Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters











Publication year range
1.
Br J Pharmacol ; 181(22): 4546-4570, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39081110

ABSTRACT

BACKGROUND AND PURPOSE: Gastrointestinal tumours overexpress voltage-gated calcium (CaV3) channels (CaV3.1, 3.2 and 3.3). CaV3 channels regulate cell growth and apoptosis colorectal cancer. Gossypol, a polyphenolic aldehyde found in the cotton plant, has anti-tumour properties and inhibits CaV3 currents. A systematic study was performed on gossypol blocking mechanism on CaV3 channels and its potential anticancer effects in colon cancer cells, which express CaV3 isoforms. EXPERIMENTAL APPROACH: Transcripts for CaV3 proteins were analysed in gastrointestinal cancers using public repositories and in human colorectal cancer cell lines HCT116, SW480 and SW620. The gossypol blocking mechanism on CaV3 channels was investigated by combining heterologous expression systems and patch-clamp experiments. The anti-tumoural properties of gossypol were estimated by cell proliferation, viability and cell cycle assays. Ca2+ dynamics were evaluated with cytosolic and endoplasmic reticulum (ER) Ca2+ indicators. KEY RESULTS: High levels of CaV3 transcripts correlate with poor prognosis in gastrointestinal cancers. Gossypol blockade of CaV3 isoforms is concentration- and use-dependent interacting with the closed, activated and inactivated conformations of CaV3 channels. Gossypol and CaV3 channels down-regulation inhibit colorectal cancer cell proliferation by arresting cell cycles at the G0/G1 and G2/M phases, respectively. CaV3 channels underlie the vectorial Ca2+ uptake by endoplasmic reticulum in colorectal cancer cells. CONCLUSION AND IMPLICATIONS: Gossypol differentially blocked CaV3 channel and its anticancer activity was correlated with high levels of CaV3.1 and CaV3.2 in colorectal cancer cells. The CaV3 regulates cell proliferation and Ca2+ dynamics in colorectal cancer cells. Understanding this blocking mechanism maybe improve cancer therapies.


Subject(s)
Calcium Channel Blockers , Calcium Channels, T-Type , Cell Proliferation , Colonic Neoplasms , Gossypol , Humans , Gossypol/pharmacology , Gossypol/analogs & derivatives , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Colonic Neoplasms/metabolism , Calcium Channel Blockers/pharmacology , Cell Proliferation/drug effects , Calcium Channels, T-Type/metabolism , Calcium Channels, T-Type/genetics , G1 Phase Cell Cycle Checkpoints/drug effects , Calcium/metabolism , Cell Line, Tumor , Resting Phase, Cell Cycle/drug effects , Antineoplastic Agents/pharmacology
2.
Sci Rep ; 13(1): 16745, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37798364

ABSTRACT

Pathophysiological response after acute myocardial infarction (AMI) is described as a three-stage model involving temporal phenotypic modifications of both immune cells and fibroblasts: a primary inflammatory phase, followed by a reparative phase and a fibrous scar maturation phase. Purinergic receptors, particularly the P2Y11 receptor, have been reported to be involved in the regulation of inflammation after ischemia and could act for the resolution of inflammation after AMI. For the first time, we characterized the immuno-inflammatory and P2Y11 expression profiles of peripheral blood mononuclear cells (PBMC) from AMI patients and analyzed the consequences of presenting these cells to cardiac fibroblasts in vitro. PBMC from 178 patients were collected at various times after reperfused ST-segment elevation AMI, from H0 to M12. Expression level of P2RY11 and genes involved in tolerogenic profile of dendritic cells and T cell polarization were evaluated by RT-PCR. P2Y11 protein expression was assessed by flow cytometry. PBMC and human cardiac fibroblasts (HCF) were cocultured and α-SMA/vimentin ratio was analyzed by flow cytometry. Within the first 48 h after AMI, expression levels of HMOX1, STAT3 and CD4 increased while IDO1 and TBX21/GATA3 ratio decreased. Concomitantly, the expression of P2RY11 increased in both T and B cells. In vitro, PBMC collected at H48 after AMI induced an increase in α-SMA/vimentin ratio in HCF. Our results suggest that human PBMC display an evolving inflammatory profile with reparative characteristics the first two days after AMI and secrete soluble mediators leading to the fibroblastic proteins modification, thus participating to myocardial fibrosis.


Subject(s)
Leukocytes, Mononuclear , Myocardial Infarction , Humans , Leukocytes, Mononuclear/metabolism , Vimentin/metabolism , Myocardial Infarction/metabolism , Inflammation/metabolism , Phenotype , Fibroblasts/metabolism
3.
Metabolomics ; 19(9): 83, 2023 09 13.
Article in English | MEDLINE | ID: mdl-37704888

ABSTRACT

INTRODUCTION: Ischemia-reperfusion injury (IRI) induces several perturbations that alter immediate kidney graft function after transplantation and may affect long-term graft outcomes. Given the IRI-dependent metabolic disturbances previously reported, we hypothesized that proximal transporters handling endo/exogenous substrates may be victims of such lesions. OBJECTIVES: This study aimed to determine the impact of hypoxia/reoxygenation on the human proximal transport system through two semi-targeted omics analyses. METHODS: Human proximal tubular cells were cultured in hypoxia (6 or 24 h), each followed by 2, 24 or 48-h reoxygenation. We investigated the transcriptomic modulation of transporters. Using semi-targeted LC-MS/MS profiling, we characterized the extra/intracellular metabolome. Statistical modelling was used to identify significant metabolic variations. RESULTS: The expression profile of transporters was impacted during hypoxia (y + LAT1 and OCTN2), reoxygenation (MRP2, PEPT1/2, rBAT, and OATP4C1), or in both conditions (P-gp and GLUT1). The P-gp and GLUT1 transcripts increased (FC (fold change) = 2.93 and 4.11, respectively) after 2-h reoxygenation preceded by 24-h hypoxia. We observed a downregulation (FC = 0.42) of y+LAT1 after 24-h hypoxia, and of PEPT2 after 24-h hypoxia followed by 2-h reoxygenation (FC = 0.40). Metabolomics showed that hypoxia altered the energetic pathways. However, intracellular metabolic homeostasis and cellular exchanges were promptly restored after reoxygenation. CONCLUSION: This study provides insight into the transcriptomic response of the tubular transporters to hypoxia/reoxygenation. No correlation was found between the expression of transporters and the metabolic variations observed. Given the complexity of studying the global tubular transport systems, we propose that further studies focus on targeted transporters.


Subject(s)
Metabolomics , Tandem Mass Spectrometry , Humans , Glucose Transporter Type 1 , Chromatography, Liquid , Metabolome , Kidney , Cell Line , Hypoxia
4.
EMBO J ; 42(13): e112198, 2023 07 03.
Article in English | MEDLINE | ID: mdl-37278161

ABSTRACT

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.


Subject(s)
Prostatic Neoplasms , Sodium , Male , Humans , Sodium/metabolism , Ion Channels/metabolism , Ion Transport , Membrane Proteins/genetics , Membrane Proteins/metabolism
5.
Cell Immunol ; 384: 104658, 2023 02.
Article in English | MEDLINE | ID: mdl-36566700

ABSTRACT

Recent evidence showed that in response to elevated sodium dietary intakes, many body tissues retain Na+ ions for long periods of time and can reach concentrations up to 200 mM. This could modulate the immune system and be responsible for several diseases. However, studies brought contrasted results and the effects of external sodium on human dendritic cell (DC) responses to danger signals remain largely unknown. Considering their central role in triggering T cell response, we tested how NaCl-enriched medium influences human DCs properties. We found that DCs submitted to high extracellular Na+ concentrations up to 200 mM remain viable and maintain the expression of specific DC markers, however, their maturation, chemotaxis toward CCL19, production of pro-inflammatory cytokines and ROS in response to LPS were also partially inhibited. In line with these results, the T-cell allostimulatory capacity of DCs was also inhibited. Finally, our data indicate that high NaCl concentrations triggered the phosphorylation of SGK1 and ERK1/2 kinases. These results raised the possibility that the previously reported pro-inflammatory effects of high NaCl concentrations on T cells might be counterbalanced by a downregulation of DC activation.


Subject(s)
Lipopolysaccharides , Sodium Chloride , Humans , Sodium Chloride/pharmacology , Sodium Chloride/metabolism , Lipopolysaccharides/pharmacology , Lipopolysaccharides/metabolism , Cell Differentiation , Chlorides/metabolism , Chlorides/pharmacology , Dendritic Cells , Cytokines/metabolism , Sodium/metabolism , Sodium/pharmacology , Cells, Cultured
6.
Oncogene ; 41(21): 2920-2931, 2022 05.
Article in English | MEDLINE | ID: mdl-35411034

ABSTRACT

Metastatic progression is a major burden for breast cancer patients and is associated with the ability of cancer cells to overcome stressful conditions, such as nutrients deprivation and hypoxia, and to gain invasive properties. Autophagy and epithelial-to-mesenchymal transition are critical contributors to these processes. Here, we show that the P2X4 purinergic receptor is upregulated in breast cancer biopsies from patients and it is primarily localised in endolysosomes. We demonstrate that P2X4 enhanced invasion in vitro, as well as mammary tumour growth and metastasis in vivo. The pro-malignant role of P2X4 was mediated by the regulation of lysosome acidity, the promotion of autophagy and cell survival. Furthermore, the autophagic activity was associated with epithelial-to-mesenchymal transition (EMT), and this role of P2X4 was even more pronounced under metabolic challenges. Pharmacological and gene silencing of P2X4 inhibited both autophagy and EMT, whereas its rescue in knocked-down cells led to the restoration of the aggressive phenotype. Together, our results demonstrate a previously unappreciated role for P2X4 in regulating lysosomal functions and fate, promoting breast cancer progression and aggressiveness.


Subject(s)
Breast Neoplasms , Receptors, Purinergic P2X4 , Autophagy/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Receptors, Purinergic P2X4/genetics , Receptors, Purinergic P2X4/metabolism
7.
Cancers (Basel) ; 15(1)2022 Dec 22.
Article in English | MEDLINE | ID: mdl-36612049

ABSTRACT

Colorectal cancer (CRC) is the second leading cause of death worldwide, with 0.9 million deaths per year. The metastatic stage of the disease is identified in about 20% of cases at the first diagnosis and is associated with low patient-survival rates. Voltage-gated sodium channels (NaV) are abnormally overexpressed in several carcinomas including CRC and are strongly associated with the metastatic behavior of cancer cells. Acidification of the extracellular space by Na+/H+ exchangers (NHE) contributes to extracellular matrix degradation and cell invasiveness. In this study, we assessed the expression levels of pore-forming α-subunits of NaV channels and NHE exchangers in tumor and adjacent non-malignant tissues from colorectal cancer patients, CRC cell lines and primary tumor cells. In all cases, SCN5A (gene encoding for NaV1.5) was overexpressed and positively correlated with cancer stage and poor survival prognosis for patients. In addition, we identified an anatomical differential expression of SCN5A and SLC9A1 (gene encoding for NHE-1) being particularly relevant for tumors that originated on the sigmoid colon epithelium. The functional activity of NaV1.5 channels was characterized in CRC cell lines and the primary cells of colon tumors obtained using tumor explant methodologies. Furthermore, we assessed the performance of two new small-molecule NaV1.5 inhibitors on the reduction of sodium currents, as well as showed that silencing SCN5A and SLC9A1 substantially reduced the 2D invasive capabilities of cancer cells. Thus, our findings show that both NaV1.5 and NHE-1 represent two promising targetable membrane proteins against the metastatic progression of CRC.

8.
Cells ; 10(7)2021 06 29.
Article in English | MEDLINE | ID: mdl-34209614

ABSTRACT

The SCN4B gene, coding for the NaVß4 subunit of voltage-gated sodium channels, was recently found to be expressed in normal epithelial cells and down-regulated in several cancers. However, its function in normal epithelial cells has not been characterized. In this study, we demonstrated that reducing NaVß4 expression in MCF10A non-cancer mammary epithelial cells generated important morphological changes observed both in two-dimensional cultures and in three-dimensional cysts. Most notably, the loss of NaVß4 induced a complete loss of epithelial organisation in cysts and increased proteolytic activity towards the extracellular matrix. Loss of epithelial morphology was associated with an increased degradation of ß-catenin, reduced E-cadherin expression and induction of mesenchymal markers N-cadherin, vimentin, and α-SMA expression. Overall, our results suggest that Navß4 may participate in the maintenance of the epithelial phenotype in mammary cells and that its downregulation might be a determining step in early carcinogenesis.


Subject(s)
Epithelial Cells/metabolism , Mammary Glands, Animal/cytology , Protein Subunits/metabolism , Voltage-Gated Sodium Channel beta-4 Subunit/metabolism , Animals , Cell Line , Cell Polarity , Down-Regulation , Epithelial Cells/cytology , Female , Humans , Mesoderm/metabolism , Phenotype , Proteolysis , beta Catenin/metabolism
9.
Eur J Med Chem ; 218: 113258, 2021 Jun 05.
Article in English | MEDLINE | ID: mdl-33813152

ABSTRACT

Herein, we report the design, synthesis and evaluation of novel bioinspired imidazo[1,2-a:4,5c']dipyridines. The structural optimization identified four anti-proliferative compounds. Compounds 11, 18, 19 and 20 exhibited excellent anticancer activities in vitro with IC50 of 0.4-5 µM against three human cancer cell lines (MDA-MB-468, MDA-MB-435s and MDA-MB-231). These four compounds induced apoptosis in MDA-MB-231 cells in a dose-dependent manner, targeting different apoptotic proteins expression: 11 increased the expression of pro-apoptotic Bax protein while 18-20 reduced the level of anti-apoptotic Bcl-2 protein. Compounds 18 and 19 also reduced MDA-MB-231 cells proliferation as measured by Ki-67 staining. Furthermore, compounds were also tested for the ability to inhibit cell migration in the highly aggressive human MDA-MB-435s cell line. Six compounds of this series (8, 15, 18, 22, 23, 24) inhibited cell migration by 41-50% while four compounds (20, 25, 27, 30) inhibited the migration by 53-62% in wound-healing experiments. Interestingly, compound 20 presented both antiproliferative and anti-migration activities and might be a promising anti-metastatic agent for cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Structure-Activity Relationship , Tumor Cells, Cultured
10.
Int J Mol Sci ; 22(2)2021 Jan 16.
Article in English | MEDLINE | ID: mdl-33467058

ABSTRACT

Vascular dysfunction in cardiovascular diseases includes vasomotor response impairments, endothelial cells (ECs) activation, and smooth muscle cells (SMCs) proliferation and migration to the intima. This results in intimal hyperplasia and vessel failure. We previously reported that activation of the P2Y11 receptor (P2Y11R) in human dendritic cells, cardiofibroblasts and cardiomyocytes was protective against hypoxia/reoxygenation (HR) lesions. In this study, we investigated the role of P2Y11R signaling in vascular dysfunction. P2Y11R activity was modulated using its pharmacological agonist NF546 and antagonist NF340. Rat aortic rings were exposed to angiotensin II (AngII) and evaluated for their vasomotor response. The P2Y11R agonist NF546 reduced AngII-induced vascular dysfunction by promoting EC-dependent vasorelaxation, through an increased nitric oxide (NO) bioavailability and reduced AngII-induced H2O2 release; these effects were prevented by the use of the P2Y11R antagonist NF340. Human vascular SMCs and ECs were subjected to AngII or H/R simulation in vitro. P2Y11R agonist modulated vasoactive factors in human ECs, that is, endothelial nitric oxide synthase (eNOS) and endothelin-1, reduced SMC proliferation and prevented the switch towards a synthetic phenotype. H/R and AngII increased ECs secretome-induced SMC proliferation, an effect prevented by P2Y11R activation. Thus, our data suggest that P2Y11R activation may protect blood vessels from HR-/AngII-induced injury and reduce vascular dysfunctions. These results open the way for new vasculoprotective interventions.


Subject(s)
Diphosphonates/pharmacology , Naphthalenesulfonates/pharmacology , Purinergic P2 Receptor Agonists/pharmacology , Receptors, Purinergic P2/metabolism , Reperfusion Injury/metabolism , Tunica Intima/pathology , Angiotensin II/toxicity , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Aorta/physiopathology , Diphosphonates/therapeutic use , Endothelin-1/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hyperplasia/prevention & control , Male , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Naphthalenesulfonates/therapeutic use , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Purinergic P2 Receptor Agonists/therapeutic use , Rats , Rats, Wistar , Reperfusion Injury/drug therapy , Tunica Intima/drug effects , Tunica Intima/metabolism , Vasodilation , Water/metabolism
11.
Sci Rep ; 10(1): 13597, 2020 08 12.
Article in English | MEDLINE | ID: mdl-32788683

ABSTRACT

Infarct size is a major prognostic factor in ST-segment elevation myocardial infarction (STEMI). It is often assessed using repeated blood sampling and the estimation of biomarker area under the concentration versus time curve (AUC) in translational research. We aimed at developing limited sampling strategies (LSS) to accurately estimate biomarker AUC using only a limited number of blood samples in STEMI patients. This retrospective study was carried out on pooled data from five clinical trials of STEMI patients (TIMI blood flow 0/1) studies where repeated blood samples were collected within 72 h after admission to assess creatine kinase (CK), cardiac troponin I (cTnI) and muscle-brain CK (CK-MB). Biomarker kinetics was assessed using previously described biomarker kinetic models. A number of LSS models including combinations of 1 to 3 samples were developed to identify sampling times leading to the best estimation of AUC. Patients were randomly assigned to either learning (2/3) or validation (1/3) subsets. Descriptive and predictive performances of LSS models were compared using learning and validation subsets, respectively. An external validation cohort was used to validate the model and its applicability to different cTnI assays, including high-sensitive (hs) cTnI. 132 patients had full CK and cTnI dataset, 49 patients had CK-MB. For each biomarker, 180 LSS models were tested. Best LSS models were obtained for the following sampling times: T4-16 for CK, T8-T20 for cTnI and T8-T16 for CK-MB for 2-sample LSS; and T4-T16-T24 for CK, T4-T12-T20 for cTnI and T8-T16-T20 for CK-MB for 3-sample LSS. External validation was achieved on 103 anterior STEMI patients (TIMI flow 0/1), and the cTnI model applicability to recommended hs cTnI confirmed. Biomarker kinetics can be assessed with a limited number of samples using kinetic modelling. This opens the way for substantial simplification of future cardioprotection studies, more acceptable for the patients.


Subject(s)
Biomarkers/metabolism , Creatine Kinase/metabolism , Myocardium/pathology , ST Elevation Myocardial Infarction/metabolism , Troponin I/metabolism , Aged , Area Under Curve , Clinical Trials as Topic , Female , Humans , Kinetics , Male , Middle Aged , Myocardium/metabolism , Necrosis , Retrospective Studies , ST Elevation Myocardial Infarction/pathology
12.
Sci Rep ; 10(1): 13350, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32770034

ABSTRACT

The acquisition of invasive capacities by carcinoma cells, i.e. their ability to migrate through and to remodel extracellular matrices, is a determinant process leading to their dissemination and to the development of metastases. these cancer cell properties have often been associated with an increased Rho-ROCK signalling, and ROCK inhibitors have been proposed for anticancer therapies. In this study we used the selective ROCK inhibitor, Y-27632, to address the participation of the Rho-ROCK signalling pathway in the invasive properties of SW620 human colon cancer cells. Contrarily to initial assumptions, Y-27632 induced the acquisition of a pro-migratory cell phenotype and increased cancer cell invasiveness in both 3- and 2-dimensions assays. This effect was also obtained using the other ROCK inhibitor Fasudil as well as with knocking down the expression of ROCK-1 or ROCK-2, but was prevented by the inhibition of NaV1.5 voltage-gated sodium channel activity. Indeed, ROCK inhibition enhanced the activity of the pro-invasive NaV1.5 channel through a pathway that was independent of gene expression regulation. In conclusions, our evidence identifies voltage-gated sodium channels as new targets of the ROCK signalling pathway, as well as responsible for possible deleterious effects of the use of ROCK inhibitors in the treatment of cancers.


Subject(s)
Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Neoplasm Invasiveness/pathology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Amides/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Colonic Neoplasms/drug therapy , Humans , Protein Serine-Threonine Kinases/metabolism , Pyridines/pharmacology , Signal Transduction/drug effects
13.
Cancers (Basel) ; 12(9)2020 Aug 19.
Article in English | MEDLINE | ID: mdl-32825056

ABSTRACT

The P2X7 receptor is an ATP-gated cation channel with a still ambiguous role in cancer progression, proposed to be either pro- or anti-cancerous, depending on the cancer or cell type in the tumour. Its role in mammary cancer progression is not yet defined. Here, we show that P2X7 receptor is functional in highly aggressive mammary cancer cells, and induces a change in cell morphology with fast F-actin reorganization and formation of filopodia, and promotes cancer cell invasiveness through both 2- and 3-dimensional extracellular matrices in vitro. Furthermore, P2X7 receptor sustains Cdc42 activity and the acquisition of a mesenchymal phenotype. In an immunocompetent mouse mammary cancer model, we reveal that the expression of P2X7 receptor in cancer cells, but not in the host mice, promotes tumour growth and metastasis development, which were reduced by treatment with specific P2X7 antagonists. Our results demonstrate that P2X7 receptor drives mammary tumour progression and represents a pertinent target for mammary cancer treatment.

14.
Eur J Pharmacol ; 876: 173060, 2020 Jun 05.
Article in English | MEDLINE | ID: mdl-32142768

ABSTRACT

Chronic inflammation is the hallmark of cardiovascular pathologies with a major role in both disease progression and occurrence of long-term complications. The massive release of ATP during the inflammatory process activates various purinergic receptors, including P2Y11. This receptor is less studied but ubiquitously expressed in all cells relevant for cardiovascular pathology: cardiomyocytes, fibroblasts, endothelial and immune cells. While several studies suggested a potential pro-inflammatory role for P2Y11 receptors, recent literature data are supportive of an anti-inflammatory profile characterized by the immunosuppression of dendritic cells, inhibition of fibroblast proliferation and of cytokines and ATP secretion. Moreover, modulation of its activity appears to mediate the positive inotropic effect of ATP and mitigate endothelial dysfunction, thus rendering this receptor a promising therapeutic target in the cardiovascular disease armamentarium. The aim of the present review is to summarize the current available knowledge on P2Y11-related purinergic signaling in the setting of inflammation and cardio-metabolic diseases.


Subject(s)
Cardiovascular Diseases/metabolism , Metabolic Diseases/metabolism , Receptors, Purinergic P2/metabolism , Animals , Cardiovascular Diseases/etiology , Cardiovascular Diseases/immunology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Fibroblasts/immunology , Fibroblasts/metabolism , Humans , Inflammation , Metabolic Diseases/complications , Metabolic Diseases/immunology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/metabolism , Receptors, Purinergic P2/genetics , Signal Transduction
15.
Sci Rep ; 9(1): 18652, 2019 12 09.
Article in English | MEDLINE | ID: mdl-31819138

ABSTRACT

Loss of epithelial polarity and gain in invasiveness by carcinoma cells are critical events in the aggressive progression of cancers and depend on phenotypic transition programs such as the epithelial-to-mesenchymal transition (EMT). Many studies have reported the aberrant expression of voltage-gated sodium channels (NaV) in carcinomas and specifically the NaV1.5 isoform, encoded by the SCN5A gene, in breast cancer. NaV1.5 activity, through an entry of sodium ions, in breast cancer cells is associated with increased invasiveness, but its participation to the EMT has to be clarified. In this study, we show that reducing the expression of NaV1.5 in highly aggressive human MDA-MB-231 breast cancer cells reverted the mesenchymal phenotype, reduced cancer cell invasiveness and the expression of the EMT-promoting transcription factor SNAI1. The heterologous expression of NaV1.5 in weakly invasive MCF-7 breast cancer cells induced their expression of both SNAI1 and ZEB1 and increased their invasive capacities. In MCF-7 cells the stimulation with the EMT-activator signal TGF-ß1 increased the expression of SCN5A. Moreover, the reduction of the salt-inducible kinase 1 (SIK1) expression promoted NaV1.5-dependent invasiveness and expression of EMT-associated transcription factor SNAI1. Altogether, these results indicated a prominent role of SIK1 in regulating NaV1.5-dependent EMT and invasiveness.


Subject(s)
Breast Neoplasms/genetics , NAV1.5 Voltage-Gated Sodium Channel/genetics , Protein Serine-Threonine Kinases/genetics , Transforming Growth Factor beta1/genetics , Breast Neoplasms/pathology , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Snail Family Transcription Factors/genetics , Zinc Finger E-box-Binding Homeobox 1/genetics
16.
Sci Rep ; 9(1): 11613, 2019 08 12.
Article in English | MEDLINE | ID: mdl-31406184

ABSTRACT

Sterile inflammation is a key determinant of myocardial reperfusion injuries. It participates in infarct size determination in acute myocardial infarction and graft rejection following heart transplantation. We previously showed that P2Y11 exerted an immunosuppressive role in human dendritic cells, modulated cardiofibroblasts' response to ischemia/reperfusion in vitro and delayed graft rejection in an allogeneic heterotopic heart transplantation model. We sought to investigate a possible role of P2Y11 in the cellular response of cardiomyocytes to ischemia/reperfusion. We subjected human AC16 cardiomyocytes to 5 h hypoxia/1 h reoxygenation (H/R). P2Y11R (P2Y11 receptor) selective agonist NF546 and/or antagonist NF340 were added at the onset of reoxygenation. Cellular damages were assessed by LDH release, MTT assay and intracellular ATP level; intracellular signaling pathways were explored. The role of P2Y11R in mitochondria-derived ROS production and mitochondrial respiration was investigated. In vitro H/R injuries were significantly reduced by P2Y11R stimulation at reoxygenation. This protection was suppressed with P2Y11R antagonism. P2Y11R stimulation following H2O2-induced oxidative stress reduced mitochondria-derived ROS production and damages through PKCε signaling pathway activation. Our results suggest a novel protective role of P2Y11 in cardiomyocytes against reperfusion injuries. Pharmacological post-conditioning targeting P2Y11R could therefore contribute to improve myocardial ischemia/reperfusion outcomes in acute myocardial infarction and cardiac transplantation.


Subject(s)
Myocytes, Cardiac/drug effects , Protein Kinase C-epsilon/metabolism , Receptors, Purinergic P2/drug effects , Reperfusion Injury/prevention & control , Signal Transduction , Adenosine Triphosphate/administration & dosage , Cardiotonic Agents/pharmacology , Heart Transplantation , Humans , Myocardial Infarction/prevention & control , Myocytes, Cardiac/enzymology , Oxygen/metabolism , Purinergic P2 Receptor Agonists/pharmacology
17.
J Thorac Cardiovasc Surg ; 158(3): 780-790.e1, 2019 09.
Article in English | MEDLINE | ID: mdl-30711276

ABSTRACT

OBJECTIVE: Myocardial ischemia reperfusion is a major cause of cell injury during cardiac transplantation and is responsible for increased graft rejection. Several in vitro studies demonstrated the protective effect of P2Y11-like purinoreceptor stimulation in the context of myocardial ischemia/reperfusion. In this study, we hypothesized a possible cardioprotective role of P2Y11R stimulation against ischemia/reperfusion lesions and validated its clinical effect in vivo in a heart transplantation model. METHODS: We subjected H9c2 rat cardiomyocyte-derived cell line to 5 hours of hypoxia and 1 hour of reoxygenation. P2Y11R selective agonist NF546 and antagonist NF340 were added at the onset of reoxygenation. Cell injuries were assessed by microculture tetrazolium reduction and intracellular adenosine triphosphate level. Clinical effect of P2Y11R stimulation was further investigated in vivo. Hearts from BALB/c mice were transplanted intra-abdominally into allogenic C57BL/6 mice (n = 104). Recipient mice were injected with P2Y11R agonist. Mice in the sham group were injected with saline solution. In the control group, hearts from C57BL/6 were transplanted into syngeneic C57BL/6 mice. Rejection lesions were investigated using histology and immunohistochemistry at days 3, 5, and 7 after transplantation. We measured caspase activities to quantify apoptosis. Production of proinflammatory and anti-inflammatory cytokines was investigated. RESULTS: P2Y11R stimulation at the onset of reoxygenation significantly reduced in vitro hypoxia/reoxygenation injuries. This protection was suppressed with P2Y11R antagonist. In vivo, cardiac allograft survival was significantly prolonged after P2Y11R stimulation. Rejection lesions, classified according to the International Society of Heart Lung Transplantation guidelines and quantified using the mean number of inflammatory cells per field, were significantly reduced in the treated group. At day 5 after transplantation, P2Y11R agonist pretreated allografts also demonstrated less apoptotic lesions. CONCLUSIONS: Our data suggest a novel cardioprotective role of P2Y11R at the onset of reoxygenation/reperfusion against reperfusion injuries. Pharmacologic conditioning using P2Y11 agonist may be beneficial after cardiac transplantation in improving myocardial ischemia/reperfusion outcomes and decreasing graft rejection lesions.


Subject(s)
Diphosphonates/pharmacology , Graft Rejection/prevention & control , Heart Transplantation/adverse effects , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Naphthalenesulfonates/pharmacology , Purinergic P2 Receptor Agonists/pharmacology , Receptors, Purinergic P2/drug effects , Animals , Apoptosis/drug effects , Caspases/metabolism , Cell Line , Cytokines/metabolism , Disease Models, Animal , Female , Graft Rejection/metabolism , Graft Rejection/pathology , Graft Survival/drug effects , Mice, Inbred BALB C , Mice, Inbred C57BL , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats , Receptors, Purinergic P2/metabolism , Signal Transduction/drug effects , Time Factors
18.
J Mol Cell Cardiol ; 121: 212-222, 2018 08.
Article in English | MEDLINE | ID: mdl-30031814

ABSTRACT

Cardiac fibroblasts are important regulators of myocardial structure and function. Their implications in pathological processes such as Ischemia/Reperfusion are well characterized. Cardiac fibroblasts respond to stress by excessive proliferation and secretion of pro-inflammatory cytokines and other factors, e.g. ATP, leading to purinergic receptors activation. P2Y11 receptor (P2Y11R) is an ATP-sensitive GPCR playing an immunomodulatory role in human dendritic cells (DC). We hypothesized that P2Y11R stimulation modulated the pro-inflammatory responses of human cardiac fibroblasts (HCF) to Hypoxia/Reoxygenation (H/R) mainly by acting on their secretome. P2Y11R stimulation in HCF at the onset of reoxygenation significantly limited H/R-induced proliferation (-19%) and pro-inflammatory cytokines and ATP secretion (-44% and -83% respectively). Exposure of DC to HCF secretome increased their expression of CD83, CD25 and CD86, suggesting a switch from immature to mature phenotype. Under LPS stimulation, DC had a pro-inflammatory profile (high IL-12/IL-10 ratio) that was decreased by HCF secretome (-3,8-fold), indicating induction of a tolerogenic profile. Moreover, P2Y11R inhibition in HCF led to high IL-12 secretion in DC, suggesting that the immunomodulatory effect of HCF secretome is P2Y11R-dependant. HCF secretome reduced H/R-induced cardiomyocytes death (-23%) through RISK pathway activation. P2Y11R inhibition in HCF induced a complete loss of HCF secretome protective effect, highlighting the cardioprotective role of P2Y11R. Our data demonstrated paracrine interactions between HCF, cardiomyocytes and DC following H/R, suggesting a key role of HCF in the cellular responses to reperfusion. These results also demonstrated a beneficial role of P2Y11R in HCF during H/R and strongly support the hypothesis that P2Y11R is a modulator of I/R injury.


Subject(s)
Myocardial Reperfusion Injury/genetics , Myocardium/metabolism , Receptors, Purinergic P2/genetics , Reperfusion Injury/genetics , Cell Proliferation/genetics , Cell Survival/genetics , Dendritic Cells/metabolism , Dendritic Cells/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Hypoxia/genetics , Hypoxia/pathology , Immunologic Factors/metabolism , Interleukin-12/genetics , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Paracrine Communication/genetics , Receptors, Purinergic P2/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
19.
Cell Calcium ; 59(4): 198-207, 2016 04.
Article in English | MEDLINE | ID: mdl-27020659

ABSTRACT

Migration capacity is essential for dendritic cells (DCs) to present antigen to T cells for the induction of immune response. The DC migration is supposed to be a calcium-dependent process, while not fully understood. Here, we report a role of the KCa3.1/IK1/SK4 channels in the migration capacity of both immature (iDC) and mature (mDC) human CD14(+)-derived DCs. KCa3.1 channels were shown to control the membrane potential of human DC and the Ca(2+) entry, which is directly related to migration capacities. The expression of migration marker such as CCR5 and CCR7 was modified in both types of DCs by TRAM-34 (100nM). But, only the migration of iDC was decreased by use of both TRAM-34 and KCa3.1 siRNA. Confocal analyses showed a close localization of CCR5 with KCa3.1 in the steady state of iDC. Finally, the implication of KCa3.1 seems to be limited to the migration capacities as T cell activation of DCs appeared unchanged. Altogether, these results demonstrated that KCa3.1 channels have a pro-migratory effect on iDC migration. Our findings suggest that KCa3.1 in human iDC play a major role in their migration and constitute an attractive target for the cell therapy optimization.


Subject(s)
Calcium/metabolism , Cell Movement , Dendritic Cells/cytology , Dendritic Cells/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Cell Movement/drug effects , Dendritic Cells/drug effects , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Pyrazoles/pharmacology
20.
J Immunol ; 195(2): 651-60, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26078273

ABSTRACT

High concentrations of extracellular ATP (eATP) resulting from cell damage may be found during an ischemia/reperfusion (I/R) episode at the site of injury. eATP activates purinergic receptors in dendritic cells (DCs) and may inhibit inflammation. This immunosuppressive activity could be of interest in the field of I/R, which is an inflammatory condition involved in myocardial infarction, stroke, and solid organ transplantation. However, the specific purinergic receptor responsible for this effect remains to be identified. In this study, we report that eATP induced maturation of human monocyte-derived DCs. Additionally, eATP inhibited IL-12 production whereas IL-10 levels remained unchanged in activated DCs. These effects were prevented by the P2Y11R antagonist NF340. Interestingly, a 5-h hypoxia prevented the effects of eATP on cytokine production whereas a 1-h hypoxia did not affect the eATP-mediated decrease of IL-12 and IL-6. We showed a time-dependent downregulation of P2Y11R at both mRNA and protein levels that was prevented by knocking down hypoxia-inducible factor-1α. In this study, we showed an immunosuppressive role of P2Y11R in human DCs. Additionally, we demonstrated that the time-dependent downregulation of P2Y11R by hypoxia orientates DCs toward a proinflammatory phenotype that may be involved in post-I/R injuries as observed after organ transplantation.


Subject(s)
Dendritic Cells/immunology , Oxygen/pharmacology , Receptors, Purinergic P2/immunology , Adenosine Triphosphate/pharmacology , Cell Hypoxia , Dendritic Cells/cytology , Dendritic Cells/drug effects , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Immunosuppression Therapy , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-12/antagonists & inhibitors , Interleukin-12/genetics , Interleukin-12/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Primary Cell Culture , Purinergic Antagonists/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Receptors, Purinergic P2/genetics , Signal Transduction , Time Factors , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL