Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Biochem Soc Trans ; 48(1): 327-336, 2020 02 28.
Article in English | MEDLINE | ID: mdl-31922198

ABSTRACT

The respiratory tract is lined by a pseudo-stratified epithelium from the nose to terminal bronchioles. This first line of defense of the lung against external stress includes five main cell types: basal, suprabasal, club, goblet and multiciliated cells, as well as rare cells such as ionocytes, neuroendocrine and tuft/brush cells. At homeostasis, this epithelium self-renews at low rate but is able of fast regeneration upon damage. Airway epithelial cell lineages during regeneration have been investigated in the mouse by genetic labeling, mainly after injuring the epithelium with noxious agents. From these approaches, basal cells have been identified as progenitors of club, goblet and multiciliated cells, but also of ionocytes and neuroendocrine cells. Single-cell RNA sequencing, coupled to lineage inference algorithms, has independently allowed the establishment of comprehensive pictures of cell lineage relationships in both mouse and human. In line with genetic tracing experiments in mouse trachea, studies using single-cell RNA sequencing (RNAseq) have shown that basal cells first differentiate into club cells, which in turn mature into goblet cells or differentiate into multiciliated cells. In the human airway epithelium, single-cell RNAseq has identified novel intermediate populations such as deuterosomal cells, 'hybrid' mucous-multiciliated cells and progenitors of rare cells. Novel differentiation dynamics, such as a transition from goblet to multiciliated cells have also been discovered. The future of cell lineage relationships in the respiratory tract now resides in the combination of genetic labeling approaches with single-cell RNAseq to establish, in a definitive manner, the hallmarks of cellular lineages in normal and pathological situations.


Subject(s)
Cell Lineage/genetics , RNA-Seq , Single-Cell Analysis/methods , Trachea/cytology , Trachea/physiology , Animals , Cell Differentiation/genetics , Epithelial Cells/metabolism , Homeostasis , Humans , Mice , Regeneration
2.
Leukemia ; 31(3): 555-564, 2017 03.
Article in English | MEDLINE | ID: mdl-27686867

ABSTRACT

Recent advances in genomic technologies have revolutionized acute myeloid leukemia (AML) understanding by identifying potential novel actionable genomic alterations. Consequently, current risk stratification at diagnosis not only relies on cytogenetics, but also on the inclusion of several of these abnormalities. Despite this progress, AML remains a heterogeneous and complex malignancy with variable response to current therapy. Although copy-number alterations (CNAs) are accepted prognostic markers in cancers, large-scale genomic studies aiming at identifying specific prognostic CNA-based markers in AML are still lacking. Using 367 AML, we identified four recurrent CNA on chromosomes 11 and 21 that predicted outcome even after adjusting for standard prognostic risk factors and potentially delineated two new subclasses of AML with poor prognosis. ERG amplification, the most frequent CNA, was related to cytarabine resistance, a cornerstone drug of AML therapy. These findings were further validated in The Cancer Genome Atlas data. Our results demonstrate that specific CNA are of independent prognostic relevance, and provide new molecular information into the genomic basis of AML and cytarabine response. Finally, these CNA identified two potential novel risk groups of AML, which when confirmed prospectively, may improve the clinical risk stratification and potentially the AML outcome.


Subject(s)
Biomarkers, Tumor , DNA Copy Number Variations , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cohort Studies , Drug Resistance, Neoplasm , Female , Gene Dosage , Genes, p53 , Genetic Association Studies , Genetic Predisposition to Disease , Genome-Wide Association Study , Genomics/methods , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/therapy , Male , Middle Aged , Mutation , Polymorphism, Single Nucleotide , Prognosis , Proportional Hazards Models , Treatment Outcome
3.
Cell Death Dis ; 4: e544, 2013 Mar 14.
Article in English | MEDLINE | ID: mdl-23492775

ABSTRACT

The resistance of hypoxic cells to radiotherapy and chemotherapy is a major problem in the treatment of cancer. Recently, an additional mode of hypoxia-inducible factor (HIF)-dependent transcriptional regulation, involving modulation of a specific set of micro RNAs (miRNAs), including miR-210, has emerged. We have recently shown that HIF-1 induction of miR-210 also stabilizes HIF-1 through a positive regulatory loop. Therefore, we hypothesized that by stabilizing HIF-1 in normoxia, miR-210 may protect cancer cells from radiation. We developed a non-small cell lung carcinoma (NSCLC)-derived cell line (A549) stably expressing miR-210 (pmiR-210) or a control miRNA (pmiR-Ctl). The miR-210-expressing cells showed a significant stabilization of HIF-1 associated with mitochondrial defects and a glycolytic phenotype. Cells were subjected to radiation levels ranging from 0 to 10 Gy in normoxia and hypoxia. Cells expressing miR-210 in normoxia had the same level of radioresistance as control cells in hypoxia. Under hypoxia, pmiR-210 cells showed a low mortality rate owing to a decrease in apoptosis, with an ability to grow even at 10 Gy. This miR-210 phenotype was reproduced in another NSCLC cell line (H1975) and in HeLa cells. We have established that radioresistance was independent of p53 and cell cycle status. In addition, we have shown that genomic double-strand breaks (DSBs) foci disappear faster in pmiR-210 than in pmiR-Ctl cells, suggesting that miR-210 expression promotes a more efficient DSB repair. Finally, HIF-1 invalidation in pmiR-210 cells removed the radioresistant phenotype, showing that this mechanism is dependent on HIF-1. In conclusion, miR-210 appears to be a component of the radioresistance of hypoxic cancer cells. Given the high stability of most miRNAs, this advantage could be used by tumor cells in conditions where reoxygenation has occurred and suggests that strategies targeting miR-210 could enhance tumor radiosensitization.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Gene Expression Regulation, Neoplastic/radiation effects , Hypoxia-Inducible Factor 1/genetics , Hypoxia/genetics , Lung Neoplasms/genetics , MicroRNAs/genetics , Apoptosis/radiation effects , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Hypoxia/genetics , Cell Hypoxia/radiation effects , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA Repair , Gamma Rays , Humans , Hypoxia/metabolism , Hypoxia/pathology , Hypoxia-Inducible Factor 1/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , MicroRNAs/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/radiation effects , Radiation Tolerance , Signal Transduction/radiation effects , Transcription, Genetic/radiation effects
4.
Cell Death Differ ; 20(6): 800-11, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23429262

ABSTRACT

Disruption of contact inhibition and serum afflux that occur after a tissue injury activate cell cycle, which then stops when confluence is reached again. Although the events involved in cell cycle entry have been widely documented, those managing cell cycle exit have remained so far ill defined. We have identified that the final stage of wound closure is preceded in keratinocytes by a strong accumulation of miR-483-3p, which acts as a mandatory signal triggering cell cycle arrest when confluence is reached. Blocking miR-483-3p accumulation strongly delays cell cycle exit, maintains cells into a proliferative state and retards their differentiation program. Using two models of cell cycle synchronization (i.e. mechanical injury and serum addition), we show that an ectopic upregulation of miR-483-3p blocks cell cycle progression in early G1 phase. This arrest results from a direct targeting of the CDC25A phosphatase by miR-483-3p, which can be impeded using an anti-miRNA against miR-483-3p or a protector that blocks the complex formation between miR-483-3p and the 3'-untranslated region (UTR) of CDC25A transcript. We show that the miRNA-induced silencing of CDC25A increases the tyrosine phosphorylation status of CDK4/6 cyclin-dependent kinases which, in turn, abolishes CDK4/6 capacity to associate with D-type cyclins. This prevents CDK4/6 kinases' activation, impairs downstream events such as cyclin E stimulation and sequesters cells in early G1. We propose this new regulatory process of cyclin-CDK association as a general mechanism coupling miRNA-mediated CDC25A invalidation to CDK post-transcriptional modifications and cell cycle control.


Subject(s)
Cell Cycle Checkpoints/physiology , Cyclin E/metabolism , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , MicroRNAs/administration & dosage , Oncogene Proteins/metabolism , cdc25 Phosphatases/antagonists & inhibitors , cdc25 Phosphatases/metabolism , 3T3 Cells , Animals , Cell Cycle Checkpoints/genetics , Cell Growth Processes/physiology , Cyclin E/genetics , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 6/genetics , HeLa Cells , Humans , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Oncogene Proteins/genetics , Transfection , cdc25 Phosphatases/genetics
5.
Cell Death Differ ; 18(3): 465-78, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20885442

ABSTRACT

Following the identification of a set of hypoxia-regulated microRNAs (miRNAs), recent studies have highlighted the importance of miR-210 and of its transcriptional regulation by the transcription factor hypoxia-inducible factor-1 (HIF-1). We report here that miR-210 is overexpressed at late stages of non-small cell lung cancer. Expression of miR-210 in lung adenocarcinoma A549 cells caused an alteration of cell viability associated with induction of caspase-3/7 activity. miR-210 induced a loss of mitochondrial membrane potential and the apparition of an aberrant mitochondrial phenotype. The expression profiling of cells overexpressing miR-210 revealed a specific signature characterized by enrichment for transcripts related to 'cell death' and 'mitochondrial dysfunction', including several subunits of the electron transport chain (ETC) complexes I and II. The transcript coding for one of these ETC components, SDHD, subunit D of succinate dehydrogenase complex (SDH), was validated as a bona fide miR-210 target. Moreover, SDHD knockdown mimicked miR-210-mediated mitochondrial alterations. Finally, miR-210-dependent targeting of SDHD was able to activate HIF-1, in line with previous studies linking loss-of-function SDH mutations to HIF-1 activation. miR-210 can thus regulate mitochondrial function by targeting key ETC component genes with important consequences on cell metabolism, survival and modulation of HIF-1 activity. These observations help explain contradictory data regarding miR-210 expression and its putative function in solid tumors.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung Neoplasms/genetics , MicroRNAs/metabolism , Mitochondria/pathology , Apoptosis , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/ultrastructure , Caspase 3/metabolism , Caspase 7/metabolism , Cell Hypoxia/genetics , Cell Line, Tumor , Cell Survival/genetics , Down-Regulation/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Lung Neoplasms/ultrastructure , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Mitochondria/enzymology , Mitochondria/ultrastructure , Mitochondrial Proteins/metabolism , Neoplasm Staging , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Succinate Dehydrogenase/metabolism , Up-Regulation/genetics
6.
Oncogene ; 29(37): 5171-81, 2010 Sep 16.
Article in English | MEDLINE | ID: mdl-20581866

ABSTRACT

Cancer cells frequently express genes normally active in male germ cells. ATAD2 is one of them encoding a conserved factor harbouring an AAA type ATPase domain and a bromodomain. We show here that ATAD2 is highly expressed in testis as well as in many cancers of different origins and that its high expression is a strong predictor of rapid mortality in lung and breast cancers. These observations suggest that ATAD2 acts on upstream and basic cellular processes to enhance oncogenesis in a variety of unrelated cell types. Accordingly, our functional studies show that ATAD2 controls chromatin dynamics, genome transcriptional activities and apoptotic cell response. We could also highlight some of the important intrinsic properties of its two regulatory domains, including a functional cross-talk between the AAA ATPase domain and the bromodomain. Altogether, these data indicate that ATAD2 overexpression in somatic cells, by acting on basic properties of chromatin, may contribute to malignant transformation.


Subject(s)
Adenosine Triphosphatases/physiology , Breast Neoplasms/physiopathology , DNA-Binding Proteins/physiology , Lung Neoplasms/physiopathology , Testis/metabolism , ATPases Associated with Diverse Cellular Activities , Acetylation , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Amino Acid Sequence , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Molecular Sequence Data , Prognosis , Sequence Homology, Amino Acid
8.
Vet Microbiol ; 133(1-2): 105-14, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18640795

ABSTRACT

Staphylococcus aureus is recognized worldwide as a major pathogen causing clinical or subclinical intramammary infections in lactating sheep, goats and cows. The present study was carried out to compare 65 S. aureus isolates mainly obtained from nasal carriage and subclinical mastitis in dairy sheep and 43 isolates obtained from subclinical mastitis from 22 goats and 21 cows. A DNA microarray, containing probes against 190 true or putative virulence factors, was used to detect the presence of the virulence genes. Their presence/absence was independently assessed by PCR for the genes of interest. Sheep isolates obtained from the nostrils or the udders did not show any significant tissue specific virulence factor. The dominant pulse-field electrophoresis profile (OV/OV'), associated with spa clonal complex spa-CC 1773, matched mainly with the agr group III and was only found in ovine and caprine isolates. This clone was more specifically characterized by the prevalence of the following virulence genes: lpl4, ssl6, bsaA1, bsaB, bsaP, SAV0812. Moreover, seven virulence-associated genes (lpl1, sel, sec, tst, lukF-PV-like component, lukM, SAV0876) were associated with isolates from small ruminants, while the egc cluster, fhuD1, abiF and SAV2496 with bovine isolates. This genomic study suggests the existence of lineage- and host-specific genes leading to the development of host-specific pathogenic traits of S. aureus isolates.


Subject(s)
Mastitis/veterinary , Oligonucleotide Array Sequence Analysis/veterinary , Staphylococcal Infections/veterinary , Staphylococcus aureus/genetics , Virulence Factors/genetics , Animals , Cattle , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Electrophoresis, Gel, Pulsed-Field , Female , Goat Diseases/microbiology , Goats , Mastitis/microbiology , Mastitis, Bovine/microbiology , Oligonucleotide Array Sequence Analysis/methods , Sheep , Sheep Diseases/microbiology , Species Specificity , Staphylococcal Infections/microbiology
9.
Am J Transplant ; 8(6): 1221-36, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18522548

ABSTRACT

Liver ischemia-reperfusion injury occurring in orthotopic liver transplantation (OLT) may be responsible for early graft failure. Molecular mechanisms underlying initial poor graft function (IPGF) have been poorly documented in human. The purpose of this study was to identify the major transcriptional alterations occurring in human livers during OLT. Twenty-one RNA extracts derived from liver transplant biopsies taken after graft reperfusion were compared with 7 RNA derived from normal control livers. Three hundred seventy-one genes were significantly modulated and classified in molecular pathways relevant to liver metabolism, inflammatory response, cell proliferation and liver protection. Grafts were then subdivided into two groups based on their peak levels of serum aspartate amino transferase within 72 h after OLT (group 1, non-IPGF: 14 patients; group 2, IPGF: 7 patients). The two corresponding data sets were compared using a supervised prediction method. A new set of genes able to correctly classify 71% of the patients was defined. These genes were functionally associated with oxidative stress, inflammation and inhibition of cell proliferation. This study provides a comprehensive picture of the transcriptional events associated with human OLT and IPGF. We anticipate that such alterations provide a framework for the elucidation of the molecular mechanisms leading to IPGF.


Subject(s)
Delayed Graft Function/genetics , Gene Expression Profiling , Liver Diseases/genetics , Liver Transplantation , Reperfusion Injury/genetics , Adult , Aged , Female , Graft Survival/genetics , Humans , Liver , Male , Middle Aged , Transplants
10.
J Physiol ; 564(Pt 3): 671-82, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15746174

ABSTRACT

The cystic fibrosis transmembrane conductance regulator (CFTR) plays a crucial role in regulating fluid secretion by the airways, intestines, sweat glands and other epithelial tissues. It is well established that the CFTR is a cAMP-activated, nucleotide-dependent anion channel, but additional functions are often attributed to it, including regulation of the epithelial sodium channel (ENaC). The absence of CFTR-dependent ENaC inhibition and the resulting sodium hyperabsorption were postulated to be a major electrolyte transport abnormality in cystic fibrosis (CF)-affected epithelia. Several ex vivo studies, including those that used the Xenopus oocyte expression system, have reported ENaC inhibition by activated CFTR, but contradictory results have also been obtained. Because CFTR-ENaC interactions have important implications in the pathogenesis of CF, the present investigation was undertaken by our three independent laboratories to resolve whether CFTR regulates ENaC in oocytes and to clarify potential sources of previously reported dissimilar observations. Using different experimental protocols and a wide range of channel expression levels, we found no evidence that activated CFTR regulates ENaC when oocyte membrane potential was carefully clamped. We determined that an apparent CFTR-dependent ENaC inhibition could be observed when resistance in series with the oocyte membrane was not low enough or the feedback voltage gain was not high enough. We suggest that the inhibitory effect of CFTR on ENaC reported in some earlier oocyte studies could be attributed to problems arising from high levels of channel expression and suboptimal recording conditions, that is, large series resistance and/or insufficient feedback voltage gain.


Subject(s)
Cyclic AMP/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Ion Channel Gating/physiology , Oocytes/metabolism , Patch-Clamp Techniques/methods , Sodium Channels/metabolism , Animals , Epithelial Sodium Channels , Humans , Membrane Potentials/physiology , Recombinant Proteins/metabolism , Xenopus laevis
11.
J Gen Physiol ; 119(2): 199-207, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11815669

ABSTRACT

The active absorption of fluid from the airspaces of the lung is important for the resolution of clinical pulmonary edema. Although ENaC channels provide a major route for Na(+) absorption, the route of Cl(-) transport has been unclear. We applied a series of complementary approaches to define the role of Cl(-) transport in fluid clearance in the distal airspaces of the intact mouse lung, using wild-type and cystic fibrosis Delta F508 mice. Initial studies in wild-type mice showed marked inhibition of fluid clearance by Cl(-) channel inhibitors and Cl(-) ion substitution, providing evidence for a transcellular route for Cl(-) transport. In response to cAMP stimulation by isoproterenol, clearance was inhibited by the CFTR inhibitor glibenclamide in both wild-type mice and the normal human lung. Although isoproterenol markedly increased fluid absorption in wild-type mice, there was no effect in Delta F508 mice. Radioisotopic clearance studies done at 23 degrees C (to block active fluid absorption) showed approximately 20% clearance of (22)Na in 30 min both without and with isoproterenol. However, the clearance of (36)Cl was increased by 47% by isoproterenol in wild-type mice but was not changed in Delta F508 mice, providing independent evidence for involvement of CFTR in cAMP-stimulated Cl(-) transport. Further, CFTR played a major role in fluid clearance in a mouse model of acute volume-overload pulmonary edema. After infusion of saline (40% body weight), the lung wet-to-dry weight ratio increased by 28% in wild-type versus 64% in Delta F508 mice. These results provide direct evidence for a functionally important role for CFTR in the distal airspaces of the lung.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Isotonic Solutions/pharmacokinetics , Lung/physiology , Absorption/physiology , Animals , Bronchodilator Agents/pharmacology , Chloride Channels/antagonists & inhibitors , Chloride Channels/metabolism , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Humans , In Vitro Techniques , Lung/drug effects , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Pulmonary Edema/genetics , Pulmonary Edema/metabolism , Sodium Chloride/metabolism
12.
Am J Physiol Lung Cell Mol Physiol ; 281(1): L52-7, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11404245

ABSTRACT

This study was designed to test the in vivo efficacy of the chemical chaperone trimethylamine oxide (TMAO) in correcting the Cl- transport defect in a mouse model of cystic fibrosis (CF). Rectal potential difference (RPD) measurements were done in matched wild-type and DeltaF508 CF mice. Mice were treated by subcutaneous injections of TMAO. Wild-type mice demonstrated a forskolin-stimulated, Cl--dependent hyperpolarization of -6.4 +/- 0.8 mV (n = 11), which was significantly increased to -13.1 +/- 1.4 mV after treatment with TMAO. DeltaF508 CF mice showed no significant responses to forskolin. Treatment with TMAO recovered a forskolin-activated RPD in DeltaF508 CF mice (-1.1 +/- 0.2 mV; n = 17) but not in CFTR null mice. The effects of TMAO were dose dependent, resulting in a slope of -0.4 +/- 0.1 mV x g(-1) x kg(-1) in DeltaF508 CF mice. The forskolin-stimulated RPD in TMAO-treated DeltaF508 CF mice was partially blocked by glibenclamide and further stimulated by apigenin. The total response to forskolin plus apigenin was -2.5 +/- 0.45 mV (n = 6 mice), corresponding to 39% of the response evoked by forskolin only in wild-type mice.


Subject(s)
Chloride Channels/drug effects , Chloride Channels/physiology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/genetics , Cystic Fibrosis/physiopathology , Methylamines/pharmacology , Oxidants/pharmacology , Animals , Apigenin , Colforsin/pharmacology , Dose-Response Relationship, Drug , Electric Conductivity , Electrophysiology , Flavonoids/pharmacology , Glyburide/pharmacology , Mice , Rectum/drug effects , Rectum/physiopathology , Reference Values
13.
Am J Physiol Renal Physiol ; 280(2): F303-13, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11208606

ABSTRACT

The epithelial Na+ channel (ENaC) constitutes the rate-limiting step for Na+ transport across tight epithelia and is the principal target of hormonal regulation, particularly by insulin and mineralocorticoids. Recently, the serine-threonine kinase (SGK) was identified as a rapidly mineralocorticoid-responsive gene, the product of which stimulates ENaC-mediated Na+ transport. Like its close relative, protein kinase B (also called Akt), SGK's kinase activity is dependent on phosphatidylinositol 3-kinase (PI3K), a key mediator of insulin signaling. In our study we show that PI3K is required for SGK-dependent stimulation of ENaC-mediated Na+ transport as well as for the production of the phosphorylated form of SGK. In A6 kidney cells, mineralocorticoid induction of the phosphorylated form of SGK preceded the increase in Na+ transport, and specific inhibition of PI3K inhibited both phosphorylation of SGK and mineralocorticoid-induced Na+ transport. Insulin both augmented SGK phosphorylation and synergized with mineralocorticoids in stimulating Na+ transport. In a Xenopus laevis oocyte coexpression assay, SGK-stimulated ENaC activity was also markedly reduced by PI3K inhibition. Finally, in vitro-translated SGK specifically interacted with the ENaC subunits expressed in Escherichia coli as glutathione S-transferase fusion proteins. These data suggest that SGK is a PI3K-dependent integrator of insulin and mineralocorticoid actions that interacts with ENaC subunits to control Na+ entry into kidney collecting duct cells.


Subject(s)
Epithelial Cells/metabolism , Kidney/metabolism , Nuclear Proteins , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Sodium Channels/metabolism , Animals , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Immediate-Early Proteins , Insulin/pharmacology , Kidney/cytology , Kidney/drug effects , Mineralocorticoids/pharmacology , Peptides/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Serine-Threonine Kinases/drug effects , Rats , Sodium Channel Blockers , Xenopus laevis
15.
Am J Physiol Cell Physiol ; 279(3): C762-70, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10942727

ABSTRACT

There is considerable interest in identifying the basic mechanisms by which dexamethasone alters ion transport across the adult alveolar epithelium. Herein, we incubated synchronized A549 cells, a human alveolar epithelial cell line, with dexamethasone (1 microM) for 24-48 h. When normalized to HPRT (a housekeeping gene), A549 beta- and gamma-subunit mRNA levels for the human amiloride-sensitive epithelial sodium channel (hENaC), assessed by RT-PCR, increased by 1.6- and 17-fold respectively, compared with control values (P < 0.05). These changes were abolished by actinomycin D, indicating transcriptional regulation. Western blotting studies revealed that dexamethasone also increased expression of beta- and gamma-hENaC protein levels. In contrast, alpha-hENaC mRNA increased by onefold (P > 0.05) and alpha-hENaC protein level was unchanged. Incubation of A549 cells with dexamethasone increased their whole cell amiloride-sensitive sodium currents twofold and decreased the K(0.5) for amiloride from 833 +/- 69 to 22 +/- 5.4 nM (mean +/- SE; P < 0.01). Single channel recordings in the cell-attached mode showed that dexamethasone treatment increased single channel open time and open probability threefold and decreased channel conductance from 8.63 +/- 0.036 to 4. 4 +/- 0.027 pS (mean +/- SE; P < 0.01). We concluded that dexamethasone modulates the amiloride-sensitive Na(+) channels by differentially regulating the expression of beta- and gamma-subunits at the mRNA and protein levels in the human A549 cell line, with little effect on alpha-hENaC subunit.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Dexamethasone/pharmacology , Lung/metabolism , Sodium Channels/drug effects , Sodium Channels/physiology , Administration, Topical , Cell Line , Electric Conductivity , Electrophysiology , Epithelial Sodium Channels , Glucocorticoids , Humans , Lung/cytology , Lung/physiology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sodium Channels/genetics
16.
Am J Physiol Lung Cell Mol Physiol ; 278(1): L177-84, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10645905

ABSTRACT

The amiloride-sensitive epithelial Na(+) channel (ENaC) is an apical membrane protein complex involved in active Na(+) absorption and in control of fluid composition in airways. There are no data reporting the distribution of its pore-forming alpha-, beta-, and gamma-subunits in the developing human lung. With use of two different rabbit polyclonal antisera raised against beta- and gamma-ENaC, immunohistochemical localization of the channel was performed in fetal (10-35 wk) and in adult human airways. Both subunits were detected after 17 wk of gestation on the apical domain of bronchial ciliated cells, in glandular ducts, and in bronchiolar ciliated and Clara cells. After 30 wk, the distribution of beta- and gamma-subunits was similar in fetal and adult airways. In large airways, the two subunits were detected in ciliated cells, in cells lining glandular ducts, and in the serous gland cells. In the distal bronchioles, beta- and gamma-subunits were identified in ciliated and Clara cells. Ultrastructural immunogold labeling confirmed the identification of beta- and gamma-ENaC proteins in submucosal serous cells and bronchiolar Clara cells. Early expression of ENaC proteins in human fetal airways suggests that Na(+) absorption might begin significantly before birth, even if secretion is still dominant.


Subject(s)
Aging/metabolism , Bronchi/metabolism , Sodium Channels/metabolism , Adult , Bronchi/embryology , Bronchi/growth & development , Epithelial Sodium Channels , Fetus/metabolism , Humans , Immunohistochemistry , Protein Isoforms/metabolism
17.
J Hypertens ; 17(5): 639-45, 1999 May.
Article in English | MEDLINE | ID: mdl-10403607

ABSTRACT

OBJECTIVE: The gamma subunit of the epithelial Na channel (gammaENaC) has been implicated in Liddle's syndrome. The objective of this study was to examine its status in essential hypertension. DESIGN AND METHODS: The search for molecular variants was performed using the SSCP technique after determination of the intron-exon boundaries of the transcribed sequence. We found an additional 205 bp intron splitting the published exon 10 in two. The last exon of gammaENaC was tested with samples from a series of 245 normotensive patients and 453 hypertensive subjects (383 Caucasians, 70 Afro-Caribbeans), all probands of hypertensive families in the HYPERGENE data set. The search was extended to the other 11 transcribed exons in a subset of 65 patients with low-renin profile. RESULTS: Four neutral polymorphisms were detected, three in the third exon of the gene (T387C, T474C, C549T) and one in the last exon (C1990G). These four variants were found with similar frequencies in hypertensive and normotensive Caucasian subjects as well as in patients with low-renin profile. Hypertensive Caucasians and hypertensive subjects of African ancestry also had similar frequencies. Lastly, we found two rare mutations, one the insertion of a proline residue at position 594 of the mature protein (594insP), the other an Arg-to-His substitution at position 631 (R631H). Compared to wild-type (1.00 +/- 0.42, n = 26), expression of the 594insP (1.10 +/- 0.43, n = 26) and R631H (0.97 +/- 0.43, n = 26) variants in Xenopus oocytes produced no significant increase in Na+ current. CONCLUSIONS: Screening of the entire coding sequence of gammaENaC does not suggest that this subunit is frequently involved in essential hypertension.


Subject(s)
Hypertension/genetics , Polymorphism, Genetic , Sodium Channels/genetics , Animals , Epithelial Sodium Channels , Female , Humans , Hypertension/metabolism , Male , Middle Aged , Mutation , Polymorphism, Single-Stranded Conformational , Recombinant Proteins , Xenopus
18.
J Histochem Cytochem ; 47(1): 51-64, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9857212

ABSTRACT

Salt taste signals from the rat anterior tongue are probably transduced via epithelial sodium channels (ENaCs) residing in the apical cellular pole of taste cells. The signals are blocked by mucosal amiloride in low microM concentrations. In contrast, the rat vallate papilla does not contribute to amiloride-blockable salt taste. Two approaches were used to probe for the three subunits of ENaC in the anterior and posterior tongue of the rats in sodium balance. (a) Immunohistochemistry with antibodies against ENaC subunits and against amiloride binding sites. In the anterior tongue, reactivity for alpha-, beta-, and gamma-subunits was present in taste buds and lingual epithelium. In the posterior tongue vallate papilla, reactivity for alpha-subunit and for amiloride binding sites was easily demonstrable, whereas that for beta-subunit and especially for gamma-subunit was weaker than in the anterior tongue. (b) RT-PCR techniques were used to probe for the presence of ENaC subunit mRNA. In isolated taste buds of the anterior tongue, mRNA of all three subunits was found, whereas in isolated taste buds of the vallate papilla only mRNA of the alpha-subunit was easily detectable. That of beta- and gamma-subunits was much less abundant. RNA of all three subunits was abundant only in taste buds of the anterior tongue. Therefore, subsets of elongated taste cells do express ENaC, but regional differences exist in the transcription and expression of subunits. The regional differences suggest that amiloride-sensitive salt taste, which requires all three subunits, is present in the anterior but not the posterior tongue of rats, as functional studies indicate.


Subject(s)
Membrane Proteins , Sodium Channels/biosynthesis , Taste Buds/metabolism , Acid Sensing Ion Channels , Alternative Splicing , Amiloride/metabolism , Animals , Degenerin Sodium Channels , Epithelial Sodium Channels , Immunohistochemistry , Ion Channels/genetics , Kidney/metabolism , Nerve Tissue Proteins/genetics , RNA/biosynthesis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Salivary Glands/metabolism , Sodium Channels/genetics
19.
Hypertension ; 32(1): 129-37, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9674649

ABSTRACT

Mutations of the last exon of the beta subunit of the amiloride-sensitive epithelial Na+ channel (betaENaC) can lead to Liddle's syndrome, a rare monogenic form of hypertension. The objective of this study was to test whether more subtle changes of betaENaC could be implicated in essential hypertension. After determination of the betaENaC coding gene organization (12 exons spanning 23.5 kb), a systematic screening of the last exon of the gene was performed in 525 subjects (475 whites, 50 Afro-Caribbeans), all probands of hypertensive families. This search was extended to the remaining 11 exons in a subset of 101 probands with low-renin hypertension. Seven amino acid changes were detected: G589S, T594M, R597H, R624C, E632G (last exon), G442V, and V434M (exon 8). These genetic variants were more frequent in subjects of African origin (44%) than in whites (1%). The functional properties of the variants were analyzed in Xenopus oocytes by two independent techniques, ie, electrophysiology and 22Na+ uptake. Small but not significant differences were observed between the variants and wild type. The clinical evaluation of the family bearing the G589S variant, which provided the highest relative ENaC activity, did not show a cosegregation between the mutation and hypertension. The present study illustrates the difficulty in establishing a relation of causality between a susceptibility gene and hypertension. Furthermore, it does not favor a substantial role of the betaENaC gene in essential hypertension.


Subject(s)
Hypertension/genetics , Sodium Channels/genetics , Adult , Animals , Base Sequence , Epithelium/metabolism , Exons/genetics , Female , Genetic Variation , Humans , Hypertension/metabolism , Introns/genetics , Male , Middle Aged , Molecular Sequence Data , Mutagenesis , Oocytes/metabolism , Pedigree , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational , Sequence Analysis, DNA , Sodium/metabolism , Sodium Channels/metabolism , Xenopus laevis
20.
J Biol Chem ; 273(16): 9424-9, 1998 Apr 17.
Article in English | MEDLINE | ID: mdl-9545267

ABSTRACT

Amiloride-sensitive sodium channels have been implicated in reproductive and early developmental processes of several species. These include the fast block of polyspermy in Xenopus oocytes that follows the sperm binding to the egg or blastocoel expansion in mammalian embryo. We have now identified a gene called dGNaC1 that is specifically expressed in the gonads and early embryo in Drosophila melanogaster. The corresponding protein belongs to the superfamily of cationic channels blocked by amiloride that includes Caenorhabditis elegans degenerins, the Helix aspersa FMRF-amide ionotropic receptor (FaNaC), the mammalian epithelial Na+ channel (ENaC), and acid-sensing ionic channels (ASIC, DRASIC, and MDEG). Expression of dGNaC1 in Xenopus oocytes generates a constitutive current that does not discriminate between Na+ and Li+, but is selective for Na+ over K+. This current is blocked by amiloride (IC50 = 24 microM), benzamil (IC50 = 2 microM), and ethylisopropyl amiloride (IC50 = 49 microM). These properties are clearly different from those obtained after expression of the previously cloned members of this family, including ENaC and the human alphaENaC-like subunit, deltaNaC. Interestingly, the pharmacology of dGNaC1 is not very different from that found for the Na+ channel characterized in rabbit preimplantation embryos. We postulate that this channel may participate in gametogenesis and early embryonic development in Drosophila.


Subject(s)
Sodium Channels/biosynthesis , Sodium Channels/physiology , Testis/metabolism , Amiloride/analogs & derivatives , Amiloride/metabolism , Amiloride/pharmacology , Amino Acid Sequence , Animals , Caenorhabditis elegans , Cations/metabolism , Drosophila Proteins , Drosophila melanogaster , Embryo, Nonmammalian/physiology , Epithelial Sodium Channels , Female , Helix, Snails , Humans , Male , Membrane Potentials/physiology , Molecular Sequence Data , Multigene Family , Oocytes/physiology , Rabbits , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Sequence Alignment , Sequence Homology, Amino Acid , Sex Characteristics , Sodium/metabolism , Sodium Channels/chemistry , Sodium Channels/genetics , Sperm-Ovum Interactions , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...