Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Gut ; 72(4): 722-735, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36882214

RESUMEN

OBJECTIVE: Intercellular communication within pancreatic ductal adenocarcinoma (PDAC) dramatically contributes to metastatic processes. The underlying mechanisms are poorly understood, resulting in a lack of targeted therapy to counteract stromal-induced cancer cell aggressiveness. Here, we investigated whether ion channels, which remain understudied in cancer biology, contribute to intercellular communication in PDAC. DESIGN: We evaluated the effects of conditioned media from patient-derived cancer-associated fibroblasts (CAFs) on electrical features of pancreatic cancer cells (PCC). The molecular mechanisms were deciphered using a combination of electrophysiology, bioinformatics, molecular and biochemistry techniques in cell lines and human samples. An orthotropic mouse model where CAF and PCC were co-injected was used to evaluate tumour growth and metastasis dissemination. Pharmacological studies were carried out in the Pdx1-Cre, Ink4afl/fl LSL-KrasG12D (KICpdx1) mouse model. RESULTS: We report that the K+ channel SK2 expressed in PCC is stimulated by CAF-secreted cues (8.84 vs 2.49 pA/pF) promoting the phosphorylation of the channel through an integrin-epidermal growth factor receptor (EGFR)-AKT (Protein kinase B) axis. SK2 stimulation sets a positive feedback on the signalling pathway, increasing invasiveness in vitro (threefold) and metastasis formation in vivo. The CAF-dependent formation of the signalling hub associating SK2 and AKT requires the sigma-1 receptor chaperone. The pharmacological targeting of Sig-1R abolished CAF-induced activation of SK2, reduced tumour progression and extended the overall survival in mice (11.7 weeks vs 9.5 weeks). CONCLUSION: We establish a new paradigm in which an ion channel shifts the activation level of a signalling pathway in response to stromal cues, opening a new therapeutic window targeting the formation of ion channel-dependent signalling hubs.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Animales , Ratones , Proteínas Proto-Oncogénicas c-akt , Carcinogénesis , Transformación Celular Neoplásica , Transducción de Señal , Neoplasias Pancreáticas
2.
J Urban Health ; 96(6): 823-834, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31728900

RESUMEN

Adverse pregnancy outcomes increase infants' risk for mortality and future health problems. Neighborhood physical disorder may contribute to adverse pregnancy outcomes by increasing maternal chronic stress. Google Street View technology presents a novel method for assessing neighborhood physical disorder but has not been previously examined in the context of birth outcomes. In this cross-sectional study, trained raters used Google's Street View imagery to virtually audit a randomly sampled block within each Chicago census tract (n = 809) for nine indicators of physical disorder. We used an item-response theory model and spatial interpolation to calculate tract-level neighborhood physical disorder scores across Chicago. We linked these data with geocoded electronic health record data from a large, academic women's hospital in Chicago (2015-2017, n = 14,309 births). We used three-level hierarchical Poisson regression to estimate prevalence ratios for the associations of neighborhood physical disorder with preterm birth (overall and spontaneous), small for gestational age (SGA), and hypertensive disorder of pregnancy (HDP). After adjustment for maternal sociodemographics, multiparity, and season of birth, living in a neighborhood with high physical disorder was associated with higher prevalence of PTB, SGA, and HDP (prevalence ratios and 95% confidence intervals 1.21 (1.06, 1.39) for PTB, 1.13 (1.01, 1.37) for SGA, and 1.23 (1.07, 1.42) for HDP). Adjustment for neighborhood poverty and maternal health conditions (e.g., hypertension, diabetes, asthma, substance use) attenuated associations. Results suggest that an adverse neighborhood physical environment may contribute to adverse pregnancy outcomes. However, future work is needed to disentangle the unique contribution of physical disorder from other characteristics of disadvantaged neighborhoods.


Asunto(s)
Madres/psicología , Pobreza/estadística & datos numéricos , Resultado del Embarazo , Nacimiento Prematuro/epidemiología , Características de la Residencia/estadística & datos numéricos , Salud de la Mujer/estadística & datos numéricos , Adulto , Chicago/epidemiología , Estudios Transversales , Registros Electrónicos de Salud , Femenino , Humanos , Recién Nacido , Embarazo , Prevalencia
3.
Proc Natl Acad Sci U S A ; 114(16): 4159-4164, 2017 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-28373572

RESUMEN

The K+ channel KCNQ1 has been proposed as a tumor suppressor in colorectal cancer (CRC). We investigated the molecular mechanisms regulating KCNQ1:ß-catenin bidirectional interactions and their effects on CRC differentiation, proliferation, and invasion. Molecular and pharmacologic approaches were used to determine the influence of KCNQ1 expression on the Wnt/ß-catenin signaling and epithelial-to-mesenchymal transition (EMT) in human CRC cell lines of varying stages of differentiation. The expression of KCNQ1 was lost with increasing mesenchymal phenotype in poorly differentiated CRC cell lines as a consequence of repression of the KCNQ1 promoter by ß-catenin:T-cell factor (TCF)-4. In well-differentiated epithelial CRC cell lines, KCNQ1 was localized to the plasma membrane in a complex with ß-catenin and E-cadherin. The colocalization of KCNQ1 with adherens junction proteins was lost with increasing EMT phenotype. ShRNA knock-down of KCNQ1 caused a relocalization of ß-catenin from the plasma membrane and a loss of epithelial phenotype in CRC spheroids. Overexpression of KCNQ1 trapped ß-catenin at the plasma membrane, induced a patent lumen in CRC spheroids, and slowed CRC cell invasion. The KCNQ1 ion channel inhibitor chromanol 293B caused membrane depolarization, redistribution of ß-catenin into the cytosol, and a reduced transepithelial electrical resistance, and stimulated CRC cell proliferation. Analysis of human primary CRC tumor patient databases showed a positive correlation between KCNQ1:KCNE3 channel complex expression and disease-free survival. We conclude that the KCNQ1 ion channel is a target gene and regulator of the Wnt/ß-catenin pathway, and its repression leads to CRC cell proliferation, EMT, and tumorigenesis.


Asunto(s)
Diferenciación Celular , Movimiento Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Regulación Neoplásica de la Expresión Génica , Canal de Potasio KCNQ1/metabolismo , beta Catenina/metabolismo , Animales , Apoptosis , Carcinogénesis , Proliferación Celular , Neoplasias Colorrectales/genética , Transición Epitelial-Mesenquimal , Humanos , Canal de Potasio KCNQ1/genética , Masculino , Invasividad Neoplásica , Pronóstico , Regiones Promotoras Genéticas , Ratas Sprague-Dawley , Tasa de Supervivencia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/genética
4.
Cancer Res ; 76(3): 607-18, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26645564

RESUMEN

The sigma 1 receptor (Sig1R) is a stress-activated chaperone that regulates ion channels and is associated with pathologic conditions, such as stroke, neurodegenerative diseases, and addiction. Aberrant expression levels of ion channels and Sig1R have been detected in tumors and cancer cells, such as myeloid leukemia and colorectal cancer, but the link between ion channel regulation and Sig1R overexpression during malignancy has not been established. In this study, we found that Sig1R dynamically controls the membrane expression of the human voltage-dependent K(+) channel human ether-à-go-go-related gene (hERG) in myeloid leukemia and colorectal cancer cell lines. Sig1R promoted the formation of hERG/ß1-integrin signaling complexes upon extracellular matrix stimulation, triggering the activation of the PI3K/AKT pathway. Consequently, the presence of Sig1R in cancer cells increased motility and VEGF secretion. In vivo, Sig1R expression enhanced the aggressiveness of tumor cells by potentiating invasion and angiogenesis, leading to poor survival. Collectively, our findings highlight a novel function for Sig1R in mediating cross-talk between cancer cells and their microenvironment, thus driving oncogenesis by shaping cellular electrical activity in response to extracellular signals. Given the involvement of ion channels in promoting several hallmarks of cancer, our study also offers a potential strategy to therapeutically target ion channel function through Sig1R inhibition.


Asunto(s)
Neoplasias/metabolismo , Neoplasias/patología , Receptores sigma/biosíntesis , Animales , Adhesión Celular/fisiología , Línea Celular Tumoral , Membrana Celular/metabolismo , Membrana Celular/fisiología , Movimiento Celular/fisiología , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Células HCT116 , Células HEK293 , Humanos , Células K562 , Ratones , Células 3T3 NIH , Invasividad Neoplásica , Neoplasias/genética , Receptores sigma/genética , Transducción de Señal , Receptor Sigma-1
5.
Int J Cell Biol ; 2011: 136802, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21876696

RESUMEN

Missense mutations in the erythroid band 3 protein (Anion Exchanger 1) have been associated with hereditary stomatocytosis. Features of cation leaky red cells combined with functional expression of the mutated protein led to the conclusion that the AE1 point mutations were responsible for Na(+) and K(+) leak through a conductive mechanism. A molecular mechanism explaining mutated AE1-linked stomatocytosis involves changes in AE1 transport properties that become leaky to Na(+) and K(+). However, another explanation suggests that point-mutated AE1 could regulate a cation leak through other transporters. This short paper intends to discuss these two alternatives.

6.
J Biol Chem ; 286(32): 27947-58, 2011 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-21680736

RESUMEN

Sig1R (Sigma-1receptor) is a 25-kDa protein structurally unrelated to other mammalian proteins. Sig1R is present in brain, liver, and heart and is overexpressed in cancer cells. Studies using exogenous sigma ligands have shown that Sig1R interacts with a variety of ion channels, but its intrinsic function and mechanism of action remain unclear. The human ether-à-gogo related gene (hERG) encodes a cardiac channel that is also abnormally expressed in many primary human cancers, potentiating tumor progression through the modulation of extracellular matrix adhesive interactions. We show herein that sigma ligands inhibit hERG current density and cell adhesion to fibronectin in K562 myeloid leukemia cells. Heterologous expression in Xenopus oocytes demonstrates that Sig1R potentiates hERG current by stimulating channel subunit biosynthesis. Silencing Sig1R in leukemic K562 cells depresses hERG current density and cell adhesion to fibronectin by reducing hERG membrane expression. In K562 cells, Sig1R silencing does not modify hERG mRNA contents but reduces hERG mature form densities. In HEK cells expressing hERG and Sig1R, both proteins co-immunoprecipitate, demonstrating a physical association. Finally, Sig1R expression enhances both channel protein maturation and stability. Altogether, these results demonstrate for the first time that Sig1R controls ion channel expression through the regulation of subunit trafficking activity.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/biosíntesis , Regulación Leucémica de la Expresión Génica , Leucemia Mieloide/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores sigma/metabolismo , Animales , Adhesión Celular/genética , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/genética , Femenino , Fibronectinas/genética , Fibronectinas/metabolismo , Humanos , Transporte Iónico , Células K562 , Leucemia Mieloide/genética , Leucemia Mieloide/patología , Proteínas de Neoplasias/genética , Estabilidad Proteica , Receptores sigma/genética , Xenopus laevis , Receptor Sigma-1
7.
Biochem J ; 426(3): 379-88, 2010 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-20028337

RESUMEN

dRTA (distal renal tubular acidosis) and HS (hereditary spherocytosis) are two diseases that can be caused by mutations in the gene encoding the AE1 (anion exchanger 1; Band 3). dRTA is characterized by defective urinary acidification, leading to metabolic acidosis, renal stones and failure to thrive. HS results in anaemia, which may require regular blood transfusions and splenectomy. Mutations in the gene encoding AE1 rarely cause both HS and dRTA. In the present paper, we describe a novel AE1 mutation, Band 3 Edmonton I, which causes dominant HS and recessive dRTA. The patient is a compound heterozygote with the new mutation C479W and the previously described mutation G701D. Red blood cells from the patient presented a reduced amount of AE1. Expression in a kidney cell line showed that kAE1 (kidney AE1) C479W is retained intracellularly. As kAE1 is a dimer, we performed co-expression studies and found that, in kidney cells, kAE1 C479W and G701D proteins traffic independently from each other despite their ability to form heterodimers. Therefore the patient carries one kAE1 mutant that is retained in the Golgi (G701D) and another kAE1 mutant (C479W) located in the endoplasmic reticulum of kidney cells, and is thus probably unable to reabsorb bicarbonate into the blood. We conclude that the C479W mutant is a novel trafficking mutant of AE1, which causes HS due to a decreased cell-surface AE1 protein and results in dRTA due to its intracellular retention in kidney.


Asunto(s)
Acidosis Tubular Renal/genética , Proteína 1 de Intercambio de Anión de Eritrocito/genética , Mutación , Esferocitosis Hereditaria/genética , Acidosis Tubular Renal/metabolismo , Acidosis Tubular Renal/patología , Animales , Proteína 1 de Intercambio de Anión de Eritrocito/metabolismo , Secuencia de Bases , Línea Celular , Análisis Mutacional de ADN , Salud de la Familia , Femenino , Genotipo , Humanos , Masculino , Oocitos/citología , Oocitos/metabolismo , Linaje , Esferocitosis Hereditaria/metabolismo , Esferocitosis Hereditaria/patología , Xenopus , Adulto Joven
8.
J Cell Physiol ; 213(1): 70-8, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17520699

RESUMEN

In this study, we devised a cysteine-focused point mutation analysis of the chloride channel function of trout anion exchanger 1 (tAE1) expressed in X. laevis oocytes. Seven cysteines, belonging to the transmembrane domain of tAE1, were mutated into serines (either individually or in groups) and the effects of these mutations on the chloride conductance of injected oocytes were measured. We showed that three cysteines were essential for the functional expression of tAE1. Namely, mutations C462S, C583S and C588S reduced Cl(-) conductance by 68%, 52% and 83%, respectively, when compared to wild type tAE1. These residual conductances were still inhibited by 0.5 mM niflumic acid. Western blot experiments demonstrated that C462 was involved in protein expression onto the plasma membrane. A mutant devoid of this residue was unable to express onto the plasma membrane, especially if several other cysteines were missing: consequently, the cysteine-less mutant of tAE1 was not functional. C583 and C588 were involved in the channel function of tAE1 as shown by anion substitution experiments proving that selectivity of the mutated pore differs from the wild type one. On the contrary, they were not involved in the Cl(-)/HCO(3)(-) exchange function of tAE1, as demonstrated by intracellular pH measurements. These and several complementary mutations allow us to conclude that a mutant of tAE1 containing the sole C462 can drive a marginal Cl(-) current; however, the minimal configuration necessary to get optimal functional expression of the tAE1 chloride channel is that of a mutant containing unaffected residues C462, C583 and C588.


Asunto(s)
Antiportadores de Cloruro-Bicarbonato/química , Antiportadores de Cloruro-Bicarbonato/metabolismo , Secuencia de Aminoácidos , Animales , Antiportadores de Cloruro-Bicarbonato/genética , Cisteína/química , Femenino , Concentración de Iones de Hidrógeno , Técnicas In Vitro , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Oocitos/metabolismo , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Trucha/genética , Trucha/metabolismo , Xenopus laevis
9.
J Cell Physiol ; 207(3): 829-35, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16508976

RESUMEN

In this study, we have shown that, when expressed in Xenopus oocytes, trout anion exchanger 1 (tAE1) was able to act as a bifunctional protein, either an anion exchanger or a chloride conductance. Point mutations of tAE1 were carried out and their effect on Cl- conductance and Cl- unidirectional flux were studied. We have shown that mutations made in transmembrane domain 7 had dramatic effects on tAE1 function. Indeed, when these residues were mutated, either individually or together (mutants E632K, D633G, and ED/KG), Cl- conductance was reduced to 28-44% that of wild-type tAE1. Moreover, ion substitution experiments showed that anion selectivity was altered. However, the exchanger function was unchanged, as evidenced by the fact that Cl- influx and K(m) were identical for each of these mutants and similar to the wild-type protein parameters. By contrast, mutations made in the C-terminal domains of the protein (R819M, Q829K) affected both transport functions. Cl- conductance was increased by approximately 200% with respect to tAE1 and anion selectivity was impaired. Likewise, Cl- influx was increased by approximately 260% and was no longer saturable. These and other mutations carried out in transmembrane domains 7, 8, 12-14 of tAE1 allow us to demonstrate without doubt that, in addition to its anion exchanger activity, tAE1 can also function as a chloride channel. Above all, this work led us to identify amino acids involved in this double function organization.


Asunto(s)
Proteína 1 de Intercambio de Anión de Eritrocito/metabolismo , Membrana Celular/genética , Membrana Celular/metabolismo , Canales de Cloruro/metabolismo , Trucha/genética , Trucha/metabolismo , Secuencia de Aminoácidos , Animales , Proteína 1 de Intercambio de Anión de Eritrocito/química , Proteína 1 de Intercambio de Anión de Eritrocito/genética , Canales de Cloruro/química , Canales de Cloruro/genética , Cloro/química , Cloro/metabolismo , Electrofisiología , Humanos , Datos de Secuencia Molecular , Oocitos/metabolismo , Técnicas de Placa-Clamp , Mutación Puntual/genética , Alineación de Secuencia , Xenopus laevis
10.
Biochim Biophys Acta ; 1664(1): 80-7, 2004 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15238261

RESUMEN

It was previously shown that expressed in Xenopus oocyte the trout (tAE1) and the mouse (mAE1) anion exchangers behave differently: both elicit anion exchange activity but only tAE1 induces a transport of organic solutes correlated with an anion conductance. In order to identify the structural domains involved in the induction of tAE1 channel activity, chimeras have been prepared between mouse and trout AE1. As some constructs were not expressed at the plasma membrane, skate exchanger (skAE1) was used instead of mouse exchanger to complete the structure-function analysis. The present paper shows that skAE1, highly similar to mAE1, does not induce a chloride conductance when expressed in Xenopus oocyte. Construct expression analysis showed that only tAE1 transmembrane domain is linked to the anion conductance. More precisely, we identified two regions composed of helices 6, 7 and 8 and putative helices 12 and 13 which are required for this function.


Asunto(s)
Proteína 1 de Intercambio de Anión de Eritrocito/química , Proteína 1 de Intercambio de Anión de Eritrocito/genética , Electrofisiología/métodos , Oocitos/metabolismo , Xenopus laevis/metabolismo , Animales , Bicarbonatos/química , Western Blotting , Membrana Celular/metabolismo , Canales de Cloruro/química , Ratones , Modelos Biológicos , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , ARN Complementario/metabolismo , Proteínas Recombinantes de Fusión/química , Relación Estructura-Actividad , Trucha
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA