Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 107
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(23)2023 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-38069171

RESUMEN

Micro-environmental factors, including stromal and immune cells, cytokines, and circulating hormones are well recognized to determine cancer progression. Melanoma cell growth was recently shown to be suppressed by cholecystokinin/gastrin (CCK) receptor antagonists, and our preliminary data suggested that melanoma patients with Helicobacter gastritis (which is associated with elevated serum gastrin) might have an increased risk of cancer progression. Therefore, in the present study, we examined how gastrin may act on melanoma cells. In 89 melanoma patients, we found a statistically significant association between circulating gastrin concentrations and melanoma thickness and metastasis, which are known risk factors of melanoma progression and prognosis. Immunocytochemistry using a validated antibody confirmed weak to moderate CCK2R expression in both primary malignant melanoma cells and the melanoma cell lines SK-MEL-2 and G361. Furthermore, among the 219 tumors in the Skin Cutaneous Melanoma TCGA Pan-Cancer dataset showing gastrin receptor (CCKBR) expression, significantly higher CCKBR mRNA levels were linked to stage III-IV than stage I-II melanomas. In both cell lines, gastrin increased intracellular calcium levels and stimulated cell migration and invasion through mechanisms inhibited by a CCK2 receptor antagonist. Proteomic studies identified increased MMP-2 and reduced TIMP-3 levels in response to gastrin that were likely to contribute to the increased migration of both cell lines. However, the effects of gastrin on tumor cell invasion were relatively weak in the presence of the extracellular matrix. Nevertheless, dermal fibroblasts/myofibroblasts, known also to express CCK2R, increased gastrin-induced cancer cell invasion. Our data suggest that in a subset of melanoma patients, an elevated serum gastrin concentration is a risk factor for melanoma tumor progression, and that gastrin may act on both melanoma and adjacent stromal cells through CCK2 receptors to promote mechanisms of tumor migration and invasion.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Humanos , Melanoma/metabolismo , Gastrinas/farmacología , Gastrinas/metabolismo , Proteómica , Receptores de Colecistoquinina , Receptor de Colecistoquinina B/genética , Receptor de Colecistoquinina B/metabolismo
2.
Mol Cell Proteomics ; 17(9): 1837-1849, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29915148

RESUMEN

Analysis of secretomes critically underpins the capacity to understand the mechanisms determining interactions between cells and between cells and their environment. In the context of cancer cell micro-environments, the relevant interactions are recognized to be an important determinant of tumor progression. Global proteomic analyses of secretomes are often performed at a single time point and frequently identify both classical secreted proteins (possessing an N-terminal signal sequence), as well as many intracellular proteins, the release of which is of uncertain biological significance. Here, we describe a mass spectrometry-based method for stable isotope dynamic labeling of secretomes (SIDLS) that, by dynamic SILAC, discriminates the secretion kinetics of classical secretory proteins and intracellular proteins released from cancer and stromal cells in culture. SIDLS is a robust classifier of the different cellular origins of proteins within the secretome and should be broadly applicable to nonproliferating cells and cells grown in short term culture.


Asunto(s)
Marcaje Isotópico/métodos , Neoplasias/metabolismo , Proteoma/metabolismo , Línea Celular Tumoral , Ontología de Genes , Humanos , Espacio Intracelular/metabolismo , Cinética , Reproducibilidad de los Resultados , Transducción de Señal , Células del Estroma/metabolismo , Factores de Tiempo
3.
Carcinogenesis ; 40(4): 500-512, 2019 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-30624614

RESUMEN

There is increasing evidence that stromal myofibroblasts play a key role in the tumour development however, the mechanisms by which they become reprogrammed to assist in cancer progression remain unclear. As cultured cancer-associated myofibroblasts (CAMs) retain an ability to enhance the proliferation and migration of cancer cells in vitro, it is possible that epigenetic reprogramming of CAMs within the tumour microenvironment may confer long-term pro-tumourigenic changes in gene expression. This study reports the first comparative multi-omics analysis of cancer-related changes in gene expression and DNA methylation in primary myofibroblasts derived from gastric and oesophageal tumours. In addition, we identify novel CAM-specific DNA methylation signatures, which are not observed in patient-matched adjacent tissue-derived myofibroblasts, or corresponding normal tissue-derived myofibroblasts. Analysis of correlated changes in DNA methylation and gene expression shows that different patterns of gene-specific DNA methylation have the potential to confer pro-tumourigenic changes in metabolism, cell signalling and differential responses to hypoxia. These molecular signatures provide new insights into potential mechanisms of stromal reprogramming in gastric and oesophageal cancer, while also providing a new resource to facilitate biomarker identification and future hypothesis-driven studies into mechanisms of stromal reprogramming and tumour progression in solid tumours.


Asunto(s)
Biomarcadores de Tumor/genética , Epigénesis Genética , Neoplasias Esofágicas/patología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Miofibroblastos/patología , Neoplasias Gástricas/patología , Movimiento Celular , Proliferación Celular , Metilación de ADN , Epigenómica , Neoplasias Esofágicas/genética , Humanos , Miofibroblastos/metabolismo , Neoplasias Gástricas/genética , Células Tumorales Cultivadas , Microambiente Tumoral
4.
PLoS Pathog ; 13(11): e1006653, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29095917

RESUMEN

Several conditions associated with reduced gastric acid secretion confer an altered risk of developing a gastric malignancy. Helicobacter pylori-induced atrophic gastritis predisposes to gastric adenocarcinoma, autoimmune atrophic gastritis is a precursor of type I gastric neuroendocrine tumours, whereas proton pump inhibitor (PPI) use does not affect stomach cancer risk. We hypothesised that each of these conditions was associated with specific alterations in the gastric microbiota and that this influenced subsequent tumour risk. 95 patients (in groups representing normal stomach, PPI treated, H. pylori gastritis, H. pylori-induced atrophic gastritis and autoimmune atrophic gastritis) were selected from a cohort of 1400. RNA extracted from gastric corpus biopsies was analysed using 16S rRNA sequencing (MiSeq). Samples from normal stomachs and patients treated with PPIs demonstrated similarly high microbial diversity. Patients with autoimmune atrophic gastritis also exhibited relatively high microbial diversity, but with samples dominated by Streptococcus. H. pylori colonisation was associated with decreased microbial diversity and reduced complexity of co-occurrence networks. H. pylori-induced atrophic gastritis resulted in lower bacterial abundances and diversity, whereas autoimmune atrophic gastritis resulted in greater bacterial abundance and equally high diversity compared to normal stomachs. Pathway analysis suggested that glucose-6-phospahte1-dehydrogenase and D-lactate dehydrogenase were over represented in H. pylori-induced atrophic gastritis versus autoimmune atrophic gastritis, and that both these groups showed increases in fumarate reductase. Autoimmune and H. pylori-induced atrophic gastritis were associated with different gastric microbial profiles. PPI treated patients showed relatively few alterations in the gastric microbiota compared to healthy subjects.


Asunto(s)
Aclorhidria/microbiología , Mucosa Gástrica/microbiología , Microbioma Gastrointestinal , Aclorhidria/inducido químicamente , Aclorhidria/etiología , Aclorhidria/inmunología , Adulto , Anciano , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/microbiología , Análisis por Conglomerados , Estudios de Cohortes , Inglaterra/epidemiología , Femenino , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/inmunología , Gastritis Atrófica/tratamiento farmacológico , Gastritis Atrófica/inmunología , Gastritis Atrófica/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Infecciones por Helicobacter/tratamiento farmacológico , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/efectos de los fármacos , Helicobacter pylori/crecimiento & desarrollo , Helicobacter pylori/inmunología , Helicobacter pylori/aislamiento & purificación , Hospitales Universitarios , Humanos , Masculino , Persona de Mediana Edad , Inhibidores de la Bomba de Protones/efectos adversos , Inhibidores de la Bomba de Protones/uso terapéutico , Riesgo , Neoplasias Gástricas/epidemiología
5.
Analyst ; 143(24): 5912-5917, 2018 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-30191233

RESUMEN

This work reports the first images obtained by combining an infrared aperture scanning near-field optical microscope (SNOM) with a quantum cascade laser (QCL). The future potential of this set-up is demonstrated by a preliminary study on an OE33 human oesophageal adenocarcinoma cell in which the cell is imaged at 1751 cm-1, 1651 cm-1, 1539 cm-1 and 1242 cm-1. In addition to the 1651 cm-1 image, three other images were acquired within the Amide I band (1689 cm-1, 1675 cm-1 and 1626 cm-1) chosen to correspond to secondary structures of proteins. The four images obtained within the Amide I band show distinct differences demonstrating the potential of this approach to reveal subtle changes in the chemical composition of a cell.


Asunto(s)
Adenocarcinoma/diagnóstico por imagen , Células Epiteliales/patología , Láseres de Semiconductores , Microscopía/instrumentación , Microscopía/métodos , Adenocarcinoma/patología , Línea Celular Tumoral , Humanos
6.
Cereb Cortex ; 27(3): 2318-2334, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27073217

RESUMEN

Cholecystokinin-expressing interneurons (CCK-INs) mediate behavior state-dependent inhibition in cortical circuits and themselves receive strong GABAergic input. However, it remains unclear to what extent GABAB receptors (GABABRs) contribute to their inhibitory control. Using immunoelectron microscopy, we found that CCK-INs in the rat hippocampus possessed high levels of dendritic GABABRs and KCTD12 auxiliary proteins, whereas postsynaptic effector Kir3 channels were present at lower levels. Consistently, whole-cell recordings revealed slow GABABR-mediated inhibitory postsynaptic currents (IPSCs) in most CCK-INs. In spite of the higher surface density of GABABRs in CCK-INs than in CA1 principal cells, the amplitudes of IPSCs were comparable, suggesting that the expression of Kir3 channels is the limiting factor for the GABABR currents in these INs. Morphological analysis showed that CCK-INs were diverse, comprising perisomatic-targeting basket cells (BCs), as well as dendrite-targeting (DT) interneurons, including a previously undescribed DT type. GABABR-mediated IPSCs in CCK-INs were large in BCs, but small in DT subtypes. In response to prolonged activation, GABABR-mediated currents displayed strong desensitization, which was absent in KCTD12-deficient mice. This study highlights that GABABRs differentially control CCK-IN subtypes, and the kinetics and desensitization of GABABR-mediated currents are modulated by KCTD12 proteins.


Asunto(s)
Colecistoquinina/metabolismo , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Potenciales Postsinápticos Inhibidores/fisiología , Interneuronas/metabolismo , Canales de Potasio/metabolismo , Receptores de GABA-A/metabolismo , Animales , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/ultraestructura , Dendritas/metabolismo , Dendritas/ultraestructura , Inmunohistoquímica , Interneuronas/ultraestructura , Masculino , Microscopía Inmunoelectrónica , Técnicas de Placa-Clamp , Ratas Wistar , Técnicas de Cultivo de Tejidos
7.
Cereb Cortex ; 27(4): 2453-2468, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-27102657

RESUMEN

Although extensively studied postnatally, the functional differentiation of cholecystokinin (CCK)-containing interneurons en route towards the cerebral cortex during fetal development is incompletely understood. Here, we used CCKBAC/DsRed mice encoding a CCK promoter-driven red fluorescent protein to analyze the temporal dynamics of DsRed expression, neuronal identity, and positioning through high-resolution developmental neuroanatomy. Additionally, we developed a dual reporter mouse line (CCKBAC/DsRed::GAD67gfp/+) to differentiate CCK-containing interneurons from DsRed+ principal cells during prenatal development. We show that DsRed is upregulated in interneurons once they exit their proliferative niche in the ganglionic eminence and remains stably expressed throughout their long-distance migration towards the cerebrum, particularly in the hippocampus. DsRed+ interneurons, including a cohort coexpressing calretinin, accumulated at the palliosubpallial boundary by embryonic day 12.5. Pioneer DsRed+ interneurons already reached deep hippocampal layers by embryonic day 14.5 and were morphologically differentiated by birth. Furthermore, we probed migrating interneurons entering and traversing the cortical plate, as well as stationary cells in the hippocampus by patch-clamp electrophysiology to show the first signs of Na+ and K+ channel activity by embryonic day 12.5 and reliable adult-like excitability by embryonic day 18.5. Cumulatively, this study defines key positional, molecular, and biophysical properties of CCK+ interneurons in the prenatal brain.


Asunto(s)
Diferenciación Celular/fisiología , Corteza Cerebral/citología , Colecistoquinina/metabolismo , Interneuronas/citología , Neurogénesis/fisiología , Animales , Movimiento Celular , Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Inmunohistoquímica , Hibridación in Situ , Interneuronas/metabolismo , Ratones , Ratones Transgénicos , Microscopía Confocal , Técnicas de Placa-Clamp
8.
Br J Clin Pharmacol ; 83(3): 466-475, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27704617

RESUMEN

AIMS: Netazepide, a gastrin/cholecystokinin 2 receptor antagonist, once daily for 12 weeks reduced the number of tumours and size of the largest one in 16 patients with autoimmune chronic atrophic gastritis (CAG), achlorhydria, hypergastrinaemia and multiple gastric neuroendocrine tumours (type 1 gastric NETs), and normalized circulating chromogranin A (CgA) produced by enterochromaffin-like cells, the source of the tumours. The aim was to assess whether longer-term netazepide treatment can eradicate type 1 gastric NETs. METHODS: After a mean 14 months off netazepide, 13 of the 16 patients took it for another 52 weeks. Assessments were: gastroscopy; gene-transcript expression in corpus biopsies using quantitative polymerase chain reaction; blood CgA and gastrin concentrations; and safety assessments. RESULTS: While off-treatment, the number of tumours, the size of the largest one, and CgA all increased again. Netazepide for 52 weeks: cleared all tumours in 5 patients; cleared all but one tumour in one patient; reduced the number of tumours and size of the largest one in the other patients; normalized CgA in all patients; and reduced mRNA abundances of CgA and histidine decarboxylase in biopsies. Gastrin did not increase further, confirming that the patients had achlorhydria. Netazepide was safe and well tolerated. CONCLUSIONS: A gastrin/cholecystokinin 2 receptor antagonist is a potential medical and targeted treatment for type 1 gastric NETs, and an alternative to regular gastroscopy or surgery. Treatment should be continuous because the tumours will regrow if it is stopped. Progress can be monitored by CgA in blood or biomarkers in mucosal biopsies.


Asunto(s)
Enfermedades Autoinmunes/tratamiento farmacológico , Benzodiazepinonas/uso terapéutico , Gastritis Atrófica/tratamiento farmacológico , Tumores Neuroendocrinos/tratamiento farmacológico , Compuestos de Fenilurea/uso terapéutico , Aclorhidria/complicaciones , Aclorhidria/tratamiento farmacológico , Aclorhidria/metabolismo , Anciano , Enfermedades Autoinmunes/sangre , Enfermedades Autoinmunes/complicaciones , Benzodiazepinonas/efectos adversos , Cromogranina A/biosíntesis , Cromogranina A/sangre , Gastrinas/sangre , Gastritis Atrófica/sangre , Gastritis Atrófica/complicaciones , Histidina Descarboxilasa/biosíntesis , Humanos , Persona de Mediana Edad , Tumores Neuroendocrinos/sangre , Tumores Neuroendocrinos/complicaciones , Tumores Neuroendocrinos/metabolismo , Compuestos de Fenilurea/efectos adversos
9.
Br J Cancer ; 114(10): 1152-9, 2016 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-27092781

RESUMEN

BACKGROUND: Stromal cells, including cancer-associated myofibroblasts (CAMs), are recognised to be determinants of cancer progression, but the mechanisms remain uncertain. The chemokine-like protein, chemerin, is upregulated in oesophageal squamous cancer (OSC) CAMs compared with adjacent tissue myofibroblasts (ATMs). In this study, we hypothesised that chemerin stimulates OSC cell invasion. METHODS: Expression of the chemerin receptor, ChemR23, in OSC was examined by immunohistochemistry. The invasion of OSC cells was studied using Boyden chambers and organotypic assays, and the role of chemerin was explored using siRNA, immunoneutralisation and a ChemR23 receptor antagonist. Matrix metalloproteinases (MMPs) were detected by western blot, enzyme assays or immunohistochemistry. RESULTS: Immunohistochemistry indicated expression of the putative chemerin receptor ChemR23 in OSC. It was also expressed in the OSC cell line, OE21. Chemerin stimulated OE21 cell migration and invasion in Boyden chambers. Conditioned medium (CM) from OSC CAMs also stimulated OE21 cell invasion and this was inhibited by chemerin immunoneutralisation, the ChemR23 antagonist CCX832, and by pretreatment of CAMs with chemerin siRNA. In organotypic cultures of OE21 cells on Matrigel seeded with either CAMs or ATMs, there was increased OE21 cell invasion by CAMs that was again inhibited by CCX832. Chemerin increased MMP-1, MMP-2 and MMP-3 abundance, and activity in OE21 cell media, and this was decreased by inhibiting protein kinase C and p44/42 MAPK kinase but not PI-3 kinase. CONCLUSIONS: The data indicate that OSC myofibroblasts release chemerin that stimulates OSC cell invasion. Treatments directed at inhibiting chemerin-ChemR23 interactions might be therapeutically useful in delaying progression in OSC.


Asunto(s)
Fibroblastos Asociados al Cáncer/citología , Carcinoma de Células Escamosas/metabolismo , Quimiocinas/metabolismo , Medios de Cultivo Condicionados/farmacología , Neoplasias Esofágicas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Receptores de Quimiocina/metabolismo , Anciano , Anciano de 80 o más Años , Fibroblastos Asociados al Cáncer/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Carcinoma de Células Escamosas de Esófago , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Metaloproteinasas de la Matriz/metabolismo , Persona de Mediana Edad , Invasividad Neoplásica , Células Tumorales Cultivadas
10.
Am J Physiol Gastrointest Liver Physiol ; 310(9): G696-704, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26939869

RESUMEN

Stromal cells influence epithelial function in both health and disease. Myofibroblasts are abundant stromal cells that influence the cellular microenvironment by release of extracellular matrix (ECM) proteins, growth factors, proteases, cytokines, and chemokines. Cancer-associated myofibroblasts (CAMs) differ from adjacent tissue (ATMs) and normal tissue myofibroblasts (NTMs), but the basis of this is incompletely understood. We report now the differential expression of miRNAs in gastric cancer CAMs. MicroRNA arrays identified differences in the miRNA profile in gastric and esophageal NTMs and in CAMs from stomach compared with NTMs. miR-181d was upregulated in gastric CAMs. Analysis of differentially regulated miRNAs indicated an involvement in Wnt signaling. Examination of a microarray data set then identified Wnt5a as the only consistently upregulated Wnt ligand in gastric CAMs. Wnt5a stimulated miR-181d expression, and knockdown of miR-181d inhibited Wnt5a stimulation of CAM proliferation and migration. Analysis of miR-181d targets suggested a role in chemotaxis. Conditioned medium from CAMs stimulated gastric cancer cell (AGS) migration more than that from ATMs, and miR-181d knockdown reduced the effect of CAM-CM on AGS cell migration but had no effect on AGS cell responses to ATM conditioned media. The data suggest that dysregulation of miRNA expression in gastric CAMs, secondary to Wnt5a signaling, accounts at least in part for the effect of CAMs in promoting cancer cell migration.


Asunto(s)
MicroARNs/genética , Miofibroblastos/metabolismo , Neoplasias Gástricas/metabolismo , Vía de Señalización Wnt , Proliferación Celular , Células Cultivadas , Quimiotaxis , Humanos , Miofibroblastos/fisiología , Neoplasias Gástricas/genética , Proteína Wnt-5a/genética , Proteína Wnt-5a/metabolismo
11.
Biochem Biophys Res Commun ; 475(1): 119-24, 2016 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-27179776

RESUMEN

Connective tissue growth factor (CTGF) has been reported in gastric adenocarcinoma and in carcinoid tumors. The aim of this study was to explore a possible link between CTGF and gastrin in gastric epithelial cells and to study the role of CTGF in gastrin induced migration and invasion of AGS-GR cells. The effects of gastrin were studied using RT-qPCR, Western blot and assays for migration and invasion. We report an association between serum gastrin concentrations and CTGF abundancy in the gastric corpus mucosa of hypergastrinemic subjects and mice. We found a higher expression of CTGF in gastric mucosa tissue adjacent to tumor compared to normal control tissue. We showed that gastrin induced expression of CTGF in gastric epithelial AGS-GR cells via MEK, PKC and PKB/AKT pathways. CTGF inhibited gastrin induced migration and invasion of AGS-GR cells. We conclude that CTGF expression is stimulated by gastrin and involved in remodeling of the gastric epithelium.


Asunto(s)
Adenocarcinoma/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Gastrinas/metabolismo , Neoplasias Gástricas/metabolismo , Estómago/patología , Adenocarcinoma/patología , Animales , Movimiento Celular , Mucosa Gástrica/metabolismo , Humanos , Ratones , Invasividad Neoplásica/patología , Transducción de Señal , Neoplasias Gástricas/patología
12.
Am J Physiol Gastrointest Liver Physiol ; 309(2): G78-86, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25977510

RESUMEN

The pyloric antral hormone gastrin plays a role in remodeling of the gastric epithelium, but the specific targets of gastrin that mediate these effects are poorly understood. Glandular epithelial cells of the gastric corpus express matrix metalloproteinase (MMP)-1, which is a potential determinant of tissue remodeling; some of these cells express the CCK-2 receptor at which gastrin acts. We have now examined the hypothesis that gastrin stimulates expression of MMP-1 in the stomach. We determined MMP-1 transcript abundance in gastric mucosal biopsies from Helicobacter pylori negative human subjects with normal gastric mucosal histology, who had a range of serum gastrin concentrations due in part to treatment with proton pump inhibitors (PPI). The effects of gastrin were studied on gastric epithelial AGS-GR cells using Western blot and migration assays. In human subjects with increased serum gastrin due to PPI usage, MMP-1 transcript abundance was increased 2-fold; there was also increased MMP-7 transcript abundance but not MMP-3. In Western blots, gastrin increased proMMP-1 abundance, as well that of a minor band corresponding to active MMP-1, in the media of AGS-GR cells, and the response was mediated by protein kinase C and p42/44 MAP kinase. There was also increased MMP-1 enzyme activity. Gastrin-stimulated AGS-GR cell migration in both scratch wound and Boyden chamber assays was inhibited by MMP-1 immunoneutralization. We conclude that MMP-1 expression is a target of gastrin implicated in mucosal remodeling.


Asunto(s)
Movimiento Celular , Células Epiteliales/enzimología , Mucosa Gástrica/enzimología , Gastrinas/metabolismo , Metaloproteinasa 1 de la Matriz/metabolismo , Animales , Estudios de Casos y Controles , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Mucosa Gástrica/efectos de los fármacos , Gastrinas/sangre , Gastrinas/genética , Humanos , Metaloproteinasa 1 de la Matriz/genética , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Quinasa C/metabolismo , Inhibidores de la Bomba de Protones/farmacología , ARN Mensajero/metabolismo , Ratas , Transducción de Señal , Transfección , Regulación hacia Arriba
13.
Gastroenterology ; 147(3): 655-666.e9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24859162

RESUMEN

BACKGROUND & AIMS: Loss of expression of Sonic Hedgehog (Shh) from parietal cells results in hypergastrinemia in mice, accompanied by increased expression of Indian Hedgehog (Ihh) and hyperproliferation of surface mucous cells. We investigated whether hypergastrinemia induces gastric epithelial proliferation by activating Ihh signaling in mice. METHODS: We studied mice with parietal cell-specific deletion of Shh (PC-Shh(KO)) and hypergastrinemia, crossed with gastrin-deficient (GKO) mice (PC-Shh(KO)/GKO). When mice were 3-4 months old, gastric tissues were collected and analyzed by histology, for incorporation of bromodeoxyuridine, and for expression of the surface mucous cell marker Ulex europaeus. PC-Shh(KO)/GKO mice were given gastrin infusions for 7 days; gastric surface epithelium was collected and expression of Ihh was quantified by laser capture microdissection followed by quantitative reverse transcriptase polymerase chain reaction. Mouse stomach-derived organoids were incubated with or without inhibitors of WNT (DKK1) or Smoothened (vismodegib) and then cocultured with immortalized stomach mesenchymal cells, to assess proliferative responses to gastrin. RESULTS: Gastric tissues from PC-Shh(KO)/GKO mice with hypergastrinemia had an expanded surface pit epithelium, indicated by a significant increase in numbers of bromodeoxyuridine- and Ulex europaeus-positive cells, but there was no evidence for hyperproliferation. Gastrin infusion of PC PC-Shh(KO)/GKO mice increased expression of Ihh and proliferation within the surface epithelium compared with mice given infusions of saline. In gastric organoids cocultured with immortalized stomach mesenchymal cells, antagonists of WNT and Smoothened inhibited gastrin-induced proliferation and WNT activity. Activity of WNT in media collected from immortalized stomach mesenchymal cells correlated with increased expression of glioma-associated oncogene homolog 1, and was inhibited by DKK1 or vismodegib. CONCLUSIONS: Ihh signaling mediates gastrin-induced proliferation of epithelial cells in stomachs of adult mice.


Asunto(s)
Proliferación Celular , Células Epiteliales/metabolismo , Mucosa Gástrica/metabolismo , Gastrinas/metabolismo , Proteínas Hedgehog/metabolismo , Gastropatías/metabolismo , Animales , Línea Celular , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Células Epiteliales/patología , Mucosa Gástrica/patología , Gastrinas/administración & dosificación , Gastrinas/deficiencia , Gastrinas/genética , Proteínas Hedgehog/deficiencia , Proteínas Hedgehog/genética , Infusiones Parenterales , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Organoides , Receptores Acoplados a Proteínas G/metabolismo , Receptor Smoothened , Gastropatías/genética , Gastropatías/patología , Factores de Tiempo , Vía de Señalización Wnt , Proteína con Dedos de Zinc GLI1
14.
Carcinogenesis ; 35(8): 1798-806, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24710625

RESUMEN

Stromal cells influence cancer progression. Myofibroblasts are an important stromal cell type, which influence the tumour microenvironment by release of extracellular matrix (ECM) proteins, proteases, cytokines and chemokines. The mechanisms of secretion are poorly understood. Here, we describe the secretion of marker proteins in gastric cancer and control myofibroblasts in response to insulin-like growth factor (IGF) stimulation and, using functional genomic approaches, we identify proteins influencing the secretory response. IGF rapidly increased myofibroblast secretion of an ECM protein, TGFßig-h3. The secretory response was not blocked by inhibition of protein synthesis and was partially mediated by increased intracellular calcium (Ca(2+)). The capacity for evoked secretion was associated with the presence of dense-core secretory vesicles and was lost in cells from patients with advanced gastric cancer. In cells responding to IGF-II, the expression of neuroendocrine marker proteins, including secretogranin-II and proenkephalin, was identified by gene array and LC-MS/MS respectively, and verified experimentally. The expression of proenkephalin was decreased in cancers from patients with advanced disease. Inhibition of secretogranin-II expression decreased the secretory response to IGF, and its over-expression recovered the secretory response consistent with a role in secretory vesicle biogenesis. We conclude that normal and some gastric cancer myofibroblasts have a neuroendocrine-like phenotype characterized by Ca(2+)-dependent regulated secretion, dense-core secretory vesicles and expression of neuroendocrine marker proteins; loss of the phenotype is associated with advanced cancer. A failure to regulate myofibroblast protein secretion may contribute to cancer progression.


Asunto(s)
Factor II del Crecimiento Similar a la Insulina/metabolismo , Miofibroblastos/patología , Sistemas Neurosecretores/patología , Secretogranina II/metabolismo , Neoplasias Gástricas/patología , Western Blotting , Estudios de Casos y Controles , Células Cultivadas , Progresión de la Enfermedad , Exocitosis/fisiología , Mucosa Gástrica/metabolismo , Humanos , Técnicas para Inmunoenzimas , Marcaje Isotópico , Miofibroblastos/metabolismo , Sistemas Neurosecretores/metabolismo , Fenotipo , ARN Interferente Pequeño/genética , Secretogranina II/antagonistas & inhibidores , Secretogranina II/genética , Neoplasias Gástricas/metabolismo , Espectrometría de Masas en Tándem
15.
J Physiol ; 592(14): 2951-8, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24665102

RESUMEN

The existence of the hormone gastrin in the distal stomach (antrum) has been known for almost 110 years, and the physiological function of this amidated peptide in regulating gastric acid secretion via the CCK2 receptor is now well established. In this brief review we consider important additional roles of gastrin, including regulation of genes encoding proteins such as plasminogen activator inhibitors and matrix metalloproteinases that have important actions on extracellular matrix remodelling. These actions are, at least in part, effected by paracrine signalling pathways and make important contributions to maintaining functional integrity of the gastric epithelium. Recent studies also provide support for the idea that gastrin, in concert with other hormones, could potentially contribute a post-prandial incretin effect. We also review recent developments in the biology of other gastrin gene products, including the precursor progastrin, which causes proliferation of the colonic epithelium and in certain circumstances may induce cancer formation. Glycine-extended biosynthetic processing intermediates also have proliferative effects in colonic mucosa and in some oesophageal cancer cell lines. Whether these additional gene products exert their effects through the CCK2 receptor or a separate entity is currently a matter of debate.


Asunto(s)
Gastrinas/fisiología , Animales , Mucosa Gástrica/metabolismo , Humanos , Incretinas/metabolismo , Receptor de Colecistoquinina B/metabolismo , Neoplasias Gástricas/metabolismo
16.
Gastroenterology ; 145(1): 197-208.e3, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23523669

RESUMEN

BACKGROUND & AIMS: Many colon cancers produce the hormone progastrin, which signals via autocrine and paracrine pathways to promote tumor growth. Transgenic mice that produce high circulating levels of progastrin (hGAS) have increased proliferation of colonic epithelial cells and are more susceptible to colon carcinogenesis than control mice. We investigated whether progastrin affects signaling between colonic epithelial and myofibroblast compartments to regulate tissue homeostasis and cancer susceptibility. METHODS: Colonic myofibroblast numbers were assessed in hGAS and C57BL/6 mice by immunohistochemistry. Human CCD18Co myofibroblasts were incubated with recombinant human progastrin (rhPG)(1-80) for 18 hours, and proliferation was assessed in the presence of pharmacologic inhibitors. The proliferation of human HT29 colonic epithelial cells was assessed after addition of conditioned media from CCD18Co cells incubated with progastrin. The effects of the insulin-like growth factor (IGF)-I receptor antagonist AG1024 were investigated in cultured HT29 cells and on the colonic epithelium of hGAS mice compared with mice that did not express transgenic progastrin (controls). RESULTS: The colonic mucosa of hGAS mice contained greater numbers of myofibroblasts that expressed α-smooth muscle actin and vimentin than controls. Incubation of CCD18Co myofibroblasts with 0.1 nmol/L rhPG(1-80) increased their proliferation, which required activation of protein kinase C and phosphatidylinositol-3 kinase. CCD18Co cells secreted IGF-II in response to rhPG(1-80), and conditioned media from CCD18Co cells that had been incubated with rhPG(1-80) increased the proliferation of HT29 cells. The colonic epithelial phenotype of hGAS mice (crypt hyperplasia, increased proliferation, and altered proportions of goblet and enteroendocrine cells) was inhibited by AG1024. CONCLUSIONS: Progastrin stimulates colonic myofibroblasts to release IGF-II, which increases proliferation of colonic epithelial cells. Progastrin might therefore alter colonic epithelial cells via indirect mechanisms to promote neoplasia.


Asunto(s)
Colon/citología , Gastrinas/fisiología , Factor II del Crecimiento Similar a la Insulina/metabolismo , Mucosa Intestinal/citología , Miofibroblastos/metabolismo , Precursores de Proteínas/fisiología , Animales , Proliferación Celular , Células Cultivadas , Quinasas Similares a Doblecortina , Células HT29 , Humanos , Péptidos y Proteínas de Señalización Intracelular/análisis , Ratones , Ratones Endogámicos C57BL , Fosfatidilinositol 3-Quinasas/fisiología , Proteína Quinasa C/fisiología , Proteínas Serina-Treonina Quinasas/análisis , Tirfostinos/farmacología
17.
Helicobacter ; 19(5): 387-99, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24827414

RESUMEN

BACKGROUND: Antibiotic resistance in Helicobacter pylori contributes to failure in eradicating the infection and is most often due to point and missense mutations in a few key genes. METHODS: The antibiotic susceptibility profiles of H. pylori isolates from 46 Pakistani patients were determined by Etest. Resistance and pathogenicity genes were amplified, and sequences were analyzed to determine the presence of mutations. RESULTS: A high percentage of isolates (73.9%) were resistant to metronidazole (MTZ), with considerable resistance to clarithromycin (CLR; 47.8%) and amoxicillin (AML; 54.3%) also observed. Relatively few isolates were resistant to tetracycline (TET; 4.3%) or to ciprofloxacin (CIP; 13%). However, most isolates (n = 43) exhibited resistance to one or more antibiotics. MTZ-resistant isolates contained missense mutations in oxygen-independent NADPH nitroreductase (RdxA; 8 mutations found) and NADH flavin oxidoreductase (FrxA; 4 mutations found). In the 23S rRNA gene, responsible for CLR resistance, a new point mutation (A2181G) and 4 previously reported mutations were identified. Pathogenicity genes cagA, dupA, and vacA s1a/m1 were detected frequently in isolates which were also found to be resistant to MTZ, CLR, and AML. A high percentage of CagA and VacA seropositivity was also observed in these patients. Phylogenetic analysis of partial sequences showed uniform distribution of the 3' region of cagA throughout the tree. CONCLUSIONS: We have identified H. pylori isolates in Pakistan which harbor pathogenicity genes and worrying antibiotic resistance profiles as a result of having acquired multiple point and missense mutations. H. pylori eradication regimens should therefore be reevaluated in this setting.


Asunto(s)
Farmacorresistencia Bacteriana , Infecciones por Helicobacter/epidemiología , Helicobacter pylori/aislamiento & purificación , Adulto , Anciano , Anciano de 80 o más Años , Proteínas Bacterianas/genética , Secuencia de Bases , Claritromicina/farmacología , Estudios de Cohortes , Femenino , Infecciones por Helicobacter/microbiología , Helicobacter pylori/efectos de los fármacos , Helicobacter pylori/genética , Helicobacter pylori/patogenicidad , Humanos , Masculino , Metronidazol/farmacología , Pruebas de Sensibilidad Microbiana , Persona de Mediana Edad , Datos de Secuencia Molecular , Mutación , Pakistán/epidemiología , Filogenia , Análisis de Secuencia de ADN , Virulencia , Adulto Joven
18.
J Proteome Res ; 12(7): 3413-22, 2013 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-23705892

RESUMEN

Cancer progression involves changes in extracellular proteolysis, but the contribution of stromal cell secretomes to the cancer degradome remains uncertain. We have now defined the secretome of a specific stromal cell type, the myofibroblast, in gastric cancer and its modification by proteolysis. SILAC labeling and COFRADIC isolation of methionine containing peptides allowed us to quantify differences in gastric cancer-derived myofibroblasts compared with myofibroblasts from adjacent tissue, revealing increased abundance of several proteases in cancer myofibroblasts including matrix metalloproteinases (MMP)-1 and -3. Moreover, N-terminal COFRADIC analysis identified cancer-restricted proteolytic cleavages, including liberation of the active forms of MMP-1, -2, and -3 from their inactive precursors. In vivo imaging confirmed increased MMP activity when gastric cancer cells were xenografted in mice together with gastric cancer myofibroblasts. Western blot and enzyme activity assays confirmed increased MMP-1, -2, and -3 activity in cancer myofibroblasts, and cancer cell migration assays indicated stimulation by MMP-1, -2, and -3 in cancer-associated myofibroblast media. Thus, cancer-derived myofibroblasts differ from their normal counterparts by increased production and activation of MMP-1, -2, and -3, and this may contribute to the remodelling of the cancer cell microenvironment.


Asunto(s)
Metaloproteinasa 1 de la Matriz/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Miofibroblastos/metabolismo , Neoplasias Gástricas/metabolismo , Animales , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Metaloproteinasa 1 de la Matriz/biosíntesis , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 3 de la Matriz/biosíntesis , Ratones , Miofibroblastos/patología , Proteolisis , Neoplasias Gástricas/patología , Microambiente Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Am J Physiol Gastrointest Liver Physiol ; 304(9): G814-22, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23494120

RESUMEN

Gastric mucosal health is maintained in response to potentially damaging luminal factors. Aspirin and nonsteroidal anti-inflammatory drugs (NSAIDs) disrupt protective mechanisms leading to bleeding and ulceration. The plasminogen activator system has been implicated in fibrinolysis following gastric ulceration, and an inhibitor of this system, plasminogen activator inhibitor (PAI)-1, is expressed in gastric epithelial cells. In Helicobacter pylori-negative patients with normal gastric histology taking aspirin or NSAIDs, we found elevated gastric PAI-1 mRNA abundance compared with controls; the increase in patients on aspirin was independent of whether they were also taking proton pump inhibitors. In the same patients, aspirin tended to lower urokinase plasminogen activator mRNA. Immunohistochemistry indicated PAI-1 localization to epithelial cells. In a model system using MKN45 or AGS-GR cells transfected with a PAI-1 promoter-luciferase reporter construct, we found no evidence for upregulation of PAI-1 expression by indomethacin, and, in fact, cyclooxygenase products such as PGE2 and PGI2 weakly stimulated expression. Increased gastric PAI-1 mRNA was also found in mice following gavage with ethanol or indomethacin, but plasma PAI-1 was unaffected. In PAI-1(-/-) mice, gastric hemorrhagic lesions in response to ethanol or indomethacin were increased compared with C57BL/6 mice. In contrast, in PAI-1-H/Kß mice in which PAI-1 is overexpressed in parietal cells, there were decreased lesions in response to ethanol and indomethacin. Thus, PAI-1 expression is increased in gastric epithelial cells in response to mucosal irritants such as aspirin and NSAIDs probably via an indirect mechanism, and PAI-1 acts as a local autoregulator to minimize mucosal damage.


Asunto(s)
Mucosa Gástrica/efectos de los fármacos , Inhibidor 1 de Activador Plasminogénico/fisiología , Animales , Aspirina/farmacología , Dinoprostona , Etanol/toxicidad , Femenino , Humanos , Indometacina/toxicidad , Masculino , Ratones , Inhibidor 1 de Activador Plasminogénico/biosíntesis , ARN Mensajero/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/biosíntesis
20.
Am J Physiol Gastrointest Liver Physiol ; 305(8): G552-63, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23907822

RESUMEN

Gastrointestinal myofibroblasts are contractile, electrically nonexcitable, transitional cells that play a role in extracellular matrix production, in ulcer healing, and in pathophysiological conditions they contribute to chronic inflammation and tumor development. Na+/Ca2+ exchangers (NCX) are known to have a crucial role in Ca2+ homeostasis of contractile cells, however, no information is available concerning the role of NCX in the proliferation and migration of gastrointestinal myofibroblasts. In this study, our aim was to investigate the role of NCX in the Ca2+ homeostasis, migration, and proliferation of human gastrointestinal myofibroblasts, focusing on human gastric myofibroblasts (HGMs). We used microfluorometric measurements to investigate the intracellular Ca2+ and Na+ concentrations, PCR analysis and immunostaining to show the presence of the NCX, patch clamp for measuring NCX activity, and proliferation and migration assays to investigate the functional role of the exchanger. We showed that 53.0±8.1% of the HGMs present Ca2+ oscillations, which depend on extracellular Ca2+ and Na+, and can be inhibited by NCX inhibitors. NCX1, NCX2, and NCX3 were expressed at both mRNA and protein levels in HGMs, and they contribute to the intracellular Ca2+ and Na+ homeostasis as well, regardless of the oscillatory activity. NCX inhibitors significantly blocked the basal and insulin-like growth factor II-stimulated migration and proliferation rates of HGMs. In conclusion, we showed that NCX plays a pivotal role in regulating the Ca2+ homeostasis, migration, and proliferation of HGMs. The inhibition of NCX activity may be a potential therapeutic target in hyperproliferative gastric diseases.


Asunto(s)
Movimiento Celular/fisiología , Proliferación Celular , Miofibroblastos/citología , Miofibroblastos/fisiología , Intercambiador de Sodio-Calcio/metabolismo , Estómago/citología , Calcio/metabolismo , Regulación de la Expresión Génica/fisiología , Humanos , Sodio/metabolismo , Intercambiador de Sodio-Calcio/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA