Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters











Publication year range
1.
Harm Reduct J ; 20(1): 110, 2023 08 16.
Article in English | MEDLINE | ID: mdl-37587466

ABSTRACT

BACKGROUND: The opioid crisis continues in full force, as physicians and caregivers are desperate for resources to help patients with opioid use and chronic pain disorders find safer and more accessible non-opioid tools. MAIN BODY: The purpose of this article is to review the current state of the opioid epidemic; the shifting picture of cannabinoids; and the research, policy, and current events that make opioid risk reduction an urgent public health challenge. The provided table contains an evidence-based clinical framework for the utilization of cannabinoids to treat patients with chronic pain who are dependent on opioids, seeking alternatives to opioids, and tapering opioids. CONCLUSION: Based on a comprehensive review of the literature and epidemiological evidence to date, cannabinoids stand to be one of the most interesting, safe, and accessible tools available to attenuate the devastation resulting from the misuse and abuse of opioid narcotics. Considering the urgency of the opioid epidemic and broadening of cannabinoid accessibility amidst absent prescribing guidelines, the authors recommend use of this clinical framework in the contexts of both clinical research continuity and patient care.


Subject(s)
Chronic Pain , Epidemics , Humans , Analgesics, Opioid/therapeutic use , Opioid Epidemic , Chronic Pain/drug therapy , Narcotics
2.
Am J Physiol Lung Cell Mol Physiol ; 321(5): L837-L843, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34494468

ABSTRACT

Bronchoalveolar lavage (BAL) samples from Severe Asthma Research Program (SARP) patients display suppression of a module of genes involved in cAMP-signaling pathways (BALcAMP) correlating with severity, therapy, and macrophage constituency. We sought to establish if gene expression changes were specific to macrophages and compared gene expression trends from multiple sources. Datasets included single-cell RNA sequencing (scRNA-seq) from lung specimens including a fatal exacerbation of severe Asthma COPD Overlap Syndrome (ACOS) after intense therapy and controls without lung disease, bulk RNA sequencing from cultured macrophage (THP-1) cells after acute or prolonged ß-agonist exposure, SARP datasets, and data from the Immune Modulators of Severe Asthma (IMSA) cohort. THP monocytes suppressed BALcAMP network gene expression after prolonged relative to acute ß-agonist exposure, corroborating SARP observations. scRNA-seq from healthy and diseased lung tissue revealed 13 cell populations enriched for macrophages. In severe ACOS, BALcAMP gene network expression scores were decreased in many cell populations, most significantly for macrophage populations (P < 3.9e-111). Natural killer (NK) cells and type II alveolar epithelial cells displayed less robust network suppression (P < 9.2e-8). Alveolar macrophages displayed the most numerous individual genes affected and the highest amplitude of modulation. Key BALcAMP genes demonstrate significantly decreased expression in severe asthmatics in the IMSA cohort. We conclude that suppression of the BALcAMP gene module identified from SARP BAL samples is validated in the IMSA patient cohort with physiological parallels observed in a monocytic cell line and in a severe ACOS patient sample with effects preferentially localizing to macrophages.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Asthma-Chronic Obstructive Pulmonary Disease Overlap Syndrome/drug therapy , Asthma-Chronic Obstructive Pulmonary Disease Overlap Syndrome/pathology , Bronchodilator Agents/pharmacology , Cyclic AMP/biosynthesis , Macrophages, Alveolar/immunology , Bronchoalveolar Lavage Fluid/chemistry , Cell Line , Cyclic AMP/genetics , Gene Expression/genetics , Gene Expression Regulation/genetics , Humans , Killer Cells, Natural/immunology , Lung/pathology , Macrophages, Alveolar/metabolism , Single-Cell Analysis , THP-1 Cells
3.
JCI Insight ; 5(11)2020 06 04.
Article in English | MEDLINE | ID: mdl-32493843

ABSTRACT

Mitochondrial quality control is mediated by the PTEN-induced kinase 1 (PINK1), a cytoprotective protein that is dysregulated in inflammatory lung injury and neurodegenerative diseases. Here, we show that a ubiquitin E3 ligase receptor component, FBXO7, targets PINK1 for its cellular disposal. FBXO7, by mediating PINK1 ubiquitylation and degradation, was sufficient to induce mitochondrial injury and inflammation in experimental pneumonia. A computational simulation-based screen led to the identification of a small molecule, BC1464, which abrogated FBXO7 and PINK1 association, leading to increased cellular PINK1 concentrations and activities, and limiting mitochondrial damage. BC1464 exerted antiinflammatory activity in human tissue explants and murine lung inflammation models. Furthermore, BC1464 conferred neuroprotection in primary cortical neurons, human neuroblastoma cells, and patient-derived cells in several culture models of Parkinson's disease. The data highlight a unique opportunity to use small molecule antagonists that disrupt PINK1 interaction with the ubiquitin apparatus to enhance mitochondrial quality, limit inflammatory injury, and maintain neuronal viability.


Subject(s)
F-Box Proteins/antagonists & inhibitors , Mitochondrial Proteins/metabolism , Neuroprotective Agents/pharmacology , Protein Kinases/metabolism , Proteolysis/drug effects , Ubiquitination/drug effects , Animals , Cell Line, Tumor , Enzyme Stability , F-Box Proteins/metabolism , Humans , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Mice , Neuroprotective Agents/chemistry , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Parkinson Disease/pathology , Pneumonia/drug therapy , Pneumonia/metabolism , Pneumonia/pathology
4.
Mol Cell Biol ; 40(12)2020 05 28.
Article in English | MEDLINE | ID: mdl-32205409

ABSTRACT

FBXL2 is an important ubiquitin E3 ligase component that modulates inflammatory signaling and cell cycle progression, but its molecular regulation is largely unknown. Here, we show that tumor necrosis factor alpha (TNF-α), a critical cytokine linked to the inflammatory response during skeletal muscle regeneration, suppressed Fbxl2 mRNA expression in C2C12 myoblasts and triggered significant alterations in cell cycle, metabolic, and protein translation processes. Gene silencing of Fbxl2 in skeletal myoblasts resulted in increased proliferative responses characterized by activation of mitogen-activated protein (MAP) kinases and nuclear factor kappa B and decreased myogenic differentiation, as reflected by reduced expression of myogenin and impaired myotube formation. TNF-α did not destabilize the Fbxl2 transcript (half-life [t1/2], ∼10 h) but inhibited SP1 transactivation of its core promoter, localized to bp -160 to +42 within the proximal 5' flanking region of the Fbxl2 gene. Chromatin immunoprecipitation and gel shift studies indicated that SP1 interacted with the Fbxl2 promoter during cellular differentiation, an effect that was less pronounced during proliferation or after TNF-α exposure. TNF-α, via activation of JNK, mediated phosphorylation of SP1 that impaired its binding to the Fbxl2 promoter, resulting in reduced transcriptional activity. The results suggest that SP1 transcriptional activation of Fbxl2 is required for skeletal muscle differentiation, a process that is interrupted by a key proinflammatory myopathic cytokine.IMPORTANCE Skeletal muscle regeneration and repair involve the recruitment and proliferation of resident satellite cells that exit the cell cycle during the process of myogenic differentiation to form myofibers. We demonstrate that the ubiquitin E3 ligase subunit FBXL2 is essential for skeletal myogenesis through its important effects on cell cycle progression and cell proliferative signaling. Further, we characterize a new mechanism whereby sustained stimulation by a major proinflammatory cytokine, TNF-α, regulates skeletal myogenesis by inhibiting the interaction of SP1 with the Fbxl2 core promoter in proliferating myoblasts. Our findings contribute to the understanding of skeletal muscle regeneration through the identification of Fbxl2 as both a critical regulator of myogenic proliferative processes and a susceptible gene target during inflammatory stimulation by TNF-α in skeletal muscle. Modulation of Fbxl2 activity may have relevance to disorders of muscle wasting associated with sustained proinflammatory signaling.


Subject(s)
F-Box Proteins/genetics , Muscle Development , Muscle, Skeletal/physiology , Sp1 Transcription Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism , Cells, Cultured , Humans , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/metabolism , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid
5.
Crit Care Med ; 47(12): 1724-1734, 2019 12.
Article in English | MEDLINE | ID: mdl-31634231

ABSTRACT

OBJECTIVES: Classification of patients with acute respiratory distress syndrome into hyper- and hypoinflammatory subphenotypes using plasma biomarkers may facilitate more effective targeted therapy. We examined whether established subphenotypes are present not only in patients with acute respiratory distress syndrome but also in patients at risk for acute respiratory distress syndrome (ARFA) and then assessed the prognostic information of baseline subphenotyping on the evolution of host-response biomarkers and clinical outcomes. DESIGN: Prospective, observational cohort study. SETTING: Medical ICU at a tertiary academic medical center. PATIENTS: Mechanically ventilated patients with acute respiratory distress syndrome or ARFA. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: We performed longitudinal measurements of 10 plasma biomarkers of host injury and inflammation. We applied unsupervised latent class analysis methods utilizing baseline clinical and biomarker variables and demonstrated that two-class models (hyper- vs hypoinflammatory subphenotypes) offered improved fit compared with one-class models in both patients with acute respiratory distress syndrome and ARFA. Baseline assignment to the hyperinflammatory subphenotype (39/104 [38%] acute respiratory distress syndrome and 30/108 [28%] ARFA patients) was associated with higher severity of illness by Sequential Organ Failure Assessment scores and incidence of acute kidney injury in patients with acute respiratory distress syndrome, as well as higher 30-day mortality and longer duration of mechanical ventilation in ARFA patients (p < 0.0001). Hyperinflammatory patients exhibited persistent elevation of biomarkers of innate immunity for up to 2 weeks postintubation. CONCLUSIONS: Our results suggest that two distinct subphenotypes are present not only in patients with established acute respiratory distress syndrome but also in patients at risk for its development. Hyperinflammatory classification at baseline is associated with higher severity of illness, worse clinical outcomes, and trajectories of persistently elevated biomarkers of host injury and inflammation during acute critical illness compared with hypoinflammatory patients. Our findings provide strong rationale for examining treatment effect modifications by subphenotypes in randomized clinical trials to inform precision therapeutic approaches in critical care.


Subject(s)
Respiratory Distress Syndrome/blood , Respiratory Distress Syndrome/complications , Adult , Aged , Biomarkers/blood , Female , Humans , Inflammation/blood , Inflammation/complications , Male , Middle Aged , Phenotype , Prognosis , Prospective Studies , Respiratory Distress Syndrome/classification , Respiratory Distress Syndrome/genetics , Risk Assessment
6.
Am J Respir Crit Care Med ; 200(7): 837-856, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31161938

ABSTRACT

Rationale: Gene expression of BAL cells, which samples the cellular milieu within the lower respiratory tract, has not been well studied in severe asthma.Objectives: To identify new biomolecular mechanisms underlying severe asthma by an unbiased, detailed interrogation of global gene expression.Methods: BAL cell expression was profiled in 154 asthma and control subjects. Of these participants, 100 had accompanying airway epithelial cell gene expression. BAL cell expression profiles were related to participant (age, sex, race, and medication) and sample traits (cell proportions), and then severity-related gene expression determined by correlating transcripts and coexpression networks to lung function, emergency department visits or hospitalizations in the last year, medication use, and quality-of-life scores.Measurements and Main Results: Age, sex, race, cell proportions, and medications strongly influenced BAL cell gene expression, but leading severity-related genes could be determined by carefully identifying and accounting for these influences. A BAL cell expression network enriched for cAMP signaling components most differentiated subjects with severe asthma from other subjects. Subsequently, an in vitro cellular model showed this phenomenon was likely caused by a robust upregulation in cAMP-related expression in nonsevere and ß-agonist-naive subjects given a ß-agonist before cell collection. Interestingly, ELISAs performed on BAL lysates showed protein levels may partly disagree with expression changes.Conclusions: Gene expression in BAL cells is influenced by factors seldomly considered. Notably, ß-agonist exposure likely had a strong and immediate impact on cellular gene expression, which may not translate to important disease mechanisms or necessarily match protein levels. Leading severity-related genes were discovered in an unbiased, system-wide analysis, revealing new targets that map to asthma susceptibility loci.


Subject(s)
Asthma/genetics , Bronchoalveolar Lavage Fluid/cytology , Gene Expression/genetics , Adrenergic beta-Agonists/pharmacology , Adult , Asthma/metabolism , Case-Control Studies , Cyclic AMP/metabolism , Eosinophils/metabolism , Epithelial Cells/metabolism , Female , Gene Expression/drug effects , Humans , In Vitro Techniques , Lymphocytes/metabolism , Macrophages, Alveolar/metabolism , Male , Neutrophils/metabolism , Sequence Analysis, RNA , Severity of Illness Index , Signal Transduction/genetics , THP-1 Cells/metabolism
7.
EBioMedicine ; 45: 553-562, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31204278

ABSTRACT

BACKGROUND: The Single immunoglobin interleukin-1 (IL-1)-related receptor (Sigirr), also known as IL-1R8, has been shown to exhibit broad anti-inflammatory effects against inflammatory diseases including acute lung injury, while molecular regulation of IL-1R8/Sigirr protein stability has not been reported. This study is designed to determine whether stabilization of IL-1R8/Sigirr by a deubiquitinating enzyme (DUB) is sufficient to suppress inflammatory responses and lessen lung inflammation. METHODS: A molecular signature of ubiquitination and degradation of IL-1R8/Sigirr was determined using a receptor ligation chase model. The anti-inflammatory effects on USP13 were investigated. USP13 knockout mice were evaluated for stabilization of IL-1R8/Sigirr and disease phenotype in an acute lung injury model. FINDINGS: IL-1R8/Sigirr degradation is mediated by the ubiquitin-proteasome system, through site-specific ubiquitination. This effect was antagonized by the DUB USP13. USP13 levels correlate directly with IL-1R8/Sigirr, and both proteins were reduced in cells and tissue from mice subjected to inflammatory injury by the TLR4 agonist lipopolysaccharide (LPS). Knockdown of USP13 in cells increased IL-1R8/Sigirr poly-ubiquitination and reduced its stability, which enhanced LPS-induced TLR4 signaling and cytokine release. Likewise, USP13-deficient mice were highly susceptible to LPS or Pseudomonas aeruginosa models of inflammatory lung injury. IL-1R8/Sigirr overexpression in cells or by pulmonary viral transduction attenuated the inflammatory phenotype conferred by the USP13-/- genotype. INTERPRETATION: Stabilization of IL-1R8/Sigirr by USP13 describes a novel anti-inflammatory pathway in diseases that could provide a new strategy to modulate immune activation. FUND: This study was supported by the US National Institutes of Health (R01HL131665, HL136294 to Y.Z., R01 GM115389 to J.Z.).


Subject(s)
Endopeptidases/genetics , Lung Diseases/genetics , Pneumonia/genetics , Receptors, Interleukin-1/genetics , Animals , Anti-Inflammatory Agents/therapeutic use , Disease Models, Animal , Humans , Lipopolysaccharides/therapeutic use , Lung Diseases/chemically induced , Lung Diseases/pathology , Mice , Mice, Knockout , Pneumonia/chemically induced , Pneumonia/pathology , Pneumonia/therapy , Protein Stability , Signal Transduction/genetics , Toll-Like Receptor 4/genetics , Ubiquitin-Specific Proteases
8.
JCI Insight ; 3(19)2018 10 04.
Article in English | MEDLINE | ID: mdl-30282819

ABSTRACT

The acute respiratory distress syndrome (ARDS) causes an estimated 70,000 US deaths annually. Multiple pharmacologic interventions for ARDS have been tested and failed. An unmet need is a suitable laboratory human model to predictively assess emerging therapeutics on organ function in ARDS. We previously demonstrated that the small molecule BC1215 blocks actions of a proinflammatory E3 ligase-associated protein, FBXO3, to suppress NF-κB signaling in animal models of lung injury. Ex vivo lung perfusion (EVLP) is a clinical technique that maintains lung function for possible transplant after organ donation. We used human lungs unacceptable for transplant to model endotoxemic injury with EVLP for 6 hours. LPS infusion induced inflammatory injury with impaired oxygenation of pulmonary venous circulation. BC1215 treatment after LPS rescued oxygenation and decreased inflammatory cytokines in bronchoalveolar lavage. RNA sequencing transcriptomics from biopsies taken during EVLP revealed robust inflammatory gene induction by LPS with a strong signal for NF-κB-associated transcripts. BC1215 treatment reduced the LPS induction of genes associated with inflammatory and host defense gene responses by Gene Ontology (GOterm) and pathways analysis. BC1215 also significantly antagonized LPS-mediated NF-κB activity. EVLP may provide a unique human platform for preclinical study of chemical entities such as FBXO3 inhibitors on tissue physiology.


Subject(s)
Benzylamines/pharmacology , F-Box Proteins/antagonists & inhibitors , Lung/drug effects , Perfusion/methods , Pyridines/pharmacology , Respiratory Distress Syndrome/drug therapy , Adolescent , Adult , Benzylamines/therapeutic use , Drug Evaluation, Preclinical/methods , F-Box Proteins/metabolism , Female , Humans , Lipopolysaccharides/toxicity , Lung/pathology , Male , Middle Aged , Pyridines/therapeutic use , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/pathology , Signal Transduction/drug effects
9.
J Biol Chem ; 293(41): 15790-15800, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30093408

ABSTRACT

Interleukin (IL)-1ß plays a critical role in IL-6ß- and transforming growth factor ß (TGFß)-initiated Th17 differentiation and induction of Th17-mediated autoimmunity. However, the means by which IL-1 regulates various aspects of Th17 development remain poorly understood. We recently reported that IL-1ß enhances STAT3 phosphorylation via NF-κB-mediated repression of SOCS3 to facilitate Il17 transcription and Th17 differentiation, identifying an effect of IL-1 signaling on proximal events of STAT3 signaling. Here, we show that IL-1ß promotes STAT3 binding to key cis-elements that control IL-17 expression. Additionally, we demonstrate that the IL-1-induced NF-κB factor RelA directly regulates the Il17a/f loci in cooperation with STAT3. Our findings reveal that IL-1 impacts both proximal signaling events and downstream interactions between transcription factors and cis-regulatory elements to promote Il17a/f transcription and Th17 differentiation.


Subject(s)
Interleukin-17/metabolism , Receptors, Interleukin-1 Type II/metabolism , STAT3 Transcription Factor/metabolism , Transcription Factor RelA/metabolism , Animals , DNA/chemistry , DNA/genetics , Interleukin-17/genetics , Mice, Inbred C57BL , Regulatory Sequences, Nucleic Acid/genetics , STAT3 Transcription Factor/genetics , Signal Transduction/physiology , Th17 Cells , Transcription Factor RelA/genetics , Transcriptional Activation
10.
FASEB J ; 32(1): 230-242, 2018 01.
Article in English | MEDLINE | ID: mdl-28874458

ABSTRACT

The tumor necrosis factor receptor-associated factor 2 (TRAF2) is a second messenger adaptor protein that plays an essential role in propagating TNF-α-mediated signaling pathways. Modulation of TRAF2 activity by ubiquitination is well studied; however, the deubiquitinating enzyme (DUB), which regulates TRAF2 stability, has not been identified. Here we reveal USP48 as the first identified DUB to deubiquitinate and stabilize TRAF2 in epithelial cells. Down-regulation of USP48 increases K48-linked polyubiquitination of TRAF2 and reduces TRAF2 protein levels. Interestingly, USP48 only targets the TRAF2 related to JNK pathway, not the TRAF2 related to NF-κB and p38 pathways. USP48 is serine phosphorylated in response to TNF-α. The phosphorylation is catalyzed by glycogen synthase kinase 3ß (GSK3ß), ultimately resulting in increases in USP48 DUB activity. Furthermore, we reveal a new biologic function of TRAF2 that contributes to epithelial barrier dysfunction, which is attenuated by knockdown of USP48. Inhibition of TRAF2/JNK pathway increases E (epithelial)-cadherin expression and enhances epithelial barrier integrity, while knockdown of USP48 attenuates TNF-α/JNK pathway and increases E-cadherin expression and cell-cell junction in epithelial cells. These data, taken together, indicate that USP48 stabilizes TRAF2, which is promoted by GSK3ß-mediated phosphorylation. Further, down-regulation of USP48 increases E-cadherin expression and epithelial barrier integrity through reducing TRAF2 stability.-Li, S., Wang, D., Zhao, J., Weathington, N. M., Shang, D., Zhao, Y. The deubiquitinating enzyme USP48 stabilizes TRAF2 and reduces E-cadherin-mediated adherens junctions.


Subject(s)
Adherens Junctions/metabolism , Cadherins/metabolism , TNF Receptor-Associated Factor 2/metabolism , Ubiquitin-Specific Proteases/metabolism , A549 Cells , Animals , Antigens, CD , Cell Line , Epithelial Cells/metabolism , Gene Knockdown Techniques , Glycogen Synthase Kinase 3 beta/metabolism , HEK293 Cells , Humans , MAP Kinase Signaling System , Mice , Phosphorylation , Protein Stability , TNF Receptor-Associated Factor 2/chemistry , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin-Specific Proteases/antagonists & inhibitors , Ubiquitin-Specific Proteases/genetics , Ubiquitination
11.
J Mol Cell Biol ; 10(1): 60-73, 2018 02 01.
Article in English | MEDLINE | ID: mdl-28992046

ABSTRACT

Phosphorylation affects ubiquitination, stability, and activity of transcriptional factors, thus regulating various cellular functions. E2F transcriptional factor 1 (E2F1) regulates paternally expressed imprinted gene 10 (Peg10) expression, thereby promoting cell proliferation. However, the effect of E2F1 stability on Peg10 expression and the molecular regulation of E2F1 stability by its phosphorylation have not been well demonstrated. Here, we describe a new pathway in which phosphorylation of E2F1 by GSK3ß increases E2F1 association with the deubiquitinating enzyme, ubiquitin-specific protease 11 (USP11), which removes K63-linked ubiquitin chains thereby preventing E2F1 degradation in the nuclei. Downregulation of USP11 increases E2F1 ubiquitination and reduces E2F1 stability and protein levels, thereby decreasing Peg10 mRNA levels. Physiologically, USP11 depletion suppresses cell proliferation and wound healing in lung epithelial cells, and these effects are reversed by E2F1 and PEG10 overexpression. Thus, our study reveals a new molecular model that phosphorylation promotes substrate stability through increasing its association with a deubiquitinating enzyme. The data suggest that GSK3ß and USP11 act in concert to modulate E2F1 abundance and PEG10 expression in lung epithelial cells to affect cell wound healing. This study provides new therapeutic targets to lessen lung injury by improving lung epithelial cell repair and remodeling after injury.


Subject(s)
E2F1 Transcription Factor/metabolism , Epithelial Cells/cytology , Lung/cytology , Proteins/genetics , Thiolester Hydrolases/metabolism , Up-Regulation , A549 Cells , Apoptosis Regulatory Proteins , Cell Line , Cell Proliferation , DNA-Binding Proteins , Down-Regulation , Epithelial Cells/metabolism , Humans , Lung/metabolism , Phosphorylation , Protein Stability , Proteolysis , RNA-Binding Proteins , Thiolester Hydrolases/genetics , Ubiquitination
12.
Biochem J ; 474(20): 3543-3557, 2017 10 10.
Article in English | MEDLINE | ID: mdl-28883123

ABSTRACT

The IFN gamma receptor 1 (IFNGR1) binds IFN-γ and activates gene transcription pathways crucial for controlling bacterial and viral infections. Although decreases in IFNGR1 surface levels have been demonstrated to inhibit IFN-γ signaling, little is known regarding the molecular mechanisms controlling receptor stability. Here, we show in epithelial and monocytic cell lines that IFNGR1 displays K48 polyubiquitination, is proteasomally degraded, and harbors three ubiquitin acceptor sites at K277, K279, and K285. Inhibition of glycogen synthase kinase 3 beta (GSK3ß) destabilized IFNGR1 while overexpression of GSK3ß increased receptor stability. We identified critical serine and threonine residues juxtaposed to ubiquitin acceptor sites that impacted IFNGR1 stability. In CRISPR-Cas9 IFNGR1 generated knockout cell lines, cellular expression of IFNGR1 plasmids encoding ubiquitin acceptor site mutations demonstrated significantly impaired STAT1 phosphorylation and decreased STAT1-dependent gene induction. Thus, IFNGR1 undergoes rapid site-specific polyubiquitination, a process modulated by GSK3ß. Ubiquitination appears to be necessary for efficient IFNGR1-dependent gamma gene induction and represents a relatively uncharacterized regulatory mechanism for this receptor.


Subject(s)
Protein Processing, Post-Translational/physiology , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , Signal Transduction/physiology , CRISPR-Cas Systems/genetics , HEK293 Cells , Humans , Interferon-gamma/pharmacology , Protein Processing, Post-Translational/drug effects , Protein Stability/drug effects , Receptors, Interferon/chemistry , Signal Transduction/drug effects , Interferon gamma Receptor
13.
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
14.
Nat Commun ; 8: 15203, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28492230

ABSTRACT

Inflammasomes regulate innate immune responses by facilitating maturation of inflammatory cytokines, interleukin (IL)-1ß and IL-18. NACHT, LRR and PYD domains-containing protein 7 (NALP7) is one inflammasome constituent, but little is known about its cellular handling. Here we show a mechanism for NALP7 protein stabilization and activation of the inflammasome by Toll-like receptor (TLR) agonism with bacterial lipopolysaccharide (LPS) and the synthetic acylated lipopeptide Pam3CSK4. NALP7 is constitutively ubiquitinated and recruited to the endolysosome for degradation. With TLR ligation, the deubiquitinase enzyme, STAM-binding protein (STAMBP) impedes NALP7 trafficking to lysosomes to increase NALP7 abundance. STAMBP deubiquitinates NALP7 and STAMBP knockdown abrogates LPS or Pam3CSK4-induced increases in NALP7 protein. A small-molecule inhibitor of STAMBP deubiquitinase activity, BC-1471, decreases NALP7 protein levels and suppresses IL-1ß release after TLR agonism. These findings describe a unique pathway of inflammasome regulation with the identification of STAMBP as a potential therapeutic target to reduce pro-inflammatory stress.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Endosomal Sorting Complexes Required for Transport/genetics , Enzyme Inhibitors/pharmacology , Inflammasomes/genetics , Leukocytes, Mononuclear/drug effects , Ubiquitin Thiolesterase/genetics , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/immunology , Cell Line , Endosomal Sorting Complexes Required for Transport/antagonists & inhibitors , Endosomal Sorting Complexes Required for Transport/immunology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/immunology , Gene Expression Regulation , HeLa Cells , Humans , Immunity, Innate , Inflammasomes/antagonists & inhibitors , Inflammasomes/immunology , Interleukin-18/genetics , Interleukin-18/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Lipopeptides/antagonists & inhibitors , Lipopeptides/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Lysosomes/drug effects , Lysosomes/metabolism , Molecular Docking Simulation , Primary Cell Culture , Signal Transduction , THP-1 Cells , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/immunology , Ubiquitination/drug effects
15.
Am J Respir Cell Mol Biol ; 57(3): 346-354, 2017 09.
Article in English | MEDLINE | ID: mdl-28421819

ABSTRACT

IL-25 and IL-4 signaling in the setting of infection or allergic responses can drive Type 2 inflammation. IL-25 requires the IL-17 receptor B (IL-17Rb) to mediate signaling through nuclear factor κ B (NF-κB) transcriptional activation. Despite the known coexistence of these two cytokines in the Type 2 inflammatory environment, collaborative signaling between the IL-4 and IL-25 axes is poorly explored. Here we demonstrate IL-4 induction of both IL-25 and IL-17Rb protein in human lung tissue culture, primary alveolar macrophages, and the THP-1 monocytic cell line. IL-4 treatment triggers gene transcription for both IL-25 and IL-17Rb but does not alter the receptor mRNA stability. Genetic antagonism of the IL-4 second messenger, signal transducer and activator of transcription 6 (STAT6), with small interfering RNA (siRNA) blunts IL-17Rb mRNA induction by IL-4. IL-25 induces signaling through the canonical NF-κB pathway, and STAT6 or NF-κB signaling inhibitors prevent IL-17Rb expression. Blockade of IL-25 with monoclonal antibody suppresses NF-κB activation after IL-4 treatment, and IL-4-mediated induction of IL-17Rb is suppressed by IL-25 siRNA. IL-25 and IL-17Rb promoter regions harbor putative NF-κB and STAT6 consensus sites, and chromatin immunoprecipitation identified these transcription factors in complex with the IL-17Rb 5' untranslated region. In bronchoalveolar lavage RNA preparations, IL-25 and IL-17Rb mRNA transcripts are increased in asthmatics compared with healthy control subjects, and IL-25 transcript abundance correlates strongly with IL-4 mRNA levels. Thus, these results indicate that IL-4 signaling up-regulates the IL-25 axis in human monocytic cells, and that IL-25 may provide autocrine signals in monocytes and macrophages to sustain IL-17Rb expression and predispose to alternative activation.


Subject(s)
Autocrine Communication/genetics , Interleukin-17/metabolism , Interleukin-4/metabolism , Monocytes/metabolism , Receptors, Interleukin-17/genetics , Transcription, Genetic , Asthma/genetics , Asthma/pathology , Base Sequence , Cell Line , Humans , Macrophages, Alveolar/metabolism , NF-kappa B/metabolism , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Interleukin-17/metabolism , STAT6 Transcription Factor/metabolism
16.
J Clin Invest ; 126(9): 3176-84, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27584731

ABSTRACT

The extracellular matrix (ECM) is the noncellular component critical in the maintenance of organ structure and the regulation of tissue development, organ structure, and cellular signaling. The ECM is a dynamic entity that undergoes continuous degradation and resynthesis. In addition to compromising structure, degradation of the ECM can liberate bioactive fragments that cause cellular activation and chemotaxis of a variety of cells. These fragments are termed matrikines, and their cellular activities are sentinel in the development and progression of tissue injury seen in chronic lung disease. Here, we discuss the matrikines that are known to be active in lung biology and their roles in lung disease. We also consider the use of matrikines as disease markers and potential therapeutic targets in lung disease.


Subject(s)
Chemotaxis , Extracellular Matrix Proteins/metabolism , Extracellular Matrix/metabolism , Lung Diseases/metabolism , Lung/metabolism , Animals , Collagen/chemistry , Glycine/chemistry , Humans , Hyaluronic Acid/metabolism , Laminin/chemistry , Proline/chemistry , Signal Transduction
17.
Mar Drugs ; 14(7)2016 Jul 21.
Article in English | MEDLINE | ID: mdl-27455285

ABSTRACT

Long-term cigarette smoking increases the risk for chronic obstructive pulmonary disease (COPD), characterized by irreversible expiratory airflow limitation. The pathogenesis of COPD involves oxidative stress and chronic inflammation. Various natural marine compounds possess both anti-oxidant and anti-inflammatory properties, but few have been tested for their efficacy in COPD models. In this study, we conducted an in vitro screening test to identify natural compounds isolated from various brown algae species that might provide protection against cigarette smoke extract (CSE)-induced cytotoxicity. Among nine selected natural compounds, apo-9'-fucoxanthinone (Apo9F) exhibited the highest protection against CSE-induced cytotoxicity in immortalized human bronchial epithelial cells (HBEC2). Furthermore, the protective effects of Apo9F were observed to be associated with a significant reduction in apoptotic cell death, DNA damage, and the levels of mitochondrial reactive oxygen species (ROS) released from CSE-exposed HBEC2 cells. These results suggest that Apo9F protects against CSE-induced DNA damage and apoptosis by regulating mitochondrial ROS production.


Subject(s)
Apoptosis/drug effects , Epithelial Cells/drug effects , Nicotiana/adverse effects , Oxidative Stress/drug effects , Rhodophyta/chemistry , Smoke/adverse effects , Terpenes/pharmacology , Cell Line , DNA Damage/drug effects , Epithelial Cells/metabolism , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Protective Agents/chemistry , Protective Agents/pharmacology , Pulmonary Disease, Chronic Obstructive/etiology , Pulmonary Disease, Chronic Obstructive/metabolism , Reactive Oxygen Species/metabolism , Smoking/adverse effects , Terpenes/chemistry
18.
Ann Am Thorac Soc ; 12(12): S222-7, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26653202

ABSTRACT

With the expected rapid growth of the aging population worldwide, there is a clear need to understand the complex process of aging to develop interventions that might extend the health span in this group of patients. Aging is associated with increased susceptibility to a variety of chronic diseases, and lung pathologies are no exception. The prevalence of lung diseases such as idiopathic pulmonary fibrosis and chronic obstructive pulmonary disease has been found to increase considerably with age. In October 2014, the Division of Pulmonary, Allergy, and Critical Care of the University of Pittsburgh cohosted the Pittsburgh-Munich Lung Conference focused in aging and lung disease with the Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Ludwig-Maximilians University and Helmholtz Zentrum Munich Germany. The purpose of the conference was to disseminate novel concepts in aging mechanisms that have an impact in lung physiology and pathogenesis of pulmonary diseases that commonly occur in older populations. The conference included 28 presentations on diverse topics, which are summarized in this report. The participants identified priorities for future basic and translational investigations that will assist in the identification of molecular insights involved in the pathogenesis of age-related pulmonary diseases and the design of therapeutic interventions for these lung conditions.


Subject(s)
Aging , Disease Management , Disease Susceptibility , Lung Diseases/therapy , Humans
19.
Sci Transl Med ; 7(311): 311ra171, 2015 Oct 28.
Article in English | MEDLINE | ID: mdl-26511508

ABSTRACT

Unchecked epithelial cell death is fundamental to the pathogenesis of pneumonia. The recognition of unique signaling pathways that preserve epithelial cell viability may present new opportunities for interventional strategies. We describe that mortality factor 4 like 1 (Morf4l1), a protein involved in chromatin remodeling, is constitutively expressed at low levels in the lung because of its continuous degradation mediated by an orphan ubiquitin E3 ligase subunit, Fbxl18. Expression of Morf4l1 increases in humans with pneumonia and is up-regulated in lung epithelia after exposure to Pseudomonas aeruginosa or lipopolysaccharide. In a mouse model of pneumonia induced by P. aeruginosa, Morf4l1 is stabilized by acetylation that protects it from Fbxl18-mediated degradation. After P. aeruginosa infection of mice, overexpression of Morf4l1 resulted in lung epithelial cell death, whereas its depletion restored cell viability. Using in silico modeling and drug-target interaction studies, we identified that the U.S. Food and Drug Administration-approved thrombin inhibitor argatroban is a Morf4l1 antagonist. Argatroban inhibited Morf4l1-dependent histone acetylation, reduced its cytotoxicity, and improved survival of mice with experimental lung injury at doses that had no anticoagulant activity. These studies uncover a previously unrecognized biological mechanism whereby pathogens subvert cell viability by extending the life span of a cytotoxic host protein. Morf4l1 may be a potential molecular target for non-antibiotic pharmacotherapy during severe pulmonary infection.


Subject(s)
Pneumonia/metabolism , Transcription Factors/metabolism , Animals , Disease Models, Animal , Epithelial Cells/metabolism , Humans , In Vitro Techniques , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Pneumonia/microbiology , Pseudomonas Infections/metabolism , Pseudomonas aeruginosa/pathogenicity , Transcription Factors/genetics
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL