Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Mol Cell Proteomics ; 17(6): 1126-1143, 2018 06.
Article in English | MEDLINE | ID: mdl-29531019

ABSTRACT

High grade gliomas are the most common brain tumors in adult. These tumors are characterized by a high infiltration in microglial cells and macrophages. The immunosuppressive tumor environment is known to orient immune cells toward a pro-tumoral and anti-inflammatory phenotype. Therefore, the current challenge for cancer therapy is to find a way to reorient macrophages toward an antitumoral phenotype. Previously, we demonstrated that macrophages secreted antitumoral factors when they were invalidated for the proprotein converstase 1/3 (PC1/3) and treated with LPS. However, achieving an activation of macrophages via LPS/TLR4/Myd88-dependent pathway appears yet unfeasible in cancer patients. On the contrary, the antitumor drug Paclitaxel is also known to activate the TLR4 MyD88-dependent signaling pathway and mimics LPS action. Therefore, we evaluated if PC1/3 knock-down (KD) macrophages could be activated by Paclitaxel and efficient against glioma. We report here that such a treatment of PC1/3 KD macrophages drove to the overexpression of proteins mainly involved in cytoskeleton rearrangement. In support of this finding, we found that these cells exhibited a Ca2+ increase after Paclitaxel treatment. This is indicative of a possible depolymerization of microtubules and may therefore reflect an activation of inflammatory pathways in macrophages. In such a way, we found that PC1/3 KD macrophages displayed a repression of the anti-inflammatory pathway STAT3 and secreted more pro-inflammatory cytokines. Extracellular vesicles isolated from these PC1/3 KD cells inhibited glioma growth. Finally, the supernatant collected from the coculture between glioma cells and PC1/3 KD macrophages contained more antitumoral factors. These findings unravel the potential value of a new therapeutic strategy combining Paclitaxel and PC1/3 inhibition to switch macrophages toward an antitumoral immunophenotype.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Brain Neoplasms/therapy , Glioma/therapy , Paclitaxel/pharmacology , Proprotein Convertase 1/genetics , Animals , Brain Neoplasms/metabolism , Cell Line , Cell Survival/drug effects , Coculture Techniques , Cytokines/metabolism , Glioma/metabolism , Macrophages/drug effects , Macrophages/metabolism , Proteomics , Rats
2.
Genet Med ; 21(2): 441-450, 2019 02.
Article in English | MEDLINE | ID: mdl-29930394

ABSTRACT

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.


Subject(s)
Calcium Signaling , Genetic Predisposition to Disease , Malignant Hyperthermia/genetics , TRPV Cation Channels/genetics , Anesthetics/pharmacology , Animals , Calcium , Disease Models, Animal , Female , Gene Expression/drug effects , Gene Knock-In Techniques , HEK293 Cells , Homeostasis , Humans , Male , Malignant Hyperthermia/metabolism , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , TRPV Cation Channels/metabolism
3.
Biochim Biophys Acta ; 1863(6 Pt B): 1337-43, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26658643

ABSTRACT

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.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , Calcium Signaling/physiology , Calcium/metabolism , Endoplasmic Reticulum/metabolism , Animals , Humans , Models, Biological , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Unfolded Protein Response/physiology
4.
Mol Cell Proteomics ; 14(11): 2857-77, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26330543

ABSTRACT

Macrophages provide the first line of host immune defense. Their activation triggers the secretion of pro-inflammatory cytokines and chemokines recruiting other immune cells. In cancer, macrophages present an M2 anti-inflammatory phenotype promoting tumor growth. In this way, strategies need to be develop to reactivate macrophages. Previously thought to be expressed only in cells with a neural/neuroendocrine phenotype, the proprotein convertase 1/3 has been shown to also be expressed in macrophages and regulated as a function of the Toll-like receptor immune response. Here, we investigated the intracellular impact of the down-regulation of the proprotein convertase 1/3 in NR8383 macrophages and confirmed the results on macrophages from PC1/3 deficient mice. A complete proteomic study of secretomes and intracellular proteins was undertaken and revealed that inhibition of proprotein convertase 1/3 orient macrophages toward an M1 activated phenotype. This phenotype is characterized by filopodial extensions, Toll-like receptor 4 MyD88-dependent signaling, calcium entry augmentation and the secretion of pro-inflammatory factors. In response to endotoxin/lipopolysaccharide, these intracellular modifications increased, and the secreted factors attracted naïve T helper lymphocytes to promote the cytotoxic response. Importantly, the application of these factors onto breast and ovarian cancer cells resulted in a decrease viability or resistance. Under inhibitory conditions using interleukin 10, PC1/3-knockdown macrophages continued to secrete inflammatory factors. These data indicate that targeted inhibition of proprotein convertase 1/3 could represent a novel type of immune therapy to reactivate intra-tumoral macrophages.


Subject(s)
Immunotherapy/methods , Macrophages, Alveolar/immunology , Macrophages, Peritoneal/immunology , Proprotein Convertase 1/antagonists & inhibitors , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Calcium/metabolism , Cell Line , Cell Line, Tumor , Cytokines/biosynthesis , Cytokines/immunology , Gene Expression Regulation , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Lipopolysaccharides/pharmacology , Macrophages, Alveolar/cytology , Macrophages, Alveolar/drug effects , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/drug effects , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Primary Cell Culture , Proprotein Convertase 1/genetics , Proprotein Convertase 1/immunology , Protein Array Analysis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Signal Transduction , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/pathology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
5.
Proc Natl Acad Sci U S A ; 111(37): E3870-9, 2014 Sep 16.
Article in English | MEDLINE | ID: mdl-25172921

ABSTRACT

Transient receptor potential vanilloid subfamily member 6 (TRPV6) is a highly selective calcium channel that has been considered as a part of store-operated calcium entry (SOCE). Despite its first discovery in the early 2000s, the role of this channel in prostate cancer (PCa) remained, until now, obscure. Here we show that TRPV6 mediates calcium entry, which is highly increased in PCa due to the remodeling mechanism involving the translocation of the TRPV6 channel to the plasma membrane via the Orai1/TRPC1-mediated Ca(2+)/Annexin I/S100A11 pathway, partially contributing to SOCE. The TRPV6 calcium channel is expressed de novo by the PCa cell to increase its survival by enhancing proliferation and conferring apoptosis resistance. Xenografts in nude mice and bone metastasis models confirmed the remarkable aggressiveness of TRPV6-overexpressing tumors. Immunohistochemical analysis of these demonstrated the increased expression of clinical markers such as Ki-67, prostate specific antigen, synaptophysin, CD31, and CD56, which are strongly associated with a poor prognosis. Thus, the TRPV6 channel acquires its oncogenic potential in PCa due to the remodeling mechanism via the Orai1-mediated Ca(2+)/Annexin I/S100A11 pathway.


Subject(s)
Calcium Channels/metabolism , Cell Membrane/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , TRPV Cation Channels/metabolism , Animals , Annexin A1/metabolism , Apoptosis , Bone Neoplasms/diagnostic imaging , Bone Neoplasms/secondary , Calcium/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Survival , Disease Progression , Endoplasmic Reticulum/metabolism , HEK293 Cells , Humans , Immunohistochemistry , Male , Mice, Nude , Neoplasm Invasiveness , Neoplasm Proteins/metabolism , ORAI1 Protein , Phenotype , Protein Transport , Radiography , S100 Proteins/metabolism , Signal Transduction , Xenograft Model Antitumor Assays
6.
J Cell Sci ; 126(Pt 19): 4479-89, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23943870

ABSTRACT

The mechanisms by which volatile general anaesthetics (VAs) produce a depression of central nervous system are beginning to be better understood, but little is known about a number of side effects. Here, we show that the cold receptor transient receptor potential melastatin 8 (TRPM8) undergoes a complex modulation by clinical concentrations of VAs in dorsal root ganglion neurons and HEK-293 cells heterologously expressing TRPM8. VAs produced a transient enhancement of TRPM8 through a depolarizing shift of its activation towards physiological membrane potentials, followed by a sustained TRPM8 inhibition. The stimulatory action of VAs engaged molecular determinants distinct from those used by the TRPM8 agonist. Transient TRPM8 activation by VAs could explain side effects such as inhibition of respiratory drive, shivering and the cooling sensation during the beginning of anaesthesia, whereas the second phase of VA action, that associated with sustained TRPM8 inhibition, might be responsible for hypothermia. Consistent with this, both hypothermia and the inhibition of respiratory drive induced by VAs are partially abolished in Trpm8-knockout animals. Thus, we propose TRPM8 as a new clinical target for diminishing common and serious complications of general anaesthesia.


Subject(s)
Anesthesia, General/adverse effects , Anesthetics, Inhalation/adverse effects , Anesthetics, Inhalation/pharmacology , Ganglia, Spinal/drug effects , Neurons/drug effects , TRPM Cation Channels/metabolism , Animals , Ganglia, Spinal/metabolism , HEK293 Cells , Humans , Hypothermia/chemically induced , Membrane Potentials/drug effects , Mice , Mice, Knockout , Neurons/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , TRPM Cation Channels/biosynthesis , TRPM Cation Channels/genetics , Transfection
7.
Cell Death Dis ; 14(8): 526, 2023 08 16.
Article in English | MEDLINE | ID: mdl-37587118

ABSTRACT

The dogma "One gene, one protein" is clearly obsolete since cells use alternative splicing and generate multiple transcripts which are translated into protein isoforms, but also use alternative translation initiation sites (TISs) and termination sites on a given transcript. Alternative open reading frames for individual transcripts give proteins originate from the 5'- and 3'-UTR mRNA regions, frameshifts of mRNA ORFs or from non-coding RNAs. Longtime considered as non-coding, recent in-silico translation prediction methods enriched the protein databases allowing the identification of new target structures that have not been identified previously. To gain insight into the role of these newly identified alternative proteins in the regulation of cellular functions, it is crucial to assess their dynamic modulation within a framework of altered physiological modifications such as experimental spinal cord injury (SCI). Here, we carried out a longitudinal proteomic study on rat SCI from 12 h to 10 days. Based on the alternative protein predictions, it was possible to identify a plethora of newly predicted protein hits. Among these proteins, some presented a special interest due to high homology with variable chain regions of immunoglobulins. We focus our interest on the one related to Kappa variable light chains which is similarly highly produced by B cells in the Bence jones disease, but here expressed in astrocytes. This protein, name Heimdall is an Intrinsically disordered protein which is secreted under inflammatory conditions. Immunoprecipitation experiments showed that the Heimdall interactome contained proteins related to astrocyte fate keepers such as "NOTCH1, EPHA3, IPO13" as well as membrane receptor protein including "CHRNA9; TGFBR, EPHB6, and TRAM". However, when Heimdall protein was neutralized utilizing a specific antibody or its gene knocked out by CRISPR-Cas9, sprouting elongations were observed in the corresponding astrocytes. Interestingly, depolarization assays and intracellular calcium measurements in Heimdall KO, established a depolarization effect on astrocyte membranes KO cells were more likely that the one found in neuroprogenitors. Proteomic analyses performed under injury conditions or under lipopolysaccharides (LPS) stimulation, revealed the expression of neuronal factors, stem cell proteins, proliferation, and neurogenesis of astrocyte convertor factors such as EPHA4, NOTCH2, SLIT3, SEMA3F, suggesting a role of Heimdall could regulate astrocytic fate. Taken together, Heimdall could be a novel member of the gatekeeping astrocyte-to-neuroprogenitor conversion factors.


Subject(s)
Astrocytes , Proteome , Animals , Rats , Proteome/genetics , Proteomics , Antibodies , Neurogenesis , 3' Untranslated Regions
8.
Cell Death Dis ; 14(4): 237, 2023 04 04.
Article in English | MEDLINE | ID: mdl-37015912

ABSTRACT

Using multi-omics analyses including RNAseq, RT-PCR, RACE-PCR, and shotgun proteomic with enrichment strategies, we demonstrated that newborn rat astrocytes produce neural immunoglobulin constant and variable heavy chains as well as light chains. However, their edification is different from the ones found in B cells and they resemble aberrant immunoglobulins observed in several cancers. Moreover, the complete enzymatic V(D)J recombination complex has also been identified in astrocytes. In addition, the constant heavy chain is also present in adult rat astrocytes, whereas in primary astrocytes from human fetus we identified constant and variable kappa chains as well as the substitution lambda chains known to be involved in pre-B cells. To gather insights into the function of these neural IgGs, CRISPR-Cas9 of IgG2B constant heavy chain encoding gene (Igh6), IgG2B overexpression, proximal labeling of rat astrocytes IgG2B and targets identification through 2D gels were performed. In Igh6 KO astrocytes, overrepresentation of factors involved in hematopoietic cells, neural stem cells, and the regulation of neuritogenesis have been identified. Moreover, overexpression of IgG2B in astrocytes induces the CRTC1-CREB-BDNF signaling pathway known to be involved in gliogenesis, whereas Igh6 KO triggers the BMP/YAP1/TEAD3 pathway activated in astrocytes dedifferentiation into neural progenitors. Proximal labeling experiments revealed that IgG2B is N-glycosylated by the OST complex, addressed to vesicle membranes containing the ATPase complex, and behaves partially like CD98hc through its association with LAT1. These experiments also suggest that proximal IgG2B-LAT1 interaction occurs concomitantly with MACO-1 and C2CD2L, at the heart of a potentially novel cell signaling platform. Finally, we demonstrated that these chains are synthesized individually and associated to recognize specific targets. Indeed, intermediate filaments Eif4a2 and Pdia6 involved in astrocyte fate constitute targets for these neural IgGs. Taken together, we hypothese that neural aberrant IgG chains may act as gatekeepers of astrocytes' fate.


Subject(s)
Astrocytes , Neural Stem Cells , Rats , Humans , Animals , Astrocytes/metabolism , Proteomics , Neurons/metabolism , Immunoglobulin G/genetics , Transcription Factors/metabolism
9.
Cancer Cell ; 1(2): 169-79, 2002 Mar.
Article in English | MEDLINE | ID: mdl-12086875

ABSTRACT

Antiapoptotic oncoprotein Bcl-2 has extramitochondrial actions due to its localization on the endoplasmic reticulum (ER); however, the specific mechanisms of such actions remain unclear. Here we show that Bcl-2 overexpression in LNCaP prostate cancer epithelial cells results in downregulation of store-operated Ca(2+) current by decreasing the number of functional channels and inhibiting ER Ca(2+) uptake through a reduction in the expression of calreticulin and SERCA2b, two key proteins controlling ER Ca(2+) content. Furthermore, we demonstrate that Ca(2+) store depletion by itself is not sufficient to induce apoptosis in Bcl-2 overexpressing cells, and that sustained Ca(2+) entry via activated store-operated channels (SOCs) is required as well. Our data therefore suggest the pivotal role of SOCs in apoptosis and cancer progression.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Homeostasis , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Apoptosis , Blotting, Western , Calcium-Binding Proteins/metabolism , Calcium-Transporting ATPases/metabolism , Calreticulin , Electric Conductivity , Electrophysiology , Endoplasmic Reticulum/metabolism , Humans , Male , Patch-Clamp Techniques , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Ribonucleoproteins/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Tumor Cells, Cultured
10.
Front Immunol ; 13: 882830, 2022.
Article in English | MEDLINE | ID: mdl-35784350

ABSTRACT

Spinal cord injury (SCI) represents a major medical challenge. At present, there is still no cure to treat it efficiently and enable functional recovery below the injury site. Previously, we demonstrated that inflammation determines the fate of the physiopathology. To decipher the molecular mechanisms involved in this process, we performed a meta-analysis of our spatio-temporal proteomic studies in the time course of SCI. This highlighted the presence of IgG isotypes in both spinal cord explants and their secretomes. These IgGs were detected in the spinal cord even if no SCI occurred. However, during the time course following SCI, abundance of IgG1 and IgG2 subclasses (a, b, c) varied according to the spatial repartition. IgG1 was clearly mostly abundant at 12 h, and a switch to IgG2a was observed after 24 h. This IgG stayed predominant 3, 7, and 10 days after SCI. A protein related to IgM as well as a variable heavy chain were only detected 12 h after lesion. Interestingly, treatment with RhoA inhibitor influenced the abundance of the various IgG isotypes and a preferential switch to IgG2c was observed. By data reuse of rat dorsal root ganglion (DRG) neurons RNAseq datasets and RT-PCR experiments performed on cDNA from DRG sensory neurons ND7/23 and N27 dopaminergic neural cell lines, we confirmed expression of immunoglobulin heavy and light chains (constant and variable) encoding genes in neurons. We then identified CD16 and CD32b as their specific receptors in sensory neuron cell line ND7/23 and their activation regulated neurites outgrowth. These results suggest that during SCI, neuronal IgG isotypes are released to modulate neurites outgrowth. Therefore, we propose a new view of the SCI response involving an antibody dependent neurite outgrowth modulation (ADNM) which could be a precursor to the neuroinflammatory response in pathological conditions.


Subject(s)
Proteomics , Spinal Cord Injuries , Animals , Immunoglobulin G/pharmacology , Neuronal Outgrowth , Rats , Sensory Receptor Cells/metabolism , Spinal Cord Injuries/pathology
11.
J Cell Biol ; 174(4): 535-46, 2006 Aug 14.
Article in English | MEDLINE | ID: mdl-16908669

ABSTRACT

Although human pannexins (PanX) are homologous to gap junction molecules, their physiological function in vertebrates remains poorly understood. Our results demonstrate that overexpression of PanX1 results in the formation of Ca(2+)-permeable gap junction channels between adjacent cells, thus, allowing direct intercellular Ca(2+) diffusion and facilitating intercellular Ca(2+) wave propagation. More intriguingly, our results strongly suggest that PanX1 may also form Ca(2+)-permeable channels in the endoplasmic reticulum (ER). These channels contribute to the ER Ca(2+) leak and thereby affect the ER Ca(2+) load. Because leakage remains the most enigmatic of those processes involved in intracellular calcium homeostasis, and the molecular nature of the leak channels is as yet unknown, the results of this work provide new insight into calcium signaling mechanisms. These results imply that for vertebrates, a new protein family, referred to as pannexins, may not simply duplicate the connexin function but may also provide additional pathways for intra- and intercellular calcium signaling and homeostasis.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling/physiology , Cell Membrane Permeability/physiology , Cell Membrane/metabolism , Connexins/metabolism , Gap Junctions/metabolism , Calcium/metabolism , Cell Communication/physiology , Cell Line, Tumor , Connexins/genetics , Diffusion , Endoplasmic Reticulum/metabolism , Fluorescent Dyes , Homeostasis/physiology , Humans , Intracellular Membranes/metabolism , Microscopy, Confocal , Nerve Tissue Proteins , RNA, Messenger/metabolism , Time Factors
12.
Cells ; 10(11)2021 11 04.
Article in English | MEDLINE | ID: mdl-34831241

ABSTRACT

Store-operated calcium entry (SOCE) provided through channels formed by ORAI proteins is a major regulator of several cellular processes. In immune cells, it controls fundamental processes such as proliferation, cell adhesion, and migration, while in cancer, SOCE and ORAI1 gene expression are dysregulated and lead to abnormal migration and/or cell proliferation. In the present study, we used the CRISPR/Cas9 technique to delete the ORAI1 gene and to identify its role in proliferative and migrative properties of the model cell line HEK-293. We showed that ORAI1 deletion greatly reduced SOCE. Thereby, we found that this decrease and the absence of ORAI1 protein did not affect HEK-293 proliferation. In addition, we determined that ORAI1 suppression did not affect adhesive properties but had a limited impact on HEK-293 migration. Overall, we showed that ORAI1 and SOCE are largely dispensable for cellular proliferation, migration, and cellular adhesion of HEK-293 cells. Thus, despite its importance in providing Ca2+ entry in non-excitable cells, our results indicate that the lack of SOCE does not deeply impact HEK-293 cells. This finding suggests the existence of compensatory mechanism enabling the maintenance of their physiological function.


Subject(s)
Calcium/metabolism , Cell Movement , Gene Knockout Techniques , ORAI1 Protein/deficiency , Calcium Channels/genetics , Calcium Channels/metabolism , Cell Adhesion , Cell Proliferation , Genome, Human , HEK293 Cells , Humans , ORAI1 Protein/metabolism , ORAI2 Protein/genetics , ORAI2 Protein/metabolism
13.
Biochim Biophys Acta Mol Cell Res ; 1868(7): 119023, 2021 06.
Article in English | MEDLINE | ID: mdl-33798603

ABSTRACT

Changes in cytosolic free Ca2+ concentration play a central role in many fundamental cellular processes including muscle contraction, neurotransmission, cell proliferation, differentiation, gene transcription and cell death. Many of these processes are known to be regulated by store-operated calcium channels (SOCs), among which ORAI1 is the most studied in cancer cells, leaving the role of other ORAI channels yet inadequately addressed. Here we demonstrate that ORAI3 channels are expressed in both normal (HPDE) and pancreatic ductal adenocarcinoma (PDAC) cell lines, where they form functional channels, their knockdown affecting store operated calcium entry (SOCE). More specifically, ORAI3 silencing increased SOCE in PDAC cell lines, while decreasing SOCE in normal pancreatic cell line. We also show the role of ORAI3 in proliferation, cell cycle, viability, mitotic catastrophe and cell death. Finally, we demonstrate that ORAI3 silencing impairs pancreatic tumor growth and induces cell death in vivo, suggesting that ORAI3 could represent a potential therapeutic target in PDAC treatment.


Subject(s)
Calcium Channels/metabolism , Pancreatic Neoplasms/genetics , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Apoptosis/genetics , Calcium/metabolism , Calcium Channels/genetics , Calcium Signaling/physiology , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Silencing/physiology , Humans , Mitosis/genetics , ORAI1 Protein/metabolism , Pancreatic Neoplasms/metabolism , RNA, Small Interfering/metabolism , Pancreatic Neoplasms
14.
Biochim Biophys Acta ; 1793(3): 528-39, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19321128

ABSTRACT

The physiological role, the mechanisms of activation, as well as the endogenous regulators for the non-selective cationic channel TRPV2 are not known so far. In the present work we report that endogenous lysophospholipids such as lysophosphatidylcholine (LPC) and lysophosphatidylinositol (LPI) induce a calcium influx via TRPV2 channel. This activation is dependent on the length of the side-chain and the nature of the lysophospholipid head-group. TRPV2-mediated calcium uptake stimulated by LPC and LPI occurred via Gq/Go-protein and phosphatidylinositol-3,4 kinase (PI3,4K) signalling. We have shown that the mechanism of TRPV2 activation induced by LPC and LPI is due to the TRPV2 channel translocation to the plasma membrane. The activation of TRPV2 channel by LPC and LPI leads to an increase in the cell migration of the prostate cancer cell line PC3. We have demonstrated that TRPV2 is directly involved in both steady-state and lysophospholipid-stimulated cancer cell migration. Thus, for the first time, we have identified one of the natural regulators of TRPV2 channel, one of the mechanisms of TRPV2 activation and regulation, as well as its pathophysiological role in cancer.


Subject(s)
Cell Movement/drug effects , Lysophospholipids/pharmacology , Prostatic Neoplasms/metabolism , TRPV Cation Channels/metabolism , Animals , CHO Cells , Calcium/metabolism , Calcium Channels/metabolism , Cell Line , Cricetinae , Cricetulus , GTP-Binding Proteins/metabolism , Humans , Male , Mice , Phosphatidylinositol 3-Kinases/metabolism , Transfection
16.
iScience ; 23(7): 101263, 2020 Jul 24.
Article in English | MEDLINE | ID: mdl-32585596

ABSTRACT

Mitochondria are important cell death checkpoints, and mitochondrial Ca2+ overload is considered as a potent apoptotic intrinsic pathway inducer. Here, we report that this Ca2+ apoptosis link is largely ineffective in inducing cell-death just by itself and required a concomitant inhibition of autophagy to counteract its pro-survival action. In such condition, an acute mitochondrial stress revealed by a DRP1-mediated mitochondrial dynamic remodeling is observed concomitantly with mitochondrial depolarization, release of cytochrome c, and efficient apoptosis induction. We also uncover that mitochondrial Ca2+ status modulates the function of autophagy as a sensitizer for chemotherapies. This priming mediated by mitochondrial Ca2+ overload and inhibition of autophagy sensitizes many cancer cells types to different chemotherapies with independent mechanisms of action. Collectively, our results redefine an important cell signaling pathway, uncovering new combined therapies for the treatment of diseases associated with mitochondrial Ca2+ homeostasis disorders such as cancer.

17.
Elife ; 92020 02 24.
Article in English | MEDLINE | ID: mdl-32091387

ABSTRACT

Control of cell death/survival balance is an important feature to maintain tissue homeostasis. Dependence receptors are able to induce either survival or cell death in presence or absence of their ligand, respectively. However, their precise mechanism of action and their physiological importance are still elusive for most of them including the MET receptor. We evidence that pro-apoptotic fragment generated by caspase cleavage of MET localizes to the mitochondria-associated membrane region. This fragment triggers a calcium transfer from endoplasmic reticulum to mitochondria, which is instrumental for the apoptotic action of the receptor. Knock-in mice bearing a mutation of MET caspase cleavage site highlighted that p40MET production is important for FAS-driven hepatocyte apoptosis, and demonstrate that MET acts as a dependence receptor in vivo. Our data shed light on new signaling mechanisms for dependence receptors' control of cell survival/death balance, which may offer new clues for the pathophysiology of epithelial structures.


Subject(s)
Cell Death/physiology , Cell Survival/physiology , Proto-Oncogene Proteins c-met/physiology , Animals , Cells, Cultured , Endoplasmic Reticulum/metabolism , Humans , Mice , Mice, Transgenic , Mitochondria/metabolism , Protein Transport , Proteolysis
18.
Biochem J ; 401(1): 49-55, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-16965263

ABSTRACT

Prolactin (PRL) has been shown to be involved in the differentiation and proliferation of numerous tissues, including the prostate gland. Moreover, variations in [Ca2+]ER (calcium concentration within the endoplasmic reticulum) may play a role in cell growth. However, few studies have focused on the regulation of calcium homoeostasis by prolactin. The present study evaluates the regulation of calcium pools as well as the possible role of [Ca2+]ER variations as a signal for growth modulation by PRL. We show that PRL stimulates the proliferation of normal SV40 immortalized epithelial prostate (PNT1A) cells with a maximum effect at a dose of 100 ng/ml. We also show that 100 ng/ml PRL increases the [Ca2+]ER when measured either by indirect quantification with Fura-2AM after application of 1 mM thapsigargin or by direct quantification with Mag-Fura-2AM within the endoplas-mic reticulum. Western blot analysis shows that the SERCA 2b (sarcoendoplasmic calcium ATPase 2b) is over-expressed in PNT1A cells treated with 100 ng/ml PRL for 24 h. A small inter-fering RNA SERCA 2a/b, used to down-regulate endogenous SERCA 2b expression, reduced both PNT1A cell proliferation and [Ca2+]ER. We thus identify [Ca2+]ER and SERCA 2b as protagonists in PRL-induced proliferation.


Subject(s)
Cell Division/drug effects , Endoplasmic Reticulum/enzymology , Prolactin/pharmacology , Prostate/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Calcium/metabolism , Cell Line , Gene Expression Regulation, Enzymologic/drug effects , Humans , Male , Prostate/cytology , Prostate/enzymology , RNA, Small Interfering/genetics , Transfection
19.
Sci Rep ; 7(1): 15896, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-29162859

ABSTRACT

Despite the tremendous progress in medicine, cancer remains one of the most serious global health problems awaiting new effective therapies. Here we present ferroquine (FQ), the next generation antimalarial drug, as a promising candidate for repositioning as cancer therapeutics. We report that FQ potently inhibits autophagy, perturbs lysosomal function and impairs prostate tumor growth in vivo. We demonstrate that FQ negatively regulates Akt kinase and hypoxia-inducible factor-1α (HIF-1α) and is particularly effective in starved and hypoxic conditions frequently observed in advanced solid cancers. FQ enhances the anticancer activity of several chemotherapeutics suggesting its potential application as an adjuvant to existing anticancer therapy. Alike its parent compound chloroquine (CQ), FQ accumulates within and deacidifies lysosomes. Further, FQ induces lysosomal membrane permeabilization, mitochondrial depolarization and caspase-independent cancer cell death. Overall, our work identifies ferroquine as a promising new drug with a potent anticancer activity.


Subject(s)
Aminoquinolines/pharmacology , Antimalarials/pharmacology , Antineoplastic Agents/pharmacology , Ferrous Compounds/pharmacology , Aminoquinolines/chemistry , Animals , Antimalarials/chemistry , Autophagy/drug effects , Caspases/metabolism , Cell Cycle Checkpoints/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chloroquine/chemistry , Chloroquine/pharmacology , Female , Ferrous Compounds/chemistry , Hydrogen-Ion Concentration , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Metallocenes , Mice, Nude , Neoplasms/pathology , Permeability , Stress, Physiological , Xenograft Model Antitumor Assays
20.
Oncotarget ; 7(20): 29063-80, 2016 May 17.
Article in English | MEDLINE | ID: mdl-27074561

ABSTRACT

Since its cloning a decade ago, TRPM8 channel has emerged as a promising prognostic marker and a putative therapeutic target in prostate cancer (PCa). However, recent studies have brought to light the complexity of TRPM8 isoforms in PCa. Consequently, the respective role of each TRPM8 isoform needs to be deciphered prior to considering TRPM8 as an attractive therapeutic target. Full-length (6 transmembrane (TM)-domain) TRPM8 channel is overexpressed in early PCa and repressed in advanced prostate tumors whereas the localization of the truncated, 4TM-TRPM8 channel (4 transmembrane (TM)-domain), in the membranes of endoplasmic reticulum (ER) is independent of the pathogenic status of epithelial cells. In the same line, expression of non-channel cytoplasmic small TRPM8 isoforms (namely sM8) is conserved in cancer cells. In this study, we identify sM8s as putative regulator of PCa cell death. Indeed, suppression of sM8 isoforms was found to induce concomitantly ER stress, oxidative stress, p21 expression and apoptosis in human epithelial prostate cancer cells. We furthermore demonstrate that induction of such mechanisms required the activity of 4TM-TRPM8 channels at the ER-mitochondria junction. Our study thus suggests that targeting sM8 could be an appropriate strategy to fight prostate cancer.


Subject(s)
Prostatic Neoplasms/pathology , TRPM Cation Channels/metabolism , Animals , Apoptosis/physiology , Cell Line, Tumor , Female , Heterografts , Humans , Male , Mice , Mice, Nude , Prostatic Neoplasms/metabolism , Protein Isoforms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL