Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 26
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Proc Natl Acad Sci U S A ; 121(11): e2307810121, 2024 Mar 12.
Article de Anglais | MEDLINE | ID: mdl-38437545

RÉSUMÉ

Treating pregnancy-related disorders is exceptionally challenging because the threat of maternal and/or fetal toxicity discourages the use of existing medications and hinders new drug development. One potential solution is the use of lipid nanoparticle (LNP) RNA therapies, given their proven efficacy, tolerability, and lack of fetal accumulation. Here, we describe LNPs for efficacious mRNA delivery to maternal organs in pregnant mice via several routes of administration. In the placenta, our lead LNP transfected trophoblasts, endothelial cells, and immune cells, with efficacy being structurally dependent on the ionizable lipid polyamine headgroup. Next, we show that LNP-induced maternal inflammatory responses affect mRNA expression in the maternal compartment and hinder neonatal development. Specifically, pro-inflammatory LNP structures and routes of administration curtailed efficacy in maternal lymphoid organs in an IL-1ß-dependent manner. Further, immunogenic LNPs provoked the infiltration of adaptive immune cells into the placenta and restricted pup growth after birth. Together, our results provide mechanism-based structural guidance on the design of potent LNPs for safe use during pregnancy.


Sujet(s)
Cellules endothéliales , Foetus , Liposomes , Nanoparticules , Femelle , Grossesse , Humains , Animaux , Souris , ARN messager/génétique , Prise en charge prénatale
2.
J Immunol ; 209(9): 1788-1795, 2022 11 01.
Article de Anglais | MEDLINE | ID: mdl-36113884

RÉSUMÉ

Chronic lung allograft dysfunction is the major barrier to long-term survival in lung transplant recipients. Evidence supports type 1 alloimmunity as the predominant response in acute/chronic lung rejection, but the immunoregulatory mechanisms remain incompletely understood. We studied the combinatorial F-box E3 ligase system: F-box protein 3 (FBXO3; proinflammatory) and F-box and leucine-rich repeat protein 2 (FBXL2; anti-inflammatory and regulates TNFR-associated factor [TRAF] protein). Using the mouse orthotopic lung transplant model, we evaluated allografts from BALB/c → C57BL/6 (acute rejection; day 10) and found significant induction of FBXO3 and diminished FBXL2 protein along with elevated T-bet, IFN-γ, and TRAF proteins 1-5 compared with isografts. In the acute model, treatment with costimulation blockade (MR1/CTLA4-Ig) resulted in attenuated FBXO3, preserved FBXL2, and substantially reduced T-bet, IFN-γ, and TRAFs 1-5, consistent with a key role for type 1 alloimmunity. Immunohistochemistry revealed significant changes in the FBXO3/FBXL2 balance in airway epithelia and infiltrating mononuclear cells during rejection compared with isografts or costimulation blockade-treated allografts. In the chronic lung rejection model, DBA/2J/C57BL/6F1 > DBA/2J (day 28), we observed persistently elevated FBXO3/FBXL2 balance and T-bet/IFN-γ protein and similar findings from lung transplant recipient lungs with chronic lung allograft dysfunction versus controls. We hypothesized that FBXL2 regulated T-bet and found FBXL2 was sufficient to polyubiquitinate T-bet and coimmunoprecipitated with T-bet on pulldown experiments and vice versa in Jurkat cells. Transfection with FBXL2 diminished T-bet protein in a dose-dependent manner in mouse lung epithelial cells. In testing type 1 cytokines, TNF-α was found to negatively regulate FBXL2 protein and mRNA levels. Together, our findings show the combinatorial E3 ligase FBXO3/FBXL2 system plays a role in the regulation of T-bet through FBXL2, with negative cross-regulation of TNF-α on FBXL2 during lung allograft rejection.


Sujet(s)
Protéines F-box , Animaux , Souris , Abatacept , Allogreffes , Cytokines/métabolisme , Modèles animaux de maladie humaine , Protéines F-box/génétique , Protéines F-box/métabolisme , Rejet du greffon , Poumon/métabolisme , Souris de lignée BALB C , Souris de lignée C57BL , Souris de lignée DBA , ARN messager , Protéines et peptides associés aux récepteurs des facteurs de nécrose tumorale/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Ubiquitin-protein ligases/métabolisme
3.
JCI Insight ; 5(2)2020 01 30.
Article de Anglais | MEDLINE | ID: mdl-31996486

RÉSUMÉ

Our integrative genomic and functional analysis identified transforming acidic coiled-coil-containing protein 2 (TACC2) as a chronic obstructive pulmonary disease (COPD) candidate gene. Here, we found that smokers with COPD exhibit a marked decrease in lung TACC2 protein levels relative to smokers without COPD. Single cell RNA sequencing reveals that TACC2 is expressed primarily in lung epithelial cells in normal human lungs. Furthermore, suppression of TACC2 expression impairs the efficiency of homologous recombination repair and augments spontaneous and cigarette smoke extract-induced (CSE-induced) DNA damage and cytotoxicity in immortalized human bronchial epithelial cells. By contrast, enforced expression of TACC2 attenuates the CSE effects. We also found that CSE enhances TACC2 degradation via the ubiquitin-proteasome system mediated by the ubiquitin E3 ligase subunit, F box L7. Furthermore, cellularly expressed TACC2 proteins harboring naturally occurring mutations exhibited altered protein lifespan coupled with modified DNA damage repair and cytotoxic responses. CS triggers emphysematous changes accompanied by accumulated DNA damage, apoptosis of alveolar epithelia, and lung inflammation in Tacc2-/- compared with Tacc2+/+ mice. Our results suggest that CS destabilizes TACC2 protein in lung epithelia by the ubiquitin proteasome system, leading to subsequent DNA damage, cytotoxicity, and emphysema.


