Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
1.
Medicine (Baltimore) ; 100(21): e25861, 2021 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-34032698

RESUMO

RATIONALE: Gastric adenocarcinoma of fundic gland (chief cell predominant type) (GA-FG-CCP) is a new, rare variant of gastric adenocarcinoma, which is characterized by mild nuclear atypia and specific immunohistochemical markers. PATIENT CONCERNS: An 84-year-old Chinese man was referred to our hospital for endoscopic resection of a gastric lesion. INTERVENTIONS: We performed endoscopic submucosal dissection, and successfully removed the lesion. DIAGNOSIS: Esophago gastroduodenoscopy showed a slightly elevated lesion with a diameter of 22 mm in the posterior wall of cardia. Magnifying endoscopy with narrow band imaging revealed an abnormal microsurface and microvessels on the tumor surface. Endoscopic ultrasonography revealed a hypoechoic mass located in the first layer. The pathological diagnosis of the biopsy specimens indicated that the tumor was high grade intraepithelial neoplasia. The pathological diagnosis differed between the superficial and deeper part of the lesion. The superficial part was composed of a tubular structure with prominent atypia and was diagnosed as well differentiated intestinal adenocarcinoma. The deeper part was composed of a well-differentiated tubular adenocarcinoma mimicking the fundic gland cells, mainly the chief cells. The tumor cells showed mild nuclear atypia and was positive for pepsinogen-I (PG-I) and mucin-6 (MUC6). This deeper part was diagnosed as GA-FG-CCP. OUTCOMES: The tumor was successfully removed. This patient had no discomfort during the follow-up period (10 months). LESSONS: We present a rare case of GA-FG-CCP coexisted with well-differentiated tubular adenocarcinoma. GA-FG-CCP exists in the deep mucosal layer and the muscularis mucosa, which could not be found under endoscopy, but could be discerned in pathology with mild nuclear atypia and special biomarkers.


Assuntos
Adenocarcinoma in Situ/diagnóstico , Adenocarcinoma/diagnóstico , Fundo Gástrico/patologia , Neoplasias Complexas Mistas/diagnóstico , Neoplasias Gástricas/diagnóstico , Adenocarcinoma/patologia , Adenocarcinoma/cirurgia , Adenocarcinoma in Situ/patologia , Adenocarcinoma in Situ/cirurgia , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/análise , Biópsia , Celulas Principais Gástricas/patologia , Endoscopia do Sistema Digestório , Endossonografia , Gastrectomia , Fundo Gástrico/citologia , Fundo Gástrico/diagnóstico por imagem , Fundo Gástrico/cirurgia , Humanos , Mucosa Intestinal/patologia , Masculino , Mucina-6/análise , Neoplasias Complexas Mistas/patologia , Neoplasias Complexas Mistas/cirurgia , Pepsinogênio A/análise , Neoplasias Gástricas/patologia , Neoplasias Gástricas/cirurgia , Resultado do Tratamento
2.
Neurogastroenterol Motil ; 32(6): e13824, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32096330

RESUMO

BACKGROUND: BALB/c and C57BL/6 mice are widely used in biomedical research; however, the differences between strains are still underestimated. Our aims were to develop an experimental protocol to evaluate the duodenal contractility and gastrointestinal transit in mice using the Alternating Current Biosusceptometry (ACB) technique and to compare gastrointestinal motor function and morphology between BALB/c and C57BL/6 strains. METHODS: Male mice were used in experiments (a) duodenal contractility: animals which had a magnetic marker surgically fixed in the duodenum to determine the frequency and amplitude of contractions and (b) gastrointestinal transit: animals which ingested a magnetically marked chow to calculate the Oro-Anal Transit Time (OATT) and the Fecal Pellet Elimination Rate (FPER). The animals were killed after the experiments for organ collection and morphometric analysis. KEY RESULTS: BALB/c and C57BL/6 had two different duodenal frequencies (high and low) with similar amplitudes. After 10 hours of monitoring, BALB/c eliminated around 89% of the ingested marker and C57BL/6 eliminated 33%; OATT and FPER were slower for C57BL/6 compared with BALB/c. The OATT and amplitude of low frequency had a strong positive correlation in C57BL/6. For BALB/c, the gastric muscular layer was thicker compared to that measured for C57BL/6. CONCLUSIONS AND INFERENCES: The experimental protocol to evaluate duodenal contractility and fecal magnetic pellets output using the ACB technique in mice was successfully established. BALB/c strains had higher duodenal frequencies and a shorter time to eliminate the ingested marker. Our results showed differences in both motor function and gastrointestinal morphology between BALB/c and C57BL/6 strains.


Assuntos
Duodeno/citologia , Duodeno/fisiologia , Fundo Gástrico/citologia , Motilidade Gastrointestinal , Animais , Trânsito Gastrointestinal , Laparotomia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Contração Muscular , Especificidade da Espécie
3.
Obes Surg ; 29(1): 76-83, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30178156

RESUMO

BACKGROUND: Weight loss after laparoscopic sleeve gastrectomy (LSG) has been mainly attributed to the restriction of gastric volume; however; other factors may contribute to weight loss after LSG. This study aimed to investigate the correlation between the number of ghrelin-secreting cells in the gastric fundus and excess weight loss (EWL) at 12 months after LSG. METHODS: The surface area of the gastric fundus was measured postoperatively in square centimeter. Histopathologic examination of the gastric fundus was made to estimate the number of ghrelin-secreting cells per square centimeter then was multiplied by the surface area of the fundus to calculate the total number of ghrelin-secreting cells in the fundus. The number of ghrelin-secreting cells was correlated with EWL and BMI at 12 months postoperatively. RESULTS: The present study included 39 patients of a mean age of 33.7 years. The mean %EWL at 12 months was 59.7 ± 12.7. The mean total number of ghrelin-producing cells in the gastric fundus was 26,228.4 ± 16,995.3. The total number of ghrelin-secreting cells had a weak positive correlation with BMI at 12 months (r = 0.2891, p = 0.07), and weak negative correlation with %EWL (r = - 0.1592, p = 0.33). CONCLUSION: There was a weak correlation between the total number of ghrelin-producing cells in the gastric fundus and plasma ghrelin levels with EWL after LSG.


Assuntos
Gastrectomia/estatística & dados numéricos , Fundo Gástrico , Grelina , Obesidade Mórbida , Redução de Peso/fisiologia , Adulto , Estudos de Coortes , Fundo Gástrico/citologia , Fundo Gástrico/metabolismo , Fundo Gástrico/cirurgia , Grelina/metabolismo , Humanos , Obesidade Mórbida/epidemiologia , Obesidade Mórbida/metabolismo , Obesidade Mórbida/cirurgia
4.
Cell Tissue Res ; 376(1): 37-49, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30467709

RESUMO

Gastric endocrine cell hormones contribute to the control of the stomach and to signalling to the brain. In other gut regions, enteroendocrine cells (EECs) exhibit extensive patterns of colocalisation of hormones. In the current study, we characterise EECs in the human gastric fundus and corpus. We utilise immunohistochemistry to investigate EECs with antibodies to ghrelin, serotonin (5-HT), somatostatin, peptide YY (PYY), glucagon-like peptide 1, calbindin, gastrin and pancreastatin, the latter as a marker of enterochromaffin-like (ECL) cells. EECs were mainly located in regions of the gastric glands populated by parietal cells. Gastrin cells were absent and PYY cells were very rare. Except for about 25% of 5-HT cells being a subpopulation of ECL cells marked by pancreastatin, colocalisation of hormones in gastric EECs was infrequent. Ghrelin cells were distributed throughout the fundus and corpus; most were basally located in the glands, often very close to parietal cells and were closed cells i.e., not in contact with the lumen. A small proportion had long processes located close to the base of the mucosal epithelium. The 5-HT cells were of at least three types: small, round, closed cells; cells with multiple, often very long, processes; and a subgroup of ECL cells. Processes were in contact with their surrounding cells, including parietal cells. Mast cells had very weak or no 5-HT immunoreactivity. Somatostatin cells were a closed type with long processes. In conclusion, four major chemically defined EEC types occurred in the human oxyntic mucosa. Within each group were cells with distinct morphologies and relationships to other mucosal cells.


Assuntos
Células Enteroendócrinas , Fundo Gástrico , Hormônios Gastrointestinais/análise , Células Enteroendócrinas/química , Células Enteroendócrinas/citologia , Feminino , Fundo Gástrico/citologia , Fundo Gástrico/metabolismo , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Obesidade/cirurgia
5.
Nat Protoc ; 14(1): 28-50, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30470820

RESUMO

The human stomach contains two primary domains: the corpus, which contains the fundic epithelium, and the antrum. Each of these domains has distinct cell types and functions, and therefore each presents with unique disease pathologies. Here, we detail two protocols to differentiate human pluripotent stem cells (hPSCs) into human gastric organoids (hGOs) that recapitulate both domains. Both protocols begin with the differentiation of hPSCs into definitive endoderm (DE) using activin A, followed by the generation of free-floating 3D posterior foregut spheroids using FGF4, Wnt pathway agonist CHIR99021 (CHIR), BMP pathway antagonist Noggin, and retinoic acid. Embedding spheroids in Matrigel and continuing 3D growth in epidermal growth factor (EGF)-containing medium for 4 weeks results in antral hGOs (hAGOs). To obtain fundic hGOs (hFGOs), spheroids are additionally treated with CHIR and FGF10. Induced differentiation of acid-secreting parietal cells in hFGOs requires temporal treatment of BMP4 and the MEK inhibitor PD0325901 for 48 h on protocol day 30. In total, it takes ~34 d to generate hGOs from hPSCs. To date, this is the only approach that generates functional human differentiated gastric cells de novo from hPSCs.


Assuntos
Técnicas de Cultura de Células , Endoderma/citologia , Células Epiteliais/citologia , Fundo Gástrico/citologia , Organoides/citologia , Células-Tronco Pluripotentes/citologia , Antro Pilórico/citologia , Ativinas/farmacologia , Benzamidas/farmacologia , Proteínas de Transporte/farmacologia , Diferenciação Celular , Colágeno/química , Meios de Cultura/química , Meios de Cultura/farmacologia , Difenilamina/análogos & derivados , Difenilamina/farmacologia , Combinação de Medicamentos , Endoderma/efeitos dos fármacos , Endoderma/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Fator 10 de Crescimento de Fibroblastos/farmacologia , Fator 4 de Crescimento de Fibroblastos/farmacologia , Fundo Gástrico/metabolismo , Humanos , Laminina/química , Especificidade de Órgãos , Organoides/efeitos dos fármacos , Organoides/metabolismo , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo , Proteoglicanas/química , Antro Pilórico/metabolismo , Piridinas/farmacologia , Pirimidinas/farmacologia , Tretinoína/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos
6.
Anat Rec (Hoboken) ; 301(12): 2128-2144, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30382612

RESUMO

The fundic glands of the stomach contain two types of mucous cells: surface mucous cells (SMCs) located at the surface of the stomach and the pits, and mucous neck cells (MNCs) situated in the neck of the glands. They produce mucins, highly glycosylated proteins. Very little is known about the glycan composition of these mucins and of gastric secretion in general. We used several lectins combined with deglycosylation pretreatments to analyze the glycan composition of SMCs and MNCs. The results showed the presence of terminal sialic acid and subterminal Gal and GalNAc, which is consistent with previous knowledge about glycosylation in mucins. Our results also support previous reports that showed a different expression of mucins in the SMCs, depending on their superficial or deep location in the pit. Some lectins labeled only the perinuclear region of the SMCs, but not the apical region, where the secretory granules are stored. This suggests that the lectins are labeling sugar residues that are accessible to lectins during the first steps of glycan synthesis, which occurs in the endoplasmic reticulum and Golgi apparatus. Our results indicate that SMCs and MNCs produce a mucus secretion with a different glycoconjugate composition. The secretion is more varied in SMCs. As our results coincide with what we know about glycosylation of mucins, we can conclude that most of the glycans detected belong to mucins, and the differences in glycosylation observed in each cell type may be due, mainly, to the different secreted mucins. Anat Rec, 301:2128-2144, 2018. © 2018 Wiley Periodicals, Inc.


Assuntos
Fundo Gástrico/citologia , Fundo Gástrico/metabolismo , Mucosa Gástrica/citologia , Mucosa Gástrica/metabolismo , Glicoconjugados/metabolismo , Muco/metabolismo , Animais , Fundo Gástrico/química , Mucosa Gástrica/química , Glicoconjugados/análise , Masculino , Mucinas/análise , Mucinas/metabolismo , Muco/química , Ratos , Ratos Sprague-Dawley
7.
J Physiol ; 596(11): 2131-2146, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29528115

RESUMO

KEY POINTS: Activation of focal adhesion kinase (FAK) by integrin signalling facilitates smooth muscle contraction by transmitting the force generated by myofilament activation to the extracellular matrix and throughout the smooth muscle tissue. Here we report that electrical field stimulation (EFS) of cholinergic motor neurons activates FAK in gastric fundus smooth muscles, and that FAK activation by EFS is atropine-sensitive but nicardipine-insensitive. PDBu and calyculin A contracted gastric fundus muscles Ca2+ -independently and also activated FAK. Inhibition of FAK activation inhibits the contractile responses evoked by EFS, and inhibits CPI-17 phosphorylation at T38. This study indicates that mechanical force or tension is sufficient to activate FAK, and that FAK appears to be involved in the activation of the protein kinase C-CPI-17 Ca2+ sensitization pathway in gastric fundus smooth muscles. These results reveal a novel role for FAK in gastric fundus smooth muscle contraction by facilitating CPI-17 phosphorylation. ABSTRACT: Smooth muscle contraction involves regulating myosin light chain phosphorylation and dephosphorylation by myosin light chain kinase and myosin light chain phosphatase. C-kinase potentiated protein phosphatase-1 inhibitor of 17 kDa (CPI-17) and myosin phosphatase targeting subunit of myosin light-chain phosphatase (MYPT1) are crucial for regulating gastrointestinal smooth muscle contraction by inhibiting myosin light chain phosphatase. Integrin signalling involves the dynamic recruitment of several proteins, including focal adhesion kinase (FAK), to focal adhesions. FAK tyrosine kinase activation is involved in cell adhesion to the extracellular matrix via integrin signalling. FAK participates in linking the force generated by myofilament activation to the extracellular matrix and throughout the smooth muscle tissue. Here, we show that cholinergic stimulation activates FAK in gastric fundus smooth muscles. Electrical field stimulation in the presence of Nω -nitro-l-arginine methyl ester and MRS2500 contracted gastric fundus smooth muscle strips and increased FAK Y397 phosphorylation (pY397). Atropine blocked the contractions and prevented the increase in pY397. The FAK inhibitor PF-431396 inhibited the contractions and the increase in pY397. PF-431396 also inhibited the electrical field stimulation-induced increase in CPI-17 T38 phosphorylation, and reduced MYPT1 T696 and T853, and myosin light chain S19 phosphorylation. Ca2+ influx was unaffected by PF-431396. Nicardipine inhibited the contractions but had no effect on the increase in pY397. Phorbol 12,13-dibutyrate or calyculin A contracted gastric fundus smooth muscle strips Ca2+ independently and increased pY397. Our findings suggest that FAK is activated by mechanical forces during contraction and reveal a novel role of FAK in the regulation of CPI-17 phosphorylation.


Assuntos
Quinase 1 de Adesão Focal/metabolismo , Quinase 2 de Adesão Focal/metabolismo , Fundo Gástrico/fisiologia , Contração Muscular , Proteínas Musculares/metabolismo , Músculo Liso/fisiologia , Animais , Cálcio/metabolismo , Células Cultivadas , Neurônios Colinérgicos/citologia , Neurônios Colinérgicos/fisiologia , Estimulação Elétrica , Fundo Gástrico/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios Motores/citologia , Neurônios Motores/fisiologia , Músculo Liso/citologia , Fosforilação , Transdução de Sinais
8.
J Physiol ; 596(9): 1549-1574, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29430647

RESUMO

KEY POINTS: Enteric neurotransmission is essential for gastrointestinal (GI) motility, although the cells and conductances responsible for post-junctional responses are controversial. The calcium-activated chloride conductance (CaCC), anoctamin-1 (Ano1), was expressed by intramuscular interstitial cells of Cajal (ICC-IM) in proximal stomach and not resolved in smooth muscle cells (SMCs). Cholinergic nerve fibres were closely apposed to ICC-IM. Conductances activated by cholinergic stimulation in isolated ICC-IM and SMCs were determined. A CaCC was activated by carbachol in ICC-IM and a non-selective cation conductance in SMCs. Responses to cholinergic nerve stimulation were studied. Excitatory junction potentials (EJPs) and mechanical responses were evoked in wild-type mice but absent or greatly reduced with knockout/down of Ano1. Drugs that block Ano1 inhibited the conductance activated by carbachol in ICC-IM and EJPs and mechanical responses in tissues. The data of the present study suggest that electrical and mechanical responses to cholinergic nerve stimulation are mediated by Ano1 expressed in ICC-IM and not SMCs. ABSTRACT: Enteric motor neurotransmission is essential for normal gastrointestinal (GI) motility. Controversy exists regarding the cells and ionic conductance(s) that mediate post-junctional neuroeffector responses to motor neurotransmitters. Isolated intramuscular ICC (ICC-IM) and smooth muscle cells (SMCs) from murine fundus muscles were used to determine the conductances activated by carbachol (CCh) in each cell type. The calcium-activated chloride conductance (CaCC), anoctamin-1 (Ano1) is expressed by ICC-IM but not resolved in SMCs, and CCh activated a Cl- conductance in ICC-IM and a non-selective cation conductance in SMCs. We also studied responses to nerve stimulation using electrical-field stimulation (EFS) of intact fundus muscles from wild-type and Ano1 knockout mice. EFS activated excitatory junction potentials (EJPs) in wild-type mice, although EJPs were absent in mice with congenital deactivation of Ano1 and greatly reduced in animals in which the CaCC-Ano1 was knocked down using Cre/loxP technology. Contractions to cholinergic nerve stimulation were also greatly reduced in Ano1 knockouts. SMCs cells also have receptors and ion channels activated by muscarinic agonists. Blocking acetylcholine esterase with neostigmine revealed a slow depolarization that developed after EJPs in wild-type mice. This depolarization was still apparent in mice with genetic deactivation of Ano1. Pharmacological blockers of Ano1 also inhibited EJPs and contractile responses to muscarinic stimulation in fundus muscles. The data of the present study are consistent with the hypothesis that ACh released from motor nerves binds muscarinic receptors on ICC-IM with preference and activates Ano1. If metabolism of acetylcholine is inhibited, ACh overflows and binds to extrajunctional receptors on SMCs, eliciting a slower depolarization response.


Assuntos
Acetilcolina/metabolismo , Células Intersticiais de Cajal/fisiologia , Miócitos de Músculo Liso/fisiologia , Estômago/fisiologia , Transmissão Sináptica , Animais , Anoctamina-1/fisiologia , Canais de Cloreto/fisiologia , Estimulação Elétrica , Fundo Gástrico/citologia , Fundo Gástrico/fisiologia , Células Intersticiais de Cajal/citologia , Camundongos , Camundongos Knockout , Contração Muscular , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Estômago/citologia
9.
Tissue Cell ; 49(6): 746-750, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29089090

RESUMO

The epithelium of the gastric mucosa and its glands in the corpus of rat stomach contains mucous surface cells (MSCs), parietal cells, mucous neck cells (MNCs), zymogenic or chief cells (ZCs), several types of enteroendocrine cells, and intermediate cells with characteristics between MNCs and ZCs also called transitional or prezymogenic cells (pre-ZCs). The aim of our work was to analyze the expression of Mannose (Man) in the rat gastric glands by means of Galanthus nivalis lectin (GNA) histochemistry to identify the differences between MNC, pre-ZCs and ZCs and to establish the relationships between these cells. Most of the cytoplasm of MNCs was negative for GNA histochemistry. Intensity of GNA labeling in the gastric gland showed a graduation from pre-ZCs (weak labeling) to ZCs (moderate labeling). Labeling of ZCs was stronger at the perinuclear and apical cytoplasm. In the last years, strong evidence has been reported supporting that ZCs differentiate from MNCs. Our work also supports the origin of ZCs from MNCs, because the GNA labeling graduation might be due to oligosaccharides which are not expressed in MNCs, start to express in pre-ZCs and are more abundant in ZCs, indicating that differentiation from MNCs to ZCs is a process in which glycans with Man moieties are synthesized.


Assuntos
Transdiferenciação Celular , Celulas Principais Gástricas/citologia , Mucosa Gástrica/citologia , Animais , Células Epiteliais/citologia , Fundo Gástrico/citologia , Imuno-Histoquímica , Masculino , Manose/análise , Lectinas de Ligação a Manose , Lectinas de Plantas , Ratos , Ratos Sprague-Dawley
10.
Nature ; 541(7636): 182-187, 2017 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-28052057

RESUMO

Despite the global prevalence of gastric disease, there are few adequate models in which to study the fundus epithelium of the human stomach. We differentiated human pluripotent stem cells (hPSCs) into gastric organoids containing fundic epithelium by first identifying and then recapitulating key events in embryonic fundus development. We found that disruption of Wnt/ß-catenin signalling in mouse embryos led to conversion of fundic to antral epithelium, and that ß-catenin activation in hPSC-derived foregut progenitors promoted the development of human fundic-type gastric organoids (hFGOs). We then used hFGOs to identify temporally distinct roles for multiple signalling pathways in epithelial morphogenesis and differentiation of fundic cell types, including chief cells and functional parietal cells. hFGOs are a powerful model for studying the development of the human fundus and the molecular bases of human gastric physiology and pathophysiology, and also represent a new platform for drug discovery.


Assuntos
Fundo Gástrico/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Animais , Padronização Corporal , Diferenciação Celular , Linhagem da Célula , Descoberta de Drogas/métodos , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Epitélio/embriologia , Epitélio/metabolismo , Feminino , Fundo Gástrico/citologia , Fundo Gástrico/embriologia , Proteínas de Homeodomínio/metabolismo , Humanos , Masculino , Camundongos , Organoides/citologia , Organoides/embriologia , Organoides/metabolismo , Células Parietais Gástricas/citologia , Células Parietais Gástricas/metabolismo , Células-Tronco Pluripotentes/citologia , Fatores de Transcrição SOXB1/metabolismo , Esferoides Celulares/citologia , Esferoides Celulares/metabolismo , Transativadores/metabolismo , Via de Sinalização Wnt/genética , beta Catenina/agonistas
11.
World J Gastroenterol ; 22(48): 10523-10531, 2016 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-28082804

RESUMO

Gastric adenocarcinoma of the fundic gland (chief cell-predominant type, GA-FG-CCP) is a rare variant of well-differentiated adenocarcinoma, and has been proposed to be a novel disease entity. GA-FG-CCP originates from the gastric mucosa of the fundic gland region without chronic gastritis or intestinal metaplasia. The majority of GA-FG-CCPs exhibit either a submucosal tumor-like superficial elevated shape or a flat shape on macroscopic examination. Narrow-band imaging with endoscopic magnification may reveal a regular or an irregular microvascular pattern, depending on the degree of tumor exposure to the mucosal surface. Pathological analysis of GA-FG-CCPs is characterized by a high frequency of submucosal invasion, rare occurrences of lymphatic and venous invasion, and low-grade malignancy. Detection of diffuse positivity for pepsinogen-I by immunohistochemistry is specific for GA-FG-CCP. Careful endoscopic examination and detailed pathological evaluation are essential for early and accurate diagnosis of GA-FG-CCP. Nearly all GA-FG-CCPs are treated by endoscopic resection due to their small tumor size and low risk of recurrence or metastasis.


Assuntos
Adenocarcinoma/diagnóstico , Adenocarcinoma/patologia , Celulas Principais Gástricas/patologia , Fundo Gástrico/patologia , Gastroscopia/métodos , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/patologia , Biomarcadores Tumorais , Biópsia , Diagnóstico Diferencial , Fundo Gástrico/citologia , Fundo Gástrico/cirurgia , Humanos , Imuno-Histoquímica , Laparoscopia/métodos , Imagem de Banda Estreita/métodos , Recidiva Local de Neoplasia , Pepsinogênio A/imunologia , Pólipos/diagnóstico , Guias de Prática Clínica como Assunto , Neoplasias Gástricas/cirurgia
12.
J Physiol ; 591(24): 6193-208, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-24144881

RESUMO

Platelet-derived growth factor receptor α positive (PDGFRα(+)) cells are suggested to mediate purinergic inputs in GI muscles, but the responsiveness of these cells to purines in situ has not been evaluated. We developed techniques to label and visualize PDGFRα(+) cells in murine gastric fundus, load cells with Ca(2+) indicators, and follow their activity via digital imaging. Immunolabelling demonstrated a high density of PDGFRα(+) cells in the fundus. Cells were isolated and purified by fluorescence-activated cell sorting (FACS) using endogenous expression of enhanced green fluorescent protein (eGFP) driven off the Pdgfra promoter. Quantitative PCR showed high levels of expression of purinergic P2Y1 receptors and SK3 K(+) channels in PDGFRα(+) cells. Ca(2+) imaging was used to characterize spontaneous Ca(2+) transients and responses to purines in PDGFRα(+) cells in situ. ATP, ADP, UTP and ß-NAD elicited robust Ca(2+) transients in PDGFRα(+) cells. Ca(2+) transients were also elicited by the P2Y1-specific agonist (N)-methanocarba-2MeSADP (MRS-2365), and inhibited by MRS-2500, a P2Y1-specific antagonist. Responses to ADP, MRS-2365 and ß-NAD were absent in PDGFRα(+) cells from P2ry1((-/-)) mice, but responses to ATP were retained. Purine-evoked Ca(2+) transients were mediated through Ca(2+) release mechanisms. Inhibitors of phospholipase C (U-73122), IP3 (2-APB), ryanodine receptors (Ryanodine) and SERCA pump (cyclopiazonic acid and thapsigargin) abolished Ca(2+) transients elicited by purines. This study provides a link between purine binding to P2Y1 receptors and activation of SK3 channels in PDGFRα(+) cells. Activation of Ca(2+) release is likely to be the signalling mechanism in PDGFRα(+) cells responsible for the transduction of purinergic enteric inhibitory input in gastric fundus muscles.


Assuntos
Sinalização do Cálcio , Cálcio/metabolismo , Fibroblastos/metabolismo , Fundo Gástrico/citologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Difosfato de Adenosina/farmacologia , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Fibroblastos/efeitos dos fármacos , Citometria de Fluxo/métodos , Fundo Gástrico/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , NAD/farmacologia , Purinérgicos/farmacologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Fosfolipases Tipo C/antagonistas & inibidores
13.
PLoS One ; 8(9): e74608, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24058600

RESUMO

Regulation of myosin light chain phosphatase (MLCP) via protein kinase C (PKC) and the 17 kDa PKC-potentiated inhibitor of myosin light chain phosphatase (CPI-17) has been reported as a Ca(2+) sensitization signaling pathway in smooth muscle (SM), and thus may be involved in tonic vs. phasic contractions. This study examined the protein expression and spatial-temporal distribution of PKCα and CPI-17 in intact SM tissues. KCl or carbachol (CCh) stimulation of tonic stomach fundus SM generates a sustained contraction while the phasic stomach antrum generates a transient contraction. In addition, the tonic fundus generates greater relative force than phasic antrum with 1 µM phorbol 12, 13-dibutyrate (PDBu) stimulation which is reported to activate the PKCα - CPI-17 pathway. Western blot analyses demonstrated that this contractile difference was not caused by a difference in the protein expression of PKCα or CPI-17 between these two tissues. Immunohistochemical results show that the distribution of PKCα in the longitudinal and circular layers of the fundus and antrum do not differ, being predominantly localized near the SM cell plasma membrane. Stimulation of either tissue with 1 µM PDBu or 1 µM CCh does not alter this peripheral PKCα distribution. There are no differences between these two tissues for the CPI-17 distribution, but unlike the PKCα distribution, CPI-17 appears to be diffusely distributed throughout the cytoplasm under relaxed tissue conditions but shifts to a primarily peripheral distribution at the plasma membrane with stimulation of the tissues with 1 µM PDBu or 1 µM CCh. Results from double labeling show that neither PKCα nor CPI-17 co-localize at the adherens junction (vinculin/talin) at the membrane but they do co-localize with each other and with caveoli (caveolin) at the membrane. This lack of difference suggests that the PKCα - CPI-17 pathway is not responsible for the tonic vs. phasic contractions observed in stomach fundus and antrum.


Assuntos
Fundo Gástrico/enzimologia , Contração Muscular/fisiologia , Músculo Liso/fisiologia , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Proteína Quinase C-alfa/metabolismo , Antro Pilórico/enzimologia , Transdução de Sinais , Animais , Fenômenos Biomecânicos/efeitos dos fármacos , Carbacol/farmacologia , Fundo Gástrico/citologia , Fundo Gástrico/efeitos dos fármacos , Fundo Gástrico/fisiologia , Contração Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Dibutirato de 12,13-Forbol/farmacologia , Antro Pilórico/citologia , Antro Pilórico/efeitos dos fármacos , Antro Pilórico/fisiologia , Transdução de Sinais/efeitos dos fármacos , Sus scrofa
14.
Am J Physiol Gastrointest Liver Physiol ; 305(4): G314-24, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23764893

RESUMO

Previous studies have identified differences in the expression of proteins that regulate myosin light chain phosphorylation and contraction in tonic and phasic smooth muscle. cGMP plays a critical role in smooth muscle relaxation and is important for optimal function of phasic and tonic smooth muscle. The intracellular cGMP levels are regulated by its hydrolysis via phosphodiesterase 5 (PDE5) and efflux via novel multidrug resistance protein 5 (MRP5). In the present study we tested the hypothesis that the differences in the phasic and tonic behavior of smooth muscles may be related to differences in mechanisms that terminate cGMP signaling. Expression of PDE5 and MRP5 was significantly (more than 2-fold) higher in fundus compared with antrum. The NO donor S-nitrosoglutathione (GSNO) caused an increase in PDE5 activity and intra- and extracellular cGMP levels in both fundus and antrum. Stimulation of PDE5 activity and increase in extracellular cGMP were significantly higher in fundus, whereas increase in intracellular cGMP was significantly higher in antrum. GSNO-induced increase in extracellular cGMP was blocked in dispersed cells by the cyclic nucleotide export blocker probenecid and in cultured muscle cells by depletion of ATP or suppression of MRP5 by siRNA, providing evidence that cGMP efflux was mediated by ATP-dependent export via MRP5. Consistent with the higher expression and activity levels of PDE5 and MRP5, GSNO-induced PKG activity and muscle relaxation were significantly lower in muscle cells from fundus compared with antrum. Thus higher expression of PDE5 and MRP5 in muscle cells from fundus correlates with tonic phenotype of muscle.


Assuntos
GMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/metabolismo , Fundo Gástrico/enzimologia , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Contração Muscular , Músculo Liso/enzimologia , Miócitos de Músculo Liso/enzimologia , Antro Pilórico/enzimologia , Trifosfato de Adenosina/metabolismo , Animais , Células Cultivadas , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/genética , Relação Dose-Resposta a Droga , Fundo Gástrico/citologia , Fundo Gástrico/efeitos dos fármacos , Hidrólise , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Contração Muscular/efeitos dos fármacos , Relaxamento Muscular , Músculo Liso/citologia , Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/farmacologia , Fenótipo , Antro Pilórico/citologia , Antro Pilórico/efeitos dos fármacos , Interferência de RNA , RNA Mensageiro/metabolismo , Coelhos , Sistemas do Segundo Mensageiro , Fatores de Tempo , Transfecção
15.
In Vivo ; 24(5): 705-7, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20952737

RESUMO

BACKGROUND: Gastric parietal cells in a baboon were recently found to be auto-fluorescent. AIM: To study gastric sections with a fluorescent microscope in a cohort of baboons. MATERIAL AND METHODS: Gastric sections from 38 baboons were stained with hematoxylin-eosin (H&E) and examined in a fluorescence microscope (FLM). The thickness of the parietal cell population was assessed at x 10 magnification. RESULTS: H&E stained all mucosal cells: fovelar, parietal and chief cells. When the same sections were analyzed with an FLM, only parietal cells were auto-fluorescent, whereas fovelar and chief cells remained non-fluorescent. Parietal cells formed a distinct, continuous auto-fluorescent band. The ratio of the auto-fluorescent parietal cell band/total mucosa ranged between 0.20 and 0.30. CONCLUSION: Gastric parietal cells became auto-fluorescent when H&E-stained sections from baboon stomachs were observed with an FLM. Eosin was the stain responsible for this optical phenomenon.


Assuntos
Amarelo de Eosina-(YS) , Corantes Fluorescentes , Fundo Gástrico/citologia , Microscopia de Fluorescência/métodos , Células Parietais Gástricas/citologia , Animais , Celulas Principais Gástricas/citologia , Corantes , Hematoxilina , Papio
16.
Am J Physiol Cell Physiol ; 299(5): C1036-46, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20702689

RESUMO

Geometric measures of localized calcium release (LCR) events have been used to understand their biophysical basis. We found power law scaling between three such metrics-maximum amplitude (MA), mass above half-maximum amplitude (MHM), and area at half-maximum amplitude (AHM). In an effort to understand this scaling a minimal analytic model was employed to simulate LCR events recorded by confocal line scan. The distribution of logMHM as a function of logAHM, pMHM(pAHM), was dependent on model parameters such as channel open time, current size, line scan offset, and apparent diffusion coefficient. The distribution of log[MHM/AHM] as a function of logMA, p[MHM/AHM](pMA), was invariant, reflecting the gross geometry of the LCR event. The findings of the model were applied to real LCR line scan data from rabbit portal vein myocytes, rat cerebral artery myocytes, and guinea pig fundus knurled cells. pMHM(pAHM) could be used to distinguish two populations of LCR events in portal vein, even at the scale of "calcium noise," and to calculate the relative current of the two. The relative current was 2. pMHM(pAHM) could also be used to study pharmacological effects. The pMHM(pAHM) distribution of knurled cell LCR events was markedly contracted by ryanodine, suggesting a reduction in channel open time. The p[MHM/AHM](pMA) distributions were invariant across all cell types and were consistent with the model, underlying the common physical basis of their geometry. The geometric scaling of LCR events demonstrated here may help with their mechanistic characterization.


Assuntos
Cálcio/metabolismo , Modelos Biológicos , Miócitos de Músculo Liso/metabolismo , Algoritmos , Animais , Células Cultivadas , Simulação por Computador , Fundo Gástrico/citologia , Cobaias , Microscopia Confocal/métodos , Artéria Cerebral Média/citologia , Veia Porta/citologia , Coelhos , Ratos , Rianodina/metabolismo , Rianodina/farmacologia , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo
17.
Am J Physiol Gastrointest Liver Physiol ; 297(4): G814-24, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19643953

RESUMO

Intracellular recordings were made from isolated circular muscle bundles of rat gastric fundus. The majority of cells generated an ongoing discharge of electrical activity that were 15 min) resulted in the spread of dye between CSMC, between ICC-IM, and between CSMC and ICC-IM. Two types of STDs were observed, regularly occurring continuous STDs and irregular noisy bursting STDs. The amplitude of STDs varied between the two types of STDs. Single units summed to develop STDs with a maximum amplitude of 30 mV. Sodium nitroprusside (3 microM) induced membrane hyperpolarization and abolished unitary potentials generated by CSMC. In contrast, the amplitude of STDs generated by ICC-IM was increased with membrane hyperpolarization. Hyperpolarization induced by pinacidil (10 microM) also increased the amplitude of STDs and enhanced dV/dt(max). These observations indicate that STDs generated in ICC-IM spread passively to the adjacent CSMC to evoke the discharge of unitary potentials in the gastric fundus.


Assuntos
Comunicação Celular , Fundo Gástrico/fisiologia , Células Intersticiais de Cajal/fisiologia , Animais , Comunicação Celular/efeitos dos fármacos , Feminino , Corantes Fluorescentes/metabolismo , Fundo Gástrico/citologia , Fundo Gástrico/efeitos dos fármacos , Fundo Gástrico/metabolismo , Imuno-Histoquímica , Técnicas In Vitro , Células Intersticiais de Cajal/efeitos dos fármacos , Células Intersticiais de Cajal/metabolismo , Canais KATP/agonistas , Canais KATP/metabolismo , Masculino , Potenciais da Membrana , Microeletrodos , Miócitos de Músculo Liso/fisiologia , Doadores de Óxido Nítrico/farmacologia , Nitroprussiato/farmacologia , Perfusão , Pinacidil/farmacologia , Propídio/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Ratos , Ratos Wistar , Fatores de Tempo
18.
Am J Physiol Gastrointest Liver Physiol ; 296(6): G1370-81, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19372102

RESUMO

Populations of interstitial cells of Cajal (ICC) are altered in several gastrointestinal neuromuscular disorders. ICC are identified typically by ultrastructure and expression of Kit (CD117), a protein that is also expressed on mast cells. No other molecular marker currently exists to independently identify ICC. The expression of ANO1 (DOG1, TMEM16A), a Ca(2+)-activated Cl(-) channel, in gastrointestinal stromal tumors suggests it may be useful as an ICC marker. The aims of this study were therefore to determine the distribution of Ano1 immunoreactivity compared with Kit and to establish whether Ano1 is a reliable marker for human and mouse ICC. Expression of Ano1 in human and mouse stomach, small intestine, and colon was investigated by immunofluorescence labeling using antibodies to Ano1 alone and in combination with antibodies to Kit. Colocalization of immunoreactivity was demonstrated by epifluorescence and confocal microscopy. In the muscularis propria, Ano1 immunoreactivity was restricted to cells with the morphology and distribution of ICC. All Ano1-positive cells in the muscularis propria were also Kit positive. Kit-expressing mast cells were not Ano1 positive. Some non-ICC in the mucosa and submucosa of human tissues were Ano1 positive but Kit negative. A few (3.2%) Ano1-positive cells in the human gastric muscularis propria were labeled weakly for Kit. Ano1 labels all classes of ICC and represents a highly specific marker for studying the distribution of ICC in mouse and human tissues with an advantage over Kit since it does not label mast cells.


Assuntos
Canais de Cloreto/análise , Trato Gastrointestinal/química , Trato Gastrointestinal/citologia , Proteínas de Membrana/análise , Proteínas de Neoplasias/análise , Adulto , Animais , Anoctamina-1 , Antígenos de Superfície/análise , Colo/química , Colo/citologia , Feminino , Fundo Gástrico/química , Fundo Gástrico/citologia , Mucosa Gástrica/química , Mucosa Gástrica/citologia , Humanos , Mucosa Intestinal/química , Mucosa Intestinal/citologia , Intestino Delgado/química , Intestino Delgado/citologia , Jejuno/química , Jejuno/citologia , Masculino , Mastócitos/química , Mastócitos/citologia , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , Proteínas Proto-Oncogênicas c-kit/análise , Antro Pilórico/química , Antro Pilórico/citologia , Estômago/química , Estômago/citologia , Triptases/análise
19.
Anat Rec (Hoboken) ; 292(1): 79-86, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18951509

RESUMO

The source of carbon dioxide for the chemical reaction leading to the production of gastric acid is unknown. The decarboxylation of an amino acid releases carbon dioxide. Pepsinogens provide a rich source of the amino acid arginine. Both the source of carbon dioxide, arginine, and the consequence of arginine decarboxylation, agmatine, have been studied. The site of carbon dioxide production has been related to the survival of the parietal cell. An immunohistochemical study has been carried out on glycol methacrylate embedded gastric biopsies from the normal stomach of 38 adult patients. The sections have been stained using polyclonal antibody to pepsinogen II, polyclonal antibody to agmatine, and polyclonal antibody to Helicobacter pylori. Pepsinogen II and agmatine are found in the parietal cell canaliculi. This is consistent with the production of carbon dioxide from arginine in the parietal cell canaliculi. Evidence is presented for the decarboxylation of arginine derived from the activation segment of pepsinogen as the source of carbon dioxide for the production of gastric acid. The production of carbon dioxide by the decarboxylation of arginine in the parietal cell canaliculus enables the extracellular hydration of carbon dioxide at the known site of carbonic anhydrase activity. The extracellular production of acid in the canaliculus together with the presence of agmatine helps to explain why the parietal cells are not destroyed during the formation of gastric acid. Agmatine is found in the mucus secreting cells of the stomach and its role in acid protection of the stomach is discussed. Anat Rec, 2009. (c) 2008 Wiley-Liss, Inc.


Assuntos
Dióxido de Carbono/metabolismo , Ácido Gástrico/metabolismo , Adulto , Agmatina/química , Agmatina/metabolismo , Arginina/química , Arginina/metabolismo , Dióxido de Carbono/química , Ácido Gástrico/química , Fundo Gástrico/citologia , Fundo Gástrico/metabolismo , Mucosa Gástrica/química , Mucosa Gástrica/citologia , Mucosa Gástrica/metabolismo , Humanos , Células Parietais Gástricas/citologia , Células Parietais Gástricas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...