Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.278
Filtrar
1.
Am J Physiol Gastrointest Liver Physiol ; 326(4): G438-G459, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38193195

RESUMO

The calcium-sensing receptor (CaSR), a G protein-coupled receptor, regulates Ca2+ concentration in plasma by regulating parathyroid hormone secretion. In other tissues, it is reported to play roles in cellular differentiation and migration and in secretion and absorption. We reported previously that CaSR can be conditionally deleted in the mouse esophagus. This conditional knockout (KO) (EsoCaSR-/-) model showed a significant reduction in the levels of adherens and tight junction proteins and had a marked buildup of bacteria on the luminal esophageal surface. To further examine the role of CaSR, we used RNA sequencing to determine gene expression profiles in esophageal epithelia of control and EsoCaSR-/-mice RNA Seq data indicated upregulation of gene sets involved in DNA replication and cell cycle in EsoCaSR-/-. This is accompanied by the downregulation of gene sets involved in the innate immune response and protein homeostasis including peptide elongation and protein trafficking. Ingenuity pathway analysis (IPA) demonstrated that these genes are mapped to important biological networks including calcium and Ras homologus A (RhoA) signaling pathways. To further explore the bacterial buildup in EsoCaSR-/- esophageal tissue, 16S sequencing of the mucosal-associated bacterial microbiome was performed. Three bacterial species, g_Rodentibacter, s_Rodentibacter_unclassified, and s_Lactobacillus_hilgardi were significantly increased in EsoCaSR-/-. Furthermore, metagenomic analysis of 16S sequences indicated that pathways related to oxidative phosphorylation and metabolism were downregulated in EsoCaSR-/- tissues. These data demonstrate that CaSR impacts major pathways of cell proliferation, differentiation, cell cycle, and innate immune response in esophageal epithelium. The disruption of these pathways causes inflammation and significant modifications of the microbiome.NEW & NOTEWORTHY Calcium-sensing receptor (CaSR) plays a significant role in maintaining the barrier function of esophageal epithelium. Using RNA sequencing, we show that conditional deletion of CaSR from mouse esophagus causes upregulation of genes involved in DNA replication and cell cycle and downregulation of genes involved in the innate immune response, protein translation, and cellular protein synthesis. Pathway analysis shows disruption of signaling pathways of calcium and actin cytoskeleton. These changes caused inflammation and esophageal dysbiosis.


Assuntos
Cálcio , Microbiota , Animais , Camundongos , Cálcio/metabolismo , Receptores de Detecção de Cálcio/genética , Receptores de Detecção de Cálcio/metabolismo , Esôfago/metabolismo , Inflamação , Expressão Gênica
2.
Pathologie (Heidelb) ; 45(1): 51-58, 2024 Feb.
Artigo em Alemão | MEDLINE | ID: mdl-38170268

RESUMO

As a result of the high approval dynamics and the growing number of immuno-oncological therapy concepts, the complexity of therapy decisions and control in the area of carcinomas of the esophagus, gastroesophageal junction and stomach is constantly increasing. Since the treatment indication for PD­1 inhibitors that are currently approved in the European Union is often linked to the expression of PD-L1 (programmed cell death-ligand 1), the evaluation of tissue-based predictive markers by the pathologist is of crucial importance for treatment stratification. Even though the immunohistochemical analysis of the PD-L1 expression status is one of the best studied, therapy-relevant biomarkers for an immuno-oncological treatment, due to the high heterogeneity of carcinomas of the upper gastrointestinal tract, there are challenges in daily clinical diagnostic work with regard to implementation, standardization and interpretation of testing. An interdisciplinary group of experts from Germany has taken a position on relevant questions from daily pathological and clinical practice, which concern the starting material, quality-assured testing and the interpretation of pathological findings, and has developed recommendations for structured reporting.


Assuntos
Carcinoma , Neoplasias Gástricas , Humanos , Antígeno B7-H1/metabolismo , Neoplasias Gástricas/diagnóstico , Biomarcadores , Esôfago/metabolismo
3.
Cell Cycle ; 22(18): 2088-2096, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37749911

RESUMO

Alcohol contributes to cellular accumulation of acetaldehyde, a primary metabolite of alcohol and a major human carcinogen. Acetaldehyde can form DNA adducts and induce interstrand crosslinks (ICLs) that are repaired by the Fanconi anemia DNA repair pathway (FA pathway). Individuals with deficiency in acetaldehyde detoxification or in the FA pathway have an increased risk of squamous-cell carcinomas (SCCs) including those of the esophagus. In a recent report, we described the molecular basis of acetaldehyde-induced DNA damage in esophageal keratinocytes [1]. We demonstrated that, at physiologically relevant concentrations, acetaldehyde induces DNA damage at the DNA replication fork. This resulted in replication stress, leading to activation of the ATR-Chk1-dependent cell cycle checkpoints. We also reported that the p53 DNA damage response is elevated in response to acetaldehyde and that the FA pathway limits acetaldehyde-induced genomic instability. Here, we highlight these findings and present additional results to discuss the role of the FA pathway and p53 DNA damage response in the protection against genomic instability and esophageal carcinogenesis.


Assuntos
Acetaldeído , Anemia de Fanconi , Humanos , Acetaldeído/toxicidade , Acetaldeído/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Dano ao DNA , Etanol , Instabilidade Genômica , Reparo do DNA , Esôfago/metabolismo , Queratinócitos/metabolismo , Replicação do DNA
4.
Neuron ; 111(14): 2184-2200.e7, 2023 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-37192624

RESUMO

Vagal sensory neurons monitor mechanical and chemical stimuli in the gastrointestinal tract. Major efforts are underway to assign physiological functions to the many distinct subtypes of vagal sensory neurons. Here, we use genetically guided anatomical tracing, optogenetics, and electrophysiology to identify and characterize vagal sensory neuron subtypes expressing Prox2 and Runx3 in mice. We show that three of these neuronal subtypes innervate the esophagus and stomach in regionalized patterns, where they form intraganglionic laminar endings. Electrophysiological analysis revealed that they are low-threshold mechanoreceptors but possess different adaptation properties. Lastly, genetic ablation of Prox2 and Runx3 neurons demonstrated their essential roles for esophageal peristalsis in freely behaving mice. Our work defines the identity and function of the vagal neurons that provide mechanosensory feedback from the esophagus to the brain and could lead to better understanding and treatment of esophageal motility disorders.


Assuntos
Subunidade alfa 3 de Fator de Ligação ao Core , Esôfago , Motilidade Gastrointestinal , Proteínas de Homeodomínio , Células Receptoras Sensoriais , Nervo Vago , Animais , Camundongos , Subunidade alfa 3 de Fator de Ligação ao Core/genética , Subunidade alfa 3 de Fator de Ligação ao Core/metabolismo , Esôfago/inervação , Esôfago/metabolismo , Esôfago/fisiologia , Motilidade Gastrointestinal/genética , Motilidade Gastrointestinal/fisiologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Mecanorreceptores/fisiologia , Neurônios Aferentes/fisiologia , Células Receptoras Sensoriais/metabolismo , Células Receptoras Sensoriais/fisiologia , Estômago/inervação , Estômago/metabolismo , Estômago/fisiologia , Nervo Vago/fisiologia
5.
Acta Cir Bras ; 38: e381723, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37098927

RESUMO

PURPOSE: The effects of hesperidin application on the wound caused by esophageal burns were investigated in this study. METHODS: Wistar albino rats were divided into three groups: Control group: only 1 mL of 0.09% NaCl was administered i.p. for 28 days; Burn group: An alkaline esophageal burn model was created with 0.2 mL of 25% NaOH orally by gavage-1 mL of 0.09% NaCl was administered i.p. for 28 days; Burn+Hesperidin group: 1 mL of 50 mL/kg of hesperidin was given i.p. for 28 days to rats after burn injury. Blood samples were collected for biochemical analysis. Esophagus samples were processed for histochemical staining and immunohistochemistry. RESULTS: Malondialdehyde (MDA) and myeloperoxidase (MPO) levels were significantly increased in Burn group. Glutathione (GSH) content and histological scores of epithelialization, collagen formation, neovascularization was decreased. After hesperidin treatment, these values were significantly improved in the Burn+Hesperidin group. In the Burn group, epithelial cells and muscular layers were degenerated. Hesperidin treatment restored these pathologies in Burn+Hesperidin group. Ki-67 and caspase-3 expressions were mainly negative in control group; however, the expression was increased in the Burn group. In the Burn+Hesperidin group, Ki-67 and caspase-3 immune activities were reduced. CONCLUSIONS: Hesperidin dosage and application methods can be developed as an alternative treatment for burn healing and treatment.


Assuntos
Hesperidina , Animais , Ratos , Hesperidina/farmacologia , Antígeno Ki-67 , Caspase 3 , Cloreto de Sódio/farmacologia , Ratos Wistar , Cicatrização , Glutationa/metabolismo , Esôfago/lesões , Esôfago/metabolismo , Esôfago/patologia
6.
Cancer Discov ; 13(6): 1346-1363, 2023 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-36929873

RESUMO

Intestinal metaplasia in the esophagus (Barrett's esophagus IM, or BE-IM) and stomach (GIM) are considered precursors for esophageal and gastric adenocarcinoma, respectively. We hypothesize that BE-IM and GIM follow parallel developmental trajectories in response to differing inflammatory insults. Here, we construct a single-cell RNA-sequencing atlas, supported by protein expression studies, of the entire gastrointestinal tract spanning physiologically normal and pathologic states including gastric metaplasia in the esophagus (E-GM), BE-IM, atrophic gastritis, and GIM. We demonstrate that BE-IM and GIM share molecular features, and individual cells simultaneously possess transcriptional properties of gastric and intestinal epithelia, suggesting phenotypic mosaicism. Transcriptionally E-GM resembles atrophic gastritis; genetically, it is clonal and has a lower mutational burden than BE-IM. Finally, we show that GIM and BE-IM acquire a protumorigenic, activated fibroblast microenvironment. These findings suggest that BE-IM and GIM can be considered molecularly similar entities in adjacent organs, opening the path for shared detection and treatment strategies. SIGNIFICANCE: Our data capture the gradual molecular and phenotypic transition from a gastric to intestinal phenotype (IM) in the esophagus and stomach. Because BE-IM and GIM can predispose to cancer, this new understanding of a common developmental trajectory could pave the way for a more unified approach to detection and treatment. See related commentary by Stachler, p. 1291. This article is highlighted in the In This Issue feature, p. 1275.


Assuntos
Esôfago de Barrett , Gastrite Atrófica , Humanos , RNA , Metaplasia/genética , Esôfago/metabolismo , Esôfago/patologia , Esôfago de Barrett/genética , Esôfago de Barrett/metabolismo , Esôfago de Barrett/patologia , Análise de Sequência de RNA , Microambiente Tumoral
7.
Cells ; 11(19)2022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-36230907

RESUMO

Esophageal reconstruction through bio-engineered allografts that highly resemble the peculiar properties of the tissue extracellular matrix (ECM) is a prospective strategy to overcome the limitations of current surgical approaches. In this work, human esophagus was decellularized for the first time in the literature by comparing three detergent-enzymatic protocols. After decellularization, residual DNA quantification and histological analyses showed that all protocols efficiently removed cells, DNA (<50 ng/mg of tissue) and muscle fibers, preserving collagen/elastin components. The glycosaminoglycan fraction was maintained (70-98%) in the decellularized versus native tissues, while immunohistochemistry showed unchanged expression of specific ECM markers (collagen IV, laminin). The proteomic signature of acellular esophagi corroborated the retention of structural collagens, basement membrane and matrix-cell interaction proteins. Conversely, decellularization led to the loss of HLA-DR expression, producing non-immunogenic allografts. According to hydroxyproline quantification, matrix collagen was preserved (2-6 µg/mg of tissue) after decellularization, while Second-Harmonic Generation imaging highlighted a decrease in collagen intensity. Based on uniaxial tensile tests, decellularization affected tissue stiffness, but sample integrity/manipulability was still maintained. Finally, the cytotoxicity test revealed that no harmful remnants/contaminants were present on acellular esophageal matrices, suggesting allograft biosafety. Despite the different outcomes showed by the three decellularization methods (regarding, for example, tissue manipulability, DNA removal, and glycosaminoglycans/hydroxyproline contents) the ultimate validation should be provided by future repopulation tests and in vivo orthotopic implant of esophageal scaffolds.


Assuntos
Detergentes , Elastina , Colágeno , DNA/metabolismo , Esôfago/metabolismo , Glicosaminoglicanos/metabolismo , Humanos , Hidroxiprolina , Laminina , Proteômica
9.
J Mol Histol ; 53(3): 589-598, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35661289

RESUMO

Interstitial cells of Cajal (ICCs) function as pacemaker cells in the gastrointestinal tract. Acute thoracic trauma is a common and lethal cause of death due to physical trauma caused by traffic accidents. This study aimed to explore the distribution of esophageal ICCs and distribution changes observed after acute thoracic trauma. Thirty rabbits were randomly divided into a control group and two study groups. The control group animals underwent an esophagectomy. All animals in the study groups underwent right chest puncture using the Hopkinson bar technique. The study groups were subjected to esophagectomy 24 and 72 h after chest puncture. Distribution, morphology, and density of esophageal ICCs were detected using transmission electron microscopy, toluidine blue staining, and immunohistochemistry. Apoptosis of esophageal ICCs was evaluated using the terminal deoxynucleotidyl transferase (TdT) dUTP Nick-End Labeling assay. Western blotting and reverse transcription polymerase chain reaction were used to detect changes in the SCF/c-kit signaling pathway. Esophageal ICCs distribution and SCF/c-kit signal pathway decreased from the upper part to the lower part in both physiological state and after thoracic trauma. In contrast, death of ICCs increased from the upper part to the lower part, both in physiological and injured state (P < 0.05). After thoracic trauma, increased ICCs and decreased death of ICCs in all parts of the esophagus (P < 0.05) were observed. The observed distribution and changes in esophageal ICCs would have an impact on motility and motility disorders of the esophagus.


Assuntos
Células Intersticiais de Cajal , Animais , Western Blotting , Esôfago/metabolismo , Imuno-Histoquímica , Células Intersticiais de Cajal/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Coelhos
10.
J Mol Cell Biol ; 14(6)2022 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-35709398

RESUMO

Somatic stem cells are essential for the maintenance of tissue homeostasis. Despite its importance, how the esophageal stratified squamous epithelium executes its self-renewal and maintenance remains elusive. In this study, using 5-bromo-2'-deoxyuridine label-chase in rats in vivo and rat esophageal organoids in vitro together with genome-wide DNA methylation and single-cell RNA sequencing, we identified a slow-cycling/quiescent stem cell population that contained high levels of hemidesmosomes (HDs) and low levels of Wnt signaling localized spatially and randomly at the basal layer of the esophageal epithelium. Pseudotime cell trajectory analysis indicated that tissue cells originated from quiescent basal stem cells in the basal layer. Perturbations of HD component expression and/or Wnt signaling reduced the stem cell population in the basal layer of esophageal keratinocyte organoids, resulting in alterations in the organoid formation rate, size, morphogenesis, and proliferation-differentiation homeostasis. Furthermore, not only high levels of HDs and low levels of Wnt signaling but also an interplay between HD and Wnt signaling defined the stem cells of the basal layer. Hence, HDs and Wnt signaling are critical determinants for defining the stem cells of the basal layer required for tissue homeostasis in mammalian esophagi.


Assuntos
Carcinoma de Células Escamosas , Células-Tronco , Ratos , Animais , Células-Tronco/metabolismo , Epitélio/metabolismo , Esôfago/metabolismo , Diferenciação Celular , Carcinoma de Células Escamosas/metabolismo , Via de Sinalização Wnt , Proliferação de Células , Mamíferos
11.
Int J Mol Sci ; 23(9)2022 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-35562940

RESUMO

Transient receptor potential vanilloid 4 (TRPV4) is a non-selective cation channel that is broadly expressed in different human tissues, including the digestive system, where it acts as a molecular sensor and a transducer that regulates a variety of functional activities. Despite the extensive research to determine the role of this channel in the physiology and pathophysiology of different organs, the unique morphological and functional features of TRPV4 in the esophagus remain largely unknown. Ten years ago, TRPV4 was shown to be highly expressed in esophageal epithelial cells where its activation induces Ca2+-dependent ATP release, which, in turn, mediates several functions, ranging from mechanosensation to wound healing. This review summarizes the research progress on TRPV4, and focuses on the functional expression of TRPV4 in esophageal epithelium and its possible role in different esophageal diseases that would support TRPV4 as a candidate target for future therapeutic approaches to treat patients with these conditions.


Assuntos
Esôfago , Canais de Cátion TRPV , Células Epiteliais/metabolismo , Mucosa Esofágica/metabolismo , Esôfago/metabolismo , Humanos , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo
12.
Transgenic Res ; 31(3): 341-349, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35570234

RESUMO

Myostatin (MSTN) is a member of the transforming growth factor-ß superfamily that inhibits skeletal muscle growth and development. The esophagus is composed of skeletal muscle and smooth muscle, but the effect of MSTN on esophagus striated muscle (ESM) is unknown. The present study investigated the role of MSTN in ESM using MSTN mutant pigs through histological, gene and protein expression analysis in ESM of MSTN knockout (MSTN-/-) pigs and their wild type (WT) littermates. Hematoxylin-eosin staining showed that the fiber cross-sectional areas in ESM of MSTN-/- pigs were significantly larger than WT pigs (P < 0.05). Immunofluorescence staining showed that the percentage of type I muscle fibers in MSTN-/- pigs were significantly lower than WT pigs (P < 0.01) and type IIA muscle fibers in MSTN-/- pigs were significantly higher than WT pigs (21% higher, P < 0.01). However, type IIB muscle fibers were not detected in the ESM of MSTN-/- or WT pigs indicating that muscle fiber types in pig ESM was composed of type I and IIA. The mRNA levels of myogenic regulatory factors (MRFs) including myogenic differentiation (MyoD), myogenin (MyoG), myogenic factor 5 (Myf5) and myogenic regulatory factor 4 (MRF4) in ESM of MSTN-/- pigs showed a significant increase (P < 0.05 at least) when compared to WT pigs while mRNA level of myocyte enhancer factor 2C (MEF2C) displayed a decrease (P < 0.001). Protein expression of myosin heavy chain I (MHC-I) in MSTN-/- ESM was decreased and myosin heavy chain IIA (MHC-IIA) was increased (P < 0.01, P < 0.05). These findings indicate that MSTN plays an important role in esophageal striated muscle development and regulates muscle fiber types.


Assuntos
Cadeias Pesadas de Miosina , Miostatina , Animais , Esôfago/metabolismo , Hipertrofia/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Miostatina/genética , RNA Mensageiro , Suínos/genética
13.
Poult Sci ; 101(7): 101859, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35561461

RESUMO

The esophagus is a tubular organ which act as a passage for food from oral cavity to stomach. Telocytes (TCs) are a unique type of interstitial cell whose existence in many organs of various species still remains unknown. In the present study, we used transmission electron microscopy (TEM) and immunohistochemistry (CD34, Vimentin, PDGFR-α) to identify subepithelial TCs in the esophageal wall of chickens. TEM micrographs confirmed the presence of TCs in the lamina propria, tunica submucosa, and tunica muscularis muscular layer of the esophageal wall. A large population of TCs were observed just beneath the epithelial layer of the esophageal wall, and the TCs demonstrated structural heterogenicity, featuring various cell body shapes of cell bodies and telopodes (Tps) with podoms, podomeres, and dichotomous branching. Furthermore, a large number of extracellular vesicles were found to be associated with TCs/Tps. Cellular extensions from TCs were observed in close proximity to blood vessels, immune cells, and mucosal glands. In the submucosa, Tps and immune cells were in very close contact. Immunohistochemical results showed that there were CD34+ cells, vimentin+ cells, and PDGFR-α+ cells in the subepithelium, lamina propria, and mucosal glands of the chicken esophageal wall, which was consistent with the TEM results. Overall, our data confirmed the existence of TCs in the chicken esophagus and suggested that TCs might contribute to epithelial regeneration and tissue homeostasis.


Assuntos
Galinhas , Telócitos , Animais , Antígenos CD34/análise , Antígenos CD34/metabolismo , Galinhas/metabolismo , Esôfago/metabolismo , Telócitos/química , Telócitos/metabolismo , Vimentina/análise , Vimentina/metabolismo
14.
Cell Rep ; 38(11): 110510, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35294885

RESUMO

The mechanisms coupling fate specification of distinct tissues to their physical separation remain to be understood. The trachea and esophagus differentiate from a single tube of definitive endoderm, requiring the transcription factors SOX2 and NKX2-1, but how the dorsoventral site of tissue separation is defined to allocate tracheal and esophageal cell types is unknown. Here, we show that the EPH/EPHRIN signaling gene Efnb2 regulates tracheoesophageal separation by controlling the dorsoventral allocation of tracheal-fated cells. Ventral loss of NKX2-1 results in disruption of separation and expansion of Efnb2 expression in the trachea independent of SOX2. Through chromatin immunoprecipitation and reporter assays, we find that NKX2-1 likely represses Efnb2 directly. Lineage tracing shows that loss of NKX2-1 results in misallocation of ventral foregut cells into the esophagus, while mosaicism for Nkx2-1 generates ectopic NKX2-1/EPHRIN-B2 boundaries that organize ectopic tracheal separation. Together, these data demonstrate that NKX2-1 coordinates tracheal specification with tissue separation through the regulation of EPHRIN-B2 and tracheoesophageal cell sorting.


Assuntos
Endoderma , Traqueia , Sistema Digestório/metabolismo , Endoderma/metabolismo , Efrina-B2/metabolismo , Esôfago/metabolismo , Traqueia/metabolismo
15.
Radiat Res ; 197(5): 480-490, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-35172004

RESUMO

Radiation-induced esophageal injury (RIEI) is a major dose-limiting complication of radiotherapy, especially for esophageal and thoracic cancers. RIEI is a multi-factorial and multi-step process, which is regulated by a complex network of DNA, RNA, protein and metabolite. However, it is unclear which esophageal metabolites are altered by ionizing radiation and how these changes affect RIEI progression. In this work, we established a rat model of RIEI with 0-40 Gy X-ray irradiation. Esophageal irradiation using ≥25 Gy induced significant changes to rats, such as body weight, food intake, water intake and esophageal structure. The metabolic changes and related pathways of rat esophageal metabolites were investigated by liquid chromatography-mass spectrometry (LC-MS). One hundred eighty metabolites showed an up-regulation in a dose-dependent manner (35 Gy ≥ 25 Gy > controls), and 199 metabolites were downregulated with increasing radiation dose (35 Gy ≤ 25 Gy < controls). The KEGG analysis showed that ionizing radiation seriously disrupted multiple metabolic pathways, and arachidonic acid metabolism was the most significantly enriched pathway. 20 metabolites were dysregulated in arachidonic acid metabolism, including up-regulation of five prostaglandins (PGA2, PGJ2, PGD2, PGH2, and PGI2) in 25 or 35 Gy groups. Cyclooxygenase-2 (COX-2), the key enzyme in catalyzing the biosynthesis of prostaglandins from arachidonic acid, was highly expressed in the esophagus of irradiated rats. Additionally, receiver operating characteristic (ROC) curve analysis revealed that PGJ2 may serve as a promising tissue biomarker for RIEI diagnosis. Taken together, these findings indicate that ionizing radiation induces esophageal metabolic alterations, which advance our understanding of the pathophysiology of RIEI from the perspective of metabolism.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Metabolômica , Lesões por Radiação , Animais , Ácido Araquidônico/química , Ácido Araquidônico/metabolismo , Esôfago/metabolismo , Prostaglandinas , Lesões por Radiação/etiologia , Ratos
16.
Sci Total Environ ; 824: 153761, 2022 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-35151731

RESUMO

A representative polycyclic aromatic hydrocarbon, benzo[a]pyrene (B[a]P), has been widely detected in environmental compartments and is highly carcinogenic to humans. Oral ingestion of B[a]P is the dominant exposure pathway. The esophagus acts as the first contact point when B[a]P enters the human body. However, its role in the development of human esophageal cancer is rarely discussed. Herein, we employed untargeted metabolomics in combination with proteomics to explore B[a]P-related intracellular responses in human esophageal cell lines. Our results demonstrated that B[a]P exposure induced significant metabolic disorders, further leading to overproduction of reactive oxygen species (ROS) and disturbance of the cellular viability process and migration ability of esophageal cells. In response, glutathione (GSH) was consumed to meet the demand for cellular detoxification, and thioredoxin (TXN) was upregulated to balance the cellular redox. These alterations caused the reregulation of some specific protein families, including S100A proteins, ribosomal proteins, and histone H1 proteins. Such changes impeded the viability and migration of esophageal cells, which could adversely affect wound healing of the epithelium. These cellular responses indicate that B[a]P will cause serious cellular damage to esophageal cells and increase the carcinogenic risk even as a result of short-term exposure. SYNOPSIS: Our omics study demonstrated how benzo[a]pyrene hampered the migration of esophageal cells and proposed a plausible mechanism underlying its carcinogenicity, which may contribute to our understanding of environmental pollutants.


Assuntos
Benzo(a)pireno , Hidrocarbonetos Policíclicos Aromáticos , Benzo(a)pireno/metabolismo , Benzo(a)pireno/toxicidade , Carcinógenos , Esôfago/metabolismo , Glutationa , Humanos , Metabolômica , Proteômica
17.
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
18.
Am J Gastroenterol ; 117(2): 346-351, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34928874

RESUMO

INTRODUCTION: The aim of this study was to investigate the effect of spontaneous sleep positions on the occurrence of nocturnal gastroesophageal reflux. METHODS: In patients referred for ambulatory pH-impedance reflux monitoring, the concurrent sleep position was measured using a sleep position measurement device (measuring left, right, supine, and prone positions). RESULTS: Fifty-seven patients were included. We observed a significantly shorter acid exposure time in the left (median 0.0%, P25-P75, 0.0%-3.0%), compared with the right lateral position (median 1.2%, 0.0%-7.5%, P = 0.022) and the supine position (median 0.6%, 0.00%-8.3%, P = 0.022). The esophageal acid clearance time was significantly shorter in the left lateral decubitus position (median 35 seconds, 16-115 seconds), compared with the supine (median 76 seconds, 22-257 seconds, P = 0.030) and right lateral positions (median 90 seconds, 26-250 seconds, P = 0.002). DISCUSSION: The left lateral decubitus position is associated with significantly shorter nocturnal esophageal acid exposure time and faster esophageal acid clearance compared with the supine and right lateral decubitus positions (see visual abstract).


Assuntos
Esôfago/metabolismo , Refluxo Gastroesofágico/fisiopatologia , Postura/fisiologia , Sono/fisiologia , Impedância Elétrica , Monitoramento do pH Esofágico/métodos , Esôfago/fisiopatologia , Feminino , Refluxo Gastroesofágico/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Masculino , Pessoa de Meia-Idade , Polissonografia
19.
FASEB J ; 36(1): e22083, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34918385

RESUMO

Proton pump inhibitors (PPIs) are the mainstay of therapy for gastroesophageal reflux disease (GERD) but up to 60% of patients have inadequate response to therapy. Acid sensing ion channels (ASICs) play important roles in nociception. This study aimed to investigate whether the increased expression of ASICs results in neuronal hyperexcitability in GERD. Esophageal biopsies were taken from GERD patients and healthy subjects to compare expression of ASIC1 and 3. Next, gene and protein expression of ASIC1 and 3 from esophageal mucosa and dorsal root ganglia (DRG) neurons were measured by qPCR, Western-blot and immunofluorescence in rodent models of reflux esophagitis (RE), non-erosive reflux disease (NERD), and sham operated groups. Excitability of DRG neurons in the GERD and sham groups were also tested by whole-cell patch-clamp recordings. We demonstrated that ASIC1 and 3 expression were significantly increased in patients with RE compared with healthy controls. This correlated positively with symptom severity of heartburn and regurgitation (p < .001). Next, ASIC1 and 3 gene and protein expression in rodent models of RE and NERD were similarly increased in esophageal mucosa as well as T3-T5 DRG neurons compared with sham operation. DRG neurons from RE animals showed hyperexcitability compared with sham group. However, intrathecal injection of ASIC specific inhibitors, PcTx1 and APTEx-2, as well as silencing ASIC1 and 3 genes with specific siRNAs prevented visceral hypersensitivity. Overall, upregulation of ASIC1 and 3 may lead to visceral hypersensitivity in RE and NERD and may be a potential therapeutic target for PPI non-responsive patients.


Assuntos
Canais Iônicos Sensíveis a Ácido/biossíntese , Esôfago/metabolismo , Refluxo Gastroesofágico/metabolismo , Azia/metabolismo , Regulação para Cima , Canais Iônicos Sensíveis a Ácido/genética , Animais , Refluxo Gastroesofágico/genética , Azia/genética , Humanos , Masculino , Ratos , Ratos Sprague-Dawley
20.
Mucosal Immunol ; 15(2): 327-337, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34903876

RESUMO

Fibroblasts mediate tissue remodeling in eosinophilic esophagitis (EoE), a chronic allergen-driven inflammatory pathology. Diverse fibroblast subtypes with homeostasis-regulating or inflammatory profiles have been recognized in various tissues, but which mediators induce these alternate differentiation states remain largely unknown. We recently identified that TNFSF14/LIGHT promotes an inflammatory esophageal fibroblast in vitro. Herein we used esophageal biopsies and primary fibroblasts to investigate the role of the LIGHT receptors, herpes virus entry mediator (HVEM) and lymphotoxin-beta receptor (LTßR), and their downstream activated pathways, in EoE. In addition to promoting inflammatory gene expression, LIGHT down-regulated homeostatic factors including WNTs, BMPs and type 3 semaphorins. In vivo, WNT2B+ fibroblasts were decreased while ICAM-1+ and IL-34+ fibroblasts were expanded in EoE, suggesting that a LIGHT-driven gene signature was imprinted in EoE versus normal esophageal fibroblasts. HVEM and LTßR overexpression and deficiency experiments demonstrated that HVEM regulates a limited subset of LIGHT targets, whereas LTßR controls all transcriptional effects. Pharmacologic blockade of the non-canonical NIK/p100/p52-mediated NF-κB pathway potently silenced LIGHT's transcriptional effects, with a lesser role found for p65 canonical NF-κB. Collectively, our results show that LIGHT promotes differentiation of esophageal fibroblasts toward an inflammatory phenotype and represses homeostatic gene expression via a LTßR-NIK-p52 NF-κB dominant pathway.


Assuntos
Esôfago , Inflamação , Transcriptoma , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral , Esôfago/metabolismo , Fibroblastos/metabolismo , Homeostase , Humanos , Inflamação/genética , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...