Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 113
Filtrar
1.
Gastroenterology ; 164(4): 550-566, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36587707

RESUMEN

BACKGROUND & AIMS: Infection with Helicobacter pylori strongly affects global health by causing chronic gastritis, ulcer disease, and gastric cancer. Although extensive research into the strong immune response against this persistently colonizing bacterium exists, the specific role of CD8+ T cells remains elusive. METHODS: We comprehensively characterize gastric H pylori-specific CD8+ T-cell responses in mice and humans by flow cytometry, RNA-sequencing, immunohistochemistry, and ChipCytometry, applying functional analyses including T-cell depletion, H pylori eradication, and ex vivo restimulation. RESULTS: We define CD8+ T-cell populations bearing a tissue-resident memory (TRM) phenotype, which infiltrate the gastric mucosa shortly after infection and mediate pathogen control by executing antigen-specific effector properties. These induced CD8+ tissue-resident memory T cells (TRM cells) show a skewed T-cell receptor beta chain usage and are mostly specific for cytotoxin-associated gene A, the distinctive oncoprotein injected by H pylori into host cells. As the infection progresses, we observe a loss of the TRM phenotype and replacement of CD8+ by CD4+ T cells, indicating a shift in the immune response during the chronic infection phase. CONCLUSIONS: Our results point toward a hitherto unknown role of CD8+ T-cell response in this bacterial infection, which may have important clinical implications for treatment and vaccination strategies against H pylori.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Humanos , Animales , Ratones , Linfocitos T CD8-positivos , Linfocitos T CD4-Positivos , Estómago , Mucosa Gástrica/microbiología , Infecciones por Helicobacter/microbiología , Antígenos Bacterianos , Proteínas Bacterianas
2.
Gut ; 72(7): 1258-1270, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37015754

RESUMEN

OBJECTIVE: Helicobacter pylori infection is the most prevalent bacterial infection worldwide. Besides being the most important risk factor for gastric cancer development, epidemiological data show that infected individuals harbour a nearly twofold increased risk to develop colorectal cancer (CRC). However, a direct causal and functional connection between H. pylori infection and colon cancer is lacking. DESIGN: We infected two Apc-mutant mouse models and C57BL/6 mice with H. pylori and conducted a comprehensive analysis of H. pylori-induced changes in intestinal immune responses and epithelial signatures via flow cytometry, chip cytometry, immunohistochemistry and single cell RNA sequencing. Microbial signatures were characterised and evaluated in germ-free mice and via stool transfer experiments. RESULTS: H. pylori infection accelerated tumour development in Apc-mutant mice. We identified a unique H. pylori-driven immune alteration signature characterised by a reduction in regulatory T cells and pro-inflammatory T cells. Furthermore, in the intestinal and colonic epithelium, H. pylori induced pro-carcinogenic STAT3 signalling and a loss of goblet cells, changes that have been shown to contribute-in combination with pro-inflammatory and mucus degrading microbial signatures-to tumour development. Similar immune and epithelial alterations were found in human colon biopsies from H. pylori-infected patients. Housing of Apc-mutant mice under germ-free conditions ameliorated, and early antibiotic eradication of H. pylori infection normalised the tumour incidence to the level of uninfected controls. CONCLUSIONS: Our studies provide evidence that H. pylori infection is a strong causal promoter of colorectal carcinogenesis. Therefore, implementation of H. pylori status into preventive measures of CRC should be considered.


Asunto(s)
Neoplasias del Colon , Infecciones por Helicobacter , Helicobacter pylori , Microbiota , Neoplasias Gástricas , Humanos , Ratones , Animales , Helicobacter pylori/genética , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/microbiología , Ratones Endogámicos C57BL , Carcinogénesis/patología , Neoplasias Gástricas/patología , Neoplasias del Colon/patología , Moco , Mucosa Gástrica/patología
3.
Am J Physiol Gastrointest Liver Physiol ; 322(6): G583-G597, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35319286

RESUMEN

Intestinal ganglionic cells in the adult enteric nervous system (ENS) are continually exposed to stimuli from the surrounding microenvironment and need at times to respond to disturbed homeostasis following acute intestinal injury. The kinase DCLK1 and intestinal Dclk1-positive cells have been reported to contribute to intestinal regeneration. Although Dclk1-positive cells are present in adult enteric ganglia, their cellular identity and response to acute injury have not been investigated in detail. Here, we reveal the presence of distinct Dclk1-tdTom+/CD49b+ glial-like and Dclk1-tdTom+/CD49b- neuronal cell types in adult myenteric ganglia. These ganglionic cells demonstrate distinct patterns of tracing over time yet show a similar expansion in response to elevated serotonergic signaling. Interestingly, Dclk1-tdTom+ glial-like and neuronal cell types appear resistant to acute irradiation injury-mediated cell death. Moreover, Dclk1-tdTom+/CD49b+ glial-like cells show prominent changes in gene expression profiles induced by injury, in contrast to Dclk1-tdTom+/CD49b- neuronal cell types. Finally, subsets of Dclk1-tdTom+/CD49b+ glial-like cells demonstrate prominent overlap with Nestin and p75NTR and strong responses to elevated serotonergic signaling or acute injury. These findings, together with their role in early development and their neural crest-like gene expression signature, suggest the presence of reserve progenitor cells in the adult Dclk1 glial cell lineage.NEW & NOTEWORTHY The kinase DCLK1 identifies glial-like and neuronal cell types in adult murine enteric ganglia, which resist acute injury-mediated cell death yet differ in their cellular response to injury. Interestingly, Dclk1-labeled glial-like cells show prominent transcriptional changes in response to injury and harbor features reminiscent of previously described enteric neural precursor cells. Our data thus add to recently emerging evidence of reserve cellular plasticity in the adult enteric nervous system.


Asunto(s)
Sistema Nervioso Entérico , Células-Madre Neurales , Animales , Sistema Nervioso Entérico/fisiología , Integrina alfa2/metabolismo , Ratones , Ratones Transgénicos , Neuroglía/metabolismo , Neuronas/metabolismo
4.
Am J Gastroenterol ; 117(4): 603-604, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35103021

RESUMEN

ABSTRACT: T1 carcinoma is often not recognized as such, and inappropriate endoscopic resection techniques are selected, resulting in positive (R1) or nonassessable (Rx) resection margins. Full-thickness resection has been proposed as an alternative to completion surgery. Gijsbers et al. compared oncological outcomes of both strategies. The main finding was that colorectal cancer recurrence was significantly higher in the full-thickness excision of the scar compared with the completion surgery group (9.0% vs 2.2%). However, metastasis-free survival and overall survival were not significantly different in both groups. The results of this study favor full-thickness excision of the scar as the first-line approach for Rx/R1-resected margins but otherwise low-risk tumors.


Asunto(s)
Neoplasias Colorrectales , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/cirugía , Endoscopía , Humanos , Recurrencia Local de Neoplasia/cirugía , Estudios Retrospectivos
5.
Eur J Nucl Med Mol Imaging ; 49(6): 2049-2063, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34882260

RESUMEN

PURPOSE: The incidence of esophageal adenocarcinoma (EAC) has been increasing for decades without significant improvements in treatment. Barrett's esophagus (BE) is best established risk factor for EAC, but current surveillance with random biopsies cannot predict progression to cancer in most BE patients due to the low sensitivity and specificity of high-definition white light endoscopy. METHODS: Here, we evaluated the membrane-bound highly specific Hsp70-specific contrast agent Tumor-Penetrating Peptide (Hsp70-TPP) in guided fluorescence molecular endoscopy biopsy. RESULTS: Hsp70 was significantly overexpressed as determined by IHC in dysplasia and EAC compared with non-dysplastic BE in patient samples (n = 12) and in high-grade dysplastic lesions in a transgenic (L2-IL1b) mouse model of BE. In time-lapse microscopy, Hsp70-TPP was rapidly taken up and internalized  by human BE dysplastic patient-derived organoids. Flexible fluorescence endoscopy of the BE mouse model allowed a specific detection of Hsp70-TPP-Cy5.5 that corresponded closely with the degree of dysplasia but not BE. Ex vivo application of Hsp70-TPP-Cy5.5 to freshly resected whole human EAC specimens revealed a high (> 4) tumor-to-background ratio and a specific detection of previously undetected tumor infiltrations. CONCLUSION: In summary, these findings suggest that Hsp70-targeted imaging using fluorescently labeled TPP peptide may improve tumor surveillance in BE patients.


Asunto(s)
Adenocarcinoma , Esófago de Barrett , Neoplasias Esofágicas , Adenocarcinoma/patología , Animales , Esófago de Barrett/diagnóstico por imagen , Esófago de Barrett/epidemiología , Biopsia , Neoplasias Esofágicas/diagnóstico por imagen , Esofagoscopía/métodos , Humanos , Ratones
6.
Carcinogenesis ; 42(3): 405-413, 2021 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-33068426

RESUMEN

Chronic inflammation induces Barrett's Esophagus (BE) which can advance to esophageal adenocarcinoma. Elevated levels of interleukin (IL)-1b, IL-6 and IL-8 together with activated nuclear factor-kappaB (NF-κB), have been identified as important mediators of tumorigenesis. The inflammatory milieu apart from cancer cells and infiltrating immune cells contains myofibroblasts (MFs) that express aSMA and Vimentin. As we observed that increased NF-κB activation and inflammation correlates with increased MF recruitment and an accelerated phenotype we here analyze the role of NF-κB in MF during esophageal carcinogenesis in our L2-IL-1B mouse model. To analyze the effect of NF-κB signaling in MFs, we crossed L2-IL-1B mice to tamoxifen inducible Vim-Cre (Vim-CreTm) mice and floxed RelA (p65fl/fl) mice to specifically eliminate NF-κB signaling in MF (IL-1b.Vim-CreTm.p65fl/fl). The interaction of epithelial cells and stromal cells was further analyzed in mouse BE organoids and patient-derived human organoids. Histological scoring of IL-1b.Vim-CreTm.p65fl/fl mice showed a significantly attenuated phenotype compared with L2-IL-1B mice, with mild inflammation, decreased metaplasia and no dysplasia. This correlated with decreased proliferation and increased differentiation in cardia tissue of IL-1b.Vim-CreTm.p65fl/fl compared with L2-IL-1B mice. Distinct changes of cytokines and chemokines within the local microenvironment in IL-1b.Vim-CreTm.p65fl/fl mice reflected the histopathological abrogated phenotype. Co-cultured NF-κB inhibitor treated MF with mouse BE organoids demonstrated NF-κB-dependent growth and migration. MFs are essential to form an inflammatory and procarcinogenic microenvironment and NF-κB signaling in stromal cells emerges as an important driver of esophageal carcinogenesis. Our data suggest anti-inflammatory approaches as preventive strategies during surveillance of BE patients.


Asunto(s)
Adenocarcinoma/inmunología , Esófago de Barrett/inmunología , Transformación Celular Neoplásica/inmunología , Neoplasias Esofágicas/inmunología , Transducción de Señal/inmunología , Factor de Transcripción ReIA/metabolismo , Adenocarcinoma/patología , Adenocarcinoma/prevención & control , Animales , Antiinflamatorios/uso terapéutico , Esófago de Barrett/tratamiento farmacológico , Esófago de Barrett/patología , Biopsia , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Neoplasias Esofágicas/patología , Neoplasias Esofágicas/prevención & control , Esófago/inmunología , Esófago/patología , Humanos , Ratones , Ratones Noqueados , Miofibroblastos/inmunología , Miofibroblastos/patología , Organoides , Cultivo Primario de Células , Transducción de Señal/efectos de los fármacos , Células del Estroma/inmunología , Células del Estroma/patología , Factor de Transcripción ReIA/genética , Microambiente Tumoral/inmunología , Vimentina/metabolismo
7.
Carcinogenesis ; 42(8): 1068-1078, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-33878160

RESUMEN

Barrett's esophagus (BE) is the main known precursor condition of esophageal adenocarcinoma (EAC). BE is defined by the presence of metaplasia above the normal squamous columnar junction and has mainly been attributed to gastroesophageal reflux disease and chronic reflux esophagitis. Thus, the rising incidence of EAC in the Western world is probably mediated by chronic esophageal inflammation, secondary to gastroesophageal reflux disease in combination with environmental risk factors such as a Western diet and obesity. However, (at present) risk prediction tools and endoscopic surveillance have shown limited effectiveness. Chemoprevention as an adjunctive approach remains an attractive option to reduce the incidence of neoplastic disease. Here, we investigate the feasibility of chemopreventive approaches in BE and EAC via inhibition of inflammatory signaling in a transgenic mouse model of BE and EAC (L2-IL1B mice), with accelerated tumor formation on a high-fat diet (HFD). L2-IL1B mice were treated with the IL-1 receptor antagonist Anakinra and the nonsteroidal anti-inflammatory drugs (NSAIDs) aspirin or Sulindac. Interleukin-1b antagonism reduced tumor progression in L2-IL1B mice with or without a HFD, whereas both NSAIDs were effective chemoprevention agents in the accelerated HFD-fed L2-IL1B mouse model. Sulindac treatment also resulted in a marked change in the immune profile of L2-IL1B mice. In summary, anti-inflammatory treatment of HFD-treated L2-IL1B mice acted protectively on disease progression. These results from a mouse model of BE support results from clinical trials that suggest that anti-inflammatory medication may be effective in the chemoprevention of EAC.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Quimioprevención/métodos , Sulindac/uso terapéutico , Adenocarcinoma , Animales , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Neoplasias Esofágicas , Ratones , Fenotipo
8.
Gastroenterology ; 159(2): 575-590, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32325086

RESUMEN

BACKGROUND & AIMS: Studies are needed to determine the mechanism by which Barrett's esophagus (BE) progresses to esophageal adenocarcinoma (EAC). Notch signaling maintains stem cells in the gastrointestinal tract and is dysregulated during carcinogenesis. We explored the relationship between Notch signaling and goblet cell maturation, a feature of BE, during EAC pathogenesis. METHODS: We measured goblet cell density and levels of Notch messenger RNAs in BE tissues from 164 patients, with and without dysplasia or EAC, enrolled in a multicenter study. We analyzed the effects of conditional expression of an activated form of NOTCH2 (pL2.Lgr5.N2IC), conditional deletion of NOTCH2 (pL2.Lgr5.N2fl/fl), or loss of nuclear factor κB (NF-κB) (pL2.Lgr5.p65fl/fl), in Lgr5+ (progenitor) cells in L2-IL1B mice (which overexpress interleukin 1 beta in esophagus and squamous forestomach and are used as a model of BE). We collected esophageal and stomach tissues and performed histology, immunohistochemistry, flow cytometry, transcriptome, and real-time polymerase chain reaction analyses. Cardia and forestomach tissues from mice were cultured as organoids and incubated with inhibitors of Notch or NF-kB. RESULTS: Progression of BE to EAC was associated with a significant reduction in goblet cell density comparing nondysplastic regions of tissues from patients; there was an inverse correlation between goblet cell density and levels of NOTCH3 and JAG2 messenger RNA. In mice, expression of the activated intracellular form of NOTCH2 in Lgr5+ cells reduced goblet-like cell maturation, increased crypt fission, and accelerated the development of tumors in the squamocolumnar junction. Mice with deletion of NOTCH2 from Lgr5+ cells had increased maturation of goblet-like cells, reduced crypt fission, and developed fewer tumors. Esophageal tissues from in pL2.Lgr5.N2IC mice had increased levels of RelA (which encodes the p65 unit of NF-κB) compared to tissues from L2-IL1B mice, and we found evidence of increased NF-κB activity in Lgr5+ cells. Esophageal tissues from pL2.Lgr5.p65fl/fl mice had lower inflammation and metaplasia scores than pL2.Lgr5.N2IC mice. In organoids derived from pL2-IL1B mice, the NF-κB inhibitor JSH-23 reduced cell survival and proliferation. CONCLUSIONS: Notch signaling contributes to activation of NF-κB and regulates differentiation of gastric cardia progenitor cells in a mouse model of BE. In human esophageal tissues, progression of BE to EAC was associated with reduced goblet cell density and increased levels of Notch expression. Strategies to block this pathway might be developed to prevent EAC in patients with BE.


Asunto(s)
Adenocarcinoma/patología , Esófago de Barrett/patología , Carcinogénesis/patología , Neoplasias Esofágicas/patología , Células Caliciformes/patología , Receptores Notch/metabolismo , Adenocarcinoma/diagnóstico , Adenocarcinoma/genética , Anciano , Animales , Esófago de Barrett/diagnóstico , Esófago de Barrett/genética , Biopsia , Carcinogénesis/genética , Diferenciación Celular/genética , Estudios Transversales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Mucosa Esofágica/citología , Mucosa Esofágica/diagnóstico por imagen , Mucosa Esofágica/patología , Neoplasias Esofágicas/diagnóstico , Neoplasias Esofágicas/genética , Esofagoscopía , Femenino , Mucosa Gástrica/citología , Mucosa Gástrica/patología , Humanos , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , FN-kappa B/metabolismo , Estudios Prospectivos , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Receptores Notch/genética , Transducción de Señal
9.
Z Gastroenterol ; 59(9): 964-982, 2021 Sep.
Artículo en Alemán | MEDLINE | ID: mdl-34507375

RESUMEN

Throughout the past decades, considerable progress has been made in the (early) diagnosis and treatment of gastrointestinal cancers. However, the prognosis for advanced stages of gastrointestinal tumors remains limited for many patients and approximately one third of all tumor patients die as a result of gastrointestinal tumors. The prevention and early detection of gastrointestinal tumors is therefore of great importance.For this reason, we summarize the current state of knowledge and recommendations for the primary, secondary and tertiary prevention of esophageal, stomach, pancreas, liver and colorectal cancer in the following.


Asunto(s)
Neoplasias Esofágicas , Neoplasias Gastrointestinales , Neoplasias Gástricas , Tracto Gastrointestinal Superior , Neoplasias Esofágicas/diagnóstico , Neoplasias Esofágicas/prevención & control , Neoplasias Gastrointestinales/diagnóstico , Neoplasias Gastrointestinales/prevención & control , Humanos , Páncreas , Pronóstico
10.
Gut ; 69(9): 1598-1607, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-31857433

RESUMEN

OBJECTIVE: Gastrointestinal microbiota may be involved in Helicobacter pylori-associated gastric cancer development. The aim of this study was to explore the possible microbial mechanisms in gastric carcinogenesis and potential dysbiosis arising from H. pylori infection. DESIGN: Deep sequencing of the microbial 16S ribosomal RNA gene was used to investigate alterations in paired gastric biopsies and stool samples in 58 subjects with successful and 57 subjects with failed anti-H. pylori treatment, relative to 49 H. pylori negative subjects. RESULTS: In H. pylori positive subjects, richness and Shannon indexes increased significantly (both p<0.001) after successful eradication and showed no difference to those of negative subjects (p=0.493 for richness and p=0.420 for Shannon index). Differential taxa analysis identified 18 significantly altered gastric genera after eradication. The combination of these genera into a Microbial Dysbiosis Index revealed that the dysbiotic microbiota in H. pylori positive mucosa was associated with advanced gastric lesions (chronic atrophic gastritis and intestinal metaplasia/dysplasia) and could be reversed by eradication. Strong coexcluding interactions between Helicobacter and Fusobacterium, Neisseria, Prevotella, Veillonella, Rothia were found only in advanced gastric lesion patients, and were absent in normal/superficial gastritis group. Changes in faecal microbiota included increased Bifidobacterium after successful H. pylori eradication and more upregulated drug-resistant functional orthologs after failed treatment. CONCLUSION: H. pylori infection contributes significantly to gastric microbial dysbiosis that may be involved in carcinogenesis. Successful H. pylori eradication potentially restores gastric microbiota to a similar status as found in uninfected individuals, and shows beneficial effects on gut microbiota.


Asunto(s)
Disbiosis , Gastritis Atrófica , Microbioma Gastrointestinal/genética , Infecciones por Helicobacter , Helicobacter pylori , Neoplasias Gástricas , Antibacterianos/uso terapéutico , Biopsia/métodos , Disbiosis/diagnóstico , Disbiosis/microbiología , Heces/microbiología , Femenino , Mucosa Gástrica/microbiología , Mucosa Gástrica/patología , Gastritis Atrófica/microbiología , Gastritis Atrófica/patología , Infecciones por Helicobacter/tratamiento farmacológico , Infecciones por Helicobacter/patología , Helicobacter pylori/aislamiento & purificación , Helicobacter pylori/patogenicidad , Humanos , Masculino , Metaplasia/microbiología , Metaplasia/patología , Interacciones Microbianas , Persona de Mediana Edad , ARN Ribosómico 16S/aislamiento & purificación , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/patología
11.
Gastroenterology ; 157(2): 492-506.e2, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30998992

RESUMEN

BACKGROUND & AIMS: Barrett's esophagus (BE) is a precursor to esophageal adenocarcinoma (EAC). Progression from BE to cancer is associated with obesity, possibly due to increased abdominal pressure and gastroesophageal reflux disease, although this pathogenic mechanism has not been proven. We investigated whether environmental or dietary factors associated with obesity contribute to the progression of BE to EAC in mice. METHODS: Tg(ED-L2-IL1RN/IL1B)#Tcw mice (a model of BE, called L2-IL1B mice) were fed a chow (control) or high-fat diet (HFD) or were crossbred with mice that express human interleukin (IL) 8 (L2-IL1B/IL8 mice). Esophageal tissues were collected and analyzed for gene expression profiles and by quantitative polymerase chain reaction, immunohistochemistry, and flow cytometry. Organoids were established from BE tissue of mice and cultured with serum from lean or obese individuals or with neutrophils from L2-IL1B mice. Feces from mice were analyzed by 16s ribosomal RNA sequencing and compared to 16s sequencing data from patients with dysplasia or BE. L2-IL1B were mice raised in germ-free conditions. RESULTS: L2-IL1B mice fed an HFD developed esophageal dysplasia and tumors more rapidly than mice fed the control diet; the speed of tumor development was independent of body weight. The acceleration of dysplasia by the HFD in the L2-IL1B mice was associated with a shift in the gut microbiota and an increased ratio of neutrophils to natural killer cells in esophageal tissues compared with mice fed a control diet. We observed similar differences in the microbiomes from patients with BE that progressed to EAC vs patients with BE that did not develop into cancer. Tissues from dysplasias of L2-IL1B mice fed the HFD contained increased levels of cytokines that are produced in response to CXCL1 (the functional mouse homolog of IL8, also called KC). Serum from obese patients caused organoids from L2-IL1B/IL8 mice to produce IL8. BE tissues from L2-IL1B mice fed the HFD and from L2-IL1B/IL8 mice contained increased numbers of myeloid cells and cells expressing Cxcr2 and Lgr5 messenger RNAs (epithelial progenitors) compared with mice fed control diets. BE tissues from L2-IL1B mice raised in germ-free housing had fewer progenitor cells and developed less dysplasia than in L2-IL1 mice raised under standard conditions; exposure of fecal microbiota from L2-IL1B mice fed the HFD to L2-IL1B mice fed the control diet accelerated tumor development. CONCLUSIONS: In a mouse model of BE, we found that an HFD promoted dysplasia by altering the esophageal microenvironment and gut microbiome, thereby inducing inflammation and stem cell expansion, independent of obesity.


Asunto(s)
Adenocarcinoma/patología , Esófago de Barrett/patología , Neoplasias Esofágicas/patología , Microbioma Gastrointestinal/fisiología , Interleucina-8/metabolismo , Obesidad/patología , Adenocarcinoma/inmunología , Adulto , Anciano , Animales , Esófago de Barrett/inmunología , Carcinogénesis/inmunología , Carcinogénesis/patología , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Neoplasias Esofágicas/inmunología , Esófago/inmunología , Esófago/patología , Heces/microbiología , Femenino , Voluntarios Sanos , Humanos , Interleucina-8/genética , Interleucina-8/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Obesidad/sangre , Obesidad/inmunología , Organoides , Suero/inmunología , Suero/metabolismo , Factores de Tiempo , Técnicas de Cultivo de Tejidos
12.
Cancer Immunol Immunother ; 69(12): 2635-2649, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32613271

RESUMEN

Barrett's esophagus (BE) is an inflammatory condition and a neoplastic precursor to esophageal adenocarcinoma (EAC). Inflammasome signaling, which contributes to acute and chronic inflammation, results in caspase-1 activation leading to the secretion of IL-1ß and IL-18, and inflammatory cell death (pyroptosis). This study aimed to characterize caspase-1 expression, and its functional importance, during disease progression to BE and EAC. Three models of disease progression (Normal-BE-EAC) were employed to profile caspase-1 expression: (1) a human esophageal cell line model; (2) a murine model of BE; and (3) resected tissue from BE-associated EAC patients. BE patient biopsies and murine BE organoids were cultured ex vivo in the presence of a caspase-1 inhibitor, to determine the importance of caspase-1 for inflammatory cytokine and chemokine secretion.Epithelial caspase-1 expression levels were significantly enhanced in BE (p < 0.01). In contrast, stromal caspase-1 levels correlated with histological inflammation scores during disease progression (p < 0.05). Elevated secretion of IL-1ß from BE explanted tissue, compared to adjacent normal tissue (p < 0.01), confirmed enhanced activity of caspase-1 in BE tissue. Caspase-1 inhibition in LPS-stimulated murine BE organoids caused a significant reduction in IL-1ß (p < 0.01) and CXCL1 (p < 0.05) secretion, confirming the importance of caspase-1 in the production of cytokines and chemokines associated with disease progression from BE to EAC. Targeting caspase-1 activity in BE patients should therefore be tested as a novel strategy to prevent inflammatory complications associated with disease progression.


Asunto(s)
Adenocarcinoma/inmunología , Esófago de Barrett/inmunología , Caspasa 1/metabolismo , Mucosa Esofágica/patología , Neoplasias Esofágicas/inmunología , Inflamasomas/inmunología , Adenocarcinoma/patología , Adenocarcinoma/cirugía , Anciano , Animales , Esófago de Barrett/genética , Esófago de Barrett/patología , Biopsia , Caspasa 1/inmunología , Inhibidores de Caspasas/farmacología , Línea Celular Tumoral , Células Cultivadas , Quimiocina CXCL1/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Mucosa Esofágica/citología , Mucosa Esofágica/inmunología , Neoplasias Esofágicas/patología , Neoplasias Esofágicas/cirugía , Esofagectomía , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Inflamasomas/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Cultivo Primario de Células , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
13.
Br J Cancer ; 121(12): 1050-1057, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31690830

RESUMEN

BACKGROUND: Cellular Dissociation Grade (CDG) composed of tumour budding and cell nest size has been shown to independently predict prognosis in pre-therapeutic biopsies and primary resections of oesophageal squamous cell carcinoma (ESCC). Here, we aimed to evaluate the prognostic impact of CDG in ESCC after neoadjuvant therapy. METHODS: We evaluated cell nest size and tumour budding activity in 122 post-neoadjuvant ESCC resections, correlated the results with tumour regression groups and patient survival and compared the results with data from primary resected cases as well as pre-therapeutic biopsies. RESULTS: CDG remained stable when results from pre-therapeutic biopsies and post-therapeutic resections from the same patient were compared. CDG was associated with therapy response and a strong predictor of overall, disease-specific (DSS) and disease-free (DFS) survival in univariate analysis and-besides metastasis-remained the only significant survival predictor for DSS and DFS in multivariate analysis. Multivariate DFS hazard ratios reached 3.3 for CDG-G2 and 4.9 for CDG-G3 neoplasms compared with CDG-G1 carcinomas (p = 0.016). CONCLUSIONS: CDG is the only morphology-based grading algorithm published to date, which in concert with regression grading, is able to contribute relevant prognostic information in the post-neoadjuvant setting of ESCC.


Asunto(s)
Tamaño de la Célula , Carcinoma de Células Escamosas de Esófago/tratamiento farmacológico , Carcinoma de Células Escamosas de Esófago/patología , Pronóstico , Adulto , Anciano , Anciano de 80 o más Años , Biopsia , Supervivencia sin Enfermedad , Carcinoma de Células Escamosas de Esófago/epidemiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Terapia Neoadyuvante/métodos , Clasificación del Tumor , Metástasis de la Neoplasia , Modelos de Riesgos Proporcionales
14.
Gastroenterology ; 154(2): 406-420, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29037468

RESUMEN

Although researchers have identified genetic alterations that contribute to development of esophageal adenocarcinoma, we know little about features of patients or environmental factors that mediate progression of chronic acid biliary reflux to Barrett's esophagus and cancer. Increasing our understanding of the mechanisms by which normal squamous epithelium progresses to early-stage invasive cancer will help formulate rational surveillance guidelines and allow us to divest resources away from patients at low risk of malignancy. We review the cellular and genetic alterations that occur during progression of Barrett's esophagus, based on findings from clinical studies and mouse models of disease. We review the features of the luminal and mucosal microenvironment of Barrett's esophagus that promote, in a small proportion of patients, development of esophageal adenocarcinoma. Markers of clonal evolution can be used to determine patient risk for cancer and set surveillance intervals.


Asunto(s)
Adenocarcinoma/etiología , Esófago de Barrett/etiología , Carcinogénesis/patología , Neoplasias Esofágicas/etiología , Reflujo Gastroesofágico/etiología , Microambiente Tumoral , Adenocarcinoma/epidemiología , Adenocarcinoma/patología , Animales , Esófago de Barrett/patología , Transformación Celular Neoplásica/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Exposición a Riesgos Ambientales/efectos adversos , Mucosa Esofágica/patología , Neoplasias Esofágicas/epidemiología , Neoplasias Esofágicas/patología , Reflujo Gastroesofágico/patología , Microbioma Gastrointestinal , Genes Supresores de Tumor , Infecciones por Helicobacter/epidemiología , Infecciones por Helicobacter/microbiología , Humanos , Incidencia , Interleucinas/metabolismo , Ratones , Mucinas/metabolismo
15.
Dis Esophagus ; 32(8)2019 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-31329831

RESUMEN

Risk stratification in patients with Barrett's esophagus (BE) to prevent the development of esophageal adenocarcinoma (EAC) is an unsolved task. The incidence of EAC and BE is increasing and patients are still at unknown risk. BarrettNET is an ongoing multicenter prospective cohort study initiated to identify and validate molecular and clinical biomarkers that allow a more personalized surveillance strategy for patients with BE. For BarrettNET participants are recruited in 20 study centers throughout Germany, to be followed for progression to dysplasia (low-grade dysplasia or high-grade dysplasia) or EAC for >10 years. The study instruments comprise self-administered epidemiological information (containing data on demographics, lifestyle factors, and health), as well as biological specimens, i.e., blood-based samples, esophageal tissue biopsies, and feces and saliva samples. In follow-up visits according to the individual surveillance plan of the participants, sample collection is repeated. The standardized collection and processing of the specimen guarantee the highest sample quality. Via a mobile accessible database, the documentation of inclusion, epidemiological data, and pathological disease status are recorded subsequently. Currently the BarrettNET registry includes 560 participants (23.1% women and 76.9% men, aged 22-92 years) with a median follow-up of 951 days. Both the design and the size of BarrettNET offer the advantage of answering research questions regarding potential causes of disease progression from BE to EAC. Here all the integrated methods and materials of BarrettNET are presented and reviewed to introduce this valuable German registry.


Asunto(s)
Adenocarcinoma/diagnóstico , Esófago de Barrett/complicaciones , Detección Precoz del Cáncer/métodos , Neoplasias Esofágicas/diagnóstico , Vigilancia de la Población/métodos , Medición de Riesgo/métodos , Adenocarcinoma/etiología , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/análisis , Reglas de Decisión Clínica , Progresión de la Enfermedad , Neoplasias Esofágicas/etiología , Femenino , Alemania , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Sistema de Registros , Factores de Riesgo , Adulto Joven
17.
Cells Tissues Organs ; 206(1-2): 35-45, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30630170

RESUMEN

In-depth knowledge of the mechanisms induced by early postischemic cardiac endogenous mesenchymal stem cells (MSCs) in the acutely ischemic heart could advance our understanding of cardiac regeneration. Herein, we aimed to identify, isolate, and initially characterize the origin, kinetics and fate of cardiac MSCs. This was facilitated by in vivo genetic cell fate mapping through green fluorescent protein (GFP) expression under the control of vimentin induction after acute myocardial infarction (MI). Following permanent ligation of the left anterior descending coronary artery in CreER+ mTom/mGFP+ mice, vimentin/GFP+ cells revealed ischemia-responsive activation, survival, and local enrichment inside the peri-infarction border zone. Fluorescence-activated cell sorting (FACS)-isolated vimentin/GFP+ cells could be strongly expanded in vitro with clonogenic precursor formation and revealed MSC-typical cell morphology. Flow-cytometric analyses demonstrated an increase in cardiac vimentin/GFP+ cells in the ischemic heart, from a 0.6% cardiac mononuclear cell (MNC) fraction at 24 h to 1.6% at 72 h following MI. Sca-1+CD45- cells within the vimentin/GFP+ subtype of this MNC fraction increased from 35.2% at 24 h to 74.6% at 72 h after MI. The cardiac postischemic vimentin/GFP+ MNC subtype showed multipotent adipogenic, chondrogenic, and osteogenic differentiation potential, which is distinctive for MSCs. In conclusion, we demonstrated a seemingly proliferative first response of vimentin- induced cardiac endogenous MSCs in the acutely ischemic heart. Genetically, GFP-targeted in vivo cell tracking, isolation, and in vitro expansion of this cardiac MSC subtype could help to clarify their reparative status in inflammation, fibrogenesis, cell turnover, tissue homeostasis, and myocardial regeneration.


Asunto(s)
Células Madre Mesenquimatosas/citología , Infarto del Miocardio/patología , Miocardio/patología , Vimentina/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Separación Celular , Supervivencia Celular , Células Cultivadas , Femenino , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/metabolismo , Antígenos Comunes de Leucocito/análisis , Antígenos Comunes de Leucocito/metabolismo , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Vimentina/análisis
18.
Dig Dis Sci ; 63(8): 2013-2021, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29948557

RESUMEN

The incidence of esophageal adenocarcinoma (EAC) and its precursor lesion Barrett's esophagus (BE) has been increasing steadily in the western world in recent decades. Understanding the cellular origins of BE and the conditions responsible for their malignant transformation would greatly facilitate risk assessment and identification of patients at risk of progression, but this topic remains a source of debate. Here, we review recent findings that have provided support for the gastroesophageal junction (GEJ) as the main source of stem cells that give rise to BE and EAC. These include both gastric cardia cells and transitional basal cells. Furthermore, we discuss the role of chronic injury and inflammation in a tumor microenvironment as a major factor in promoting stem cell expansion and proliferation as well as transformation of the GEJ-derived stem cells and progression to EAC. We conclude that there exists a large amount of empirical support for the GEJ as the likely source of BE stem cells. While BE seems to resemble a successful adaptation to esophageal damage, carcinogenesis appears as a consequence of natural selection at the level of GEJ stem cells, and later glands, that expand into the esophagus wherein the local ecology creates the selective landscape for cancer progression.


Asunto(s)
Adenocarcinoma , Esófago de Barrett , Neoplasias Esofágicas , Unión Esofagogástrica/patología , Células Madre , Adenocarcinoma/etiología , Adenocarcinoma/patología , Esófago de Barrett/complicaciones , Esófago de Barrett/patología , Progresión de la Enfermedad , Neoplasias Esofágicas/etiología , Neoplasias Esofágicas/patología , Humanos , Metaplasia , Células Madre/patología , Células Madre/fisiología
19.
Am J Physiol Gastrointest Liver Physiol ; 313(4): G285-G299, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28684459

RESUMEN

Dclk1-expressing tuft cells constitute a unique intestinal epithelial lineage that is distinct from enterocytes, Paneth cells, goblet cells, and enteroendocrine cells. Tuft cells express taste-related receptors and distinct transcription factors and interact closely with the enteric nervous system, suggesting a chemosensory cell lineage. In addition, recent work has shown that tuft cells interact closely with cells of the immune system, with a critical role in the cellular regulatory network governing responses to luminal parasites. Importantly, ablation of tuft cells severely impairs epithelial proliferation and tissue regeneration after injury, implicating tuft cells in the modulation of epithelial stem/progenitor function. Finally, tuft cells expand during chronic inflammation and in preneoplastic tissues, suggesting a possible early role in inflammation-associated tumorigenesis. Hence, we outline and discuss emerging evidence that strongly supports tuft cells as key regulatory cells in the complex network of the intestinal microenvironment.


Asunto(s)
Microambiente Celular/fisiología , Células Quimiorreceptoras/fisiología , Mucosa Intestinal/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células Madre Multipotentes/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Nicho de Células Madre/fisiología , Animales , Quinasas Similares a Doblecortina , Humanos , Modelos Biológicos , Regeneración/fisiología
20.
Gut ; 65(1): 9-18, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25986943

RESUMEN

OBJECTIVE: To clarify the full range of benefits and adverse consequences of Helicobacter pylori eradication as a strategy for gastric cancer prevention, the community-based intervention trial was launched in Linqu County, China. DESIGN: A total of 184,786 residents aged 25-54 years were enrolled in this trial and received (13)C-urea breath test. H. pylori positive participants were assigned into two groups, either receiving a 10-day quadruple anti-H. pylori treatment or lookalike placebos together with a single dosage of omeprazole and bismuth. RESULTS: The prevalence of H. pylori in trial participants was 57.6%. A total of 94,101 subjects completed the treatment. The overall H. pylori eradication rate was 72.9% in the active group. Gender, body mass index, history of stomach disease, baseline delta over baseline-value of (13)C-urea breath test, missed medication doses, smoking and drinking were independent predictors of eradication failure. The missed doses and high baseline delta over baseline-value were important contributors in men and women (all Ptrend<0.001). However, a dose-response relationship between failure rate and smoking or drinking index was found in men (all Ptrend<0.001), while high body mass index (Ptrend<0.001) and history of stomach disease were significant predictors in women. The treatment failure rate increased up to 48.8% (OR 2.87, 95% CI 2.24 to 3.68) in men and 39.4% (OR 2.67, 95% CI 1.61 to 4.42) in women with multiple factors combined. CONCLUSIONS: This large community-based intervention trial to eradicate H. pylori is feasible and acceptable. The findings of this trial lead to a distinct evaluation of factors influencing eradication that should be generally considered for future eradication therapies. TRIAL REGISTRATION NUMBER: ChiCTR-TRC-10000979 in accordance with WHO ICTRP requirements.


Asunto(s)
Antibacterianos/uso terapéutico , Infecciones por Helicobacter/tratamiento farmacológico , Helicobacter pylori , Neoplasias Gástricas/prevención & control , Adulto , Antiulcerosos/uso terapéutico , China , Método Doble Ciego , Quimioterapia Combinada , Femenino , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/diagnóstico , Humanos , Masculino , Metronidazol/uso terapéutico , Persona de Mediana Edad , Estudios Prospectivos , Neoplasias Gástricas/microbiología , Tetraciclina/uso terapéutico , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda