Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
1.
Alcohol Clin Exp Res (Hoboken) ; 47(10): 1839-1849, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37864530

ABSTRACT

BACKGROUND: Chronic alcohol ingestion predisposes to lung injury and disrepair during sepsis. Our previous studies outlined roles for transforming growth factor-beta 1 (TGFß1) and granulocyte-macrophage colony-stimulating factor (GM-CSF) in epithelial barrier homeostasis and how alcohol perturbs their expression and signaling. Here we hypothesize that ethanol-exposed lung fibroblasts (LF) are a source of dysregulated TGFß1 and GM-CSF and thereby alter airway epithelial barrier function. METHODS: Human or rat LF were cultured ± ethanol for 2 weeks and then co-cultured with human or rat airway epithelial cells (AEC) seeded on Transwell permeable supports. In selected groups, a TGFß1 receptor type 1 (TGFßR1) inhibitor (SB431542) or a TGFß1 neutralizing antibody was applied. Transepithelial electrical resistance (TER) was measured prior to co-culture and on day 5 of co-culture. AEC were then analyzed for the expression of selected tight junction and mesenchymal proteins, and transwell membranes were analyzed by immunofluorescence microscopy for ZO-1 expression and localization. TGFß1 and GM-CSF levels in conditioned media from the co-cultures were quantified by ELISA. RESULTS: AEC co-cultured with ethanol-exposed LF (ELF) showed a significant reduction in TER and corresponding decreases in ZO-1 expression, whereas collagen type 1A1 and α-smooth muscle actin protein expression were increased. In parallel, in conditioned media from the ELF + AEC co-cultures, activated TGFß1 levels increased and GM-CSF levels decreased. Notably, all the effects of ELF on the AEC were prevented by blocking TGFß1 activity. CONCLUSIONS: Prior ethanol exposure to LF induces barrier dysfunction in naive AEC in a paracrine fashion through activation of TGFß1 signaling and suppression of GM-CSF. These experimental findings provide a potential mechanism by which chronic alcohol ingestion impairs airway epithelial integrity and renders individuals susceptible to lung injury.

2.
Shock ; 59(4): 612-620, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36640152

ABSTRACT

ABSTRACT: Increased epithelial permeability in sepsis is mediated via disruptions in tight junctions, which are closely associated with the perijunctional actin-myosin ring. Genetic deletion of myosin light chain kinase (MLCK) reverses sepsis-induced intestinal hyperpermeability and improves survival in a murine model of intra-abdominal sepsis. In an attempt to determine the generalizability of these findings, this study measured the impact of MLCK deletion on survival and potential associated mechanisms following pneumonia-induced sepsis. MLCK -/- and wild-type mice underwent intratracheal injection of Pseudomonas aeruginosa . Unexpectedly, survival was significantly worse in MLCK -/- mice than wild-type mice. This was associated with increased permeability to Evans blue dye in bronchoalveolar lavage fluid but not in tissue homogenate, suggesting increased alveolar epithelial leak. In addition, bacterial burden was increased in bronchoalveolar lavage fluid. Cytokine array using whole-lung homogenate demonstrated increases in multiple proinflammatory and anti-inflammatory cytokines in knockout mice. These local pulmonary changes were associated with systemic inflammation with increased serum levels of IL-6 and IL-10 and a marked increase in bacteremia in MLCK -/- mice. Increased numbers of both bulk and memory CD4 + T cells were identified in the spleens of knockout mice, with increased early and late activation. These results demonstrate that genetic deletion of MLCK unexpectedly increases mortality in pulmonary sepsis, associated with worsened alveolar epithelial leak and both local and systemic inflammation. This suggests that caution is required in targeting MLCK for therapeutic gain in sepsis.


Subject(s)
Lung , Myosin-Light-Chain Kinase , Pneumonia , Sepsis , Animals , Mice , Cytokines , Inflammation , Intestinal Mucosa , Lung/metabolism , Lung/pathology , Mice, Knockout , Myosin-Light-Chain Kinase/genetics , Permeability , Pneumonia/complications , Sepsis/pathology , Tight Junctions/physiology
3.
Alcohol Clin Exp Res ; 46(12): 2214-2224, 2022 12.
Article in English | MEDLINE | ID: mdl-36281822

ABSTRACT

BACKGROUND: Alcohol impairs pulmonary innate immune function and is associated with an increased risk of tuberculosis (TB). Toll-like receptor 2 (TLR2) is a pattern recognition receptor on alveolar macrophages that recognizes Mycobacterium tuberculosis (Mtb). The expression of TLR2 depends, in part, on granulocyte-macrophage colony-stimulating factor (GM-CSF) signaling. Given our prior work demonstrating the suppression of GM-CSF signaling following chronic alcohol ingestion, we hypothesized that alcohol impairs TLR2 expression via the suppression of GM-CSF and thereby reduces the ability of the macrophage to recognize and phagocytose Mtb. METHODS: Primary alveolar macrophages were isolated from control-fed and alcohol-fed rats. Prior to cell isolation, some alcohol-fed rats were treated with intranasal GM-CSF and then endotracheally inoculated with an attenuated strain of Mtb. Primary macrophages were then isolated and immunofluorescence was used to determine phagocytic efficiency and TLR2 expression in the presence and absence of GM-CSF treatment and phagocytic efficiency in the presence and absence of TLR2 neutralization. RESULTS: TLR2 expression and phagocytosis of Mtb were significantly lower in the alveolar macrophages of alcohol-fed rats than control-fed rats. In parallel, blocking TLR2 signaling recapitulated this decreased phagocytosis of Mtb. In contrast, intranasal GM-CSF treatment restored TLR2 expression and Mtb phagocytosis in the alveolar macrophages of alcohol-fed rats to levels comparable to those of control-fed rats. CONCLUSIONS: Chronic alcohol ingestion reduces TLR2 protein expression and phagocytosis of Mtb, likely due to impaired GM-CSF signaling. GM-CSF restores membrane-bound TLR2 expression and phagocytic function.


Subject(s)
Ethanol , Macrophages, Alveolar , Mycobacterium tuberculosis , Phagocytosis , Toll-Like Receptor 2 , Animals , Rats , Ethanol/adverse effects , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Mycobacterium tuberculosis/metabolism , Toll-Like Receptor 2/metabolism , Phagocytosis/drug effects
4.
Am J Med Sci ; 362(6): 535-536, 2021 12.
Article in English | MEDLINE | ID: mdl-34634263
5.
Am J Med Sci ; 361(1): 90-97, 2021 01.
Article in English | MEDLINE | ID: mdl-32773107

ABSTRACT

BACKGROUND: Despite anti-retroviral therapy, HIV-1 infection increases the risk of pneumonia and causes oxidative stress and defective alveolar macrophage (AM) immune function. We have previously determined that HIV-1 proteins inhibit antioxidant defenses and impair AM phagocytosis by suppressing nuclear factor (erythroid-derived 2)-like 2 (Nrf2). Given its known effects on Nrf2, we hypothesize miR-144 mediates the HIV-1 induced suppression of Nrf2. METHODS: Primary AMs isolated from HIV-1 transgenic (HIV-1 Tg) rats and wild type littermates (WT) as well as human monocyte-derived macrophages (MDMs) infected ex vivo with HIV-1 were used. We modulated miR-144 expression using a miR-144 mimic or an inhibitor to assay its effects on Nrf2/ARE activity and AM functions in vitro and in vivo. RESULTS: MiR-144 expression was increased in AMs from HIV-1 Tg rats and in HIV-1-infected human MDMs compared to cells from WT rats and non-infected human MDMs, respectively. Increasing miR-144 with a miR-144 mimic inhibited the expression of Nrf2 and its downstream effectors in WT rat macrophages and consequently impaired their bacterial phagocytic capacity and H2O2 scavenging ability. These effects on Nrf2 expression and AM function were reversed by antagonizing miR-144 ex vivo or in the airways of HIV-1 Tg rats in vivo, but this protection was abrogated by silencing Nrf2 expression. CONCLUSIONS: Our results suggest that inhibiting miR-144 or interfering with its deleterious effects on Nrf2 attenuates HIV-1-mediated AM immune dysfunction and improves lung health in individuals with HIV.


Subject(s)
HIV Infections/physiopathology , HIV/physiology , Macrophages, Alveolar/metabolism , MicroRNAs/metabolism , NF-E2-Related Factor 2/metabolism , Animals , Female , HIV Infections/metabolism , Male , Rats
7.
Sci Rep ; 10(1): 11696, 2020 07 16.
Article in English | MEDLINE | ID: mdl-32678115

ABSTRACT

Pulmonary Arterial Hypertension (PAH) is overrepresented in People Living with Human Immunodeficiency Virus (PLWH). HIV protein gp120 plays a key role in the pathogenesis of HIV-PAH. Genetic changes in HIV gp120 determine viral interactions with chemokine receptors; specifically, HIV-X4 viruses interact with CXCR4 while HIV-R5 interact with CCR5 co-receptors. Herein, we leveraged banked samples from patients enrolled in the NIH Lung HIV studies and used bioinformatic analyses to investigate whether signature sequences in HIV-gp120 that predict tropism also predict PAH. Further biological assays were conducted in pulmonary endothelial cells in vitro and in HIV-transgenic rats. We found that significantly more persons living with HIV-PAH harbor HIV-X4 variants. Multiple HIV models showed that recombinant gp120-X4 as well as infectious HIV-X4 remarkably increase arachidonate 5-lipoxygenase (ALOX5) expression. ALOX5 is essential for the production of leukotrienes; we confirmed that leukotriene levels are increased in bronchoalveolar lavage fluid of HIV-infected patients. This is the first report associating HIV-gp120 genotype to a pulmonary disease phenotype, as we uncovered X4 viruses as potential agents in the pathophysiology of HIV-PAH. Altogether, our results allude to the supplementation of antiretroviral therapy with ALOX5 antagonists to rescue patients with HIV-X4 variants from fatal PAH.


Subject(s)
Arachidonate 5-Lipoxygenase/metabolism , HIV Infections/complications , HIV-1/genetics , Lung/metabolism , Pulmonary Arterial Hypertension/complications , Viral Tropism/genetics , Adult , Animals , Anti-HIV Agents/therapeutic use , Cells, Cultured , Cohort Studies , Disease Models, Animal , Endothelial Cells/metabolism , Female , Genotype , HIV Envelope Protein gp120/genetics , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/metabolism , Humans , Male , Middle Aged , Phenotype , Pulmonary Arterial Hypertension/virology , Pulmonary Artery/cytology , Rats , Rats, Inbred F344 , Rats, Transgenic , Receptors, CXCR4/metabolism
9.
Cell Death Dis ; 10(8): 580, 2019 08 02.
Article in English | MEDLINE | ID: mdl-31371699

ABSTRACT

Antiretroviral therapy extends survival but does not eliminate HIV from its cellular reservoirs. Between immune and stromal cells in the tissue microenvironment, a dynamic intercellular communication might influence host viral immune responses via intercellular transfer of extracellular vehicles (EVs) (microvesicles, exosome, or apoptotic bodies). It is increasingly recognized that HIV-infected macrophage-secreted nucleotide-rich exosomes might play a critical role in mediating communication between macrophages and other structural cells; however, molecular mechanisms underlying cell-cell crosstalk remain unknown. Here we show that HIV-1-infected macrophages and HIV-1 proteins Tat or gp120-treated macrophages express high levels of microRNAs, including miR-23a and miR-27a. Identical miRNAs expression patterns were detected in macrophage-secreted exosomes isolated from bronchoalveolar lavage fluid of HIV transgenic rats. Tat-treated macrophage-derived exosomal miR-23a attenuated posttranscriptional modulation of key tight junction protein zonula occludens (ZO-1) 3'-UTR in epithelial cells. In parallel, exosomal miR-27a released from Tat-treated macrophages altered the mitochondrial bioenergetics of recipient lung epithelial cells by targeting peroxisome proliferator-activated receptor gamma (PPARγ), while simultaneously stimulating glycolysis. Together, exosomal miRNAs shuttle from macrophages to epithelial cells and thereby explain in part HIV-mediated lung epithelial barrier dysfunction. These studies suggest that targeting miRNAs may be of therapeutic value to enhance lung health in HIV.


Subject(s)
Lung/metabolism , MicroRNAs/genetics , Mitochondria/metabolism , Cell Movement/drug effects , Energy Metabolism/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/virology , Extracellular Vesicles/genetics , Glycolysis/genetics , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/pharmacology , HIV-1/genetics , HIV-1/pathogenicity , Humans , Lung/pathology , Lung/virology , Macrophages/metabolism , Macrophages/pathology , Macrophages/virology , Mitochondria/pathology , Mitochondria/virology , PPAR gamma/genetics , Zonula Occludens-1 Protein/genetics , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/pharmacology
10.
Am J Physiol Cell Physiol ; 317(2): C390-C397, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31091144

ABSTRACT

Chronic HIV infection causes redox stress and increases the risk of acute and chronic lung injury, even when individuals are adherent to antiretroviral therapy. HIV-1 transgene expression in rats inhibits nuclear factor (erythroid-derived 2)-like 2 (Nrf2), which regulates antioxidant defenses and alveolar epithelial cell (AEC) barrier function, but the mechanism is unknown. In this study, we present novel evidence that these pathological effects of HIV are mediated by microRNA-144 (miR-144). HIV-1 transgene expression in vivo increases the expression of miR-144 in the alveolar epithelium, and this can be replicated by direct exposure of naïve primary AECs to either Tat or gp120 ex vivo. Further, treating naïve primary AECs with a miR-144 mimic decreased the expression and activity of Nrf2 and inhibited their barrier formation. In contrast, treatment with a miR-144 antagomir increased the expression and activity of Nrf2 and improved barrier function in primary AECs isolated from HIV-1 transgenic rats. Importantly, either delivering the miR-144 antagomir intratracheally, or directly activating Nrf2 by dietary treatment with PB123, increased Nrf2 expression and barrier formation in HIV-1 transgenic rat AECs. This study provides new experimental evidence that HIV-induced inhibition of Nrf2 and consequent AEC barrier dysfunction are mediated via miR-144, and that these pathophysiological effects can be mitigated in vivo by either directly antagonizing miR-144 or activating Nrf2. Our findings suggest that targeting the inhibition of Nrf2 in individuals living with HIV could enhance their lung health and decrease the lung-specific morbidity and mortality that persists despite antiretroviral therapy.


Subject(s)
Alveolar Epithelial Cells/metabolism , HIV Infections/metabolism , HIV-1/metabolism , MicroRNAs/metabolism , NF-E2-Related Factor 2/metabolism , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/virology , Animals , Antagomirs/pharmacology , Cells, Cultured , Disease Models, Animal , HIV Infections/drug therapy , HIV Infections/genetics , HIV Infections/virology , HIV-1/genetics , Host-Pathogen Interactions , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , NF-E2-Related Factor 2/agonists , NF-E2-Related Factor 2/genetics , Rats, Inbred F344 , Rats, Transgenic , Signal Transduction
11.
Alcohol Clin Exp Res ; 43(7): 1427-1438, 2019 07.
Article in English | MEDLINE | ID: mdl-31081931

ABSTRACT

BACKGROUND: Alcohol exposure induces TGFß1 and renders the lung susceptible to injury and disrepair. We determined that TGFß1 regulates myofibroblast differentiation through the loss of Thy-1 expression and consequent induction of α-SMA. TGFß1 is important for T helper 17 (Th17) differentiation and IL-17 secretion, which in turn participates in tissue repair. We hypothesized that alcohol induces Th17 differentiation via TGFß1 and that IL-17 produced by these cells contributes to the development of profibrotic lung myofibroblasts. METHODS: Primary lung fibroblasts (PLFs) were treated with alcohol, TGFß1, and IL-17 and then analyzed for Thy-1 expression and cell morphology. Naïve and Th17-polarized CD4+ T cells were exposed to alcohol and assessed for IL-17 expression. CD4+ T cells from alcohol-fed mice were analyzed for Th17 and IL-17 expression. Lungs of control-fed, bleomycin-treated and alcohol-fed, bleomycin-treated mice were analyzed for IL-17 protein expression. RESULTS: Alcohol-treated PLFs expressed lower levels of Thy-1 than untreated cells. TGFß1 or IL-17 exposure suppressed PLF Thy-1 expression. When administered together, TGFß1 and IL-17 additively down-regulated Thy-1 expression. Exposure of naïve and Th17-polarized CD4+ T cells to alcohol induced the Th17 phenotype and augmented their production of IL-17. CD4+ Th17+ levels are elevated in the peripheral compartment but not in the lungs of alcohol-fed animals. Treatment of the PLFs with IL-17 and alcohol induced α-SMA expression. Induction of α-SMA and myofibroblast morphology by IL-17 occurred selectively in a Thy-1- fibroblast subpopulation. Chronic alcohol ingestion augmented lung-specific IL-17 expression following bleomycin-induced lung injury. CONCLUSIONS: Alcohol exposure skews T cells toward a Th17 immune response that in turn primes the lung for fibroproliferative disrepair through loss of Thy-1 expression and induction of myofibroblast differentiation. These effects suggest that IL-17 and TGFß1 contribute to fibroproliferative disrepair in the lung and targeting these proteins could limit morbidity and mortality following lung injury in alcoholic individuals.


Subject(s)
Cell Differentiation/drug effects , Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Fibroblasts/drug effects , Interleukin-17/biosynthesis , Lung/pathology , Myofibroblasts/drug effects , Thy-1 Antigens/biosynthesis , Thy-1 Antigens/genetics , Actins/biosynthesis , Actins/genetics , Animals , CD4 Lymphocyte Count , Cell Transdifferentiation/drug effects , Down-Regulation/drug effects , Lung/drug effects , Lymphotoxin-alpha/biosynthesis , Mice , Mice, Inbred C57BL , T-Lymphocytes/drug effects
12.
Am J Med Sci ; 357(6): 483-491, 2019 06.
Article in English | MEDLINE | ID: mdl-31000424

ABSTRACT

BACKGROUND: Individuals with HIV have ∼2-fold increased risk of developing pulmonary fibrosis. The mechanism(s) by which this occurs has yet to be determined. HIV-1 protein gp120 activates CXCR4 in the lymphocyte, promoting a variety of intracellular signaling pathways including those common to TGFß1 associated with lung fibroblast-to-myofibroblast transdifferentiation. We hypothesized that gp120 promotes pulmonary fibrotic changes via activation of CXCR4 in the lung fibroblast. METHODS: Mouse primary lung fibroblasts (PLFs) were cultured ± gp120, then analyzed for α-SMA expression and stress fiber formation. In parallel, PLFs were cultured ± gp120 ± AMD3100 (a CXCR4 antagonist), and α-SMA, pan and phospho-Akt, and total and phospho-MAPK (or ERK1/2) protein expression was quantified. Finally, lungs and PLFs from wild-type and HIV-1 transgenic mice were analyzed for hydroxyproline and α-SMA content. RESULTS: gp120 treatment increased α-SMA expression and myofibroblast differentiation in PLFs. gp120 treatment activated phosphorylation of ERK1/2, but not PI3K-Akt. Pretreatment with AMD3100 inhibited gp120-induced ERK1/2 phosphorylation and gp120-induced α-SMA expression. In parallel, there was a significant increase in hydroxyproline content in lungs from older HIV-1 transgenic mice and a >3-fold increase in α-SMA expression in PLFs isolated from HIV-1 transgenic mice. CONCLUSIONS: gp120 induces α-SMA expression and fibroblast-to-myofibroblast transdifferentiation by activating the CXCR4-ERK1/2 signaling pathway in mouse PLFs. Lungs of older HIV-1 transgenic mice contain higher hydroxyproline content and their PLFs have a striking increase in α-SMA expression. These results suggest a mechanism by which individuals with HIV are at increased risk of developing pulmonary fibrotic changes as they age.


Subject(s)
Cell Transdifferentiation , Fibroblasts/metabolism , HIV Envelope Protein gp120/physiology , Pulmonary Fibrosis/etiology , Receptors, CXCR4/metabolism , Actins/metabolism , Animals , Benzylamines , Cyclams , Female , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/therapeutic use , Hydroxyproline/metabolism , MAP Kinase Signaling System , Male , Mice, Transgenic , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/prevention & control , Receptors, CXCR4/antagonists & inhibitors
13.
Int Forum Allergy Rhinol ; 8(10): 1184-1189, 2018 10.
Article in English | MEDLINE | ID: mdl-29897665

ABSTRACT

BACKGROUND: The endocannabinoid system represents a highly conserved, innate signaling network with direct and indirect control of eicosanoid-mediated inflammation. Activation of the type 2 cannabinoid receptor (CB2R) leads to decreased type 2 inflammation and reduced production of arachidonic acid (AA). Given that altered AA metabolism is associated with aspirin-exacerbated respiratory disease (AERD), we hypothesized that expression of the CB2R gene CNR2 is increased in AERD. METHODS: Nasal polyps from consecutive patients undergoing endoscopic sinus surgery for AERD or allergic fungal rhinosinusitis (AFRS) were prospectively evaluated. Control sphenoid mucosa was collected from patients undergoing endoscopic skull base procedures. Expression and localization of endocannabinoid receptors were evaluated by quantitative reverse transcript-polymerase chain reaction (qRT-PCR) and immunohistochemistry. A 2-group unpaired t test with unequal variances was used to evaluate group differences. RESULTS: Thirteen subjects were included in this pilot study, including 5 controls, 5 AFRS patients, and 3 AERD patients. Upregulated expression of CNR2 was detected in subjects with AERD vs both AFRS (p = 0.049) and controls (p = 0.047), with a mean increase of 5.2-fold. No significant differences in expression of the CB1R gene CNR1 were detected between control and AFRS groups. Immunohistochemistry predominantly localized CB1R and CB2R expression to the surface epithelium in all subjects. CONCLUSION: The endocannabinoid system is an emerging immunomodulatory network that may be involved in AERD. This is the first study of CB2R in sinonasal disease, showing significantly increased transcription in nasal polyps from subjects with AERD. Additional study is warranted to further evaluate the contribution and therapeutic potential of this novel finding in chronic rhinosinusitis.


Subject(s)
Asthma, Aspirin-Induced/genetics , Receptor, Cannabinoid, CB2/genetics , Up-Regulation , Adolescent , Adult , Asthma, Aspirin-Induced/metabolism , Asthma, Aspirin-Induced/pathology , Chronic Disease , Epithelium/metabolism , Female , Humans , Male , Middle Aged , Nasal Mucosa/metabolism , Nasal Mucosa/pathology , Nasal Polyps/metabolism , Nasal Polyps/pathology , Pilot Projects , Receptor, Cannabinoid, CB2/metabolism , Rhinitis, Allergic/genetics , Rhinitis, Allergic/metabolism , Rhinitis, Allergic/pathology , Sinusitis/metabolism , Sinusitis/pathology , Young Adult
14.
Am J Med Sci ; 355(5): 497-505, 2018 05.
Article in English | MEDLINE | ID: mdl-29753380

ABSTRACT

BACKGROUND: Alcohol significantly impairs antioxidant defenses and innate immune function in the lung and increases matrix metalloproteinase 9 (MMP-9) activity. The receptor for advanced glycation end products (RAGE) is a well-characterized marker of lung injury that is cleaved by MMP-9 into soluble RAGE and has not yet been examined in the alcoholic lung. We hypothesized that chronic alcohol ingestion would impair RAGE signaling via MMP-9 in the alveolar macrophage and thereby impair innate immune function. MATERIALS AND METHODS: Primary alveolar macrophages were isolated from control-fed or alcohol-fed rats. Real-time polymerase chain reaction (qRT-PCR), Western blotting, and enzyme-linked immunosorbent assays were performed to evaluate RAGE expression. Silencing of MMP-9 ribonucleic acid (RNA) in a rat alveolar macrophage cell line was confirmed by qRT-PCR, and immunofluorescence (IF) was used to assess the association between alcohol, MMP-9, and RAGE. Phagocytosis was assessed using flow cytometry. Sulforaphane and glutathione were used to assess the relationship between oxidative stress and RAGE. RESULTS: RAGE messenger RNA expression was significantly increased in the alveolar macrophages of alcohol-fed rats, but IF showed that membrane-bound RAGE protein expression was decreased. Lavage fluid demonstrated increased levels of soluble RAGE (sRAGE). Decreasing MMP-9 expression using si-MMP-9 abrogated the effects of alcohol on RAGE protein. Phagocytic function was suppressed by direct RAGE inhibition, and the impairment was reversed by antioxidant treatment. CONCLUSIONS: Chronic alcohol ingestion reduces RAGE protein expression and increases the amount of sRAGE in alveolar lavage fluid, likely via cleavage by MMP-9. In addition, it impairs phagocytic function. Antioxidants restore membrane-bound RAGE and phagocytic function.


Subject(s)
Alcoholism/immunology , Immunity, Innate , Macrophages, Alveolar/immunology , Phagocytosis/immunology , Receptor for Advanced Glycation End Products/metabolism , Alcoholism/metabolism , Animals , Cells, Cultured , Ethanol/administration & dosage , Immunity, Innate/drug effects , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Male , Matrix Metalloproteinase 9/metabolism , Phagocytosis/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/immunology
15.
Alcohol Res ; 38(2): 243-254, 2017.
Article in English | MEDLINE | ID: mdl-28988576

ABSTRACT

Among the many organ systems affected by harmful alcohol use, the lungs are particularly susceptible to infections and injury. The mechanisms responsible for rendering people with alcohol use disorder (AUD) vulnerable to lung damage include alterations in host defenses of the upper and lower airways, disruption of alveolar epithelial barrier integrity, and alveolar macrophage immune dysfunction. Collectively, these derangements encompass what has been termed the "alcoholic lung" phenotype. Alcohol-related reductions in antioxidant levels also may contribute to lung disease in people with underlying AUD. In addition, researchers have identified several regulatory molecules that may play crucial roles in the alcohol-induced disease processes. Although there currently are no approved therapies to combat the detrimental effects of chronic alcohol consumption on the respiratory system, these molecules may be potential therapeutic targets to guide future investigation.


Subject(s)
Alcohol-Related Disorders/complications , Lung Diseases/etiology , Animals , Humans , Lung Diseases/drug therapy , Lung Diseases/metabolism
16.
17.
J Leukoc Biol ; 102(2): 517-525, 2017 08.
Article in English | MEDLINE | ID: mdl-28550120

ABSTRACT

Respiratory complications occur frequently in individuals living with human immunodeficiency-1 virus (HIV) infection, and there is evidence that HIV-related oxidative stress impairs alveolar macrophage immune function. We hypothesized that nuclear factor (erythroid-derived 2)-like 2 (Nrf2), a master transcription factor that activates the antioxidant response element (ARE) and regulates antioxidant defenses, has an important role in alveolar macrophage (AMs) immune dysfunction in individuals with HIV infections. To test that hypothesis, we analyzed human monocyte-derived macrophages (MDMs) that were either infected with HIV-1 or were exposed to the HIV-related proteins gp120 and Tat ex vivo and determined that either stress affected the expression of Nrf2 and the Nrf2-ARE-dependent genes for NAD(P)H dehydrogenase, quinone 1 (NQO1) and glutamate-cysteine ligase, catalytic subunit (GCLC). We then determined that the expression of Nrf2, NQO1, and GCLC was significantly decreased in primary AMs isolated from HIV-1 transgenic rats. In parallel, treating a rat macrophage cell line (NR8383 cells) with the HIV-related proteins gp120 or Tat similarly decreased the gene and protein expression of Nrf2, NQO1, and GCLC. Further, phagocytic function was decreased in both human MDMs infected with HIV-1 and primary AMs from HIV-1 transgenic rats. Importantly, treating HIV-1-infected human MDMs or AMs from HIV-1 transgenic rats with sulforaphane (SFN, an Nrf2 activator) significantly improved their phagocytic function. The salutary effects of SFN were abrogated by silencing RNA to Nrf2 in wild-type rat macrophages. Our findings demonstrate that HIV-1 infection and exposure to HIV-1-related proteins inhibit Nrf2-ARE activity in the AMs and impair their phagocytic function. Treatments targeted at increasing Nrf2-ARE activity could, therefore, enhance lung innate immunity in people living with HIV-1.


Subject(s)
Antioxidant Response Elements/immunology , Gene Expression Regulation/immunology , HIV Infections/immunology , Macrophages, Alveolar/immunology , NF-E2-Related Factor 2/immunology , Animals , Blotting, Western , HIV-1/immunology , Humans , Macrophages, Alveolar/virology , NF-E2-Related Factor 2/metabolism , Phagocytosis/immunology , Rats , Rats, Inbred F344 , Rats, Transgenic , Real-Time Polymerase Chain Reaction
18.
AIDS Res Hum Retroviruses ; 33(10): 1018-1026, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28314381

ABSTRACT

Despite antiretroviral therapy (ART), respiratory infections increase mortality in individuals living with chronic human immunodeficiency virus (HIV) infection. In experimental and clinical studies of chronic HIV infection, alveolar macrophages (AMs) exhibit impaired phagocytosis and bacterial clearance. Peroxisome proliferator-activated receptor (PPAR)γ, NADPH oxidase (Nox) isoforms Nox1, Nox2, Nox4, and transforming growth factor-beta 1 (TGFß1) are critical mediators of AM oxidative stress and phagocytic dysfunction. Therefore, we hypothesized that HIV alters AM expression of these targets, resulting in chronic lung oxidative stress and subsequent immune dysfunction. A cross-sectional study of HIV-infected (n = 22) and HIV-uninfected (n = 6) subjects was conducted. Bronchoalveolar lavage (BAL) was performed, and AMs were isolated. Lung H2O2 generation was determined by measuring H2O2 in the BAL fluid. In AMs, PPARγ, Nox1, Nox2, Nox4, and TGFß1 mRNA (quantitative real-time polymerase chain reaction) and protein (fluorescent immunomicroscopy) levels were assessed. Compared with HIV-uninfected (control) subjects, HIV-infected subjects were relatively older and the majority were African American; ∼86% were on ART, and their median CD4 count was 445, with a median viral load of 0 log copies/ml. HIV infection was associated with increased H2O2 in the BAL, decreased AM mRNA and protein levels of PPARγ, and increased AM mRNA and protein levels of Nox1, Nox2, Nox4, and TGFß1. PPARγ attenuation and increases in Nox1, Nox2, Nox4, and TGFß1 contribute to AM oxidative stress and immune dysfunction in the AMs of otherwise healthy HIV-infected subjects. These findings provide novel insights into the molecular mechanisms by which HIV increases susceptibility to pulmonary infections.


Subject(s)
HIV Infections/pathology , Macrophages, Alveolar/immunology , NADPH Oxidases/metabolism , Oxidative Stress/immunology , PPAR gamma/metabolism , Transforming Growth Factor beta1/metabolism , Adult , Anti-HIV Agents/therapeutic use , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , CD4 Lymphocyte Count , Cells, Cultured , Cross-Sectional Studies , Female , HIV Infections/drug therapy , HIV Infections/virology , Humans , Hydrogen Peroxide/analysis , Male , Middle Aged , NADPH Oxidases/genetics , PPAR gamma/genetics , Phagocytosis/immunology , RNA, Messenger/genetics , Transforming Growth Factor beta1/genetics , Viral Load
19.
Sci Rep ; 7: 42028, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28181540

ABSTRACT

Triggering receptor expressed on myeloid cells-1(TREM-1) is a member of the superimmunoglobulin receptor family. We have previously shown that TREM-1 prolongs survival of macrophages treated with lipoolysaccharide through Egr2-Bcl2 signaling. Recent studies suggest a role for TREM-1 in viral immunity. Human immunodeficiency virus-1 (HIV) targets the monocyte/macrophage lineage at varying stages of infection. Emerging data suggest that macrophages are key reservoirs for latent HIV even in individuals on antiretroviral therapy. Here, we investigated the potential role of TREM-1 in HIV latency in macrophages. Our data show that human macrophages infected with HIV show an increased expression of TREM-1. In parallel, direct exposure to the HIV-related proteins Tat or gp120 induces TREM-1 expression in macrophages and confers anti-apoptotic attributes.NF-κB p65 silencing identified that these proteins induce TREM-1 in p65-dependent manner. TREM-1 silencing in macrophages exposed to HIV-related proteins led to increased caspase 3 activation and reduced Bcl-2 expression, rendering them susceptible to apotosis. These novel data reveal that TREM-1 may play a critical role in establishing HIV reservoir in macrophages by inhibiting apoptosis. Therefore, targeting TREM-1 could be a novel therapeutic approach to enhance clearance of the HIV reservoir, at least within the macrophage pools.


Subject(s)
HIV Envelope Protein gp120/metabolism , HIV Infections/virology , Host-Pathogen Interactions , Macrophages/virology , Triggering Receptor Expressed on Myeloid Cells-1/metabolism , Virus Latency , tat Gene Products, Human Immunodeficiency Virus/metabolism , Cell Survival , Cells, Cultured , Humans , Macrophages/physiology
20.
Am J Respir Cell Mol Biol ; 56(5): 563-567, 2017 05.
Article in English | MEDLINE | ID: mdl-27911588

ABSTRACT

The advent of antiretroviral therapy has transformed infection by the type 1 human immunodeficiency virus (HIV) from a rapidly fatal disease to a chronic illness with excellent long-term survival rates. Although HIV primarily targets the adaptive arm of host immunity, it simultaneously impacts the innate immune system, and has profound implications for lung health, even when viral suppression is achieved with antiretroviral therapy. The lung has evolved a unique array of innate immune defenses, and the pathophysiological interactions between HIV and the pulmonary innate immune system deserve particular attention. In this review, we discuss work that elucidates how the components of innate immunity both respond to and are perturbed by infection with HIV.


Subject(s)
HIV/immunology , Immunity, Innate/immunology , Humans , Lung/immunology , Models, Biological
SELECTION OF CITATIONS
SEARCH DETAIL
...