Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Biology (Basel) ; 13(7)2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-39056656

RESUMO

Fibroblast heterogeneity remains undefined in eosinophilic esophagitis (EoE), an allergic inflammatory disorder complicated by fibrosis. We utilized publicly available single-cell RNA sequencing data (GSE201153) of EoE esophageal biopsies to identify fibroblast sub-populations, related transcriptomes, disease status-specific pathways and cell-cell interactions. IL13-treated fibroblast cultures were used to model active disease. At least 2 fibroblast populations were identified, F_A and F_B. Several genes including ACTA2 were more enriched in F_A. F_B percentage was greater than F_A and epithelial-mesenchymal transition upregulated in F_B vs. F_A in active and remission EoE. Epithelial-mesenchymal transition was also upregulated in F_B in active vs. remission EoE and TNF-α signaling via NFKB was downregulated in F_A. IL-13 treatment upregulated ECM-related genes more profoundly in ACTA2- fibroblasts than ACTA2+ myofibroblasts. After proliferating epithelial cells, F_B and F_A contributed most to cell-cell communication networks. ECM-Receptor interaction strength was stronger than secreted or cell-cell contact signaling in active vs. remission EoE and significant ligand-receptor pairs were driven mostly by F_B. This unbiased analysis identifies at least 2 fibroblast sub-populations in EoE in vivo, distinguished in part by ACTA2. Fibroblasts play a critical role in cell-cell interactions in EoE, most profoundly via ECM-receptor signaling via the F_B sub-group.

2.
Curr Opin Gastroenterol ; 40(4): 291-298, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38661722

RESUMO

PURPOSE OF REVIEW: Eosinophilic esophagitis (EoE) is a Th2 immune/antigen-mediated disorder characterized by esophageal dysfunction and eosinophilic inflammation. Worsening dysphagia and food impactions are significant complications associated with esophageal remodeling and fibrostenotic disease. This review highlights the most recent research findings pertaining to mechanisms of sub-epithelial fibrosis in EoE, current diagnostic tools, and therapeutic approaches. RECENT FINDINGS: Recent studies leveraging publicly available single cell sequencing databases and comparative proteomics have furthered our understanding of the mechanisms mediating fibrosis. Fibroblast crosstalk with the extracellular matrix and with epithelial, endothelial, and T cells have been implicated, with the likely existence of multiple fibroblast sub-types. Accurate diagnosis of remodeling with biopsies remains a challenge due to inadequate depth of sampling. Web-based tools incorporating epithelial findings show promise in predicting subepithelial fibrosis. Impedance planimetry with esophageal distensibility measurements are increasingly utilized tools to assess fibrostenotic severity. Immunostaining and luminal captured proteins associated with remodeling show promise as potential molecular markers of fibrosis. Anti-inflammatory therapy may improve esophageal fibrosis and distensibility, although specific fibrosis-targeted therapy is lacking. SUMMARY: Recent studies highlight novel mechanisms of fibrosis in EoE. Improved understanding of these mechanisms may lead to novel diagnostic strategies and therapies, and thereby inform treatment decisions.


Assuntos
Esofagite Eosinofílica , Esôfago , Fibrose , Esofagite Eosinofílica/fisiopatologia , Esofagite Eosinofílica/diagnóstico , Esofagite Eosinofílica/terapia , Esofagite Eosinofílica/patologia , Humanos , Esôfago/patologia , Esôfago/fisiopatologia
3.
ACG Case Rep J ; 10(12): e01239, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38130481

RESUMO

Colorectal cancer (CRC) metastasizing to the stomach and duodenum is rare. Even rarer is when the CRC subtype is signet-ring cell carcinoma (SRCC). Endoscopic findings of CRC metastasis to the stomach have been described as solitary and submucosal while duodenal metastasis has been observed to be exophytic. In this report, we describe a case of a middle-aged man with colon SRCC presenting with oral intolerance. He was found to have concurrent metastases to the stomach and duodenum and died 8 months after his SRCC diagnosis.

4.
Alcohol Clin Exp Res (Hoboken) ; 47(7): 1297-1311, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37128647

RESUMO

BACKGROUND: Mechanisms by which alcohol increases the risk of esophageal squamous cell carcinoma remain undefined. Human esophageal myofibroblasts (HEMFs) subjacent to the squamous epithelium are exposed directly to these agents via epithelial barrier defects and indirectly via factors derived from the exposed epithelium. Our aim was to investigate the cellular biology of HEMFs and HEMF-esophageal epithelial cell interactions in response to alcohol and its toxic metabolite acetaldehyde. METHODS: An immortalized HEMF and a human esophageal epithelial cell line (Epi) were treated with alcohol (0 to 200 mM) or acetaldehyde (0 to 100 µM) in a cyclic fashion or incubated with supernatants collected from treated cells. Healthy cell %, reactive oxygen species (ROS), and proliferation were assessed via flow cytometry, luminescence, scratch wound, and colorimetric assays, respectively. A 15-plex multiplex assay was performed on cell supernatants, followed by IL-6 and IL-8 qRT-PCR and ELISA. RESULTS: Healthy HEMF decreased to less than 80% at 30 mM alcohol and 70 µM acetaldehyde, with microscopic changes at 40 µM acetaldehyde. HEMF ROS was detected at 100 mM alcohol and 80 µM acetaldehyde. Supernatants from 30 mM alcohol- or 40 µM acetaldehyde-treated HEMFs increased Epi proliferation more than two-fold that of lower doses. In the complementary studies, healthy Epi cells decreased to less than 80% at 50 mM and 70 µM acetaldehyde, with microscopic changes at 40 µM. Supernatants from Epi treated with 50 mM alcohol or 40 µM acetaldehyde increased HEMF proliferation more than two-fold that of lower doses. A multiplex assay of supernatants showed the greatest increase in concentrations of IL-6 and IL-8 in HEMFs and in Epi treated with higher doses of alcohol or acetaldehyde. Neutralization of IL-6 and IL-8 in supernatants of HEMFS and esophageal epithelial cells inhibited the proliferation of Epi and HEMFs, respectively. CONCLUSIONS: Alcohol and acetaldehyde doses in which the majority of HEMFs and epithelial cells are healthy, elicit the production of paracrine mediators with pro-proliferative effects on neighboring cells. Understanding the effect of alcohol and acetaldehyde on HEMFs and HEMF-epithelial interactions could help to identify the molecular basis by which alcohol increases the risk for esophageal cancer.

5.
Int J Mol Sci ; 23(18)2022 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-36142285

RESUMO

Subepithelial human esophageal myofibroblasts (HEMFs) in gastroesophageal reflux disease (GERD) are exposed to luminal contents via impaired squamous epithelium barrier integrity. The supernatant of HEMFs treated with acidic bile salts reflective of in vivo reflux increases squamous epithelial thickness. We aimed to identify the involved mechanisms using an unbiased approach. Acidic-bile-salt-treated primary HEMF cultures (n = 4) were submitted for RNA-Seq and analyzed with Partek Flow followed by Ingenuity Pathway Analysis (IPA). A total of 1165 molecules (579 downregulated, 586 upregulated) were differentially expressed, with most top regulated molecules either extracellular or in the plasma membrane. Increases in HEMF CXCL-8, IL-6, AREG, and EREG mRNA, and protein secretion were confirmed. Top identified canonical pathways were agranulocyte and granulocyte adhesion and diapedesis, PI3K/AKT signaling, CCR5 signaling in macrophages, and the STAT3 pathway. Top diseases and biological functions were cellular growth and development, hematopoiesis, immune cell trafficking, and cell-mediated response. The targets of the top upstream regulator ErbB2 included CXCL-8, IL-6, and AREG and the inhibition of CXCL-8 in the HEMF supernatant decreased squamous epithelial proliferation. Our work shows an inflammatory/immune cell and proliferative pathways activation in HEMFs in the GERD environment and identifies CXCL-8 as a HEMF-derived chemokine with paracrine proliferative effects on squamous epithelium.


Assuntos
Carcinoma de Células Escamosas , Refluxo Gastroesofágico , Ácidos e Sais Biliares/metabolismo , Ácidos e Sais Biliares/farmacologia , Carcinoma de Células Escamosas/metabolismo , Humanos , Interleucina-6/metabolismo , Miofibroblastos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/genética
6.
JGH Open ; 5(7): 809-812, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34263076

RESUMO

BACKGROUND AND AIM: Methane levels in methane-positive lactulose breath tests are frequently elevated at time zero. We hypothesized that baseline methane level is sufficient to detect excessive methane production and thereby avoid extended testing. Our aim was to determine if baseline methane levels were sufficient to identify methane-positive individuals as defined by current guidelines. METHODS: A retrospective study of lactulose breath tests was conducted at an open access motility lab. A methane-positive study was defined as a methane level ≥10 ppm at any time. Small intestinal bacterial overgrowth (SIBO) was defined as a ≥20 ppm rise in hydrogen from baseline by 90 min. Dual-positive SIBO and methane studies were identified. Demographics, symptoms, and indications were recorded. RESULTS: Of 745 tests, 33.1%, 15.0%, and 3.1% were SIBO, methane, and dual-positive, respectively. Precisely 96.4% of methane-positive studies had methane levels ≥10 ppm within 90 min and 75.9% had levels ≥10 ppm at time 0. An additional elevation of ≥20 ppm over baseline within 90 min was observed in 32.1%. Of 22 methane-positive patients with constipation, methane levels were ≥10 ppm at baseline in 81.8% and were ≥10 ppm within 90 min in all cases. CONCLUSIONS: Nearly 25% of methane-positive studies were not identified by a fasting methane level, but 96% were identified within 90 min. Most methane-positive studies did not have a rise of 20 ppm above baseline. Our findings suggest the lactulose breath test for hydrogen and methane can be complete at 90 min.

8.
Dig Dis Sci ; 66(6): 1940-1948, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32691385

RESUMO

BACKGROUND: Defining factors associated with severe reflux esophagitis allows for identification of subgroups most at risk for complications of strictures and esophageal malignancy. We hypothesized there might be unique clinical features in patients with reflux esophagitis in a predominantly Hispanic population of a large, safety-net hospital. AIM: Define clinical and endoscopic features of reflux esophagitis in a predominantly Hispanic population of a large, safety-net hospital. METHODS: This is retrospective comparative study of outpatients and hospitalized patients identified with mild (Los Angeles Grade A/B) and severe (Los Angeles Grade C/D) esophagitis through an endoscopy database review. The electronic medical record was reviewed for demographic and clinical data. RESULTS: Reflux esophagitis was identified in 382/5925 individuals: 56.5% males and 79.8% Hispanic. Multivariable logistic regression model adjusted for age, gender, race, body mass index (BMI), tobacco and alcohol use, and hospitalization status with severity as the outcome showed an interaction between gender and BMI (p ≤ 0.01). Stratification by gender showed that obese females had decreased odds of severe esophagitis compared to normal BMI females (OR = 0.18, 95% CI = 0.07-0.47; p < 0.01). In males, the odds of esophagitis were higher in inpatient status (OR = 2.84, 95% CI = 1.52 - 5.28; p < 0.01) and as age increased (OR = 1.37, 95% CI = 1.03 - 1.83; p = 0.03). CONCLUSIONS: We identify gender-specific associations with severe esophagitis in a predominantly Hispanic cohort. In females, obese BMI appears to be protective against severe esophagitis compared to normal BMI, while in men inpatient status and increasing age were associated with increased odds of severe esophagitis.


Assuntos
Esofagite Péptica/diagnóstico , Esofagite Péptica/fisiopatologia , Hispânico ou Latino , Hospitais de Condado/tendências , Provedores de Redes de Segurança/tendências , Caracteres Sexuais , Adulto , Idoso , Esofagite Péptica/terapia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Fatores de Risco
9.
PLoS One ; 15(9): e0238852, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32925965

RESUMO

The pathogenesis of esophageal injury in gastroesophageal reflux disease (GERD) is incompletely understood. We modeled exposure of human esophageal myofibroblasts (HEMFs) to gastroesophageal reflux by repeated treatment with pH 4.5 and pH 4.5 bile salts and determined the effects on the epithelium in a 3D organotypic-like air-liquid interface model. Total, basal and supra-basal thickness of the epithelium were measured and immunostaining for p63, for basal (CK 14) and supra-basal (CK 4) squamous differentiation markers, and for cell proliferation (PCNA) were performed. Epithelial cell proliferation in response to HEMF conditioned media was also assessed in 2D culture. In the 3D organotypic model, total epithelial thickness increased similarly with pH 4.5 and pH 4.5 bile salt treated versus untreated and bile salt treated HEMF conditioned media. Epithelial p63 immunostaining was increased and multilayered. There was expansion of the CK14+ basal and CK4+ supra-basal layers in the epithelium established with conditioned media from pH 4.5 and pH 4.5 bile salt treated HEMFs versus untreated HEMF conditioned media. PCNA + cells per µm of tissue were unchanged in the basal layer across all treatment conditions while PCNA + cells per total DAPI + cells were decreased. In 2D culture, basal epithelial proliferation decreased with conditioned media from pH 4.5 and pH 4.5 bile salt treated HEMFs compared to conditioned media from untreated HEMF conditioned media. Secreted factors from HEMFs treated with acidic stimuli encountered in GERD increase epithelial thickness compared to secreted factors from untreated HEMFs and expand both basal and supra-basal layers. Our findings demonstrate for the first time paracrine regulation of the squamous epithelium from acid stimulated HEMFs. The effects of secreted factors from acid treated HEMFs on basal cell proliferation in this model and the mechanism mediating the increase in epithelial thickness merit further investigation.


Assuntos
Ácidos e Sais Biliares/farmacologia , Meios de Cultivo Condicionados/farmacologia , Células Epiteliais/patologia , Esôfago/patologia , Refluxo Gastroesofágico/patologia , Miofibroblastos/citologia , Comunicação Parácrina , Técnicas de Cultura de Células , Células Cultivadas , Células Epiteliais/efeitos dos fármacos , Esôfago/efeitos dos fármacos , Humanos , Técnicas In Vitro
10.
Sci Rep ; 8(1): 12354, 2018 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-30120338

RESUMO

We have previously shown myofibroblasts subjacent to the squamous epithelium in the normal human esophagus and an increase in esophagitis. Myofibroblast contribution to bone morphogenetic protein (BMP) signaling and to paracrine mediated epithelial-mesenchymal interactions in the human esophagus remains incompletely defined. We investigated BMP4 and BMP inhibitor GREM1 gene expression and protein levels in previously characterized human esophageal myofibroblast primary cultures and in a human esophageal myofibroblast cell line. We adapted human esophageal myofibroblast conditioned media into a 3D organotypic model to investigate the effect of myofibroblast secreted factors on squamous epithelial morphology, proliferation, differentiation and BMP signaling. Human esophageal myofibroblasts constitutively secrete GREM1 and increase BMP4 expression and BMP4 secretion in response to epithelial Hedgehog ligand SHH. Detection of secreted BMP4 is decreased in the presence of GREM1. Myofibroblast conditioned media increases epithelial proliferation and expression of basal markers p63 and CK14 leading to an overall increase in epithelial thickness. Epithelial BMP signaling increases with myofibroblast conditioned media. These findings were partially reversed with GREM1 inhibition. Our results demonstrate that myofibroblasts are potential sources of GREM1 and of BMP4 in the human esophagus and that human esophageal myofibroblast-epithelial paracrine interactions contribute in part to the regulation of epithelial growth.


Assuntos
Proteínas Morfogenéticas Ósseas/biossíntese , Mucosa Esofágica/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Miofibroblastos/metabolismo , Comunicação Parácrina , Proteínas Morfogenéticas Ósseas/genética , Linhagem Celular , Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Transdução de Sinais
11.
Ann N Y Acad Sci ; 1380(1): 183-194, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27441783

RESUMO

Gastroesophageal reflux disease (GERD) has become the most commonly seen gastrointestinal disorder in outpatient clinics. In the United States, around 20% of the general population experience heartburn on a weekly basis. Although clinical complaints can be mild or moderate, patients with GERD may develop further complications, such as peptic strictures, Barrett's esophagus (BE), and even esophageal adenocarcinoma. Pathologically, GERD is developed as a result of chronic and enhanced exposure of the esophageal epithelium to noxious gastric refluxate. In this review article, we provide an overview of GERD and then focus on the roles of stromal cells, interleukin 4, and adiponectin in GERD and BE. The importance of inflammation and immunomodulators in GERD pathogenesis is highlighted. Targeting the immunomodulators or inflammation in general may improve the therapeutic outcome of GERD, in particular, in those refractory to proton pump inhibitors.


Assuntos
Adiponectina/metabolismo , Refluxo Gastroesofágico/complicações , Refluxo Gastroesofágico/imunologia , Fatores Imunológicos/metabolismo , Interleucina-4/metabolismo , Adiponectina/antagonistas & inibidores , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Refluxo Gastroesofágico/diagnóstico , Humanos , Fatores Imunológicos/antagonistas & inibidores , Interleucina-4/antagonistas & inibidores , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/metabolismo
12.
Ann N Y Acad Sci ; 1380(1): 58-66, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27442914

RESUMO

Treatment of esophageal pain remains a major challenge for the clinician. Although many patients have heartburn and may respond to proton pump inhibitors, there in an unmet need for other treatment modalities in patients where there are no obvious pathological findings. Although analgesics are the mainstay in esophageal pain treatment, many patients are nonresponders to these drugs. The current concise review focuses on other systems affecting pain processing, where better understanding may serve as a framework for therapy. These are the parasympathetic nervous system, exercise, and personality profiles. Finally, treatment with analgesics for functional chest pain remains a challenge, and an overview of treatment with antidepressive drugs is provided.


Assuntos
Dor no Peito/diagnóstico , Dor no Peito/terapia , Esôfago/patologia , Esôfago/fisiologia , Analgésicos/farmacologia , Analgésicos/uso terapêutico , Antidepressivos/farmacologia , Antidepressivos/uso terapêutico , Dor no Peito/fisiopatologia , Terapia Combinada/métodos , Esôfago/efeitos dos fármacos , Exercício Físico/fisiologia , Exercício Físico/psicologia , Refluxo Gastroesofágico/diagnóstico , Refluxo Gastroesofágico/fisiopatologia , Refluxo Gastroesofágico/terapia , Fármacos Gastrointestinais/farmacologia , Fármacos Gastrointestinais/uso terapêutico , Humanos , Personalidade/efeitos dos fármacos , Personalidade/fisiologia , Resultado do Tratamento
13.
Ann N Y Acad Sci ; 1380(1): 33-47, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27311061

RESUMO

Standard tests in clinical practice commonly fail to demonstrate a clear esophageal etiology for symptoms such as heartburn, dysphagia, or chest pain. Over the years, various provocative measures have been developed to provide a better understanding of the origins of such symptoms. Some measures, such as esophageal acid infusion or changing bolus consistency, can be easily incorporated into clinical practice. Others, such as multimodal stimulation systems, are more technically demanding. They have contributed to a better understanding of esophageal physiology in health and disease. Their role in clinical decision making is still evolving. This focused review provides a summary of the esophageal nociceptive pathways and how provocative testing can be used to interrogate their integrity.


Assuntos
Testes Diagnósticos de Rotina/métodos , Esôfago/fisiologia , Refluxo Gastroesofágico/diagnóstico , Refluxo Gastroesofágico/fisiopatologia , Dor no Peito/diagnóstico , Dor no Peito/fisiopatologia , Testes Diagnósticos de Rotina/tendências , Previsões , Humanos , Manometria/métodos , Manometria/tendências
14.
PLoS One ; 11(4): e0153185, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27055018

RESUMO

Stromal cells with a myofibroblast phenotype present in the normal human esophagus are increased in individuals with gastro-esophageal reflux disease (GERD). We have previously demonstrated that myofibroblasts stimulated with acid and TLR4 agonists increase IL-6 and IL-8 secretion using primary cultures of myofibroblasts established from normal human esophagus. While primary cultures have the advantage of reflecting the in vivo environment, a short life span and unavoidable heterogeneity limits the usefulness of this model in larger scale in vitro cellular signaling studies. The major aim of this paper therefore was to generate a human esophageal myofibroblast line with an extended lifespan. In the work presented here we have generated and characterized an immortalized human esophageal myofibroblast line by transfection with a commercially available GFP-hTERT lentivirus. Immortalized human esophageal myofibroblasts demonstrate phenotypic, genotypic and functional similarity to primary cultures of esophageal myofibroblasts we have previously described. We found that immortalized esophageal myofibroblasts retain myofibroblast spindle-shaped morphology at low and high confluence beyond passage 80, and express α-SMA, vimentin, and CD90 myofibroblast markers. Immortalized human esophageal myofibroblasts also express the putative acid receptor TRPV1 and TLR4 and retain the functional capacity to respond to stimuli encountered in GERD with secretion of IL-6. Finally, immortalized human esophageal myofibroblasts also support the stratified growth of squamous esophageal epithelial cells in 3D organotypic cultures. This newly characterized immortalized human esophageal myofibroblast cell line can be used in future cellular signaling and co-culture studies.


Assuntos
Biomarcadores/análise , Linhagem Celular Transformada/citologia , Esôfago/citologia , Refluxo Gastroesofágico , Miofibroblastos/citologia , Western Blotting , Linhagem Celular Transformada/metabolismo , Células Cultivadas , Técnicas de Cocultura , Citocinas/genética , Citocinas/metabolismo , Esôfago/metabolismo , Imunofluorescência , Humanos , Técnicas Imunoenzimáticas , Miofibroblastos/metabolismo , Fenótipo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Am J Physiol Gastrointest Liver Physiol ; 308(11): G904-23, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25882613

RESUMO

The pathophysiology of esophageal injury, repair, and inflammation in gastroesophageal reflux-disease (GERD) is complex. Whereas most studies have focused on the epithelial response to GERD injury, we are interested in the stromal response. We hypothesized that subepithelial esophageal myofibroblasts in GERD secrete proinflammatory cytokines in response to injurious agents encountered via epithelial barrier breaches or through dilated epithelial intercellular spaces. We determined the percentage of myofibroblasts [-smooth muscle actin (-SMA)+vimentin+CD31-] in the subepithelial GERD and normal esophageal stroma by immunomorphologic analysis. We performed -SMA coimmunostaining with IL-6 and p65. We established and characterized primary cultures of -SMA+vimentin+CD31-CD45- human esophageal myofibroblasts (HuEso MFs). We modeled GERD by treatment with pH 4.5-acidified media and Toll-like receptor 4 (TLR4) ligands, LPS and high-mobility group box 1 protein (HMGB1), and determined myofibroblast cytokine secretion in response to GERD injury. We demonstrate that spindle-shaped cell myofibroblasts are located near the basement membrane of stratified squamous epithelium in normal esophagus. We identify an increase in subepithelial myofibroblasts and activation of proinflammatory pathways in patients with GERD. Primary cultures of stromal cells obtained from normal esophagus retain myofibroblast morphology and express the acid receptor transient receptor potential channel vanilloid subfamily 1 (TRPV1) and TLR4. HuEso MFs stimulated with acid and TLR4 agonists LPS and HMGB1 increase IL-6 and IL-8 secretion via TRPV1 and NF-B activation. Our work implicates a role for human subepithelial stromal cells in the pathogenesis of GERD-related esophageal injury. Findings of this study can be extended to the investigation of epithelial-stromal interactions in inflammatory esophageal mucosal disorders.


Assuntos
Actinas/metabolismo , Esôfago , Refluxo Gastroesofágico , Interleucina-6 , Interleucina-8 , Miofibroblastos , Receptor 4 Toll-Like/metabolismo , Membrana Basal/metabolismo , Membrana Basal/patologia , Técnicas de Cultura de Células , Esôfago/metabolismo , Esôfago/patologia , Refluxo Gastroesofágico/metabolismo , Refluxo Gastroesofágico/patologia , Proteína HMGB1/metabolismo , Humanos , Inflamação/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Estimulação Química , Vimentina/metabolismo
16.
J Vis Exp ; (95): 52215, 2015 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-25650889

RESUMO

Murine and human esophageal myofibroblasts are generated via enzymatic digestion. Neonate (8-12 day old) murine esophagus is harvested, minced, washed, and subjected to enzymatic digestion with collagenase and dispase for 25 min. Human esophageal resection specimens are stripped of muscularis propria and adventitia and the remaining mucosa is minced, and subjected to enzymatic digestion with collagenase and dispase for up to 6 hr. Cultured cells express α-SMA and vimentin and express desmin weakly or not at all. Culture conditions are not conducive to growth of epithelial, hematopoietic, or endothelial cells. Culture purity is further confirmed by flow cytometric evaluation of cell surface marker expression of potential contaminating hematopoietic and endothelial cells. The described technique is straightforward and results in consistent generation of non-hematopoieitc, non-endothelial stromal cells. Limitations of this technique are inherent to the use of primary cultures in molecular biology studies, i.e., the unavoidable variability encountered among cultures established across different mice or humans. Primary cultures however are a more representative reflection of the in vivo state compared to cell lines. These methods also provide investigators the ability to isolate and culture stromal cells from different clinical and experimental conditions, allowing comparisons between groups. Characterized esophageal stromal cells can also be used in functional studies investigating epithelial-stromal interactions in esophageal disorders.


Assuntos
Esôfago/citologia , Miofibroblastos/citologia , Actinas/biossíntese , Animais , Biomarcadores/metabolismo , Linhagem Celular , Técnicas Citológicas/métodos , Desmina/biossíntese , Esôfago/metabolismo , Humanos , Camundongos , Músculo Liso/citologia , Músculo Liso/metabolismo , Miofibroblastos/metabolismo , Fenótipo , Células Estromais/citologia , Células Estromais/metabolismo , Vimentina/biossíntese
17.
Transl Res ; 164(1): 70-83, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24731292

RESUMO

Epimorphin (Epim), a member of the syntaxin family of membrane-bound, intracellular vesicle-docking proteins, is expressed in intestinal myofibroblasts and macrophages. We demonstrated previously that Epimorphin(-/-)(Epim(-/-)) mice are protected, in part, from dextran sodium sulfate (DSS)-induced colitis. Although interleukin (IL)-6/p-Stat3 signaling has been implicated in the pathogenesis of colitis, the myofibroblast contribution to IL-6 signaling in colitis remains unexplored. Our aim was to investigate the IL-6 pathway in Epim(-/-) mice in the DSS colitis model. Whole colonic tissue, epithelium, and stroma of WT and congenic Epim(-/-) mice treated with 5% DSS for 7 days were analyzed for IL-6 and a downstream effector, p-Stat3, by immunostaining and immunoblot. Colonic myofibroblast and peritoneal macrophage IL-6 secretion were evaluated by enzyme-linked immunosorbent assay. IL-6 and p-Stat3 expression were decreased in Epim(-/-) vs WT colon. A relative increase in stromal vs epithelial p-Stat3 expression was observed in WT mice but not in Epim(-/-) mice. Epim deletion abrogates IL-6 secretion from colonic myofibroblasts treated with IL-1ß and decreases IL-6 secretion from peritoneal macrophages in a subset of DSS-treated mice. Epim deletion inhibits IL-6 secretion most profoundly from colonic myofibroblasts. Distribution of Stat3 activation is altered in DSS-treated Epim(-/-) mice. Our findings support the notion that myofibroblasts modulate IL-6/p-Stat3 signaling in DSS-treated Epim(-/-) mice.


Assuntos
Colite/induzido quimicamente , Interleucina-6/metabolismo , Glicoproteínas de Membrana/metabolismo , Transdução de Sinais , Animais , Sulfato de Dextrana/toxicidade , Regulação da Expressão Gênica/fisiologia , Interleucina-6/genética , Mucosa Intestinal/patologia , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
18.
Am J Gastroenterol ; 108(11): 1706-12, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24019081

RESUMO

OBJECTIVES: Dysphagia may develop following antireflux surgery as a consequence of poor esophageal peristaltic reserve. We hypothesized that suboptimal contraction response following multiple rapid swallows (MRS) could be associated with chronic transit symptoms following antireflux surgery. METHODS: Wet swallow and MRS responses on esophageal high-resolution manometry (HRM) were characterized collectively in the esophageal body (distal contractile integral (DCI)), and individually in each smooth muscle contraction segment (S2 and S3 amplitudes) in 63 patients undergoing antireflux surgery and in 18 healthy controls. Dysphagia was assessed using symptom questionnaires. The MRS/wet swallow ratios were calculated for S2 and S3 peak amplitudes and DCI. MRS responses were compared in patients with and without late postoperative dysphagia following antireflux surgery. RESULTS: Augmentation of smooth muscle contraction (MRS/wet swallow ratios >1.0) as measured collectively by DCI was seen in only 11.1% with late postoperative dysphagia, compared with 63.6% in those with no dysphagia and 78.1% in controls (P≤0.02 for each comparison). Similar results were seen with S3 but not S2 peak amplitude ratios. Receiver operating characteristics identified a DCI MRS/wet swallow ratio threshold of 0.85 in segregating patients with late postoperative dysphagia from those with no postoperative dysphagia with a sensitivity of 0.67 and specificity of 0.64. CONCLUSIONS: Lack of augmentation of smooth muscle contraction following MRS is associated with late postoperative dysphagia following antireflux surgery, suggesting that MRS responses could assess esophageal smooth muscle peristaltic reserve. Further research is warranted to determine if antireflux surgery needs to be tailored to the MRS response.


Assuntos
Transtornos de Deglutição/diagnóstico , Deglutição/fisiologia , Esôfago/fisiopatologia , Fundoplicatura/efeitos adversos , Refluxo Gastroesofágico/cirurgia , Peristaltismo/fisiologia , Transtornos de Deglutição/etiologia , Transtornos de Deglutição/fisiopatologia , Refluxo Gastroesofágico/fisiopatologia , Humanos , Manometria , Contração Muscular/fisiologia , Músculo Liso/fisiopatologia
19.
Am J Physiol Gastrointest Liver Physiol ; 305(8): G564-72, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-23886856

RESUMO

Interactions between the epithelium and surrounding mesenchyme/stroma play an important role in normal gut morphogenesis, the epithelial response to injury, and epithelial carcinogenesis. The tumor microenvironment, composed of stromal cells including myofibroblasts and immune cells, regulates tumor growth and the cancer stem cell niche. Deletion of epimorphin (Epim), a syntaxin family member expressed in myofibroblasts and macrophages, results in partial protection from colitis and from inflammation-induced colon cancer in mice. We sought to determine whether epimorphin deletion protects from polyposis in the Apcmin/+ mouse model of intestinal carcinogenesis. Epim-/- mice were crossed to Apcmin/+ mice; Apcmin/+ and Apcmin/+/Epim-/- mice were killed at 3 mo of age. Polyp numbers and sizes were quantified in small intestine and colon, and gene expression analyses for pathways relevant to epithelial carcinogenesis were performed. Primary myofibroblast cultures were isolated, and expression and secretion of selected growth factors from Apcmin/+ and Apcmin/+/Epim-/- myofibroblasts were examined by ELISA. Small bowel polyposis was significantly inhibited in Apcmin/+/Epim-/- compared with Apcmin/+ mice. Apcmin/+/Epim-/- compared with Apcmin/+ polyps and adjacent uninvolved intestinal mucosa had increased transforming growth factor-ß (TGF-ß) expression and signaling with increased P-Smad2/3 expression. Myofibroblasts isolated from Apcmin/+/Epim-/- vs. Apcmin/+ mice had markedly decreased hepatocyte growth factor (HGF) expression and secretion. We concluded that Epim deletion inhibits polyposis in Apcmin/+ mice, associated with increased mucosal TGF-ß signaling and decreased myofibroblast HGF expression and secretion. Our data suggest that Epim deletion reduces tumorigenicity of the stromal microenvironment.


Assuntos
Proteína da Polipose Adenomatosa do Colo/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Glicoproteínas de Membrana/metabolismo , Miofibroblastos/metabolismo , Polipose Adenomatosa do Colo/genética , Polipose Adenomatosa do Colo/metabolismo , Polipose Adenomatosa do Colo/patologia , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Neoplasias do Colo/metabolismo , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Fator de Crescimento de Hepatócito/genética , Mucosa Intestinal/fisiologia , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
20.
Am J Physiol Gastrointest Liver Physiol ; 304(7): G662-72, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23370675

RESUMO

We identified α-smooth muscle actin (α-SMA)- and vimentin-expressing spindle-shaped esophageal mesenchymal cells in the adult and neonate murine esophageal lamina propria. We hypothesized that these esophageal mesenchymal cells express and secrete signaling and inflammatory mediators in response to injury. We established primary cultures of esophageal mesenchymal cells using mechanical and enzymatic digestion. We demonstrate that these primary cultures are nonhematopoietic, nonendothelial, stromal cells with myofibroblast-like features. These cells increase secretion of IL-6 in response to treatment with acidified media and IL-1ß. They also increase bone morphogenetic protein (Bmp)-4 secretion in response to sonic hedgehog. The location of these cells and their biological functions demonstrate their potential role in regulating esophageal epithelial responses to injury and repair.


Assuntos
Esôfago/metabolismo , Interleucina-1beta/farmacologia , Células-Tronco Mesenquimais/fisiologia , Miofibroblastos/fisiologia , Animais , Proteína Morfogenética Óssea 4 , Células Cultivadas , Esôfago/citologia , Interleucina-6/metabolismo , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA