Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Gastroenterology ; 158(6): 1650-1666.e15, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32032583

RESUMO

BACKGROUND & AIMS: Gastric chief cells, a mature cell type that secretes digestive enzymes, have been proposed to be the origin of metaplasia and cancer through dedifferentiation or transdifferentiation. However, studies supporting this claim have had technical limitations, including issues with the specificity of chief cell markers and the toxicity of drugs used. We therefore sought to identify genes expressed specifically in chief cells and establish a model to trace these cells. METHODS: We performed transcriptome analysis of Mist1-CreERT-traced cells, with or without chief cell depletion. Gpr30-rtTA mice were generated and crossed to TetO-Cre mice, and lineage tracing was performed after crosses to R26-TdTomato mice. Additional lineage tracing experiments were performed using Mist1-CreERT, Kitl-CreERT, Tff1-Cre, and Tff2-Cre mice crossed to reporter mice. Mice were given high-dose tamoxifen or DMP-777 or were infected with Helicobacter pylori to induce gastric metaplasia. We studied mice that expressed mutant forms of Ras in gastric cells, using TetO-KrasG12D, LSL-KrasG12D, and LSL-HrasG12V mice. We analyzed stomach tissues from GPR30-knockout mice. Mice were given dichloroacetate to inhibit pyruvate dehydrogenase kinase (PDK)-dependent cell competition. RESULTS: We identified GPR30, the G-protein-coupled form of the estrogen receptor, as a cell-specific marker of chief cells in gastric epithelium of mice. Gpr30-rtTA mice crossed to TetO-Cre;R26-TdTomato mice had specific expression of GPR30 in chief cells, with no expression noted in isthmus stem cells or lineage tracing of glands. Expression of mutant Kras in GPR30+ chief cells did not lead to the development of metaplasia or dysplasia but, instead, led to a reduction in labeled numbers of chief cells and a compensatory expansion of neck lineage, which was derived from upper Kitl+ clones. Administration of high-dose tamoxifen, DMP-777, or H pylori decreased the number of labeled chief cells. Chief cells were eliminated from epithelia via GPR30- and PDK-dependent cell competition after metaplastic stimuli, whereas loss of GRP30 or inhibition of PDK activity preserved chief cell numbers and attenuated neck lineage cell expansion. CONCLUSIONS: In tracing studies of mice, we found that most chief cells are lost during metaplasia and therefore are unlikely to contribute to gastric carcinogenesis. Expansion of cells that coexpress neck and chief lineage markers, known as spasmolytic polypeptide-expressing metaplasia, does not occur via dedifferentiation from chief cells but, rather, through a compensatory response from neck progenitors to replace the eliminated chief cells.


Assuntos
Celulas Principais Gástricas/fisiologia , Mucosa Gástrica/patologia , Infecções por Helicobacter/patologia , Helicobacter pylori/patogenicidade , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Azetidinas/toxicidade , Comunicação Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Linhagem da Célula/fisiologia , Ácido Dicloroacético/administração & dosagem , Modelos Animais de Doenças , Mucosa Gástrica/citologia , Mucosa Gástrica/efeitos dos fármacos , Infecções por Helicobacter/microbiologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Metaplasia/induzido quimicamente , Metaplasia/microbiologia , Metaplasia/patologia , Camundongos , Camundongos Knockout , Piperazinas/toxicidade , Piruvato Desidrogenase Quinase de Transferência de Acetil/antagonistas & inibidores , Receptores de Estrogênio/genética , Receptores Acoplados a Proteínas G/genética , Células-Tronco/fisiologia , Tamoxifeno/toxicidade
2.
Gastroenterology ; 158(3): 598-609.e5, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31589873

RESUMO

BACKGROUND & AIMS: Adult zymogen-producing (zymogenic) chief cells (ZCs) in the mammalian gastric gland base are believed to arise from descending mucous neck cells, which arise from stem cells. Gastric injury, such as from Helicobacter pylori infection in patients with chronic atrophic gastritis, can cause metaplasia, characterized by gastric cell expression of markers of wound-healing; these cells are called spasmolytic polypeptide-expressing metaplasia (SPEM) cells. We investigated differentiation and proliferation patterns of neck cells, ZCs, and SPEM cells in mice. METHODS: C57BL/6 mice were given intraperitoneal injections of high-dose tamoxifen to induce SPEM or gavaged with H pylori (PMSS1) to induce chronic gastric injury. Mice were then given pulses of 5-bromo-2'-deoxyuridine (BrdU) in their drinking water, followed by chase periods without BrdU, or combined with intraperitoneal injections of 5-ethynyl-2'-deoxyuridine. We collected gastric tissues and performed immunofluorescence and immunohistochemical analyses to study gastric cell proliferation, differentiation, and turnover. RESULTS: After 8 weeks of continuous BrdU administration, fewer than 10% of homeostatic ZCs incorporated BrdU, whereas 88% of neck cells were labeled. In pulse-chase experiments, various chase periods decreased neck cell label but did not increase labeling of ZCs. When mice were given BrdU at the same time as tamoxifen, more than 90% of cells were labeled in all gastric lineages. After 3 months' recovery (no tamoxifen), ZCs became the predominant BrdU-labeled population, whereas other cells, including neck cells, were mostly negative. When we tracked the labeled cells in such mice over time, we observed that the proportion of BrdU-positive ZCs remained greater than 60% up to 11 months. In mice whose ZCs were the principal BrdU-positive population, acute injury by tamoxifen or chronic injury by H pylori infection resulted in SPEM cells becoming the principal BrdU-positive population. After withdrawal of tamoxifen, BrdU-positive ZCs reappeared. CONCLUSIONS: We studied mice in homeostasis or with tamoxifen- or H pylori-induced SPEM. Our findings indicated that mucous neck cells do not contribute substantially to generation of ZCs during homeostasis and that ZCs maintain their own census, likely through infrequent self-replication. After metaplasia-inducing injury, ZCs can become SPEM cells, and then redifferentiate into ZCs on injury resolution.


Assuntos
Diferenciação Celular , Proliferação de Células , Celulas Principais Gástricas/patologia , Celulas Principais Gástricas/fisiologia , Mucosa Gástrica/patologia , Animais , Bromodesoxiuridina , Feminino , Imunofluorescência , Mucosa Gástrica/metabolismo , Infecções por Helicobacter/complicações , Helicobacter pylori , Homeostase , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Metaplasia/etiologia , Metaplasia/metabolismo , Metaplasia/patologia , Metaplasia/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Tamoxifeno
4.
J Clin Invest ; 123(4): 1475-91, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23478405

RESUMO

After cell fate specification, differentiating cells must amplify the specific subcellular features required for their specialized function. How cells regulate such subcellular scaling is a fundamental unanswered question. Here, we show that the E3 ubiquitin ligase Mindbomb 1 (MIB1) is required for the apical secretory apparatus established by gastric zymogenic cells as they differentiate from their progenitors. When Mib1 was deleted, death-associated protein kinase-1 (DAPK1) was rerouted to the cell base, microtubule-associated protein 1B (MAP1B) was dephosphorylated, and the apical vesicles that normally support mature secretory granules were dispersed. Consequently, secretory granules did not mature. The transcription factor MIST1 bound the first intron of Mib1 and regulated its expression. We further showed that loss of MIB1 and dismantling of the apical secretory apparatus was the earliest quantifiable aberration in zymogenic cells undergoing transition to a precancerous metaplastic state in mouse and human stomach. Our results reveal a mechanistic pathway by which cells can scale up a specific, specialized subcellular compartment to alter function during differentiation and scale it down during disease.


Assuntos
Diferenciação Celular , Celulas Principais Gástricas/fisiologia , Ubiquitina-Proteína Ligases/fisiologia , Adenocarcinoma/enzimologia , Adenocarcinoma/patologia , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Sequência de Bases , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Crescimento Celular , Polaridade Celular , Celulas Principais Gástricas/enzimologia , Sequência Conservada , Proteínas Quinases Associadas com Morte Celular , Humanos , Neoplasias Intestinais/enzimologia , Neoplasias Intestinais/patologia , Metaplasia/induzido quimicamente , Metaplasia/enzimologia , Metaplasia/patologia , Camundongos , Camundongos Knockout , Microtúbulos/genética , Microtúbulos/metabolismo , Transporte Proteico , Estômago/patologia , Tamoxifeno , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
5.
Exp Cell Res ; 317(19): 2759-64, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21907708

RESUMO

The digestive-enzyme secreting, gastric epithelial chief (zymogenic) cell is remarkable and underappreciated. Here, we discuss how all available evidence suggests that mature chief cells in the adult, mammalian stomach are postmitotic, slowly turning over cells that arise via a relatively long-lived progenitor, the mucous neck cell, The differentiation of chief cells from neck cells does not involve cell division, and the neck cell has its own distinct pattern of gene expression and putative physiological function. Thus, the ontogeny of the normal chief cell lineage exemplifies transdifferentiation. Furthermore, under pathophysiogical loss of acid-secreting parietal cell, the chief cell lineage can itself trasndifferentiate into a mucous cell metaplasia designated Spasmolytic Polypeptide Expressing Metaplasia (SPEM). Especially in the presence of inflammation, this metaplastic lineage can regain proliferative capacity and, in humans may also further differentiate into intestinal metaplasia. The results indicate that gastric fundic lineages display remarkable plasticity in both physiological ontogeny and pathophysiological pre-neoplastic metaplasia.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Celulas Principais Gástricas/fisiologia , Lesões Pré-Cancerosas/etiologia , Estômago/patologia , Adulto , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Carcinoma/etiologia , Carcinoma/genética , Carcinoma/patologia , Transdiferenciação Celular/genética , Transdiferenciação Celular/fisiologia , Celulas Principais Gástricas/metabolismo , Celulas Principais Gástricas/patologia , Mucosa Gástrica/metabolismo , Humanos , Metaplasia , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/metabolismo , Lesões Pré-Cancerosas/patologia , Neoplasias Gástricas/etiologia , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia
6.
Gastroenterology ; 139(6): 2038-49, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20816838

RESUMO

BACKGROUND & AIMS: The transition of gastric epithelial mucous neck cells (NCs) to digestive enzyme-secreting zymogenic cells (ZCs) involves an increase in rough endoplasmic reticulum (ER) and formation of many large secretory vesicles. The transcription factor MIST1 is required for granulogenesis of ZCs. The transcription factor XBP1 binds the Mist1 promoter and induces its expression in vitro and expands the ER in other cell types. We investigated whether XBP1 activates Mist1 to regulate ZC differentiation. METHODS: Xbp1 was inducibly deleted in mice using a tamoxifen/Cre-loxP system; effects on ZC size and structure (ER and granule formation) and gastric differentiation were studied and quantified for up to 13 months after deletion using morphologic, immunofluorescence, quantitative reverse-transcriptase polymerase chain reaction, and immunoblot analyses. Interactions between XBP1 and the Mist1 promoter were studied by chromatin immunoprecipitation from mouse stomach and in XBP1-transfected gastric cell lines. RESULTS: Tamoxifen-induced deletion of Xbp1 (Xbp1Δ) did not affect survival of ZCs but prevented formation of their structure. Xbp1Δ ZCs shrank 4-fold, compared with those of wild-type mice, with granulogenesis and cell shape abnormalities and disrupted rough ER. XBP1 was required and sufficient for transcriptional activation of MIST1. ZCs that developed in the absence of XBP1 induced ZC markers (intrinsic factor, pepsinogen C) but showed abnormal retention of progenitor NC markers. CONCLUSIONS: XBP1 controls the transcriptional regulation of ZC structural development; it expands the lamellar rough ER and induces MIST1 expression to regulate formation of large granules. XBP1 is also required for loss of mucous NC markers as ZCs form.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Celulas Principais Gástricas/citologia , Celulas Principais Gástricas/fisiologia , Proteínas de Ligação a DNA/genética , Retículo Endoplasmático Rugoso/fisiologia , Fatores de Transcrição/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/fisiologia , Linhagem Celular , Celulas Principais Gástricas/ultraestrutura , Proteínas de Ligação a DNA/metabolismo , Integrases/genética , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Regiões Promotoras Genéticas/fisiologia , Fatores de Transcrição de Fator Regulador X , Vesículas Secretórias/fisiologia , Células-Tronco/citologia , Células-Tronco/fisiologia , Fatores de Transcrição/metabolismo , Proteína 1 de Ligação a X-Box
7.
Gastroenterology ; 139(6): 2028-2037.e9, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20854822

RESUMO

BACKGROUND & AIMS: Gastric cancer evolves in the setting of a pathologic mucosal milieu characterized by both loss of acid-secreting parietal cells and mucous cell metaplasias. Indeed, mucous cell metaplasia is considered the critical preneoplastic lesion for gastric cancer. Previous investigations have shown that infection of mice with Helicobacter felis or induction of acute parietal cell loss with the drug DMP-777 leads to the emergence of a type of metaplasia designated spasmolytic polypeptide-expressing metaplasia (SPEM). We have hypothesized that SPEM arises from proliferating cells in gland bases, either from a cryptic progenitor cell or by transdifferentiation of mature chief cells. METHODS: Taking advantage of the chief cell-restricted expression of Mist1-Cre-ER(T2), we used lineage mapping to examine whether SPEM lineages were derived from chief cells in 3 independent models of induction by DMP-777 treatment, L-635 treatment, or H felis infection. RESULTS: Treatment of mice with L-635 for 3 days led to rapid parietal cell loss, induction of a prominent inflammatory infiltrate, and emergence of SPEM. In all 3 models, SPEM developed, at least in part, from transdifferentiation of chief cells. We further found that acute parietal cell loss in the setting of inflammation (L-635 treatment) led to more rapid induction and expansion of SPEM derived from transdifferentiation of chief cells. CONCLUSIONS: These studies provide direct evidence by lineage tracing that SPEM evolves from differentiated chief cells. Thus, mature gastric chief cells have the ability to act as cryptic progenitors and reacquire proliferative capacity within the context of mucosal injury and inflammation.


Assuntos
Celulas Principais Gástricas/patologia , Gastrite/patologia , Lesões Pré-Cancerosas/patologia , Células-Tronco/patologia , Neoplasias Gástricas/patologia , Doença Aguda , Animais , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Linhagem da Célula/fisiologia , Celulas Principais Gástricas/fisiologia , Doença Crônica , Modelos Animais de Doenças , Gastrite/microbiologia , Gastrite/fisiopatologia , Infecções por Helicobacter/patologia , Infecções por Helicobacter/fisiopatologia , Helicobacter felis , Peptídeos e Proteínas de Sinalização Intercelular , Óperon Lac/genética , Metaplasia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Parietais Gástricas/patologia , Células Parietais Gástricas/fisiologia , Peptídeos/genética , Peptídeos/metabolismo , Lesões Pré-Cancerosas/microbiologia , Lesões Pré-Cancerosas/fisiopatologia , Células-Tronco/fisiologia , Neoplasias Gástricas/microbiologia , Neoplasias Gástricas/fisiopatologia
8.
Gastroenterology ; 139(6): 2018-2027.e2, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20708616

RESUMO

BACKGROUND & AIMS: Gastric stem cells are located in the isthmus of the gastric glands and give rise to epithelial progenitors that undergo bipolar migration and differentiation into pit and oxyntic lineages. Although gastric mucus neck cells located below the isthmus express trefoil factor family 2 (TFF2) protein, TFF2 messenger RNA transcripts are concentrated in cells above the neck region in normal corpus mucosa, suggesting that TFF2 transcription is a marker of gastric progenitor cells. METHODS: Using a BAC strategy, we generated a transgenic mouse with a tamoxifen-inducible Cre under the control of the TFF2 promoter (TFF2-BAC-Cre(ERT2)) and analyzed the lineage derivation from TFF2 mRNA transcript-expressing (TTE) cells. RESULTS: TTE cells were localized to the isthmus, above and distinct from TFF2 protein-expressing mucus neck cells. Lineage tracing revealed that these cells migrated toward the bottom of the gland within 20 days, giving rise to parietal, mucous neck, and chief cells, but not to enterochromaffin-like-cell. Surface mucus cells were not derived from TTE cells and the progeny of the TTE lineage did not survive beyond 200 days. TTE cells were localized in the isthmus adjacent to doublecortin CaM kinase-like-1(+) putative progenitor cells. Induction of spasmolytic polypeptide-expressing metaplasia with DMP-777-induced acute parietal cell loss revealed that this metaplastic phenotype might arise in part through transdifferentiation of chief cells as opposed to expansion of mucus neck or progenitor cells. CONCLUSIONS: TFF2 transcript-expressing cells are progenitors for mucus neck, parietal and zymogenic, but not for pit or enterochromaffin-like cell lineages in the oxyntic gastric mucosa.


Assuntos
Mucosa Gástrica/citologia , Mucosa Gástrica/fisiologia , Mucinas/genética , Proteínas Musculares/genética , Peptídeos/genética , Células-Tronco/citologia , Células-Tronco/fisiologia , Animais , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Celulas Principais Gástricas/citologia , Celulas Principais Gástricas/fisiologia , Duodeno/citologia , Duodeno/fisiologia , Células Enterocromafins/citologia , Células Enterocromafins/fisiologia , Integrases/genética , Rim/citologia , Rim/fisiologia , Pulmão/citologia , Pulmão/fisiologia , Camundongos , Camundongos Transgênicos , Células Parietais Gástricas/citologia , Células Parietais Gástricas/fisiologia , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , Transcrição Gênica/fisiologia , Fator Trefoil-2
9.
Dig Dis Sci ; 55(12): 3339-48, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20437101

RESUMO

BACKGROUND: The widespread use of proton pump inhibitors (PPIs) is known to cause sporadic gastric fundic gland polyps (FGPs). Altered expression and localization of the water or ion transport proteins might contribute to the excess fluid secretion into the cystic lumen for the development of FGPs. AIMS: We investigated the alteration of the murine gastric fundic mucosa after PPI treatment, and examined the expression of water channel aquaporin-4 (AQP4) and potassium channel KCNQ1, which are expressed only in the parietal cells in the gastric mucosa. METHODS: Male 5-week-old C57BL/6J mice were administered lansoprazole (LPZ) by subcutaneous injection for 8 weeks. The expression of AQP4 and KCNQ1 were investigated by Western blotting, quantitative RT-PCR, and immunohistochemistry. The expression of mucin-6 (Muc6), pepsinogen, and sonic hedgehog (Shh) were also investigated as mucosal cell lineage markers. RESULTS: Gastric mucosal hyperplasia with multiple cystic dilatations, exhibiting similar histological findings to the FGPs, was observed in the LPZ-treated mice. An increase in the number of AQP4-positive parietal cells and KCNQ1-positive parietal cells was observed. The extension of the distribution of AQP4-positive cells toward the surface of the fundic glands was also observed. The expression levels of AQP4 mRNA and protein were significantly enhanced. The expression of KCNQ1 mRNA was correlated with that of AQP4 mRNA in the LPZ-treated mice. Mucous neck-to-zymogenic cell lineage differentiation was delayed in association with decreased expression of Shh in the LPZ-treated mice. CONCLUSIONS: PPI administration increased the number of parietal cells with enhanced expression of AQP4 and KCNQ1.


Assuntos
2-Piridinilmetilsulfinilbenzimidazóis/farmacologia , Aquaporina 4/metabolismo , Ácido Gástrico/metabolismo , Canal de Potássio KCNQ1/metabolismo , Células Parietais Gástricas/metabolismo , Inibidores da Bomba de Prótons/farmacologia , Animais , Western Blotting , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula , Celulas Principais Gástricas/fisiologia , Hiperplasia , Imuno-Histoquímica , Lansoprazol , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células Parietais Gástricas/patologia
10.
Am J Physiol Gastrointest Liver Physiol ; 289(3): G521-9, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15933222

RESUMO

Muscarinic cholinergic mechanisms play a key role in stimulating gastric pepsinogen secretion. Studies using antagonists suggested that the M3 receptor subtype (M3R) plays a prominent role in mediating pepsinogen secretion, but in situ hybridization indicated expression of M1 receptor (M1R) in rat chief cells. We used mice that were deficient in either the M1 (M1R-/-) or M3 (M3R-/-) receptor or that lacked both receptors (M(1/3)R-/-) to determine the role of M1R and M3R in mediating cholinergic agonist-induced pepsinogen secretion. Pepsinogen secretion from murine gastric glands was determined by adapting methods used for rabbit and rat stomach. In wild-type (WT) mice, maximal concentrations of carbachol and CCK caused a 3.0- and 2.5-fold increase in pepsinogen secretion, respectively. Maximal carbachol-induced secretion from M1R-/- mouse gastric glands was decreased by 25%. In contrast, there was only a slight decrease in carbachol potency and no change in efficacy when comparing M3R-/- with WT glands. To explore the possibility that both M1R and M3R are involved in carbachol-mediated pepsinogen secretion, we examined secretion from glands prepared from M(1/3)R-/- double-knockout mice. Strikingly, carbachol-induced pepsinogen secretion was nearly abolished in glands from M(1/3)R-/- mice, whereas CCK-induced secretion was not altered. In situ hybridization for murine M1R and M3R mRNA in gastric mucosa from WT mice revealed abundant signals for both receptor subtypes in the cytoplasm of chief cells. These data clearly indicate that, in gastric chief cells, a mixture of M1 and M3 receptors mediates cholinergic stimulation of pepsinogen secretion and that no other muscarinic receptor subtypes are involved in this activity. The development of a murine secretory model facilitates use of transgenic mice to investigate the regulation of pepsinogen secretion.


Assuntos
Carbacol/farmacologia , Celulas Principais Gástricas/fisiologia , Agonistas Colinérgicos/farmacologia , Pepsinogênio A/metabolismo , Receptor Muscarínico M1/fisiologia , Receptor Muscarínico M3/fisiologia , Animais , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Receptor Muscarínico M1/genética , Receptor Muscarínico M3/genética
11.
J Infect Dis ; 191(4): 571-8, 2005 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-15655781

RESUMO

BACKGROUND: Infection of the gastric mucosa with Helicobacter pylori leads to increased apoptosis. Cytokines and receptors of the tumor necrosis factor (TNF) family are known to be involved in this process. The role that the death-inducing TNF- alpha -related apoptosis-inducing ligand (TRAIL) and its receptors play, in the context of H. pylori infection, is unknown. METHODS: In 74 H. pylori-infected and 51 H. pylori-uninfected gastric antral biopsy specimens, levels of TRAIL mRNA and TRAIL receptor mRNA were determined quantitatively by TaqMan reverse-transcriptase polymerase chain reaction. Recombinant TRAIL-induced apoptosis was measured in human and rat gastric epithelial cells by end-labeling of DNA with fluorescein-dTUP and by fluorescence-activated cell sorter analysis. RESULTS: In patients infected with cagA+/vacAs1+ H. pylori strains, expression of TRAIL and the proapoptotic receptors TRAIL-R1 and -R2 was down-regulated, whereas expression of the antiapoptotic receptors TRAIL-R3 and -R4 was up-regulated. Furthermore, expression of TRAIL and TRAIL-R1 and -R2 correlated inversely with the severity of gastric inflammation. Significant apoptosis of isolated human gastric epithelial cells and highly enriched rat parietal and chief cells was induced by 100 ng/mL TRAIL. CONCLUSIONS: Down-regulation of the TRAIL system, in the context of H. pylori infection, may limit exaggerated apoptosis of gastric epithelial cells and destruction of tissue and, therefore, may enable H. pylori to maintain its niche.


Assuntos
Apoptose , Mucosa Gástrica/patologia , Infecções por Helicobacter/metabolismo , Helicobacter pylori/patogenicidade , Glicoproteínas de Membrana/biossíntese , Receptores do Fator de Necrose Tumoral/biossíntese , Fator de Necrose Tumoral alfa/biossíntese , Adulto , Idoso , Animais , Antígenos de Bactérias/biossíntese , Proteínas Reguladoras de Apoptose , Proteínas de Bactérias/biossíntese , Células Cultivadas , Celulas Principais Gástricas/citologia , Celulas Principais Gástricas/fisiologia , DNA/metabolismo , Regulação para Baixo , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Feminino , Citometria de Fluxo , Proteínas Ligadas por GPI , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiologia , Regulação da Expressão Gênica , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/patologia , Humanos , Masculino , Glicoproteínas de Membrana/fisiologia , Pessoa de Meia-Idade , Células Parietais Gástricas/citologia , Células Parietais Gástricas/fisiologia , RNA Mensageiro/análise , Ratos , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Membro 10c de Receptores do Fator de Necrose Tumoral , Ligante Indutor de Apoptose Relacionado a TNF , Receptores Chamariz do Fator de Necrose Tumoral , Fator de Necrose Tumoral alfa/fisiologia
12.
Oncogene ; 23(20): 3572-9, 2004 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-15116092

RESUMO

Reg (regenerating gene product) was originally identified as a growth factor involved in pancreatic regeneration. During the healing course of gastric erosion, Reg expression is highly increased in the enterochromaffin-like (ECL) cells surrounding the ulcer crater, suggesting its role as a regulator of gastric mucosal regeneration. However, there has been no direct in vivo evidence of a growth-promoting role of Reg for the gastric mucosal cells. In the current study, Reg-transgenic mice were created and gastric mucosa were analysed for histological changes. Transgenic mice showed a marked increase in the thickness of the fundic mucosa. Anti-proliferating cell nuclear antigen (PCNA) staining of the fundic mucosa demonstrated the enlargement of the proliferating neck zone and the lower PCNA-negative zone. Histological analysis employing antibodies against cell-type markers revealed expansion of the chief cell and parietal cell populations and no change in the number of surface mucus-producing cells, ECL cells, or G cells. In conclusion, Reg has a growth-promoting effect on gastric progenitor cells and an activity to direct the differentiation of the cells into chief cell and parietal cell lineages. This was in contrast to other factors, all of which had been shown to drive differentiation towards mucus producing cells in vivo. In the injured gastric mucosa, Reg may play a unique and important part in the reconstruction of the properly organized mucosal architecture.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Diferenciação Celular/fisiologia , Celulas Principais Gástricas/fisiologia , Glicoproteínas/genética , Células Parietais Gástricas/fisiologia , Animais , Apoptose/fisiologia , Proteínas de Ligação ao Cálcio/metabolismo , Divisão Celular/fisiologia , Mucosa Gástrica/citologia , Mucosa Gástrica/crescimento & desenvolvimento , Mucosa Gástrica/patologia , Glicoproteínas/metabolismo , Litostatina , Camundongos , Camundongos Transgênicos
13.
Dig Dis Sci ; 48(8): 1431-44, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12924634

RESUMO

Besides their known physiological actions, bile acids are signaling molecules that alter cell function by interacting with muscarinic and nuclear receptors. Bile acid interaction with nuclear receptors modulates bile acid and cholesterol metabolism, whereas the potential consequences of muscarinic receptor activation are much broader. This review examines recent discoveries regarding bile acid interaction with muscarinic receptors. Selective and functional bile acid interaction has been reported with M3 receptors expressed in guinea pig gastric chief cells, human colon cancer cells, and transfected Chinese hamster ovary cells. Interaction of bile acids with chief cells may contribute to mucosal damage and other pathophysiological consequences of bile reflux. Bile acid-induced stimulation of muscarinic receptors on colon cancer cells may contribute to cellular proliferation and neoplasia. Potential consequences of bile acid interaction with muscarinic receptors on gastrointestinal myocytes, biliary epithelium, vascular endothelium and dermal neurons are discussed. Elucidation of molecular mechanisms underlying interaction of bile acids with muscarinic receptors may suggest new treatments for conditions that result from such interactions.


Assuntos
Celulas Principais Gástricas/fisiologia , Neoplasias do Colo/fisiopatologia , Ácido Litocólico/análogos & derivados , Receptores Muscarínicos/fisiologia , Transdução de Sinais/fisiologia , Acetilcolina/antagonistas & inibidores , Acetilcolina/fisiologia , Animais , Células CHO , Linhagem Celular Transformada , Celulas Principais Gástricas/efeitos dos fármacos , Cricetinae , Cobaias , Humanos , Ácido Litocólico/farmacologia , Receptores Muscarínicos/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
14.
Stem Cells ; 21(3): 322-36, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12743327

RESUMO

In the human stomach, the oxyntic epithelium includes numerous tubular invaginations consisting of short pits opening into long glands. The pit is lined by pit cells, whereas the gland is composed of three regions: the base, containing zymogenic cells; the neck, containing neck cells; and the isthmus, composed of little known immature cells and of parietal cells, which are also scattered through the neck and base. The aim of this study was to examine the ultrastructure of the immature cells and to determine their relation to mature cells. To do so, normal oxyntic mucosal biopsies from subjects ranging from 20-43 years old were fixed in aldehydes and postfixed in reduced osmium for electron microscopy and morphometric analysis. The immature cells were sorted out into four classes, whose roles were clarified by comparison with the thoroughly investigated mouse oxyntic epithelium. The first class was composed of the least differentiated immature cells, which were rare and characterized by minute, dense, or cored secretory granules and were accordingly named mini-granule cells. Their function was not clarified. The second class consisted of pre-pit cells, which were characterized by few dense mucous granules and give rise to pit cells that ascend the pit wall and, after reaching the luminal surface, die or are extruded. Both pre-pit and pit cells underwent continuous renewal and, therefore, together constituted a renewal system referred to as pit cell lineage. The third class, or pre-neck cells, characterized by cored secretory granules, give rise to neck cells that descend toward the base region and differentiate further into pre-zymogenic cells, which finally become zymogenic cells. The latter eventually degenerate and die. Thus pre-neck cells and their progeny constitute a renewing system, designated zymogenic cell lineage. The fourth class, or pre-parietal cells, characterized by long microvilli and few tubulovesicles, differentiate into parietal cells that descend along the neck and base regions and eventually degenerate and die. Pre-parietal and parietal cells represent a renewing system referred to as parietal cell lineage. While the origin of the last three classes of progenitor cells has not been elucidated, it is likely that they arise either from an unidentified multipotential stem cell, possibly the mini-granule cell itself, or from the mitotic activity of pre-pit and pre-neck cells. In conclusion, the human oxyntic epithelium is composed of continually renewing cells organized in distinct cell lineages.


Assuntos
Células Epiteliais/ultraestrutura , Mucosa Gástrica/ultraestrutura , Células Parietais Gástricas/ultraestrutura , Células-Tronco/ultraestrutura , Adulto , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Núcleo Celular/fisiologia , Núcleo Celular/ultraestrutura , Polaridade Celular/fisiologia , Celulas Principais Gástricas/fisiologia , Celulas Principais Gástricas/ultraestrutura , Citoplasma/fisiologia , Citoplasma/ultraestrutura , Células Epiteliais/fisiologia , Mucosa Gástrica/fisiologia , Humanos , Microscopia Eletrônica , Células-Tronco Multipotentes/fisiologia , Células-Tronco Multipotentes/ultraestrutura , Organelas/fisiologia , Organelas/ultraestrutura , Células Parietais Gástricas/fisiologia , Vesículas Secretórias/fisiologia , Vesículas Secretórias/ultraestrutura , Células-Tronco/fisiologia
16.
Am J Physiol ; 274(6): G997-1004, 1998 06.
Artigo em Inglês | MEDLINE | ID: mdl-9696723

RESUMO

Although bile acids damage gastric mucosa, the mechanisms underlying tissue injury induced by these agents are not well understood. To determine whether bile acids alter gastric secretory function, we investigated the actions of sodium cholate, deoxycholate, lithocholate, and their taurine and glycine conjugates on a highly homogeneous population of gastric chief cells. Lithocholyltaurine (LCT), a particularly injurious bile acid, caused a threefold increase in pepsinogen secretion (detectable with 100 nM and maximal with 10 microM LCT). When combined with other secretagogues, increasing concentrations of LCT caused progressive inhibition of carbamylcholine (carbachol)-induced pepsinogen secretion but did not alter CCK- or 8-bromo-cAMP-induced secretion. Taurine and unconjugated lithocholate did not alter basal or carbachol-induced secretion. These observations suggested that LCT is a partial cholinergic agonist. To test this hypothesis, we examined the actions of the cholinergic antagonist atropine on LCT-induced pepsinogen secretion. Atropine (10 microM) abolished carbachol- and LCT-induced pepsinogen secretion. Likewise, carbachol (0.1 mM) and LCT (1 mM) induced an atropine-sensitive, two- to threefold increase in cellular levels of inositol 1,4,5-trisphosphate. We examined the actions of LCT on binding of the cholinergic radioligand [N-methyl-3H]scopolamine ([3H]NMS) to chief cells. Half-maximal inhibition of [3H]NMS binding was observed with approximately 0.5 mM carbachol and 1 mM LCT. These results indicate that the bile acid LCT is a partial agonist for muscarinic cholinergic receptors on gastric chief cells.


Assuntos
Celulas Principais Gástricas/fisiologia , Colagogos e Coleréticos/farmacologia , Receptores Colinérgicos/efeitos dos fármacos , Receptores Colinérgicos/fisiologia , Ácido Taurolitocólico/farmacologia , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Animais , Atropina/farmacologia , Carbacol/farmacologia , Colecistocinina/farmacologia , Cobaias , Masculino , N-Metilescopolamina/metabolismo , Parassimpatolíticos/farmacologia , Pepsinogênios/metabolismo , Sincalida/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA