Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Gut ; 71(6): 1068-1077, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34497145

RESUMEN

OBJECTIVE: Metaplasia arises from differentiated cell types in response to injury and is considered a precursor in many cancers. Heterogeneous cell lineages are present in the reparative metaplastic mucosa with response to injury, including foveolar cells, proliferating cells and spasmolytic polypeptide-expressing metaplasia (SPEM) cells, a key metaplastic cell population. Zymogen-secreting chief cells are long-lived cells in the stomach mucosa and have been considered the origin of SPEM cells; however, a conflicting paradigm has proposed isthmal progenitor cells as an origin for SPEM. DESIGN: Gastric intrinsic factor (GIF) is a stomach tissue-specific gene and exhibits protein expression unique to mature mouse chief cells. We generated a novel chief cell-specific driver mouse allele, GIF-rtTA. GIF-GFP reporter mice were used to validate specificity of GIF-rtTA driver in chief cells. GIF-Cre-RnTnG mice were used to perform lineage tracing during homoeostasis and acute metaplasia development. L635 treatment was used to induce acute mucosal injury and coimmunofluorescence staining was performed for various gastric lineage markers. RESULTS: We demonstrated that mature chief cells, rather than isthmal progenitor cells, serve as the predominant origin of SPEM cells during the metaplastic process after acute mucosal injury. Furthermore, we observed long-term label-retaining chief cells at 1 year after the GFP labelling in chief cells. However, only a very small subset of the long-term label-retaining chief cells displayed the reprogramming ability in homoeostasis. In contrast, we identified chief cell-originating SPEM cells as contributing to lineages within foveolar cell hyperplasia in response to the acute mucosal injury. CONCLUSION: Our study provides pivotal evidence for cell plasticity and lineage contributions from differentiated gastric chief cells during acute metaplasia development.


Asunto(s)
Células Principales Gástricas , Neoplasias Gástricas , Animales , Plasticidad de la Célula , Células Principales Gástricas/metabolismo , Mucosa Gástrica/metabolismo , Humanos , Metaplasia/metabolismo , Ratones , Estómago , Neoplasias Gástricas/metabolismo
2.
Cell Mol Gastroenterol Hepatol ; 13(3): 843-856, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34915204

RESUMEN

The epithelial lining of the intestine, particularly the stem cell compartment, is affected by harsh conditions in the luminal environment and also is susceptible to genotoxic agents such as radiation and chemotherapy. Therefore, the ability for intestinal epithelial cells to revert to a stem cell state is an important physiological damage response to regenerate the intestinal epithelium at sites of mucosal injury. Many signaling networks involved in maintaining the stem cell niche are activated as part of the damage response to promote cellular plasticity and regeneration. The relative contribution of each cell type and signaling pathway is a critical area of ongoing research, likely dependent on the nature of injury as well as the regional specification within the intestine. Here, we review the current understanding of the multicellular cooperation to restore the intestinal epithelium after damage.


Asunto(s)
Plasticidad de la Célula , Nicho de Células Madre , Homeostasis , Intestinos , Regeneración
3.
Cell Mol Gastroenterol Hepatol ; 12(1): 59-80, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33548596

RESUMEN

BACKGROUND & AIMS: The molecular motor, Myosin Vb (MYO5B), is well documented for its role in trafficking cargo to the apical membrane of epithelial cells. Despite its involvement in regulating apical proteins, the role of MYO5B in cell polarity is less clear. Inactivating mutations in MYO5B result in microvillus inclusion disease (MVID), a disorder characterized by loss of key apical transporters and the presence of intracellular inclusions in enterocytes. We previously identified that inclusions in Myo5b knockout (KO) mice form from invagination of the apical brush border via apical bulk endocytosis. Herein, we sought to elucidate the role of polarity complexes and tight junction proteins during the formation of inclusions. METHODS: Intestinal tissue from neonatal control and Myo5b KO littermates was analyzed by immunofluorescence to determine the localization of polarity complexes and tight junction proteins. RESULTS: Proteins that make up the apical polarity complexes-Crumbs3 and Pars complexes-were associated with inclusions in Myo5b KO mice. In addition, tight junction proteins were observed to be concentrated over inclusions that were present at the apical membrane of Myo5b-deficient enterocytes in vivo and in vitro. Our mouse findings are complemented by immunostaining in a large animal swine model of MVID genetically engineered to express a human MVID-associated mutation that shows an accumulation of Claudin-2 over forming inclusions. The findings from our swine model of MVID suggest that a similar mechanism of tight junction accumulation occurs in patients with MVID. CONCLUSIONS: These data show that apical bulk endocytosis involves the altered localization of apical polarity proteins and tight junction proteins after loss of Myo5b.


Asunto(s)
Enterocitos/metabolismo , Miosina Tipo V/metabolismo , Proteínas de Uniones Estrechas/metabolismo , Animales , Endocitosis , Absorción Intestinal , Ratones , Ratones Noqueados , Miosina Tipo V/deficiencia , Proteínas de Uniones Estrechas/genética
4.
Gastroenterology ; 159(6): 2077-2091.e8, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32891625

RESUMEN

BACKGROUND & AIMS: Severe injury to the lining of the stomach leads to changes in the epithelium (reprogramming) that protect and promote repair of the tissue, including development of spasmolytic polypeptide-expressing metaplasia (SPEM) and tuft and foveolar cell hyperplasia. Acute gastric damage elicits a type-2 inflammatory response that includes production of type-2 cytokines and infiltration by eosinophils and alternatively activated macrophages. Stomachs of mice that lack interleukin 33 (IL33) or interleukin 13 (IL13) did not undergo epithelial reprogramming after drug-induced injury. We investigated the role of group 2 innate lymphoid cells (ILC2s) in gastric epithelial repair. METHODS: Acute gastric injury was induced in C57BL/6J mice (wild-type and RAG1 knockout) by administration of L635. We isolated ILC2s by flow cytometry from stomachs of mice that were and were not given L635 and performed single-cell RNA sequencing. ILC2s were depleted from wild-type and RAG1-knockout mice by administration of anti-CD90.2. We assessed gastric cell lineages, markers of metaplasia, inflammation, and proliferation. Gastric tissue microarrays from patients with gastric adenocarcinoma were analyzed by immunostaining. RESULTS: There was a significant increase in the number of GATA3-positive ILC2s in stomach tissues from wild-type mice after L635-induced damage, but not in stomach tissues from IL33-knockout mice. We characterized a marker signature of gastric mucosal ILC2s and identified a transcription profile of metaplasia-associated ILC2s, which included changes in expression of Il5, Il13, Csf2, Pd1, and Ramp3; these changes were validated by quantitative polymerase chain reaction and immunocytochemistry. Depletion of ILC2s from mice blocked development of metaplasia after L635-induced injury in wild-type and RAG1-knockout mice and prevented foveolar and tuft cell hyperplasia and infiltration or activation of macrophages after injury. Numbers of ILC2s were increased in stomach tissues from patients with SPEM compared with patients with normal corpus mucosa. CONCLUSIONS: In analyses of stomach tissues from mice with gastric tissue damage and patients with SPEM, we found evidence of type 2 inflammation and increased numbers of ILC2s. Our results suggest that ILC2s coordinate the metaplastic response to severe gastric injury.


Asunto(s)
Mucosa Gástrica/patología , Inmunidad Innata , Subgrupos Linfocitarios/inmunología , Animales , Modelos Animales de Enfermedad , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/inmunología , Humanos , Interleucina-33/genética , Metaplasia/inducido químicamente , Metaplasia/genética , Metaplasia/inmunología , Ratones , Ratones Noqueados
5.
Gastroenterology ; 158(8): 2236-2249.e9, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32112796

RESUMEN

BACKGROUND & AIMS: Microvillus inclusion disease (MVID) is caused by inactivating mutations in the myosin VB gene (MYO5B). MVID is a complex disorder characterized by chronic, watery, life-threatening diarrhea that usually begins in the first hours to days of life. We developed a large animal model of MVID to better understand its pathophysiology. METHODS: Pigs were cloned by transfer of chromatin from swine primary fetal fibroblasts, which were edited with TALENs and single-strand oligonucleotide to introduce a P663-L663 substitution in the endogenous swine MYO5B (corresponding to the P660L mutation in human MYO5B, associated with MVID) to fertilized oocytes. We analyzed duodenal tissues from patients with MVID (with the MYO5B P660L mutation) and without (controls), and from pigs using immunohistochemistry. Enteroids were generated from pigs with MYO5B(P663L) and without the substitution (control pigs). RESULTS: Duodenal tissues from patients with MVID lacked MYO5B at the base of the apical membrane of intestinal cells; instead MYO5B was intracellular. Intestinal tissues and derived enteroids from MYO5B(P663L) piglets had reduced apical levels and diffuse subapical levels of sodium hydrogen exchanger 3 and SGLT1, which regulate transport of sodium, glucose, and water, compared with tissues from control piglets. However, intestinal tissues and derived enteroids from MYO5B(P663L) piglets maintained CFTR on apical membranes, like tissues from control pigs. Liver tissues from MYO5B(P663L) piglets had alterations in bile salt export pump, a transporter that facilitates bile flow, which is normally expressed in the bile canaliculi in the liver. CONCLUSIONS: We developed a large animal model of MVID that has many features of the human disease. Studies of this model could provide information about the functions of MYO5B and MVID pathogenesis, and might lead to new treatments.


Asunto(s)
Duodeno/metabolismo , Edición Génica , Mucosa Intestinal/metabolismo , Síndromes de Malabsorción/genética , Microvellosidades/patología , Mucolipidosis/genética , Cadenas Pesadas de Miosina/genética , Miosina Tipo V/genética , Transportador 1 de Sodio-Glucosa/metabolismo , Intercambiador 3 de Sodio-Hidrógeno/metabolismo , Animales , Animales Modificados Genéticamente , Células Cultivadas , Técnicas de Cocultivo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Modelos Animales de Enfermedad , Duodeno/patología , Predisposición Genética a la Enfermedad , Humanos , Mucosa Intestinal/patología , Síndromes de Malabsorción/metabolismo , Síndromes de Malabsorción/patología , Microvellosidades/genética , Microvellosidades/metabolismo , Mucolipidosis/metabolismo , Mucolipidosis/patología , Mutación Missense , Fenotipo , Sodio/metabolismo , Transportador 1 de Sodio-Glucosa/genética , Intercambiador 3 de Sodio-Hidrógeno/genética , Sus scrofa
6.
J Cell Biol ; 218(11): 3647-3662, 2019 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-31562230

RESUMEN

In patients with inactivating mutations in myosin Vb (Myo5B), enterocytes show large inclusions lined by microvilli. The origin of inclusions in small-intestinal enterocytes in microvillus inclusion disease is currently unclear. We postulated that inclusions in Myo5b KO mouse enterocytes form through invagination of the apical brush border membrane. 70-kD FITC-dextran added apically to Myo5b KO intestinal explants accumulated in intracellular inclusions. Live imaging of Myo5b KO-derived enteroids confirmed the formation of inclusions from the apical membrane. Treatment of intestinal explants and enteroids with Dyngo resulted in accumulation of inclusions at the apical membrane. Inclusions in Myo5b KO enterocytes contained VAMP4 and Pacsin 2 (Syndapin 2). Myo5b;Pacsin 2 double-KO mice showed a significant decrease in inclusion formation. Our results suggest that apical bulk endocytosis in Myo5b KO enterocytes resembles activity-dependent bulk endocytosis, the primary mechanism for synaptic vesicle uptake during intense neuronal stimulation. Thus, apical bulk endocytosis mediates the formation of inclusions in neonatal Myo5b KO enterocytes.


Asunto(s)
Endocitosis , Enterocitos/citología , Enterocitos/metabolismo , Miosina Tipo V/metabolismo , Animales , Ratones , Ratones Noqueados , Miosina Tipo V/deficiencia
7.
Cell Mol Gastroenterol Hepatol ; 8(3): 379-405, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31071489

RESUMEN

BACKGROUND & AIMS: Many differentiated epithelial cell types are able to reprogram in response to tissue damage. Although reprogramming represents an important physiological response to injury, the regulation of cellular plasticity is not well understood. Damage to the gastric epithelium initiates reprogramming of zymogenic chief cells into a metaplastic cell lineage known as spasmolytic polypeptide-expressing metaplasia (SPEM). The present study seeks to identify the role of xCT, a cystine/glutamate antiporter, in chief cell reprogramming after gastric injury. We hypothesize that xCT-dependent reactive oxygen species (ROS) detoxification is required for the reprogramming of chief cells into SPEM. METHODS: Sulfasalazine (an xCT inhibitor) and small interfering RNA knockdown were used to target xCT on metaplastic cells in vitro. Sulfasalazine-treated wild-type mice and xCT knockout mice were analyzed. L635 or DMP-777 treatment was used to chemically induce acute gastric damage. The anti-inflammatory metabolites of sulfasalazine (sulfapyridine and mesalazine) were used as controls. Normal gastric lineages, metaplastic markers, autophagy, proliferation, xCT activity, ROS, and apoptosis were assessed. RESULTS: xCT was up-regulated early as chief cells transitioned into SPEM. Inhibition of xCT or small interfering RNA knockdown blocked cystine uptake and decreased glutathione production by metaplastic cells and prevented ROS detoxification and proliferation. Moreover, xCT activity was required for chief cell reprogramming into SPEM after gastric injury in vivo. Chief cells from xCT-deficient mice showed decreased autophagy, mucus granule formation and proliferation, as well as increased levels of ROS and apoptosis compared with wild-type mice. On the other hand, the anti-inflammatory metabolites of sulfasalazine did not affect SPEM development. CONCLUSIONS: The results presented here suggest that maintaining redox balance is crucial for progression through the reprogramming process and that xCT-mediated cystine uptake is required for chief cell plasticity and ROS detoxification.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/genética , Azetidinas/efectos adversos , Mucosa Gástrica/patología , Piperazinas/efectos adversos , Sulfasalazina/farmacología , Sistema de Transporte de Aminoácidos y+/metabolismo , Animales , Línea Celular , Plasticidad de la Célula , Reprogramación Celular , Células Principales Gástricas/citología , Células Principales Gástricas/efectos de los fármacos , Células Principales Gástricas/metabolismo , Mucosa Gástrica/citología , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/metabolismo , Técnicas de Inactivación de Genes , Humanos , Ratones , Células Parietales Gástricas/citología , Células Parietales Gástricas/efectos de los fármacos , Células Parietales Gástricas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba
8.
J Histochem Cytochem ; 67(1): 53-63, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29969055

RESUMEN

Gastric adenocarcinoma develops in metaplastic mucosa associated with Helicobacter pylori infection in the stomach. We have sought to evaluate the precise lineage changes in the stomachs of insulin-gastrin (INS-GAS) mice infected with H. pylori and/or intestinal flora (Altered Schaedler's Flora; ASF). Stomachs from groups infected with H. pylori contained progressive spasmolytic polypeptide-expressing metaplasia (SPEM) compared with germ-free and mice infected with ASF alone. The overall phenotype of the H. pylori-infected mice was dominated by Ulex europaeus lectin (UEAI)-positive foveolar hyperplasia that was distinct from GSII/CD44v9-positive SPEM. However, in the mice with H. pylori co-infected with ASF, we identified a subpopulation of UEAI-positive foveolar cells that co-expressed intestinal mucin 4 (MUC4). These regions of foveolar cells were variably positive for CD44v9 as well as TFF3. Interestingly, an intravascular lesion identified in a dual H. pylori/ASF-infected mouse expressed both UEAI and Muc4. Finally, we identified an increase in the number of tuft cells within the mucosa of H. pylori-infected groups. Our findings suggest that H. pylori infection promotes foveolar hyperplasia as well as metaplasia, while co-infection may promote progressive foveolar and metaplastic lesions as well as dysplasia. Grading of gastric lesions in mice as preneoplastic requires multiple immunostaining markers to assign lineage derivation and behavior.


Asunto(s)
Mucosa Gástrica/patología , Infecciones por Helicobacter/patología , Helicobacter pylori/aislamiento & purificación , Adenocarcinoma/microbiología , Adenocarcinoma/patología , Animales , Quinasas Similares a Doblecortina , Mucosa Gástrica/microbiología , Microbioma Gastrointestinal , Infecciones por Helicobacter/microbiología , Helicobacter pylori/fisiología , Receptores de Hialuranos/análisis , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intercelular/análisis , Masculino , Ratones , Mucina 4/análisis , Proteínas Serina-Treonina Quinasas/análisis , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/patología
9.
Cell Mol Gastroenterol Hepatol ; 6(3): 257-276, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30109253

RESUMEN

Background & Aims: Chronic inflammation is a predisposing condition for colorectal cancer. Many studies to date have focused on proinflammatory signaling pathways in the colon. Understanding the mechanisms that suppress inflammation, particularly in epithelial cells, is critical for developing therapeutic interventions. Here, we explored the roles of transforming growth factor ß (TGFß) family signaling through SMAD4 in colonic epithelial cells. Methods: The Smad4 gene was deleted specifically in adult murine intestinal epithelium. Colitis was induced by 3 rounds of dextran sodium sulfate in drinking water, after which mice were observed for up to 3 months. Nontransformed mouse colonocyte cell lines and colonoid cultures and human colorectal cancer cell lines were analyzed for responses to TGFß1 and bone morphogenetic protein 2. Results: Dextran sodium sulfate treatment was sufficient to drive carcinogenesis in mice lacking colonic Smad4 expression, with resulting tumors bearing striking resemblance to human colitis-associated carcinoma. Loss of SMAD4 protein was observed in 48% of human colitis-associated carcinoma samples as compared with 19% of sporadic colorectal carcinomas. Loss of Smad4 increased the expression of inflammatory mediators within nontransformed mouse colon epithelial cells in vivo. In vitro analysis of mouse and human colonic epithelial cell lines and organoids indicated that much of this regulation was cell autonomous. Furthermore, TGFß signaling inhibited the epithelial inflammatory response to proinflammatory cytokines. Conclusions: TGFß suppresses the expression of proinflammatory genes in the colon epithelium, and loss of its downstream mediator, SMAD4, is sufficient to initiate inflammation-driven colon cancer. Transcript profiling: GSE100082.


Asunto(s)
Carcinoma/inmunología , Colitis/inmunología , Neoplasias Colorrectales/inmunología , Inflamación/inmunología , Proteína Smad4/inmunología , Animales , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/metabolismo , Carcinoma/etiología , Carcinoma/patología , Línea Celular , Línea Celular Tumoral , Colitis/inducido químicamente , Colitis/complicaciones , Neoplasias Colorrectales/etiología , Neoplasias Colorrectales/patología , Sulfato de Dextran/farmacología , Humanos , Inflamación/inducido químicamente , Inflamación/complicaciones , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Smad4/genética , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
10.
Gastroenterology ; 155(6): 1883-1897.e10, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30144427

RESUMEN

BACKGROUND & AIMS: Inactivating mutations in MYO5B cause microvillus inclusion disease (MVID), but the physiological cause of the diarrhea associated with this disease is unclear. We investigated whether loss of MYO5B results in aberrant expression of apical enterocyte transporters. METHODS: We studied alterations in apical membrane transporters in MYO5B-knockout mice, as well as mice with tamoxifen-inducible, intestine-specific disruption of Myo5b (VilCreERT2;Myo5bflox/flox mice) or those not given tamoxifen (controls). Intestinal tissues were collected from mice and analyzed by immunostaining, immunoelectron microscopy, or cultured enteroids were derived. Functions of brush border transporters in intestinal mucosa were measured in Ussing chambers. We obtained duodenal biopsy specimens from individuals with MVID and individuals without MVID (controls) and compared transporter distribution by immunocytochemistry. RESULTS: Compared to intestinal tissues from littermate controls, intestinal tissues from MYO5B-knockout mice had decreased apical localization of SLC9A3 (also called NHE3), SLC5A1 (also called SGLT1), aquaporin (AQP) 7, and sucrase isomaltase, and subapical localization of intestinal alkaline phosphatase and CDC42. However, CFTR was present on apical membranes of enterocytes from MYO5B knockout and control mice. Intestinal biopsies from patients with MVID had subapical localization of NHE3, SGLT1, and AQP7, but maintained apical CFTR. After tamoxifen administration, VilCreERT2;Myo5bflox/flox mice lost apical NHE3, SGLT1, DRA, and AQP7, similar to germline MYO5B knockout mice. Intestinal tissues from VilCreERT2;Myo5bflox/flox mice had increased CFTR in crypts and CFTR localized to the apical membranes of enterocytes. Intestinal mucosa from VilCreERT2;Myo5bflox/flox mice given tamoxifen did not have an intestinal barrier defect, based on Ussing chamber analysis, but did have decreased SGLT1 activity and increased CFTR activity. CONCLUSIONS: Although trafficking of many apical transporters is regulated by MYO5B, trafficking of CFTR is largely independent of MYO5B. Decreased apical localization of NHE3, SGLT1, DRA, and AQP7 might be responsible for dysfunctional water absorption in enterocytes of patients with MVID. Maintenance of apical CFTR might exacerbate water loss by active secretion of chloride into the intestinal lumen.


Asunto(s)
Cloruros/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Enterocitos/metabolismo , Síndromes de Malabsorción/genética , Microvellosidades/patología , Mucolipidosis/genética , Miosina Tipo V/genética , Intercambiadores de Sodio-Hidrógeno/metabolismo , Animales , Acuaporinas/metabolismo , Duodeno/metabolismo , Duodeno/patología , Silenciador del Gen , Humanos , Mucosa Intestinal , Intestinos/citología , Intestinos/patología , Síndromes de Malabsorción/patología , Ratones , Ratones Noqueados , Microvellosidades/genética , Mucolipidosis/patología , Transporte de Proteínas , Transportador 1 de Sodio-Glucosa/metabolismo , Intercambiador 3 de Sodio-Hidrógeno/metabolismo , Complejo Sacarasa-Isomaltasa/metabolismo , Tamoxifeno/administración & dosificación
11.
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
12.
Gut ; 67(5): 805-817, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-28196875

RESUMEN

OBJECTIVE: Alternatively activated macrophages (M2) are associated with the progression of spasmolytic polypeptide-expressing metaplasia (SPEM) in the stomach. However, the precise mechanism(s) and critical mediators that induce SPEM are unknown. DESIGN: To determine candidate genes important in these processes, macrophages from the stomach corpus of mice with SPEM (DMP-777-treated) or advanced SPEM (L635-treated) were isolated and RNA sequenced. Effects on metaplasia development after acute parietal cell loss induced by L635 were evaluated in interleukin (IL)-33, IL-33 receptor (ST2) and IL-13 knockout (KO) mice. RESULTS: Profiling of metaplasia-associated macrophages in the stomach identified an M2a-polarised macrophage population. Expression of IL-33 was significantly upregulated in macrophages associated with advanced SPEM. L635 induced metaplasia in the stomachs of wild-type mice, but not in the stomachs of IL-33 and ST2 KO mice. While IL-5 and IL-9 were not required for metaplasia induction, IL-13 KO mice did not develop metaplasia in response to L635. Administration of IL-13 to ST2 KO mice re-established the induction of metaplasia following acute parietal cell loss. CONCLUSIONS: Metaplasia induction and macrophage polarisation after parietal cell loss is coordinated through a cytokine signalling network of IL-33 and IL-13, linking a combined response to injury by both intrinsic mucosal mechanisms and infiltrating M2 macrophages.


Asunto(s)
Interleucina-13/metabolismo , Interleucina-33/metabolismo , Macrófagos/metabolismo , Metaplasia/metabolismo , Estómago/citología , Animales , Citometría de Flujo , Mucosa Gástrica/metabolismo , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intercelular , Proteína 1 Similar al Receptor de Interleucina-1 , Interleucina-13/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Parietales Gástricas/citología , Péptidos/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Interleucina/genética , Transducción de Señal
13.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...