Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 141
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 42(13): e112198, 2023 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-37278161

RESUMEN

There is growing evidence that ion channels are critically involved in cancer cell invasiveness and metastasis. However, the molecular mechanisms of ion signaling promoting cancer behavior are poorly understood and the complexity of the underlying remodeling during metastasis remains to be explored. Here, using a variety of in vitro and in vivo techniques, we show that metastatic prostate cancer cells acquire a specific Na+ /Ca2+ signature required for persistent invasion. We identify the Na+ leak channel, NALCN, which is overexpressed in metastatic prostate cancer, as a major initiator and regulator of Ca2+ oscillations required for invadopodia formation. Indeed, NALCN-mediated Na+ influx into cancer cells maintains intracellular Ca2+ oscillations via a specific chain of ion transport proteins including plasmalemmal and mitochondrial Na+ /Ca2+ exchangers, SERCA and store-operated channels. This signaling cascade promotes activity of the NACLN-colocalized proto-oncogene Src kinase, actin remodeling and secretion of proteolytic enzymes, thus increasing cancer cell invasive potential and metastatic lesions in vivo. Overall, our findings provide new insights into an ion signaling pathway specific for metastatic cells where NALCN acts as persistent invasion controller.


Asunto(s)
Neoplasias de la Próstata , Sodio , Masculino , Humanos , Sodio/metabolismo , Canales Iónicos/metabolismo , Transporte Iónico , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo
2.
Physiol Rev ; 98(2): 559-621, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29412049

RESUMEN

Genomic instability is a primary cause and fundamental feature of human cancer. However, all cancer cell genotypes generally translate into several common pathophysiological features, often referred to as cancer hallmarks. Although nowadays the catalog of cancer hallmarks is quite broad, the most common and obvious of them are 1) uncontrolled proliferation, 2) resistance to programmed cell death (apoptosis), 3) tissue invasion and metastasis, and 4) sustained angiogenesis. Among the genes affected by cancer, those encoding ion channels are present. Membrane proteins responsible for signaling within cell and among cells, for coupling of extracellular events with intracellular responses, and for maintaining intracellular ionic homeostasis ion channels contribute to various extents to pathophysiological features of each cancer hallmark. Moreover, tight association of these hallmarks with ion channel dysfunction gives a good reason to classify them as special type of channelopathies, namely oncochannelopathies. Although the relation of cancer hallmarks to ion channel dysfunction differs from classical definition of channelopathies, as disease states causally linked with inherited mutations of ion channel genes that alter channel's biophysical properties, in a broader context of the disease state, to which pathogenesis ion channels essentially contribute, such classification seems absolutely appropriate. In this review the authors provide arguments to substantiate such point of view.


Asunto(s)
Canalopatías/genética , Inestabilidad Genómica/genética , Canales Iónicos/genética , Neoplasias/genética , Animales , Humanos , Canales Iónicos/metabolismo , Mutación/genética , Neoplasias/patología , Transducción de Señal/genética
3.
Mol Ther ; 31(4): 970-985, 2023 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-36641622

RESUMEN

Nonsense mutations are responsible for around 10% of cases of genetic diseases, including cystic fibrosis. 2,6-diaminopurine (DAP) has recently been shown to promote efficient readthrough of UGA premature stop codons. In this study, we show that DAP can correct a nonsense mutation in the Cftr gene in vivo in a new CF mouse model, in utero, and through breastfeeding, thanks, notably, to adequate pharmacokinetic properties. DAP turns out to be very stable in plasma and is distributed throughout the body. The ability of DAP to correct various endogenous UGA nonsense mutations in the CFTR gene and to restore its function in mice, in organoids derived from murine or patient cells, and in cells from patients with cystic fibrosis reveals the potential of such readthrough-stimulating molecules in developing a therapeutic approach. The fact that correction by DAP of certain nonsense mutations reaches a clinically relevant level, as judged from previous studies, makes the use of this compound all the more attractive.


Asunto(s)
Codón sin Sentido , Fibrosis Quística , Ratones , Animales , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/genética , Codón de Terminación/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética
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-32737754

RESUMEN

In the last three decades, a growing number of studies have implicated ion channels in all essential processes of prostate carcinogenesis, including cell proliferation, apoptosis, migration, and angiogenesis. The changes in the expression of individual ion channels show a specific profile, making these proteins promising clinical biomarkers that may enable better molecular subtyping of the disease and lead to more rapid and accurate clinical decision-making. Expression profiles and channel function are mainly based on the tumoral tissue itself, in this case, the epithelial cancer cell population. To date, little data on the ion channel profile of the cancerous prostate stroma are available, even though tumor interactions with the microenvironment are crucial in carcinogenesis and each distinct population plays a specific role in tumor progression. In this review, we describe ion channel expression profiles specific for the distinct cell population of the tumor microenvironment (stromal, endothelial, neuronal, and neuroendocrine cell populations) and the technical approaches used for efficient separation and screening of these cell populations.


Asunto(s)
Detección Precoz del Cáncer , Neoplasias de la Próstata , Carcinogénesis , Humanos , Canales Iónicos , Masculino , Neoplasias de la Próstata/diagnóstico , Microambiente Tumoral
6.
Int J Mol Sci ; 23(13)2022 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-35806388

RESUMEN

Solid tumours are characterised by an altered microenvironment (TME) from the physicochemical point of view, displaying a highly hypoxic and acidic interstitial fluid. Hypoxia results from uncontrolled proliferation, aberrant vascularization and altered cancer cell metabolism. Tumour cellular apparatus adapts to hypoxia by altering its metabolism and behaviour, increasing its migratory and metastatic abilities by the acquisition of a mesenchymal phenotype and selection of aggressive tumour cell clones. Extracellular acidosis is considered a cancer hallmark, acting as a driver of cancer aggressiveness by promoting tumour metastasis and chemoresistance via the selection of more aggressive cell phenotypes, although the underlying mechanism is still not clear. In this context, Ca2+ channels represent good target candidates due to their ability to integrate signals from the TME. Ca2+ channels are pH and hypoxia sensors and alterations in Ca2+ homeostasis in cancer progression and vascularization have been extensively reported. In the present review, we present an up-to-date and critical view on Ca2+ permeable ion channels, with a major focus on TRPs, SOCs and PIEZO channels, which are modulated by tumour hypoxia and acidosis, as well as the consequent role of the altered Ca2+ signals on cancer progression hallmarks. We believe that a deeper comprehension of the Ca2+ signalling and acidic pH/hypoxia interplay will break new ground for the discovery of alternative and attractive therapeutic targets.


Asunto(s)
Acidosis , Neoplasias , Humanos , Hipoxia , Neoplasias/metabolismo , Transducción de Señal , Microambiente Tumoral
7.
Int J Mol Sci ; 24(1)2022 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-36613866

RESUMEN

Though the first discovery of TRPV6 channel expression in various tissues took place in the early 2000s, reliable tools for its protein detection in various cells and tissues are still missing. Here we show the generation and validation of rabbit polyclonal anti-TRPV6 channel antibodies (rb79-82) against four epitopes of 15 amino acids. Among them, only one antibody, rb79, was capable of detecting the full-length glycosylated form of the TRPV6 channel at around 100 kDa. The generated antibody was shown to be suitable for all in vitro applications, such as immunoblotting, immunoprecipitation, immunocytochemistry, immunofluorescence, etc. One of the most important applications is immunohistochemistry using the paraffin-embedded sections from cancer resection specimens. Using prostate cancer resection specimens, we have confirmed the absence of the TRPV6 protein in both healthy and benign hyperplasia, as well as its expression and correlation to the prostate cancer grades. Thus, the generated rabbit polyclonal anti-TRPV6 channel antibody rb79 is suitable for all in vitro diagnostic applications and particularly for the diagnosis in clinics using paraffin-embedded sections from patients suffering from various diseases and disorders involving the TRPV6 channel.


Asunto(s)
Neoplasias de la Próstata , Canales Catiónicos TRPV , Humanos , Masculino , Animales , Conejos , Canales Catiónicos TRPV/metabolismo , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/metabolismo , Inmunohistoquímica , Calcio/metabolismo
8.
FASEB J ; 34(6): 7483-7499, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32277850

RESUMEN

Recent studies have revealed gender differences in cold perception, and pointed to a possible direct action of testosterone (TST) on the cold-activated TRPM8 (Transient Receptor Potential Melastatin Member 8) channel. However, the mechanisms by which TST influences TRPM8-mediated sensory functions remain elusive. Here, we show that TST inhibits TRPM8-mediated mild-cold perception through the noncanonical engagement of the Androgen Receptor (AR). Castration of both male rats and mice increases sensitivity to mild cold, and this effect depends on the presence of intact TRPM8 and AR. TST in nanomolar concentrations suppresses whole-cell TRPM8-mediated currents and single-channel activity in native dorsal root ganglion (DRG) neurons and HEK293 cells co-expressing recombinant TRPM8 and AR, but not TRPM8 alone. AR cloned from rat DRGs shows no difference from standard AR. However, biochemical assays and confocal imaging reveal the presence of AR on the cell surface and its interaction with TRPM8 in response to TST, leading to an inhibition of channel activity.


Asunto(s)
Receptores Androgénicos/metabolismo , Canales Catiónicos TRPM/metabolismo , Testosterona/metabolismo , Andrógenos/metabolismo , Animales , Línea Celular , Frío , Femenino , Ganglios Espinales/metabolismo , Células HEK293 , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Ratas , Ratas Wistar
9.
Int J Mol Sci ; 22(21)2021 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-34768857

RESUMEN

The mammalian exclusive Orai3 channel participates in the generation and/or modulation of two independent Ca2+ currents, the store-operated current, Icrac, involving functional interactions between the stromal interaction molecules (STIM), STIM1/STIM2, and Orai1/Orai2/Orai3, as well as the store-independent arachidonic acid (AA) (or leukotriene C4)-regulated current Iarc, which involves Orai1, Orai3 and STIM1. Overexpression of functional Orai3 has been described in different neoplastic cells and cancer tissue samples as compared to non-tumor cells or normal adjacent tissue. In these cells, Orai3 exhibits a cell-specific relevance in Ca2+ influx. In estrogen receptor-positive breast cancer cells and non-small cell lung cancer (NSCLC) cells store-operated Ca2+ entry (SOCE) is strongly dependent on Orai3 expression while in colorectal cancer and pancreatic adenocarcinoma cells Orai3 predominantly modulates SOCE. On the other hand, in prostate cancer cells Orai3 expression has been associated with the formation of Orai1/Orai3 heteromeric channels regulated by AA and reduction in SOCE, thus leading to enhanced proliferation. Orai3 overexpression is associated with supporting several cancer hallmarks, including cell cycle progression, proliferation, migration, and apoptosis resistance. This review summarizes the current knowledge concerning the functional role of Orai3 in the pathogenesis of cancer.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio/fisiología , Neoplasias/patología , Animales , Apoptosis/fisiología , Calcio/metabolismo , Canales de Calcio/genética , Ciclo Celular/fisiología , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Humanos
10.
Biochim Biophys Acta Mol Cell Res ; 1865(7): 981-994, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29678654

RESUMEN

Calcium (Ca2+) release from the endoplasmic reticulum plays an important role in many cell-fate defining cellular processes. Traditionally, this Ca2+ release was associated with the ER Ca2+ release channels, inositol 1,4,5­triphosphate receptor (IP3R) and ryanodine receptor (RyR). Lately, however, other calcium conductances have been found to be intracellularly localized and to participate in cell fate regulation. Nonetheless, molecular identity and functional properties of the ER Ca2+ release mechanisms associated with multiple diseases, e.g. prostate cancer, remain unknown. Here we identify a new family of transient receptor potential melastatine 8 (TRPM8) channel isoforms as functional ER Ca2+ release channels expressed in mitochondria-associated ER membranes (MAMs). These TRPM8 isoforms exhibit an unconventional structure with 4 transmembrane domains (TMs) instead of 6 TMs characteristic of the TRP channel archetype. We show that these 4TM-TRPM8 isoforms form functional channels in the ER and participate in regulation of the steady-state Ca2+ concentration ([Ca2+]) in mitochondria and the ER. Thus, our study identifies 4TM-TRPM8 isoforms as ER Ca2+ release mechanism distinct from classical Ca2+ release channels.


Asunto(s)
Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Mitocondrias/metabolismo , Neoplasias de la Próstata/metabolismo , Canales Catiónicos TRPM/genética , Canales Catiónicos TRPM/metabolismo , Anciano , Empalme Alternativo , Línea Celular Tumoral , Células Epiteliales/citología , Células Epiteliales/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Próstata/citología , Próstata/metabolismo , Neoplasias de la Próstata/genética , Dominios Proteicos , Canales Catiónicos TRPM/química
12.
Genet Med ; 21(2): 441-450, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29930394

RESUMEN

PURPOSE: Malignant hyperthermia (MH) is a pharmacogenetic disorder arising from uncontrolled muscle calcium release due to an abnormality in the sarcoplasmic reticulum (SR) calcium-release mechanism triggered by halogenated inhalational anesthetics. However, the molecular mechanisms involved are still incomplete. METHODS: We aimed to identify transient receptor potential vanilloid 1 (TRPV1) variants within the entire coding sequence in patients who developed sensitivity to MH of unknown etiology. In vitro and in vivo functional studies were performed in heterologous expression system, trpv1-/- mice, and a murine model of human MH. RESULTS: We identified TRPV1 variants in two patients and their heterologous expression in muscles of trpv1-/- mice strongly enhanced calcium release from SR upon halogenated anesthetic stimulation, suggesting they could be responsible for the MH phenotype. We confirmed the in vivo significance by using mice with a knock-in mutation (Y524S) in the type I ryanodine receptor (Ryr1), a mutation analogous to the Y522S mutation associated with MH in humans. We showed that the TRPV1 antagonist capsazepine slows the heat-induced hypermetabolic response in this model. CONCLUSION: We propose that TRPV1 contributes to MH and could represent an actionable therapeutic target for prevention of the pathology and also be responsible for MH sensitivity when mutated.


Asunto(s)
Señalización del Calcio , Predisposición Genética a la Enfermedad , Hipertermia Maligna/genética , Canales Catiónicos TRPV/genética , Anestésicos/farmacología , Animales , Calcio , Modelos Animales de Enfermedad , Femenino , Expresión Génica/efectos de los fármacos , Técnicas de Sustitución del Gen , Células HEK293 , Homeostasis , Humanos , Masculino , Hipertermia Maligna/metabolismo , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Canales Catiónicos TRPV/metabolismo
13.
Mol Phylogenet Evol ; 136: 104-118, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30980935

RESUMEN

Genes showing versatile functions or subjected to fast expansion and contraction during the adaptation of species to specific ecological conditions, like sensory receptors for odors, pheromones and tastes, are characterized by a great plasticity through evolution. One of the most fascinating sensory receptors in the family of TRP channels, the cold and menthol receptor TRPM8, has received significant attention in the literature. Recent studies have reported the existence of TRPM8 channel isoforms encoded by alternative mRNAs transcribed from alternative promoters and processed by alternative splicing. Since the first draft of the human genome was accomplished in 2000, alternative transcription, alternative splicing and alternative translation have appeared as major sources of gene product diversity and are thought to participate in the generation of complexity in higher organisms. In this study, we investigate whether alternative transcription has been a driving force in the evolution of the human forms of the cold receptor TRPM8. We identified 33 TRPM8 alternative mRNAs (24 new sequences) and their associated protein isoforms in human tissues. Using comparative genomics, we described the evolution of the human TRPM8 sequences in eight ancestors since the origin of Amniota, and estimated in which ancestors the new TRPM8 variants originated. In order to validate the estimated origins of this receptor, we performed experimental validations of predicted exons in mouse tissues. Our results suggest a first diversification event of the cold receptor in the Boreoeutheria ancestor, and a subsequent divergence at the origin of Simiiformes.


Asunto(s)
Frío , Evolución Molecular , Mentol/metabolismo , Canales Catiónicos TRPM/genética , Empalme Alternativo/genética , Animales , Línea Celular Tumoral , Exones/genética , Variación Genética , Células HEK293 , Humanos , Ratones , Sistemas de Lectura Abierta/genética , Filogenia , Isoformas de Proteínas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Canales Catiónicos TRPM/metabolismo
14.
Pflugers Arch ; 470(10): 1419-1429, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29926226

RESUMEN

While originally cloned from the prostate in 2001, transient receptor potential, melastatin member 8 (TRPM8) has since been identified as the cold/menthol receptor in the peripheral nervous system. This discovery has led to hundreds of studies regarding the role of this channel in pain and thermosensation phenomena, while relegating TRPM8 involvement in cancer to a secondary role. Despite these findings, there is growing evidence that TRPM8 should be carefully studied within the frame of carcinogenesis, especially in the prostate, where it is highly expressed and where many teams have confirmed variations in its expression during cancer progression. Its regulation by physiological factors, such as PSA and androgens, has proved that TRPM8 can exhibit an activity beyond that of a cold receptor, thus explaining how the channel can be activated in organs not exposed to temperature variations. With this review, we aim to provide a brief overview of the current knowledge regarding the complex roles of TRPM8 in prostate carcinogenesis and to show that this research path still represents a "hot" topic with potential clinical applications in the short term.


Asunto(s)
Neoplasias de la Próstata/metabolismo , Canales Catiónicos TRPM/metabolismo , Andrógenos/metabolismo , Animales , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Antígeno Prostático Específico/metabolismo , Neoplasias de la Próstata/genética , Canales Catiónicos TRPM/química , Canales Catiónicos TRPM/genética
15.
Proc Natl Acad Sci U S A ; 112(26): E3345-54, 2015 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-26080404

RESUMEN

Deviation of the ambient temperature is one of the most ubiquitous stimuli that continuously affect mammals' skin. Although the role of the warmth receptors in epidermal homeostasis (EH) was elucidated in recent years, the mystery of the keratinocyte mild-cold sensor remains unsolved. Here we report the cloning and characterization of a new functional epidermal isoform of the transient receptor potential M8 (TRPM8) mild-cold receptor, dubbed epidermal TRPM8 (eTRPM8), which is localized in the keratinocyte endoplasmic reticulum membrane and controls mitochondrial Ca(2+) concentration ([Ca(2+)]m). In turn, [Ca(2+)]m modulates ATP and superoxide (O2(·-)) synthesis in a cold-dependent manner. We report that this fine tuning of ATP and O2(·-) levels by cooling controls the balance between keratinocyte proliferation and differentiation. Finally, to ascertain eTRPM8's role in EH in vivo we developed a new functional knockout mouse strain by deleting the pore domain of TRPM8 and demonstrated that eTRPM8 knockout impairs adaptation of the epidermis to low temperatures.


Asunto(s)
Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Frío , Epidermis/metabolismo , Queratinocitos/citología , Isoformas de Proteínas/fisiología , Canales Catiónicos TRPM/fisiología , Adenosina Trifosfato/metabolismo , Animales , Calcio/metabolismo , Canales de Calcio/metabolismo , Células Cultivadas , Humanos , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Superóxidos/metabolismo
16.
J Physiol ; 595(10): 3063-3075, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28304082

RESUMEN

Intracellular calcium (Ca2+ ) signals are key regulators of multiple cellular functions, both healthy and physiopathological. It is therefore unsurprising that several cancers present a strong Ca2+ homeostasis deregulation. Among the various hallmarks of cancer disease, a particular role is played by metastasis, which has a critical impact on cancer patients' outcome. Importantly, Ca2+ signalling has been reported to control multiple aspects of the adaptive metastatic cancer cell behaviour, including epithelial-mesenchymal transition, cell migration, local invasion and induction of angiogenesis (see Abstract Figure). In this context Ca2+ signalling is considered to be a substantial intracellular tool that regulates the dynamicity and complexity of the metastatic cascade. In the present study we review the spatial and temporal organization of Ca2+ fluxes, as well as the molecular mechanisms involved in metastasis, analysing the key steps which regulate initial tumour spread.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Invasividad Neoplásica , Movimiento Celular , Transición Epitelial-Mesenquimal , Humanos
17.
Biochim Biophys Acta ; 1863(6 Pt B): 1337-43, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26658643

RESUMEN

Here, we review the role of Ca(2+) in apoptosis, namely that ER Ca(2+) depletion or a sustained elevation of cytosolic or mitochondrial Ca(2+) concentration are sufficient to trigger apoptosis. These concepts have emerged by the use of ER stressor agents that decrease the ER Ca(2+) pool by inhibiting SERCA pumps. However, aside from their well-known actions on Ca(2+) homeostasis disruption leading to apoptosis, new evidence show that some ER Ca(2+) modulators have significant implications in other Ca(2+)-mediated or Ca(2+)-independent pathways determining cell fate suggesting a more complex regulation of apoptosis by intracellular Ca(2+). Here, we discuss the crucial interplay between Ca(2+) mediated apoptosis, the Unfold Protein Response and autophagy determining cell fate, and the molecular compounds that have been used to depict these pathways. This review of the literature clearly shows the need for new inhibitors that do not interfere concomitantly with autophagy and Ca(2+) signaling. This article is part of a Special Issue entitled: Calcium and Cell Fate. Guest Editors: Jacques Haiech, Claus Heizmann, Joachim Krebs, Thierry Capiod and Olivier Mignen.


Asunto(s)
Apoptosis/fisiología , Autofagia/fisiología , Señalización del Calcio/fisiología , Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Animales , Humanos , Modelos Biológicos , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Respuesta de Proteína Desplegada/fisiología
18.
Pflugers Arch ; 469(12): 1567-1577, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28849300

RESUMEN

Pancreatic cancer is characterized by a massive fibrosis (desmoplasia), which is primarily caused by activated pancreatic stellate cells (PSCs). This leads to a hypoxic tumor microenvironment further reinforcing the activation of PSCs by stimulating their secretion of growth factors and chemokines. Since many of them elicit their effects via G-protein-coupled receptors (GPCRs), we tested whether TRPC6 channels, effector proteins of many G-protein-coupled receptor pathways, are required for the hypoxic activation of PSCs. Thus far, the function of ion channels in PSCs is virtually unexplored. qPCR revealed TRPC6 channels to be one of the most abundant TRPC channels in primary cultures of murine PSCs. TRPC6 channel function was assessed by comparing PSCs from TRPC6-/- mice and wildtype (wt) littermates. Cell migration, Ca2+ signaling, and cytokine secretion were analyzed as readout for PSC activation. Hypoxia was induced by incubating PSCs for 24 h in 1% O2 or chemically with dimethyloxalylglycine (DMOG). PSCs migrate faster in response to hypoxia. Due to reduced autocrine stimulation, TRPC6-/- PSCs fail to increase their rate of migration to the same level as wt PSCs under hypoxic conditions. This defect could not be overcome by the stimulation with platelet-derived growth factor. In line with these results, calcium influx is increased in wt but not TRPC6-/- PSCs under hypoxia. We conclude that TRPC6 channels of PSCs are major effector proteins in an autocrine stimulation pathway triggered by hypoxia.


Asunto(s)
Células Estrelladas Pancreáticas/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Hipoxia de la Célula , Células Cultivadas , Femenino , Masculino , Ratones , Ratones Noqueados , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Canal Catiónico TRPC6 , Microambiente Tumoral/fisiología
19.
Mol Carcinog ; 56(8): 1851-1867, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28277613

RESUMEN

Previous studies showed the effects of resveratrol (RES) on several cancer cells, including prostate cancer (PCa) cell apoptosis without taking into consideration the impact of the tumor microenvironment (TME). The TME is composed of cancer cells, endothelial cells, blood cells, and cancer-associated fibroblasts (CAF), the main source of growth factors. The latter cells might modify in the TME the impact of RES on tumor cells via secreted factors. Recent data clearly show the impact of CAF on cancer cells apoptosis resistance via secreted factors. However, the effects of RES on PCa CAF have not been studied so far. We have investigated here for the first time the effects of RES on the physiology of PCa CAF in the context of TME. Using a prostate cancer CAF cell line and primary cultures of CAF from prostate cancers, we show that RES activates the N-terminal mutated Transient Receptor Potential Ankyrin 1 (TRPA1) channel leading to an increase in intracellular calcium concentration and the expression and secretion of growth factors (HGF and VEGF) without inducing apoptosis in these cells. Interestingly, in the present work, we also show that when the prostate cancer cells were co-cultured with CAF, the RES-induced cancer cell apoptosis was reduced by 40%, an apoptosis reduction canceled in the presence of the TRPA1 channel inhibitors. The present work highlights CAF TRPA1 ion channels as a target for RES and the importance of the channel in the epithelial-stromal crosstalk in the TME leading to resistance to the RES-induced apoptosis.


Asunto(s)
Anticarcinógenos/farmacología , Antioxidantes/farmacología , Canales de Calcio/metabolismo , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Próstata/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Estilbenos/farmacología , Canales de Potencial de Receptor Transitorio/metabolismo , Secuencia de Aminoácidos , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Canales de Calcio/análisis , Canales de Calcio/genética , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Línea Celular Tumoral , Humanos , Masculino , Mutación , Proteínas del Tejido Nervioso/análisis , Proteínas del Tejido Nervioso/genética , Próstata/metabolismo , Próstata/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Resveratrol , Canal Catiónico TRPA1 , Canales de Potencial de Receptor Transitorio/análisis , Canales de Potencial de Receptor Transitorio/genética , Microambiente Tumoral/efectos de los fármacos
20.
FASEB J ; 30(9): 3155-70, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27317670

RESUMEN

Testes of most male mammals present the particularity of being externalized from the body and are consequently slightly cooler than core body temperature (4-8°C below). Although, hypothermia of the testis is known to increase germ cells apoptosis, little is known about the underlying molecular mechanisms, including cold sensors, transduction pathways, and apoptosis triggers. In this study, using a functional knockout mouse model of the cold and menthol receptors, dubbed transient receptor potential melastatine 8 (TRPM8) channels, we found that TRPM8 initiated the cold-shock response by differentially modulating cold- and heat-shock proteins. Besides, apoptosis of germ cells increased in proportion to the cooling level in control mice but was independent of temperature in knockout mice. We also observed that the rate of germ cell death correlated positively with the reactive oxygen species level and negatively with the expression of the detoxifying enzymes. This result suggests that the TRPM8 sensor is a key determinant of germ cell fate under hypothermic stimulation.-Borowiec, A.-S., Sion, B., Chalmel, F., Rolland, A. D., Lemonnier, L., De Clerck, T., Bokhobza, A., Derouiche, S., Dewailly, E., Slomianny, C., Mauduit, C., Benahmed, M., Roudbaraki, M., Jégou, B., Prevarskaya, N., Bidaux, G. Cold/menthol TRPM8 receptors initiate the cold-shock response and protect germ cells from cold-shock-induced oxidation.


Asunto(s)
Especies Reactivas de Oxígeno/metabolismo , Canales Catiónicos TRPM/metabolismo , Testículo/fisiología , Animales , Frío , Regulación de la Expresión Génica , Células HEK293 , Humanos , Masculino , Meiosis , Ratones , Ratones Noqueados , Oxidación-Reducción , Canales Catiónicos TRPM/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA