed RNA-sequencing analysis. We measured organoid-forming capacity and ISC percentages using intestinal tissues from Lkb1Lgr5-KO mice. We analyzed human Ls174t cells with knockdown of LKB1 or other proteins by immunoblotting, real-time quantitative polymerase chain reaction, and the Seahorse live-cell metabolic assay. RESULTS: Some intestinal crypts from Lkb1Lgr5-KO mice lost ISCs compared with crypts from control mice. However, most crypts from Lkb1Lgr5-KO mice contained functional ISCs that expressed increased levels of Atoh1 messenger RNA (mRNA), acquired a gene expression signature associated with secretory cells, and generated more cells in the secretory lineage compared with control mice. Knockdown of LKB1 in Ls174t cells induced expression of Atoh1 mRNA and a phenotype of increased mucin production; knockdown of ATOH1 prevented induction of this phenotype. The increased expression of Atoh1 mRNA after LKB1 loss from ISCs or Ls174t cells did not involve Notch or Wnt signaling. Knockdown of pyruvate dehydrogenase kinase 4 (PDK4) or inhibition with dichloroacetate reduced the up-regulation of Atoh1 mRNA after LKB1 knockdown in Ls174t cells. Cells with LKB1 knockdown had a reduced rate of oxygen consumption, which was partially restored by PDK4 inhibition with dichloroacetate. ISCs with knockout of LKB1 increased the expression of PDK4 and had an altered metabolic profile. CONCLUSIONS: LKB1 represses transcription of ATOH1, via PDK4, in ISCs, restricting their differentiation into secretory lineages. These findings provide a connection between metabolism and the fate determination of ISCs.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Energy Metabolism/physiology , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Stem Cells/physiology , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Dichloroacetic Acid/pharmacology , Gene Knockdown Techniques , HEK293 Cells , Humans , Intestinal Mucosa/cytology , Intestine, Small/cytology , Mice , Mice, Knockout , Primary Cell Culture , Protein Serine-Threonine Kinases/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/antagonists & inhibitors , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , RNA-Seq , Transcription, Genetic , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...