Sujet(s)
Emphysème pulmonaire/induit chimiquement , Emphysème pulmonaire/génétique , Fumée/effets indésirables , Fumer/effets indésirables , Protéines suppresseurs de tumeurs/effets des médicaments et des substances chimiques , Protéines suppresseurs de tumeurs/génétique , Protéines suppresseurs de tumeurs/métabolisme , Sujet âgé , Animaux , Apoptose/effets des médicaments et des substances chimiques , Protéines de transport , Lignée cellulaire , Altération de l'ADN/effets des médicaments et des substances chimiques , Réparation de l'ADN , Emphysème , Cellules épithéliales/métabolisme , Femelle , Humains , Poumon/métabolisme , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Adulte d'âge moyen , Mutagenèse dirigée , Proteasome endopeptidase complex/métabolisme , Nicotiana/effets indésirables , Transcriptome , Ubiquitine/métabolisme , Ubiquitin-protein ligases/métabolisme
4.
JCI Insight ; 4(19)2019 10 03.
Article de Anglais | MEDLINE | ID: mdl-31578312

RÉSUMÉ

Dysregulated proinflammatory cytokine release has been implicated in the pathogenesis of several life-threatening acute lung illnesses such as pneumonia, sepsis, and acute respiratory distress syndrome. Suppressors of cytokine signaling proteins, particularly SOCS2, have recently been described as antiinflammatory mediators. However, the regulation of SOCS2 protein has not been described. Here we describe a mechanism of SOCS2 regulation by the action of the ubiquitin E3 ligase KIAA0317. KIAA0317-mediated degradation of SOCS2 exacerbated inflammation in vitro, and depletion of KIAA0317 in vivo ameliorated pulmonary inflammation. KIAA0317-knockout mice exhibited resistance to LPS-induced pulmonary inflammation, while KIAA03017 reexpression mitigated this effect. We uncovered a small molecule inhibitor of KIAA0317 protein (BC-1365) that prevented SOCS2 degradation and attenuated LPS- and P. aeruginosa-induced lung inflammation in vivo. These studies show KIAA0317 to be a critical mediator of pulmonary inflammation through its degradation of SOCS2 and a potential candidate target for therapeutic inhibition.


Sujet(s)
Poumon/métabolisme , Pneumopathie infectieuse/métabolisme , Protéines SOCS/métabolisme , Ubiquitin-protein ligases/génétique , Ubiquitin-protein ligases/métabolisme , Adulte , Sujet âgé , Animaux , Cytokines/métabolisme , Femelle , Humains , Immunité innée , Poumon/anatomopathologie , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Adulte d'âge moyen , Simulation de docking moléculaire , Pneumopathie infectieuse/immunologie , Pneumopathie infectieuse/anatomopathologie , Liaison aux protéines , Transcriptome
5.
Respir Res ; 18(1): 131, 2017 06 28.
Article de Anglais | MEDLINE | ID: mdl-28659178

RÉSUMÉ

BACKGROUND: Azithromycin, an antibiotic used for multiple infectious disorders, exhibits anti-inflammatory effects, but the molecular basis for this activity is not well characterized. Azithromycin inhibits IL-1ß-mediated inflammation that is dependent, in part, on inflammasome activity. Here, we investigated the effects of azithromycin on the NACHT, LRR, and PYD domains-containing protein 3 (NALP3) protein, which is the sensing component of the NALP3 inflammasome, in human monocytes. METHODS: THP-1 cells were treated with azithromycin alone, LPS alone, or both. NALP3 and IL-1ß protein levels were determined by immunoblotting. NLRP3 gene (encoding NALP3) transcript levels were determined by quantitative qPCR. In order to measure NLRP3 transcript decay, actinomycin D was used to impair gene transcription. THP-1 Lucia cells which contain an NF-κB responsive luciferase element were used to assess NF-κB activity in response to azithromycin, LPS, and azithromycin/LPS by measuring luminescence. To confirm azithromycin's effects on NLRP3 mRNA and promoter activity conclusively, HEK cells were lipofected with luciferase reporter constructs harboring either the 5' untranslated region (UTR) of the NLRP3 gene which included the promoter, the 3' UTR of the gene, or an empty plasmid prior to treatment with azithromycin and/or LPS, and luminescence was measured. RESULTS: Azithromycin decreased IL-1ß levels and reduced NALP3 protein levels in LPS-stimulated THP-1 monocytes through a mechanism involving decreased mRNA stability of the NALP3 - coding NLRP3 gene transcript as well as by decreasing NF-κB activity. Azithromycin accelerated NLRP3 transcript decay confirmed by mRNA stability and 3'UTR luciferase reporter assays, and yet the antibiotic had no effect on NLRP3 promoter activity in cells containing a 5' UTR reporter. CONCLUSIONS: These studies provide a unique mechanism whereby azithromycin exerts immunomodulatory actions in monocytes by destabilizing mRNA levels for a key inflammasome component, NALP3, leading to decreased IL-1ß-mediated inflammation.


Sujet(s)
Azithromycine/pharmacologie , Inflammasomes/métabolisme , Monocytes/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , ARN messager/métabolisme , Antibactériens/pharmacologie , Antibactériens/usage thérapeutique , Azithromycine/usage thérapeutique , Lignée cellulaire , Cellules HEK293 , Humains , Inflammasomes/antagonistes et inhibiteurs , Inflammasomes/génétique , Inflammation/traitement médicamenteux , Inflammation/génétique , Inflammation/métabolisme , Monocytes/effets des médicaments et des substances chimiques , Facteur de transcription NF-kappa B/antagonistes et inhibiteurs , Facteur de transcription NF-kappa B/génétique , Facteur de transcription NF-kappa B/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/antagonistes et inhibiteurs , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Stabilité protéique/effets des médicaments et des substances chimiques , ARN messager/antagonistes et inhibiteurs , ARN messager/génétique
6.
J Infect Dis ; 214(10): 1520-1530, 2016 11 15.
Article de Anglais | MEDLINE | ID: mdl-27613775

RÉSUMÉ

BACKGROUND: Lung CD4+ T-cell depletion and dysfunction, CD8+ T-cell alveolitis, smoking, and poor control of human immunodeficiency virus (HIV) are features of HIV-associated chronic obstructive pulmonary disease (COPD), but these changes have not been evaluated in smokers at risk for COPD. We evaluated the impact of viral suppression following initiation of antiretroviral therapy (ART) on HIV-specific immunity and the balance of the CD4+ T-cell to CD8+ T-cell ratio in the lung. METHODS: Using flow cytometry, we assessed the T-cell immune response in lung and blood specimens obtained from 12 actively smoking HIV-positive patients before ART initiation and after ART-associated viral suppression. RESULTS: HIV suppression resulted in enhanced lung and systemic HIV-specific CD4+ T-cell immune responses without significant changes in CD8+ T-cell responses. We observed an increase in lung ratios of CD4+ T cells to CD8+ T cells and CD4+ T-cell frequencies, decreased CD8+ T-cell numbers, and resolution of CD8+ T-cell alveolitis after ART in 9 of 12 individuals. Viral suppression reduced Fas receptor and programmed death 1 expression in lung CD4+ T cells, correlating with enhanced effector function and reduced susceptibility to apoptosis. HIV suppression rescued peripheral but not lung HIV-specific CD4+ T-cell proliferation, resulting in augmented effector multifunction. DISCUSSION: Together, our results demonstrate that HIV suppression restores lung mucosal HIV-specific CD4+ T-cell multifunctional immunity and balance in the ratio of CD4+ T cells to CD8+ T cells, often resolving CD8+ T-cell alveolitis in active smokers. Peripheral expansion and redistribution of CD4+ T cells and increased resistance to apoptosis are 2 mechanisms contributing to immunologic improvement following viral suppression in patients at risk for HIV-associated COPD.


Sujet(s)
Antirétroviraux/usage thérapeutique , Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , Infections à VIH/traitement médicamenteux , Poumon/anatomopathologie , Broncho-pneumopathie chronique obstructive/prévention et contrôle , Fumer/effets indésirables , Adulte , Rapport CD4-CD8 , Femelle , Cytométrie en flux , Infections à VIH/complications , Humains , Poumon/immunologie , Mâle , Adulte d'âge moyen , Broncho-pneumopathie chronique obstructive/anatomopathologie , Résultat thérapeutique
7.
J Exp Med ; 213(6): 1029-46, 2016 05 30.
Article de Anglais | MEDLINE | ID: mdl-27162139

RÉSUMÉ

The E3 small ubiquitin-like modifier (SUMO) protein ligase protein inhibitor of activated STAT 4 (PIAS4) is a pivotal protein in regulating the TGFß pathway. In this study, we discovered a new protein isoform encoded by KIAA0317, termed fibrosis-inducing E3 ligase 1 (FIEL1), which potently stimulates the TGFß signaling pathway through the site-specific ubiquitination of PIAS4. FIEL1 targets PIAS4 using a double locking mechanism that is facilitated by the kinases PKCζ and GSK3ß. Specifically, PKCζ phosphorylation of PIAS4 and GSK3ß phosphorylation of FIEL1 are both essential for the degradation of PIAS4. FIEL1 protein is highly expressed in lung tissues from patients with idiopathic pulmonary fibrosis (IPF), whereas PIAS4 protein levels are significantly reduced. FIEL1 overexpression significantly increases fibrosis in a bleomycin murine model, whereas FIEL1 knockdown attenuates fibrotic conditions. Further, we developed a first-in-class small molecule inhibitor toward FIEL1 that is highly effective in ameliorating fibrosis in mice. This study provides a basis for IPF therapeutic intervention by modulating PIAS4 protein abundance.


Sujet(s)
Poumon/métabolisme , Inhibiteurs de STAT activés/métabolisme , Fibrose pulmonaire/métabolisme , Ubiquitin-protein ligases/métabolisme , Ubiquitination , Animaux , Bléomycine/effets indésirables , Bléomycine/pharmacologie , Lignée cellulaire , Modèles animaux de maladie humaine , Glycogen synthase kinase 3 beta/génétique , Glycogen synthase kinase 3 beta/métabolisme , Humains , Isoenzymes/génétique , Isoenzymes/métabolisme , Poumon/anatomopathologie , Souris , Protéines liant le poly-adp-ribose , Inhibiteurs de STAT activés/génétique , Protéine kinase C/génétique , Protéine kinase C/métabolisme , Protein Kinase C-theta , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/génétique , Fibrose pulmonaire/anatomopathologie , Ubiquitin-protein ligases/génétique
8.
Sci Transl Med ; 7(295): 295ra109, 2015 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-26157031

RÉSUMÉ

Invading pathogens may trigger overactivation of the innate immune system, which results in the release of large amounts of proinflammatory cytokines (cytokine storm) and leads to the development of pulmonary edema, multiorgan failure, and shock. PIAS1 is a multifunctional and potent anti-inflammatory protein that negatively regulates several key inflammatory pathways such as Janus kinase (JAK)-signal transducer and activator of transcription (STAT) and nuclear factor κB (NF-κB). We discovered a ubiquitin E3 ligase, HECTD2, which ubiquitinated and mediated the degradation of PIAS1, thus increasing inflammation in an experimental pneumonia model. We found that GSK3ß phosphorylation of PIAS1 provided a phosphodegron for HECTD2 targeting. We also identified a mislocalized HECTD2 polymorphism, HECTD2(A19P), that was present in 8.5% of the population and functioned to reduce inflammation. This polymorphism prevented HECTD2/PIAS1 nuclear interaction, thus preventing PIAS1 degradation. The HECTD2(A19P) polymorphism was also protective toward acute respiratory distress syndrome (ARDS). We then developed a small-molecule inhibitor, BC-1382, that targeted HECTD2 and attenuated lipopolysaccharide (LPS)- and Pseudomonas aeruginosa-induced lung inflammation. These studies describe an unreported innate immune pathway and suggest that mutation or antagonism of the E3 ligase HECTD2 results in reduced severity of lung inflammation by selectively modulating the abundance of the anti-inflammatory protein PIAS1.


Sujet(s)
Immunité innée/physiologie , Inflammation/physiopathologie , Lésion pulmonaire/immunologie , Lésion pulmonaire/physiopathologie , Ubiquitin-protein ligases/physiologie , Animaux , Techniques de knock-down de gènes , Lésion pulmonaire/microbiologie , Souris , Phosphorylation , Inhibiteurs de STAT activés/génétique , Inhibiteurs de STAT activés/métabolisme , Pseudomonas/pathogénicité
9.
Am J Respir Cell Mol Biol ; 52(5): 622-33, 2015 May.
Article de Anglais | MEDLINE | ID: mdl-25286244

RÉSUMÉ

Acute cellular rejection is a known risk factor for the development of obliterative bronchiolitis, which limits the long-term survival of lung transplant recipients. However, the T cell effector mechanisms in both of these processes remain incompletely understood. Using the mouse orthotopic lung transplant model, we investigated whether C57BL/6 T-bet(-/-) recipients of major histocompatibility complex (MHC)-mismatched BALB/c lung grafts develop rejection pathology and allospecific cytokine responses that differ from wild-type mice. T-bet(-/-) recipients demonstrated vigorous allograft rejection at 10 days, characterized by neutrophilic inflammation and predominantly CD8(+) T cells producing allospecific IL-17 and/or IFN-γ, in contrast to IFN-γ-dominant responses in WT mice. CD4(+) T cells produced IL-17 but not IFN-γ responses in T-bet(-/-) recipients, in contrast to WT controls. Costimulation blockade using anti-CD154 Ab significantly reduced allospecific CD8(+)IFN-γ(+) responses in both T-bet(-/-) and WT mice but had no attenuating effect on lung rejection pathology in T-bet(-/-) recipients or on the development of obliterative airway inflammation that occurred only in T-bet(-/-) recipients. However, neutralization of IL-17A significantly attenuated costimulation blockade-resistant rejection pathology and airway inflammation in T-bet(-/-) recipients. In addition, CXCL1 (neutrophil chemokine) was increased in T-bet(-/-) allografts, and IL-17 induced CXCL1 from mouse lung epithelial cells in vitro. Taken together, our data show that T-bet-deficient recipients of complete MHC-mismatched lung allografts develop costimulation blockade-resistant rejection characterized by neutrophilia and obliterative airway inflammation that is predominantly mediated by CD8(+)IL-17(+) T cells. Our data support T-bet-deficient mouse recipients of lung allografts as a viable animal model to study the immunopathogenesis of small airway injury in lung transplantation.


Sujet(s)
Lymphocytes T CD8+/métabolisme , Rejet du greffon/étiologie , Médiateurs de l'inflammation/métabolisme , Interleukine-17/métabolisme , Transplantation pulmonaire/effets indésirables , Poumon/métabolisme , Granulocytes neutrophiles/métabolisme , Pneumopathie infectieuse/étiologie , Protéines à domaine boîte-T/métabolisme , Maladie aigüe , Allogreffes , Animaux , Anticorps/pharmacologie , Ligand de CD40/immunologie , Ligand de CD40/métabolisme , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/immunologie , Chimiotaxie des leucocytes , Modèles animaux de maladie humaine , Rejet du greffon/immunologie , Rejet du greffon/métabolisme , Rejet du greffon/anatomopathologie , Rejet du greffon/prévention et contrôle , Histocompatibilité , Médiateurs de l'inflammation/immunologie , Interféron gamma/immunologie , Interféron gamma/métabolisme , Interleukine-17/immunologie , Poumon/effets des médicaments et des substances chimiques , Poumon/immunologie , Poumon/anatomopathologie , Souris de souche-129 , Souris de lignée BALB C , Souris de lignée C57BL , Souris transgéniques , Granulocytes neutrophiles/immunologie , Pneumopathie infectieuse/immunologie , Pneumopathie infectieuse/métabolisme , Pneumopathie infectieuse/anatomopathologie , Pneumopathie infectieuse/prévention et contrôle , Protéines à domaine boîte-T/déficit , Protéines à domaine boîte-T/génétique
10.
PLoS One ; 9(12): e115937, 2014.
Article de Anglais | MEDLINE | ID: mdl-25549252

RÉSUMÉ

Cyclic adenosine monophosphate (cAMP) is a crucial intracellular second messenger molecule that converts extracellular molecules to intracellular signal transduction pathways generating cell- and stimulus-specific effects. Importantly, specific phosphodiesterase (PDE) subtypes control the amplitude and duration of cAMP-induced physiological processes and are therefore a prominent pharmacological target currently used in a variety of fields. Here we tested the extracts from traditional Chinese medicine, Forsythia suspense seeds, which have been used for more than 2000 years to relieve respiratory symptoms. Using structural-functional analysis we found its major lignin, Forsynthin, acted as an immunosuppressant by inhibiting PDE4 in inflammatory and immune cell. Moreover, several novel, selective small molecule derivatives of Forsythin were tested in vitro and in murine models of viral and bacterial pneumonia, sepsis and cytokine-driven systemic inflammation. Thus, pharmacological targeting of PDE4 may be a promising strategy for immune-related disorders characterized by amplified host inflammatory response.


Sujet(s)
Forsythia/composition chimique , Lignine/pharmacologie , Inhibiteurs de la phosphodiestérase-4/isolement et purification , Animaux , Simulation numérique , Cyclic Nucleotide Phosphodiesterases, Type 4/métabolisme , Inflammation/traitement médicamenteux , Lignine/isolement et purification , Lésion pulmonaire/traitement médicamenteux , Souris , Souris de lignée C57BL , Inhibiteurs de la phosphodiestérase-4/composition chimique , Inhibiteurs de la phosphodiestérase-4/pharmacologie , Inhibiteurs de la phosphodiestérase-4/usage thérapeutique , Plantes médicinales/composition chimique , Choc septique/traitement médicamenteux , Relation structure-activité
11.
Am J Respir Crit Care Med ; 190(7): 744-55, 2014 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-25137293

RÉSUMÉ

RATIONALE: As overall survival improves, individuals with HIV infection become susceptible to other chronic diseases, including accelerated chronic obstructive pulmonary disease (COPD). OBJECTIVES: To determine whether individuals with HIV-associated COPD exhibit dysregulated lung mucosal T-cell immunity compared with control subjects. METHODS: Using flow cytometry, we evaluated peripheral blood and lung mucosal T-cell immunity in 14 HIV(+)COPD(+), 13 HIV(+)COPD(-), and 7 HIV(-)COPD(+) individuals. MEASUREMENTS AND MAIN RESULTS: HIV(+)COPD(+) individuals demonstrated profound CD4(+) T-cell depletion with reduced CD4/CD8 T-cell ratios in bronchoalveolar lavage-derived lung mononuclear cells, not observed in peripheral blood mononuclear cells, and diminished CD4(+) T cell absolute numbers, compared with control subjects. Furthermore, HIV(+)COPD(+) individuals demonstrated decreased pulmonary HIV-specific and staphylococcal enterotoxin B-reactive CD4(+) memory responses, including loss of multifunctionality, compared with HIV(+)COPD(-) control subjects. In contrast, lung mucosal HIV-specific CD8(+) T-cell responses were preserved. Lung CD4(+) T cells from HIV(+)COPD(+) individuals expressed increased surface Fas death receptor (CD95) and programmed death-1, but similar bronchoalveolar lavage viral loads as control subjects. However, programmed death-1 expression inversely correlated with HIV-specific lung CD4(+)IFN-γ(+) T-cell responses, suggesting functional exhaustion. Moreover, lung CD4(+) T cells from HIV(+)COPD(+) patients demonstrated increased basal and HIV antigen-induced expression of the early apoptosis marker annexin V compared with control subjects, which was significantly attenuated with anti-Fas blockade. Lastly, lung mucosal, but not blood, CD4(+)/CD8(+) ratios from HIV(+) patients significantly correlated with the FEV1, but not in HIV(-)COPD(+) patients. CONCLUSIONS: Together, our results provide evidence for profound lung mucosal CD4(+) T-cell depletion via a Fas-dependent activation-induced cell death mechanism, along with impaired HIV-specific CD4(+) immunity as immunologic features of HIV-associated COPD.


Sujet(s)
Apoptose/immunologie , Lymphocytes T CD4+/immunologie , Infections à VIH/immunologie , Poumon/immunologie , Broncho-pneumopathie chronique obstructive/immunologie , Liquide de lavage bronchoalvéolaire/immunologie , Lymphocytes T CD8+/immunologie , Mort cellulaire , Études de cohortes , Femelle , Cytométrie en flux/méthodes , Infections à VIH/complications , Humains , Immunité muqueuse/immunologie , Agranulocytes/immunologie , Études longitudinales , Mâle , Adulte d'âge moyen , Broncho-pneumopathie chronique obstructive/complications , Charge virale/immunologie , Antigènes CD95/immunologie
12.
Cell Rep ; 7(2): 476-487, 2014 Apr 24.
Article de Anglais | MEDLINE | ID: mdl-24703837

RÉSUMÉ

Acute lung injury (ALI) is linked to mitochondrial injury, resulting in impaired cellular oxygen utilization; however, it is unknown how these events are linked on the molecular level. Cardiolipin, a mitochondrial-specific lipid, is generated by cardiolipin synthase (CLS1). Here, we show that S. aureus activates a ubiquitin E3 ligase component, Fbxo15, that is sufficient to mediate proteasomal degradation of CLS1 in epithelia, resulting in decreased cardiolipin availability and disrupted mitochondrial function. CLS1 is destabilized by the phosphatase and tensin homolog (PTEN)-induced putative kinase 1 (PINK1), which binds CLS1 to phosphorylate and regulates CLS1 disposal. Like Fbxo15, PINK1 interacts with and regulates levels of CLS1 through a mechanism dependent upon Thr219. S. aureus infection upregulates this Fbxo15-PINK1 pathway to impair mitochondrial integrity, and Pink1 knockout mice are less prone to S. aureus-induced ALI. Thus, ALI-associated disruption of cellular bioenergetics involves bioeffectors that utilize a phosphodegron to elicit ubiquitin-mediated disposal of a key mitochondrial enzyme.


Sujet(s)
Antigène CD86/métabolisme , Protéines F-box/métabolisme , Mitochondries/métabolisme , Pneumopathie infectieuse/métabolisme , Protein kinases/métabolisme , Adolescent , Adulte , Animaux , Antigène CD86/génétique , Études cas-témoins , Lignée cellulaire , Cellules cultivées , Enfant , Stabilité enzymatique , Protéines F-box/génétique , Femelle , Humains , Mâle , Souris , Souris de lignée C57BL , Adulte d'âge moyen , Protein kinases/génétique , Sous-unités de protéines/génétique , Sous-unités de protéines/métabolisme
13.
Am J Physiol Lung Cell Mol Physiol ; 306(6): L534-42, 2014 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-24441869

RÉSUMÉ

MicroRNAs are small noncoding RNAs that inhibit protein expression. We have previously shown that the inhibition of the microRNA let-7d in epithelial cells caused changes consistent with epithelial-to-mesenchymal transition (EMT) both in vitro and in vivo. The aim of this study was to determine whether the introduction of let-7d into fibroblasts alters their mesenchymal properties. Transfection of primary fibroblasts with let-7d caused a decrease in expression of the mesenchymal markers α-smooth muscle actin, N-cadherin, fibroblast-specific protein-1, and fibronectin, as well as an increase in the epithelial markers tight junction protein-1 and keratin 19. Phenotypic changes were also present, including a delay in wound healing, reduced motility, and proliferation of fibroblasts following transfection. In addition, we examined the effects of transfection on fibroblast responsiveness to TGF-ß, an important factor in many fibrotic processes such as lung fibrosis and found that let-7d transfection significantly attenuated high-mobility group-A2 protein induction by TGF-ß. Our results indicate that administration of the epithelial microRNA let-7d can significantly alter the phenotype of primary fibroblasts.


Sujet(s)
Transition épithélio-mésenchymateuse , Fibroblastes/cytologie , Poumon/métabolisme , microARN/génétique , Myofibroblastes/métabolisme , Fibrose pulmonaire/métabolisme , Facteur de croissance transformant bêta/métabolisme , Actines/métabolisme , Cadhérines/métabolisme , Protéines de liaison au calcium/métabolisme , Mouvement cellulaire/génétique , Prolifération cellulaire , Cellules cultivées , Fibroblastes/métabolisme , Fibronectines/métabolisme , Protéine HMGA2/métabolisme , Protéine HMG2/métabolisme , Humains , Fibrose pulmonaire idiopathique/métabolisme , Fibrose pulmonaire idiopathique/anatomopathologie , Kératine-19/métabolisme , Poumon/cytologie , Alvéoles pulmonaires/métabolisme , Fibrose pulmonaire/génétique , Protéine S100A4 liant le calcium , Facteurs de transcription de la famille Snail , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Transfection , Cicatrisation de plaie/génétique , Protéine-1 de la zonula occludens/métabolisme
14.
J Immunol ; 191(10): 5247-55, 2013 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-24123678

RÉSUMÉ

Cytokine-driven inflammation underlies the pathobiology of a wide array of infectious and immune-related disorders. The TNFR-associated factor (TRAF) proteins have a vital role in innate immunity by conveying signals from cell surface receptors to elicit transcriptional activation of genes encoding proinflammatory cytokines. We discovered that a ubiquitin E3 ligase F box component, termed Fbxo3, potently stimulates cytokine secretion from human inflammatory cells by mediating the degradation of the TRAF inhibitory protein, Fbxl2. Analysis of the Fbxo3 C-terminal structure revealed that the bacterial-like ApaG molecular signature was indispensible for mediating Fbxl2 disposal and stimulating cytokine secretion. By targeting this ApaG motif, we developed a highly unique, selective genus of small-molecule Fbxo3 inhibitors that by reducing TRAF protein levels, potently inhibited cytokine release from human blood mononuclear cells. The Fbxo3 inhibitors effectively lessened the severity of viral pneumonia, septic shock, colitis, and cytokine-driven inflammation systemically in murine models. Thus, pharmacological targeting of Fbxo3 might be a promising strategy for immune-related disorders characterized by a heightened host inflammatory response.


Sujet(s)
Benzylamines/usage thérapeutique , Protéines F-box/métabolisme , Inflammation/traitement médicamenteux , Pyridines/usage thérapeutique , Protéines et peptides associés aux récepteurs des facteurs de nécrose tumorale/métabolisme , Animaux , Benzylamines/pharmacologie , Lignée cellulaire , Colite/traitement médicamenteux , Cytokines/biosynthèse , Cytokines/métabolisme , Protéines F-box/antagonistes et inhibiteurs , Inflammation/immunologie , Sous-type H1N1 du virus de la grippe A/effets des médicaments et des substances chimiques , Agranulocytes/effets des médicaments et des substances chimiques , Agranulocytes/immunologie , Lésion pulmonaire/traitement médicamenteux , Lésion pulmonaire/virologie , Mâle , Souris , Souris de lignée C57BL , Infections à Orthomyxoviridae/traitement médicamenteux , Pyridines/pharmacologie , Protéines et peptides associés aux récepteurs des facteurs de nécrose tumorale/antagonistes et inhibiteurs , Ubiquitin-protein ligases/métabolisme
15.
Cell Signal ; 25(10): 2047-59, 2013 Oct.
Article de Anglais | MEDLINE | ID: mdl-23707388

RÉSUMÉ

Cell cycle progression through its regulatory control by changes in intracellular Ca(2+) levels at the G1/S transition mediates cellular proliferation and viability. Ca(2+)/CaM-dependent kinase 1 (CaMKI) appears critical in regulating the assembly of the cyclin D1/cdk4 complex essential for G1 progression, but how this occurs is unknown. Cyclin D1/cdk4 assembly in the early G1 phase is also regulated via binding to p27. Here, we show that a ubiquitin E3 ligase component, F-box protein Fbxl12, mediates CaMKI degradation via a proteasome-directed pathway leading to disruption of cyclin D1/cdk4 complex assembly and resultant G1 arrest in lung epithelia. We also demonstrate that i) CaMKI phosphorylates p27 at Thr(157) and Thr(198) in human cells and at Thr(170) and Thr(197) in mouse cells to modulate its subcellular localization; ii) Fbxl12-induced CaMKI degradation attenuates p27 phosphorylation at these sites in early G1 and iii) activation of CaMKI during G1 transition followed by p27 phosphorylation appears to be upstream to other p27 phosphorylation events, an effect abrogated by Fbxl12 overexpression. Lastly, known inducers of G1 arrest significantly increase Fbxl12 levels in cells. Thus, Fbxl12 may be a previously uncharacterized, functional growth inhibitor regulating cell cycle progression that might be used for mechanism-based therapy.


Sujet(s)
Signalisation calcique/génétique , Calcium-Calmodulin-Dependent Protein Kinases/métabolisme , Cycline D1/métabolisme , Protéines F-box/métabolisme , Points de contrôle de la phase G1 du cycle cellulaire/génétique , Animaux , Calcium-Calmodulin-Dependent Protein Kinases/génétique , Points de contrôle du cycle cellulaire/génétique , Cycline D1/génétique , Kinase-4 cycline-dépendante/génétique , Kinase-4 cycline-dépendante/métabolisme , Protéines F-box/génétique , Régulation de l'expression des gènes , Humains , Souris , Proteasome endopeptidase complex/génétique , Proteasome endopeptidase complex/métabolisme , Protéolyse
16.
Nat Immunol ; 14(5): 470-9, 2013 May.
Article de Anglais | MEDLINE | ID: mdl-23542741

RÉSUMÉ

Uncontrolled activation of tumor necrosis factor receptor-associated factor (TRAF) proteins may result in profound tissue injury by linking surface signals to cytokine release. Here we show that a ubiquitin E3 ligase component, Fbxo3, potently stimulates cytokine secretion from human inflammatory cells by destabilizing a sentinel TRAF inhibitor, Fbxl2. Fbxo3 and TRAF protein in circulation positively correlated with cytokine responses in subjects with sepsis, and we identified a polymorphism in human Fbxo3, with one variant being hypofunctional. A small-molecule inhibitor targeting Fbxo3 was sufficient to lessen severity of cytokine-driven inflammation in several mouse disease models. These studies identified a pathway of innate immunity that may be useful to detect subjects with altered immune responses during critical illness or provide a basis for therapeutic intervention targeting TRAF protein abundance.


Sujet(s)
Protéines F-box/métabolisme , Infections à Pseudomonas/immunologie , Pseudomonas aeruginosa/immunologie , Sepsie/immunologie , Protéines et peptides associés aux récepteurs des facteurs de nécrose tumorale/métabolisme , Animaux , Caecum/immunologie , Caecum/chirurgie , Lignée cellulaire , Cytokines/métabolisme , Modèles animaux de maladie humaine , Motifs F-box/génétique , Protéines F-box/génétique , Humains , Immunomodulation , Inflammation/génétique , Souris , Souris de lignée C57BL , Polymorphisme génétique , Stabilité protéique , Infections à Pseudomonas/génétique , Pseudomonas aeruginosa/génétique , Petit ARN interférent/génétique , Sepsie/génétique , Transgènes/génétique
17.
FASEB J ; 27(7): 2611-9, 2013 Jul.
Article de Anglais | MEDLINE | ID: mdl-23512198

RÉSUMÉ

Rac1, a member of the Rho family of GTPases, regulates diverse cellular functions, including cytoskeleton reorganization and cell migration. F-box proteins are major subunits within the Skp1-Cul1-F-box (SCF) E3 ubiquitin ligases that recognize specific substrates for ubiquitination. The role of F-box proteins in regulating Rac1 stability has not been studied. Mouse lung epithelial (MLE12) cells were used to investigate Rac1 stability and cell migration. Screening of an F-box protein library and in vitro ubiquitination assays identified FBXL19, a relatively new member of the F-box protein family that targets Rac1 for its polyubiquitination and proteasomal degradation. Overexpression of FBXL19 decreased both Rac1 active and inactive forms and significantly reduced cellular migration. Protein kinase AKT-mediated phosphorylation of Rac1 at serine(71) was essential for FBXL19-mediated Rac1 ubiquitination and depletion. Lysine(166) within Rac1 was identified as a polyubiquitination acceptor site. Rac1(S71A) and Rac1(K166R) mutant proteins were resistant to FBXL19-mediated ubiquitination and degradation. Further, ectopically expressed FBXL19 reduced cell migration in Rac1-overexpressing cells (P<0.01, Rac1 cells vs. FBXL19+Rac1 cells), but not in Rac1 lysine(166) mutant-overexpressing cells. FBXL19 diminished formation of the migratory leading edge. Thus, SCF(FBXL19) targets Rac1 for its disposal, a process regulated by AKT. These findings provide the first evidence of an F-box protein targeting a small G protein for ubiquitination and degradation to modulate cell migration.


Sujet(s)
Mouvement cellulaire , Protéines de liaison à l'ADN/métabolisme , Protéines F-box/métabolisme , Neuropeptides/métabolisme , Protéines G rac/métabolisme , Animaux , Lignée cellulaire , Protéines de liaison à l'ADN/génétique , Protéines F-box/génétique , Humains , Immunotransfert , Souris , Mutation , Neuropeptides/génétique , Phosphorylation , Polyubiquitine/métabolisme , Proteasome endopeptidase complex/métabolisme , Liaison aux protéines , Protéolyse , Protéines proto-oncogènes c-akt/génétique , Protéines proto-oncogènes c-akt/métabolisme , Interférence par ARN , Sérine/génétique , Sérine/métabolisme , Ubiquitination , Protéines G rac/génétique , Protéine G rac1
18.
Cell Cycle ; 12(4): 663-73, 2013 Feb 15.
Article de Anglais | MEDLINE | ID: mdl-23370391

RÉSUMÉ

Aurora B kinase is an integral regulator of cytokinesis as it stabilizes the intercellular canal within the midbody to ensure proper chromosomal segregation during cell division. Here we identified an E3 ligase subunit, F box protein FBXL2, that by recognizing a calmodulin binding signature within Aurora B, ubiquitinates and removes the kinase from the midbody. Calmodulin, by competing with the F box protein for access to the calmodulin binding signature, protected Aurora B from FBXL2. Calmodulin co-localized with Aurora B on the midbody, preserved Aurora B levels in cells, and stabilized intercellular canals during delayed abscission. Genetic or pharmaceutical depletion of endogenous calmodulin significantly reduced Aurora B protein levels at the midbody resulting in tetraploidy and multi-spindle formation. The calmodulin inhibitor, calmidazolium, reduced Aurora B protein levels resulting in tetraploidy, mitotic arrest, and apoptosis of tumorigenic cells and profoundly inhibiting tumor formation in athymic nude mice. These observations indicate molecular interplay between Aurora B and calmodulin in telophase and suggest that calmodulin acts as a checkpoint sensor for chromosomal segregation errors during mitosis.


Sujet(s)
Calmoduline/génétique , Cytocinèse/génétique , Protéines F-box/génétique , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Protein-Serine-Threonine Kinases/génétique , Télophase/génétique , Animaux , Apoptose/effets des médicaments et des substances chimiques , Aurora kinase B , Aurora kinases , Sites de fixation , Calmoduline/métabolisme , Lignée cellulaire tumorale , Ségrégation des chromosomes/effets des médicaments et des substances chimiques , Cytocinèse/effets des médicaments et des substances chimiques , Antienzymes/pharmacologie , Cellules épithéliales/cytologie , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme , Protéines F-box/métabolisme , Humains , Imidazoles/pharmacologie , Souris , Souris nude , Transplantation tumorale , Liaison aux protéines , Protein-Serine-Threonine Kinases/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Télophase/effets des médicaments et des substances chimiques , Charge tumorale/effets des médicaments et des substances chimiques
19.
J Biol Chem ; 288(9): 6306-16, 2013 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-23319590

RÉSUMÉ

Histone acetyltransferase binding to origin recognition complex (HBO1) plays a crucial role in DNA replication licensing and cell proliferation, yet its molecular regulation in cells is relatively unknown. Here an uncharacterized protein, Fbxw15, directly interacts with HBO1, a labile protein (t½ = ∼3 h), to mediate its ubiquitination (Lys(338)) and degradation in the cytoplasm. Fbxw15-mediated HBO1 depletion required mitogen-activated protein kinase 1 (Mek1), which was sufficient to trigger HBO1 phosphorylation and degradation in cells. Mek1 ability to produce HBO1 degradation was blocked by Fbxw15 silencing. Lipopolysaccharide induced HBO1 degradation, an effect abrogated by Fbxw15 or Mek1 cellular depletion. Modulation of Fbxw15 levels was able to differentially regulate histone H3K14 acetylation and cellular proliferation by altering HBO1 levels. These studies authenticate Fbxw15 as a ubiquitin E3 ligase subunit that mediates endotoxin-induced HBO1 depletion in cells, thereby controlling cell replicative capacity.


Sujet(s)
Prolifération cellulaire , Protéines F-box/métabolisme , Histone acetyltransferases/métabolisme , Proteasome endopeptidase complex/métabolisme , Protéolyse , Ubiquitine/métabolisme , Acétylation , Animaux , Lignée cellulaire , Protéines F-box/génétique , Extinction de l'expression des gènes , Histone acetyltransferases/génétique , Histone/génétique , Histone/métabolisme , Humains , MAP Kinase Kinase 1/génétique , MAP Kinase Kinase 1/métabolisme , Souris , Proteasome endopeptidase complex/génétique , Ubiquitine/génétique , Ubiquitin-protein ligases/génétique , Ubiquitin-protein ligases/métabolisme
20.
Nat Immunol ; 13(7): 651-8, 2012 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-22660580

RÉSUMÉ

The ST2L receptor for interleukin 33 (IL-33) mediates pulmonary inflammation and immune system-related disorders, such as asthma and rheumatoid arthritis. At present, very little is known about the molecular regulation of ST2L expression. Here we found that FBXL19, an 'orphan' member of the Skp1-Cullin-F-box family of E3 ubiquitin ligases, selectively bound to ST2L to mediate its polyubiquitination and elimination in the proteasome. Degradation of ST2L involved phosphorylation of ST2L at Ser442 catalyzed by the kinase GSK3ß. Overexpression of FBXL19 abrogated the proapoptotic and inflammatory effects of IL-33 and lessened the severity of lung injury in mouse models of pneumonia. Our results suggest that modulation of the IL-33-ST2L axis by ubiquitin ligases might serve as a unique strategy for lessening pulmonary inflammation.


Sujet(s)
Protéines de liaison à l'ADN/immunologie , Protéines F-box/immunologie , Pneumopathie infectieuse/immunologie , Récepteurs aux interleukines/immunologie , Ubiquitin-protein ligases/métabolisme , Ubiquitination/immunologie , Animaux , Cellules cultivées , Protéines de liaison à l'ADN/métabolisme , Protéines F-box/métabolisme , Glycogen Synthase Kinase 3/métabolisme , Glycogen synthase kinase 3 beta , Protéine-1 analogue au récepteur de l'interleukin-1 , Souris , Souris de lignée C57BL , Phosphorylation , Pneumopathie infectieuse/anatomopathologie , Proteasome endopeptidase complex/métabolisme , Sérine/métabolisme , Indice de gravité de la maladie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...