Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.250
Filtrar
1.
Histochem Cell Biol ; 157(2): 217-238, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34984523

RESUMO

The microanatomical features of the oesophageal gastric tract in tetrapod representatives and their function, especially those related to the mucosal layer, have not yet been fully investigated. The mucosal layer cells and their function in the oesophageal gastric tract differ structurally and functionally in tetrapod representatives based on interspecies difference and the type of food and feeding habits. The present study was, therefore, postulated to compare the mucosal microanatomical structure and histochemical biodistribution of different mucin types in oesophageal gastric tract tissues of four tetrapod species. A representative of each tetrapod class was selected, as follows: the Egyptian toad Bufo regularis, the lizard Trachylepis quinquetaeniata, the domestic pigeon Columba livia domestica and the albino mouse Mus musculus for Amphibia, Reptilia, Aves and Mammalia, respectively. Microanatomically, in lower tetrapods (toad and lizard), the mucosal layer of the oesophagus was composed of simple ciliated columnar epithelium with goblet cells, whereas in higher tetrapods (pigeon and mouse) it was composed of stratified squamous epithelium, with non-keratinised epithelium in the pigeon but keratinised epithelium in the mouse. However, the gastric mucosal layer of the stomach in lower tetrapods consists of simple columnar epithelium and gastric glands. Similarly, the mucosa of the pigeon's proventriculus consists of simple columnar epithelium with proventricular glands opened into the lumen, whereas mouse mucosa consists of simple columnar epithelium which folds and forms gastric glands with gastric pits having a variety of cell types. Histochemically, the neutral mucin profile biodistribution in the oesophagus mucosal layer was variable. It was strongly positive in the toad and lizard, but was weak in the pigeon and completely negative in the mouse. In contrast it was strongly positive in the gastric mucosa of the toad, lizard and pigeon, but was weak in the mouse's gastric mucosa. On the other hand, the signals of carboxylated and sulfated mucins were found to be different. They were strong in the mucosa of the lizard oesophagus. In contrast, the carboxylated mucins in the gastric mucosa were positive in all representatives except the mouse. The sulfated mucins were, however, seen localised in the mucosal layer cells of the lizard and pigeon only. The study revealed that the microanatomical structures and functions as well as mucin distribution profiles in the oesophageal gastric tract are in line with interspecies difference and the type of food and feeding habits. However, this may need further investigations including more tetrapod representatives.


Assuntos
Esôfago/química , Mucosa Gástrica/química , Mucinas/metabolismo , Animais , Bufonidae , Columbidae , Esôfago/citologia , Esôfago/metabolismo , Mucosa Gástrica/citologia , Mucosa Gástrica/metabolismo , Lagartos , Camundongos , Distribuição Tecidual
2.
Gastroenterology ; 162(2): 415-430, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34728185

RESUMO

The mucosa of the body of the stomach (ie, the gastric corpus) uses 2 overlapping, depth-dependent mechanisms to respond to injury. Superficial injury heals via surface cells with histopathologic changes like foveolar hyperplasia. Deeper, usually chronic, injury/inflammation, most frequently induced by the carcinogenic bacteria Helicobacter pylori, elicits glandular histopathologic alterations, initially manifesting as pyloric (also known as pseudopyloric) metaplasia. In this pyloric metaplasia, corpus glands become antrum (pylorus)-like with loss of acid-secreting parietal cells (atrophic gastritis), expansion of foveolar cells, and reprogramming of digestive enzyme-secreting chief cells into deep antral gland-like mucous cells. After acute parietal cell loss, chief cells can reprogram through an orderly stepwise progression (paligenosis) initiated by interleukin-13-secreting innate lymphoid cells (ILC2s). First, massive lysosomal activation helps mitigate reactive oxygen species and remove damaged organelles. Second, mucus and wound-healing proteins (eg, TFF2) and other transcriptional alterations are induced, at which point the reprogrammed chief cells are recognized as mucus-secreting spasmolytic polypeptide-expressing metaplasia cells. In chronic severe injury, glands with pyloric metaplasia can harbor both actively proliferating spasmolytic polypeptide-expressing metaplasia cells and eventually intestine-like cells. Gastric glands with such lineage confusion (mixed incomplete intestinal metaplasia and proliferative spasmolytic polypeptide-expressing metaplasia) may be at particular risk for progression to dysplasia and cancer. A pyloric-like pattern of metaplasia after injury also occurs in other gastrointestinal organs including esophagus, pancreas, and intestines, and the paligenosis program itself seems broadly conserved across tissues and species. Here we discuss aspects of metaplasia in stomach, incorporating data derived from animal models and work on human cells and tissues in correlation with diagnostic and clinical implications.


Assuntos
Plasticidade Celular/fisiologia , Reprogramação Celular/fisiologia , Mucosa Gástrica/fisiologia , Regeneração/fisiologia , Estômago/fisiologia , Animais , Mucosa Gástrica/citologia , Mucosa Gástrica/patologia , Infecções por Helicobacter/fisiopatologia , Humanos , Hiperplasia , Metaplasia , Células Parietais Gástricas/fisiologia , Estômago/citologia , Estômago/patologia
3.
Bioengineered ; 13(1): 71-82, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34898361

RESUMO

The present study aimed to investigate the protective effects and molecular mechanisms of Dendrobium officinale polysaccharides on gastric mucosal injuries. Following one week of continuous intragastric administration, a gastric mucosal injury model was established using intragastric administration of anhydrous ethanol. The area of gastric ulcer was measured, the contents of interleukin- 6 (IL-6), epidermal growth factor receptor (EGFR), and thyroid transcription factor 1 (TFF-1) in serum were detected by enzyme linked immunosorbent assay (ELISA), and the expressions of EGFR, TFF-1, IL-6, Raf-2, MAP kinase kinase 1 (MEK1), MEK2, and ERK1 in the gastric tissue were determined utilizing qPCR, Western blotting and immunohistochemistry. Simultaneously, Dendrobium officinale polysaccharides and anhydrous ethanol were added to the gastric mucosal cells (GES1) cultured in vitro, and the protective effects of Dendrobium officinale polysaccharides on cell viability was detected using Cell Counting Kit (CCK)-8. The addition of Dendrobium officinale polysaccharides markedly improved the gastric epithelial defect, inflammatory cell infiltration, and redness and swelling stemmed from gastric mucosal injuries and greatly reduced the area of gastric ulcer. The inhibition rates of gastric ulcer were 48.12 ± 2.98, 42.95 ± 1.52, and 27.96 ± 2.05% in the high, medium, and low concentration Dendrobium officinale polysaccharide groups, respectively. Dendrobium officinale polysaccharides could increase the expressions of EGFR and TFF-1 and decrease the expressions of IL-6, Raf-2, MEK1, MEK2, and ERK1. Dendrobium officinale polysaccharides could reduce the level of inflammatory factors and protect gastric mucosa by inhibiting the expression of MAPK pathway genes and proteins.


Assuntos
Anti-Inflamatórios/administração & dosagem , Dendrobium/química , Etanol/efeitos adversos , Mucosa Gástrica/citologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Polissacarídeos/administração & dosagem , Úlcera Gástrica/tratamento farmacológico , Animais , Anti-Inflamatórios/farmacologia , Linhagem Celular , Sobrevivência Celular , Modelos Animais de Doenças , Mucosa Gástrica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Genes erbB-1/efeitos dos fármacos , Humanos , Interleucina-6/metabolismo , Masculino , Extratos Vegetais , Polissacarídeos/farmacologia , Ratos , Úlcera Gástrica/induzido quimicamente , Úlcera Gástrica/metabolismo , Úlcera Gástrica/patologia , Fator Trefoil-1/efeitos dos fármacos , Fator Trefoil-1/metabolismo
4.
Nutrients ; 13(12)2021 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-34959833

RESUMO

Helicobacter pylori (H. pylori) infection promotes gastric carcinogenesis by increasing oxidative stress, inflammation, and dysregulation of cell survival and proliferation of gastric epithelial cells. Astaxanthin (ASTX), a bioactive carotenoid, exhibits antioxidant and anticancer effects by modulating aberrant signaling pathways that lead to dysregulation of cell death and proliferation. To elucidate the molecular mechanism of H. pylori-induced gastric carcinogenesis and to examine the inhibitory effect of ASTX on H. pylori-induced gastric epithelial cell gene expression changes, we performed comparative RNA-sequencing (RNA-Seq) analysis for H. pylori-infected gastric epithelial cells treated with or without ASTX. RNA-Seq results reveal that differentially expressed genes (DEGs) in H. pylori-infected cells were mainly associated with the Wnt/ß-catenin signaling pathway, which is related to cell proliferation. ASTX significantly reversed H. pylori-induced transcriptional alterations of the key mediators involved in ß-catenin signaling, notably, porcupine (gene symbol, PORCN), spermine oxidase (SMOX), bone morphogenetic protein (BMP) and activin membrane-bound inhibitor (BAMBI), SMAD family member 4 (SMAD4), transforming growth factor-ß1 (TGFB1), Fos-like 1 (FOSLI), and c-myc (MYC). We suggest that ASTX may be a potential therapeutic agent that can suppress H. pylori-induced proliferation-associated gene expression changes, in part, by counter-regulating the Wnt/ß-catenin signaling pathway.


Assuntos
Anticarcinógenos/farmacologia , Células Epiteliais/efeitos dos fármacos , Infecções por Helicobacter/complicações , Helicobacter pylori , Antioxidantes/farmacologia , Carcinogênese/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Epiteliais/microbiologia , Mucosa Gástrica/citologia , Expressão Gênica/efeitos dos fármacos , Humanos , Análise de Sequência de RNA , Neoplasias Gástricas/microbiologia , Neoplasias Gástricas/prevenção & controle , Via de Sinalização Wnt/efeitos dos fármacos , Xantofilas/farmacologia
5.
Nat Commun ; 12(1): 5709, 2021 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-34588453

RESUMO

The gastric H+,K+-ATPase mediates electroneutral exchange of 1H+/1K+ per ATP hydrolysed across the membrane. Previous structural analysis of the K+-occluded E2-P transition state of H+,K+-ATPase showed a single bound K+ at cation-binding site II, in marked contrast to the two K+ ions occluded at sites I and II of the closely-related Na+,K+-ATPase which mediates electrogenic 3Na+/2K+ translocation across the membrane. The molecular basis of the different K+ stoichiometry between these K+-counter-transporting pumps is elusive. We show a series of crystal structures and a cryo-EM structure of H+,K+-ATPase mutants with changes in the vicinity of site I, based on the structure of the sodium pump. Our step-wise and tailored construction of the mutants finally gave a two-K+ bound H+,K+-ATPase, achieved by five mutations, including amino acids directly coordinating K+ (Lys791Ser, Glu820Asp), indirectly contributing to cation-binding site formation (Tyr340Asn, Glu936Val), and allosterically stabilizing K+-occluded conformation (Tyr799Trp). This quintuple mutant in the K+-occluded E2-P state unambiguously shows two separate densities at the cation-binding site in its 2.6 Å resolution cryo-EM structure. These results offer new insights into how two closely-related cation pumps specify the number of K+ accommodated at their cation-binding site.


Assuntos
Mucosa Gástrica/enzimologia , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Potássio/metabolismo , Sítios de Ligação/genética , Cátions Monovalentes/metabolismo , Membrana Celular/enzimologia , Microscopia Crioeletrônica , Cristalização , Ensaios Enzimáticos , Mucosa Gástrica/citologia , ATPase Trocadora de Hidrogênio-Potássio/genética , ATPase Trocadora de Hidrogênio-Potássio/isolamento & purificação , ATPase Trocadora de Hidrogênio-Potássio/ultraestrutura , Células HEK293 , Humanos , Modelos Moleculares , Mutação , Engenharia de Proteínas , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Especificidade por Substrato/genética
6.
FEBS Lett ; 595(17): 2257-2270, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34278574

RESUMO

HIV preferentially infects α4 ß7+ CD4 T cells, forming latent reservoirs that contribute to HIV persistence during antiretroviral therapy. However, the properties of α4 ß7+ CD4 T cells in blood and mucosal compartments remain understudied. Employing two distinct models of HIV infection, HIV-infected humans and simian-human immunodeficiency virus (SHIV)-infected rhesus macaques, we show that α4 ß7+ CD4 T cells in blood are enriched for genes regulating cell cycle progression and cellular metabolism. Unlike their circulating counterparts, rectal α4 ß7+ CD4 T cells exhibited a core tissue-residency gene expression program. These features were conserved across primate species, indicating that the environment influences memory T-cell transcriptional networks. Our findings provide an important molecular foundation for understanding the role of α4 ß7 in HIV infection.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/virologia , Infecções por HIV/sangue , Integrinas/metabolismo , Adulto , Animais , COVID-19/sangue , COVID-19/virologia , Ciclo Celular , Proliferação de Células , Mucosa Gástrica/citologia , Mucosa Gástrica/virologia , Regulação da Expressão Gênica , Humanos , Imunização , Macaca mulatta , Masculino , Síndrome de Imunodeficiência Adquirida dos Símios/sangue , Síndrome de Imunodeficiência Adquirida dos Símios/virologia
7.
BMC Pharmacol Toxicol ; 22(1): 44, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34266494

RESUMO

BACKGROUND: Helicobacter pylori (H. pylori) is a common pathogen in development of peptic ulcers with pyroptosis. Rabeprazole, a critical component of standard triple therapy, has been widely used as the first-line regimen for H. pylori infectious treatment. The aim of this study to explore the function of Rabeprazole on cell pyroptosis in vitro. METHODS: The clinical sample from patients diagnosed with or without H. pylori-infection were collected to analyze by Immunohistochemistry (IHC). Real-time quantitative PCR (qPCR), western blot (WB) and enzyme linked immunosorbent assay (Elisa) were performed to analyze the effect of Rabeprazole on cell pyroptosis, including LDH, IL-1ß and IL-18. RESULTS: In this study, we showed that Rabeprazole regulated a phenomenon of cell pyroptosis as confirmed by lactate dehydrogenase (LDH) assay. Further results showed that Rabeprazole inhibited cell pyroptosis in gastric epithelial cells by alleviating GSDMD-executed pyroptosis, leading to decrease IL-1ß and IL-18 mature and secretion, which is attributed to NLRP3 inflammasome activation inhibition. Further analysis showed that ASC, NLRP3 and Caspase-1, was significantly repressed in response to Rabeprazole stimulation, resulting in decreasing cleaved-caspase-1 expression. Most important, NLRP3 and GSDMD is significantly increased in gastric tissue of patients with H. pylori infection. CONCLUSION: These findings revealed a critical role of Rabeprazole in cell pyroptosis in patients with H. pylori infection, suggesting that targeting cell pyroptosis is an alternative strategy in improving H. pylori treatment.


Assuntos
Anti-Inflamatórios/uso terapêutico , Antiulcerosos/uso terapêutico , Células Epiteliais/efeitos dos fármacos , Infecções por Helicobacter/tratamento farmacológico , Inibidores da Bomba de Prótons/uso terapêutico , Rabeprazol/uso terapêutico , Adolescente , Anti-Inflamatórios/farmacologia , Antiulcerosos/farmacologia , Linhagem Celular , Criança , Pré-Escolar , Células Epiteliais/metabolismo , Feminino , Mucosa Gástrica/citologia , Mucosa Gástrica/metabolismo , Infecções por Helicobacter/metabolismo , Humanos , Interleucina-18/genética , Interleucina-18/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , L-Lactato Desidrogenase/metabolismo , Masculino , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Proteínas de Ligação a Fosfato/genética , Proteínas de Ligação a Fosfato/metabolismo , Inibidores da Bomba de Prótons/farmacologia , Piroptose/efeitos dos fármacos , Rabeprazol/farmacologia
8.
Chembiochem ; 22(18): 2783-2790, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34169626

RESUMO

Chronic infection with Helicobacter pylori increases risk of gastric diseases including gastric cancer. Despite development of a robust immune response, H. pylori persists in the gastric niche. Progression of gastric inflammation to serious disease outcomes is associated with infection with H. pylori strains which encode the cag Type IV Secretion System (cag T4SS). The cag T4SS is responsible for translocating the oncogenic protein CagA into host cells and inducing pro-inflammatory and carcinogenic signaling cascades. Our previous work demonstrated that nutrient iron modulates the activity of the T4SS and biogenesis of T4SS pili. In response to H. pylori infection, the host produces a variety of antimicrobial molecules, including the iron-binding glycoprotein, lactoferrin. Our work shows that apo-lactoferrin exerts antimicrobial activity against H. pylori under iron-limited conditions, while holo-lactoferrin enhances bacterial growth. Culturing H. pylori in the presence of holo-lactoferrin prior to co-culture with gastric epithelial cells, results in repression of the cag T4SS activity. Concomitantly, a decrease in biogenesis of cag T4SS pili at the host-pathogen interface was observed under these culture conditions by high-resolution electron microscopy analyses. Taken together, these results indicate that acquisition of alternate sources of nutrient iron plays a role in regulating the pro-inflammatory activity of a bacterial secretion system and present novel therapeutic targets for the treatment of H. pylori-related disease.


Assuntos
Helicobacter pylori/efeitos dos fármacos , Lactoferrina/farmacologia , Sistemas de Secreção Tipo IV/metabolismo , Animais , Modelos Animais de Doenças , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Mucosa Gástrica/citologia , Mucosa Gástrica/metabolismo , Gerbillinae , Infecções por Helicobacter/tratamento farmacológico , Infecções por Helicobacter/microbiologia , Helicobacter pylori/metabolismo , Imunidade Inata , Interleucina-8/metabolismo , Ferro/metabolismo , Lactoferrina/química , Lactoferrina/metabolismo , Lactoferrina/uso terapêutico , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/farmacologia , Isoformas de Proteínas/uso terapêutico , Sistemas de Secreção Tipo IV/antagonistas & inibidores
9.
Aging (Albany NY) ; 13(8): 11808-11821, 2021 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-33883305

RESUMO

There has been increasing evidence that microRNAs (miRNAs) are related to glioma progression, and that genetically engineered mesenchymal stem cells (MSCs) can inhibit the growth of gliomas. However, the underlying mechanism of bone marrow-MSCs (BM--MSCs) and miRs in gastric cancer still remains unclear. Patients with gastric cancer treated in Shijiazhuang First Hospital as well as healthy individuals undergoing physical examinations were recruited to measure the expression of exosomal miR-1228. Receiver operating characteristic (ROC) curves were plotted and the patients were followed up. BM--MSCs from healthy subjects were collected and exosomes were extracted. The MSC cells were transfected with lentiviral vectors carrying miR-1228 and MMP-14 over-expression sequences and scramble sequence, followed by exosome extraction. The exosomes were co-cultured with SGC-7901 and MGC-823 cells to detect cell proliferation, invasion, apoptosis and migration. The correlation between miR-1228 and MMP-14 was determined by dual-luciferase reporter assay. miR-1228 was highly expressed in serum exosomes of patients with gastric cancer with a area under ROC curve (AUC) of 0.865. The exosomes derived from BM-MSCs are expected to be efficient nanocarriers. Up-regulation of miR-1228 can down-regulate the expression of MMP-14 and effectively hinders the development and progression of gastric cancer.


Assuntos
Biomarcadores Tumorais/metabolismo , Metaloproteinase 14 da Matriz/genética , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/metabolismo , Neoplasias Gástricas/genética , Idoso , Medula Óssea/patologia , Estudos de Casos e Controles , Linhagem Celular Tumoral , Proliferação de Células/genética , Técnicas de Cocultura , Regulação para Baixo , Exossomos/genética , Exossomos/metabolismo , Feminino , Seguimentos , Mucosa Gástrica/citologia , Mucosa Gástrica/patologia , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Voluntários Saudáveis , Humanos , Masculino , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/patologia , Regulação para Cima
10.
Peptides ; 141: 170551, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33862165

RESUMO

Energy homeostasis is is determined by food intake and energy expenditure, which are partly regulated by the cross-talk between central and peripheral hormonal signals. Phoenixin (PNX) is a recently discovered pleiotropic neuropeptide with isoforms of 14 (PNX-14) and 20 (PNX-20) amino acids. It is a potent reproductive peptide in vertebrates, regulating the hypothalamo-pituitary-gonadal axis (HPG). It has been identified as a regulator of food intake during light phase when injected intracerebroventricularly in rats. In addition, plasma levels of PNX also increased after food intake in rats, suggesting that it might have possible roles in energy homeostasis. We hypothesized that gut is a source and site of action of PNX in mice. Immunoreactivity for PNX and its putative receptor, super-conserved receptor expressed in brain (SREB3; also known as the G-protein coupled receptor 173/GPR 173) was found in the stomach and intestine of male C57/BL6 J mice, and in MGN3-1 (mouse stomach endocrine) cells and STC-1 (mouse enteroendocrine) cells. In MGN3-1 cells, PNX-20 significantly upregulated ghrelin (10 nM) and ghrelin-O-acyl transferase (GOAT) mRNAs (1000 nM) at 6 h. In STC-1 cells, it significantly suppressed CCK (100 nM) at 2 h. No effects were found on other intestinal hormones tested (glucagon like peptide-1, glucose dependent insulinotropic polypeptide, and peptide YY). Together, these results indicate that PNX-20 is produced in the gut, and it could act directly on gut cells to regulate metabolic hormones.


Assuntos
Mucosa Gástrica , Hormônios Peptídicos , Animais , Masculino , Camundongos , Aciltransferases/genética , Colecistocinina/genética , Células Endócrinas/fisiologia , Mucosa Gástrica/citologia , Mucosa Gástrica/fisiologia , Regulação da Expressão Gênica , Grelina/genética , Proteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Hormônios Peptídicos/genética , Hormônios Peptídicos/metabolismo
11.
Methods Mol Biol ; 2283: 153-173, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33765317

RESUMO

Helicobacter pylori chronically infects the gastric mucosa of humans and diseases associated with infection include gastritis, peptic ulceration, and development of gastric cancer. The organism displays a distinct tropism for the gastric mucosa of humans and for the gastric mucin MUC5AC. While the majority of organisms are found in the mucus layer overlying the epithelial cells in the stomach, adherence of the organism to the gastric epithelium is necessary for the development of disease. The interaction of H. pylori with epithelial cells results in subversion of host cell signaling and induction of an inflammatory response. Factors that influence the outcome of infection include host genetics, environmental factors, and the phenotype of the infecting strain. In this chapter, we describe cell culture assays to assess the interaction of H. pylori with epithelial cells, immunofluorescent staining to detect H. pylori in infected human gastric biopsy specimens and the use of flow cytometry to detect mucin binding to H. pylori.


Assuntos
Técnicas de Cultura de Células/métodos , Mucosa Gástrica/citologia , Helicobacter pylori/patogenicidade , Mucina-5AC/metabolismo , Aderência Bacteriana , Células Cultivadas , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiologia , Humanos
12.
Eur J Pharmacol ; 900: 174020, 2021 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-33741381

RESUMO

Gastric cancer is one of the most common and deadly cancers among men and women and is the third leading cause of cancer mortality worldwide. Thus, discovering and developing novel therapeutics for gastric cancer has become a global priority. In this study, we synthesized two novel anthraquinone-based aspirin derivatives, Asp-X3 and Asp-X3-CH3, with therapeutic potential for gastric cancer. The structures of the two compounds were determined by 1D, 2D-NMR, and High-Resolution Mass (HRSM). Asp-X3 and Asp-X3-CH3 could inhibit the growth of gastric cancer cells (SGC7901), yielding IC50 values 10-fold lower than that of Aspirin. Asp-X3 and Asp-X3-CH3 were less toxic to gastric mucosal cells, yielding IC50 values that were about 2-fold higher than the corresponding IC50 values determined with SGC7901 cells. Asp-X3-CH3 and Asp-X3 also induced SGC7901 cells to undergo apoptosis, yielding apoptotic rates that were about twice the rate induced by Aspirin. Asp-X3-CH3 did not cause significant loss of COX-1 expression in gastric mucosal cells, whereas Asp-X3 and Aspirin both caused significant loss of COX-1 expression as demonstrated by Western blot, consistent with their effects on the content of PGE2 in these cells as determined by ELISA assay. However, both Asp-X3-CH3 and Asp-X3 exerted a similar effect on the level of COX-2 in gastric cancer cells, causing as much as 90% and 95% reduction in COX-2 expression, respectively. Taken together, the results suggested that Asp-X3-CH3 and Asp-X3 were potentially better agents than Aspirin for the inhibition of gastric cancer cell growth, but Asp-X3-CH3 was more effective.


Assuntos
Antraquinonas/síntese química , Antraquinonas/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Aspirina/análogos & derivados , Aspirina/farmacologia , Neoplasias Gástricas/prevenção & controle , Apoptose/efeitos dos fármacos , Aspirina/síntese química , Linhagem Celular Tumoral , Ciclo-Oxigenase 1/metabolismo , Inibidores de Ciclo-Oxigenase 2/farmacologia , Dinoprostona/metabolismo , Mucosa Gástrica/citologia , Mucosa Gástrica/efeitos dos fármacos , Humanos , Espectroscopia de Ressonância Magnética , Neoplasias Gástricas/induzido quimicamente , Relação Estrutura-Atividade
13.
mBio ; 12(1)2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33563833

RESUMO

The human gastrointestinal mucosal surface consists of a eukaryotic epithelium, a prokaryotic microbiota, and a carbohydrate-rich interface that separates them. In the gastrointestinal tract, the interaction of bacteriophages (phages) and their prokaryotic hosts influences the health of the mammalian host, especially colonization with invasive pathobionts. Antibiotics may be used, but they also kill protective commensals. Here, we report a novel phage whose lytic cycle is enhanced in intestinal environments. The tail fiber gene, whose protein product binds human heparan sulfated proteoglycans and localizes the phage to the epithelial cell surface, positions it near its bacterial host, a type of locational targeting mechanism. This finding offers the prospect of developing mucosal targeting phage to selectively remove invasive pathobiont species from mucosal surfaces.IMPORTANCE Invasive pathobionts or microbes capable of causing disease can reside deep within the mucosal epithelium of our gastrointestinal tract. Targeted effective antibacterial therapies are needed to combat these disease-causing organisms, many of which may be multidrug resistant. Here, we isolated a lytic bacteriophage (phage) that can localize to the epithelial surface by binding heparan sulfated glycans, positioning it near its host, Escherichia coli This targeted therapy can be used to selectively remove invasive pathobionts from the gastrointestinal tract, preventing the development of disease.


Assuntos
Bacteriófagos/metabolismo , Mucosa Gástrica/citologia , Trato Gastrointestinal/virologia , Proteoglicanas de Heparan Sulfato/metabolismo , Interações Microbianas , Polissacarídeos/metabolismo , Proteínas da Cauda Viral/metabolismo , Animais , Bacteriófagos/genética , Bacteriófagos/isolamento & purificação , Bacteriófagos/patogenicidade , Técnicas de Cultura de Células , Escherichia coli/metabolismo , Feminino , Mucosa Gástrica/virologia , Trato Gastrointestinal/fisiologia , Humanos , Masculino , Camundongos Endogâmicos BALB C , Microbiota , Organoides/citologia , Organoides/virologia , Organismos Livres de Patógenos Específicos , Simbiose , Proteínas da Cauda Viral/genética
14.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-33479180

RESUMO

An ability to safely harness the powerful regenerative potential of adult stem cells for clinical applications is critically dependent on a comprehensive understanding of the underlying mechanisms regulating their activity. Epithelial organoid cultures accurately recapitulate many features of in vivo stem cell-driven epithelial renewal, providing an excellent ex vivo platform for interrogation of key regulatory mechanisms. Here, we employed a genome-scale clustered, regularly interspaced, short palindromic repeats (CRISPR) knockout (KO) screening assay using mouse gastric epithelial organoids to identify modulators of Wnt-driven stem cell-dependent epithelial renewal in the gastric mucosa. In addition to known Wnt pathway regulators, such as Apc, we found that KO of Alk, Bclaf3, or Prkra supports the Wnt independent self-renewal of gastric epithelial cells ex vivo. In adult mice, expression of these factors is predominantly restricted to non-Lgr5-expressing stem cell zones above the gland base, implicating a critical role for these factors in suppressing self-renewal or promoting differentiation of gastric epithelia. Notably, we found that Alk inhibits Wnt signaling by phosphorylating the tyrosine of Gsk3ß, while Bclaf3 and Prkra suppress regenerating islet-derived (Reg) genes by regulating the expression of epithelial interleukins. Therefore, Alk, Bclaf3, and Prkra may suppress stemness/proliferation and function as novel regulators of gastric epithelial differentiation.


Assuntos
Células-Tronco Adultas/metabolismo , Quinase do Linfoma Anaplásico/genética , Células Epiteliais/metabolismo , Edição de Genes/métodos , Organoides/metabolismo , Proteínas de Ligação a RNA/genética , Via de Sinalização Wnt/genética , Proteína da Polipose Adenomatosa do Colo/genética , Proteína da Polipose Adenomatosa do Colo/metabolismo , Células-Tronco Adultas/citologia , Quinase do Linfoma Anaplásico/metabolismo , Animais , Sistemas CRISPR-Cas , Proliferação de Células , Células Epiteliais/citologia , Mucosa Gástrica/citologia , Mucosa Gástrica/metabolismo , Regulação da Expressão Gênica , Biblioteca Gênica , Glicogênio Sintase Quinase 3 beta/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Células HEK293 , Humanos , Interleucinas/genética , Interleucinas/metabolismo , Camundongos , Organoides/citologia , Proteínas de Ligação a RNA/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Estômago/citologia
15.
Exp Anim ; 70(1): 63-72, 2021 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-32981898

RESUMO

Aromatase, an estrogen synthase, exists in the gastric parietal cells of Wistar rats. The stomach synthesizes large amounts of estrogens and secretes them into the portal vein. We have been particularly studying gastric estrogen synthesis using Wistar rats. However, estrogen synthesis in the stomach of Sprague-Dawley (SD) rats, which are used as frequently as those of the Wistar strain, has not been clarified. We examined steroid synthesis in the stomach of SD rats using immunohistochemistry, in situ hybridization, Western blotting, real-time PCR, and LC-MS/MS. Aromatase also exists in the stomach of SD rats. Its distribution was not found to be different from that of Wistar rats. Results show that H+/K+-ATPase ß-subunit and aromatase colocalized in double immunofluorescence staining. Each steroid synthase downstream from progesterone was present in the gastric mucosa. These results suggest that steroid hormones are synthesized in the parietal cells in the same pathway as Wistar rats. Although mRNA expression of steroid synthases were higher in SD, no significant difference was found in the amount of protein and each steroid hormone level in the portal vein. Although differences between strains might exist in steroid hormone synthesis, results show that SD rats are as useful as Wistar rats for gastric estrogen synthesis experimentation.


Assuntos
Estrogênios/biossíntese , Células Parietais Gástricas/metabolismo , Ratos Sprague-Dawley , Animais , Aromatase/genética , Aromatase/metabolismo , Aromatase/fisiologia , Estrogênios/metabolismo , Mucosa Gástrica/citologia , Mucosa Gástrica/enzimologia , Mucosa Gástrica/metabolismo , Expressão Gênica , Masculino , Células Parietais Gástricas/enzimologia , Veia Porta/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Wistar
16.
Neurogastroenterol Motil ; 33(3): e13993, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33020982

RESUMO

BACKGROUND AND AIM: Muscularis macrophages (MMs) not only mediate the innate immunity, but also functionally interact with cells important for gastrointestinal motility. The aim of this study was to determine the spatial relationship and types of contacts between the MMs and neighboring cells in the muscularis propria of human and mouse stomach, small intestine, and large intestine. METHODS: The distribution and morphology of MMs and their contacts with other cells were investigated by immunohistochemistry and transmission electron microscopy. KEY RESULTS: Immunohistochemistry showed variable shape and number of MMs according to their location in different portions of the muscle coat. By double labeling, a close association between MMs and neighboring cells, that is, neurons, smooth muscle cells, interstitial cells of Cajal (ICCs), telocytes (TCs)/PDGFRα-positive cells, was seen. Electron microscopy demonstrated that in the muscle layers of both animal species, MMs have similar ultrastructural features and have specialized cell-to-cell contacts with smooth muscle cells and TCs/PDGFRα-positive cells but not with ICCs and enteric neurons. CONCLUSION & INFERENCES: This study describes varying patterns of distribution of MMs between different regions of the gut, and reports the presence of distinct and extended cell-to-cell contacts between MMs and smooth muscle cells and between MMs and TCs/PDGFRα-positive cells. In contrast, MMs, although close to ICCs and nerve elements, did not make contact with them. These findings indicate specialized and variable roles for MMs in the modulation of gastrointestinal motility whose significance should be more closely investigated in normal and pathological conditions.


Assuntos
Mucosa Gástrica/citologia , Junções Intercelulares/ultraestrutura , Mucosa Intestinal/citologia , Macrófagos/citologia , Miócitos de Músculo Liso/citologia , Telócitos/citologia , Animais , Comunicação Celular , Sistema Nervoso Entérico , Feminino , Mucosa Gástrica/metabolismo , Mucosa Gástrica/ultraestrutura , Humanos , Células Intersticiais de Cajal/citologia , Células Intersticiais de Cajal/metabolismo , Células Intersticiais de Cajal/ultraestrutura , Mucosa Intestinal/metabolismo , Mucosa Intestinal/ultraestrutura , Macrófagos/metabolismo , Macrófagos/ultraestrutura , Masculino , Camundongos , Microscopia Eletrônica de Transmissão , Pessoa de Meia-Idade , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/ultraestrutura , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Telócitos/metabolismo , Telócitos/ultraestrutura
17.
Gastroenterology ; 160(4): 1256-1268.e9, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33189701

RESUMO

BACKGROUND & AIMS: Inflammation in the gastrointestinal tract may lead to the development of cancer. Dicarbonyl electrophiles, such as isolevuglandins (isoLGs), are generated from lipid peroxidation during the inflammatory response and form covalent adducts with amine-containing macromolecules. Thus, we sought to determine the role of dicarbonyl electrophiles in inflammation-associated carcinogenesis. METHODS: The formation of isoLG adducts was analyzed in the gastric tissues of patients infected with Helicobacter pylori from gastritis to precancerous intestinal metaplasia, in human gastric organoids, and in patients with colitis and colitis-associated carcinoma (CAC). The effect on cancer development of a potent scavenger of dicarbonyl electrophiles, 5-ethyl-2-hydroxybenzylamine (EtHOBA), was determined in transgenic FVB/N insulin-gastrin (INS-GAS) mice and Mongolian gerbils as models of H pylori-induced carcinogenesis and in C57BL/6 mice treated with azoxymethane-dextran sulfate sodium as a model of CAC. The effect of EtHOBA on mutations in gastric epithelial cells of H pylori-infected INS-GAS mice was assessed by whole-exome sequencing. RESULTS: We show increased isoLG adducts in gastric epithelial cell nuclei in patients with gastritis and intestinal metaplasia and in human gastric organoids infected with H pylori. EtHOBA inhibited gastric carcinoma in infected INS-GAS mice and gerbils and attenuated isoLG adducts, DNA damage, and somatic mutation frequency. Additionally, isoLG adducts were elevated in tissues from patients with colitis, colitis-associated dysplasia, and CAC as well as in dysplastic tumors of C57BL/6 mice treated with azoxymethane-dextran sulfate sodium. In this model, EtHOBA significantly reduced adduct formation, tumorigenesis, and dysplasia severity. CONCLUSIONS: Dicarbonyl electrophiles represent a link between inflammation and somatic genomic alterations and are thus key targets for cancer chemoprevention.


Assuntos
Transformação Celular Neoplásica/imunologia , Neoplasias Associadas a Colite/imunologia , Lipídeos/imunologia , Lesões Pré-Cancerosas/imunologia , Neoplasias Gástricas/imunologia , Animais , Benzilaminas/farmacologia , Benzilaminas/uso terapêutico , Núcleo Celular/metabolismo , Transformação Celular Neoplásica/efeitos dos fármacos , Neoplasias Associadas a Colite/microbiologia , Neoplasias Associadas a Colite/patologia , Neoplasias Associadas a Colite/prevenção & controle , Modelos Animais de Doenças , Células Epiteliais , Mucosa Gástrica/citologia , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/imunologia , Mucosa Gástrica/patologia , Gastrite/imunologia , Gastrite/microbiologia , Gastrite/patologia , Gerbillinae , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/patologia , Helicobacter pylori/imunologia , Helicobacter pylori/isolamento & purificação , Humanos , Lipídeos/antagonistas & inibidores , Metaplasia/imunologia , Metaplasia/microbiologia , Metaplasia/patologia , Camundongos , Camundongos Transgênicos , Organoides , Lesões Pré-Cancerosas/tratamento farmacológico , Lesões Pré-Cancerosas/microbiologia , Lesões Pré-Cancerosas/patologia , Neoplasias Gástricas/microbiologia , Neoplasias Gástricas/patologia , Neoplasias Gástricas/prevenção & controle
18.
Gut ; 70(10): 1833-1846, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33293280

RESUMO

OBJECTIVE: Tissue stem cells are central regulators of organ homoeostasis. We looked for a protein that is exclusively expressed and functionally involved in stem cell activity in rapidly proliferating isthmus stem cells in the stomach corpus. DESIGN: We uncovered the specific expression of Iqgap3 in proliferating isthmus stem cells through immunofluorescence and in situ hybridisation. We performed lineage tracing and transcriptomic analysis of Iqgap3 +isthmus stem cells with the Iqgap3-2A-tdTomato mouse model. Depletion of Iqgap3 revealed its functional importance in maintenance and proliferation of stem cells. We further studied Iqgap3 expression and the associated gene expression changes during tissue repair after tamoxifen-induced damage. Immunohistochemistry revealed elevated expression of Iqgap3 in proliferating regions of gastric tumours from patient samples. RESULTS: Iqgap3 is a highly specific marker of proliferating isthmus stem cells during homoeostasis. Iqgap3+isthmus stem cells give rise to major cell types of the corpus unit. Iqgap3 expression is essential for the maintenance of stem potential. The Ras pathway is a critical partner of Iqgap3 in promoting strong proliferation in isthmus stem cells. The robust induction of Iqgap3 expression following tissue damage indicates an active role for Iqgap3 in tissue regeneration. CONCLUSION: IQGAP3 is a major regulator of stomach epithelial tissue homoeostasis and repair. The upregulation of IQGAP3 in gastric cancer suggests that IQGAP3 plays an important role in cancer cell proliferation.


Assuntos
Proteínas Ativadoras de GTPase/metabolismo , Mucosa Gástrica/citologia , Homeostase/fisiologia , Células-Tronco/citologia , Neoplasias Gástricas/metabolismo , Animais , Biomarcadores Tumorais/metabolismo , Proliferação de Células/fisiologia , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Neoplasias Gástricas/tratamento farmacológico , Tamoxifeno/toxicidade
19.
Carcinogenesis ; 42(2): 180-189, 2021 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-33284950

RESUMO

Cancer-associated fibroblasts (CAFs) tend to have tumor-promoting capacity, and can provide therapeutic targets. Even without cancer cells, CAF phenotypes are stably maintained, and DNA methylation and H3K27me3 changes have been shown to be involved. Here, we searched for a potential therapeutic target in primary CAFs from gastric cancer and a mechanism for its dysregulation. Expression microarray using eight CAFs and seven non-CAFs (NCAFs) revealed that serum amyloid A1 (SAA1), which encodes an acute phase secreted protein, was second most upregulated in CAFs, following IGF2. Conditioned medium (CM) derived from SAA1-overexpressing NCAFs was shown to increase migration of gastric cancer cells compared with that from control NCAFs, and its tumor-promoting effect was comparable to that of CM from CAFs. In addition, increased migration of cancer cells by CM from CAFs was mostly canceled with CM from CAFs with SAA1 knockdown. Chromatin immunoprecipitation (ChIP)-quantitative PCR showed that CAFs had higher levels of H3K27ac, an active enhancer mark, in the promoter and the two far upstream regions of SAA1 than NCAFs. Also, BET bromodomain inhibitors, JQ1 and mivebresib, decreased SAA1 expression and tumor-promoting effects in CAFs, suggesting SAA1 upregulation by enhancer activation in CAFs. Our present data showed that SAA1 is a candidate therapeutic target from gastric CAFs and indicated that increased enhancer acetylation is important for its overexpression.


Assuntos
Fibroblastos Associados a Câncer/metabolismo , Proteína Amiloide A Sérica/genética , Neoplasias Gástricas/patologia , Acetilação , Azepinas/farmacologia , Azepinas/uso terapêutico , Fibroblastos Associados a Câncer/efeitos dos fármacos , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Meios de Cultivo Condicionados/metabolismo , Elementos Facilitadores Genéticos , Gastrectomia , Mucosa Gástrica/citologia , Mucosa Gástrica/patologia , Mucosa Gástrica/cirurgia , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Cultura Primária de Células , Piridonas/farmacologia , Piridonas/uso terapêutico , Proteína Amiloide A Sérica/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/cirurgia , Sulfonamidas/farmacologia , Sulfonamidas/uso terapêutico , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo , Triazóis/farmacologia , Triazóis/uso terapêutico , Regulação para Cima
20.
Cell Mol Gastroenterol Hepatol ; 11(3): 783-801, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33069918

RESUMO

BACKGROUND & AIMS: Tight junctions form a barrier to the paracellular passage of luminal antigens. Although most tight junction proteins reside within the apical tight junction complex, claudin-18 localizes mainly to the basolateral membrane where its contribution to paracellular ion transport is undefined. Claudin-18 loss in mice results in gastric neoplasia development and tumorigenesis that may or may not be due to tight junction dysfunction. The aim here was to investigate paracellular permeability defects in stomach mucosa from claudin-18 knockout (Cldn18-KO) mice. METHODS: Stomach tissue from wild-type, heterozygous, or Cldn18-KO mice were stripped of the external muscle layer and mounted in Ussing chambers. Transepithelial resistance, dextran 4 kDa flux, and potential difference (PD) were calculated from the chambered tissues after identifying differences in tissue histopathology that were used to normalize these measurements. Marker expression for claudins and ion transporters were investigated by transcriptomic and immunostaining analysis. RESULTS: No paracellular permeability defects were evident in stomach mucosa from Cldn18-KO mice. RNAseq identified changes in 4 claudins from Cldn18-KO mice, particularly the up-regulation of claudin-2. Although claudin-2 localized to tight junctions in cells at the base of gastric glands, its presence did not contribute overall to mucosal permeability. Stomach tissue from Cldn18-KO mice also had no PD versus a lumen-negative PD in tissues from wild-type mice. This difference resulted from changes in transcellular Cl- permeability with the down-regulation of Cl- loading and Cl- secreting anion transporters. CONCLUSIONS: Our findings suggest that Cldn18-KO has no effect on tight junction permeability in the stomach from adult mice but rather affects anion permeability. The phenotype in these mice may thus be secondary to transcellular anion transporter expression/function in the absence of claudin-18.


Assuntos
Cloretos/metabolismo , Claudinas/deficiência , Células Epiteliais/metabolismo , Mucosa Gástrica/metabolismo , Junções Íntimas/metabolismo , Animais , Permeabilidade da Membrana Celular , Claudinas/genética , Claudinas/metabolismo , Células Epiteliais/citologia , Feminino , Mucosa Gástrica/citologia , Íons/metabolismo , Masculino , Camundongos , Camundongos Knockout , Modelos Animais , RNA-Seq , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...