Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 48
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Nature ; 620(7975): 890-897, 2023 Aug.
Article de Anglais | MEDLINE | ID: mdl-37558881

RÉSUMÉ

Alveolar epithelial type 1 (AT1) cells are necessary to transfer oxygen and carbon dioxide between the blood and air. Alveolar epithelial type 2 (AT2) cells serve as a partially committed stem cell population, producing AT1 cells during postnatal alveolar development and repair after influenza A and SARS-CoV-2 pneumonia1-6. Little is known about the metabolic regulation of the fate of lung epithelial cells. Here we report that deleting the mitochondrial electron transport chain complex I subunit Ndufs2 in lung epithelial cells during mouse gestation led to death during postnatal alveolar development. Affected mice displayed hypertrophic cells with AT2 and AT1 cell features, known as transitional cells. Mammalian mitochondrial complex I, comprising 45 subunits, regenerates NAD+ and pumps protons. Conditional expression of yeast NADH dehydrogenase (NDI1) protein that regenerates NAD+ without proton pumping7,8 was sufficient to correct abnormal alveolar development and avert lethality. Single-cell RNA sequencing revealed enrichment of integrated stress response (ISR) genes in transitional cells. Administering an ISR inhibitor9,10 or NAD+ precursor reduced ISR gene signatures in epithelial cells and partially rescued lethality in the absence of mitochondrial complex I function. Notably, lung epithelial-specific loss of mitochondrial electron transport chain complex II subunit Sdhd, which maintains NAD+ regeneration, did not trigger high ISR activation or lethality. These findings highlight an unanticipated requirement for mitochondrial complex I-dependent NAD+ regeneration in directing cell fate during postnatal alveolar development by preventing pathological ISR induction.


Sujet(s)
Pneumocytes , Différenciation cellulaire , Lignage cellulaire , Poumon , Mitochondries , Stress physiologique , Animaux , Souris , Pneumocytes/cytologie , Pneumocytes/métabolisme , Pneumocytes/anatomopathologie , Poumon/cytologie , Poumon/métabolisme , Poumon/anatomopathologie , Mitochondries/enzymologie , Mitochondries/métabolisme , NAD/métabolisme , NADH dehydrogenase/métabolisme , Protons , RNA-Seq , Saccharomyces cerevisiae/génétique , Saccharomyces cerevisiae/métabolisme , Analyse de l'expression du gène de la cellule unique
2.
JCI Insight ; 8(4)2023 02 22.
Article de Anglais | MEDLINE | ID: mdl-36626234

RÉSUMÉ

Persistent symptoms and radiographic abnormalities suggestive of failed lung repair are among the most common symptoms in patients with COVID-19 after hospital discharge. In mechanically ventilated patients with acute respiratory distress syndrome (ARDS) secondary to SARS-CoV-2 pneumonia, low tidal volumes to reduce ventilator-induced lung injury necessarily elevate blood CO2 levels, often leading to hypercapnia. The role of hypercapnia on lung repair after injury is not completely understood. Here - using a mouse model of hypercapnia exposure, cell lineage tracing, spatial transcriptomics, and 3D cultures - we show that hypercapnia limits ß-catenin signaling in alveolar type II (AT2) cells, leading to their reduced proliferative capacity. Hypercapnia alters expression of major Wnts in PDGFRα+ fibroblasts from those maintaining AT2 progenitor activity toward those that antagonize ß-catenin signaling, thereby limiting progenitor function. Constitutive activation of ß-catenin signaling in AT2 cells or treatment of organoid cultures with recombinant WNT3A protein bypasses the inhibitory effects of hypercapnia. Inhibition of AT2 proliferation in patients with hypercapnia may contribute to impaired lung repair after injury, preventing sealing of the epithelial barrier and increasing lung flooding, ventilator dependency, and mortality.


Sujet(s)
Hypercapnie , Voie de signalisation Wnt , Souris , bêta-Caténine/métabolisme , Prolifération cellulaire , COVID-19/complications , Hypercapnie/métabolisme , Animaux
3.
Am J Respir Cell Mol Biol ; 66(5): 564-576, 2022 05.
Article de Anglais | MEDLINE | ID: mdl-35202558

RÉSUMÉ

Epithelial polyploidization after injury is a conserved phenomenon recently shown to improve barrier restoration during wound healing. Whether lung injury can induce alveolar epithelial polyploidy is not known. We show that bleomycin injury induces alveolar type 2 cell (AT2) hypertrophy and polyploidy. AT2 polyploidization is also seen in short term ex vivo cultures, where AT2-to-AT1 transdifferentiation is associated with substantial binucleation due to failed cytokinesis. Both hypertrophic and polyploid features of AT2 cells can be attenuated by inhibiting the integrated stress response using the small molecule ISRIB. These data suggest that AT2 hypertrophic growth and polyploidization may be a feature of alveolar epithelial injury. Because AT2 cells serve as facultative progenitors for the distal lung epithelium, a propensity for injury-induced binucleation has implications for AT2 self-renewal and regenerative potential upon reinjury, which may benefit from targeting the integrated stress response.


Sujet(s)
Lésion pulmonaire , Pneumocytes/métabolisme , Différenciation cellulaire , Humains , Hypertrophie/métabolisme , Lésion pulmonaire/induit chimiquement , Lésion pulmonaire/génétique , Lésion pulmonaire/métabolisme , Polyploïdie
4.
Front Cell Dev Biol ; 9: 689983, 2021.
Article de Anglais | MEDLINE | ID: mdl-34277634

RÉSUMÉ

Several acute and chronic lung diseases are associated with alveolar hypoventilation leading to accumulation of CO2 (hypercapnia). The ß-subunit of the Na,K-ATPase plays a pivotal role in maintaining epithelial integrity by functioning as a cell adhesion molecule and regulating cell surface stability of the catalytic α-subunit of the transporter, thereby, maintaining optimal alveolar fluid balance. Here, we identified the E3 ubiquitin ligase for the Na,K-ATPase ß-subunit, which promoted polyubiquitination, subsequent endocytosis and proteasomal degradation of the protein upon exposure of alveolar epithelial cells to elevated CO2 levels, thus impairing alveolar integrity. Ubiquitination of the Na,K-ATPase ß-subunit required lysine 5 and 7 and mutating these residues (but not other lysines) prevented trafficking of Na,K-ATPase from the plasma membrane and stabilized the protein upon hypercapnia. Furthermore, ubiquitination of the Na,K-ATPase ß-subunit was dependent on prior phosphorylation at serine 11 by protein kinase C (PKC)-ζ. Using a protein microarray, we identified the tumor necrosis factor receptor-associated factor 2 (TRAF2) as the E3 ligase driving ubiquitination of the Na,K-ATPase ß-subunit upon hypercapnia. Of note, prevention of Na,K-ATPase ß-subunit ubiquitination was necessary and sufficient to restore the formation of cell-cell junctions under hypercapnic conditions. These results suggest that a hypercapnic environment in the lung may lead to persistent epithelial dysfunction in affected patients. As such, the identification of the E3 ligase for the Na,K-ATPase may provide a novel therapeutic target, to be employed in patients with acute or chronic hypercapnic respiratory failure, aiming to restore alveolar epithelial integrity.

5.
J Membr Biol ; 254(5-6): 447-457, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34114062

RÉSUMÉ

The Na,K-ATPase establishes the electrochemical gradient of cells by driving an active exchange of Na+ and K+ ions while consuming ATP. The minimal functional transporter consists of a catalytic α-subunit and a ß-subunit with chaperon activity. The Na,K-ATPase also functions as a cell adhesion molecule and participates in various intracellular signaling pathways. The maturation and trafficking of the Na,K-ATPase include co- and post-translational processing of the enzyme in the endoplasmic reticulum (ER) and the Golgi apparatus and subsequent delivery to the plasma membrane (PM). The ER folding of the enzyme is considered as the rate-limiting step in the membrane delivery of the protein. It has been demonstrated that only assembled Na,K-ATPase α:ß-complexes may exit the organelle, whereas unassembled, misfolded or unfolded subunits are retained in the ER and are subsequently degraded. Loss of function of the Na,K-ATPase has been associated with lung, heart, kidney and neurological disorders. Recently, it has been shown that ER dysfunction, in particular, alterations in the homeostasis of the organelle, as well as impaired ER-resident chaperone activity may impede folding of Na,K-ATPase subunits, thus decreasing the abundance and function of the enzyme at the PM. Here, we summarize our current understanding on maturation and subsequent processing of the Na,K-ATPase in the ER under physiological and pathophysiological conditions.


Sujet(s)
Réticulum endoplasmique , Membrane cellulaire/métabolisme , Réticulum endoplasmique/métabolisme , Ions/métabolisme , Pliage des protéines , Sodium-Potassium-Exchanging ATPase/métabolisme
7.
Int J Mol Sci ; 21(4)2020 Feb 21.
Article de Anglais | MEDLINE | ID: mdl-32098115

RÉSUMÉ

Alveolar edema, impaired alveolar fluid clearance, and elevated CO2 levels (hypercapnia) are hallmarks of the acute respiratory distress syndrome (ARDS). This study investigated how hypercapnia affects maturation of the Na,K-ATPase (NKA), a key membrane transporter, and a cell adhesion molecule involved in the resolution of alveolar edema in the endoplasmic reticulum (ER). Exposure of human alveolar epithelial cells to elevated CO2 concentrations caused a significant retention of NKA-ß in the ER and, thus, decreased levels of the transporter in the Golgi apparatus. These effects were associated with a marked reduction of the plasma membrane (PM) abundance of the NKA-α/ß complex as well as a decreased total and ouabain-sensitive ATPase activity. Furthermore, our study revealed that the ER-retained NKA-ß subunits were only partially assembled with NKA α-subunits, which suggests that hypercapnia modifies the ER folding environment. Moreover, we observed that elevated CO2 levels decreased intracellular ATP production and increased ER protein and, particularly, NKA-ß oxidation. Treatment with α-ketoglutaric acid (α-KG), which is a metabolite that has been shown to increase ATP levels and rescue mitochondrial function in hypercapnia-exposed cells, attenuated the deleterious effects of elevated CO2 concentrations and restored NKA PM abundance and function. Taken together, our findings provide new insights into the regulation of NKA in alveolar epithelial cells by elevated CO2 levels, which may lead to the development of new therapeutic approaches for patients with ARDS and hypercapnia.


Sujet(s)
Pneumocytes/enzymologie , Dioxyde de carbone/métabolisme , Réticulum endoplasmique/enzymologie , Hypercapnie/enzymologie , Pliage des protéines , Sodium-Potassium-Exchanging ATPase/métabolisme , Cellules A549 , Pneumocytes/anatomopathologie , Animaux , Réticulum endoplasmique/anatomopathologie , Humains , Hypercapnie/anatomopathologie , Rats
8.
J Clin Invest ; 130(3): 1301-1314, 2020 03 02.
Article de Anglais | MEDLINE | ID: mdl-31714898

RÉSUMÉ

Influenza A virus (IAV) is among the most common causes of pneumonia-related death worldwide. Pulmonary epithelial cells are the primary target for viral infection and replication and respond by releasing inflammatory mediators that recruit immune cells to mount the host response. Severe lung injury and death during IAV infection result from an exuberant host inflammatory response. The linear ubiquitin assembly complex (LUBAC), composed of SHARPIN, HOIL-1L, and HOIP, is a critical regulator of NF-κB-dependent inflammation. Using mice with lung epithelial-specific deletions of HOIL-1L or HOIP in a model of IAV infection, we provided evidence that, while a reduction in the inflammatory response was beneficial, ablation of the LUBAC-dependent lung epithelial-driven response worsened lung injury and increased mortality. Moreover, we described a mechanism for the upregulation of HOIL-1L in infected and noninfected cells triggered by the activation of type I IFN receptor and mediated by IRF1, which was maladaptive and contributed to hyperinflammation. Thus, we propose that lung epithelial LUBAC acts as a molecular rheostat that could be selectively targeted to modulate the immune response in patients with severe IAV-induced pneumonia.


Sujet(s)
Sous-type H1N1 du virus de la grippe A/immunologie , Poumon/immunologie , Complexes multiprotéiques/immunologie , Infections à Orthomyxoviridae/immunologie , Pneumopathie virale/immunologie , Muqueuse respiratoire/immunologie , Cellules A549 , Animaux , Chiens , Humains , Sous-type H1N1 du virus de la grippe A/génétique , Facteur-1 de régulation d'interféron/génétique , Facteur-1 de régulation d'interféron/immunologie , Poumon/anatomopathologie , Poumon/virologie , Cellules rénales canines Madin-Darby , Souris , Souris knockout , Complexes multiprotéiques/génétique , Infections à Orthomyxoviridae/génétique , Infections à Orthomyxoviridae/anatomopathologie , Pneumopathie virale/génétique , Pneumopathie virale/anatomopathologie , Muqueuse respiratoire/anatomopathologie , Muqueuse respiratoire/virologie , Ubiquitin-protein ligases/déficit , Ubiquitin-protein ligases/immunologie
9.
Sci Rep ; 9(1): 18251, 2019 12 03.
Article de Anglais | MEDLINE | ID: mdl-31796806

RÉSUMÉ

Carbon dioxide (CO2) is sensed by cells and can trigger signals to modify gene expression in different tissues leading to changes in organismal functions. Despite accumulating evidence that several pathways in various organisms are responsive to CO2 elevation (hypercapnia), it has yet to be elucidated how hypercapnia activates genes and signaling pathways, or whether they interact, are integrated, or are conserved across species. Here, we performed a large-scale transcriptomic study to explore the interaction/integration/conservation of hypercapnia-induced genomic responses in mammals (mice and humans) as well as invertebrates (Caenorhabditis elegans and Drosophila melanogaster). We found that hypercapnia activated genes that regulate Wnt signaling in mouse lungs and skeletal muscles in vivo and in several cell lines of different tissue origin. Hypercapnia-responsive Wnt pathway homologues were similarly observed in secondary analysis of available transcriptomic datasets of hypercapnia in a human bronchial cell line, flies and nematodes. Our data suggest the evolutionarily conserved role of high CO2 in regulating Wnt pathway genes.


Sujet(s)
Caenorhabditis elegans/métabolisme , Dioxyde de carbone/pharmacologie , Drosophila melanogaster/métabolisme , Voie de signalisation Wnt/effets des médicaments et des substances chimiques , Animaux , Bronches/cytologie , Bronches/métabolisme , Caenorhabditis elegans/effets des médicaments et des substances chimiques , Lignée cellulaire , Drosophila melanogaster/effets des médicaments et des substances chimiques , Analyse de profil d'expression de gènes , Humains , Hypercapnie/métabolisme , Mâle , Souris , Souris de lignée C57BL , Réaction de polymérisation en chaine en temps réel , Analyse sur puce à tissus
10.
J Immunol ; 202(2): 484-493, 2019 01 15.
Article de Anglais | MEDLINE | ID: mdl-30530483

RÉSUMÉ

Muscle dysfunction is common in patients with adult respiratory distress syndrome and is associated with morbidity that can persist for years after discharge. In a mouse model of severe influenza A pneumonia, we found the proinflammatory cytokine IL-6 was necessary for the development of muscle dysfunction. Treatment with a Food and Drug Administration-approved Ab antagonist to the IL-6R (tocilizumab) attenuated the severity of influenza A-induced muscle dysfunction. In cultured myotubes, IL-6 promoted muscle degradation via JAK/STAT, FOXO3a, and atrogin-1 upregulation. Consistent with these findings, atrogin-1+/- and atrogin-1-/- mice had attenuated muscle dysfunction following influenza infection. Our data suggest that inflammatory endocrine signals originating from the injured lung activate signaling pathways in the muscle that induce dysfunction. Inhibiting these pathways may limit morbidity in patients with influenza A pneumonia and adult respiratory distress syndrome.


Sujet(s)
Virus de la grippe A/physiologie , Grippe humaine/immunologie , Interleukine-6/métabolisme , Poumon/physiologie , Protéines du muscle/métabolisme , Muscles/anatomopathologie , Infections à Orthomyxoviridae/immunologie , Pneumopathie virale/immunologie , SKP cullin F-box protein ligases/métabolisme , Syndrome cachectique/immunologie , Animaux , Cellules cultivées , Modèles animaux de maladie humaine , Protéine O3 à motif en tête de fourche/métabolisme , Humains , Interleukine-6/génétique , Janus kinases/métabolisme , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Protéines du muscle/génétique , SKP cullin F-box protein ligases/génétique , Facteurs de transcription STAT/métabolisme , Transduction du signal
11.
Transl Res ; 198: 29-39, 2018 08.
Article de Anglais | MEDLINE | ID: mdl-29752900

RÉSUMÉ

Cell homeostasis requires precise coordination of cellular proteins function. Ubiquitination is a post-translational modification that modulates protein half-life and function and is tightly regulated by ubiquitin E3 ligases and deubiquitinating enzymes. Lung injury can progress to acute respiratory distress syndrome that is characterized by an inflammatory response and disruption of the alveolocapillary barrier resulting in alveolar edema accumulation and hypoxemia. Ubiquitination plays an important role in the pathobiology of acute lung injury as it regulates the proteins modulating the alveolocapillary barrier and the inflammatory response. Better understanding of the signaling pathways regulated by ubiquitination may lead to novel therapeutic approaches by targeting specific elements of the ubiquitination pathways.


Sujet(s)
Lésion pulmonaire/enzymologie , Proteasome endopeptidase complex/métabolisme , /enzymologie , Transduction du signal , Ubiquitine/métabolisme , Humains , Protéolyse
13.
Proc Natl Acad Sci U S A ; 114(47): E10178-E10186, 2017 11 21.
Article de Anglais | MEDLINE | ID: mdl-29109255

RÉSUMÉ

Organisms have evolved adaptive mechanisms in response to stress for cellular survival. During acute hypoxic stress, cells down-regulate energy-consuming enzymes such as Na,K-ATPase. Within minutes of alveolar epithelial cell (AEC) exposure to hypoxia, protein kinase C zeta (PKCζ) phosphorylates the α1-Na,K-ATPase subunit and triggers it for endocytosis, independently of the hypoxia-inducible factor (HIF). However, the Na,K-ATPase activity is essential for cell homeostasis. HIF induces the heme-oxidized IRP2 ubiquitin ligase 1L (HOIL-1L), which leads to PKCζ degradation. Here we report a mechanism of prosurvival adaptation of AECs to prolonged hypoxia where PKCζ degradation allows plasma membrane Na,K-ATPase stabilization at ∼50% of normoxic levels, preventing its excessive down-regulation and cell death. Mice lacking HOIL-1L in lung epithelial cells (CreSPC/HOIL-1Lfl/fl ) were sensitized to hypoxia because they express higher levels of PKCζ and, consequently, lower plasma membrane Na,K-ATPase levels, which increased cell death and worsened lung injury. In AECs, expression of an α1-Na,K-ATPase construct bearing an S18A (α1-S18A) mutation, which precludes PKCζ phosphorylation, stabilized the Na,K-ATPase at the plasma membrane and prevented hypoxia-induced cell death even in the absence of HOIL-1L. Adenoviral overexpression of the α1-S18A mutant Na,K-ATPase in vivo rescued the enhanced sensitivity of CreSPC/HOIL-1Lfl/fl mice to hypoxic lung injury. These data suggest that stabilization of Na,K-ATPase during severe hypoxia is a HIF-dependent process involving PKCζ degradation. Accordingly, we provide evidence of an important adaptive mechanism to severe hypoxia, whereby halting the exaggerated down-regulation of plasma membrane Na,K-ATPase prevents cell death and lung injury.


Sujet(s)
Protéines de transport/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Hypoxie/anatomopathologie , Lésion pulmonaire/anatomopathologie , Protéine kinase C/métabolisme , Sodium-Potassium-Exchanging ATPase/métabolisme , Cellules A549 , Animaux , Apoptose , Cellules COS , Protéines de transport/génétique , Hypoxie cellulaire , Membrane cellulaire/métabolisme , Chlorocebus aethiops , Régulation négative , Endocytose , Cellules épithéliales/anatomopathologie , Humains , Hypoxie/complications , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Lésion pulmonaire/étiologie , Mâle , Souris , Souris de lignée C57BL , Souris de lignée ICR , Souris knockout , Mutation , Phosphorylation , Culture de cellules primaires , Protéolyse , Alvéoles pulmonaires/cytologie , Alvéoles pulmonaires/anatomopathologie , Interférence par ARN , Petit ARN interférent/métabolisme , Rats , Rat Sprague-Dawley , Sodium-Potassium-Exchanging ATPase/génétique
14.
Front Immunol ; 8: 623, 2017.
Article de Anglais | MEDLINE | ID: mdl-28620381

RÉSUMÉ

The alveolar epithelium secretes cytokines and chemokines that recruit immune cells to the lungs, which is essential for fighting infections but in excess can promote lung injury. Overexpression of FXYD5, a tissue-specific regulator of the Na,K-ATPase, in mice, impairs the alveolo-epithelial barrier, and FXYD5 overexpression in renal cells increases C-C chemokine ligand-2 (CCL2) secretion in response to lipopolysaccharide (LPS). The aim of this study was to determine whether FXYD5 contributes to the lung inflammation and injury. Exposure of alveolar epithelial cells (AEC) to LPS increased FXYD5 levels at the plasma membrane, and FXYD5 silencing prevented both the activation of NF-κB and the secretion of cytokines in response to LPS. Intratracheal instillation of LPS into mice increased FXYD5 levels in the lung. FXYD5 overexpression increased the recruitment of interstitial macrophages and classical monocytes to the lung in response to LPS. FXYD5 silencing decreased CCL2 levels, number of cells, and protein concentration in bronchoalveolar lavage fluid (BALF) after LPS treatment, indicating that FXYD5 is required for the NF-κB-stimulated epithelial production of CCL2, the influx of immune cells, and the increase in alveolo-epithelial permeability in response to LPS. Silencing of FXYD5 also prevented the activation of NF-κB and cytokine secretion in response to interferon α and TNF-α, suggesting that pro-inflammatory effects of FXYD5 are not limited to the LPS-induced pathway. Furthermore, in the absence of other stimuli, FXYD5 overexpression in AEC activated NF-κB and increased cytokine production, while FXYD5 overexpression in mice increased cytokine levels in BALF, indicating that FXYD5 is sufficient to induce the NF-κB-stimulated cytokine secretion by the alveolar epithelium. The FXYD5 overexpression also increased cell counts in BALF, which was prevented by silencing the CCL2 receptor (CCR2), or by treating mice with a CCR2-blocking antibody, confirming that FXYD5-induced CCL2 production leads to the recruitment of monocytes to the lung. Taken together, the data demonstrate that FXYD5 is a key contributor to inflammatory lung injury.

15.
Cell Signal ; 38: 49-59, 2017 10.
Article de Anglais | MEDLINE | ID: mdl-28652146

RÉSUMÉ

Atypical protein kinase C ζ (PKCζ) forms an apico-basal polarity complex with Partitioning Defective (Pard) 3 and Pard6 to regulate normal epithelial cell apico-basolateral polarization. The dissociation of the PKCζ/Pard3/Pard6 complex is essential for the disassembly of the tight/adherens junction and epithelial-mesenchymal transition (EMT) that is critical for tumor spreading. Loss of cell polarity and epithelial organization is strongly correlated with malignancy and tumor progression in some other cancer types. However, it is unclear whether the PKCζ/Pard3/Pard6 complex plays a role in the progression of non-small-cell lung cancer (NSCLC). We found that hypoxia downregulated the PKCζ/Pard3/Pard6 complex, correlating with induction of lung cancer cell migration and invasion. Silencing of the PKCζ/Pard3/Pard6 polarity complex components induced lung cancer cell EMT, invasion, and colonization in vivo. Suppression of Pard3 was associated with altered expression of genes regulating wound healing, cell apoptosis/death and cell motility, and particularly upregulation of MAP3K1 and fibronectin which are known to contribute to lung cancer progression. Human lung adenocarcinoma tissues expressed less Pard6b and PKCζ than the adjacent normal tissues and in experimental mouse lung adenocarcinoma, the levels of Pard3 and PKCζ were also decreased. In addition, we showed that a methylation locus in the gene body of Pard3 is positively associated with the expression of Pard3 and that methylation of the Pard3 gene increased cellular sensitivity to carboplatin, a common chemotherapy drug. Suppression of Pard3 increased chemoresistance in lung cancer cells. Together, these results suggest that reduced expression of PKCζ/Pard3/Pard6 contributes to NSCLC EMT, invasion, and chemoresistance.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Adénocarcinome/métabolisme , Adénocarcinome/anatomopathologie , Protéines du cycle cellulaire/métabolisme , Transition épithélio-mésenchymateuse , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Protéines membranaires/métabolisme , Protéine kinase C/métabolisme , Cellules A549 , Adénocarcinome pulmonaire , Animaux , Carboplatine/pharmacologie , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Hypoxie cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Polarité de la cellule/effets des médicaments et des substances chimiques , Cisplatine/pharmacologie , Régulation négative/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Fibronectines/métabolisme , Extinction de l'expression des gènes/effets des médicaments et des substances chimiques , Humains , Poumon/effets des médicaments et des substances chimiques , Poumon/métabolisme , Poumon/anatomopathologie , MAP Kinase Kinase Kinase 1/métabolisme , Souris nude , Invasion tumorale
16.
J Biol Chem ; 291(44): 23159-23174, 2016 10 28.
Article de Anglais | MEDLINE | ID: mdl-27624940

RÉSUMÉ

The Na,K-ATPase α2 subunit plays a key role in cardiac muscle contraction by regulating intracellular Ca2+, whereas α1 has a more conventional role of maintaining ion homeostasis. The ß subunit differentially regulates maturation, trafficking, and activity of α-ß heterodimers. It is not known whether the distinct role of α2 in the heart is related to selective assembly with a particular one of the three ß isoforms. We show here by immunofluorescence and co-immunoprecipitation that α2 is preferentially expressed with ß2 in T-tubules of cardiac myocytes, forming α2ß2 heterodimers. We have expressed human α1ß1, α2ß1, α2ß2, and α2ß3 in Pichia pastoris, purified the complexes, and compared their functional properties. α2ß2 and α2ß3 differ significantly from both α2ß1 and α1ß1 in having a higher K0.5K+ and lower K0.5Na+ for activating Na,K-ATPase. These features are the result of a large reduction in binding affinity for extracellular K+ and shift of the E1P-E2P conformational equilibrium toward E1P. A screen of perhydro-1,4-oxazepine derivatives of digoxin identified several derivatives (e.g. cyclobutyl) with strongly increased selectivity for inhibition of α2ß2 and α2ß3 over α1ß1 (range 22-33-fold). Molecular modeling suggests a possible basis for isoform selectivity. The preferential assembly, specific T-tubular localization, and low K+ affinity of α2ß2 could allow an acute response to raised ambient K+ concentrations in physiological conditions and explain the importance of α2ß2 for cardiac muscle contractility. The high sensitivity of α2ß2 to digoxin derivatives explains beneficial effects of cardiac glycosides for treatment of heart failure and potential of α2ß2-selective digoxin derivatives for reducing cardiotoxicity.


Sujet(s)
Adenosine triphosphatases/métabolisme , Transporteurs de cations/métabolisme , Molécules d'adhérence cellulaire neuronale/métabolisme , Antienzymes/composition chimique , Myocarde/enzymologie , Sodium-Potassium-Exchanging ATPase/métabolisme , Adenosine triphosphatases/antagonistes et inhibiteurs , Adenosine triphosphatases/composition chimique , Animaux , Transporteurs de cations/antagonistes et inhibiteurs , Transporteurs de cations/composition chimique , Molécules d'adhérence cellulaire neuronale/antagonistes et inhibiteurs , Molécules d'adhérence cellulaire neuronale/composition chimique , Dimérisation , Antienzymes/métabolisme , Humains , Isoenzymes/antagonistes et inhibiteurs , Isoenzymes/composition chimique , Isoenzymes/génétique , Isoenzymes/métabolisme , Cinétique , Souris , Myocarde/composition chimique , Potassium/composition chimique , Potassium/métabolisme , Sodium/composition chimique , Sodium/métabolisme , Sodium-Potassium-Exchanging ATPase/antagonistes et inhibiteurs , Sodium-Potassium-Exchanging ATPase/composition chimique , Sodium-Potassium-Exchanging ATPase/génétique
17.
J Cell Sci ; 129(12): 2394-406, 2016 06 15.
Article de Anglais | MEDLINE | ID: mdl-27142834

RÉSUMÉ

FXYD5 (also known as dysadherin), a regulatory subunit of the Na,K-ATPase, impairs intercellular adhesion by a poorly understood mechanism. Here, we determined whether FXYD5 disrupts the trans-dimerization of Na,K-ATPase molecules located in neighboring cells. Mutagenesis of the Na,K-ATPase ß1 subunit identified four conserved residues, including Y199, that are crucial for the intercellular Na,K-ATPase trans-dimerization and adhesion. Modulation of expression of FXYD5 or of the ß1 subunit with intact or mutated ß1-ß1 binding sites demonstrated that the anti-adhesive effect of FXYD5 depends on the presence of Y199 in the ß1 subunit. Immunodetection of the plasma membrane FXYD5 was prevented by the presence of O-glycans. Partial FXYD5 deglycosylation enabled antibody binding and showed that the protein level and the degree of O-glycosylation were greater in cancer than in normal cells. FXYD5-induced impairment of adhesion was abolished by both genetic and pharmacological inhibition of FXYD5 O-glycosylation. Therefore, the extracellular O-glycosylated domain of FXYD5 impairs adhesion by interfering with intercellular ß1-ß1 interactions, suggesting that the ratio between FXYD5 and α1-ß1 heterodimer determines whether the Na,K-ATPase acts as a positive or negative regulator of intercellular adhesion.


Sujet(s)
Glycoprotéines membranaires/métabolisme , Protéines tumorales/métabolisme , Multimérisation de protéines , Sous-unités de protéines/métabolisme , Sodium-Potassium-Exchanging ATPase/métabolisme , Cellules A549 , Acides aminés/métabolisme , Animaux , Spécificité des anticorps , Adhérence cellulaire , Lignée cellulaire tumorale , Membrane cellulaire/métabolisme , Chiens , Cellules épithéliales/métabolisme , Techniques de knock-down de gènes , Glycosylation , Cellules HEK293 , Humains , Canaux ioniques , Cellules rénales canines Madin-Darby , Souris , Protéines des microfilaments , Liaison aux protéines , Sous-unités de protéines/composition chimique , Rats , Sodium-Potassium-Exchanging ATPase/composition chimique
18.
J Biol Chem ; 291(21): 11072-82, 2016 May 20.
Article de Anglais | MEDLINE | ID: mdl-27006401

RÉSUMÉ

The FXYD proteins are a family of small membrane proteins that share an invariant four amino acid signature motif F-X-Y-D and act as tissue-specific regulatory subunits of the Na,K-ATPase. FXYD5 (also termed dysadherin or RIC) is a structurally and functionally unique member of the FXYD family. As other FXYD proteins, FXYD5 specifically interacts with the Na,K-ATPase and alters its kinetics by increasing Vmax However, unlike other family members FXYD5 appears to have additional functions, which cannot be readily explained by modulation of transport kinetics. Knockdown of FXYD5 in MDA-MB-231 breast cancer cells largely decreases expression and secretion of the chemokine CCL2 (MCP-1). A related effect has also been observed in renal cell carcinoma cells. The current study aims to further characterize the relationship between the expression of FXYD5 and CCL2 secretion. We demonstrate that transfection of M1 epithelial cell line with FXYD5 largely increases lipopolysaccharide (LPS) stimulated CCL2 mRNA and secretion of the translated protein. We have completed a detailed analysis of the molecular events leading to the above response. Our key findings indicate that FXYD5 generates a late response by increasing the surface expression of the TNFα receptor, without affecting its total protein level, or mRNA transcription. LPS administration to mice demonstrates induced secretion of CCL2 and TNFα in FXYD5-expressing lung peripheral tissue, which suggests a possible role for FXYD5 in normal epithelia during inflammation.


Sujet(s)
Glycoprotéines membranaires/métabolisme , Protéines membranaires/métabolisme , Protéines tumorales/métabolisme , Animaux , Lignée cellulaire , Lignée cellulaire tumorale , Chimiokine CCL2/génétique , Chimiokine CCL2/métabolisme , Cellules épithéliales/métabolisme , Expression des gènes/effets des médicaments et des substances chimiques , Techniques de knock-down de gènes , Humains , Médiateurs de l'inflammation/métabolisme , Canaux ioniques , Cinétique , Lipopolysaccharides/pharmacologie , Glycoprotéines membranaires/antagonistes et inhibiteurs , Glycoprotéines membranaires/génétique , Protéines membranaires/génétique , Souris , Protéines des microfilaments , Protéines tumorales/antagonistes et inhibiteurs , Protéines tumorales/génétique , ARN messager/génétique , ARN messager/métabolisme , Récepteurs aux facteurs de nécrose tumorale/génétique , Récepteurs aux facteurs de nécrose tumorale/métabolisme , Protéines recombinantes/génétique , Protéines recombinantes/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
19.
Am J Respir Cell Mol Biol ; 54(6): 761-8, 2016 06.
Article de Anglais | MEDLINE | ID: mdl-26848516

RÉSUMÉ

The covalent attachment of ubiquitin to target proteins is one of the most prevalent post-translational modifications, regulating a myriad of cellular processes including cell growth, survival, and metabolism. Recently, a novel RING E3 ligase complex was described, called linear ubiquitin assembly complex (LUBAC), which is capable of connecting ubiquitin molecules in a novel head-to-tail fashion via the N-terminal methionine residue. LUBAC is a heteromeric complex composed of heme-oxidized iron-responsive element-binding protein 2 ubiquitin ligase-1L (HOIL-1L), HOIL-1L-interacting protein, and shank-associated RH domain-interacting protein (SHARPIN). The essential role of LUBAC-generated linear chains for activation of nuclear factor-κB (NF-κB) signaling was first described in the activation of tumor necrosis factor-α receptor signaling complex. A decade of research has identified additional pathways that use LUBAC for downstream signaling, including CD40 ligand and the IL-1ß receptor, as well as cytosolic pattern recognition receptors including nucleotide-binding oligomerization domain containing 2 (NOD2), retinoic acid-inducible gene 1 (RIG-1), and the NOD-like receptor family, pyrin domain containing 3 inflammasome (NLRP3). Even though the three components of the complex are required for full activation of NF-κB, the individual components of LUBAC regulate specific cell type- and stimuli-dependent effects. In humans, autosomal defects in LUBAC are associated with both autoinflammation and immunodeficiency, with additional disorders described in mice. Moreover, in the lung epithelium, HOIL-1L ubiquitinates target proteins independently of the other LUBAC components, adding another layer of complexity to the function and regulation of LUBAC. Although many advances have been made, the diverse functions of linear ubiquitin chains and the regulation of LUBAC are not yet completely understood. In this review, we discuss the various roles of linear ubiquitin chains and point to areas of study that would benefit from further investigation into LUBAC-mediated signaling pathways in lung pathophysiology.


Sujet(s)
Maladie , Santé , Ubiquitination , Animaux , Humains , Poumon/métabolisme , Proteasome endopeptidase complex/métabolisme , Ubiquitine/métabolisme
20.
Mol Cell Biol ; 35(23): 3962-73, 2015 Dec.
Article de Anglais | MEDLINE | ID: mdl-26370512

RÉSUMÉ

The c-Jun amino-terminal kinase (JNK) plays a role in inflammation, proliferation, apoptosis, and cell adhesion and cell migration by phosphorylating paxillin and ß-catenin. JNK phosphorylation downstream of AMP-activated protein kinase (AMPK) activation is required for high CO2 (hypercapnia)-induced Na,K-ATPase endocytosis in alveolar epithelial cells. Here, we provide evidence that during hypercapnia, JNK promotes the phosphorylation of LMO7b, a scaffolding protein, in vitro and in intact cells. LMO7b phosphorylation was blocked by exposing the cells to the JNK inhibitor SP600125 and by infecting cells with dominant-negative JNK or AMPK adenovirus. The knockdown of the endogenous LMO7b or overexpression of mutated LMO7b with alanine substitutions of five potential JNK phosphorylation sites (LMO7b-5SA) or only Ser-1295 rescued both LMO7b phosphorylation and the hypercapnia-induced Na,K-ATPase endocytosis. Moreover, high CO2 promoted the colocalization and interaction of LMO7b and the Na,K-ATPase α1 subunit at the plasma membrane, which were prevented by SP600125 or by transfecting cells with LMO7b-5SA. Collectively, our data suggest that hypercapnia leads to JNK-induced LMO7b phosphorylation at Ser-1295, which facilitates the interaction of LMO7b with Na,K-ATPase at the plasma membrane promoting the endocytosis of Na,K-ATPase in alveolar epithelial cells.


Sujet(s)
Endocytose , Protéines à homéodomaine/métabolisme , Hypercapnie/métabolisme , JNK Mitogen-Activated Protein Kinases/métabolisme , Sodium-Potassium-Exchanging ATPase/métabolisme , Facteurs de transcription/métabolisme , Séquence d'acides aminés , Animaux , Dioxyde de carbone/métabolisme , Lignée cellulaire , Activation enzymatique , Protéines à homéodomaine/analyse , Protéines à homéodomaine/génétique , Humains , JNK Mitogen-Activated Protein Kinases/analyse , Données de séquences moléculaires , Mutation , Phosphorylation , Cartes d'interactions protéiques , Rats , Sodium-Potassium-Exchanging ATPase/analyse , Facteurs de transcription/analyse , Facteurs de transcription/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...