Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 946
Filter
1.
Biochem Biophys Res Commun ; 726: 150280, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-38909534

ABSTRACT

Esophageal epithelium is one of the most proliferative and regenerative epithelia in our body, indicating robust stem cell activity. However, the underlying mechanisms regulating the self-renewal and differentiation of esophageal stem cells need to be more elucidated. Here, we identify the role of YAP1 in esophageal stem cells. YAP1 is differentially expressed in the nuclei of esophageal basal cells. Furthermore, the treatment of verteporfin, a YAP1 inhibitor, interfered with esophageal organoid formation. Consistently, YAP1 deletion decreased esophageal organoid formation and the expression of basal genes while increasing the expression of suprabasal genes. Finally, global transcriptomic analysis revealed that YAP1 inhibition induced a significant enrichment of gene sets related to keratinization and cornification, while depleting gene sets related to DNA repair and chromosome maintenance. Our data uncover a novel regulatory mechanism for esophageal stem cells, which could provide a potential strategy for esophageal regenerative medicine.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Differentiation , Cell Self Renewal , Esophagus , Stem Cells , YAP-Signaling Proteins , YAP-Signaling Proteins/metabolism , Stem Cells/metabolism , Stem Cells/cytology , Esophagus/cytology , Esophagus/metabolism , Animals , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Mice , Humans , Organoids/metabolism , Organoids/cytology
2.
J Vis Exp ; (207)2024 05 10.
Article in English | MEDLINE | ID: mdl-38801260

ABSTRACT

The squamous epithelium of the esophagus is directly exposed to the environment, continuously facing foreign antigens, including food antigens and microbes. Maintaining the integrity of the epithelial barrier is critical for preventing infections and avoiding inflammation caused by harmless food-derived antigens. This article provides simplified protocols for generating human esophageal organoids and air-liquid interface cultures from patient biopsies to study the epithelial compartment of the esophagus in the context of tissue homeostasis and disease. These protocols have been significant scientific milestones in the last decade, describing three-dimensional organ-like structures from patient-derived primary cells, organoids, and air-liquid interface cultures. They offer the possibility to investigate the function of specific cytokines, growth factors, and signaling pathways in the esophageal epithelium within a three-dimensional framework while maintaining the phenotypic and genetic properties of the donor. Organoids provide information on tissue microarchitecture by assessing the transcriptome and proteome after cytokine stimulation. In contrast, air-liquid interface cultures allow the assessment of the epithelial barrier integrity through transepithelial resistance (TEER) or macromolecule flux measurements. Combining these organoids and air-liquid interface cultures is a powerful tool to advance research in impaired esophageal epithelial barrier conditions.


Subject(s)
Eosinophilic Esophagitis , Organoids , Eosinophilic Esophagitis/pathology , Eosinophilic Esophagitis/metabolism , Humans , Organoids/pathology , Organoids/metabolism , Cell Culture Techniques, Three Dimensional/methods , Esophagus/pathology , Esophagus/cytology , Cell Culture Techniques/methods , Epithelial Cells/metabolism , Epithelial Cells/pathology
4.
Dev Cell ; 59(10): 1269-1283.e6, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38565145

ABSTRACT

Progenitor cells adapt their behavior in response to tissue demands. However, the molecular mechanisms controlling esophageal progenitor decisions remain largely unknown. Here, we demonstrate the presence of a Troy (Tnfrsf19)-expressing progenitor subpopulation localized to defined regions along the mouse esophageal axis. Lineage tracing and mathematical modeling demonstrate that Troy-positive progenitor cells are prone to undergoing symmetrical fate choices and contribute to esophageal tissue homeostasis long term. Functionally, TROY inhibits progenitor proliferation and enables commitment to differentiation without affecting fate symmetry. Whereas Troy expression is stable during esophageal homeostasis, progenitor cells downregulate Troy in response to tissue stress, enabling proliferative expansion of basal cells refractory to differentiation and reestablishment of tissue homeostasis. Our results demonstrate functional, spatially restricted progenitor heterogeneity in the esophageal epithelium and identify how dynamic regulation of Troy coordinates tissue generation.


Subject(s)
Cell Differentiation , Cell Proliferation , Esophagus , Receptors, Tumor Necrosis Factor , Stem Cells , Animals , Mice , Cell Lineage , Epithelium/metabolism , Esophageal Mucosa/metabolism , Esophageal Mucosa/cytology , Esophagus/cytology , Esophagus/metabolism , Homeodomain Proteins , Homeostasis , Stem Cells/metabolism , Stem Cells/cytology , Receptors, Tumor Necrosis Factor/analysis , Receptors, Tumor Necrosis Factor/metabolism
5.
Int J Immunopathol Pharmacol ; 38: 3946320241249397, 2024.
Article in English | MEDLINE | ID: mdl-38688472

ABSTRACT

Objectives: Resveratrol has been implicated in the differentiation and development of human umbilical cord mesenchymal stem cells. The differentiation of into esophageal fibroblasts is a promising strategy for esophageal tissue engineering. However, the pharmacological effect and underlying mechanism of resveratrol on human umbilical cord mesenchymal stem cells differentiation are unknown. Here, we investigated the effects and mechanism of resveratrol on the differentiation of human umbilical cord mesenchymal stem cells. Methods: Using a transwell-membrane coculture system to culture human umbilical cord mesenchymal stem cells and esophageal fibroblasts, we examined how resveratrol act on the differentiation of human umbilical cord mesenchymal stem cells. Immunocytochemistry, Sirius red staining, quantitative real-time PCR, and Western blotting were performed to examine collagen synthesis and possible signaling pathways in human umbilical cord mesenchymal stem cells. Results: We found that resveratrol promoted collagen synthesis and AKT phosphorylation. However, co-treatment of cells with resveratrol and the PI3K inhibitor LY294002 inhibited collagen synthesis and AKT phosphorylation. We demonstrated that resveratrol down-regulated the expression of IL-6, TGF-ß, caspase-9, and Bax by activating the AKT pathway in human umbilical cord mesenchymal stem cell. Furthermore, resveratrol inhibited phosphorylated NF-ĸB in human umbilical cord mesenchymal stem cells. Conclusion: Our data suggest that resveratrol promotes the differentiation of human umbilical cord mesenchymal stem cells into fibroblasts. The underlying mechanism is associated with the downregulation of IL-6 and TGF-ß via the AKT pathway and by inhibiting the NF-ĸB pathway. Resveratrol may be useful for esophageal tissue engineering.


Subject(s)
Cell Differentiation , Esophagus , Fibroblasts , Mesenchymal Stem Cells , Proto-Oncogene Proteins c-akt , Resveratrol , Signal Transduction , Umbilical Cord , Humans , Resveratrol/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Cell Differentiation/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Signal Transduction/drug effects , Umbilical Cord/cytology , Esophagus/drug effects , Esophagus/cytology , Collagen/metabolism , Cells, Cultured , Coculture Techniques , Interleukin-6/metabolism , Transforming Growth Factor beta/metabolism , Phosphorylation , Caspase 9/metabolism
6.
Cell Mol Gastroenterol Hepatol ; 18(1): 15-40, 2024.
Article in English | MEDLINE | ID: mdl-38452871

ABSTRACT

BACKGROUND & AIMS: Autophagy plays roles in esophageal pathologies both benign and malignant. Here, we aim to define the role of autophagy in esophageal epithelial homeostasis. METHODS: We generated tamoxifen-inducible, squamous epithelial-specific Atg7 (autophagy related 7) conditional knockout mice to evaluate effects on esophageal homeostasis and response to the carcinogen 4-nitroquinoline 1-oxide (4NQO) using histologic and biochemical analyses. We fluorescence-activated cell sorted esophageal basal cells based on fluorescence of the autophagic vesicle (AV)-identifying dye Cyto-ID and then subjected these cells to transmission electron microscopy, image flow cytometry, three-dimensional organoid assays, RNA sequencing, and cell cycle analysis. Three-dimensional organoids were subjected to passaging, single-cell RNA sequencing, cell cycle analysis, and immunostaining. RESULTS: Genetic autophagy inhibition in squamous epithelium resulted in increased proliferation of esophageal basal cells under homeostatic conditions and also was associated with significant weight loss in mice treated with 4NQO that further displayed perturbed epithelial tissue architecture. Esophageal basal cells with high AV level (Cyto-IDHigh) displayed limited organoid formation capability on initial plating but passaged more efficiently than their counterparts with low AV level (Cyto-IDLow). RNA sequencing suggested increased autophagy in Cyto-IDHigh esophageal basal cells along with decreased cell cycle progression, the latter of which was confirmed by cell cycle analysis. Single-cell RNA sequencing of three-dimensional organoids generated by Cyto-IDLow and Cyto-IDHigh cells identified expansion of 3 cell populations and enrichment of G2/M-associated genes in the Cyto-IDHigh group. Ki67 expression was also increased in organoids generated by Cyto-IDHigh cells, including in basal cells localized beyond the outermost cell layer. CONCLUSIONS: Autophagy contributes to maintenance of the esophageal proliferation-differentiation gradient. Esophageal basal cells with high AV level exhibit limited proliferation and generate three-dimensional organoids with enhanced self-renewal capacity.


Subject(s)
Autophagy , Cell Proliferation , Homeostasis , Mice, Knockout , Organoids , Animals , Mice , Organoids/metabolism , Esophagus/pathology , Esophagus/cytology , Esophagus/metabolism , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Autophagy-Related Protein 7/metabolism , Autophagy-Related Protein 7/genetics , 4-Nitroquinoline-1-oxide , Cell Self Renewal , Esophageal Mucosa/pathology , Esophageal Mucosa/metabolism , Esophageal Mucosa/cytology , Single-Cell Analysis
7.
Histochem Cell Biol ; 157(2): 217-238, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34984523

ABSTRACT

The microanatomical features of the oesophageal gastric tract in tetrapod representatives and their function, especially those related to the mucosal layer, have not yet been fully investigated. The mucosal layer cells and their function in the oesophageal gastric tract differ structurally and functionally in tetrapod representatives based on interspecies difference and the type of food and feeding habits. The present study was, therefore, postulated to compare the mucosal microanatomical structure and histochemical biodistribution of different mucin types in oesophageal gastric tract tissues of four tetrapod species. A representative of each tetrapod class was selected, as follows: the Egyptian toad Bufo regularis, the lizard Trachylepis quinquetaeniata, the domestic pigeon Columba livia domestica and the albino mouse Mus musculus for Amphibia, Reptilia, Aves and Mammalia, respectively. Microanatomically, in lower tetrapods (toad and lizard), the mucosal layer of the oesophagus was composed of simple ciliated columnar epithelium with goblet cells, whereas in higher tetrapods (pigeon and mouse) it was composed of stratified squamous epithelium, with non-keratinised epithelium in the pigeon but keratinised epithelium in the mouse. However, the gastric mucosal layer of the stomach in lower tetrapods consists of simple columnar epithelium and gastric glands. Similarly, the mucosa of the pigeon's proventriculus consists of simple columnar epithelium with proventricular glands opened into the lumen, whereas mouse mucosa consists of simple columnar epithelium which folds and forms gastric glands with gastric pits having a variety of cell types. Histochemically, the neutral mucin profile biodistribution in the oesophagus mucosal layer was variable. It was strongly positive in the toad and lizard, but was weak in the pigeon and completely negative in the mouse. In contrast it was strongly positive in the gastric mucosa of the toad, lizard and pigeon, but was weak in the mouse's gastric mucosa. On the other hand, the signals of carboxylated and sulfated mucins were found to be different. They were strong in the mucosa of the lizard oesophagus. In contrast, the carboxylated mucins in the gastric mucosa were positive in all representatives except the mouse. The sulfated mucins were, however, seen localised in the mucosal layer cells of the lizard and pigeon only. The study revealed that the microanatomical structures and functions as well as mucin distribution profiles in the oesophageal gastric tract are in line with interspecies difference and the type of food and feeding habits. However, this may need further investigations including more tetrapod representatives.


Subject(s)
Esophagus/chemistry , Gastric Mucosa/chemistry , Mucins/metabolism , Animals , Bufonidae , Columbidae , Esophagus/cytology , Esophagus/metabolism , Gastric Mucosa/cytology , Gastric Mucosa/metabolism , Lizards , Mice , Tissue Distribution
8.
Am J Physiol Cell Physiol ; 321(5): C798-C811, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34524930

ABSTRACT

Altered esophageal ion transport mechanisms play a key role in inflammatory and cancerous diseases of the esophagus, but epithelial ion processes have been less studied in the esophagus because of the lack of a suitable experimental model. In this study, we generated three-dimensional (3D) esophageal organoids (EOs) from two different mouse strains and characterized the ion transport processes of the EOs. EOs form a cell-filled structure with a diameter of 250-300 µm and were generated from epithelial stem cells as shown by FACS analysis. Using conventional PCR and immunostaining, the presence of Slc26a6 Cl-/HCO3- anion exchanger (AE), Na+/H+ exchanger (NHE), Na+/HCO3- cotransporter (NBC), cystic fibrosis transmembrane conductance regulator (CFTR), and anoctamin 1 Cl- channels was detected in EOs. Microfluorimetric techniques revealed high NHE, AE, and NBC activities, whereas that of CFTR was relatively low. In addition, inhibition of CFTR led to functional interactions between the major acid-base transporters and CFTR. We conclude that EOs provide a relevant and suitable model system for studying the ion transport mechanisms of esophageal epithelial cells, and they can be also used as preclinical tools to assess the effectiveness of novel therapeutic compounds in esophageal diseases associated with altered ion transport processes.


Subject(s)
Epithelial Cells/metabolism , Esophagus/metabolism , Membrane Transport Proteins/metabolism , Organoids/metabolism , Stem Cells/metabolism , Animals , Anoctamin-1/genetics , Anoctamin-1/metabolism , Antiporters/genetics , Antiporters/metabolism , Cell Culture Techniques , Cells, Cultured , Chloride-Bicarbonate Antiporters/genetics , Chloride-Bicarbonate Antiporters/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Esophagus/cytology , Female , Ion Transport , Male , Membrane Transport Proteins/genetics , Mice, Inbred C57BL , Organoids/cytology , Sodium-Bicarbonate Symporters/genetics , Sodium-Bicarbonate Symporters/metabolism , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism , Sulfate Transporters/genetics , Sulfate Transporters/metabolism
9.
Science ; 373(6556): 760-767, 2021 08 13.
Article in English | MEDLINE | ID: mdl-34385390

ABSTRACT

The origin of human metaplastic states and their propensity for cancer is poorly understood. Barrett's esophagus is a common metaplastic condition that increases the risk for esophageal adenocarcinoma, and its cellular origin is enigmatic. To address this, we harvested tissues spanning the gastroesophageal junction from healthy and diseased donors, including isolation of esophageal submucosal glands. A combination of single-cell transcriptomic profiling, in silico lineage tracing from methylation, open chromatin and somatic mutation analyses, and functional studies in organoid models showed that Barrett's esophagus originates from gastric cardia through c-MYC and HNF4A-driven transcriptional programs. Furthermore, our data indicate that esophageal adenocarcinoma likely arises from undifferentiated Barrett's esophagus cell types even in the absence of a pathologically identifiable metaplastic precursor, illuminating early detection strategies.


Subject(s)
Adenocarcinoma/pathology , Barrett Esophagus/pathology , Cardia/cytology , Esophageal Neoplasms/pathology , Esophagus/pathology , Barrett Esophagus/genetics , Barrett Esophagus/metabolism , Cardia/chemistry , Cell Differentiation , Cell Lineage , Cell Transformation, Neoplastic , Epigenesis, Genetic , Epithelial Cells/cytology , Epithelial Cells/metabolism , Esophagus/cytology , Esophagus/metabolism , Exocrine Glands/chemistry , Exocrine Glands/cytology , Hepatocyte Nuclear Factor 4/metabolism , Humans , Keratin-7/analysis , Metaplasia , Phenotype , Proto-Oncogene Proteins c-myc/metabolism , RNA-Seq , Single-Cell Analysis , Transcription, Genetic , Transcriptome
10.
Biomed Mater ; 16(5)2021 08 27.
Article in English | MEDLINE | ID: mdl-34384057

ABSTRACT

Perfusion-decellularization was an interesting technique to generate a natural extracellular matrix (ECM) with the complete three-dimensional anatomical structure and vascular system. In this study, the esophageal ECM (E-ECM) scaffold was successfully constructed by perfusion-decellularized technique through the vascular system for the first time. And the physicochemical and biological properties of the E-ECM scaffolds were evaluated. The bone marrow mesenchymal stem cells (BMSCs) were induced to differentiate into myocytesin vitro. E-ECM scaffolds reseeded with myocytes were implanted into the greater omenta to obtain recellular esophageal ECM (RE-ECM), a tissue-engineered esophagus. The results showed that the cells of the esophagi were completely and uniformly removed after perfusion. E-ECM scaffolds retained the original four-layer organizational structure and vascular system with excellent biocompatibility. And the E-ECM scaffolds had no significant difference in mechanical properties comparing with fresh esophagi,p> 0.05. Immunocytochemistry showed positive expression ofα-sarcomeric actin, suggesting that BMSCs had successfully differentiated into myocytes. Most importantly, we found that in the RE-ECM muscularis, the myocytes regenerated linearly and continuously and migrated to the deep, and the tissue vascularization was obvious. The cell survival rates at 1 week and 2 weeks were 98.5 ± 3.0% and 96.4 ± 4.6%, respectively. It was demonstrated that myocytes maintained the ability for proliferation and differentiation for at least 2 weeks, and the cell activity was satisfactory in the RE-ECM. It follows that the tissue-engineered esophagus based on perfusion-decellularized technique and mesenchymal stem cells has great potential in esophageal repair. It is proposed as a promising alternative for reconstruction of esophageal defects in the future.


Subject(s)
Decellularized Extracellular Matrix/chemistry , Esophagus , Mesenchymal Stem Cells/cytology , Tissue Engineering/methods , Animals , Cells, Cultured , Esophagus/chemistry , Esophagus/cytology , Esophagus/metabolism , Male , Perfusion , Rabbits
11.
Medicine (Baltimore) ; 100(21): e25993, 2021 May 28.
Article in English | MEDLINE | ID: mdl-34032716

ABSTRACT

ABSTRACT: Guanine nucleotide-binding protein-like-3-like (GNL3L) is required for processing ribosomal pre-rRNA and cell proliferation and is upregulated in many types of cancer. This study is aimed to investigate the clinical significance of GNL3L in esophageal cancer. The mRNA and protein expression levels of GNL3L were determined by using quantitative real-time polymerase chain reaction and immunohistochemistry, respectively. GNL3L was localized in both cytoplasm and nucleus. The expression levels of GNL3L in esophageal cancer tissues were significantly higher than those in adjacent nonmalignant tissues. High GNL3L expression was associated with pathologic type and poor differentiation. Patients with high GNL3L expression had shorter overall survival (OS) than those with low GNL3L expression. Multivariate Cox regression analysis revealed that GNL3L expression was an independently predictive factor for the OS of patient with esophageal cancer. The Gene Expression Profiling Interactive Analysis (GEPIA) databases also showed that GNL3L was upregulated in esophageal cancer, which was closely associated with an unfavorable prognosis of patients with esophageal cancer. Taken together, our findings suggest that GNL3L is upregulated in esophageal cancer, which is linked to the progression of the disease. As a result, GNL3L could be used as a biomarker for esophageal cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Esophageal Neoplasms/mortality , Esophageal Squamous Cell Carcinoma/mortality , GTP-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/analysis , Cell Nucleus/pathology , Cell Proliferation , Chemotherapy, Adjuvant/methods , Cytoplasm/pathology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophageal Neoplasms/therapy , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/therapy , Esophagectomy , Esophagus/cytology , Esophagus/pathology , Esophagus/surgery , Female , GTP-Binding Proteins/analysis , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Male , Middle Aged , Nuclear Proteins/analysis , Prognosis , Up-Regulation
12.
Cells ; 10(5)2021 04 29.
Article in English | MEDLINE | ID: mdl-33946915

ABSTRACT

Many decellularized extracellular matrix-derived whole organs have been widely used in studies of tissue engineering and cancer models. However, decellularizing porcine esophagus to obtain decellularized esophageal matrix (DEM) for potential biomedical applications has not been widely investigated. In this study a modified decellularization protocol was employed to prepare a porcine esophageal DEM for the study of cancer cell growth. The cellular removal and retention of matrix components in the porcine DEM were fully characterized. The microstructure of the DEM was observed using scanning electronic microscopy. Human esophageal squamous cell carcinoma (ESCC) and human primary esophageal fibroblast cells (FBCs) were seeded in the DEM to observe their growth. Results show that the decellularization process did not cause significant loss of mechanical properties and that blood ducts and lymphatic vessels in the submucosa layer were also preserved. ESCC and FBCs grew on the DEM well and the matrix did not show any toxicity to cells. When FBS and ESCC were cocultured on the matrix, they secreted more periostin, a protein that supports cell adhesion on matrix. This study shows that the modified decellularization protocol can effectively remove the cell materials and maintain the microstructure of the porcine esophageal matrix, which has the potential application of studying cell growth and migration for esophageal cancer models.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Esophageal Neoplasms/metabolism , Esophagus/cytology , Extracellular Matrix/chemistry , Tissue Scaffolds/chemistry , Animals , Carcinoma, Squamous Cell/pathology , Cell Adhesion , Cell Line, Tumor , Cells, Cultured , Coculture Techniques/methods , Elastic Modulus , Esophageal Neoplasms/pathology , Esophagus/metabolism , Fibroblasts/metabolism , Fibroblasts/physiology , Mice , Swine
13.
PLoS One ; 16(4): e0248994, 2021.
Article in English | MEDLINE | ID: mdl-33798212

ABSTRACT

Swallow is a complex behavior that consists of three coordinated phases: oral, pharyngeal, and esophageal. Esophageal distension (EDist) has been shown to elicit pharyngeal swallow, but the physiologic characteristics of EDist-induced pharyngeal swallow have not been specifically described. We examined the effect of rapid EDist on oropharyngeal swallow, with and without an oral water stimulus, in spontaneously breathing, sodium pentobarbital anesthetized cats (n = 5). Electromyograms (EMGs) of activity of 8 muscles were used to evaluate swallow: mylohyoid (MyHy), geniohyoid (GeHy), thyrohyoid (ThHy), thyropharyngeus (ThPh), thyroarytenoid (ThAr), cricopharyngeus (upper esophageal sphincter: UES), parasternal (PS), and costal diaphragm (Dia). Swallow was defined as quiescence of the UES with overlapping upper airway activity, and it was analyzed across three stimulus conditions: 1) oropharyngeal water infusion only, 2) rapid esophageal distension (EDist) only, and 3) combined stimuli. Results show a significant effect of stimulus condition on swallow EMG amplitude of the mylohyoid, geniohyoid, thyroarytenoid, diaphragm, and UES muscles. Collectively, we found that, compared to rapid cervical esophageal distension alone, the stimulus condition of rapid distension combined with water infusion is correlated with increased laryngeal adductor and diaphragm swallow-related EMG activity (schluckatmung), and post-swallow UES recruitment. We hypothesize that these effects of upper esophageal distension activate the brainstem swallow network, and function to protect the airway through initiation and/or modulation of a pharyngeal swallow response.


Subject(s)
Deglutition , Esophagus/physiology , Inhalation , Mechanoreceptors/physiology , Pharynx/physiology , Animals , Cats , Esophagus/cytology , Male , Muscle Contraction
14.
Sci Rep ; 11(1): 7257, 2021 03 31.
Article in English | MEDLINE | ID: mdl-33790338

ABSTRACT

The current study focused on the histogenesis of the esophagus in quail embryos. Formation of the gut tube occurred on the 4th day of incubation. Development of the muscular layers occurred in a sequential manner; the inner circular layer on the 7th day, the outer longitudinal layer on the 8th day and the muscularis mucosae on the 9th day. Glandular development began on the 13th day of incubation. The epithelium was pseudostratified columnar that consisted of mucous cells, dendritic cells, and keratinocyte precursors. Epithelial stratification occurred on the 15th day of incubation. We used Mallory trichrome, Weigert-Van Gieson, and Gomori silver stains to visualize fibrous components. Scanned samples showed formation of endoderm and mesoderm on the 5th day of incubation. A layer of myoblasts developed on the 8th day of incubation. Formation of mucosal folds, which contained glandular openings, occurred on the 14th to 17th days of incubation. On the 5th to 8th days of incubation, CD34 and vascular endothelial growth factor (VEGF) positive-mesodermal cells, and telocytes (TCs) were detected. On the 15th day of incubation, CD34 and VEGF positive-telocytes, and fibroblasts, were identified. The current study described the correlations between functional morphology and evolutionary biology.


Subject(s)
Embryo, Nonmammalian , Esophagus , Organogenesis/physiology , Quail/embryology , Animals , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/embryology , Epithelium/embryology , Esophagus/cytology , Esophagus/embryology
15.
Cell Rep ; 34(10): 108819, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33691112

ABSTRACT

The upper gastrointestinal tract, consisting of the esophagus, stomach, and duodenum, controls food transport, digestion, nutrient uptake, and hormone production. By single-cell analysis of healthy epithelia of these human organs, we molecularly define their distinct cell types. We identify a quiescent COL17A1high KRT15high stem/progenitor cell population in the most basal cell layer of the esophagus and detect substantial gene expression differences between identical cell types of the human and mouse stomach. Selective expression of BEST4, CFTR, guanylin, and uroguanylin identifies a rare duodenal cell type, referred to as BCHE cell, which likely mediates high-volume fluid secretion because of continual activation of the CFTR channel by guanylin/uroguanylin-mediated autocrine signaling. Serotonin-producing enterochromaffin cells in the antral stomach significantly differ in gene expression from duodenal enterochromaffin cells. We, furthermore, discover that the histamine-producing enterochromaffin-like cells in the oxyntic stomach express the luteinizing hormone, yet another member of the enteroendocrine hormone family.


Subject(s)
Duodenum/cytology , Esophagus/cytology , Stomach/cytology , Upper Gastrointestinal Tract/cytology , Animals , Autoantigens/genetics , Autoantigens/metabolism , Bestrophins/genetics , Bestrophins/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Duodenum/metabolism , Duodenum/pathology , Esophagus/metabolism , Esophagus/pathology , Gene Expression , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Keratin-15/genetics , Keratin-15/metabolism , Luteinizing Hormone/genetics , Luteinizing Hormone/metabolism , Mice , Mice, Inbred C57BL , Non-Fibrillar Collagens/genetics , Non-Fibrillar Collagens/metabolism , Single-Cell Analysis , Stem Cells/cytology , Stem Cells/metabolism , Stomach/metabolism , Stomach/pathology , Upper Gastrointestinal Tract/metabolism , Upper Gastrointestinal Tract/pathology , Collagen Type XVII
16.
Sci Rep ; 11(1): 7086, 2021 03 29.
Article in English | MEDLINE | ID: mdl-33782465

ABSTRACT

Constructive remodeling of focal esophageal defects with biodegradable acellular grafts relies on the ability of host progenitor cell populations to repopulate implant regions and facilitate growth of de novo functional tissue. Intrinsic molecular mechanisms governing esophageal repair processes following biomaterial-based, surgical reconstruction is largely unknown. In the present study, we utilized mass spectrometry-based quantitative proteomics and in silico pathway evaluations to identify signaling cascades which were significantly activated during neoepithelial formation in a Sprague Dawley rat model of onlay esophagoplasty with acellular silk fibroin scaffolds. Pharmacologic inhibitor and rescue experiments revealed that epithelialization of neotissues is significantly dependent in part on pro-survival stimuli capable of suppressing caspase activity in epithelial progenitors via activation of hepatocyte growth factor receptor (c-MET), tropomyosin receptor kinase A (TrkA), phosphoinositide 3-kinase (PI3K), and protein kinase B (Akt) signaling mechanisms. These data highlight the molecular machinery involved in esophageal epithelial regeneration following surgical repair with acellular implants.


Subject(s)
Esophagus/cytology , Fibroins/administration & dosage , Plastic Surgery Procedures/methods , Animals , Epithelial Cells/cytology , Esophagus/surgery , Humans , Rats, Sprague-Dawley , Regeneration , Signal Transduction
17.
Development ; 148(6)2021 03 29.
Article in English | MEDLINE | ID: mdl-33782045

ABSTRACT

The esophagus is derived from the anterior portion of the foregut endoderm, which also gives rise to the respiratory system. As it develops, the esophageal lining is transformed from a simple columnar epithelium into a stratified squamous cell layer, accompanied by the replacement of unspecified mesenchyme with layers of muscle cells. Studies in animal models have provided significant insights into the roles of various signaling pathways in esophageal development. More recent studies using human pluripotent stem cells (hPSCs) further demonstrate that some of these signaling pathways are conserved in human esophageal development. In addition, a combination of mouse genetics and hPSC differentiation approaches have uncovered new players that control esophageal morphogenesis. In this Review, we summarize these new findings and discuss how the esophagus is established and matures throughout different stages, including its initial specification, respiratory-esophageal separation, epithelial morphogenesis and maintenance. We also discuss esophageal muscular development and enteric nervous system innervation, which are essential for esophageal structure and function.


Subject(s)
Esophagus/cytology , Pluripotent Stem Cells/metabolism , Animals , Cell Differentiation , Endoderm/cytology , Endoderm/metabolism , Enteric Nervous System/cytology , Enteric Nervous System/growth & development , Enteric Nervous System/metabolism , Esophagus/metabolism , Hedgehog Proteins/metabolism , Humans , Pluripotent Stem Cells/cytology , Signal Transduction , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism
18.
Nat Rev Cancer ; 21(4): 239-256, 2021 04.
Article in English | MEDLINE | ID: mdl-33627798

ABSTRACT

Cancer is a clonal disorder derived from a single ancestor cell and its progenies that are positively selected by acquisition of 'driver mutations'. However, the evolution of positively selected clones does not necessarily imply the presence of cancer. On the contrary, it has become clear that expansion of these clones in phenotypically normal or non-cancer tissues is commonly seen in association with ageing and/or in response to environmental insults and chronic inflammation. Recent studies have reported expansion of clones harbouring mutations in cancer driver genes in the blood, skin, oesophagus, bronchus, liver, endometrium and bladder, where the expansion could be so extensive that tissues undergo remodelling of an almost entire tissue. The presence of common cancer driver mutations in normal tissues suggests a strong link to cancer development, providing an opportunity to understand early carcinogenic processes. Nevertheless, some driver mutations are unique to normal tissues or have a mutation frequency that is much higher in normal tissue than in cancer, indicating that the respective clones may not necessarily be destined for evolution to cancer but even negatively selected for carcinogenesis depending on the mutated gene. Moreover, tissues that are remodelled by genetically altered clones might define functionalities of aged tissues or modified inflammatory processes. In this Review, we provide an overview of major findings on clonal expansion in phenotypically normal or non-cancer tissues and discuss their biological significance not only in cancer development but also in ageing and inflammatory diseases.


Subject(s)
Aging/genetics , Cell Proliferation/genetics , Clonal Evolution , Clone Cells/cytology , Neoplasms/genetics , Aging/pathology , Anemia, Aplastic/genetics , Anemia, Aplastic/pathology , Barrett Esophagus/genetics , Barrett Esophagus/pathology , Bronchi/cytology , Bronchi/metabolism , Carcinogenesis , Clonal Hematopoiesis/genetics , Colitis, Ulcerative/genetics , Colitis, Ulcerative/pathology , Colon/cytology , Colon/metabolism , Endometrium/cytology , Endometrium/metabolism , Esophagus/cytology , Esophagus/metabolism , Female , Gastric Mucosa/metabolism , Humans , Inflammation/genetics , Inflammation/pathology , Liver/cytology , Liver/metabolism , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Male , Metaplasia , Mutation , Neoplasms/pathology , Oncogenes/genetics , Skin/cytology , Skin/metabolism , Stomach/pathology , Urothelium/cytology , Urothelium/metabolism
19.
Lab Invest ; 101(6): 745-759, 2021 06.
Article in English | MEDLINE | ID: mdl-33495575

ABSTRACT

Multiple genome-wide association studies (GWAS) have linked Forkhead Box F1 (FOXF1) to Barrett's esophagus (BE). Understanding whether FOXF1 is involved in initiation of Barrett's metaplasia could allow FOXF1 to be used for risk stratification and for therapy. Two-dimensional cell cultures and three-dimensional organoid cultures and well-annotated human biopsies were used to determine the role of FOXF1 in BE pathogenesis. Multiple established esophageal squamous and BE cell lines were tested in gain- and loss-of-function studies. Initiation of a BE-like metaplastic change was evaluated by measuring characteristic cytokeratins and global gene expression profiling and by culturing organoids. Epithelial-mesenchymal transition (EMT) was evaluated by immunostaining for E-cadherin, vimentin and Snail, and by cell motility assay. Columnar esophageal epithelium of BE patients exhibited higher expression of FOXF1 compared to normal squamous esophageal epithelium of GERD patients (P < 0.001). Acidic bile salts induced nuclear FOXF1 in esophageal squamous cells. FOXF1 overexpression in normal esophageal squamous cells: (a) increased columnar cytokeratins and decreased squamous cytokeratins, (b) converted squamous organoids to glandular organoids, and (c) switched global gene profiles to resemble that of human BE epithelium (P = 2.1685e - 06 for upregulated genes and P = 8.3378e - 09 for downregulated genes). FOXF1 inhibition in BE cell lines led to loss of BE differentiation markers, CK7, and mucin 2. Also, FOXF1 induced EMT and promoted cell motility in normal esophageal squamous epithelial cells. FOXF1-induced genes mapped to pathways such as Cancer, Cellular Assembly and Organization, DNA Replication, Recombination, and Repair. In conclusion, FOXF1 promotes a BE-like columnar phenotype and cell motility in esophageal squamous epithelial cells, which may have a critical role in BE development. FOXF1 should be studied further as a biomarker for BE and as a target for BE treatment.


Subject(s)
Barrett Esophagus/etiology , Epithelial-Mesenchymal Transition , Forkhead Transcription Factors/metabolism , Aged , Barrett Esophagus/metabolism , Cell Line, Tumor , Epithelial Cells/metabolism , Esophagus/cytology , Esophagus/metabolism , Humans , Middle Aged
20.
J Morphol ; 282(3): 355-367, 2021 03.
Article in English | MEDLINE | ID: mdl-33314288

ABSTRACT

The esophageal pouches of Chaetopleura angulata and Acanthochitona fascicularis were investigated using light and transmission electron microscopy. These pouches linked to the posterior region of the esophagus are known as sugar glands as they contain a fluid rich in polysaccharide digesting enzymes. They are the second largest glands in the digestive system of chitons, just after the digestive gland. In both species, the pouches contain a dense array of finger-shaped villi. The villi epithelium includes absorptive cells, basophilic secretory cells, mucus-secreting cells, and basal cells. Some absorptive cells were bordered by a dense cover of long microvilli, whereas other absorptive cells had short and sparse microvilli. Absorptive cells contain several lysosomes, mitochondria, peroxisomes, a few small Golgi stacks, some lipid droplets, and large amounts of glycogen. The basophilic secretory cells are characterized by the presence of many electron-dense vesicles, with a glycoprotein content, a large number of rough endoplasmic reticulum cisternae, and a highly developed Golgi apparatus. Mucus-secreting cells are characterized by large vesicles containing acid polysaccharides and wide Golgi stacks. Basal cells that were found at the base of the epithelium in contact with the basal lamina exhibit histological and ultrastructural features of enteroendocrine cells. We suggest that these glandular pouches are involved in extracellular and intracellular digestion, and accumulate lipid and glycogen reserves.


Subject(s)
Animal Structures/anatomy & histology , Esophagus/anatomy & histology , Polyplacophora/anatomy & histology , Animal Structures/cytology , Animal Structures/ultrastructure , Animals , Esophagus/cytology , Esophagus/ultrastructure , Polyplacophora/cytology , Polyplacophora/ultrastructure , Secretory Vesicles/metabolism , Staining and Labeling
SELECTION OF CITATIONS
SEARCH DETAIL