Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Pathol ; 259(4): 362-368, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36625379

RESUMEN

Most gastric cancers develop in inflamed gastric mucosa due to Helicobacter pylori infection, typically with metaplastic changes. However, the origins of gastric cancer remain unknown. Here, we present a case of intramucosal gastric carcinoma (IGC) and oxyntic gland adenoma (OGA) derived from spasmolytic polypeptide-expressing metaplasia (SPEM). Early gastric cancer adjacent to a polyp was found in the upper corpus of a 71-year-old woman without H. pylori infection and was endoscopically resected. Histological examination showed IGC and OGA, both of which had predominant MUC6 expression. Interestingly, gastric glands with enriched MUC6-positive mucous cells, referred to as SPEM, expanded between them. Whole-exome sequencing analysis revealed a truncating KRAS(G12D) mutation in IGC, OGA, and SPEM. In addition, TP53 and CDKN2A mutations and a loss of chromosome 17p were found in the IGC, whereas a GNAS mutation was observed in the OGA. These results indicated that IGC and OGA originated from the KRAS-mutated SPEM. © 2023 The Pathological Society of Great Britain and Ireland.


Asunto(s)
Adenoma , Carcinoma , Infecciones por Helicobacter , Helicobacter pylori , Neoplasias Gástricas , Femenino , Humanos , Anciano , Neoplasias Gástricas/genética , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Mucosa Gástrica , Metaplasia , Adenoma/genética
2.
EMBO Rep ; 22(9): e51806, 2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34309175

RESUMEN

Differentiated cells across multiple species and organs can re-enter the cell cycle to aid in injury-induced tissue regeneration by a cellular program called paligenosis. Here, we show that activating transcription factor 3 (ATF3) is induced early during paligenosis in multiple cellular contexts, transcriptionally activating the lysosomal trafficking gene Rab7b. ATF3 and RAB7B are upregulated in gastric and pancreatic digestive-enzyme-secreting cells at the onset of paligenosis Stage 1, when cells massively induce autophagic and lysosomal machinery to dismantle differentiated cell morphological features. Their expression later ebbs before cells enter mitosis during Stage 3. Atf3-/- mice fail to induce RAB7-positive autophagic and lysosomal vesicles, eventually causing increased death of cells en route to Stage 3. Finally, we observe that ATF3 is expressed in human gastric metaplasia and during paligenotic injury across multiple other organs and species. Thus, our findings indicate ATF3 is an evolutionarily conserved gene orchestrating the early paligenotic autodegradative events that must occur before cells are poised to proliferate and contribute to tissue repair.


Asunto(s)
Factor de Transcripción Activador 3 , Plasticidad de la Célula , Factor de Transcripción Activador 3/genética , Animales , Ciclo Celular , Diferenciación Celular , Metaplasia/genética , Ratones
3.
J Med Genet ; 57(6): 385-388, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-30877236

RESUMEN

BACKGROUND: Spasmolytic polypeptide-expressing metaplasia (SPEM) is present in more than 90% of resected gastric cancer tissues. However, although widely regarded as a pre-cancerous tissue, its genetic characteristics have not been well studied. METHODS: Immunohistochemistry using Trefoil factor 2 (TFF2) antibodies was used to identify TFF2-positive SPEM cells within SPEM glands in the stomach of Helicobacter felis (H. felis) -infected mice and human clinical samples. Laser microdissection was used to isolate specific cells from both the infected mice and the human samples. The genetic instability in these cells was examined by measuring the lengths of microsatellite (MS) markers using capillary electrophoresis. Also, genome-wide genetic variations in the SPEM cells from the clinical sample was examined using deep whole-exome sequencing. RESULTS: SPEM cells indeed exhibit not only heightened MS instability (MSI), but also genetic instabilities that extend genome-wide. Furthermore, surprisingly, we found that morphologically normal, TFF2-negative cells also contain a comparable degree of genomic instabilities as the co-resident SPEM cells within the SPEM glands. CONCLUSION: These results, for the first time, clearly establish elevated genetic instability as a critical property of SPEM glands, which may provide a greater possibility for malignant clone selection. More importantly, these results indicate that SPEM cells may not be the sole origin of carcinogenesis in the stomach and strongly suggest the common progenitor of these cells, the stem cells, as the source of these genetic instabilities, and thus, potential key players in carcinogenesis.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/genética , Metaplasia/genética , Neoplasias Gástricas/genética , Factor Trefoil-2/genética , Animales , Femenino , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patología , Regulación Neoplásica de la Expresión Génica/genética , Inestabilidad Genómica/genética , Xenoinjertos , Humanos , Masculino , Metaplasia/patología , Ratones , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , Neoplasias Gástricas/patología
4.
Am J Physiol Cell Physiol ; 319(6): C947-C954, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32755448

RESUMEN

Spasmolytic polypeptide/trefoil factor 2 (TFF2)-expressing metaplasia (SPEM) is a mucous-secreting reparative lineage that emerges at the ulcer margin in response to gastric injury. Under conditions of chronic inflammation with parietal cell loss, SPEM has been found to emerge and evolve into neoplasia. Cluster-of-differentiation gene 44 (CD44) is known to coordinate normal and metaplastic epithelial cell proliferation. In particular, CD44 variant isoform 9 (CD44v9) associates with the cystine-glutamate transporter xCT, stabilizes the protein, and provides defense against reactive oxygen species (ROS). xCT stabilization by CD44v9 leads to defense against ROS by cystine uptake, glutathione (GSH) synthesis, and maintenance of the redox balance within the intracellular environment. Furthermore, p38 signaling is a known downstream ROS target, leading to diminished cell proliferation and migration, two vital processes of gastric epithelial repair. CD44v9 emerges during repair of the gastric epithelium after injury, where it is coexpressed with other markers of SPEM. The regulatory mechanisms for the emergence of CD44v9 and the role of CD44v9 during the process of gastric epithelial regeneration are largely unknown. Inflammation and M2 macrophage infiltration have recently been demonstrated to play key roles in the induction of SPEM after injury. The following review proposes new insights into the functional role of metaplasia in the process of gastric regeneration in response to ulceration. Our insights are extrapolated from documented studies reporting oxyntic atrophy and SPEM development and our current unpublished findings using the acetic acid-induced gastric injury model.


Asunto(s)
Mucosa Gástrica/patología , Metaplasia/patología , Regeneración/fisiología , Estómago/patología , Estómago/fisiología , Ácido Acético/efectos adversos , Animales , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/fisiología , Humanos , Estómago/efectos de los fármacos
5.
Biochem Biophys Res Commun ; 524(3): 649-655, 2020 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-32033746

RESUMEN

RATIONALE: Spasmolytic polypeptide-expressing metaplasia (SPEM) is an important risk factor for the occurrence of gastric cancer. It may be driven by a chronic inflammatory environment in which macrophage is involved. Studies have shown that intestinal metaplasia may originate from SPEM, and bile acid-induced chronic inflammation plays an important role in the process of intestinal metaplasia. However, whether bile acids are involved in the development of SPEM and the specific mechanism are unclear. Meanwhile, macrophages are known to be involved in inflammation regulation by releasing various factors, including exosomes. In this study, we hypothesized that the exosomes released from macrophages stimulated by deoxycholic acid participated in the development of SPME. METHODS: In vivo, mice were gavaged with deoxycholic acid for 4 weeks, and gastric tissues were harvested. In vitro, deoxycholic acid-induced macrophage-derived exosomes were isolated by ultracentrifugation and cocultured with the gastric organoids of mice. Immunofluorescence staining and quantitative real-time PCR were used to analyze markers of macrophages and SPEM. RESULTS: In vivo, after 4 weeks of deoxycholic acid intragastric administration, macrophage markers (F4/80) and SPEM markers (TFF2 and GSII lectin) were increased in from treated mice compared with those from normal control mice. In vitro, macrophage-derived exosomes labeled with PKH67 were internalized by gastric organoids. Deoxycholic acid-induced macrophage-derived exosomes increased the expression of SPEM markers (TFF2 and GSII lectin) in gastric organoids compared to exosomes derived from macrophages without deoxycholic acid stimulation. CONCLUSION: Macrophage-derived exosomes may be a novel mechanism by which deoxycholic acid promotes SPEM.


Asunto(s)
Ácido Desoxicólico/farmacología , Exosomas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Macrófagos/metabolismo , Estómago/patología , Animales , Biomarcadores/metabolismo , Endocitosis/efectos de los fármacos , Exosomas/efectos de los fármacos , Exosomas/ultraestructura , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/patología , Macrófagos/efectos de los fármacos , Masculino , Metaplasia , Ratones , Ratones Endogámicos C57BL , Organoides/efectos de los fármacos , Organoides/metabolismo , Células RAW 264.7 , Regulación hacia Arriba/efectos de los fármacos
6.
J Pathol ; 247(4): 513-523, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30511397

RESUMEN

Chronic inflammation of the gastric mucosa, often caused by autoimmune gastritis and/or infection with Helicobacter pylori, can lead to atrophy of acid-secreting parietal cells with metaplasia of remaining cells. The histological pattern marks a critical step in the progression from chronic gastritis to gastric cancer, yet underlying mechanism(s) of inflammation-induced cell death of gastric epithelial cells are poorly understood. We investigated direct effects of a type 1 cytokine associated with autoimmunity and infection, interferon-γ (IFN-γ), on gastric epithelial cells. IFN-γ was applied to three-dimensional organoid cultures of gastric epithelial cells derived from gastric corpus gland (gastroids) of control and IFN-γ receptor-deficient mice. Gastroids were also treated with supernatants from activated immune cells isolated from a mouse model of autoimmune-mediated atrophic gastritis (TxA23) with and without IFN-γ expression. Finally, histopathological analysis of atrophy and metaplasia severity was performed in TxA23 mice and compared to TxA23 × Ifng-/- mice. Gastric epithelial cells in gastroid cultures expressed IFN-γ receptor in the basolateral membrane, and gastroids died when treated with IFN-γ in an IFN-γ receptor-dependent manner. Supernatants from immune cells containing high levels of IFN-γ were highly toxic to gastroids, and toxicity was tempered when IFN-γ was either neutralized using a monoclonal antibody or when supernatants from Ifng-/- mouse immune cells were used. Finally, TxA23 × Ifng-/- mice showed near-complete abrogation of pre-cancerous histopathological atrophy and metaplasia versus IFN-γ-sufficient controls. We identify IFN-γ as a critical promoter of parietal cell atrophy with metaplasia during the progression of gastritis to gastric atrophy and metaplasia. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Mucosa Gástrica/patología , Interferón gamma/fisiología , Neoplasias Gástricas/patología , Animales , Atrofia/patología , Muerte Celular/fisiología , Transformación Celular Neoplásica/patología , Progresión de la Enfermedad , Células Epiteliales/patología , Gastritis , Interferón gamma/deficiencia , Interferón gamma/farmacología , Metaplasia/patología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células Parietales Gástricas/patología , Células Tumorales Cultivadas
7.
Helicobacter ; 24(3): e12578, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30990573

RESUMEN

BACKGROUND AND AIMS: Spasmolytic polypeptide-expressing metaplasia (SPEM) is a preneoplastic gastric cancer lesion related to epigenetic microRNA (miRNA) expression. This study elucidated whether Helicobacter pylori-infected first-degree relatives of patients with gastric cancer (GCF) are susceptible to have SPEM and correlated with miR-21, 155, and 223 expressions. We also validated whether SPEM and these miRNAs can be regressed after H pylori eradication. METHODS: We prospectively enrolled 148 GCF and 148 nonulcer dyspepsia (NUD) subjects without gastric cancer familial history as controls. Each case had received a panendoscopy to determine H pylori status and gastric histology, including SPEM. The cases with SPEM were followed after H pylori eradication to determine SPEM regression. The total RNA was extracted to analyze tissues miR-21, 155, and 223 before and after eradication. RESULTS: GCF subjects had a higher prevalence of H pylori infection (73% vs 32%) and SPEM (42% vs 14%, P < 0.01) than controls. The tissue miR-21, 155, and 223 in antrum were higher in cases with SPEM than in those without SPEM (P <= 0.05). There was similar SPEM reversibility after H pylori eradication between GCF subjects and controls (72% vs 69%, P = 0.852). In the SPEM regressed cases, tissue miR-21, 155, and 223 decreased after H pylori eradication (P < 0.05). CONCLUSION: The H pylori-infected GCF subjects were prone to have SPEM with higher tissues miR-21, 155, and 223 expressions. H pylori eradication can result in a 70% SPEM regression, accompanied by a decline in miR-21, 155, and 233 expression levels.


Asunto(s)
Infecciones por Helicobacter/metabolismo , Helicobacter pylori/fisiología , Metaplasia/metabolismo , MicroARNs/genética , Péptidos/metabolismo , Neoplasias Gástricas/metabolismo , Adulto , Femenino , Infecciones por Helicobacter/microbiología , Humanos , Péptidos y Proteínas de Señalización Intercelular , Masculino , Metaplasia/microbiología , Persona de Mediana Edad , Neoplasias Gástricas/microbiología
8.
J Pathol ; 245(2): 132-137, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29508389

RESUMEN

The gastrointestinal mucosae provide a critical barrier between the external and internal milieu. Thus, damage to the mucosa requires an immediate response to provide appropriate wound closure and healing. Metaplastic lineages with phenotypes similar to the mucous glands of the distal stomach or Brunner's glands have been associated with various injurious scenarios in the stomach, small bowel, and colon. These lineages have been assigned various names including pyloric metaplasia, pseudopyloric metaplasia, ulcer-associated cell lineage (UACL), and spasmolytic polypeptide-expressing metaplasia (SPEM). A re-examination of the literature on these various forms of mucous cell metaplasia suggests that pyloric-type mucosal gland lineages may provide a ubiquitous response to mucosal injury throughout the gastrointestinal tract as well as in the pancreas, esophagus, and other mucosal surfaces. While the cellular origin of these putative reparative lineages likely varies in different regions of the gut, their final phenotypes may converge on a pyloric-type gland dedicated to mucous secretion. In addition to their healing properties in the setting of acute injury, these pyloric-type lineages may also represent precursors to neoplastic transitions in the face of chronic inflammatory influences. Further investigations are needed to determine how discrete molecular profiles relate to the origin and function of pyloric-type metaplasias previously described by histological characteristics in multiple epithelial mucosal systems in the setting of acute and chronic damage. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Linaje de la Célula , Colitis Ulcerosa/patología , Enfermedad de Crohn/patología , Mucosa Gástrica/patología , Neoplasias Gastrointestinales/patología , Mucosa Intestinal/patología , Péptidos/metabolismo , Lesiones Precancerosas/patología , Animales , Colitis Ulcerosa/metabolismo , Enfermedad de Crohn/metabolismo , Mucosa Gástrica/metabolismo , Neoplasias Gastrointestinales/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular , Mucosa Intestinal/metabolismo , Metaplasia , Lesiones Precancerosas/metabolismo , Transducción de Señal
9.
J Physiol ; 596(17): 3861-3867, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29427515

RESUMEN

The development of intestinal-type gastric cancer is preceded by the emergence of metaplastic cell lineages in the gastric mucosa. In particular, intestinal metaplasia and spasmolytic polypeptide-expressing metaplasia (SPEM) have been associated with the pathological progression to intestinal-type gastric cancer. The development of SPEM represents a physiological response to damage that recruits reparative cells to sites of mucosal injury. Metaplastic cell lineages are characterized by mucus secretion, adding a protective barrier to the epithelium. Increasing evidence indicates that the influence of alarmins and cytokines is required to initiate the process of metaplasia development. In particular, IL-33 derived from epithelial cells stimulates IL-13 production by specialized innate immune cells to induce chief cell transdifferentiation into SPEM following the loss of parietal cells from the corpus of the stomach. While SPEM represents a physiological healing response to acute injury, persistent injury and chronic inflammation can perpetuate a recurring pattern of reprogramming and metaplasia that is a risk factor for gastric cancer development. The transdifferentiation of zymogen secreting cells into mucous cell metaplasia may represent both a general repair mechanism in response to mucosal injury in many epithelia as well as a common pre-neoplastic pathway associated with chronic injury and inflammation.


Asunto(s)
Inflamación/complicaciones , Péptidos y Proteínas de Señalización Intercelular/efectos adversos , Interleucina-13/metabolismo , Intestinos/inmunología , Metaplasia/patología , Células Parietales Gástricas/inmunología , Estómago/inmunología , Animales , Humanos , Inflamación/metabolismo , Intestinos/patología , Metaplasia/etiología , Metaplasia/metabolismo , Metaplasia/prevención & control , Células Parietales Gástricas/metabolismo , Células Parietales Gástricas/patología , Estómago/patología
10.
Am J Physiol Gastrointest Liver Physiol ; 312(2): G133-G144, 2017 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-27932500

RESUMEN

The Notch signaling pathway is known to regulate stem cells and epithelial cell homeostasis in gastrointestinal tissues; however, Notch function in the corpus region of the stomach is poorly understood. In this study we examined the consequences of Notch inhibition and activation on cellular proliferation and differentiation and defined the specific Notch receptors functioning in the mouse and human corpus. Notch pathway activity was observed in the mouse corpus epithelium, and gene expression analysis revealed NOTCH1 and NOTCH2 to be the predominant Notch receptors in both mouse and human. Global Notch inhibition for 5 days reduced progenitor cell proliferation in the mouse corpus, as well as in organoids derived from mouse and human corpus tissue. Proliferation effects were mediated through both NOTCH1 and NOTCH2 receptors, as demonstrated by targeting each receptor alone or in combination with Notch receptor inhibitory antibodies. Analysis of differentiation by marker expression showed no change to the major cell lineages; however, there was a modest increase in the number of transitional cells coexpressing markers of mucous neck and chief cells. In contrast to reduced proliferation after pathway inhibition, Notch activation in the adult stomach resulted in increased proliferation coupled with reduced differentiation. These findings suggest that NOTCH1 and NOTCH2 signaling promotes progenitor cell proliferation in the mouse and human gastric corpus, which is consistent with previously defined roles for Notch in promoting stem and progenitor cell proliferation in the intestine and antral stomach. NEW & NOTEWORTHY: Here we demonstrate that the Notch signaling pathway is essential for proliferation of stem cells in the mouse and human gastric corpus. We identify NOTCH1 and NOTCH2 as the predominant Notch receptors expressed in both mouse and human corpus and show that both receptors are required for corpus stem cell proliferation. We show that chronic Notch activation in corpus stem cells induces hyperproliferation and tissue hypertrophy, suggesting that Notch may drive gastric tumorigenesis.


Asunto(s)
Proliferación Celular/fisiología , Células Epiteliales/fisiología , Receptor Notch1/metabolismo , Receptor Notch2/metabolismo , Estómago/fisiología , Animales , Femenino , Mucosa Gástrica/citología , Genes Reporteros , Humanos , Masculino , Ratones , Organoides/citología , Organoides/fisiología , Receptor Notch1/genética , Receptor Notch2/genética , Transducción de Señal/fisiología , Células Madre , Tamoxifeno/farmacología
11.
Am J Physiol Gastrointest Liver Physiol ; 312(1): G67-G76, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27881402

RESUMEN

The plasticity of gastric chief cells is exemplified by their ability to transdifferentiate into spasmolytic polypeptide-expressing metaplasia (SPEM) after parietal cell loss. We sought to determine if chief cell maturity is a limiting factor in the capacity to transdifferentiate. Mist1-/- mice, previously shown to form only immature chief cells, were treated with DMP-777 or L635 to study the capability of these immature chief cells to transdifferentiate into a proliferative metaplastic lineage after acute parietal cell loss. Mist1-/- mice treated with DMP-777 showed fewer chief cell to SPEM transitions. Mist1-/- mice treated with L635 demonstrated significantly fewer proliferative SPEM cells compared with control mice. Thus immature chief cells were unable to transdifferentiate efficiently into SPEM after acute parietal cell loss. To determine whether chief cell age affects transdifferentiation into SPEM, we used tamoxifen to induce YFP expression in chief cells of Mist1CreER/+;RosaYFP mice and subsequently treated the cells with L635 to induce SPEM at 1 to 3.5 mo after tamoxifen treatment. After L635 treatment to induce acute parietal cell loss, 43% of all YFP-positive cells at 1 mo posttamoxifen were SPEM cells, of which 44% of these YFP-positive SPEM cells were proliferative. By 2 mo after tamoxifen induction, only 24% of marked SPEM cells were proliferating. However, by 3.5 mo after tamoxifen induction, only 12% of marked chief cells transdifferentiated into SPEM and none were proliferative. Thus, as chief cells age, they lose their ability to transdifferentiate into SPEM and proliferate. Therefore, both functional maturation and age limit chief cell plasticity. NEW & NOTEWORTHY: Previous investigations have indicated that spasmolytic polypeptide-expressing metaplasia (SPEM) in the stomach arises from transdifferentiation of chief cells. Nevertheless, the intrinsic properties of chief cells that influence transdifferentiation have been largely unknown. We now report that the ability to transdifferentiate into SPEM is impaired in chief cells that lack full functional maturation, and as chief cells age, they lose their ability to transdifferentiate. Thus chief cell plasticity is dependent on both cell age and maturation.


Asunto(s)
Linaje de la Célula/fisiología , Transdiferenciación Celular/fisiología , Células Principales Gástricas/patología , Estómago/patología , Factores de Edad , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proliferación Celular/fisiología , Células Principales Gástricas/metabolismo , Mucosa Gástrica/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Metaplasia/metabolismo , Metaplasia/patología , Ratones , Ratones Noqueados , Células Parietales Gástricas/metabolismo , Células Parietales Gástricas/patología , Péptidos/metabolismo
12.
Helicobacter ; 22(4)2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28326664

RESUMEN

BACKGROUND: Corpus-predominant gastritis index (CGI) is an early histological marker to identify Helicobacter pylori-infected gastric cancer relatives at risk of cancer. This study validated whether CGI is more prevalent in H. pylori-infected nonulcer dyspepsia (NUD) subjects than in duodenal ulcer (DU) controls and whether it is reversible after H. pylori eradication or is correlated with noninvasive biomarkers. MATERIALS AND METHODS: In this longitudinal cohort study, 573 H. pylori-infected subjects were enrolled, including 349 NUD and 224 DU. Gastric specimens were provided to assess CGI, spasmolyic polypeptide-expressing metaplasia (SPEM), and Operative Link on Gastric Intestinal Metaplasia assessment (OLGIM). Serum pepsinogen I and II levels were assessed using enzyme-linked immunosorbent assay. CGI subjected were followed up at least 1 year after H. pylori eradication. RESULTS: NUD subjects had higher prevalence rates of CGI (47.0% vs 29.9%, P<.001) and OLGIM stages III-IV (24.1% vs 15.2%, P=.01) than controls. CGI was highly prevalent in NUD subjects after the age of 40, which was 10 years earlier than atrophic gastritis and intestinal metaplasia. NUD subjects with CGI had higher risk of SPEM (OR 2.86, P<.001) and lower serum pepsinogen I/II ratios (P<.001) than those without CGI. Serum pepsinogen I/II ratios <9 could predict CGI modestly (AUROC 0.69, 95% CI: 0.63-0.74). CGI was regressed after eradication (P<.001). CONCLUSIONS: CGI was more prevalent in H. pylori-infected NUD subjects than in controls, was correlated with SPEM, and may serve as a marker earlier than OLGIM to indicate risk of gastric cancer. Moreover, CGI could be regressed after eradication.


Asunto(s)
Dispepsia/complicaciones , Gastritis/complicaciones , Gastritis/patología , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/patología , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/epidemiología , Adulto , Anciano , Ensayo de Inmunoadsorción Enzimática , Femenino , Histocitoquímica , Humanos , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Pepsinógeno A/sangre , Pronóstico , Estudios Prospectivos , Medición de Riesgo , Estómago/patología
13.
BMC Gastroenterol ; 17(1): 145, 2017 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-29212456

RESUMEN

BACKGROUND: Although Helicobacter-induced gastric inflammation is the major predisposing factor for gastric carcinogenesis, the precise mechanism by which chronic gastritis causes gastric cancer remains unclear. Intestinal and spasmolytic polypeptide-expressing metaplasia (SPEM) is considered as precancerous lesions, changes in epithelial tissue stem/progenitor cells after chronic inflammation has not been clarified yet. In this study, we utilized three-dimensional gastric epithelial cell culture systems that could form organoids, mimicking gastric epithelial layer, and characterized the changes in epithelial cells after chronic Helicobacter felis infection. METHODS: We used three mice model; 1) long-term H. felis infection, 2) H. felis eradication, and 3) MNU chemical carcinogenesis model. We performed cRNA microarray analysis after organoid culture, and analyzed the effects of chronic gastric inflammation on tissue stem cells, by the size of organoid, mRNA expression profile and immunohistochemical analysis. RESULTS: The number of organoids cultured from gastric epithelial cells was significantly higher in organoids isolated from H. felis-infected mice compared with those from uninfected gastric mucosa. Based on the mRNA expression profile, we found that possible stem cell markers such as Cd44, Dclk1, and genes associated with the intestinal phenotype, such as Villin, were increased in organoids isolated from H. felis-infected mucosa compared with the control. The upregulation of these genes were cancelled after H. felis eradication. In a xenograft model, tumors were generated only from organoids cultured from carcinogen-treated gastric mucosa, not from H. felis infected mucosa or control organoids. CONCLUSIONS: Our results suggested that, as a possible mechanism of gastric carcinogenesis, chronic inflammation induced by H. felis infection increased the number of tissue stem/progenitor cells and the expression of stem cell markers. These findings suggest that chronic inflammation may alter the direction of differentiation toward undifferentiated state and that drawbacks may enable cells to redifferentiate to intestinal metaplasia or neoplasia.


Asunto(s)
Mucosa Gástrica/patología , Gastritis/patología , Infecciones por Helicobacter/patología , Células Madre/patología , Animales , Carcinogénesis , Recuento de Células , Proliferación Celular , Enfermedad Crónica , Citocinas/metabolismo , Modelos Animales de Enfermedad , Mucosa Gástrica/metabolismo , Gastritis/metabolismo , Expresión Génica , Infecciones por Helicobacter/metabolismo , Helicobacter felis , Masculino , Ratones Endogámicos C57BL , Organoides/metabolismo , ARN Mensajero/genética , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/patología
14.
Int J Mol Sci ; 18(10)2017 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-28953255

RESUMEN

Despite a significant decrease in the incidence of gastric cancer in Western countries over the past century, gastric cancer is still one of the leading causes of cancer-related deaths worldwide. Most human gastric cancers develop after long-term Helicobacter pylori infection via the Correa pathway: the progression is from gastritis, atrophy, intestinal metaplasia, dysplasia, to cancer. However, it remains unclear whether metaplasia is a direct precursor of gastric cancer or merely a marker of high cancer risk. Here, we review human studies on the relationship between metaplasia and cancer in the stomach, data from mouse models of metaplasia regarding the mechanism of metaplasia development, and the cellular responses induced by H. pylori infection.


Asunto(s)
Lesiones Precancerosas/patología , Neoplasias Gástricas/patología , Estómago/patología , Animales , Biomarcadores , Modelos Animales de Enfermedad , Epigénesis Genética , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiología , Mucosa Gástrica/patología , Regulación Neoplásica de la Expresión Génica , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/microbiología , Helicobacter pylori , Humanos , Metaplasia , Ratones , Clasificación del Tumor , Transducción de Señal , Neoplasias Gástricas/etiología , Neoplasias Gástricas/metabolismo
15.
Am J Physiol Gastrointest Liver Physiol ; 307(8): G777-92, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25190476

RESUMEN

Oxyntic atrophy in the stomach leads to chief cell transdifferentiation into spasmolytic polypeptide expressing metaplasia (SPEM). Investigations of preneoplastic metaplasias in the stomach are limited by the sole reliance on in vivo mouse models, owing to the lack of in vitro models for distinct normal mucosal lineages and metaplasias. Utilizing the Immortomouse, in vitro cell models of chief cells and SPEM were developed to study the characteristics of normal chief cells and metaplasia. Chief cells and SPEM cells isolated from Immortomice were cultured and characterized at both the permissive (33°C) and the nonpermissive temperature (39°C). Clones were selected on the basis of their transcriptional expression of specific stomach lineage markers (named ImChief and ImSPEM) and protein expression and growth were analyzed. The transcriptional expression profiles of ImChief and ImSPEM cells were compared further by using gene microarrays. ImChief cells transcriptionally express most chief cell markers and contain pepsinogen C and RAB3D-immunostaining vesicles. ImSPEM cells express the SPEM markers TFF2 and HE4 and constitutively secrete HE4. Whereas ImChief cells cease proliferation at the nonpermissive temperature, ImSPEM cells continue to proliferate at 39°C. Gene expression profiling of ImChief and ImSPEM revealed myelin and lymphocyte protein 2 (MAL2) as a novel marker of SPEM lineages. Our results indicate that the expression and proliferation profiles of the novel ImChief and ImSPEM cell lines resemble in vivo chief and SPEM cell lineages. These cell culture lines provide the first in vitro systems for studying the molecular mechanisms of the metaplastic transition in the stomach.


Asunto(s)
Células Principales Gástricas/metabolismo , Proteínas Proteolipídicas Asociadas a Mielina y Linfocito/metabolismo , Péptidos/metabolismo , Estómago/patología , Animales , Biomarcadores/metabolismo , Células Cultivadas , Mucosa Gástrica/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Metaplasia/diagnóstico , Ratones , Proteínas Proteolipídicas Asociadas a Mielina y Linfocito/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Pepsinógeno C/genética , Pepsinógeno C/metabolismo , Péptidos/genética , Proteínas/genética , Proteínas/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Complejos de Ubiquitina-Proteína Ligasa , Proteína 2 de Dominio del Núcleo de Cuatro Disulfuros WAP , Proteínas de Unión al GTP rab3/genética , Proteínas de Unión al GTP rab3/metabolismo
16.
Gastroenterology ; 145(3): 554-65, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23684942

RESUMEN

BACKGROUND & AIMS: Almost all gastric cancers are adenocarcinomas, which have considerable heterogeneity among patients. We sought to identify subtypes of gastric adenocarcinomas with particular biological properties and responses to chemotherapy and targeted agents. METHODS: We compared gene expression patterns among 248 gastric tumors; using a robust method of unsupervised clustering, consensus hierarchical clustering with iterative feature selection, we identified 3 major subtypes. We developed a classifier for these subtypes and validated it in 70 tumors from a different population. We identified distinct genomic and epigenomic properties of the subtypes. We determined drug sensitivities of the subtypes in primary tumors using clinical survival data, and in cell lines through high-throughput drug screening. RESULTS: We identified 3 subtypes of gastric adenocarcinoma: proliferative, metabolic, and mesenchymal. Tumors of the proliferative subtype had high levels of genomic instability, TP53 mutations, and DNA hypomethylation. Cancer cells of the metabolic subtype were more sensitive to 5-fluorouracil than the other subtypes. Furthermore, in 2 independent groups of patients, those with tumors of the metabolic subtype appeared to have greater benefits with 5-fluorouracil treatment. Tumors of the mesenchymal subtype contain cells with features of cancer stem cells, and cell lines of this subtype are particularly sensitive to phosphatidylinositol 3-kinase-AKT-mTOR inhibitors in vitro. CONCLUSIONS: Based on gene expression patterns, we classified gastric cancers into 3 subtypes, and validated these in an independent set of tumors. The subgroups have differences in molecular and genetic features and response to therapy; this information might be used to select specific treatment approaches for patients with gastric cancer.


Asunto(s)
Adenocarcinoma/clasificación , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/genética , Fluorouracilo/uso terapéutico , Regulación Neoplásica de la Expresión Génica , Inhibidores de las Quinasa Fosfoinosítidos-3 , Neoplasias Gástricas/clasificación , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Anciano , Teorema de Bayes , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Análisis por Conglomerados , Estudios de Asociación Genética , Humanos , Persona de Mediana Edad , Modelos Estadísticos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Análisis de Regresión , Estudios Retrospectivos , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Análisis de Supervivencia , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Resultado del Tratamiento
17.
Gut ; 62(9): 1270-9, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22773549

RESUMEN

OBJECTIVES: Spasmolytic polypeptide-expressing metaplasia (SPEM) develops as a preneoplastic lesion in the stomachs of mice and humans after parietal cell loss. To identify the commonalities and differences between phenotypic SPEM lineages, SPEM were studied from three different mouse models of parietal cell loss: with chronic inflammation with Helicobacter felis infection; with acute inflammation with L635 treatment; and without inflammation following DMP-777 treatment. DESIGN: RNA transcripts from laser capture microdissected normal chief cells and SPEM lineages were compared using gene microarray. Alterations in transcripts were validated by quantitative real-time PCR. Clusterin and cystic fibrosis transmembrane conductance regulator (CFTR) were selected for immunohistochemical analysis in all mouse models as well as in human SPEM, intestinal metaplasia and gastric cancer. RESULTS: Transcript expression patterns demonstrated differences among the phenotypic SPEM models. Clusterin expression was significantly upregulated in all three mouse SPEM models as well as in human SPEM. The highest clusterin expression in human gastric cancers correlated with poor survival. Conversely, CFTR expression was upregulated only in SPEM with inflammation in mice. In humans, intestinal metaplasia, but not SPEM, expressed CFTR. CONCLUSIONS: While markers such as clusterin are expressed in all phenotypic SPEM lineages, distinct patterns of upregulated genes including CFTR are present in murine metaplasia associated with inflammation, indicative of progression of metaplasia towards a more intestinalised metaplastic phenotype.


Asunto(s)
Clusterina/metabolismo , Infecciones por Helicobacter/complicaciones , Inflamación , Intestinos/patología , Células Parietales Gástricas/patología , Péptidos , Animales , Azetidinas/farmacología , Biomarcadores/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Inflamación/tratamiento farmacológico , Inflamación/etiología , Inflamación/genética , Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Captura por Microdisección con Láser , Metaplasia/diagnóstico , Metaplasia/etiología , Metaplasia/genética , Metaplasia/metabolismo , Ratones , Ratones Endogámicos CFTR , Células Parietales Gástricas/metabolismo , Péptidos/genética , Péptidos/metabolismo , Piperazinas/farmacología , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Regulación hacia Arriba
18.
Int J Oncol ; 64(3)2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38299264

RESUMEN

Gastric cancer ranks as one of the most prevalent cancers worldwide. While the incidence of gastric cancer in Western countries has notably diminished over the past century, it continues to be a leading cause of cancer­related mortality on a global scale. The majority of gastric cancers in humans are attributed to chronic Helicobacter pylori infection and the progression of gastric cancer is often preceded by gastritis, atrophy, metaplasia and dysplasia. However, the precise mechanisms underlying the development of gastric cancer remain ambiguous, including the formation of gastric polyps and precancerous lesions. In humans, two types of precancerous metaplasia have been identified in relation to gastric malignancies: Intestinal metaplasia and spasmolytic polypeptide­expressing metaplasia (SPEM). The role of SPEM in the induction of gastric cancer has gained recent attention and its link with early­stage human gastric cancer is increasingly evident. To gain insight into SPEM, the present study reviewed the role and research progress of SPEM in gastric cancer.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Péptidos y Proteínas de Señalización Intercelular , Lesiones Precancerosas , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/patología , Mucosa Gástrica/patología , Péptidos , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/patología , Lesiones Precancerosas/patología , Metaplasia
19.
Chin J Integr Med ; 2024 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-38676828

RESUMEN

The progression from gastric mucosal inflammation to cancer signifies a pivotal event in the trajectory of gastric cancer (GC) development. Chinese medicine (CM) exhibits unique advantages and holds significant promise in inhibiting carcinogenesis of the gastric mucosa. This review intricately examines the critical pathological events during the transition from gastric mucosal inflammation-cancer transformation (GMICT), with a particular focus on pathological evolution mechanisms of spasmolytic polypeptide-expressing metaplasia (SPEM). Moreover, it investigates the pioneering applications and advancements of CM in intervening within the medical research domain of precancerous transformations leading to GC. Furthermore, the analysis extends to major shortcomings and challenges confronted by current research in gastric precancerous lesions, and innovative studies related to CM are presented. We offer a highly succinct yet optimistic outlook on future developmental trends. This paper endeavors to foster a profound understanding of forefront dynamics in GMICT research and scientific implications of modernizing CM. It also introduces a novel perspective for establishing a collaborative secondary prevention system for GC that integrates both Western and Chinese medicines.

20.
Cancers (Basel) ; 16(3)2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38339273

RESUMEN

Gastric cancer is one of the leading causes of cancer deaths worldwide, with chronic gastritis representing the main predisposing factor initiating the cascade of events leading to metaplasia and eventually progressing to cancer. A widely accepted classification distinguishes between autoimmune and environmental atrophic gastritis, mediated, respectively, by T cells promoting the destruction of the oxyntic mucosa, and chronic H. pylori infection, which has also been identified as the major risk factor for gastric cancer. The original dogma posits Th1 immunity as a main causal factor for developing gastritis and metaplasia. Recently, however, it has become evident that Th2 immune responses play a major role in the events causing chronic inflammation leading to tumorigenesis, and in this context, many different cell types and cytokines are involved. In particular, the activity of cytokines, such as IL-33 and IL-13, and cell types, such as mast cells, M2 macrophages and eosinophils, are intertwined in the process, promoting chronic gastritis-dependent and more diffuse metaplasia. Herein, we provide an overview of the critical events driving the pathology of this disease, focusing on the most recent findings regarding the importance of Th2 immunity in gastritis and gastric metaplasia.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA