Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 600
Filtrar
1.
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
2.
Cells ; 10(6)2021 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-34199843

RESUMO

BACKGROUND: Lipopolysaccharide (LPS) of Helicobacter pylori (Hp) bacteria causes disintegration of gastric tissue cells in vitro. It has been suggested that interleukin (IL)-33 is involved in healing gastric injury. AIM: To elucidate whether Hp LPS affects regeneration of gastric barrier initiated by IL-33. METHODS: Primary gastric epithelial cells or fibroblasts from Caviae porcellus were transfected with siRNA IL-33. Such cells, not exposed or treated with LPS Hp, were sub-cultured in the medium with or without exogenous IL-33. Then cell migration was assessed in conjunction with oxidative stress and apoptosis, activation of extracellular signal-regulated kinase (Erk), production of collagen I and soluble ST2 (IL-33 decoy). RESULTS: Control cells not treated with LPS Hp migrated in the presence of IL-33. The pro-regenerative activity of IL-33 was related to stimulation of cells to collagen I production. Wound healing by cells exposed to LPS Hp was inhibited even in the presence of IL-33. This could be due to increased oxidative stress and apoptosis in conjunction with Erk activation, sST2 elevation and modulation of collagen I production. CONCLUSIONS: The recovery of gastric barrier cells during Hp infection potentially can be affected due to downregulation of pro-regenerative activity of IL-33 by LPS Hp.


Assuntos
Células Epiteliais/metabolismo , Fibroblastos/metabolismo , Mucosa Gástrica/fisiologia , Helicobacter pylori/química , Interleucina-33/metabolismo , Lipopolissacarídeos/farmacologia , Regeneração/efeitos dos fármacos , Animais , Cobaias , Lipopolissacarídeos/química
3.
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
5.
Nat Rev Gastroenterol Hepatol ; 18(6): 393-410, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33514916

RESUMO

The enteric nervous system (ENS) is the largest division of the peripheral nervous system and closely resembles components and functions of the central nervous system. Although the central role of the ENS in congenital enteric neuropathic disorders, including Hirschsprung disease and inflammatory and functional bowel diseases, is well acknowledged, its role in systemic diseases is less understood. Evidence of a disordered ENS has accumulated in neurodegenerative diseases ranging from amyotrophic lateral sclerosis, Alzheimer disease and multiple sclerosis to Parkinson disease as well as neurodevelopmental disorders such as autism. The ENS is a key modulator of gut barrier function and a regulator of enteric homeostasis. A 'leaky gut' represents the gateway for bacterial and toxin translocation that might initiate downstream processes. Data indicate that changes in the gut microbiome acting in concert with the individual genetic background can modify the ENS, central nervous system and the immune system, impair barrier function, and contribute to various disorders such as irritable bowel syndrome, inflammatory bowel disease or neurodegeneration. Here, we summarize the current knowledge on the role of the ENS in gastrointestinal and systemic diseases, highlighting its interaction with various key players involved in shaping the phenotypes. Finally, current flaws and pitfalls related to ENS research in addition to future perspectives are also addressed.


Assuntos
Sistema Nervoso Entérico/fisiopatologia , Neoplasias Colorretais/fisiopatologia , Diabetes Mellitus/fisiopatologia , Dieta , Sistema Nervoso Entérico/embriologia , Acalasia Esofágica/genética , Acalasia Esofágica/fisiopatologia , Mucosa Gástrica/fisiologia , Microbioma Gastrointestinal/fisiologia , Doença de Hirschsprung/genética , Doença de Hirschsprung/fisiopatologia , Humanos , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/fisiopatologia , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/fisiopatologia
6.
Development ; 147(18)2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32878924

RESUMO

The identity of embryonic gastric epithelial progenitors is unknown. We used single-cell RNA-sequencing, genetic lineage tracing and organoid assays to assess whether Axin2- and Lgr5-expressing cells are gastric progenitors in the developing mouse stomach. We show that Axin2+ cells represent a transient population of embryonic epithelial cells in the forestomach. Lgr5+ cells generate both glandular corpus and squamous forestomach organoids ex vivo Only Lgr5+ progenitors give rise to zymogenic cells in culture. Modulating the activity of the WNT, BMP and Notch pathways in vivo and ex vivo, we found that WNTs are essential for the maintenance of Lgr5+ epithelial cells. Notch prevents differentiation of the embryonic epithelial cells along all secretory lineages and hence ensures their maintenance. Whereas WNTs promote differentiation of the embryonic progenitors along the zymogenic cell lineage, BMPs enhance their differentiation along the parietal lineage. In contrast, WNTs and BMPs are required to suppress differentiation of embryonic gastric epithelium along the pit cell lineage. Thus, coordinated action of the WNT, BMP and Notch pathways controls cell fate determination in the embryonic gastric epithelium.


Assuntos
Linhagem da Célula/fisiologia , Células Epiteliais/metabolismo , Mucosa Gástrica/metabolismo , Transdução de Sinais/fisiologia , Células-Tronco/metabolismo , Estômago/fisiologia , Animais , Diferenciação Celular/fisiologia , Células Epiteliais/fisiologia , Feminino , Mucosa Gástrica/fisiologia , Camundongos , Organoides/metabolismo , Organoides/fisiologia , Células-Tronco/fisiologia
7.
Am J Physiol Cell Physiol ; 319(6): C947-C954, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32755448

RESUMO

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


Assuntos
Mucosa Gástrica/patologia , Metaplasia/patologia , Regeneração/fisiologia , Estômago/patologia , Estômago/fisiologia , Ácido Acético/efeitos adversos , Animais , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/fisiologia , Humanos , Estômago/efeitos dos fármacos
8.
Artigo em Inglês | MEDLINE | ID: mdl-32210918

RESUMO

The structurally-related peptides, gastrin and cholecystokinin (CCK), were originally discovered as humoral stimulants of gastric acid secretion and pancreatic enzyme release, respectively. With the aid of methodological advances in biochemistry, immunochemistry, and molecular biology in the past several decades, our concept of gastrin and CCK as simple gastrointestinal hormones has changed considerably. Extensive in vitro and in vivo studies have shown that gastrin and CCK play important roles in several cellular processes including maintenance of gastric mucosa and pancreatic islet integrity, neurogenesis, and neoplastic transformation. Indeed, gastrin and CCK, as well as their receptors, are expressed in a variety of tumor cell lines, animal models, and human samples, and might contribute to certain carcinogenesis. In this review, we will briefly introduce the gastrin and CCK system and highlight the effects of gastrin and CCK in the regulation of cell proliferation and apoptosis in both normal and abnormal conditions. The potential imaging and therapeutic use of these peptides and their derivatives are also summarized.


Assuntos
Fenômenos Fisiológicos Celulares , Colecistocinina/fisiologia , Gastrinas/fisiologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Fenômenos Fisiológicos Celulares/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Colecistocinina/farmacologia , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Mucosa Gástrica/fisiologia , Gastrinas/farmacologia , Humanos , Pâncreas/metabolismo , Pâncreas/patologia , Pâncreas/fisiologia , Transdução de Sinais/efeitos dos fármacos
9.
Nihon Yakurigaku Zasshi ; 153(6): 261-266, 2019.
Artigo em Japonês | MEDLINE | ID: mdl-31178530

RESUMO

Gastric proton pump (H+,K+-ATPase) which is responsible for H+ secretion of gastric acid (HCl) in gastric parietal cells is the major therapeutic target for treatment of acid-related diseases. H+,K+-ATPase consists of two subunits, a catalytic α-subunit (αHK) and a glycosylated ß-subunit (ßHK). N-glycosylation of ßHK is essential for trafficking and stability of αHK in apical membrane of gastric parietal cells. Terminal sialic acid residues on sugar chains have an important role in various cellular functions. Recently, we succeeded in visualizing the sialylation and desialylation dynamics of ßHK using a fluorescence bioimaging nanoprobe consisting of biocompatible polymers conjugated with lectins for detecting sialic acid. In H+,K+-ATPase-expressing cell lines, rat gastric mucosa, and primary culture of rat gastric parietal cells, fluorescence imaging of sialic acid with the nanoprobe showed that sialylation of ßHK is regulated by intragastric pH and that inhibition of gastric acid secretion induces desialylation of ßHK. In biochemical and pharmacological studies, we revealed that enzyme activity of αHK is negatively regulated by desialylation of ßHK. Our studies uncovered a novel negative-feedback mechanism of H+,K+-ATPase in which sialic acids of ßHK positively regulates H+,K+-ATPase activity, and acidic pH decreases the pump activity by cleaving sialic acids of ßHK. In this topic, we introduce the overview of our research using the bioimaging nanoprobe.


Assuntos
Corantes Fluorescentes , Mucosa Gástrica/fisiologia , ATPase Trocadora de Hidrogênio-Potássio/fisiologia , Ácido N-Acetilneuramínico/química , Imagem Óptica , Bombas de Próton/fisiologia , Animais , Ácido Gástrico , Nanopartículas , Ratos
10.
Mucosal Immunol ; 12(3): 784-794, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30846831

RESUMO

Helicobacter suis is the most prevalent non-Helicobacter pylori Helicobacter species in the human stomach and is associated with chronic gastritis, peptic ulcer disease, and gastric mucosa-associated lymphoid tissue (MALT) lymphoma. H. suis colonizes the gastric mucosa of 60-95% of pigs at slaughter age, and is associated with chronic gastritis, decreased weight gain, and ulcers. Here, we show that experimental H. suis infection changes the mucin composition and glycosylation, decreasing the amount of H. suis-binding glycan structures in the pig gastric mucus niche. Similarly, the H. suis-binding ability of mucins from H. pylori-infected humans is lower than that of noninfected individuals. Furthermore, the H. suis growth-inhibiting effect of mucins from both noninfected humans and pigs is replaced by a growth-enhancing effect by mucins from infected individuals/pigs. Thus, Helicobacter spp. infections impair the mucus barrier by decreasing the H. suis-binding ability of the mucins and by decreasing the antiprolific activity that mucins can have on H. suis. Inhibition of these mucus-based defenses creates a more stable and inhabitable niche for H. suis. This is likely of importance for long-term colonization and outcome of infection, and reversing these impairments may have therapeutic benefits.


Assuntos
Mucinas Gástricas/metabolismo , Mucosa Gástrica/fisiologia , Gastrite/metabolismo , Infecções por Helicobacter/metabolismo , Helicobacter heilmannii/fisiologia , Muco/fisiologia , Úlcera/metabolismo , Adulto , Animais , Proliferação de Células , Doença Crônica , Feminino , Mucosa Gástrica/microbiologia , Gastrite/microbiologia , Glicosilação , Infecções por Helicobacter/microbiologia , Humanos , Masculino , Pessoa de Meia-Idade , Ligação Proteica , Suínos , Úlcera/microbiologia
11.
Free Radic Biol Med ; 130: 35-47, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30326282

RESUMO

Oxidative stress contributes to the progression of non-steroidal anti-inflammatory drug (NSAID)-induced gastrointestinal (GI) cell apoptosis. In our previous study, we reported that nuclear factor erythroid 2-related factor 2 (Nrf2) plays a protective role against ketoprofen-induced GI mucosal oxidative injury. Recent reports suggest that Nrf2 could exhibit antioxidative and antiapoptosis responses through up-regulation of DJ-1 (PARK7). In the current study, we proposed that induction of DJ-1 expression by protocatechuic acid (PCA) might provide a potential therapeutic approach for treating oxidative stress-associated GI ulcer diseases. The results indicated that PCA increased mRNA expression of glutathione peroxidase and heme oxygenase-1 through up-regulation of DJ-1 followed by Nrf2 translocation. Furthermore, PCA protected Int-407 cells against ketoprofen-induced oxidative stress by regulating the DJ-1, PI3K, and mTOR pathways. Pretreatment with PCA inhibited mitochondrial ROS generation, up-regulated the mitochondrial membrane potential, and down-regulated pro-apoptotic Bax as well as downstream caspase-8, caspase-9, and caspase-3 activity, and reversed impaired DJ-1 and anti-apoptotic Bcl-2 protein expression in Int-407 cells induced by ketoprofen. Similar to the in vitro results, SD rats treated with PCA before administration of ketoprofen exhibited decreased caspase-3 protein expression as well as oxidative damage, and impairment of the antioxidant system and DJ-1 protein expression in the GI mucosa were reversed. The administration of lansoprazole, a type of proton pump inhibitor (PPI), strongly inhibited ketoprofen-induced GI mucosal injuries via up-regulation of DJ-1, indicating that DJ-1 is essential for the dietary antioxidant- and PPI drug-mediated mechanism of ulcer therapy. These results suggest that DJ-1 could be a novel target for protection against ketoprofen-induced GI ulcers due to its antioxidant and anti-apoptosis characteristics.


Assuntos
Anti-Inflamatórios não Esteroides/efeitos adversos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/prevenção & controle , Células Epiteliais/imunologia , Mucosa Gástrica/fisiologia , Hidroxibenzoatos/metabolismo , Cetoprofeno/efeitos adversos , Proteína Desglicase DJ-1/metabolismo , Animais , Linhagem Celular , Citoproteção , Humanos , Masculino , Estresse Oxidativo , Fosfatidilinositol 3-Quinases/metabolismo , Proteína Desglicase DJ-1/genética , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
12.
Mater Sci Eng C Mater Biol Appl ; 94: 909-919, 2019 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-30423779

RESUMO

High-performance submucosal injection materials (SIMs) contribute to the success of endoscopic therapy for early-stage gastrointestinal neoplasms. This study aimed to identify the most important factor (viscoelastic parameter) that determines SIM performance and the ease of injection. To determine the ideal viscoelastic parameters of SIMs, submucosal elevation heights (SEHs) and the ease of submucosal injection [characterized by injection pressures (IPs)] were evaluated using a newly developed ex vivo model, in which a constant tension was applied to the studied specimen. The strongest positive correlation was observed between the loss modulus determined at an oscillation frequency of 0.1 rad/s and SEH (correlation coefficient > 0.9) and between the loss modulus at 10 rad/s and IP (correlation coefficient > 0.9). SIMs with high loss moduli (0.1 rad/s) also contributed to maintenance of the submucosal elevation. Moreover, the SEHs of pseudoplastic fluid SIMs (whose loss moduli increased slightly with increasing angular frequency) were greater than those of Newtonian fluid SIMs (whose loss modulus increased drastically with increasing angular frequency). In this study, the ideal viscoelastic SIM parameters were clarified. The loss modulus (0.1 rad/s) was the most important viscoelastic factor affecting SIM performance. Additionally, the development of pseudoplastic fluid SIMs may lead to the creation of next-generation SIMs, with a performance superior to that of sodium hyaluronate, which is currently used widely in endoscopic treatments.


Assuntos
Materiais Biocompatíveis/farmacologia , Elasticidade , Mucosa Gástrica/fisiologia , Injeções , Animais , Pressão , Reologia , Sus scrofa , Viscosidade
13.
Int J Mol Sci ; 21(1)2019 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-31892140

RESUMO

Background: Gastric glands grow and cells reach differentiation at weaning in rats. By considering that early weaning (EW) can affect the timing of development, we aimed to compare molecular and cellular markers of differentiation in pups and adults. Methods: Wistar rats were separated into suckling-control (S) and EW groups at 15 days. Stomachs were collected at 15, 18, and 60 days for RNA and protein extraction, and morphology. Results: After EW, the expression of genes involved in differentiation (Atp4b, Bhlha15 and Pgc) augmented (18 days), and Atp4b and Gif were high at 60 days. EW increased the number of zymogenic cells (ZC) in pups and adults and augmented mucous neck cells only at 18 days, whereas parietal and transition cells (TC) were unchanged. Conclusions: EW affected the gastric mucosa mostly in a transient manner as the changes in gene expression and distribution of differentiated cells that were detected in pups were not fully maintained in adults, except for the size of ZC population. We concluded that though most of EW effects were immediate, such nutritional change in the infancy might affect part of gastric digestive functions in a permanent manner, as some markers were kept unbalanced in the adulthood.


Assuntos
Diferenciação Celular/fisiologia , Células Epiteliais/fisiologia , Mucosa Gástrica/fisiologia , Estômago/fisiologia , Animais , Feminino , Expressão Gênica/fisiologia , Masculino , Ratos , Ratos Wistar , Desmame
14.
Exp Mol Med ; 50(12): 1-14, 2018 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-30510283

RESUMO

DNA methylation is a regulatory mechanism in epigenetics that is frequently altered during human carcinogenesis. To detect critical methylation events associated with gastric cancer (GC), we compared three DNA methylomes from gastric mucosa (GM), intestinal metaplasia (IM), and gastric tumor (GT) cells that were microscopically dissected from an intestinal-type early gastric cancer (EGC) using methylated DNA binding domain sequencing (MBD-seq) and reduced representation bisulfite sequencing (RRBS) analysis. In this study, we focused on differentially methylated promoters (DMPs) that could be directly associated with gene expression. We detected 2,761 and 677 DMPs between the GT and GM by MBD-seq and RRBS, respectively, and for a total of 3,035 DMPs. Then, 514 (17%) of all DMPs were detected in the IM genome, which is a precancer of GC, supporting that some DMPs might represent an early event in gastric carcinogenesis. A pathway analysis of all DMPs demonstrated that 59 G protein-coupled receptor (GPCR) genes linked to the hypermethylated DMPs were significantly enriched in a neuroactive ligand-receptor interaction pathway. Furthermore, among the 59 GPCRs, six GI hormone receptor genes (NPY1R, PPYR1, PTGDR, PTGER2, PTGER3, and SSTR2) that play an inhibitory role in the secretion of gastrin or gastric acid were selected and validated as potential biomarkers for the diagnosis or prognosis of GC patients in two cohorts. These data suggest that the loss of function of gastrointestinal (GI) hormone receptors by promoter methylation may lead to gastric carcinogenesis because gastrin and gastric acid have been known to play a role in cell differentiation and carcinogenesis in the GI tract.


Assuntos
Mucosa Gástrica/fisiologia , Regiões Promotoras Genéticas/genética , Receptores dos Hormônios Gastrointestinais/genética , Neoplasias Gástricas/genética , Carcinogênese , Diferenciação Celular , Linhagem Celular Tumoral , Metilação de DNA , Ácido Gástrico/metabolismo , Gastrinas/metabolismo , Humanos , Metaplasia , Neurotransmissores/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores dos Hormônios Gastrointestinais/metabolismo , Transdução de Sinais , Neoplasias Gástricas/diagnóstico , Sulfitos , Sequenciamento Completo do Genoma
15.
J Cell Sci ; 131(16)2018 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-30072444

RESUMO

The role of the actin cytoskeleton in the sequence of physiological epithelial repair in the intact epithelium has yet to be elucidated. Here, we explore the role of actin in gastric repair in vivo and in vitro gastric organoids (gastroids). In response to two-photon-induced cellular damage of either an in vivo gastric or in vitro gastroid epithelium, actin redistribution specifically occurred in the lateral membranes of cells neighboring the damaged cell. This was followed by their migration inward to close the gap at the basal pole of the dead cell, in parallel with exfoliation of the dead cell into the lumen. The repair and focal increase of actin was significantly blocked by treatment with EDTA or the inhibition of actin polymerization. Treatment with inhibitors of myosin light chain kinase, myosin II, trefoil factor 2 signaling or phospholipase C slowed both the initial actin redistribution and the repair. While Rac1 inhibition facilitated repair, inhibition of RhoA/Rho-associated protein kinase inhibited it. Inhibitors of focal adhesion kinase and Cdc42 had negligible effects. Hence, initial actin polymerization occurs in the lateral membrane, and is primarily important to initiate dead cell exfoliation and cell migration to close the gap.


Assuntos
Actinas/metabolismo , Mucosa Gástrica/lesões , Organoides/lesões , Multimerização Proteica/fisiologia , Reepitelização/fisiologia , Estômago/citologia , Animais , Movimento Celular , Células Cultivadas , Células Epiteliais/fisiologia , Feminino , Mucosa Gástrica/metabolismo , Mucosa Gástrica/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Organoides/citologia , Organoides/fisiologia , Polimerização , Regeneração/fisiologia , Estômago/lesões
16.
Pak J Pharm Sci ; 31(3): 769-775, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29716854

RESUMO

Helicobacter pylori (H. pylori) infections are very serious health problem that are further worsened by increasing/developing resistance to the current antibiotics. Therefore, new therapeutic agents are needed for H. pylori eradication. Use of a CD46 derived peptide (P3) as bactericidal agent against H. pylori has shown high activity rate in vivo and this study examines the changes in H. pylori features in response to effect of P3 treatment.AGS cells were infected with H. pylori wild type strain 67:21 and its mice passage strains (P3 treated and untreated strains) and further examined using immunoblotting assay, FACS and Urease activity analysis. Comparatively we found increased level of Urease alpha subunit A (UreA) and alkyl hydroperoxide reductase C (AhpC) proteins for P3 treated strain of H. pylori than its wild type or untreated strain after infection of AGS cells. Conclusion These results suggest that there might be a high rate of adherence to host cells for the P3 treated passage strain than untreated or wild type strain. Our findings also indicate that either adhesins are being changed or H. pylori interaction to the host cells is affected after P3 treatment.


Assuntos
Células Epiteliais/microbiologia , Mucosa Gástrica/microbiologia , Helicobacter pylori/isolamento & purificação , Animais , Proteínas de Bactérias/isolamento & purificação , Proteínas de Bactérias/fisiologia , Proteínas de Transporte/farmacologia , Proteínas de Transporte/uso terapêutico , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/fisiologia , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/fisiologia , Infecções por Helicobacter/tratamento farmacológico , Helicobacter pylori/efeitos dos fármacos , Helicobacter pylori/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Peptídeos/farmacologia , Peptídeos/uso terapêutico
17.
Proc Natl Acad Sci U S A ; 115(6): 1328-1333, 2018 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-29358395

RESUMO

Genetic and epigenetic alterations are both involved in carcinogenesis, and their low-level accumulation in normal tissues constitutes cancer risk. However, their relative importance has never been examined, as measurement of low-level mutations has been difficult. Here, we measured low-level accumulations of genetic and epigenetic alterations in normal tissues with low, intermediate, and high cancer risk and analyzed their relative effects on cancer risk in the esophagus and stomach. Accumulation of genetic alterations, estimated as a frequency of rare base substitution mutations, significantly increased according to cancer risk in esophageal mucosae, but not in gastric mucosae. The mutation patterns reflected the exposure to lifestyle risk factors. In contrast, the accumulation of epigenetic alterations, measured as DNA methylation levels of marker genes, significantly increased according to cancer risk in both tissues. Patients with cancer (high-risk individuals) were precisely discriminated from healthy individuals with exposure to risk factors (intermediate-risk individuals) by a combination of alterations in the esophagus (odds ratio, 18.2; 95% confidence interval, 3.69-89.9) and by only epigenetic alterations in the stomach (odds ratio, 7.67; 95% confidence interval, 2.52-23.3). The relative importance of epigenetic alterations upon genetic alterations was 1.04 in the esophagus and 2.31 in the stomach. The differential impacts among tissues will be critically important for effective cancer prevention and precision cancer risk diagnosis.


Assuntos
Carcinoma de Células Escamosas/genética , Epigênese Genética , Neoplasias Esofágicas/genética , Mucosa Gástrica/fisiologia , Neoplasias Gástricas/genética , Biomarcadores Tumorais/genética , Metilação de DNA , Carcinoma de Células Escamosas do Esôfago , Feminino , Estudo de Associação Genômica Ampla , Infecções por Helicobacter/complicações , Humanos , Masculino , Taxa de Mutação , Mutação Puntual , Fatores de Risco , Fator de Transcrição AP-2/genética
18.
Biomed Pharmacother ; 97: 75-81, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29080461

RESUMO

Helicobacter pylori (H. pylori) infection is the major cause for gastritis, peptic ulcer, and gastric cancer. Elevated oxidative stress, mitochondrial dysfunction and apoptotic death of gastric epithelial cells are typical hallmarks of H. pylori infection. Ascorbic Acid 2-Glucoside (AA2G) is a stable version of Vitamin C, that binds glucose to conventional vitamin C. AA2G has free radical scavenging activities and anti-apoptotic abilities. However, the protective effect of AA2G against H. pylori-infection in gastric epithelial cells is yet unknown. In this study, we investigated the effects of AA2G in human H. pylori-infected gastric epithelial cells. AA2G could remarkably ameliorate H. pylori-induced oxidative stress, including the levels of intracellular reactive oxygen species (ROS) and 4-hydroxynonenal (4-HNE). Importantly, AA2G treatment also improved mitochondrial function by restoring the level of ATP and mitochondrial membrane potential (MMP). Furthermore, AA2G reduced apoptosis induced by H. pylori through modulation of mitochondria-dependent apoptotic pathways. Our findings suggest that AA2G has a protective effect against H. pylori infection in gastric epithelial cells.


Assuntos
Apoptose/efeitos dos fármacos , Ácido Ascórbico/análogos & derivados , Células Epiteliais/efeitos dos fármacos , Mucosa Gástrica/efeitos dos fármacos , Helicobacter pylori/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Apoptose/fisiologia , Ácido Ascórbico/farmacologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Células Epiteliais/fisiologia , Mucosa Gástrica/fisiologia , Helicobacter pylori/fisiologia , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/fisiologia , Mitocôndrias/fisiologia , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo
19.
Yi Chuan ; 39(8): 753-762, 2017 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-28903902

RESUMO

Single-cell analysis of heterogeneity has become the cutting-edge technology for profound understandings of relationships between cell populations. At present, common methods used in single cellular genomic research are mainly microfluidic technologies (Fluidigm) or based on microwells, both requiring a uniform size of cells at the entrance. However, the size of cells in specific tissues can vary from type to type. To address this issue, we need to establish a method to identify genomic features of individual cells of different sizes. In this paper, we developed a robust method in the analysis of single cellular genomic mutations among gastric tissues. Briefly, the single gastric gland was isolated from the whole tissue, and further enzymatically digested into single cells of various sizes by trypsin. These single cells were then spread on the polyethylene naphthalene slides and selected by the laser microdissection method. Whole genome amplification (WGA) and capillary electrophoresis were performed subsequently to detect single cell microsatellite. This method enabled us to detect the existence of microsatellite instability (MSI) of each single cell within the intestinal metaplasia, and to carry out a flexible and fine analysis of single cells with different sizes in tissues and glands. This reliable and practical method is well performed in both low and high-throughput genome analysis when combined with cell labeling methods, thus providing a novel and highly flexible way to study tissue heterogeneity on the single cell scale.


Assuntos
Mucosa Gástrica/metabolismo , Mucosa Gástrica/fisiologia , Variação Genética/genética , Estômago/fisiologia , Genoma/ética , Humanos , Repetições de Microssatélites/genética , Análise de Célula Única/métodos
20.
J Pathol ; 242(4): 463-475, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28497484

RESUMO

The CD44 gene encodes several protein isoforms due to alternative splicing and post translational modifications. Given that CD44 variant isoform 9 (CD44v9) is expressed within Spasmolytic Polypeptide/TFF2-Expressing Metaplasia (SPEM) glands during repair, CD44v9 may be play a funcitonal role during the process of regeneration of the gastric epithelium. Here we hypothesize that CD44v9 marks a regenerative cell lineage responsive to infiltrating macrophages during regeneration of the gastric epithelium. Ulcers were induced in CD44-deficient (CD44KO) and C57BL/6 (BL6) mice by a localized application of acetic acid to the serosal surface of the stomach. Gastric organoids expressing CD44v9 were derived from mouse stomachs and transplanted at the ulcer site of CD44KO mice. Ulcers, CD44v9 expression, proliferation and histology were measured 1, 3, 5 and 7-days post-injury. Human-derived gastric organoids were generated from stomach tissue collected from elderly (>55 years) or young (14-20 years) patients. Organoids were transplanted into the stomachs of NOD scid gamma (NSG) mice at the site of injury. Gastric injury was induced in NRG-SGM3 (NRGS) mice harboring human-derived immune cells (hnNRGS) and the immune profile anlayzed by CyTOF. CD44v9 expression emerged within regenerating glands the ulcer margin in response to injury. While ulcers in BL6 mice healed within 7-days post-injury, CD44KO mice exhibited loss of repair and epithelial regeneration. Ulcer healing was promoted in CD44KO mice by transplanted CD55v9-expressing gastric organoids. NSG mice exhibited loss of CD44v9 expression and gastric repair. Transplantation of human-derived gastric organoids from young, but not aged stomachs promoted repair in NSG mouse stomachs in response to injury. Finally, compared to NRGS mice, huNRGS animals exhibited reduced ulcer sizes, an infiltration of human CD162+ macrophages and an emergence of CD44v9 expression in SPEM. Thus, during repair of the gastic epithelium CD44v9 emerges within a regenerative cell lineage that coincides with macrophage inflitration within the injured mucosa. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Mucosa Gástrica/fisiologia , Receptores de Hialuronatos/genética , Regeneração/fisiologia , Úlcera Gástrica/metabolismo , Adolescente , Fatores Etários , Idoso , Animais , Células Cultivadas , Mucosa Gástrica/patologia , Variação Genética/fisiologia , Humanos , Receptores de Hialuronatos/metabolismo , Receptores de Hialuronatos/fisiologia , Macrófagos/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Pessoa de Meia-Idade , Organoides/citologia , Organoides/transplante , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiologia , Regeneração/genética , Úlcera Gástrica/genética , Úlcera Gástrica/patologia , Cicatrização/fisiologia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA