Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Cells ; 12(6)2023 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-36980185

RESUMEN

The mammalian 20S catalytic core of the proteasome is made of 14 different subunits (α1-7 and ß1-7) but exists as different subtypes depending on the cell type. In immune cells, for instance, constitutive catalytic proteasome subunits can be replaced by the so-called immuno-catalytic subunits, giving rise to the immunoproteasome. Proteasome activity is also altered by post-translational modifications (PTMs) and by genetic variants. Immunochemical methods are commonly used to investigate these PTMs whereby protein-tagging is necessary to monitor their effect on 20S assembly. Here, we present a new miniaturized workflow combining top-down and bottom-up mass spectrometry of immunopurified 20S proteasomes that analyze the proteasome assembly status as well as the full proteoform footprint, revealing PTMs, mutations, single nucleotide polymorphisms (SNPs) and induction of immune-subunits in different biological samples, including organoids, biopsies and B-lymphoblastoid cell lines derived from patients with proteasome-associated autoinflammatory syndromes (PRAAS). We emphasize the benefits of using top-down mass spectrometry in preserving the endogenous conformation of protein modifications, while enabling a rapid turnaround (1 h run) and ensuring high sensitivity (1-2 pmol) and demonstrate its capacity to semi-quantify constitutive and immune proteasome subunits.


Asunto(s)
Complejo de la Endopetidasa Proteasomal , Procesamiento Proteico-Postraduccional , Animales , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Citoplasma/metabolismo , Espectrometría de Masas/métodos , Línea Celular , Mamíferos/metabolismo
2.
Cell Rep ; 36(1): 109318, 2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-34233185

RESUMEN

The immunological synapse is a complex structure that decodes stimulatory signals into adapted lymphocyte responses. It is a unique window to monitor lymphocyte activity because of development of systematic quantitative approaches. Here we demonstrate the applicability of high-content imaging to human T and natural killer (NK) cells and develop a pipeline for unbiased analysis of high-definition morphological profiles. Our approach reveals how distinct facets of actin cytoskeleton remodeling shape immunological synapse architecture and affect lytic granule positioning. Morphological profiling of CD8+ T cells from immunodeficient individuals allows discrimination of the roles of the ARP2/3 subunit ARPC1B and the ARP2/3 activator Wiskott-Aldrich syndrome protein (WASP) in immunological synapse assembly. Single-cell analysis further identifies uncoupling of lytic granules and F-actin radial distribution in ARPC1B-deficient lymphocytes. Our study provides a foundation for development of morphological profiling as a scalable approach to monitor primary lymphocyte responsiveness and to identify complex aspects of lymphocyte micro-architecture.


Asunto(s)
Forma de la Célula , Imagenología Tridimensional , Células Asesinas Naturales/citología , Linfocitos T/citología , Complejo 2-3 Proteico Relacionado con la Actina/deficiencia , Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Adolescente , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/efectos de los fármacos , Línea Celular , Forma de la Célula/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Exocitosis/efectos de los fármacos , Humanos , Sinapsis Inmunológicas/efectos de los fármacos , Sinapsis Inmunológicas/metabolismo , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/metabolismo , Masculino , Compuestos de Organoselenio/farmacología , Compuestos de Organosilicio/farmacología , Análisis de la Célula Individual , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Tionas/farmacología , Uracilo/análogos & derivados , Uracilo/farmacología , Proteína del Síndrome de Wiskott-Aldrich/deficiencia , Proteína del Síndrome de Wiskott-Aldrich/metabolismo
3.
Part Fibre Toxicol ; 18(1): 26, 2021 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-34330311

RESUMEN

The gastrointestinal tract is a complex interface between the external environment and the immune system. Its ability to control uptake across the mucosa and to protect the body from damage of harmful substances from the lumen is defined as the intestinal barrier function (IBF). The IBF involves four elements: the intestinal microbiota, the mucus layer, the epithelium and the immune system. Its dysfunction is linked with human diseases including inflammatory, metabolic, infectious, autoimmune and neurologic disorders. Most of these diseases are complex and involve genetic, psychological and environmental factors. Over the past 10 years, many genetic polymorphisms predisposing to inflammatory bowel disease (IBD) have been identified. Yet, it is now clear that they are insufficient to explain the onset of these chronic diseases. Although it has been evidenced that some environmental factors such as cigarette smoking or carbohydrate intake are associated with IBD, other environmental factors also present potential health risks such as ingestion of food additives introduced in the human diet, including those composed of mineral particles, by altering the four elements of the intestinal barrier function. The aim of this review is to provide a critical opinion on the potential of TiO2 particles, especially when used as a food additive, to alter the four elements of the intestinal barrier function, and consequently to evaluate if this additive would likely play a role in the development and/or exacerbation of IBD.


Asunto(s)
Neoplasias Colorrectales , Enfermedades Inflamatorias del Intestino , Neoplasias Colorrectales/inducido químicamente , Neoplasias Colorrectales/genética , Dieta/efectos adversos , Humanos , Enfermedades Inflamatorias del Intestino/inducido químicamente , Enfermedades Inflamatorias del Intestino/genética , Mucosa Intestinal , Titanio
4.
Front Cell Dev Biol ; 9: 656795, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34026755

RESUMEN

The cytolethal distending toxin (CDT) is produced by several Gram-negative pathogenic bacteria. In addition to inflammation, experimental evidences are in favor of a protumoral role of CDT-harboring bacteria such as Escherichia coli, Campylobacter jejuni, or Helicobacter hepaticus. CDT may contribute to cell transformation in vitro and carcinogenesis in mice models, through the genotoxic action of CdtB catalytic subunit. Here, we investigate the mechanism of action by which CDT leads to genetic instability in human cell lines and colorectal organoids from healthy patients' biopsies. We demonstrate that CDT holotoxin induces a replicative stress dependent on CdtB. The slowing down of DNA replication occurs mainly in late S phase, resulting in the expression of fragile sites and important chromosomic aberrations. These DNA abnormalities induced after CDT treatment are responsible for anaphase bridge formation in mitosis and interphase DNA bridge between daughter cells in G1 phase. Moreover, CDT-genotoxic potential preferentially affects human cycling cells compared to quiescent cells. Finally, the toxin induces nuclear distension associated to DNA damage in proliferating cells of human colorectal organoids, resulting in decreased growth. Our findings thus identify CDT as a bacterial virulence factor targeting proliferating cells, such as human colorectal progenitors or stem cells, inducing replicative stress and genetic instability transmitted to daughter cells that may therefore contribute to carcinogenesis. As some CDT-carrying bacterial strains were detected in patients with colorectal cancer, targeting these bacteria could be a promising therapeutic strategy.

5.
Cancers (Basel) ; 13(8)2021 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-33916891

RESUMEN

Colorectal cancer (CRC) is the third most common cause of cancer-related death. Significant improvements in CRC treatment have been made for the last 20 years, on one hand thanks to a better detection, allowing surgical resection of the incriminated area, and on the other hand, thanks to a better knowledge of CRC's development allowing the improvement of drug strategies. Despite this crucial progress, CRC remains a public health issue. The current model for CRC initiation and progression is based on accumulation of sequential known genetic mutations in the colon epithelial cells' genome leading to a loss of control over proliferation and survival. However, increasing evidence reveals that CRC initiation is more complex. Indeed, chronic inflammatory contexts, such as inflammatory bowel diseases, have been shown to increase the risk for CRC development in mice and humans. In this manuscript, we review whether colon fibroblasts can go from the main regulators of the ISC homeostasis, regulating not only the renewal process but also the epithelial cells' differentiation occurring along the colon crypt, to the main player in the initiation of the colorectal cancer process due to chronic inflammation.

6.
Cells ; 9(12)2020 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-33297478

RESUMEN

Intestinal stem cells (ISC) are crucial players in colon epithelium physiology. The accurate control of their auto-renewal, proliferation and differentiation capacities provides a constant flow of regeneration, maintaining the epithelial intestinal barrier integrity. Under stress conditions, colon epithelium homeostasis in disrupted, evolving towards pathologies such as inflammatory bowel diseases or colorectal cancer. A specific environment, namely the ISC niche constituted by the surrounding mesenchymal stem cells, the factors they secrete and the extracellular matrix (ECM), tightly controls ISC homeostasis. Colon ECM exerts physical constraint on the enclosed stem cells through peculiar topography, stiffness and deformability. However, little is known on the molecular and cellular events involved in ECM regulation of the ISC phenotype and fate. To address this question, combining accurately reproduced colon ECM mechanical parameters to primary ISC cultures such as organoids is an appropriated approach. Here, we review colon ECM physical properties at physiological and pathological states and their bioengineered in vitro reproduction applications to ISC studies.


Asunto(s)
Colon/metabolismo , Epitelio/metabolismo , Matriz Extracelular/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Mucosa Intestinal/metabolismo , Células Madre/citología , Animales , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Células Epiteliales/citología , Homeostasis , Humanos , Inflamación , Enfermedades Inflamatorias del Intestino/patología , Intestinos/citología , Ratones , Organoides/citología , Fenotipo , Nicho de Células Madre , Andamios del Tejido/química
7.
Front Cell Dev Biol ; 8: 363, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32582690

RESUMEN

Inflammatory Bowel Diseases (IBD) are chronic inflammatory disorders, where epithelial defects drive, at least in part, some of the pathology. We reconstituted human intestinal epithelial organ, by using three-dimension culture of human colon organoids. Our aim was to characterize morphological and functional phenotypes of control (non-IBD) organoids, compared to inflamed organoids from IBD patients. The results generated describe the epithelial defects associated with IBD in primary organoid cultures, and evaluate the use of this model for pharmacological testing of anti-inflammatory approaches. Human colonic tissues were obtained from either surgical resections or biopsies, all harvested in non-inflammatory zones. Crypts were isolated from controls (non-IBD) and IBD patients and were cultured up to 12-days. Morphological (size, budding formation, polarization, luminal content), cell composition (proliferation, differentiation, immaturity markers expression), and functional (chemokine and tight junction protein expression) parameters were measured by immunohistochemistry, RT-qPCR or western-blot. The effects of inflammatory cocktail or anti-inflammatory treatments were studied in controls and IBD organoid cultures respectively. Organoid cultures from controls or IBD patients had the same cell composition after 10 to 12-days of culture, but IBD organoid cultures showed an inflammatory phenotype with decreased size and budding capacity, increased cell death, luminal debris, and inverted polarization. Tight junction proteins were also significantly decreased in IBD organoid cultures. Inflammatory cytokine cocktail reproduced this inflammatory phenotype in non-IBD organoids. Clinically used treatments (5-ASA, glucocorticoids, anti-TNF) reduced some, but not all parameters. Inflammatory phenotype is associated with IBD epithelium, and can be studied in organoid cultures. This model constitutes a reliable human pre-clinical model to investigate new strategies targeting epithelial repair.

8.
Artículo en Inglés | MEDLINE | ID: mdl-31708870

RESUMEN

Protease-activated receptors (PARs) belong to the G protein-coupled receptor (GPCR) family. Compared to other GPCRs, the specificity of the four PARs is the lack of physiologically soluble ligands able to induce their activation. Indeed, PARs are physiologically activated after proteolytic cleavage of their N-terminal domain by proteases. The resulting N-terminal end becomes a tethered activation ligand that interact with the extracellular loop 2 domain and thus induce PAR signal. PARs expression is ubiquitous and these receptors have been largely described in chronic inflammatory diseases and cancer. In this review, after describing their discovery, structure, mechanisms of activation, we then focus on the roles of PARs in the intestine and the two main diseases affecting the organ, namely inflammatory bowel diseases and cancer.

9.
Int J Mol Sci ; 20(19)2019 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-31547627

RESUMEN

The tumor bulk is composed of a highly heterogeneous population of cancer cells, as well as a large variety of resident and infiltrating host cells, extracellular matrix proteins, and secreted proteins, collectively known as the tumor microenvironment (TME). The TME is essential for driving tumor development by promoting cancer cell survival, migration, metastasis, chemoresistance, and the ability to evade the immune system responses. Therapeutically targeting tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs), regulatory T-cells (T-regs), and mesenchymal stromal/stem cells (MSCs) is likely to have an impact in cancer treatment. In this review, we focus on describing the normal physiological functions of each of these cell types and their behavior in the cancer setting. Relying on the specific surface markers and secreted molecules in this context, we review the potential targeting of these cells inducing their depletion, reprogramming, or differentiation, or inhibiting their pro-tumor functions or recruitment. Different approaches were developed for this targeting, namely, immunotherapies, vaccines, small interfering RNA, or small molecules.


Asunto(s)
Inmunoterapia , Neoplasias , Microambiente Tumoral/inmunología , Fibroblastos Asociados al Cáncer/inmunología , Fibroblastos Asociados al Cáncer/patología , Diferenciación Celular/inmunología , Humanos , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/patología , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/terapia , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología
10.
Cell Mol Gastroenterol Hepatol ; 7(2): 357-369, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30704984

RESUMEN

Nucleotide-binding oligomerization domain 2 (NOD2) is an intracellular pattern recognition receptor that senses bacterial peptidoglycan-conserved motifs in cytosol and stimulates host immune response including epithelial and immune cells. The association of NOD2 mutations with a number of inflammatory pathologies including Crohn's disease (CD), graft-versus-host diseases, or Blau syndrome, highlights its pivotal role in inflammatory response and the associated-carcinogenesis development. Since its identification in 2001 and its association with CD, the role of NOD2 in epithelial cells and immune cells has been investigated extensively but the precise mechanism by which NOD2 mutations lead to CD and the associated carcinogenesis development is largely unknown. In this review, we present and discuss recent developments about the role of NOD2 inside epithelial cells on the control of the inflammatory process and its linked carcinogenesis development.


Asunto(s)
Carcinogénesis/patología , Células Epiteliales/patología , Inflamación/patología , Intestinos/patología , Proteína Adaptadora de Señalización NOD2/metabolismo , Animales , Carcinogénesis/metabolismo , Células Epiteliales/metabolismo , Humanos , Inflamación/metabolismo , Modelos Biológicos
11.
Int J Cancer ; 145(2): 494-502, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-30628725

RESUMEN

The Focal adhesion kinase (FAK) is a ubiquitous cytoplasmic tyrosine-kinase promoting tumor progression and metastasis processes by acting in cancer cells and their tumor microenvironment partners. FAK overexpression in primary colon tumors and their metastasis is associated to poor colorectal cancer (CRC) patients' outcome. Eight FAK mRNA alternative splice variants have been described and contribute to additional level of FAK activity regulation, some of them corresponding to overactivated FAK isoforms. To date, FAK mRNA alternative splice variants expression and implication in CRC processes remain unknown. Here, using different human CRC cells lines displaying differential invasive capacities in an in vivo murine model recapitulating the different steps of CRC development from primary tumors to liver and lung metastasis, we identified three out of the eight mRNA variants (namely FAK0 , FAK28 and FAK6 ) differentially expressed along the CRC process and the tumor sites. Our results highlight an association between FAK0 and FAK6 expressions and the metastatic potential of the most aggressive cell lines HT29 and HCT116, suggesting that FAK0 and FAK6 could represent aggressiveness markers in CRC. Our findings also suggest a more specific role for FAK28 in the interactions between the tumors cells and their microenvironment. In conclusion, targeting FAK0 , the common form of FAK, might not be a good strategy based on the numerous roles of this kinase in physiological processes. In contrast, FAK6 or FAK28 splice variants, or their corresponding protein isoforms, may putatively represent future therapeutic target candidates in the development of CRC primary tumors and metastasis.


Asunto(s)
Empalme Alternativo , Neoplasias Colorrectales/patología , Quinasa 1 de Adhesión Focal/genética , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/secundario , Animales , Biomarcadores de Tumor/genética , Neoplasias Colorrectales/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Células HT29 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones , Invasividad Neoplásica , Trasplante de Neoplasias , Isoformas de ARN/genética , Regulación hacia Arriba
12.
Materials (Basel) ; 12(1)2019 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-30621089

RESUMEN

In the fields of biology and medicine, nanoproducts such as nanoparticles (NPs) are specifically interesting as theranostic tools, since they offer the double capacity to locally deliver active drugs and to image exactly where the product is delivered. Among the many described possibilities, silica nanoparticles (SiNPs) represent a good choice because of their ease of synthesis, the possibility of their vast functionalization, and their good biocompatibility. However, SiNPs' passive cell internalization by endocytosis only distributes NPs into the cell cytoplasm and is unable to target the nucleus if SiNPs are larger than a few nanometers. In this study, we demonstrate that the cell penetration of SiNPs of 28⁻30 nm in diameter can be strongly enhanced using a physical method, called electroporation or electropermeabilization (EP). The uptake of fluorescently labelled silica nanoparticles was improved in two different cancer cell lines, namely, HCT-116 (human colon cancer) cells and RL (B-lymphoma) cells. First, we studied cells' capability for the regular passive uptake of SiNPs in vitro. Then, we set EP parameters in order to induce a more efficient and rapid cell loading, also comprising the nuclear compartment, while preserving the cell viability. In the final approach, we performed in vivo experiments, and evidenced that the labeling was long-lasting, as confirmed by fluorescence imaging of labeled tumors, which enabled a 30-day follow-up. This kind of SiNPs delivery, achieved by EP, could be employed to load extensive amounts of active ingredients into the cell nucleus, and concomitantly allow the monitoring of the long-term fate of nanoparticles.

14.
Br J Pharmacol ; 175(18): 3656-3668, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29959891

RESUMEN

BACKGROUND AND PURPOSE: Thrombin is massively released upon tissue damage associated with bleeding or chronic inflammation. The effects of this thrombin on tissue regrowth and repair has been scarcely addressed and only in cancer cell lines. Hence, the purpose of the present study was to determine thrombin's pharmacological effects on human intestinal epithelium growth, proliferation and apoptosis, using three-dimensional cultures of human colon organoids. EXPERIMENTAL APPROACH: Crypts were isolated from human colonic resections and cultured for 6 days, forming human colon organoids. Cultured organoids were exposed to 10 and 50 mU·mL-1 of thrombin, in the presence or not of protease-activated receptor (PAR) antagonists. Organoid morphology, metabolism, proliferation and apoptosis were followed. KEY RESULTS: Thrombin favoured organoid maturation leading to a decreased number of immature cystic structures and a concomitant increased number of larger structures releasing cell debris and apoptotic cells. The size of budding structures, metabolic activity and proliferation were significantly reduced in organoid cultures exposed to thrombin, while apoptosis was dramatically increased. Both PAR1 and PAR4 antagonists inhibited apoptosis regardless of thrombin doses. Thrombin-induced inhibition of proliferation and metabolic activity were reversed by PAR4 antagonist for thrombin's lowest dose and by PAR1 antagonist for thrombin's highest dose. CONCLUSIONS AND IMPLICATIONS: Overall, our data suggest that the presence of thrombin in the vicinity of human colon epithelial cells favours their maturation at the expense of their regenerative capacities. Our data point to thrombin and its two receptors PAR1 and PAR4 as potential molecular targets for epithelial repair therapies.


Asunto(s)
Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Colon/efectos de los fármacos , Organoides/efectos de los fármacos , Receptor PAR-1/metabolismo , Receptores de Trombina/metabolismo , Trombina/farmacología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Colon/citología , Humanos , Organoides/citología , Organoides/crecimiento & desarrollo
15.
Pharm Biol ; 55(1): 124-131, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27925497

RESUMEN

CONTEXT AND OBJECTIVE: Diplotaxis harra (Forssk.) Boiss. (Brassicaceae) is traditionally used as an antidiabetic, anti-inflammatory or anticancer agent. In these pathologies, the glycogen synthase kinase 3 ß (GSK3ß) is overactivated and represents an interesting therapeutic target. Several flavonoids can inhibit GSK3ß and the purpose of this study was to search for the compounds in Diplotaxis harra which are able to modulate GSK3ß. MATERIALS AND METHODS: Methanol extracts from D. harra flowers were prepared and the bio-guided fractionation of their active compounds was performed using inflammatory [protease-activated receptor 2 (PAR2)-stimulated IEC6 cells] and cancer (human Caco-2 cell line) intestinal cells. 50-100 µg/mL of fractions or compounds purified by HPLC were incubated with cells whose inhibited form of GSK3ß (Pser9 GSK3ß) and survival were analyzed by Western blot at 1 h and colorimetric assay at 24 h, respectively. LC-UV-MS profiles and MS-MS spectra were used for the characterization of extracts and flavonoids-enriched fractions, and the identification of pure flavonoids was achieved by MS and NMR analysis. RESULTS: The methanol extract from D. harra flowers and its flavonoid-enriched fraction inhibit GSK3ß in PAR2-stimulated IEC6 cells. GSK3ß inhibition by the flavonoid-enriched D. harra fraction was dependent on PKC activation. The flavonoid-enriched D. harra fraction and its purified compound isorhamnetin-3,7-di-O-glucoside induced a 20% decrease of PAR2-stimulated IEC6 and Caco-2 cell survival. Importantly, normal cells (non-stimulated IEC6 cells) were spared by these treatments. CONCLUSION: This work indicates that flavonoids from D. harra display cytotoxic activity against inflammatory and cancer intestinal cells which could depend on GSK3ß inhibition.


Asunto(s)
Antiinflamatorios/farmacología , Antineoplásicos Fitogénicos/farmacología , Brassicaceae/química , Neoplasias del Colon/tratamiento farmacológico , Flavonoles/farmacología , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Glicósidos/farmacología , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Extractos Vegetales/farmacología , Antiinflamatorios/aislamiento & purificación , Antineoplásicos Fitogénicos/aislamiento & purificación , Células CACO-2 , Supervivencia Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Flavonoles/aislamiento & purificación , Flores , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Glicósidos/aislamiento & purificación , Humanos , Enfermedades Inflamatorias del Intestino/enzimología , Enfermedades Inflamatorias del Intestino/patología , Espectroscopía de Resonancia Magnética , Metanol/química , Fitoterapia , Extractos Vegetales/aislamiento & purificación , Plantas Medicinales , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Solventes/química , Espectrometría de Masas en Tándem
16.
Am J Physiol Gastrointest Liver Physiol ; 311(2): G221-36, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27313176

RESUMEN

Protease-activated receptors PAR1 and PAR2 play an important role in the control of epithelial cell proliferation and migration. However, the survival of normal and tumor intestinal stem/progenitor cells promoted by proinflammatory mediators may be critical in oncogenesis. The glycogen synthase kinase-3ß (GSK3ß) pathway is overactivated in colon cancer cells and promotes their survival and drug resistance. We thus aimed to determine PAR1 and PAR2 effects on normal and tumor intestinal stem/progenitor cells and whether they involved GSK3ß. First, PAR1 and PAR2 were identified in colon stem/progenitor cells by immunofluorescence. In three-dimensional cultures of murine crypt units or single tumor Caco-2 cells, PAR2 activation decreased numbers and size of normal or cancerous spheroids, and PAR2-deficient spheroids showed increased proliferation, indicating that PAR2 represses proliferation. PAR2-stimulated normal cells were more resistant to stress (serum starvation or spheroid passaging), suggesting prosurvival effects of PAR2 Accordingly, active caspase-3 was strongly increased in PAR2-deficient normal spheroids. PAR2 but not PAR1 triggered GSK3ß activation through serine-9 dephosphorylation in normal and tumor cells. The PAR2-triggered GSK3ß activation implicates an arrestin/PP2A/GSK3ß complex that is dependent on the Rho kinase activity. Loss of PAR2 was associated with high levels of GSK3ß nonactive form, strengthening the role of PAR2 in GSK3ß activation. GSK3 pharmacological inhibition impaired the survival of PAR2-stimulated spheroids and serum-starved cells. Altogether our data identify PAR2/GSK3ß as a novel pathway that plays a critical role in the regulation of stem/progenitor cell survival and proliferation in normal colon crypts and colon cancer.


Asunto(s)
Colon/enzimología , Células Epiteliales/enzimología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células Madre Neoplásicas/enzimología , Receptor PAR-2/metabolismo , Células Madre/enzimología , Animales , Arrestina/metabolismo , Células CACO-2 , Proliferación Celular , Supervivencia Celular , Colon/patología , Activación Enzimática , Células Epiteliales/patología , Humanos , Masculino , Ratones Endogámicos C57BL , Células Madre Neoplásicas/patología , Fosforilación , Proteína Fosfatasa 2/metabolismo , Interferencia de ARN , Receptor PAR-2/genética , Transducción de Señal , Esferoides Celulares , Nicho de Células Madre , Células Madre/patología , Transfección , Microambiente Tumoral , Quinasas Asociadas a rho/metabolismo
17.
Biochim Biophys Acta ; 1844(3): 487-96, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24334106

RESUMEN

The Src-family tyrosine kinases (SFKs) are oncogenic enzymes that contribute to the initiation and progression of many types of cancer. In normal cells, SFKs are kept in an inactive state mainly by phosphorylation of a consensus regulatory tyrosine near the C-terminus (Tyr(530) in the SFK c-Src). As recent data indicate that tyrosine modification enhances binding of metal ions, the hypothesis that SFKs might be regulated by metal ions was investigated. The c-Src C-terminal peptide bound two Fe(3+) ions with affinities at pH4.0 of 33 and 252µM, and phosphorylation increased the affinities at least 10-fold to 1.4 and 23µM, as measured by absorbance spectroscopy. The corresponding phosphorylated peptide from the SFK Lyn bound two Fe(3+) ions with much higher affinities (1.2pM and 160nM) than the Src C-terminal peptide. Furthermore, when Lyn or Hck kinases, which had been stabilised in the inactive state by phosphorylation of the C-terminal regulatory tyrosine, were incubated with Fe(3+) ions, a significant enhancement of kinase activity was observed. In contrast Lyn or Hck kinases in the unphosphorylated active state were significantly inhibited by Fe(3+) ions. These results suggest that Fe(3+) ions can regulate SFK activity by binding to the phosphorylated C-terminal regulatory tyrosine.


Asunto(s)
Compuestos Férricos/metabolismo , Familia-src Quinasas/metabolismo , Secuencia de Aminoácidos , Cationes , Activación Enzimática , Datos de Secuencia Molecular , Fosforilación , Unión Proteica , Resonancia por Plasmón de Superficie , Familia-src Quinasas/química
18.
J Biol Chem ; 287(49): 41458-68, 2012 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-23055519

RESUMEN

F1 domain of F(1)F(o)-ATPase was initially believed to be strictly expressed in the mitochondrial membrane. Interestingly, recent reports have shown that the F1 complex can serve as a cell surface receptor for apparently unrelated ligands. Here we show for the first time the presence of the F(1)-ATPase at the cell surface of normal or cancerous colonic epithelial cells. Using surface plasmon resonance technology and mass spectrometry, we identified a peptide hormone product of the gastrin gene (glycine-extended gastrin (G-gly)) as a new ligand for the F(1)-ATPase. By molecular modeling, we identified the motif in the peptide sequence (E(E/D)XY), that directly interacts with the F(1)-ATPase and the amino acids in the F(1)-ATPase that bind this motif. Replacement of the Glu-9 residue by an alanine in the E(E/D)XY motif resulted in a strong decrease of G-gly binding to the F(1)-ATPase and the loss of its biological activity. In addition we demonstrated that F(1)-ATPase mediates the growth effects of the peptide. Indeed, blocking F(1)-ATPase activity decreases G-gly-induced cell growth. The mechanism likely involves ADP production by the membrane F(1)-ATPase, which is induced by G-gly. These results suggest an important contribution of cell surface F(1)-ATPase in the pro-proliferative action of this gastrointestinal peptide.


Asunto(s)
Membrana Celular/enzimología , Colon/enzimología , Células Epiteliales/metabolismo , ATPasas de Translocación de Protón/química , Adenosina Difosfato/química , Secuencia de Aminoácidos , Animales , Células CACO-2 , Dominio Catalítico , Bovinos , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Células Endoteliales/citología , Humanos , Mitocondrias/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos/química , Homología de Secuencia de Aminoácido , Resonancia por Plasmón de Superficie
19.
Cancer Prev Res (Phila) ; 5(4): 675-84, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22366915

RESUMEN

The most frequently occurring lesions in the colon are the hyperplastic polyps. Hyperplastic polyps have long been considered as lesions with no malignant potential and colonoscopy for these patients is not recommended. However, recent works suggest that hyperplastic polyps may represent precursor lesions of some sporadic colorectal cancers. Until now, no biomarker allows to identify the subset of hyperplastic polyps that may have a malignant potential. Because the hormone precursor progastrin has been involved in colon carcinogenesis, we investigated whether its expression in hyperplastic polyps predicts the occurrence of colonic neoplasm after resection of hyperplastic polyps. We retrospectively analyzed progastrin expression in hyperplastic polyps from 74 patients without history of colorectal pathology. In our study, 41% of patients presenting an initial hyperplastic polyp subsequently developed adenomatous polyps, recognized as precursor lesions for colorectal adenocarcinomas. Progastrin was overexpressed in the hyperplastic polyps in 40% of the patients. We showed a significant association between progastrin overexpression and shortened neoplasm-free survival (P = 0.001). Patients with high overexpression of progastrin had a 5-year neoplasm-free survival rate of 38% as compared with 100% for the patients with low progastrin expression. In addition, we established a predictive test on the basis of progastrin staining and patients' age that predicts occurrence of neoplasm after developing a first hyperplastic polyp with a sensitivity of 100% [95% confidence interval (CI), 79%-100%] and a specificity of 74% (51%-90%). We show that progastrin expression evaluation in hyperplastic polyps is an efficient prognostic tool to determine patients with higher risk of metachronous neoplasms who could benefit from an adapted follow-up.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Pólipos del Colon/metabolismo , Gastrinas/biosíntesis , Precursores de Proteínas/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Cohortes , Supervivencia sin Enfermedad , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Curva ROC , Reproducibilidad de los Resultados
20.
World J Gastroenterol ; 17(40): 4488-95, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-22110279

RESUMEN

AIM: To analyse αv integrin expression induced by gastrin in pancreatic cancer models. METHODS: αv integrin mRNA expression in human pancreatic cancer cells was analysed using a "cancer genes" array and confirmed by real-time reverse transcription-polymerase chain reaction (PCR). Western blotting and semi-quantitative immunohistochemistry were used to examine protein levels in human pancreatic cancer cell lines and pancreatic tissues, respectively. The role of αv integrin on gastrin-induced cell adhesion was examined using blocking anti-αv integrin monoclonal antibodies. Adherent cells were quantified by staining with crystal violet. RESULTS: Using a "cancer genes" array we identified αv integrin as a new gastrin target gene in human pancreatic cancer cells. A quantitative real-time PCR approach was used to confirm αv integrin gene expression. We also demonstrate that Src family kinases and the PI 3-kinase, two signalling pathways specifically activated by the CCK-2 receptor (CCK2R), are involved in gastrin-mediated αv integrin expression. In contrast, inhibition of the ERK pathway was without any effect on αv integrin expression induced by gastrin. Our results also show that gastrin modulates cell adhesion via αv integrins. Indeed, in vitro adhesion assays performed on fibronectin show that gastrin significantly increases adhesion of pancreatic cancer cells. The use of blocking anti-αv integrin monoclonal antibodies completely reversed the increase in cell-substrate adhesion induced by gastrin. In addition, we showed in vivo that the targeted CCK2R expression in the pancreas of Elas-CCK2 mice, leads to the overexpression of αv integrin. This process may contribute to pancreatic tumour development observed in these transgenic animals. CONCLUSION: αv integrin is a new gastrin target in pancreatic cancer models and contributes to gastrin effects on cell adhesion.


Asunto(s)
Gastrinas/metabolismo , Integrina alfaV/metabolismo , Neoplasias Pancreáticas/metabolismo , Animales , Adhesión Celular/fisiología , Línea Celular Tumoral , Gastrinas/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Integrina alfaV/genética , Ratones , Ratones Transgénicos , Análisis por Micromatrices/métodos , Fosfatidilinositol 3-Quinasas/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Receptor de Colecistoquinina B/genética , Receptor de Colecistoquinina B/metabolismo , Transducción de Señal/fisiología , Familia-src Quinasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...