Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 940
Filtrar
1.
Histochem Cell Biol ; 157(2): 217-238, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34984523

RESUMO

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.


Assuntos
Esôfago/química , Mucosa Gástrica/química , Mucinas/metabolismo , Animais , Bufonidae , Columbidae , Esôfago/citologia , Esôfago/metabolismo , Mucosa Gástrica/citologia , Mucosa Gástrica/metabolismo , Lagartos , Camundongos , Distribuição Tecidual
2.
Am J Physiol Cell Physiol ; 321(5): C798-C811, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34524930

RESUMO

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.


Assuntos
Células Epiteliais/metabolismo , Esôfago/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Organoides/metabolismo , Células-Tronco/metabolismo , Animais , Anoctamina-1/genética , Anoctamina-1/metabolismo , Antiporters/genética , Antiporters/metabolismo , Técnicas de Cultura de Células , Células Cultivadas , Antiportadores de Cloreto-Bicarbonato/genética , Antiportadores de Cloreto-Bicarbonato/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Esôfago/citologia , Feminino , Transporte de Íons , Masculino , Proteínas de Membrana Transportadoras/genética , Camundongos Endogâmicos C57BL , Organoides/citologia , Simportadores de Sódio-Bicarbonato/genética , Simportadores de Sódio-Bicarbonato/metabolismo , Trocadores de Sódio-Hidrogênio/genética , Trocadores de Sódio-Hidrogênio/metabolismo , Transportadores de Sulfato/genética , Transportadores de Sulfato/metabolismo
3.
Science ; 373(6556): 760-767, 2021 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-34385390

RESUMO

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.


Assuntos
Adenocarcinoma/patologia , Esôfago de Barrett/patologia , Cárdia/citologia , Neoplasias Esofágicas/patologia , Esôfago/patologia , Esôfago de Barrett/genética , Esôfago de Barrett/metabolismo , Cárdia/química , Diferenciação Celular , Linhagem da Célula , Transformação Celular Neoplásica , Epigênese Genética , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Esôfago/citologia , Esôfago/metabolismo , Glândulas Exócrinas/química , Glândulas Exócrinas/citologia , Fator 4 Nuclear de Hepatócito/metabolismo , Humanos , Queratina-7/análise , Metaplasia , Fenótipo , Proteínas Proto-Oncogênicas c-myc/metabolismo , RNA-Seq , Análise de Célula Única , Transcrição Gênica , Transcriptoma
4.
Biomed Mater ; 16(5)2021 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-34384057

RESUMO

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.


Assuntos
Matriz Extracelular Descelularizada/química , Esôfago , Células-Tronco Mesenquimais/citologia , Engenharia Tecidual/métodos , Animais , Células Cultivadas , Esôfago/química , Esôfago/citologia , Esôfago/metabolismo , Masculino , Perfusão , Coelhos
5.
Medicine (Baltimore) ; 100(21): e25993, 2021 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-34032716

RESUMO

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.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias Esofágicas/mortalidade , Carcinoma de Células Escamosas do Esôfago/mortalidade , Proteínas de Ligação ao GTP/metabolismo , Proteínas Nucleares/metabolismo , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Biomarcadores Tumorais/análise , Núcleo Celular/patologia , Proliferação de Células , Quimioterapia Adjuvante/métodos , Citoplasma/patologia , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Neoplasias Esofágicas/terapia , Carcinoma de Células Escamosas do Esôfago/genética , Carcinoma de Células Escamosas do Esôfago/patologia , Carcinoma de Células Escamosas do Esôfago/terapia , Esofagectomia , Esôfago/citologia , Esôfago/patologia , Esôfago/cirurgia , Feminino , Proteínas de Ligação ao GTP/análise , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Proteínas Nucleares/análise , Prognóstico , Regulação para Cima
6.
Cells ; 10(5)2021 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-33946915

RESUMO

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.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Neoplasias Esofágicas/metabolismo , Esôfago/citologia , Matriz Extracelular/química , Tecidos Suporte/química , Animais , Carcinoma de Células Escamosas/patologia , Adesão Celular , Linhagem Celular Tumoral , Células Cultivadas , Técnicas de Cocultura/métodos , Módulo de Elasticidade , Neoplasias Esofágicas/patologia , Esôfago/metabolismo , Fibroblastos/metabolismo , Fibroblastos/fisiologia , Camundongos , Suínos
7.
Sci Rep ; 11(1): 7257, 2021 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-33790338

RESUMO

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.


Assuntos
Embrião não Mamífero , Esôfago , Organogênese/fisiologia , Codorniz/embriologia , Animais , Embrião não Mamífero/citologia , Embrião não Mamífero/embriologia , Epitélio/embriologia , Esôfago/citologia , Esôfago/embriologia
8.
PLoS One ; 16(4): e0248994, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33798212

RESUMO

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.


Assuntos
Deglutição , Esôfago/fisiologia , Inalação , Mecanorreceptores/fisiologia , Faringe/fisiologia , Animais , Gatos , Esôfago/citologia , Masculino , Contração Muscular
9.
Sci Rep ; 11(1): 7086, 2021 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-33782465

RESUMO

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.


Assuntos
Esôfago/citologia , Fibroínas/administração & dosagem , Procedimentos de Cirurgia Plástica/métodos , Animais , Células Epiteliais/citologia , Esôfago/cirurgia , Humanos , Ratos Sprague-Dawley , Regeneração , Transdução de Sinais
10.
Cell Rep ; 34(10): 108819, 2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33691112

RESUMO

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.


Assuntos
Duodeno/citologia , Esôfago/citologia , Estômago/citologia , Trato Gastrointestinal Superior/citologia , Animais , Autoantígenos/genética , Autoantígenos/metabolismo , Bestrofinas/genética , Bestrofinas/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Duodeno/metabolismo , Duodeno/patologia , Esôfago/metabolismo , Esôfago/patologia , Expressão Gênica , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Queratina-15/genética , Queratina-15/metabolismo , Hormônio Luteinizante/genética , Hormônio Luteinizante/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Colágenos não Fibrilares/genética , Colágenos não Fibrilares/metabolismo , Análise de Célula Única , Células-Tronco/citologia , Células-Tronco/metabolismo , Estômago/metabolismo , Estômago/patologia , Trato Gastrointestinal Superior/metabolismo , Trato Gastrointestinal Superior/patologia
11.
Development ; 148(6)2021 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-33782045

RESUMO

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.


Assuntos
Esôfago/citologia , Células-Tronco Pluripotentes/metabolismo , Animais , Diferenciação Celular , Endoderma/citologia , Endoderma/metabolismo , Sistema Nervoso Entérico/citologia , Sistema Nervoso Entérico/crescimento & desenvolvimento , Sistema Nervoso Entérico/metabolismo , Esôfago/metabolismo , Proteínas Hedgehog/metabolismo , Humanos , Células-Tronco Pluripotentes/citologia , Transdução de Sinais , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo
12.
Nat Rev Cancer ; 21(4): 239-256, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33627798

RESUMO

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.


Assuntos
Envelhecimento/genética , Proliferação de Células/genética , Evolução Clonal , Células Clonais/citologia , Neoplasias/genética , Envelhecimento/patologia , Anemia Aplástica/genética , Anemia Aplástica/patologia , Esôfago de Barrett/genética , Esôfago de Barrett/patologia , Brônquios/citologia , Brônquios/metabolismo , Carcinogênese , Hematopoiese Clonal/genética , Colite Ulcerativa/genética , Colite Ulcerativa/patologia , Colo/citologia , Colo/metabolismo , Endométrio/citologia , Endométrio/metabolismo , Esôfago/citologia , Esôfago/metabolismo , Feminino , Mucosa Gástrica/metabolismo , Humanos , Inflamação/genética , Inflamação/patologia , Fígado/citologia , Fígado/metabolismo , Cirrose Hepática/genética , Cirrose Hepática/patologia , Masculino , Metaplasia , Mutação , Neoplasias/patologia , Oncogenes/genética , Pele/citologia , Pele/metabolismo , Estômago/patologia , Urotélio/citologia , Urotélio/metabolismo
13.
Lab Invest ; 101(6): 745-759, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33495575

RESUMO

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.


Assuntos
Esôfago de Barrett/etiologia , Transição Epitelial-Mesenquimal , Fatores de Transcrição Forkhead/metabolismo , Idoso , Esôfago de Barrett/metabolismo , Linhagem Celular Tumoral , Células Epiteliais/metabolismo , Esôfago/citologia , Esôfago/metabolismo , Humanos , Pessoa de Meia-Idade
14.
J Surg Res ; 259: 562-568, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33261858

RESUMO

BACKGROUND: Gastroesophageal reflux and Barrett's esophagus are significant risk factors for the development of esophageal adenocarcinoma. Group IIa secretory phospholipase A2 (sPLA2) catalyzes the production of various proinflammatory metabolites and plays a critical role in promoting reflux-induced inflammatory changes within the distal esophagus. We hypothesized that inhibition of sPLA2 in human Barrett's cells would attenuate adhesion molecule expression via decreased activation of nuclear factor kappa B (NF-κB) and decrease cell proliferation, possibly mitigating the invasive potential of Barrett's esophagus. MATERIALS AND METHODS: Normal human esophageal epithelial cells (HET1A) and Barrett's cells (CPB) were assayed for baseline sPLA2 expression. CPB cells were treated with a specific inhibitor of sPLA2 followed by tumor necrosis factor-α. Protein expression was evaluated using immunoblotting. Cell proliferation was assessed using an MTS cell proliferation assay kit. Statistical analysis was performed using the Student's t-test or analysis of variance, where appropriate. RESULTS: CPB cells demonstrated higher baseline sPLA2 expression than HET1A cells (P = 0.0005). Treatment with 30 µM sPLA2 inhibitor significantly attenuated intercellular adhesion molecule-1 (P = 0.004) and vascular cell adhesion molecule-1 (P < 0.0001) expression as well as decreased NF-κB activation (P = 0.002). sPLA2 inhibition decreased cell proliferation in a dose-dependent manner (P < 0.001 for 15, 20, and 30 µM doses). CONCLUSIONS: sPLA2 inhibition in human Barrett's cells decreases cellular adhesive properties and NF-κB activation as well as decreases cell proliferation, signifying downregulation of the inflammatory response and possible attenuation of cellular malignant potential. These findings identify sPLA2 inhibition as a potential chemopreventive target for premalignant lesions of the esophagus.


Assuntos
Esôfago de Barrett/patologia , Esôfago/patologia , Fosfolipases A2 do Grupo II/antagonistas & inibidores , Ácidos Pentanoicos/farmacologia , Adenocarcinoma/patologia , Adenocarcinoma/prevenção & controle , Esôfago de Barrett/tratamento farmacológico , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Neoplasias Esofágicas/patologia , Neoplasias Esofágicas/prevenção & controle , Esôfago/citologia , Fosfolipases A2 do Grupo II/metabolismo , Humanos , Ácidos Pentanoicos/uso terapêutico
15.
J Morphol ; 282(3): 355-367, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33314288

RESUMO

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.


Assuntos
Estruturas Animais/anatomia & histologia , Esôfago/anatomia & histologia , Poliplacóforos/anatomia & histologia , Estruturas Animais/citologia , Estruturas Animais/ultraestrutura , Animais , Esôfago/citologia , Esôfago/ultraestrutura , Poliplacóforos/citologia , Poliplacóforos/ultraestrutura , Vesículas Secretórias/metabolismo , Coloração e Rotulagem
16.
Clin Immunol ; 222: 108600, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33197619

RESUMO

OBJECTIVES: To evaluate the occurrence, abundance, distribution, nature and clinical significance of multinucleated giant cell (MGC) in esophageal cancer. MATERIALS AND METHODS: MGCs were examined with conventional pathology, immunohistochemistry and immunofluorescence in 107 esophageal cancer tissues. The findings were correlated to pathological diagnosis and clinical behavior of the cancers. RESULTS: MGCs were identified in 31.7% (34/107) of the cases. MGCs were positive for CD11c, CD11b, CD32, CD16, HLA-DR and MMP9, and negative for CD163, CD206 and CD64 giving a molecular profile of proinflammatory M1 but not immunosuppressive M2. MGCs were significantly related to decreased lymph node metastasis (p = 0.011), low pTNM stage (p = 0.044), favorable survival (p = 0.04), squamous cell cancer type rather than other histopathological subtypes (p = 0.020) and associated to better differentiation (p = 0.063). CONCLUSIONS: MGCs belong to M1 macrophage and perform phagocytosis and scavenging of cancer cells that would benefit patients' survival and could serve as a prognostic marker.


Assuntos
Neoplasias Esofágicas/patologia , Esôfago/citologia , Células Gigantes/imunologia , Macrófagos/imunologia , Fagocitose/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma de Células Escamosas/patologia , China , Neoplasias Esofágicas/diagnóstico , Neoplasias Esofágicas/imunologia , Esôfago/imunologia , Esôfago/patologia , Feminino , Humanos , Metástase Linfática/patologia , Masculino , Pessoa de Meia-Idade , Prognóstico , Receptores de IgG/imunologia
17.
PLoS One ; 15(9): e0239625, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32966340

RESUMO

During alcohol consumption, the esophageal mucosa is directly exposed to high concentrations of ethanol (EtOH). We therefore investigated the response of normal human esophageal epithelial cell lines EPC1, EPC2 and EPC3 to acute EtOH exposure. While these cells were able to tolerate 2% EtOH for 8 h in both three-dimensional organoids and monolayer culture conditions, RNA sequencing suggested that EtOH induced mitochondrial dysfunction. With EtOH treatment, EPC1 and EPC2 cells also demonstrated decreased mitochondrial ATPB protein expression by immunofluorescence and swollen mitochondria lacking intact cristae by transmission electron microscopy. Mitochondrial membrane potential (ΔΨm) was decreased in a subset of EPC1 and EPC2 cells stained with ΔΨm-sensitive dye MitoTracker Deep Red. In EPC2, EtOH decreased ATP level while impairing mitochondrial respiration and electron transportation chain functions, as determined by ATP fluorometric assay, respirometry, and liquid chromatography-mass spectrometry. Additionally, EPC2 cells demonstrated enhanced oxidative stress by flow cytometry for mitochondrial superoxide (MitoSOX), which was antagonized by the mitochondria-specific antioxidant MitoCP. Concurrently, EPC1 and EPC2 cells underwent autophagy following EtOH exposure, as evidenced by flow cytometry for Cyto-ID, which detects autophagic vesicles, and immunoblots demonstrating induction of the lipidated and cleaved form of LC3B and downregulation of SQSTM1/p62. In EPC1 and EPC2, pharmacological inhibition of autophagy flux by chloroquine increased mitochondrial oxidative stress while decreasing cell viability. In EPC2, autophagy induction was coupled with phosphorylation of AMP activated protein kinase (AMPK), a cellular energy sensor responding to low ATP levels, and dephosphorylation of downstream substrates of mechanistic Target of Rapamycin Complex (mTORC)-1 signaling. Pharmacological AMPK activation by AICAR decreased EtOH-induced reduction of ΔΨm and ATP in EPC2. Taken together, acute EtOH exposure leads to mitochondrial dysfunction and oxidative stress in esophageal keratinocytes, where the AMPK-mTORC1 axis may serve as a regulatory mechanism to activate autophagy to provide cytoprotection against EtOH-induced cell injury.


Assuntos
Autofagia , Esôfago/citologia , Queratinócitos/metabolismo , Mitocôndrias/metabolismo , Estresse Oxidativo , Quinases Proteína-Quinases Ativadas por AMP , Animais , Linhagem Celular , Células Cultivadas , Etanol/farmacologia , Feminino , Queratinócitos/efeitos dos fármacos , Masculino , Potencial da Membrana Mitocondrial , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
18.
Methods Cell Biol ; 159: 1-22, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32586439

RESUMO

The human and murine esophagus have some substantial differences that limit the utility of mouse as a model to study human esophagus development and disease. Due to these limitations several recent reports describe the development of methods to generate human esophageal tissues via the directed differentiation of pluripotent stem cells. Methods for differentiation are based on knowledge of years of studying embryonic development of the esophagus in vertebrate animal models. Esophageal tissues derived from human pluripotent stem cells have been used to study both development and diseases affecting the esophagus. Here, we provide a detailed protocol for the directed differentiation of human pluripotent stem cells into human esophageal organoids and organotypic raft cultures, that are highly similar, morphologically and transcriptionally, to the human esophagus epithelium. We discuss limitations of the current esophageal models and the importance of engineering more complex tissue models with muscle and enteric nerves. Moving forward, these models might be utilized for the development of personalized treatments, as well as other therapeutic solutions.


Assuntos
Técnicas de Cultura de Células/métodos , Esôfago/citologia , Organoides/citologia , Células-Tronco Pluripotentes/citologia , Animais , Diferenciação Celular , Crioultramicrotomia , Fibroblastos/citologia , Imunofluorescência , Células-Tronco Embrionárias Humanas/citologia , Humanos , Queratinócitos/citologia , Camundongos , Fixação de Tecidos
19.
J Gastroenterol ; 55(8): 763-774, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32556644

RESUMO

BACKGROUND: The esophagus is known to be derived from the foregut. However, the mechanisms regulating this process remain unclear. In particular, the details of the human esophagus itself have been poorly researched. In this decade, studies using human induced pluripotent stem cells (hiPSCs) have proven powerful tools for clarifying the developmental biology of various human organs. Several studies using hiPSCs have demonstrated that retinoic acid (RA) signaling promotes the differentiation of foregut into tissues such as lung and pancreas. However, the effect of RA signaling on the differentiation of foregut into esophagus remains unclear. METHODS: We established a novel stepwise protocol with transwell culture and an air-liquid interface system for esophageal epithelial cell (EEC) differentiation from hiPSCs. We then evaluated the effect of all-trans retinoic acid (ATRA), which is a retinoic acid receptor (RAR)α, RARß and RARγ agonist, on the differentiation from the hiPSC-derived foregut. Finally, to identify which RAR subtype was involved in the differentiation, we used synthetic agonists and antagonists of RARα and RARγ, which are known to be expressed in esophagus. RESULTS: We successfully generated stratified layers of cells expressing EEC marker genes that were positive for lugol staining. The enhancing effect of ATRA on EEC differentiation was clearly demonstrated with quantitative reverse transcription polymerase chain reaction, immunohistology, lugol-staining and RNA sequencing analyses. RARγ agonist and antagonist enhanced and suppressed EEC differentiation, respectively. RARα agonist had no effect on the differentiation. CONCLUSION: We revealed that RARγ activation promotes the differentiation of hiPSCs-derived foregut into EECs.


Assuntos
Células Epiteliais/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Receptores do Ácido Retinoico/metabolismo , Tretinoína/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Células Epiteliais/efeitos dos fármacos , Esôfago/citologia , Esôfago/efeitos dos fármacos , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Camundongos , Receptores do Ácido Retinoico/efeitos dos fármacos , Receptor alfa de Ácido Retinoico/efeitos dos fármacos , Receptor alfa de Ácido Retinoico/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos
20.
Nat Rev Genet ; 21(8): 446-447, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32504077
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...