Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.571
Filter
1.
Sci Adv ; 10(32): eadl1584, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39110797

ABSTRACT

Histone acetyltransferases KAT2A and KAT2B are paralogs highly expressed in the intestinal epithelium, but their functions are not well understood. In this study, double knockout of murine Kat2 genes in the intestinal epithelium was lethal, resulting in robust activation of interferon signaling and interferon-associated phenotypes including the loss of intestinal stem cells. Use of pharmacological agents and sterile organoid cultures indicated a cell-intrinsic double-stranded RNA trigger for interferon signaling. Acetyl-proteomics and sequencing of immunoprecipitated double-stranded RNA were used to interrogate the mechanism behind this response, which identified mitochondria-encoded double-stranded RNA as the source of intrinsic interferon signaling. Kat2a and Kat2b therefore play an essential role in regulating mitochondrial functions and maintaining intestinal health.


Subject(s)
Histone Acetyltransferases , Interferons , Mice, Knockout , RNA, Double-Stranded , Signal Transduction , Stem Cells , Animals , RNA, Double-Stranded/metabolism , Mice , Stem Cells/metabolism , Stem Cells/cytology , Interferons/metabolism , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/cytology , Mitochondria/metabolism , Cell Self Renewal/genetics , Intestines/cytology
2.
Science ; 385(6708): eadk1679, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39088603

ABSTRACT

Neuroimmune cross-talk participates in intestinal tissue homeostasis and host defense. However, the matrix of interactions between arrays of molecularly defined neuron subsets and of immunocyte lineages remains unclear. We used a chemogenetic approach to activate eight distinct neuronal subsets, assessing effects by deep immunophenotyping, microbiome profiling, and immunocyte transcriptomics in intestinal organs. Distinct immune perturbations followed neuronal activation: Nitrergic neurons regulated T helper 17 (TH17)-like cells, and cholinergic neurons regulated neutrophils. Nociceptor neurons, expressing Trpv1, elicited the broadest immunomodulation, inducing changes in innate lymphocytes, macrophages, and RORγ+ regulatory T (Treg) cells. Neuroanatomical, genetic, and pharmacological follow-up showed that Trpv1+ neurons in dorsal root ganglia decreased Treg cell numbers via the neuropeptide calcitonin gene-related peptide (CGRP). Given the role of these neurons in nociception, these data potentially link pain signaling with gut Treg cell function.


Subject(s)
Calcitonin Gene-Related Peptide , Ganglia, Spinal , Neuroimmunomodulation , Nociceptors , T-Lymphocytes, Regulatory , TRPV Cation Channels , Th17 Cells , Animals , Mice , Calcitonin Gene-Related Peptide/metabolism , Calcitonin Gene-Related Peptide/genetics , Cholinergic Neurons/metabolism , Ganglia, Spinal/metabolism , Ganglia, Spinal/cytology , Gastrointestinal Microbiome , Intestines/immunology , Intestines/cytology , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Nociception , Nociceptors/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/immunology , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics
3.
Int Immunopharmacol ; 138: 112614, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38972212

ABSTRACT

Intestinal stem cells (ISCs) play a crucial role in maintaining the equilibrium and regenerative potential of intestinal tissue, thereby ensuring tissue homeostasis and promoting effective tissue regeneration following injury. It has been proven that targeting Toll-like receptors (TLRs) can help prevent radiation-induced damage to the intestine. In this study, we established an intestinal injury model using IR and evaluated the effects of CL429 on ISC regeneration both in vivo and in vitro. Following radiation exposure, mice treated with CL429 showed a significant increase in survival rates (100% survival in the treated group compared to 54.54% in the control group). CL429 also showed remarkable efficacy in inhibiting radiation-induced intestinal damage and promoting ISC proliferation and regeneration. In addition, CL429 protected intestinal organoids against IR-induced injury. Mechanistically, RNA sequencing and Western blot analysis revealed the activation of the Wnt and Hippo signaling pathways by CL429. Specifically, we observed a significant upregulation of YAP1, a key transcription factor in the Hippo pathway, upon CL429 stimulation. Furthermore, knockdown of YAP1 significantly attenuated the radioprotective effect of CL429 on intestinal organoids, indicating that CL429-mediated intestinal radioprotection is dependent on YAP1. In addition, we investigated the relationship between TLR2 and YAP1 using TLR2 knockout mice, and our results showed that TLR2 knockout abolished the activation of CL429 on YAP1. Taken together, our study provides evidence supporting the role of CL429 in promoting ISC regeneration through activation of TLR2-YAP1. And further investigation of the interaction between TLRs and other signaling pathways may enhance our understanding of ISC regeneration after injury.


Subject(s)
Intestines , Stem Cells , Toll-Like Receptor 2 , YAP-Signaling Proteins , Animals , Male , Mice , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Proliferation/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/radiation effects , Intestines/cytology , Mice, Inbred C57BL , Organoids/metabolism , Regeneration , Signal Transduction , Stem Cells/metabolism , Stem Cells/drug effects , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 2/genetics , Up-Regulation
4.
PLoS One ; 19(7): e0300666, 2024.
Article in English | MEDLINE | ID: mdl-39052651

ABSTRACT

Mechanistic investigation of host-microbe interactions in the human gut are hindered by difficulty of co-culturing microbes with intestinal epithelial cells. On one hand the gut bacteria are a mix of facultative, aerotolerant or obligate anaerobes, while the intestinal epithelium requires oxygen for growth and function. Thus, a coculture system that can recreate these contrasting oxygen requirements is critical step towards our understanding microbial-host interactions in the human gut. Here, we demonstrate Intestinal Organoid Physoxic Coculture (IOPC) system, a simple and cost-effective method for coculturing anaerobic intestinal bacteria with human intestinal organoids (HIOs). Using commensal anaerobes with varying degrees of oxygen tolerance, such as nano-aerobe Bacteroides thetaiotaomicron and strict anaerobe Blautia sp., we demonstrate that IOPC can successfully support 24-48 hours HIO-microbe coculture. The IOPC recapitulates the contrasting oxygen conditions across the intestinal epithelium seen in vivo. The IOPC cultured HIOs showed increased barrier integrity, and induced expression of immunomodulatory genes. A transcriptomic analysis suggests that HIOs from different donors show differences in the magnitude of their response to coculture with anaerobic bacteria. Thus, the IOPC system provides a robust coculture setup for investigating host-microbe interactions in complex, patient-derived intestinal tissues, that can facilitate the study of mechanisms underlying the role of the microbiome in health and disease.


Subject(s)
Coculture Techniques , Intestinal Mucosa , Organoids , Oxygen , Humans , Organoids/microbiology , Organoids/metabolism , Oxygen/metabolism , Coculture Techniques/methods , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/cytology , Gastrointestinal Microbiome , Host Microbial Interactions , Bacteria, Anaerobic/growth & development , Bacteria, Anaerobic/metabolism , Intestines/microbiology , Intestines/cytology , Bacteroides thetaiotaomicron/metabolism
5.
Cell Rep ; 43(7): 114305, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38906148

ABSTRACT

Planarian flatworms undergo continuous internal turnover, wherein old cells are replaced by the division progeny of adult pluripotent stem cells (neoblasts). How cell turnover is carried out at the organismal level remains an intriguing question in planarians and other systems. While previous studies have predominantly focused on neoblast proliferation, little is known about the processes that mediate cell loss during tissue homeostasis. Here, we use the planarian epidermis as a model to study the mechanisms of cell removal. We established a covalent dye-labeling assay and image analysis pipeline to quantify the cell turnover rate in the planarian epidermis. Our findings indicate that the ventral epidermis is highly dynamic and epidermal cells undergo internalization via basal extrusion, followed by a relocation toward the intestine and ultimately digestion by intestinal phagocytes. Overall, our study reveals a complex homeostatic process of cell clearance that may generally allow planarians to catabolize their own cells.


Subject(s)
Epidermis , Intestines , Planarians , Animals , Planarians/metabolism , Planarians/physiology , Epidermis/metabolism , Intestines/cytology , Epidermal Cells/metabolism , Homeostasis
6.
Life Sci ; 352: 122875, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38942359

ABSTRACT

The complex and dynamic environment of the gastrointestinal tract shapes one of the fastest renewing tissues in the human body, the intestinal epithelium. Considering the lack of human preclinical studies, reliable models that mimic the intestinal environment are increasingly explored. Patient-derived intestinal organoids are powerful tools that recapitulate in vitro many pathophysiological features of the human intestine. In this review, the possible applications of human intestinal organoids in different research fields are highlighted. From physiologically relevant to intestinal disease modeling, regenerative medicine, and toxicology studies, the potential of intestinal organoids will be here presented and discussed. Despite the remarkable opportunities offered, limitations related to ethical concerns, tissue collection, reproducibility, and methodologies may hinder the full exploitation of this cell-based model into high throughput studies and clinical practice. Currently, distinct approaches can be used to overcome the numerous challenges found along the way and to allow the full implementation of this ground-breaking technology.


Subject(s)
Intestinal Mucosa , Organoids , Humans , Organoids/cytology , Intestinal Mucosa/cytology , Regenerative Medicine/methods , Intestines/cytology , Intestines/physiology , Animals , Intestinal Diseases/pathology , Models, Biological
7.
Proc Natl Acad Sci U S A ; 121(25): e2321228121, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38857399

ABSTRACT

Ciliary defects are linked to ciliopathies, but impairments in the sensory cilia of Caenorhabditis elegans neurons extend lifespan, a phenomenon with previously unclear mechanisms. Our study reveals that neuronal cilia defects trigger the unfolded protein response of the endoplasmic reticulum (UPRER) within intestinal cells, a process dependent on the insulin/insulin-like growth factor 1 (IGF-1) signaling transcription factor and the release of neuronal signaling molecules. While inhibiting UPRER doesn't alter the lifespan of wild-type worms, it normalizes the extended lifespan of ciliary mutants. Notably, deactivating the cyclic nucleotide-gated (CNG) channel TAX-4 on the ciliary membrane promotes lifespan extension through a UPRER-dependent mechanism. Conversely, constitutive activation of TAX-4 attenuates intestinal UPRER in ciliary mutants. Administering a CNG channel blocker to worm larvae activates intestinal UPRER and increases adult longevity. These findings suggest that ciliary dysfunction in sensory neurons triggers intestinal UPRER, contributing to lifespan extension and implying that transiently inhibiting ciliary channel activity may effectively prolong lifespan.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Cilia , Longevity , Unfolded Protein Response , Animals , Caenorhabditis elegans/metabolism , Cilia/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Cyclic Nucleotide-Gated Cation Channels/metabolism , Cyclic Nucleotide-Gated Cation Channels/genetics , Intestines/cytology , Signal Transduction , Neurons/metabolism , Endoplasmic Reticulum/metabolism , Insulin-Like Growth Factor I/metabolism , Intestinal Mucosa/metabolism
8.
Cell ; 187(12): 3056-3071.e17, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38848678

ABSTRACT

The currently accepted intestinal epithelial cell organization model proposes that Lgr5+ crypt-base columnar (CBC) cells represent the sole intestinal stem cell (ISC) compartment. However, previous studies have indicated that Lgr5+ cells are dispensable for intestinal regeneration, leading to two major hypotheses: one favoring the presence of a quiescent reserve ISC and the other calling for differentiated cell plasticity. To investigate these possibilities, we studied crypt epithelial cells in an unbiased fashion via high-resolution single-cell profiling. These studies, combined with in vivo lineage tracing, show that Lgr5 is not a specific ISC marker and that stemness potential exists beyond the crypt base and resides in the isthmus region, where undifferentiated cells participate in intestinal homeostasis and regeneration following irradiation (IR) injury. Our results provide an alternative model of intestinal epithelial cell organization, suggesting that stemness potential is not restricted to CBC cells, and neither de-differentiation nor reserve ISC are drivers of intestinal regeneration.


Subject(s)
Homeostasis , Intestinal Mucosa , Receptors, G-Protein-Coupled , Regeneration , Stem Cells , Animals , Stem Cells/metabolism , Stem Cells/cytology , Mice , Intestinal Mucosa/metabolism , Receptors, G-Protein-Coupled/metabolism , Intestines/cytology , Cell Differentiation , Mice, Inbred C57BL , Epithelial Cells/metabolism , Single-Cell Analysis , Male
9.
PLoS One ; 19(6): e0304526, 2024.
Article in English | MEDLINE | ID: mdl-38857221

ABSTRACT

In vitro models, such as primary cells and continuous cell lines routinely used for evaluating drug candidates, have limitations in their translational relevance to human diseases. Organotypic cultures are increasingly being used to assess therapeutics for various cancers and infectious diseases. Monitoring drug cytotoxicity in cell cultures is crucial in drug development, and several commercially available kits for cytotoxicity assessment offer distinct advantages and limitations. Given the complexity of organoid cultures, including donor-driven variability, we investigated drug-treated, tissue stem cell-derived human intestinal organoid responses with commonly used cell cytotoxicity assay kits. Using seven different compounds, we compared the cytotoxicity assay performance of two different leaky membrane-based and two metabolism-based assays. Significant variability was seen in reported viability outcomes across assays and organoid lines. High baseline activity of lactate dehydrogenase (LDH) in four human intestinal organoid lines required modification of the standard LDH assay protocol. Additionally, the LDH assay reported unique resilience to damage in a genetically-modified line contrasting results compared to other assays. This study highlights factors that can impact the measurement of cell cytotoxicity in intestinal organoid models, which are emerging as valuable new tools for research and pre-clinical drug testing and suggest the need for using multiple assay types to ensure reliable cytotoxicity assessment.


Subject(s)
L-Lactate Dehydrogenase , Organoids , Humans , Organoids/drug effects , Organoids/metabolism , Organoids/cytology , L-Lactate Dehydrogenase/metabolism , Cell Survival/drug effects , Intestines/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism
10.
Cell ; 187(12): 2900-2902, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38848673

ABSTRACT

In tissue homeostasis, intestinal stem cells (ISCs) undergo continuous self-renewal to sustain rapid cellular turnover. In this issue of Cell, Capdevila et al.1 and Malagola, Vasciaveo, et al.2 identify a new ISC population in the upper crypt that can generate Lgr5+ stem cells during homeostasis.


Subject(s)
Intestines , Stem Cells , Stem Cells/cytology , Stem Cells/metabolism , Intestines/cytology , Animals , Humans , Homeostasis , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Receptors, G-Protein-Coupled/metabolism , Mice , Cell Differentiation
11.
Biofabrication ; 16(4)2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38914075

ABSTRACT

Accurate reproduction of human intestinal structure and functionin vitrois of great significance for understanding the development and disease occurrence of the gut. However, mostin vitrostudies are often confined to 2D models, 2.5D organ chips or 3D organoids, which cannot fully recapitulate the tissue architecture, microenvironment and cell compartmentalization foundin vivo. Herein, a centimeter-scale intestine tissue that contains intestinal features, such as hollow tubular structure, capillaries and tightly connected epithelium with invivo-likering folds, crypt-villi, and microvilli is constructed by 3D embedding bioprinting. In our strategy, a novel photocurable bioink composed of methacrylated gelatin, methacrylated sodium alginate and poly (ethylene glycol) diacrylate is developed for the fabrication of intestinal model. The Caco-2 cells implanted in the lumen are induced by the topological structures of the model to derive microvilli, crypt-villi, and tight junctions, simulating the intestinal epithelial barrier. The human umbilical vein endothelial cells encapsulated within the model gradually form microvessels, mimicking the dense capillary network in the intestine. This intestine-like tissue, which closely resembles the structure and cell arrangement of the human gut, can act as a platform to predict the therapeutic and toxic side effects of new drugs on the intestine.


Subject(s)
Bioprinting , Capillaries , Human Umbilical Vein Endothelial Cells , Intestines , Printing, Three-Dimensional , Humans , Caco-2 Cells , Capillaries/cytology , Intestines/cytology , Tissue Engineering , Alginates/chemistry , Polyethylene Glycols/chemistry , Tissue Scaffolds/chemistry , Intestinal Mucosa/cytology , Gelatin/chemistry
12.
Nat Commun ; 15(1): 5493, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38944657

ABSTRACT

JNK signaling is a critical regulator of inflammation and regeneration, but how it is controlled in specific tissue contexts remains unclear. Here we show that, in the Drosophila intestine, the TNF-type ligand, Eiger (Egr), is expressed exclusively by intestinal stem cells (ISCs) and enteroblasts (EBs), where it is induced by stress and during aging. Egr preferentially activates JNK signaling in a paracrine fashion in differentiated enterocytes (ECs) via its receptor, Grindelwald (Grnd). N-glycosylation genes (Alg3, Alg9) restrain this activation, and stress-induced downregulation of Alg3 and Alg9 correlates with JNK activation, suggesting a regulatory switch. JNK activity in ECs induces expression of the intermembrane protease Rhomboid (Rho), driving secretion of EGFR ligands Keren (Krn) and Spitz (Spi), which in turn activate EGFR signaling in progenitor cells (ISCs and EBs) to stimulate their growth and division, as well as to produce more Egr. This study uncovers an N-glycosylation-controlled, paracrine JNK-EGFR-JNK feedforward loop that sustains ISC proliferation during stress-induced gut regeneration.


Subject(s)
Drosophila Proteins , ErbB Receptors , Intestines , MAP Kinase Signaling System , Animals , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , ErbB Receptors/metabolism , ErbB Receptors/genetics , Intestines/cytology , Drosophila melanogaster/metabolism , Drosophila melanogaster/genetics , Enterocytes/metabolism , Enterocytes/cytology , Stem Cells/metabolism , Stem Cells/cytology , Intestinal Mucosa/metabolism , Intestinal Mucosa/cytology , Drosophila/metabolism , Glycosylation , Receptors, Invertebrate Peptide/metabolism , Receptors, Invertebrate Peptide/genetics , Cell Proliferation , JNK Mitogen-Activated Protein Kinases/metabolism , Signal Transduction , Cell Communication , Cell Differentiation , Epidermal Growth Factor , Membrane Proteins
13.
Cell Stem Cell ; 31(5): 591-592, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38701755

ABSTRACT

Recently in Cell Metabolism, Wei et al.1 unveiled a brain-to-gut pathway that conveys psychological stress to intestinal epithelial cells, leading to their dysfunction. This gut-brain axis involves a microbial metabolite, indole-3-acetate (IAA), as a niche signal that hampers mitochondrial respiration to skew intestinal stem cell (ISC) fate.


Subject(s)
Stem Cells , Stem Cells/metabolism , Stem Cells/cytology , Animals , Humans , Intestines/cytology , Intestines/microbiology , Stress, Physiological , Gastrointestinal Microbiome/physiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Cell Differentiation , Mitochondria/metabolism
14.
Biofabrication ; 16(3)2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38788705

ABSTRACT

Fabrication of engineered intestinal tissues with the structures and functions as humans is crucial and promising as the tools for developing drugs and functional foods. The aim of this study is to fabricate an engineered intestinal tissue from Caco-2 cells by air-liquid interface culture using a paper-based dual-layer scaffold and analyze its structure and functions. Just by simply placing on a folded paper soaked in the medium, the electrospun gelatin microfiber mesh as the upper cell adhesion layer of the dual-layer scaffold was exposed to the air, while the lower paper layer worked to preserve and supply the cell culture medium to achieve stable culture over several weeks. Unlike the flat tissue produced using the conventional commercial cultureware, Transwell, the engineered intestinal tissue fabricated in this study formed three-dimensional villous architectures. Microvilli and tight junction structures characteristic of epithelial tissue were also formed at the apical side. Furthermore, compared to the tissue prepared by Transwell, mucus production was significantly larger, and the enzymatic activities of drug metabolism and digestion were almost equivalent. In conclusion, the air-liquid interface culture using the paper-based dual-layer scaffold developed in this study was simple but effective in fabricating the engineered intestinal tissue with superior structures and functions.


Subject(s)
Mucus , Paper , Tissue Engineering , Tissue Scaffolds , Tissue Scaffolds/chemistry , Humans , Caco-2 Cells , Mucus/metabolism , Intestines/cytology , Intestines/physiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/cytology , Air , Cell Culture Techniques/methods
15.
Fish Shellfish Immunol ; 150: 109605, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38704111

ABSTRACT

Crucian carp (Carassius carassius) is an important aquatic economic animal, and the immune barrier function of its intestine has been a focus of research into oral vaccines and drugs. However, the histological structures of the intestinal barrier and its adjacent areas have not been clearly established, and little subcellular evidence is available to elucidate the spatial distribution of intracellular biological processes. In this study, the spatial distribution of autophagy and endosome formation in the intestinal epithelial cells (IECs) of crucian carp were analyzed. These two biological activities are closely related to intestinal homeostasis, immunity, and cell communication. Periodic acid-Schiff (PAS) and Masson's trichrome staining were employed to elucidate the distinctive histological framework of the Crucian carp's myoid cell network, which resides within the subepithelial layer and is characterized by gap junctions. Transmission electron microscopy (TEM), immunohistochemistry (IHC), and immunofluorescence (IF) were used to detect the structural and functional aspects of the IEC in different intestinal segments. TEM and immunohistochemical analyses captured the biogenesis and maturation of early and late endosomes as well as multivesicular bodies (MVBs), as well as the initiation and progression of autophagy, including macroautophagy and mitophagy. The endosome and MVBs-specific marker CD63 and autophagy-related protein LC3 were highly expressed in IECs and were correlated with autophagy and endosome biosynthesis in the apical and basal regions of individual cells, and differed between different intestinal segments. In summary, this study elucidated the ubiquity and morphological characteristics of autophagy and endosome formation across different intestinal segments of crucian carp. A unique myoid cell network beneath the intestinal epithelium in crucian carp was also identified, expanding the histological understanding of this animal's intestinal tract.


Subject(s)
Autophagy , Carps , Endosomes , Animals , Carps/immunology , Endosomes/immunology , Endosomes/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/cytology , Intestines/immunology , Intestines/cytology , Epithelial Cells/immunology
16.
Stem Cell Res Ther ; 15(1): 155, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38816841

ABSTRACT

In the past decade, intestinal organoid technology has paved the way for reproducing tissue or organ morphogenesis during intestinal physiological processes in vitro and studying the pathogenesis of various intestinal diseases. Intestinal organoids are favored in drug screening due to their ability for high-throughput in vitro cultivation and their closer resemblance to patient genetic characteristics. Furthermore, as disease models, intestinal organoids find wide applications in screening diagnostic markers, identifying therapeutic targets, and exploring epigenetic mechanisms of diseases. Additionally, as a transplantable cellular system, organoids have played a significant role in the reconstruction of damaged epithelium in conditions such as ulcerative colitis and short bowel syndrome, as well as in intestinal material exchange and metabolic function restoration. The rise of interdisciplinary approaches, including organoid-on-chip technology, genome editing techniques, and microfluidics, has greatly accelerated the development of organoids. In this review, VOSviewer software is used to visualize hot co-cited journal and keywords trends of intestinal organoid firstly. Subsequently, we have summarized the current applications of intestinal organoid technology in disease modeling, drug screening, and regenerative medicine. This will deepen our understanding of intestinal organoids and further explore the physiological mechanisms of the intestine and drug development for intestinal diseases.


Subject(s)
Organoids , Organoids/metabolism , Organoids/cytology , Humans , Intestines/cytology , Animals , Regenerative Medicine/methods , Intestinal Mucosa/metabolism , Intestinal Mucosa/cytology
17.
Curr Protoc ; 4(5): e1062, 2024 May.
Article in English | MEDLINE | ID: mdl-38775005

ABSTRACT

The architecture and morphology of the intestinal tissue from mice or other small animals are difficult to preserve for histological and molecular analysis due to the fragile nature of this tissue. The intestinal mucosa consists of villi and crypts lined with epithelial cells. In between the epithelial folds extends the lamina propria, a loose connective tissue that contains blood and lymph vessels, fibroblasts, and immune cells. Underneath the mucosa are two layers of contractile smooth muscle and nerves. The tissue experiences significant changes during fixation, which can impair the reliability of histologic analysis. Poor-quality histologic sections are not suitable for quantitative image-based tissue analysis. This article offers a new fixative composed of neutral buffered formalin (NBF) and acetic acid, called FA. This fixative significantly improved the histology of mouse intestinal tissue compared to traditional NBF and enabled precise, reproducible histologic molecular analyses using QuPath software. Algorithmic training of QuPath allows for automated segmentation of intestinal compartments, which can be further interrogated for cellular composition and disease-related changes. © 2024 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol: Improved preservation of mouse intestinal tissue using a formalin/acetic acid fixative Support Protocol: Quantitative tissue analysis using QuPath.


Subject(s)
Acetic Acid , Fixatives , Formaldehyde , Tissue Fixation , Animals , Mice , Tissue Fixation/methods , Intestinal Mucosa/cytology , Intestines/cytology , Intestines/pathology , Software
18.
J Sci Food Agric ; 104(12): 7417-7428, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38760970

ABSTRACT

BACKGROUND: Wuliangye strong aroma baijiu (hereafter, Wuliangye baijiu) is a traditional Chinese grain liquor containing short-chain fatty acids, ethyl caproate, ethyl lactate, other trace components, and a large proportion of ethanol. The effects of Wuliangye baijiu on intestinal stem cells and intestinal epithelial development have not been elucidated. Here, the role of Wuliangye baijiu in intestinal epithelial regeneration and gut microbiota modulation was investigated by administering a Lieber-DeCarli chronic ethanol liquid diet in a mouse model to mimic long-term (8 weeks') light/moderate alcohol consumption (1.6 g kg-1 day-1) in healthy human adults. RESULTS: Wuliangye baijiu promoted colonic crypt proliferation in mice. According to immunofluorescence and reverse transcription-quantitative polymerase chain reaction analyses, compared with the ethanol-only treatment, Wuliangye baijiu increased the number of intestinal stem cells and goblet cells and the expression of enteroendocrine cell differentiation markers in the mouse colon. Furthermore, gut microbiota analysis showed an increase in the relative abundance of microbiota related to intestinal homeostasis following Wuliangye baijiu administration. Notably, increased abundance of Bacteroidota, Faecalibaculum, Lachnospiraceae, and Blautia may play an essential role in promoting stem-cell-mediated intestinal epithelial development and maintaining intestinal homeostasis. CONCLUSIONS: In summary, these findings suggest that Wuliangye baijiu can be used to regulate intestinal stem cell proliferation and differentiation in mice and to alter gut microbiota distributions, thereby promoting intestinal homeostasis. This research elucidates the mechanism by which Wuliangye baijiu promotes intestinal health. © 2024 Society of Chemical Industry.


Subject(s)
Bacteria , Cell Differentiation , Cell Proliferation , Gastrointestinal Microbiome , Homeostasis , Intestinal Mucosa , Stem Cells , Animals , Mice , Gastrointestinal Microbiome/drug effects , Cell Proliferation/drug effects , Cell Differentiation/drug effects , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/cytology , Humans , Male , Bacteria/classification , Bacteria/isolation & purification , Bacteria/genetics , Bacteria/metabolism , Intestinal Mucosa/metabolism , Mice, Inbred C57BL , Intestines/microbiology , Intestines/cytology , Alcoholic Beverages/analysis , Ethanol , Colon/microbiology , Colon/metabolism
19.
In Vitro Cell Dev Biol Anim ; 60(6): 689-696, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38656569

ABSTRACT

Hair follicle stem cells (HFSCs) are adult stem cells located in the outer root sheath of the follicle bulge with high neural plasticity, which promise a potential for the stem cell therapy for neurological diseases. Hirschsprung's disease (HD) is characterized by the absence of ganglia in the distant bowel. In this study, we elucidated the capacity of HFSCs to differentiate into neuronal cells in the aganglionic colon from embryonic rat. HFSCs were isolated from adult Sprague-Dawley (SD) rats and formed spheres that could be passaged. The cultured HFSCs expressed neural crest stem cells (NCSCs) markers such as SOX10, CD34, and nestin, which indicated their neural crest lineage. Subsequent differentiation assays demonstrated that these cells could give rise to neural progeny that expressed neuronal or glial markers. The aganglionic colon from the embryonic intestine was applied as in vitro explant to test the capacity of proliferation and differentiation of HFSCs. The HFSCs expressing GFP or RFP integrated in intestinal explants and maintained proliferative capacity. Moreover, the HFSCs differentiated into Tuj1- or S100ß-positive cells in the cultured intestinal explants. The results proposed that the HFSCs might be an alternative source of neural stem cells for the HD therapy.


Subject(s)
Adult Stem Cells , Cell Differentiation , Hair Follicle , Neurons , Rats, Sprague-Dawley , Animals , Neurons/cytology , Neurons/metabolism , Hair Follicle/cytology , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Rats , Cell Proliferation , Intestines/cytology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Cells, Cultured
20.
BMC Mol Cell Biol ; 25(1): 14, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38689222

ABSTRACT

BACKGROUND: Emerging evidence underscores the responsiveness of the mammalian intestine to dietary cues, notably through the involvement of LGR5 + intestinal stem cells in orchestrating responses to diet-driven signals. However, the effects of high-fat diet (HFD) on these cellular dynamics and their impact on gut integrity remain insufficiently understood. Our study aims to assess the multifaceted interactions between palmitic acid (PA), cell proliferation, and the intestinal epithelial barrier using a canine colonoid model. Canine models, due to their relevance in simulating human intestinal diseases, offer a unique platform to explore the molecular mechanisms underlying HFD derived intestinal dysfunction. RESULTS: Canine colonoids were subjected to PA exposure, a surrogate for the effects of HFD. This intervention revealed a remarkable augmentation of cell proliferative activity. Furthermore, we observed a parallel reduction in transepithelial electrical resistance (TEER), indicating altered epithelium barrier integrity. While E-cadherin exhibited consistency, ZO-1 displayed a noteworthy reduction in fluorescence intensity within the PA-exposed group. CONCLUSIONS: By employing canine intestinal organoid systems, we provide compelling insights into the impact of PA on intestinal physiology. These findings underscore the importance of considering both cell proliferative activity and epithelial integrity in comprehending the repercussions of HFDs on intestinal health. Our study contributes to a deeper understanding of the consequences of HFD on intestinal homeostasis, utilizing valuable translational in vitro models derived from dogs.


Subject(s)
Cell Proliferation , Diet, High-Fat , Intestinal Mucosa , Organoids , Palmitic Acid , Animals , Dogs , Diet, High-Fat/adverse effects , Intestinal Barrier Function , Intestinal Mucosa/metabolism , Intestinal Mucosa/cytology , Intestines/cytology , Intestines/physiology , Organoids/metabolism , Organoids/cytology , Palmitic Acid/metabolism , Palmitic Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL