Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 121(11): e2307810121, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38437545

ABSTRACT

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.


Subject(s)
Endothelial Cells , Fetus , Liposomes , Nanoparticles , Female , Pregnancy , Humans , Animals , Mice , RNA, Messenger/genetics , Prenatal Care
2.
J Immunol ; 209(9): 1788-1795, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36113884

ABSTRACT

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.


Subject(s)
F-Box Proteins , Animals , Mice , Abatacept , Allografts , Cytokines/metabolism , Disease Models, Animal , F-Box Proteins/genetics , F-Box Proteins/metabolism , Graft Rejection , Lung/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , RNA, Messenger , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin-Protein Ligases/metabolism
3.
JCI Insight ; 5(2)2020 01 30.
Article in English | MEDLINE | ID: mdl-31996486

ABSTRACT

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.


Subject(s)
Pulmonary Emphysema/chemically induced , Pulmonary Emphysema/genetics , Smoke/adverse effects , Smoking/adverse effects , Tumor Suppressor Proteins/drug effects , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Aged , Animals , Apoptosis/drug effects , Carrier Proteins , Cell Line , DNA Damage/drug effects , DNA Repair , Emphysema , Epithelial Cells/metabolism , Female , Humans , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Mutagenesis, Site-Directed , Proteasome Endopeptidase Complex/metabolism , Nicotiana/adverse effects , Transcriptome , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism
4.
JCI Insight ; 4(19)2019 10 03.
Article in English | MEDLINE | ID: mdl-31578312

ABSTRACT

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.


Subject(s)
Lung/metabolism , Pneumonia/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Adult , Aged , Animals , Cytokines/metabolism , Female , Humans , Immunity, Innate , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Molecular Docking Simulation , Pneumonia/immunology , Pneumonia/pathology , Protein Binding , Transcriptome
5.
Respir Res ; 18(1): 131, 2017 06 28.
Article in English | MEDLINE | ID: mdl-28659178

ABSTRACT

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.


Subject(s)
Azithromycin/pharmacology , Inflammasomes/metabolism , Monocytes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , RNA, Messenger/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Azithromycin/therapeutic use , Cell Line , HEK293 Cells , Humans , Inflammasomes/antagonists & inhibitors , Inflammasomes/genetics , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Monocytes/drug effects , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Protein Stability/drug effects , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics
6.
J Infect Dis ; 214(10): 1520-1530, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27613775

ABSTRACT

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.


Subject(s)
Anti-Retroviral Agents/therapeutic use , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , HIV Infections/drug therapy , Lung/pathology , Pulmonary Disease, Chronic Obstructive/prevention & control , Smoking/adverse effects , Adult , CD4-CD8 Ratio , Female , Flow Cytometry , HIV Infections/complications , Humans , Lung/immunology , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/pathology , Treatment Outcome
7.
J Exp Med ; 213(6): 1029-46, 2016 05 30.
Article in English | MEDLINE | ID: mdl-27162139

ABSTRACT

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.


Subject(s)
Lung/metabolism , Protein Inhibitors of Activated STAT/metabolism , Pulmonary Fibrosis/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Animals , Bleomycin/adverse effects , Bleomycin/pharmacology , Cell Line , Disease Models, Animal , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Lung/pathology , Mice , Poly-ADP-Ribose Binding Proteins , Protein Inhibitors of Activated STAT/genetics , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Kinase C-theta , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Ubiquitin-Protein Ligases/genetics
8.
Sci Transl Med ; 7(295): 295ra109, 2015 Jul 08.
Article in English | MEDLINE | ID: mdl-26157031

ABSTRACT

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.


Subject(s)
Immunity, Innate/physiology , Inflammation/physiopathology , Lung Injury/immunology , Lung Injury/physiopathology , Ubiquitin-Protein Ligases/physiology , Animals , Gene Knockdown Techniques , Lung Injury/microbiology , Mice , Phosphorylation , Protein Inhibitors of Activated STAT/genetics , Protein Inhibitors of Activated STAT/metabolism , Pseudomonas/pathogenicity
9.
Am J Respir Cell Mol Biol ; 52(5): 622-33, 2015 May.
Article in English | MEDLINE | ID: mdl-25286244

ABSTRACT

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.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Graft Rejection/etiology , Inflammation Mediators/metabolism , Interleukin-17/metabolism , Lung Transplantation/adverse effects , Lung/metabolism , Neutrophils/metabolism , Pneumonia/etiology , T-Box Domain Proteins/metabolism , Acute Disease , Allografts , Animals , Antibodies/pharmacology , CD40 Ligand/immunology , CD40 Ligand/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Chemotaxis, Leukocyte , Disease Models, Animal , Graft Rejection/immunology , Graft Rejection/metabolism , Graft Rejection/pathology , Graft Rejection/prevention & control , Histocompatibility , Inflammation Mediators/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-17/immunology , Lung/drug effects , Lung/immunology , Lung/pathology , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neutrophils/immunology , Pneumonia/immunology , Pneumonia/metabolism , Pneumonia/pathology , Pneumonia/prevention & control , T-Box Domain Proteins/deficiency , T-Box Domain Proteins/genetics
10.
PLoS One ; 9(12): e115937, 2014.
Article in English | MEDLINE | ID: mdl-25549252

ABSTRACT

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.


Subject(s)
Forsythia/chemistry , Lignin/pharmacology , Phosphodiesterase 4 Inhibitors/isolation & purification , Animals , Computer Simulation , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Inflammation/drug therapy , Lignin/isolation & purification , Lung Injury/drug therapy , Mice , Mice, Inbred C57BL , Phosphodiesterase 4 Inhibitors/chemistry , Phosphodiesterase 4 Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/therapeutic use , Plants, Medicinal/chemistry , Shock, Septic/drug therapy , Structure-Activity Relationship
11.
Am J Respir Crit Care Med ; 190(7): 744-55, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25137293

ABSTRACT

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.


Subject(s)
Apoptosis/immunology , CD4-Positive T-Lymphocytes/immunology , HIV Infections/immunology , Lung/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Bronchoalveolar Lavage Fluid/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Death , Cohort Studies , Female , Flow Cytometry/methods , HIV Infections/complications , Humans , Immunity, Mucosal/immunology , Leukocytes, Mononuclear/immunology , Longitudinal Studies , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/complications , Viral Load/immunology , fas Receptor/immunology
12.
Cell Rep ; 7(2): 476-487, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24703837

ABSTRACT

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.


Subject(s)
B7-2 Antigen/metabolism , F-Box Proteins/metabolism , Mitochondria/metabolism , Pneumonia/metabolism , Protein Kinases/metabolism , Adolescent , Adult , Animals , B7-2 Antigen/genetics , Case-Control Studies , Cell Line , Cells, Cultured , Child , Enzyme Stability , F-Box Proteins/genetics , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Protein Kinases/genetics , Protein Subunits/genetics , Protein Subunits/metabolism
13.
Am J Physiol Lung Cell Mol Physiol ; 306(6): L534-42, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24441869

ABSTRACT

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.


Subject(s)
Epithelial-Mesenchymal Transition , Fibroblasts/cytology , Lung/metabolism , MicroRNAs/genetics , Myofibroblasts/metabolism , Pulmonary Fibrosis/metabolism , Transforming Growth Factor beta/metabolism , Actins/metabolism , Cadherins/metabolism , Calcium-Binding Proteins/metabolism , Cell Movement/genetics , Cell Proliferation , Cells, Cultured , Fibroblasts/metabolism , Fibronectins/metabolism , HMGA2 Protein/metabolism , HMGB2 Protein/metabolism , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Keratin-19/metabolism , Lung/cytology , Pulmonary Alveoli/metabolism , Pulmonary Fibrosis/genetics , S100 Calcium-Binding Protein A4 , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection , Wound Healing/genetics , Zonula Occludens-1 Protein/metabolism
14.
J Immunol ; 191(10): 5247-55, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24123678

ABSTRACT

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.


Subject(s)
Benzylamines/therapeutic use , F-Box Proteins/metabolism , Inflammation/drug therapy , Pyridines/therapeutic use , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism , Animals , Benzylamines/pharmacology , Cell Line , Colitis/drug therapy , Cytokines/biosynthesis , Cytokines/metabolism , F-Box Proteins/antagonists & inhibitors , Inflammation/immunology , Influenza A Virus, H1N1 Subtype/drug effects , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Lung Injury/drug therapy , Lung Injury/virology , Male , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/drug therapy , Pyridines/pharmacology , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/antagonists & inhibitors , Ubiquitin-Protein Ligases/metabolism
15.
Cell Signal ; 25(10): 2047-59, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23707388

ABSTRACT

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.


Subject(s)
Calcium Signaling/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cyclin D1/metabolism , F-Box Proteins/metabolism , G1 Phase Cell Cycle Checkpoints/genetics , Animals , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cell Cycle Checkpoints/genetics , Cyclin D1/genetics , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , F-Box Proteins/genetics , Gene Expression Regulation , Humans , Mice , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Proteolysis
16.
Nat Immunol ; 14(5): 470-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23542741

ABSTRACT

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.


Subject(s)
F-Box Proteins/metabolism , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Sepsis/immunology , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism , Animals , Cecum/immunology , Cecum/surgery , Cell Line , Cytokines/metabolism , Disease Models, Animal , F-Box Motifs/genetics , F-Box Proteins/genetics , Humans , Immunomodulation , Inflammation/genetics , Mice , Mice, Inbred C57BL , Polymorphism, Genetic , Protein Stability , Pseudomonas Infections/genetics , Pseudomonas aeruginosa/genetics , RNA, Small Interfering/genetics , Sepsis/genetics , Transgenes/genetics
17.
FASEB J ; 27(7): 2611-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23512198

ABSTRACT

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.


Subject(s)
Cell Movement , DNA-Binding Proteins/metabolism , F-Box Proteins/metabolism , Neuropeptides/metabolism , rac GTP-Binding Proteins/metabolism , Animals , Cell Line , DNA-Binding Proteins/genetics , F-Box Proteins/genetics , Humans , Immunoblotting , Mice , Mutation , Neuropeptides/genetics , Phosphorylation , Polyubiquitin/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Proteolysis , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Serine/genetics , Serine/metabolism , Ubiquitination , rac GTP-Binding Proteins/genetics , rac1 GTP-Binding Protein
18.
Cell Cycle ; 12(4): 663-73, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23370391

ABSTRACT

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.


Subject(s)
Calmodulin/genetics , Cytokinesis/genetics , F-Box Proteins/genetics , Gene Expression Regulation/drug effects , Protein Serine-Threonine Kinases/genetics , Telophase/genetics , Animals , Apoptosis/drug effects , Aurora Kinase B , Aurora Kinases , Binding Sites , Calmodulin/metabolism , Cell Line, Tumor , Chromosome Segregation/drug effects , Cytokinesis/drug effects , Enzyme Inhibitors/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , F-Box Proteins/metabolism , Humans , Imidazoles/pharmacology , Mice , Mice, Nude , Neoplasm Transplantation , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Telophase/drug effects , Tumor Burden/drug effects
19.
J Biol Chem ; 288(9): 6306-16, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23319590

ABSTRACT

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.


Subject(s)
Cell Proliferation , F-Box Proteins/metabolism , Histone Acetyltransferases/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitin/metabolism , Acetylation , Animals , Cell Line , F-Box Proteins/genetics , Gene Silencing , Histone Acetyltransferases/genetics , Histones/genetics , Histones/metabolism , Humans , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , Mice , Proteasome Endopeptidase Complex/genetics , Ubiquitin/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
20.
Nat Immunol ; 13(7): 651-8, 2012 Jun 03.
Article in English | MEDLINE | ID: mdl-22660580

ABSTRACT

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.


Subject(s)
DNA-Binding Proteins/immunology , F-Box Proteins/immunology , Pneumonia/immunology , Receptors, Interleukin/immunology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/immunology , Animals , Cells, Cultured , DNA-Binding Proteins/metabolism , F-Box Proteins/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Interleukin-1 Receptor-Like 1 Protein , Mice , Mice, Inbred C57BL , Phosphorylation , Pneumonia/pathology , Proteasome Endopeptidase Complex/metabolism , Serine/metabolism , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL
...