Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Front Immunol ; 15: 1374818, 2024.
Article in English | MEDLINE | ID: mdl-38827738

ABSTRACT

Activated lung ILC2s produce large quantities of IL-5 and IL-13 that contribute to eosinophilic inflammation and mucus production following respiratory syncytial virus infection (RSV). The current understanding of ILC2 activation during RSV infection, is that ILC2s are activated by alarmins, including IL-33, released from airway epithelial cells in response to viral-mediated damage. Thus, high levels of RSV neutralizing maternal antibody generated from maternal immunization would be expected to reduce IL-33 production and mitigate ILC2 activation. Here we report that lung ILC2s from mice born to RSV-immunized dams become activated despite undetectable RSV replication. We also report, for the first time, expression of activating and inhibitory Fcgamma receptors on ILC2s that are differentially expressed in offspring born to immunized versus unimmunized dams. Alternatively, ex vivo IL-33-mediated activation of ILC2s was mitigated following the addition of antibody: antigen immune complexes. Further studies are needed to confirm the role of Fcgamma receptor ligation by immune complexes as an alternative mechanism of ILC2 regulation in RSV-associated eosinophilic lung inflammation.


Subject(s)
Interleukin-33 , Lung , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections , Respiratory Syncytial Viruses , Animals , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/prevention & control , Mice , Female , Lung/immunology , Lung/virology , Interleukin-33/immunology , Respiratory Syncytial Viruses/immunology , Lymphocytes/immunology , Immunization , Receptors, IgG/immunology , Receptors, IgG/metabolism , Antibodies, Viral/immunology , Pregnancy , Respiratory Syncytial Virus Vaccines/immunology
2.
Front Immunol ; 14: 1206026, 2023.
Article in English | MEDLINE | ID: mdl-37646035

ABSTRACT

Respiratory syncytial virus (RSV) is the leading cause of childhood hospitalizations due to bronchiolitis in children under 5 years of age. Moreover, severe RSV disease requiring hospitalization is associated with the subsequent development of wheezing and asthma. Due to the young age in which viral protection is needed and risk of vaccine enhanced disease following direct infant vaccination, current approaches aim to protect young children through maternal immunization strategies that boost neutralizing maternal antibody (matAb) levels. However, there is a scarcity of studies investigating the influence of maternal immunization on secondary immune responses to RSV in the offspring or whether the subsequent development of wheezing and asthma is mitigated. Toward this goal, our lab developed a murine model of maternal RSV vaccination and repeat RSV exposure to evaluate the changes in immune response and development of exacerbated lung inflammation on secondary RSV exposure in mice born to immunized dams. Despite complete protection following primary RSV exposure, offspring born to pre-fusion F (PreF)-vaccinated dams had exaggerated secondary ILC2 and Th2 responses, characterized by enhanced production of IL-4, IL-5, and IL-13. These enhanced type 2 cellular responses were associated with exaggerated airway eosinophilia and mucus hyperproduction upon re-exposure to RSV. Importantly, depletion of CD4+ T cells led to complete amelioration of the observed type 2 pathology on secondary RSV exposure. These unanticipated results highlight the need for additional studies that look beyond primary protection to better understand how maternal immunization shapes subsequent immune responses to repeat RSV exposure.


Subject(s)
Asthma , Pneumonia , Respiratory Syncytial Virus, Human , Animals , Mice , CD4-Positive T-Lymphocytes , Immunity, Innate , Respiratory Sounds , Pneumonia/prevention & control
3.
J Clin Med ; 12(10)2023 May 09.
Article in English | MEDLINE | ID: mdl-37240472

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a disease of the airways and lungs due to an enhanced inflammatory response, commonly caused by cigarette smoking. Patients with COPD are often multimorbid, as they commonly suffer from multiple chronic (inflammatory) conditions. This intensifies the burden of individual diseases, negatively affects quality of life, and complicates disease management. COPD and comorbidities share genetic and lifestyle-related risk factors and pathobiological mechanisms, including chronic inflammation and oxidative stress. The receptor for advanced glycation end products (RAGE) is an important driver of chronic inflammation. Advanced glycation end products (AGEs) are RAGE ligands that accumulate due to aging, inflammation, oxidative stress, and carbohydrate metabolism. AGEs cause further inflammation and oxidative stress through RAGE, but also through RAGE-independent mechanisms. This review describes the complexity of RAGE signaling and the causes of AGE accumulation, followed by a comprehensive overview of alterations reported on AGEs and RAGE in COPD and in important co-morbidities. Furthermore, it describes the mechanisms by which AGEs and RAGE contribute to the pathophysiology of individual disease conditions and how they execute crosstalk between organ systems. A section on therapeutic strategies that target AGEs and RAGE and could alleviate patients from multimorbid conditions using single therapeutics concludes this review.

4.
Front Immunol ; 14: 1039997, 2023.
Article in English | MEDLINE | ID: mdl-36776857

ABSTRACT

Background: Asthma is a major public healthcare burden, affecting over 300 million people worldwide. While there has been great progress in the treatment of asthma, subsets of patients who present with airway neutrophilia, often have more severe disease, and tend to be resistant to conventional corticosteroid treatments. The receptor for advanced glycation endproducts (RAGE) plays a central role in the pathogenesis of eosinophilic asthma, however, it's role in neutrophilic asthma remains largely uninvestigated. Methods: A mouse model of severe steroid resistant neutrophilic airway disease (SSRNAD) using the common fungal allergen Alternaria alternata (AA) was employed to evaluate the effects of genetic ablation of RAGE and pharmacological inhibition of the NLRP3 inflammasome on neutrophilic airway inflammation. Results: AA exposure induced robust neutrophil-dominant airway inflammation and increased BALF levels of Th1/Th17 cytokines in wild-type mice, which was significantly reduced in RAGE-/- mice. Serum levels of IgE and IgG1 were increased similarly in both wild-type and RAGE-/- mice. Pharmacological inhibition of NLRP3 blocked the effects of AA exposure and NLRP3 inflammasome activation was RAGE-dependent. Neutrophil extracellular traps were elevated in the BALF of wild-type but not RAGE-/- mice and an atypical population of SiglecF+ neutrophils were identified in the BALF. Lastly, time-course studies found that RAGE expression promoted sustained neutrophil accumulation in the BALF of mice in response to AA.


Subject(s)
Asthma , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Receptor for Advanced Glycation End Products , Animals , Mice , Allergens , Asthma/metabolism , Inflammation , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Receptor for Advanced Glycation End Products/genetics
5.
Front Immunol ; 13: 1025341, 2022.
Article in English | MEDLINE | ID: mdl-36268035

ABSTRACT

Respiratory syncytial virus (RSV) remains the most common cause of lower respiratory tract infections in children worldwide. Development of a vaccine has been hindered due the risk of enhanced respiratory disease (ERD) following natural RSV exposure and the young age (<6 months) at which children would require protection. Risk factors linked to the development of ERD include poorly neutralizing antibody, seronegative status (never been exposed to RSV), and a Th2-type immune response. Stabilization of the more antigenic prefusion F protein (PreF) has reinvigorated hope for a protective RSV vaccine that elicits potent neutralizing antibody. While anecdotal evidence suggests that children and adults previously exposed to RSV (seropositive) are not at risk for developing vaccine associated ERD, differences in host immune responses in seropositive and seronegative individuals that may protect against ERD remain unclear. It is also unclear if vaccine formulations that skew towards Th1- versus Th2-type immune responses increase pathology or provide greater protection in seropositive individuals. Therefore, the goal of this work was to compare the host immune response to a stabilized prefusion RSV antigen formulated alone or with Th1 or Th2 skewing adjuvants in seronegative and seropositive BALB/c mice. We have developed a novel BALB/c mouse model whereby mice are first infected with RSV (seropositive) and then vaccinated during pregnancy to recapitulate maternal immunization strategies. Results of these studies show that prior RSV infection mitigates vaccine-mediated skewing by Th1- and Th2-polarizing adjuvants that was observed in seronegative animals. Moreover, vaccination with PreF plus the Th1-skewing adjuvant, Advax, increased RSV F85-93-specific CD8 T cells in both seronegative and seropositive dams. These data demonstrate the importance of utilizing seropositive animals in preclinical vaccine studies to assess both the safety and efficacy of candidate RSV vaccines.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Virus, Human , Mice , Animals , Antibodies, Viral , Lung , Antibodies, Neutralizing , Mice, Inbred BALB C , CD8-Positive T-Lymphocytes , Adjuvants, Immunologic
6.
Ther Adv Respir Dis ; 15: 17534666211016071, 2021.
Article in English | MEDLINE | ID: mdl-34275342

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal lung disease in which most patients die within 3 years of diagnosis. With an unknown etiology, IPF results in progressive fibrosis of the lung parenchyma, diminishing normal lung function, which results in respiratory failure, and eventually, death. While few therapies are available to reduce disease progression, patients continue to advance toward respiratory failure, leaving lung transplantation the only viable option for survival. As incidence and mortality rates steadily increase, the need for novel therapeutics is imperative. The receptor for advanced glycation endproducts (RAGE) is most highly expressed in the lungs and plays a significant role in a number of chronic lung diseases. RAGE has long been linked to IPF; however, confounding data from both human and experimental studies have left an incomplete and perplexing story. This review examines the present understanding of the role of RAGE in human and experimental models of IPF, drawing parallels to recent advances in RAGE biology. Moreover, this review discusses the role of RAGE in lung injury response, type 2 immunity, and cellular senescence, and how such mechanisms may relate to RAGE as both a biomarker of disease progression and potential therapeutic target in IPF.The reviews of this paper are available via the supplemental material section.


Subject(s)
Pulmonary Fibrosis , Receptor for Advanced Glycation End Products , Disease Progression , Humans , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/pathology , Receptor for Advanced Glycation End Products/metabolism
7.
Allergy ; 76(5): 1350-1366, 2021 05.
Article in English | MEDLINE | ID: mdl-32976640

ABSTRACT

Asthma is a generalized term that describes a scope of distinct pathologic phenotypes of variable severity, which share a common complication of reversible airflow obstruction. Asthma is estimated to affect almost 400 million people worldwide, and nearly ten percent of asthmatics have what is considered "severe" disease. The majority of moderate to severe asthmatics present with a "type 2-high" (T2-hi) phenotypic signature, which pathologically is driven by the type 2 cytokines Interleukin-(IL)-4, IL-5, and IL-13. However, "type 2-low" (T2-lo) phenotypic signatures are often associated with more severe, steroid-refractory neutrophilic asthma. A wide range of clinical and experimental studies have found that the receptor for advanced glycation endproducts (RAGE) plays a significant role in the pathogenesis of asthma and allergic airway disease (AAD). Current experimental data indicates that RAGE is a critical mediator of the type 2 inflammatory reactions which drive the development of T2-hi AAD. However, clinical studies demonstrate that increased RAGE ligands and signaling strongly correlate with asthma severity, especially in severe neutrophilic asthma. This review presents an overview of the current understandings of RAGE in asthma pathogenesis, its role as a biomarker of disease, and future implications for mechanistic studies, and potential therapeutic intervention strategies.


Subject(s)
Asthma , Hypersensitivity/diagnosis , Receptor for Advanced Glycation End Products , Asthma/diagnosis , Cytokines , Humans , Interleukin-13 , Lung
8.
Vaccine ; 38(50): 7885-7891, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33129608

ABSTRACT

Respiratory syncytial virus (RSV) commonly causes severe respiratory tract infections in infants, peaking between 2 and 6 months of age; an age at which direct vaccination is unlikely to be effective. Maternal immunization can deliver high levels of antibodies to newborns, providing immediate protection. Following natural infection, antibodies targeting the prefusion conformation of RSV F protein (PreF) have the greatest neutralizing capacity and thus, may provide infants with a high degree of RSV protection when acquired through maternal vaccination. However, the influence of anti-PreF maternal antibodies on infant immunity following RSV exposure has not been elucidated. To address this knowledge gap, offspring born to dams immunized with a RSV PreF vaccine formulation were challenged with RSV and their immune responses were analyzed over time. These studies demonstrated safety and efficacy for RSV-challenged, maternally-immunized offspring but high and waning maternal antibody levels were associated with differential innate and T cell immunity.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Antibodies, Neutralizing , Antibodies, Viral , Humans , Immunization , Infant , Infant, Newborn , Respiratory Syncytial Virus Infections/prevention & control , T-Lymphocytes , Vaccination , Viral Fusion Proteins
9.
Vaccine ; 38(41): 6357-6362, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32829976

ABSTRACT

Respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract infections among infants with most infections occurring in the first year of life. Multiple RSV exposures are required for children to mount adult-like immune responses. Although adult RSV immunity is associated with less severe disease, the protection induced through natural infection is short-lived. Therefore, vaccination of RSV-experienced young children may accelerate immunity and provide long-term protection from RSV reinfection. However, the extent to which different Th-biased vaccine regimens influence pre-existing humoral and cellular immunity in RSV-experienced young children is unknown. To address this question, infant BALB/c mice were RSV-infected and subsequently immunized with the prefusion RSV F (PreF) antigen formulated with either a Th2-skewing (Alum) or Th1/Th2-balanced (Advax-SM) adjuvant. These studies show that both adjuvants boosted neutralizing antibody and protected from RSV reinfection, but Advax-SM adjuvant prevented the Th2-skewed immunity observed in RSV-experienced young mice immunized with PreF/Alum.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Virus, Human , Animals , Antibodies, Viral , Lung , Mice , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections/prevention & control
10.
Front Immunol ; 11: 1673, 2020.
Article in English | MEDLINE | ID: mdl-32849580

ABSTRACT

Respiratory syncytial virus (RSV) remains the most common cause of lower respiratory tract infections in children worldwide. Development of a vaccine has been hindered by the risk of developing enhanced respiratory disease (ERD) upon natural exposure to the virus. Generation of higher quality neutralizing antibodies with stabilized pre-fusion F protein antigens has been proposed as a strategy to prevent ERD. We sought to test whether there was evidence of ERD in naïve BALB/c mice immunized with an unadjuvanted, stabilized pre-fusion F protein, and challenged with RSV line 19. We further sought to determine the extent to which formulation with a Th2-biased (alum) or a more Th1/Th2-balanced (Advax-SM) adjuvant influenced cellular responses and lung pathology. When exposed to RSV, mice immunized with pre-fusion F protein alone (PreF) exhibited increased airway eosinophilia and mucus accumulation. This was further exacerbated by formulation of PreF with Alum (aluminum hydroxide). Conversely, formulation of PreF with a Th1/Th2-balanced adjuvant, Advax-SM, not only suppressed RSV viral replication, but also inhibited airway eosinophilia and mucus accumulation. This was associated with lower numbers of lung innate lymphocyte cells (ILC2s) and CD4+ T cells producing IL-5+ or IL-13+ and increased IFNγ+ CD4+ and CD8+ T cells, in addition to RSV F-specific CD8+ T cells. These data suggest that in the absence of preimmunity, stabilized PreF antigens may still be associated with aberrant Th2 responses that induce lung pathology in response to RSV infection, and can be prevented by formulation with more Th1/Th2-balanced adjuvants that enhance CD4+ and CD8+ IFNγ+ T cell responses. This may support the use of stabilized PreF antigens with Th1/Th2-balanced adjuvants like, Advax-SM, as safer alternatives to alum in RSV vaccine candidates.


Subject(s)
Adjuvants, Immunologic/pharmacology , Aluminum Hydroxide/pharmacology , Lung/drug effects , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/pharmacology , Respiratory Syncytial Viruses/drug effects , Th2 Cells/drug effects , Viral Fusion Proteins/pharmacology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Immunity, Humoral/drug effects , Immunization , Immunogenicity, Vaccine/drug effects , Lung/immunology , Lung/pathology , Lung/virology , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/immunology , Respiratory Syncytial Viruses/pathogenicity , Th1-Th2 Balance/drug effects , Th2 Cells/immunology , Th2 Cells/metabolism , Th2 Cells/virology
11.
J Allergy Clin Immunol ; 144(3): 796-808.e12, 2019 09.
Article in English | MEDLINE | ID: mdl-30940519

ABSTRACT

BACKGROUND: Asthma is estimated to effect more than 300 million persons worldwide, leading to nearly 250,000 deaths annually. The majority of patients with mild-to-severe asthma have what is deemed "type-2 high" asthma, which is driven by the prototypical type 2 cytokines IL-4, IL-5, and IL-13. Studies have indicated that the receptor for advanced glycation end products (RAGE) is a critical molecule in the pathogenesis of experimental asthma/allergic airway inflammation. More specifically, RAGE expressed on stromal cells, rather than hematopoietic cells, is critical to induction of asthma/allergic airway inflammation by driving type 2 inflammatory responses. However, the role of RAGE in directly mediating type 2 cytokine signaling has never been investigated. OBJECTIVE: The goal of this study was to test the hypothesis that RAGE mediates type 2 cytokine-induced signal transduction, airway inflammation, and mucus metaplasia in the lungs. METHODS: Wild-type (WT) and RAGE knockout (RAGE-/-) mice, were intranasally administered rIL-5/rIL-13 or rIL-4 alone, and signal transducer and activator of transcription 6 (STAT6) signaling, airway inflammation, and mucus metaplasia were assessed. A RAGE small-molecule antagonist was used to determine the effects of pharmacologically inhibiting RAGE on type 2 cytokine-induced effects. RESULTS: Administration of type 2 cytokines induced pronounced airway inflammation and mucus metaplasia in WT mice, which was nearly completely abrogated in RAGE-/- mice. In addition, treatment with a RAGE-specific antagonist diminished the effects of type 2 cytokines in WT mice and in primary human bronchial epithelial cell cultures. Genetic ablation or pharmacologic inhibition of RAGE blocks the effects of IL-13 and IL-4 by inhibiting sustained STAT6 activation and downstream target gene expression in mice and in human bronchial epithelial cells. CONCLUSIONS: This study is the first to indicate that RAGE is a critical component of type 2 cytokine signal transduction mechanisms, which is a driving force behind type 2-high asthma.


Subject(s)
Asthma/immunology , Cytokines/immunology , Lung/immunology , Receptor for Advanced Glycation End Products/immunology , Animals , Bronchoalveolar Lavage Fluid/immunology , Cells, Cultured , Epithelial Cells/immunology , Humans , Mice, Inbred C57BL , Mice, Knockout , Mucus/immunology , Receptor for Advanced Glycation End Products/genetics , Signal Transduction
12.
Toxicol Appl Pharmacol ; 348: 43-53, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29673857

ABSTRACT

Lung epithelial cells are the first cell-type to come in contact with hazardous dust materials. Upon deposition, they invoke complex reactions in attempt to eradicate particles from the airways, and repair damage. The cell surface is composed of a heterogeneous network of matrix proteins and proteoglycans, which act as scaffold and control cell-signaling networks. These functions are controlled, in part, by the sulfation patterns of heparin-sulfate proteoglycans (HSPGs), which are enzymatically regulated. Although there is evidence of altered HSPG-sulfation in idiopathic pulmonary fibrosis (IPF), this is not investigated in silicosis. Our previous studies revealed down-regulation of Sulfatase-1 (SULF1) in human bronchial epithelial cells (BECs) by crystalline silica (CS). In this study, CS-induced down-regulation of SULF1, and increases in Sulfated-HSPGs, were determined in human BECs, and in rat lungs. By siRNA and plasmid transfection techniques the effects of SULF1 expression on silica-induced fibrogenic and proliferative gene expression were determined. These studies confirmed down-regulation of SULF1 and subsequent increases in sulfated-HSPGs in vitro. Moreover, short-term exposure of rats to CS resulted in similar changes in vivo. Conversely, effects were reversed after long term CS exposure of rats. SULF1 knockdown, and overexpression alleviated and exacerbated silica-induced decrease in cell viability, respectively. Furthermore, overexpression of SULF1 promoted silica-induced proliferative and fibrogenic gene expression, and collagen production. These findings demonstrate that the HSPG modification enzyme SULF1 and HSPG sulfation are altered by CS in vitro and in vivo. Furthermore, these changes may contribute to CS-induced lung pathogenicity by affecting injury tolerance, hyperproliferation, and fibrotic effects.


Subject(s)
Cell Proliferation/drug effects , Epithelial Cells/drug effects , Lung/drug effects , Pulmonary Fibrosis/chemically induced , Silicon Dioxide/toxicity , Silicosis/etiology , Sulfotransferases/metabolism , Animals , Cell Line , Collagen/metabolism , Crystallization , Down-Regulation , Epithelial Cells/enzymology , Epithelial Cells/pathology , Female , Heparin/analogs & derivatives , Heparin/metabolism , Humans , Lung/enzymology , Lung/pathology , Proteoglycans/metabolism , Pulmonary Fibrosis/enzymology , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Rats, Wistar , Signal Transduction/drug effects , Silicon Dioxide/chemistry , Silicosis/enzymology , Silicosis/genetics , Silicosis/pathology , Sulfotransferases/genetics , Time Factors
13.
Paediatr Respir Rev ; 23: 40-49, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28416135

ABSTRACT

The receptor for advanced glycation endproducts (RAGE) is a pro-inflammatory pattern recognition receptor (PRR) that has been implicated in the pathogenesis of numerous inflammatory diseases. It was discovered in 1992 on endothelial cells and was named for its ability to bind advanced glycation endproducts and promote vascular inflammation in the vessels of patients with diabetes. Further studies revealed that RAGE is most highly expressed in lung tissue and spurred numerous explorations into RAGE's role in the lung. These studies have found that RAGE is an important mediator in allergic airway inflammation (AAI) and asthma, pulmonary fibrosis, lung cancer, chronic obstructive pulmonary disease (COPD), acute lung injury, pneumonia, cystic fibrosis, and bronchopulmonary dysplasia. RAGE has not yet been targeted in the lungs of paediatric or adult clinical populations, but the development of new ways to inhibit RAGE is setting the stage for the emergence of novel therapeutic agents for patients suffering from these pulmonary conditions.


Subject(s)
Pneumonia , Receptor for Advanced Glycation End Products , Adult , Child , Drug Discovery , Glycation End Products, Advanced/metabolism , Humans , Pneumonia/metabolism , Pneumonia/therapy , Receptor for Advanced Glycation End Products/antagonists & inhibitors , Receptor for Advanced Glycation End Products/metabolism
14.
Am J Respir Cell Mol Biol ; 56(6): 694-699, 2017 06.
Article in English | MEDLINE | ID: mdl-28080134

ABSTRACT

Bronchial epithelial cells and pulmonary endothelial cells are thought to be the primary modulators of conducting airways and vessels, respectively. However, histological examination of both mouse and human lung tissue reveals that alveolar epithelial cells (AECs) line the adventitia of large airways and vessels and thus are also in a position to directly regulate these structures. The primary purpose of this perspective is to highlight the fact that AECs coat the adventitial surface of every vessel and airway in the lung parenchyma. This localization is ideal for transmitting signals that can contribute to physiologic and pathologic responses in vessels and airways. A few examples of mediators produced by AECs that may contribute to vascular and airway responses are provided to illustrate some of the potential effects that AECs may modulate.


Subject(s)
Alveolar Epithelial Cells/metabolism , Lung/blood supply , Lung/metabolism , Alveolar Epithelial Cells/immunology , Animals , Humans , Immunity, Innate , Lung/physiology , Models, Biological
15.
Toxicol Appl Pharmacol ; 301: 61-70, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27095093

ABSTRACT

Growth and development of the mature lung is a complex process orchestrated by a number of intricate developmental signaling pathways. Wingless-type MMTV-integration site (WNT) signaling plays critical roles in controlling branching morphogenesis cell differentiation, and formation of the conducting and respiratory airways. In addition, WNT pathways are often re-activated in mature lungs during repair and regeneration. WNT- signaling has been elucidated as a crucial contributor to the development of idiopathic pulmonary fibrosis as well as other hyper-proliferative lung diseases. Silicosis, a detrimental occupational lung disease caused by excessive inhalation of crystalline silica dust, is hallmarked by repeated cycles of damaging inflammation, epithelial hyperplasia, and formation of dense, hyalinized nodules of whorled collagen. However, mechanisms of epithelial cell hyperplasia and matrix deposition are not well understood, as most research efforts have focused on the pronounced inflammatory response. Microarray data from our previous studies has revealed a number of WNT-signaling and WNT-target genes altered by crystalline silica in human lung epithelial cells. In the present study, we utilize pathway analysis to designate connections between genes altered by silica in WNT-signaling networks. Furthermore, we confirm microarray findings by QRT-PCR and demonstrate both activation of canonical (ß-catenin) and down-regulation of non-canonical (WNT5A) signaling in immortalized (BEAS-2B) and primary (PBEC) human bronchial epithelial cells. These findings suggest that WNT-signaling and cross-talk with other pathways (e.g. Notch), may contribute to proliferative, fibrogenic and inflammatory responses to silica in lung epithelial cells.


Subject(s)
Epithelial Cells/drug effects , Silicon Dioxide/toxicity , Wnt Signaling Pathway/drug effects , Cell Line , Cells, Cultured , Epithelial Cells/metabolism , Humans , Lung/cytology , Oligonucleotide Array Sequence Analysis , Transcriptome
16.
Hum Mol Genet ; 24(5): 1374-89, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25351596

ABSTRACT

Occupational and environmental exposures to airborne asbestos and silica are associated with the development of lung fibrosis in the forms of asbestosis and silicosis, respectively. However, both diseases display distinct pathologic presentations, likely associated with differences in gene expression induced by different mineral structures, composition and bio-persistent properties. We hypothesized that effects of mineral exposure in the airway epithelium may dictate deviating molecular events that may explain the different pathologies of asbestosis versus silicosis. Using robust gene expression-profiling in conjunction with in-depth pathway analysis, we assessed early (24 h) alterations in gene expression associated with crocidolite asbestos or cristobalite silica exposures in primary human bronchial epithelial cells (NHBEs). Observations were confirmed in an immortalized line (BEAS-2B) by QRT-PCR and protein assays. Utilization of overall gene expression, unsupervised hierarchical cluster analysis and integrated pathway analysis revealed gene alterations that were common to both minerals or unique to either mineral. Our findings reveal that both minerals had potent effects on genes governing cell adhesion/migration, inflammation, and cellular stress, key features of fibrosis. Asbestos exposure was most specifically associated with aberrant cell proliferation and carcinogenesis, whereas silica exposure was highly associated with additional inflammatory responses, as well as pattern recognition, and fibrogenesis. These findings illustrate the use of gene-profiling as a means to determine early molecular events that may dictate pathological processes induced by exogenous cellular insults. In addition, it is a useful approach for predicting the pathogenicity of potentially harmful materials.


Subject(s)
Asbestos, Crocidolite/toxicity , Epithelial Cells/drug effects , Gene Expression Profiling , Lung/drug effects , Silicon Dioxide/toxicity , Carcinogenesis/chemically induced , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Cluster Analysis , Dose-Response Relationship, Drug , Epithelial Cells/pathology , Humans , Inflammation/chemically induced , Inflammation/pathology , Lung/cytology , Microarray Analysis , Signal Transduction
17.
Part Fibre Toxicol ; 11: 58, 2014 Nov 19.
Article in English | MEDLINE | ID: mdl-25406505

ABSTRACT

RATIONALE: Mineral particles in the lung cause inflammation and silicosis. In myeloid and bronchial epithelial cells the inflammasome plays a role in responses to crystalline silica. Thioredoxin (TRX) and its inhibitory protein TRX-interacting protein link oxidative stress with inflammasome activation. We investigated inflammasome activation by crystalline silica polymorphs and modulation by TRX in vitro, as well as its localization and the importance of silica surface reactivity in rats. METHODS: We exposed bronchial epithelial cells and differentiated macrophages to silica polymorphs quartz and cristobalite and measured caspase-1 activity as well as the release of IL-1ß, bFGF and HMGB1; including after TRX overexpression or treatment with recombinant TRX. Rats were intratracheally instilled with vehicle control, Dörentruper quartz (DQ12) or DQ12 coated with polyvinylpyridine N-oxide. At days 3, 7, 28, 90, 180 and 360 five animals per treatment group were sacrificed. Hallmarks of silicosis were assessed with Haematoxylin-eosin and Sirius Red stainings. Caspase-1 activity in the bronchoalveolar lavage and caspase-1 and IL-1ß localization in lung tissue were determined using Western blot and immunohistochemistry (IHC). RESULTS: Silica polymorphs triggered secretion of IL-1ß, bFGF and HMGB1 in a surface reactivity dependent manner. Inflammasome readouts linked with caspase-1 enzymatic activity were attenuated by TRX overexpression or treatment. At day 3 and 7 increased caspase-1 activity was detected in BALF of the DQ12 group and increased levels of caspase-1 and IL-1ß were observed with IHC in the DQ12 group compared to controls. DQ12 exposure revealed silicotic nodules at 180 and 360 days. Particle surface modification markedly attenuated the grade of inflammation and lymphocyte influx and attenuated the level of inflammasome activation, indicating that the development of silicosis and inflammasome activation is determined by crystalline silica surface reactivity. CONCLUSION: Our novel data indicate the pivotal role of surface reactivity of crystalline silica to activate the inflammasome in cultures of both epithelial cells and macrophages. Inhibitory capacity of the antioxidant TRX to inflammasome activation was evidenced. DQ12 quartz exposure induced acute and chronic functional activation of the inflammasome in the heterogeneous cell populations of the lung in associated with its crystalline surface reactivity.


Subject(s)
Air Pollutants/toxicity , Carrier Proteins/agonists , Inflammasomes/drug effects , Lung/drug effects , Respiratory Mucosa/drug effects , Silicon Dioxide/toxicity , Air Pollutants/chemistry , Animals , Biomarkers/metabolism , Bronchi/drug effects , Bronchi/immunology , Bronchi/metabolism , Bronchi/pathology , Carrier Proteins/metabolism , Cell Line , Cell Survival/drug effects , Female , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Inhalation Exposure/adverse effects , Lung/immunology , Lung/metabolism , Lung/pathology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , NLR Family, Pyrin Domain-Containing 3 Protein , Particle Size , Rats , Rats, Wistar , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Silicon Dioxide/administration & dosage , Silicon Dioxide/chemistry , Silicosis/immunology , Silicosis/metabolism , Silicosis/pathology , Surface Properties , Toxicity Tests, Acute , Toxicity Tests, Chronic
18.
Am J Respir Cell Mol Biol ; 51(5): 595-603, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24940987

ABSTRACT

Malignant mesothelioma (MM), lung cancers, and asbestosis are hyperproliferative diseases associated with exposures to asbestos. All have a poor prognosis; thus, the need to develop novel and effective therapies is urgent. Vandetanib (Van) (ZD6474, ZACTIMA) is a tyrosine kinase inhibitor that has shown equivocal results in clinical trials for advanced non-small cell lung cancer. However, tyrosine kinase inhibitors alone have shown no significant clinical activity in phase II trials of patients with unresectable MM. Using epithelioid (HMESO) and sarcomatoid (H2373) human MM lines, the efficacy of tumor cell killing and signaling pathways modulated by Van with and without doxorubicin (Dox) was examined. Van alone reduced total cell numbers in HMESO MM and synergistically increased the toxicity of Dox in HMESO and H2373 cells. Most importantly, we identified two novel cell survival/resistance pathways, ERK5 and cyclic AMP response element binding protein (CREB), that were inhibited by Van and Dox. After silencing of either ERK5 or CREB, significant decreases in cell numbers in the Dox-resistant sarcomatoid H2373 line were observed. Results suggest that a plethora of cell signaling pathways associated with cell survival are induced by Dox but inhibited by the addition of Van in MM. Data from our study support the combined efficacy of Van and Dox as a novel approach in the treatment of MM that is further enhanced by blocking ERK5 or CREB signaling cascades.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Doxorubicin/pharmacology , MAP Kinase Signaling System/physiology , Mesothelioma/drug therapy , Mitogen-Activated Protein Kinase 7/metabolism , Piperidines/pharmacology , Quinazolines/pharmacology , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cyclic AMP Response Element-Binding Protein/genetics , Doxorubicin/toxicity , Drug Synergism , Humans , MAP Kinase Signaling System/drug effects , Mesothelioma/metabolism , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Mitogen-Activated Protein Kinase 7/genetics , Neoplasms, Connective Tissue/drug therapy , Neoplasms, Connective Tissue/metabolism , Phosphorylation/drug effects , Phosphorylation/physiology , Piperidines/toxicity , Quinazolines/toxicity , RNA, Small Interfering/genetics , Sarcoma/drug therapy , Sarcoma/metabolism
19.
BMC Cancer ; 13: 400, 2013 Sep 11.
Article in English | MEDLINE | ID: mdl-24024776

ABSTRACT

BACKGROUND: Malignant mesotheliomas (MMs) are chemoresistant tumors related to exposure to asbestos fibers. The long latency period of MM (30-40 yrs) and heterogeneity of tumor presentation make MM difficult to diagnose and treat at early stages. Currently approved second-line treatments following surgical resection of MMs include a combination of cisplatin or carboplatin (delivered systemically) and pemetrexed, a folate inhibitor, with or without subsequent radiation. The systemic toxicities of these treatments emphasize the need for more effective, localized treatment regimens. METHODS: Acid-prepared mesoporous silica (APMS) microparticles were loaded with doxorubicin (DOX) and modified externally with a mesothelin (MB) specific antibody before repeated intraperitoneal (IP) injections into a mouse xenograft model of human peritoneal MM. The health/weight of mice, tumor volume/weight, tumor necrosis and cell proliferation were evaluated in tumor-bearing mice receiving saline, DOX high (0.2 mg/kg), DOX low (0.05 mg/kg), APMS-MB, or APMS-MB-DOX (0.05 mg/kg) in saline. RESULTS: Targeted therapy (APMS-MB-DOX at 0.05 mg/kg) was more effective than DOX low (0.05 mg/kg) and less toxic than treatment with DOX high (0.2 mg/kg). It also resulted in the reduction of tumor volume without loss of animal health and weight, and significantly decreased tumor cell proliferation. High pressure liquid chromatography (HPLC) of tumor tissue confirmed that APMS-MB-DOX particles delivered DOX to target tissue. CONCLUSIONS: Data suggest that targeted therapy results in greater chemotherapeutic efficacy with fewer adverse side effects than administration of DOX alone. Targeted microparticles are an attractive option for localized drug delivery.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Doxorubicin/administration & dosage , Drug Delivery Systems , GPI-Linked Proteins/antagonists & inhibitors , Mesothelioma/metabolism , Mesothelioma/pathology , Microspheres , Animals , Body Weight , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , GPI-Linked Proteins/metabolism , Humans , Inflammation/metabolism , Inflammation/pathology , Injections, Intraperitoneal , Ki-67 Antigen/metabolism , Macrophages/pathology , Mesothelin , Mesothelioma/drug therapy , Mice , Necrosis/drug therapy , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
20.
Part Fibre Toxicol ; 10: 39, 2013 Aug 13.
Article in English | MEDLINE | ID: mdl-23937860

ABSTRACT

BACKGROUND: Pleural fibrosis and malignant mesotheliomas (MM) occur after exposures to pathogenic fibers, yet the mechanisms initiating these diseases are unclear. RESULTS: We document priming and activation of the NLRP3 inflammasome in human mesothelial cells by asbestos and erionite that is causally related to release of IL-1ß, IL-6, IL-8, and Vascular Endothelial Growth Factor (VEGF). Transcription and release of these proteins are inhibited in vitro using Anakinra, an IL-1 receptor antagonist that reduces these cytokines in a human peritoneal MM mouse xenograft model. CONCLUSIONS: These novel data show that asbestos-induced priming and activation of the NLRP3 inflammasome triggers an autocrine feedback loop modulated via the IL-1 receptor in mesothelial cell type targeted in pleural infection, fibrosis, and carcinogenesis.


Subject(s)
Asbestos, Crocidolite/toxicity , Autocrine Communication , Carrier Proteins/metabolism , Cytokines/metabolism , Epithelium/drug effects , Inflammasomes/drug effects , Inflammation Mediators/metabolism , Mesothelioma/chemically induced , Zeolites/toxicity , Animals , Cell Line, Tumor , Cytokines/genetics , Dose-Response Relationship, Drug , Epithelium/immunology , Epithelium/pathology , Humans , Inflammasomes/immunology , Interleukin 1 Receptor Antagonist Protein/pharmacology , Mesothelioma/drug therapy , Mesothelioma/genetics , Mesothelioma/immunology , Mesothelioma/pathology , Mice , Mice, SCID , NLR Family, Pyrin Domain-Containing 3 Protein , Primary Cell Culture , RNA, Messenger/metabolism , Receptors, Interleukin-1/antagonists & inhibitors , Receptors, Interleukin-1/metabolism , Time Factors , Transcription, Genetic/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...