Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Pflugers Arch ; 476(6): 1007-1018, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38613695

RESUMEN

Neutrophil granulocytes play a crucial role in host defense against invading pathogens and in inflammatory diseases. The aim of this study was to elucidate membrane potential dynamics during the initial phase of neutrophil activation and its relation to migration and production of reactive oxygen species (ROS). We performed ROS production measurements of neutrophils from healthy C57BL/6J mice after TNFα-priming and/or C5a stimulation. The actin cytoskeleton was visualized with fluorescence microscopy. Furthermore, we combined migration assays and measurements of membrane potential dynamics after stimulating unprimed and/or TNFα-primed neutrophils with C5a. We show that C5a has a concentration-dependent effect on ROS production and chemokinetic migration. Chemokinetic migration and chemotaxis are impaired at C5a concentrations that induce ROS production. The actin cytoskeleton of unstimulated and of ROS-producing neutrophils is not distributed in a polarized way. Inhibition of the phagocytic NADPH oxidase NOX2 with diphenyleneiodonium (DPI) leads to a polarized distribution of the actin cytoskeleton and rescues chemokinetic migration of primed and C5a-stimulated neutrophils. Moreover, C5a evokes a pronounced depolarization of the cell membrane potential by 86.6 ± 4.2 mV starting from a resting membrane potential of -74.3 ± 0.7 mV. The C5a-induced depolarization occurs almost instantaneously (within less than one minute) in contrast to the more gradually developing depolarization induced by PMA (lag time of 3-4 min). This initial depolarization is accompanied by a decrease of the migration velocity. Collectively, our results show that stimulation with C5a evokes parallel changes in membrane potential dynamics, neutrophil ROS production and motility. Notably, the amplitude of membrane potential dynamics is comparable to that of excitable cells.


Asunto(s)
Complemento C5a , Potenciales de la Membrana , Ratones Endogámicos C57BL , Neutrófilos , Especies Reactivas de Oxígeno , Animales , Neutrófilos/metabolismo , Neutrófilos/efectos de los fármacos , Neutrófilos/fisiología , Complemento C5a/metabolismo , Complemento C5a/farmacología , Especies Reactivas de Oxígeno/metabolismo , Ratones , Potenciales de la Membrana/fisiología , NADPH Oxidasas/metabolismo , Citoesqueleto de Actina/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Movimiento Celular/efectos de los fármacos , Activación Neutrófila , NADPH Oxidasa 2/metabolismo
2.
Exp Dermatol ; 33(1): e14983, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38009253

RESUMEN

Tumour cell detachment from the primary tumour is an early and crucial step of the metastatic cascade. At the single cell level, it was already shown that migrating melanoma cells establish both intra- and extracellular pH gradients and that the Na+ /H+ exchanger NHE1 accumulates at the leading edges to strengthen cell-matrix interactions. However, less is known about the role of NHE1 in collective cell migration and the specific pH microenvironment at tumour cell-cell contacts. We used MV3 melanoma cells transfected with a NHE1-expressing vector or a control vector. NHE1 localization at cell-cell contacts was assessed via immunofluorescence imaging. Collective migration was analysed by live-cell imaging. The NHE1 activity and the perimembranous pH were measured both intra- and extracellularly by ratiometric fluorescence microscopy. NHE1 clearly localizes at cell-cell contacts. Its overexpression further increases migratory speed and translocation in multidirectional pathway analyses. NHE1 overexpressing MV3 cells also move further away from their neighbouring cells during wound closure assays. pH measurements revealed that the NHE1 is highly active at cell-cell contacts of melanoma cells. NHE1-mediated pH dynamics at such contact sites are more prominent in NHE1-overexpressing melanoma cells. Our findings highlight the contribution of the NHE1 towards modulation and plasticity of melanoma cell-cell contacts. We propose that its localization and functional activity at cell-cell contacts promotes evasion of single melanoma cells from the primary tumour.


Asunto(s)
Melanoma , Humanos , Intercambiador 1 de Sodio-Hidrógeno/metabolismo , Melanoma/metabolismo , Línea Celular Tumoral , Intercambiadores de Sodio-Hidrógeno/metabolismo , Comunicación Celular , Concentración de Iones de Hidrógeno , Microambiente Tumoral
3.
J Immunol ; 209(1): 136-144, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35715008

RESUMEN

Neutrophil granulocytes are the first and robust responders to the chemotactic molecules released from an inflamed acidic tissue. The aim of this study was to elucidate the role of microenvironmental pH in neutrophil chemotaxis. To this end, we used neutrophils from male C57BL/6J mice and combined live cell imaging chemotaxis assays with measurements of the intracellular pH (pHi) in varied extracellular pH (pHe). Observational studies were complemented by biochemical analyses of leukotriene B4 (LTB4) production and activation of the Cdc42 Rho GTPase. Our data show that pHi of neutrophils dose-dependently adapts to a given pH of the extracellular milieu. Neutrophil chemotaxis toward C5a has an optimum at pHi ∼7.1, and its pHi dependency is almost parallel to that of LTB4 production. Consequently, a shallow pHe gradient, resembling that encountered by neutrophils during extravasation from a blood vessel (pH ∼7.4) into the interstitium (pH ∼7.2), favors chemotaxis of stimulated neutrophils. Lowering pHe below pH 6.8, predominantly affects neutrophil chemotaxis, although the velocity is largely maintained. Inhibition of the Na+/H+ exchanger 1 (NHE1) with cariporide drastically attenuates neutrophil chemotaxis at the optimal pHi irrespective of the high LTB4 production. Neutrophil migration and chemotaxis are almost completely abrogated by inhibiting LTB4 production or blocking its receptor (BLT1). The abundance of the active GTP-bound form of Cdc42 is strongly reduced by NHE1 inhibition or pHe 6.5. In conclusion, we propose that the pH dependence of neutrophil chemotaxis toward C5a is caused by a pHi-dependent production of LTB4 and activation of Cdc42. Moreover, it requires the activity of NHE1.


Asunto(s)
Leucotrieno B4 , Neutrófilos , Animales , Quimiotaxis , Quimiotaxis de Leucocito , Concentración de Iones de Hidrógeno , Masculino , Ratones , Ratones Endogámicos C57BL , Neutrófilos/fisiología
4.
Pflugers Arch ; 475(10): 1225-1240, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37566113

RESUMEN

Pancreatic stellate cells (PSCs) that can co-metastasize with cancer cells shape the tumor microenvironment (TME) in pancreatic ductal adenocarcinoma (PDAC) by producing an excessive amount of extracellular matrix. This leads to a TME characterized by increased tissue pressure, hypoxia, and acidity. Moreover, cells within the tumor secrete growth factors. The stimuli of the TME trigger Ca2+ signaling and cellular Na+ loading. The Na+/Ca2+ exchanger (NCX) connects the cellular Ca2+ and Na+ homeostasis. The NCX is an electrogenic transporter, which shuffles 1 Ca2+ against 3 Na+ ions over the plasma membrane in a forward or reverse mode. Here, we studied how the impact of NCX activity on PSC migration is modulated by cues from the TME. NCX expression was revealed with qPCR and Western blot. [Ca2+]i, [Na+]i, and the cell membrane potential were determined with the fluorescent indicators Fura-2, Asante NaTRIUM Green-2, and DiBAC4(3), respectively. PSC migration was quantified with live-cell imaging. To mimic the TME, PSCs were exposed to hypoxia, pressure, acidic pH (pH 6.6), and PDGF. NCX-dependent signaling was determined with Western blot analyses. PSCs express NCX1.3 and NCX1.9. [Ca2+]i, [Na+]i, and the cell membrane potential are 94.4 nmol/l, 7.4 mmol/l, and - 39.8 mV, respectively. Thus, NCX1 usually operates in the forward (Ca2+ export) mode. NCX1 plays a differential role in translating cues from the TME into an altered migratory behavior. When NCX1 is operating in the forward mode, its inhibition accelerates PSC migration. Thus, NCX1-mediated extrusion of Ca2+ contributes to a slow mode of migration of PSCs.


Asunto(s)
Células Estrelladas Pancreáticas , Intercambiador de Sodio-Calcio , Humanos , Intercambiador de Sodio-Calcio/metabolismo , Células Estrelladas Pancreáticas/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Transducción de Señal , Hipoxia , Calcio/metabolismo
5.
Artículo en Inglés | MEDLINE | ID: mdl-32772273

RESUMEN

Ion channels are a major class of membrane proteins that play central roles in signaling within and among cells, as well as in the coupling of extracellular events with cellular responses. Dysregulated ion channel activity plays a causative role in many diseases including cancer. Here, we will review their role in lung cancer. Lung cancer is one of the most frequently diagnosed cancers, and it causes the highest number of deaths of all cancer types. The overall 5-year survival rate of lung cancer patients is only 19% and decreases to 5% when patients are diagnosed with stage IV. Thus, new therapeutical strategies are urgently needed. The important contribution of ion channels to the progression of various types of cancer has been firmly established so that ion channel-based therapeutic concepts are currently developed. Thus far, the knowledge on ion channel function in lung cancer is still relatively limited. However, the published studies clearly show the impact of ion channel inhibitors on a number of cellular mechanisms underlying lung cancer cell aggressiveness such as proliferation, migration, invasion, cell cycle progression, or adhesion. Additionally, in vivo experiments reveal that ion channel inhibitors diminish tumor growth in mice. Furthermore, some studies give evidence that ion channel inhibitors can have an influence on the resistance or sensitivity of lung cancer cells to common chemotherapeutics such as paclitaxel or cisplatin.


Asunto(s)
Neoplasias Pulmonares , Animales , Humanos , Canales Iónicos , Neoplasias Pulmonares/tratamiento farmacológico , Ratones , Transducción de Señal
6.
Biol Chem ; 404(4): 325-337, 2023 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-36594183

RESUMEN

Cancer is characterized by uncontrolled growth, invasion, and metastasis. In addition to solid cancer cells, cancer-associated fibroblasts (CAFs) play important roles in cancer pathophysiology. They arise from "healthy" cells but get manipulated by solid cancer cells to supply them and develop a tumor microenvironment (TME) that protects the cancer cells from the immune defense. A wide variety of cell types can differentiate into CAFs, including fibroblasts, endothelial cells, and epithelial cells. Precise Ca2+ regulation is essential for each cell including CAFs. The electrogenic Na+/Ca2+ exchanger (NCX) is one of the ubiquitously expressed regulatory Ca2+ transport proteins that rapidly responds to changes of the intracellular ion concentrations. Its transport function is also influenced by the membrane potential and thereby indirectly by the activity of ion channels. NCX transports Ca2+ out of the cell (forward mode) or allows its influx (reverse mode), always in exchange for 3 Na+ ions that are moved into the opposite direction. In this review, we discuss the functional roles NCX has in CAFs and how these depend on the properties of the TME. NCX activity modifies migration and leads to a reduced proliferation and apoptosis. The effect of the NCX in fibrosis is still largely unknown.


Asunto(s)
Fibroblastos Asociados al Cáncer , Neoplasias , Células Endoteliales/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Intercambiador de Sodio-Calcio/metabolismo , Fibroblastos/metabolismo , Calcio/metabolismo
7.
Biol Chem ; 404(4): 339-353, 2023 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-36571487

RESUMEN

Ion channels play an important role for regulation of the exocrine and the endocrine pancreas. This review focuses on the Ca2+-regulated K+ channel KCa3.1, encoded by the KCNN4 gene, which is present in both parts of the pancreas. In the islets of Langerhans, KCa3.1 channels are involved in the regulation of membrane potential oscillations characterizing nutrient-stimulated islet activity. Channel upregulation is induced by gluco- or lipotoxic conditions and might contribute to micro-inflammation and impaired insulin release in type 2 diabetes mellitus as well as to diabetes-associated renal and vascular complications. In the exocrine pancreas KCa3.1 channels are expressed in acinar and ductal cells. They are thought to play a role for anion secretion during digestion but their physiological role has not been fully elucidated yet. Pancreatic carcinoma, especially pancreatic ductal adenocarcinoma (PDAC), is associated with drastic overexpression of KCa3.1. For pharmacological targeting of KCa3.1 channels, we are discussing the possible benefits KCa3.1 channel inhibitors might provide in the context of diabetes mellitus and pancreatic cancer, respectively. We are also giving a perspective for the use of a fluorescently labeled derivative of the KCa3.1 blocker senicapoc as a tool to monitor channel distribution in pancreatic tissue. In summary, modulating KCa3.1 channel activity is a useful strategy for exo-and endocrine pancreatic disease but further studies are needed to evaluate its clinical suitability.


Asunto(s)
Diabetes Mellitus Tipo 2 , Islotes Pancreáticos , Neoplasias Pancreáticas , Humanos , Páncreas , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas
8.
PLoS Comput Biol ; 18(5): e1010089, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35584137

RESUMEN

The motility of neutrophils and their ability to sense and to react to chemoattractants in their environment are of central importance for the innate immunity. Neutrophils are guided towards sites of inflammation following the activation of G-protein coupled chemoattractant receptors such as CXCR2 whose signaling strongly depends on the activity of Ca2+ permeable TRPC6 channels. It is the aim of this study to analyze data sets obtained in vitro (murine neutrophils) and in vivo (zebrafish neutrophils) with a stochastic mathematical model to gain deeper insight into the underlying mechanisms. The model is based on the analysis of trajectories of individual neutrophils. Bayesian data analysis, including the covariances of positions for fractional Brownian motion as well as for exponentially and power-law tempered model variants, allows the estimation of parameters and model selection. Our model-based analysis reveals that wildtype neutrophils show pure superdiffusive fractional Brownian motion. This so-called anomalous dynamics is characterized by temporal long-range correlations for the movement into the direction of the chemotactic CXCL1 gradient. Pure superdiffusion is absent vertically to this gradient. This points to an asymmetric 'memory' of the migratory machinery, which is found both in vitro and in vivo. CXCR2 blockade and TRPC6-knockout cause tempering of temporal correlations in the chemotactic gradient. This can be interpreted as a progressive loss of memory, which leads to a marked reduction of chemotaxis and search efficiency of neutrophils. In summary, our findings indicate that spatially differential regulation of anomalous dynamics appears to play a central role in guiding efficient chemotactic behavior.


Asunto(s)
Quimiotaxis , Neutrófilos , Animales , Teorema de Bayes , Factores Quimiotácticos , Quimiotaxis/fisiología , Ratones , Canal Catiónico TRPC6 , Pez Cebra
9.
Mol Ther ; 30(4): 1536-1552, 2022 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-35031433

RESUMEN

Extravasation of circulating tumor cells (CTCs) is critical for metastasis and is initiated by adhesive interactions between glycoligands on CTCs and E-selectin on endothelia. Here, we show that the clinically approved proteasome inhibitor bortezomib (BZM; Velcade) counteracts the cytokine-dependent induction of E-selectin in the lung mediated by the primary tumor, thereby impairing endothelial adhesion and thus spontaneous lung metastasis in vivo. However, the efficacy of BZM crucially depends on the tumor cells' E-selectin ligands, which determine distinct adhesion patterns. The canonical ligands sialyl-Lewis A (sLeA) and sLeX mediate particularly high-affinity E-selectin binding so that the incomplete E-selectin-reducing effect of BZM is not sufficient to disrupt adhesion or metastasis. In contrast, tumor cells lacking sLeA/X nevertheless bind E-selectin, but with low affinity, so that adhesion and lung metastasis are significantly diminished. Such low-affinity E-selectin ligands apparently consist of sialylated MGAT5 products on CD44. BZM no longer has anti-metastatic activity after CD44 knockdown in sLeA/X-negative tumor cells or E-selectin knockout in mice. sLeA/X can be determined by immunohistochemistry in cancer samples, which might aid patient stratification. These data suggest that BZM might act as a drug for inhibiting extravasation and thus distant metastasis formation in malignancies expressing low-affinity E-selectin ligands.


Asunto(s)
Selectina E , Neoplasias Pulmonares , Animales , Bortezomib/farmacología , Antígeno CA-19-9/farmacología , Adhesión Celular , Selectina E/genética , Selectina E/metabolismo , Humanos , Ligandos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Ratones , Metástasis de la Neoplasia , Oligosacáridos , Antígeno Sialil Lewis X
10.
Pflugers Arch ; 474(11): 1147-1157, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36152073

RESUMEN

Lung cancer is one of the leading causes of cancer-related deaths worldwide. The Ca2+-activated K+ channel KCa3.1 contributes to the progression of non-small cell lung cancer (NSCLC). Recently, KCa3.1 channels were found in the inner membrane of mitochondria in different cancer cells. Mitochondria are the main sources for the generation of reactive oxygen species (ROS) that affect the progression of cancer cells. Here, we combined Western blotting, immunofluorescence, and fluorescent live-cell imaging to investigate the expression and function of KCa3.1 channels in the mitochondria of NSCLC cells. Western blotting revealed KCa3.1 expression in mitochondrial lysates from different NSCLC cells. Using immunofluorescence, we demonstrate a co-localization of KCa3.1 channels with mitochondria of NSCLC cells. Measurements of the mitochondrial membrane potential with TMRM reveal a hyperpolarization following the inhibition of KCa3.1 channels with the cell-permeable blocker senicapoc. This is not the case when cells are treated with the cell-impermeable peptidic toxin maurotoxin. The hyperpolarization of the mitochondrial membrane potential is accompanied by an increased generation of ROS in NSCLC cells. Collectively, our results provide firm evidence for the functional expression of KCa3.1 channels in the inner membrane of mitochondria of NSCLC cells.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo
11.
Arch Pharm (Weinheim) ; 355(12): e2200388, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36161669

RESUMEN

The calcium-activated potassium channel 3.1 (KCa 3.1) is overexpressed in many tumor entities and has predictive power concerning disease progression and outcome. Imaging of the KCa 3.1 channel in vivo using a radiotracer for positron emission tomography (PET) could therefore establish a potentially powerful diagnostic tool. Senicapoc shows high affinity and excellent selectivity toward the KCa 3.1 channel. We have successfully pursued the synthesis of the 18 F-labeled derivative [18 F]3 of senicapoc using the prosthetic group approach with 1-azido-2-[18 F]fluoroethane ([18 F]6) in a "click" reaction. The biological activity of the new PET tracer was evaluated in vitro and in vivo. Inhibition of the KCa 3.1 channel by 3 was demonstrated by patch clamp experiments and the binding pose was analyzed by docking studies. In mouse and human serum, [18 F]3 was stable for at least one half-life of [18 F]fluorine. Biodistribution experiments in wild-type mice were promising, showing rapid and predominantly renal excretion. An in vivo study using A549-based tumor-bearing mice was performed. The tumor signal could be delineated and image analysis showed a tumor-to-muscle ratio of 1.47 ± 0.24. The approach using 1-azido-2-[18 F]fluoroethane seems to be a good general strategy to achieve triarylacetamide-based fluorinated PET tracers for imaging of the KCa 3.1 channel in vivo.


Asunto(s)
Neoplasias , Canales de Potasio Calcio-Activados , Animales , Humanos , Ratones , Radioisótopos de Flúor/metabolismo , Radiofármacos/farmacología , Radiofármacos/metabolismo , Distribución Tisular , Canales de Potasio Calcio-Activados/metabolismo , Relación Estructura-Actividad , Tomografía de Emisión de Positrones/métodos , Neoplasias/metabolismo
12.
Int J Mol Sci ; 23(12)2022 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-35743115

RESUMEN

In the fight against prostate cancer (PCa), TRPM8 is one of the most promising clinical targets. Indeed, several studies have highlighted that TRPM8 involvement is key in PCa progression because of its impact on cell proliferation, viability, and migration. However, data from the literature are somewhat contradictory regarding the precise role of TRPM8 in prostatic carcinogenesis and are mostly based on in vitro studies. The purpose of this study was to clarify the role played by TRPM8 in PCa progression. We used a prostate orthotopic xenograft mouse model to show that TRPM8 overexpression dramatically limited tumor growth and metastasis dissemination in vivo. Mechanistically, our in vitro data revealed that TRPM8 inhibited tumor growth by affecting the cell proliferation and clonogenic properties of PCa cells. Moreover, TRPM8 impacted metastatic dissemination mainly by impairing cytoskeleton dynamics and focal adhesion formation through the inhibition of the Cdc42, Rac1, ERK, and FAK pathways. Lastly, we proved the in vivo efficiency of a new tool based on lipid nanocapsules containing WS12 in limiting the TRPM8-positive cells' dissemination at metastatic sites. Our work strongly supports the protective role of TRPM8 on PCa progression, providing new insights into the potential application of TRPM8 as a therapeutic target in PCa treatment.


Asunto(s)
Neoplasias de la Próstata , Canales Catiónicos TRPM , Animales , Carcinogénesis/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Metástasis de la Neoplasia/patología , Próstata/patología , Neoplasias de la Próstata/metabolismo , Canales Catiónicos TRPM/genética , Canales Catiónicos TRPM/metabolismo
13.
Mol Cancer ; 20(1): 74, 2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33941200

RESUMEN

Loss of apical-basal polarity and downregulation of cell-cell contacts is a critical step during the pathogenesis of cancer. Both processes are regulated by the scaffolding protein Pals1, however, it is unclear whether the expression of Pals1 is affected in cancer cells and whether Pals1 is implicated in the pathogenesis of the disease.Using mRNA expression data and immunostainings of cancer specimen, we show that Pals1 is frequently downregulated in colorectal cancer, correlating with poorer survival of patients. We further found that Pals1 prevents cancer cell metastasis by controlling Rac1-dependent cell migration through inhibition of Arf6, which is independent of the canonical binding partners of Pals1. Loss of Pals1 in colorectal cancer cells results in increased Arf6 and Rac1 activity, enhanced cell migration and invasion in vitro and increased metastasis of transplanted tumor cells in mice. Thus, our data reveal a new function of Pals1 as a key inhibitor of cell migration and metastasis of colorectal cancer cells. Notably, this new function is independent of the known role of Pals1 in tight junction formation and apical-basal polarity.


Asunto(s)
Factor 6 de Ribosilación del ADP/metabolismo , Neoplasias Colorrectales/patología , Proteínas de la Membrana/metabolismo , Nucleósido-Fosfato Quinasa/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Movimiento Celular/fisiología , Células HCT116 , Xenoinjertos , Humanos , Ratones , Invasividad Neoplásica/patología
14.
Cell Physiol Biochem ; 55(S3): 131-144, 2021 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-34043300

RESUMEN

The Kca3.1 channels, previously designated as IK1 or SK4 channels and encoded by the KCNN4 gene, are activated by a rise of the intracellular Ca2+ concentration. These K+ channels are widely expressed in many organs and involved in many pathologies. In particular, Kca3.1 channels have been studied intensively in the context of cancer. They are not only a marker and a valid prognostic tool for cancer patients, but have an important share in driving cancer progression. Their function is required for many characteristic features of the aggressive cancer cell behavior such as migration, invasion and metastasis as well as proliferation and therapy resistance. In the context of cancer, another property of Kca3.1 is now emerging. These channels can be a target for novel small molecule-based imaging probes, as it has been validated in case of fluorescently labeled senicapoc-derivatives. The aim of this review is (i) to give an overview on the role of Kca3.1 channels in cancer progression and in shaping the cancer microenvironment, (ii) discuss the potential of using Kca3.1 targeting drugs for cancer imaging, (iii) and highlight the possibility of combining molecular dynamics simulations to image inhibitor binding to Kca3.1 channels in order to provide a deeper understanding of Kca3.1 channel pharmacology. Alltogether, Kca3.1 is an attractive therapeutic target so that senicapoc, originally developed for the treatment of sickle cell anemia, should be repurposed for the treatment of cancer patients.


Asunto(s)
Acetamidas/uso terapéutico , Antineoplásicos/uso terapéutico , Calcio/metabolismo , Neoplasias/tratamiento farmacológico , Bloqueadores de los Canales de Potasio/uso terapéutico , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/antagonistas & inhibidores , Compuestos de Tritilo/uso terapéutico , Antineoplásicos/química , Antidrepanocíticos/química , Antidrepanocíticos/uso terapéutico , Sitios de Unión , Señalización del Calcio , Progresión de la Enfermedad , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Simulación de Dinámica Molecular , Terapia Molecular Dirigida/métodos , Metástasis de la Neoplasia , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Bloqueadores de los Canales de Potasio/química , Estructura Secundaria de Proteína , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/química , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/genética , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética
15.
BMC Cancer ; 20(1): 264, 2020 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-32228510

RESUMEN

BACKGROUND: The human pancreatic cancer cell line A818-6 can be grown in vitro either as a highly malignant, undifferentiated monolayer (ML) or as three-dimensional (3D) single layer hollow spheres (HS) simulating a benign, highly differentiated, duct-like pancreatic epithelial structure. This characteristic allowing A818-6 cells to switch from one phenotype to another makes these cells a unique system to characterize the cellular and molecular modifications during differentiation on one hand and malignant transformation on the other hand. Ion channels and transport proteins (transportome) have been implicated in malignant transformation. Therefore, the current study aimed to analyse the transportome gene expression profile in the A818-6 cells growing as a monolayer or as hollow spheres. METHODS & RESULTS: The study identified the differentially expressed transportome genes in both cellular states of A818-6 using Agilent and Nanostring arrays and some targets were validated via immunoblotting. Additionally, these results were compared to a tissue Affymetrix microarray analysis of pancreatic adenocarcinoma patients' tissues. The overall transcriptional profile of the ML and HS cells confirmed the formerly described mesenchymal features of ML and epithelial nature of HS which was further verified via high expression of E-cadherin and low expression of vimentin found in HS in comparison to ML. Among the predicted features between HS and ML was the involvement of miRNA-9 in this switch. Importantly, the bioinformatics analysis also revealed substantial number (n = 126) of altered transportome genes. Interestingly, three genes upregulated in PDAC tissue samples (GJB2, GJB5 and SLC38A6) were found to be also upregulated in ML and 3 down-regulated transportome genes (KCNQ1, TRPV6 and SLC4A) were also reduced in ML. CONCLUSION: This reversible HS/ML in vitro system might help in understanding the pathophysiological impact of the transportome in the dedifferentiation process in pancreatic carcinogenesis. Furthermore, the HS/ML model represents a novel system for studying the role of the transportome during the switch from a more benign, differentiated (HS) to a highly malignant, undifferentiated (ML) phenotype.


Asunto(s)
Adenocarcinoma/metabolismo , Neoplasias Pancreáticas/metabolismo , Transcriptoma/genética , Adenocarcinoma/patología , Sistemas de Transporte de Aminoácidos Neutros/genética , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Carcinogénesis/genética , Diferenciación Celular/genética , Línea Celular Tumoral , Plasticidad de la Célula , Biología Computacional , Conexina 26 , Conexinas/genética , Conexinas/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/genética , Neoplasias Pancreáticas/patología
16.
Angew Chem Int Ed Engl ; 59(21): 8277-8284, 2020 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-32097518

RESUMEN

Small-molecule probes for the in vitro imaging of KCa 3.1 channel-expressing cells were developed. Senicapoc, showing high affinity and selectivity for the KCa 3.1 channels, was chosen as the targeting component. BODIPY dyes 15-20 were synthesized and connected by a CuI -catalyzed azide-alkyne [3+2]cycloaddition with propargyl ether senicapoc derivative 8, yielding fluorescently labeled ligands 21-26. The dimethylpyrrole-based imaging probes 25 and 26 allow staining of KCa 3.1 channels in NSCLC cells. The specificity was shown by removing the punctate staining pattern by pre-incubation with senicapoc. The density of KCa 3.1 channels detected with 25 and by immunostaining was identical. The punctate structure of the labeled channels could also be observed in living cells. Molecular modeling showed binding of the senicapoc-targeting component towards the binding site within the ion channel and orientation of the linker with the dye along the inner surface of the ion channel.


Asunto(s)
Colorantes Fluorescentes/química , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Células A549 , Sitios de Unión , Compuestos de Boro/química , Reacción de Cicloadición , Colorantes Fluorescentes/síntesis química , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/química , Ligandos , Microscopía Fluorescente , Simulación de Dinámica Molecular
17.
Physiol Rev ; 92(4): 1865-913, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23073633

RESUMEN

Cell motility is central to tissue homeostasis in health and disease, and there is hardly any cell in the body that is not motile at a given point in its life cycle. Important physiological processes intimately related to the ability of the respective cells to migrate include embryogenesis, immune defense, angiogenesis, and wound healing. On the other side, migration is associated with life-threatening pathologies such as tumor metastases and atherosclerosis. Research from the last ≈ 15 years revealed that ion channels and transporters are indispensable components of the cellular migration apparatus. After presenting general principles by which transport proteins affect cell migration, we will discuss systematically the role of channels and transporters involved in cell migration.


Asunto(s)
Movimiento Celular/fisiología , Canales Iónicos/metabolismo , Transporte Iónico/fisiología , Proteínas de Transporte de Membrana/metabolismo , Animales
19.
FASEB J ; 32(3): 1665-1676, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29162704

RESUMEN

Podocyte malfunction is central to glomerular diseases and is marked by defective podocyte intercellular junctions and actin cytoskeletal dynamics. Podocytes share many morphologic features with neurons, so that similar sets of proteins appear to regulate cell process formation. One such protein is the tropomyosin-related kinase C (TrkC). TrkC deficiency in mice leads to proteinuria as a surrogate of defective kidney filter function. Activation of endogenous TrkC by its ligand neurotrophin-3 resulted in increased podocyte migration-a surrogate of podocyte actin dynamics in vivo. Employing a mutagenesis approach, we found that the Src homologous and collagen-like (Shc) binding site Tyr516 within the TrkC cytoplasmic domain was necessary for TrkC-induced migration of podocytes. TrkC activation led to a mobility shift of Wiskott-Aldrich syndrome family verprolin-homologous protein (WAVE)-2 which is known to orchestrate Arp2/3 activation and actin polymerization. Chemical inactivation of Erk or mutagenesis of 2 of 4 known Erk target sites within WAVE2, Thr346 and Ser351, abolished the TrkC-induced WAVE2 mobility shift. Knockdown of WAVE2 by shRNA abolished TrkC-induced podocyte migration. In summary, TrkC signals to the podocyte actin cytoskeleton to induce migration by phosphorylating WAVE2 Erk dependently. This signaling mechanism may be important for TrkC-mediated cytoskeletal dynamics in podocyte disease.-Gromnitza, S., Lepa, C., Weide, T., Schwab, A., Pavenstädt, H., George, B. Tropomyosin-related kinase C (TrkC) enhances podocyte migration by ERK-mediated WAVE2 activation.


Asunto(s)
Movimiento Celular , Citoesqueleto/metabolismo , Sistema de Señalización de MAP Quinasas , Podocitos/metabolismo , Receptor trkC/metabolismo , Familia de Proteínas del Síndrome de Wiskott-Aldrich/metabolismo , Animales , Línea Celular Transformada , Citoesqueleto/genética , Ratones , Neurotrofina 3/biosíntesis , Neurotrofina 3/genética , Podocitos/citología , Dominios Proteicos , Receptor trkC/genética , Familia de Proteínas del Síndrome de Wiskott-Aldrich/genética
20.
Bioessays ; 39(6)2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28440551

RESUMEN

We present here the hypothesis that the unique microenvironmental pH landscape of acid-base transporting epithelia is an important factor in development of epithelial cancers, by rendering the epithelial and stromal cells pre-adapted to the heterogeneous extracellular pH (pHe ) in the tumor microenvironment. Cells residing in organs with net acid-base transporting epithelia such as the pancreatic ductal and gastric epithelia are exposed to very different, temporally highly variable pHe values apically and basolaterally. This translates into spatially and temporally non-uniform intracellular pH (pHi ) patterns. Disturbed pHe - and pHi -homeostasis contributes to essentially all hallmarks of cancer. Our hypothesis, that the physiological pHe microenvironment in acid-base secreting epithelia shapes cancers arising in these tissues, can be tested using novel imaging tools. The acidic tumor pHe in turn might be exploited therapeutically. Pancreatic cancers are used as our prime example, but we propose that this concept is also relevant for other cancers of acid-base transporting epithelia.


Asunto(s)
Carcinogénesis , Neoplasias Pancreáticas/patología , Microambiente Tumoral , Animales , Progresión de la Enfermedad , Humanos , Concentración de Iones de Hidrógeno , Neoplasias Pancreáticas/química , Neoplasias Pancreáticas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA