Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(19)2023 Sep 25.
Article in English | MEDLINE | ID: mdl-37833956

ABSTRACT

Interstitial lung diseases (ILDs) are lethal lung diseases characterized by pulmonary inflammation and progressive lung interstitial scarring. We previously developed a mouse model of ILD using vanadium pentoxide (V2O5) and identified several gene candidates on chromosome 4 associated with pulmonary fibrosis. While these data indicated a significant genetic contribution to ILD susceptibility, they did not include any potential associations and interactions with the mitochondrial genome that might influence disease risk. To conduct this pilot work, we selected the two divergent strains we previously categorized as V2O5-resistant C57BL6J (B6) and -responsive DBA/2J (D2) and compared their mitochondrial genome characteristics, including DNA variants, heteroplasmy, lesions, and copy numbers at 14- and 112-days post-exposure. While we did not find changes in the mitochondrial genome at 14 days post-exposure, at 112 days, we found that the responsive D2 strain exhibited significantly fewer mtDNA copies and more lesions than control animals. Alongside these findings, mtDNA heteroplasmy frequency decreased. These data suggest that mice previously shown to exhibit increased susceptibility to pulmonary fibrosis and inflammation sustain damage to the mitochondrial genome that is evident at 112 days post-V2O5 exposure.


Subject(s)
DNA, Mitochondrial , Pulmonary Fibrosis , Mice , Animals , DNA, Mitochondrial/genetics , DNA Copy Number Variations , Heteroplasmy , Mice, Inbred DBA
2.
Cardiovasc Toxicol ; 19(2): 168-177, 2019 04.
Article in English | MEDLINE | ID: mdl-30382549

ABSTRACT

The broad list of commercial applications for multi-walled carbon nanotubes (MWCNT) can be further expanded with the addition of various surface chemistry modifications. For example, standard commercial grade MWCNT (C-grade) can be carboxylated (COOH) or nitrogen-doped (N-doped) to suite specific utilities. We previously reported dose-dependent expansions of cardiac ischemia/reperfusion (I/R) injury, 24 h after intratracheal instillation of C-grade, COOH, or N-doped MWCNT in mice. Here, we have tested the hypothesis that airway exposure to MWCNT perturbs cardiovascular adenosinergic signaling, which could contribute to exacerbation of cardiac I/R injury. 100 µL of Vehicle or identical suspension volumes containing 100 µg of C-grade, COOH, or N-doped MWCNT were instilled into the trachea of CD-1 ICR mice. 1 day later, we measured cyclic adenosine monophosphate (cAMP) concentrations in cardiac tissue and evaluated arterial adenosinergic smooth muscle signaling mechanisms related to nitric oxide synthase (NOS) and cyclooxygenase (COX) in isolated aortic tissue. We also verified cardiac I/R injury expansion and examined both lung histology and bronchoalveolar lavage fluid cellularity in MWCNT exposed mice. Myocardial cAMP concentrations were reduced (p < 0.05) in the C-grade group by 17.4% and N-doped group by 13.7% compared to the Vehicle group. Curve fits to aortic ring 2-Cl-Adenosine concentration responses were significantly greater in the MWCNT groups vs. the Vehicle group. Aortic constrictor responses were more pronounced with NOS inhibition and were abolished with COX inhibition. These findings indicate that addition of functional chemical moieties on the surface of MWCNT may alter the biological responses to exposure by influencing cardiovascular adenosinergic signaling and promoting cardiac injury.


Subject(s)
Adenosine/pharmacology , Muscle, Smooth, Vascular/drug effects , Myocardial Infarction/chemically induced , Myocardial Reperfusion Injury/chemically induced , Myocardium/metabolism , Nanotubes, Carbon/toxicity , Signal Transduction/drug effects , Adenosine/analogs & derivatives , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Cyclic AMP/metabolism , Inhalation Exposure , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Mice, Inbred ICR , Muscle, Smooth, Vascular/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/pathology , Nitric Oxide Synthase/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Vasoconstriction/drug effects , Vasodilation/drug effects
3.
FASEB J ; 30(2): 775-84, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26506979

ABSTRACT

Obesity has more than doubled in children and tripled in adolescents in the past 30 yr. The association between metabolic disorders in offspring of obese mothers with diabetes has long been known; however, a growing body of research indicates that fathers play a significant role through presently unknown mechanisms. Recent observations have shown that changes in paternal diet may result in transgenerational inheritance of the insulin-resistant phenotype. Although diet-induced epigenetic reprogramming via paternal lineage has recently received much attention in the literature, the effect of paternal physical activity on offspring metabolism has not been adequately addressed. In the current study, we investigated the effects of long-term voluntary wheel-running in C57BL/6J male mice on their offspring's predisposition to insulin resistance. Our observations revealed that fathers subjected to wheel-running for 12 wk produced offspring that were more susceptible to the adverse effects of a high-fat diet, manifested in increased body weight and adiposity, impaired glucose tolerance, and elevated insulin levels. Long-term paternal exercise also altered expression of several metabolic genes, including Ogt, Oga, Pdk4, H19, Glut4, and Ptpn1, in offspring skeletal muscle. Finally, prolonged exercise affected gene methylation patterns and micro-RNA content in the sperm of fathers, providing a potential mechanism for the transgenerational inheritance. These findings suggest that paternal exercise produces offspring with a thrifty phenotype, potentially via miRNA-induced modification of sperm.


Subject(s)
Adiposity , Energy Metabolism , Epigenesis, Genetic , Insulin Resistance , Obesity/metabolism , Physical Conditioning, Animal , Animals , Male , Mice , Muscle Proteins/biosynthesis , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Obesity/genetics , Obesity/pathology
4.
FASEB J ; 28(3): 1098-112, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24285090

ABSTRACT

Interstitial lung diseases (ILDs) are characterized by injury, inflammation, and scarring of alveoli, leading to impaired function. The etiology of idiopathic forms of ILD is not understood, making them particularly difficult to study due to the lack of appropriate animal models. Consequently, few effective therapies have emerged. We developed an inbred mouse model of ILD using vanadium pentoxide (V2O5), the most common form of a transition metal found in cigarette smoke, fuel ash, mineral ores, and steel alloys. Pulmonary responses to V2O5, including dose-dependent increases in lung permeability, inflammation, collagen content, and dysfunction, were significantly greater in DBA/2J mice compared to C57BL/6J mice. Inflammatory and fibrotic responses persisted for 4 mo in DBA/2J mice, while limited responses in C57BL/6J mice resolved. We investigated the genetic basis for differential responses through genetic mapping of V2O5-induced lung collagen content in BXD recombinant inbred (RI) strains and identified significant linkage on chromosome 4 with candidate genes that associate with V2O5-induced collagen content across the RI strains. Results suggest that V2O5 may induce pulmonary fibrosis through mechanisms distinct from those in other models of pulmonary fibrosis. These findings should further advance our understanding of mechanisms involved in ILD and thereby aid in identification of new therapeutic targets.


Subject(s)
Genetic Predisposition to Disease , Pulmonary Fibrosis/genetics , Vanadium Compounds/toxicity , Animals , Bronchoalveolar Lavage Fluid , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Pulmonary Fibrosis/chemically induced , Real-Time Polymerase Chain Reaction , Transforming Growth Factor beta/metabolism
5.
N C Med J ; 74(6): 534-6, 2013.
Article in English | MEDLINE | ID: mdl-24316785

ABSTRACT

Epigenetic modifications are gene regulatory mechanisms that allow rapid adaptation to the environment. These mitotically stable and meiotically heritable changes are sensitive to environmental conditions especially during developmental periods, and they are essential to understanding how information in the DNA sequence is utilized. Recent research in this area has led to excitement and questions about medical applications of epigenetics.


Subject(s)
Clinical Medicine , Epigenomics , Humans
6.
Part Fibre Toxicol ; 8: 24, 2011 Aug 18.
Article in English | MEDLINE | ID: mdl-21851604

ABSTRACT

BACKGROUND: Multi-walled carbon nanotubes (MWCNTs) are widely used in many disciplines due to their unique physical and chemical properties. Therefore, some concerns about the possible human health and environmental impacts of manufactured MWCNTs are rising. We hypothesized that instillation of MWCNTs impairs pulmonary function in C57BL/6 mice due to development of lung inflammation and fibrosis. METHODS: MWCNTs were administered to C57BL/6 mice by oropharyngeal aspiration (1, 2, and 4 mg/kg) and we assessed lung inflammation and fibrosis by inflammatory cell infiltration, collagen content, and histological assessment. Pulmonary function was assessed using a FlexiVent system and levels of Ccl3, Ccl11, Mmp13 and IL-33 were measured by RT-PCR and ELISA. RESULTS: Mice administered MWCNTs exhibited increased inflammatory cell infiltration, collagen deposition and granuloma formation in lung tissue, which correlated with impaired pulmonary function as assessed by increased resistance, tissue damping, and decreased lung compliance. Pulmonary exposure to MWCNTs induced an inflammatory signature marked by cytokine (IL-33), chemokine (Ccl3 and Ccl11), and protease production (Mmp13) that promoted the inflammatory and fibrotic changes observed within the lung. CONCLUSIONS: These results further highlight the potential adverse health effects that may occur following MWCNT exposure and therefore we suggest these materials may pose a significant risk leading to impaired lung function following environmental and occupational exposures.


Subject(s)
Inhalation Exposure/adverse effects , Lung/drug effects , Nanotubes, Carbon/toxicity , Pneumonia/chemically induced , Pulmonary Fibrosis/chemically induced , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Collagen/metabolism , Cytokines/immunology , Dose-Response Relationship, Drug , Instillation, Drug , Lung/immunology , Lung/pathology , Mice , Mice, Inbred C57BL , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Nanotubes, Carbon/chemistry , Particle Size , Pneumonia/immunology , Pneumonia/pathology , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/pathology , Respiratory Function Tests , Surface Properties
7.
Am J Physiol Lung Cell Mol Physiol ; 301(1): L31-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21531775

ABSTRACT

Exposure to environmental pollutants has been linked to various airway diseases and disease exacerbations. Almost all chronic airway diseases such as chronic obstructive pulmonary disease and asthma are caused by complicated interactions between gene and environment. One of the major hallmarks of those diseases is airway mucus overproduction (MO). Excessive mucus causes airway obstruction and significantly increases morbidity and mortality. Metals are major components of environmental particulate matters (PM). Among them, vanadium has been suggested to play an important role in PM-induced mucin production. Vanadium pentoxide (V(2)O(5)) is the most common commercial source of vanadium, and it has been associated with occupational chronic bronchitis and asthma, both of which are MO diseases. However, the underlying mechanism is not entirely clear. In this study, we used both in vitro and in vivo models to demonstrate the robust inductions of mucin production by V(2)O(5). Furthermore, the follow-up mechanistic study revealed a novel v-raf-1 murine leukemia viral oncogene homolog 1-IKK-NF-κB pathway that mediated V(2)O(5)-induced mucin production. Most interestingly, the reactive oxygen species and the classical mucin-inducing epidermal growth factor receptor (EGFR)-MAPK pathway appeared not to be involved in this process. Thus the V(2)O(5)-induced mucin production may represent a novel EGFR-MAPK-independent and environmental toxicant-associated MO model. Complete elucidation of the signaling pathway in this model will not only facilitate the development of the treatment for V(2)O(5)-associated occupational diseases but also advance our understanding on the EGFR-independent mucin production in other chronic airway diseases.


Subject(s)
Mucin 5AC/biosynthesis , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Vanadium Compounds/toxicity , Animals , Cells, Cultured , Enzyme Activation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Epithelial Cells/pathology , ErbB Receptors/metabolism , Free Radical Scavengers/pharmacology , Gene Expression Regulation/drug effects , Humans , I-kappa B Kinase/metabolism , MAP Kinase Signaling System/drug effects , Metaplasia , Mice , Mucin 5AC/genetics , NF-kappa B/metabolism , Pneumonia/metabolism , Pneumonia/pathology , Reactive Oxygen Species/metabolism , Respiratory Mucosa/enzymology , Respiratory Mucosa/pathology , raf Kinases/metabolism
8.
Nanotoxicology ; 5(4): 531-45, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21043986

ABSTRACT

Cerium oxide (CeO2) represents an important nanomaterial with wide ranging applications. However, little is known regarding how CeO2 exposure may influence pulmonary or systemic inflammation. Furthermore, how mast cells would influence inflammatory responses to a nanoparticle exposure is unknown. We thus compared pulmonary and cardiovascular responses between C57BL/6 and B6.Cg-Kit(W-sh) mast cell deficient mice following CeO2 nanoparticle instillation. C57BL/6 mice instilled with CeO2 exhibited mild pulmonary inflammation. However, B6.Cg-Kit(W-sh) mice did not display a similar degree of inflammation following CeO2 instillation. Moreover, C57BL/6 mice instilled with CeO2 exhibited altered aortic vascular responses to adenosine and an increase in myocardial ischemia/reperfusion injury which was absent in B6.Cg-Kit(W-sh) mice. In vitro CeO2 exposure resulted in increased production of PGD2, TNF-α, IL-6 and osteopontin by cultured mast cells. These findings demonstrate that CeO2 nanoparticles activate mast cells contributing to pulmonary inflammation, impairment of vascular relaxation and exacerbation of myocardial ischemia/reperfusion injury.


Subject(s)
Cerium/toxicity , Mast Cells/metabolism , Metal Nanoparticles/toxicity , Reperfusion Injury/metabolism , Adenosine/pharmacology , Analysis of Variance , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Bronchoalveolar Lavage Fluid/cytology , Chemokine CCL3 , Gene Expression Regulation/drug effects , Histocytochemistry , Interleukin-10 , Interleukin-13/genetics , Interleukin-13/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Lung/chemistry , Male , Mast Cells/pathology , Metal Nanoparticles/chemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction , Myocardium/chemistry , Osteopontin/genetics , Osteopontin/metabolism , Particle Size , Pneumonia/chemically induced , Pneumonia/metabolism , Pneumonia/pathology , Prostaglandin D2 , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
9.
Part Fibre Toxicol ; 7: 9, 2010 Apr 12.
Article in English | MEDLINE | ID: mdl-20385015

ABSTRACT

BACKGROUND: Elevated levels of air pollution are associated with increased risk of lung cancer. Particulate matter (PM) contains transition metals that may potentiate neoplastic development through the induction of oxidative stress and inflammation, a lung cancer risk factor. Vanadium pentoxide (V2O5) is a component of PM derived from fuel combustion as well as a source of occupational exposure in humans. In the current investigation we examined the influence of genetic background on susceptibility to V2O5-induced inflammation and evaluated whether V2O5 functions as a tumor promoter using a 2-stage (initiation-promotion) model of pulmonary neoplasia in mice. RESULTS: A/J, BALB/cJ (BALB), and C57BL/6J (B6) mice were treated either with the initiator 3-methylcholanthrene (MCA; 10 microg/g; i.p.) or corn oil followed by 5 weekly aspirations of V2O5 or PBS and pulmonary tumors were enumerated 20 weeks following MCA treatment. Susceptibility to V2O5-induced pulmonary inflammation was assessed in bronchoalveolar lavage fluid (BALF), and chemokines, transcription factor activity, and MAPK signaling were quantified in lung homogenates. We found that treatment of animals with MCA followed by V2O5 promoted lung tumors in both A/J (10.3 +/- 0.9 tumors/mouse) and BALB (2.2 +/- 0.36) mice significantly above that observed with MCA/PBS or V2O5 alone (P < 0.05). No tumors were observed in the B6 mice in any of the experimental groups. Mice sensitive to tumor promotion by V2O5 were also found to be more susceptible to V2O5-induced pulmonary inflammation and hyperpermeability (A/J>BALB>B6). Differential strain responses in inflammation were positively associated with elevated levels of the chemokines KC and MCP-1, higher NFkappaB and c-Fos binding activity, as well as sustained ERK1/2 activation in lung tissue. CONCLUSIONS: In this study we demonstrate that V2O5, an occupational and environmentally relevant metal oxide, functions as an in vivo lung tumor promoter among different inbred strains of mice. Further, we identified a positive relationship between tumor promotion and susceptibility to V2O5-induced pulmonary inflammation. These findings suggest that repeated exposures to V2O5 containing particles may augment lung carcinogenesis in susceptible individuals through oxidative stress mediated pathways.


Subject(s)
Air Pollutants/toxicity , Carcinogens/toxicity , Lung Injury/chemically induced , Lung Neoplasms/chemically induced , Vanadium Compounds/toxicity , Animals , Chemokines/metabolism , Disease Models, Animal , Genetic Predisposition to Disease , Inhalation Exposure , Lung Injury/genetics , Lung Injury/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Methylcholanthrene/toxicity , Mice , Mice, Inbred A/genetics , Mice, Inbred BALB C/genetics , Mice, Inbred C57BL/genetics , Species Specificity , Transcription Factors/metabolism
10.
Am J Respir Crit Care Med ; 179(2): 138-50, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-18931336

ABSTRACT

RATIONALE: Respiratory syncytial virus (RSV) is the most frequent cause of significant lower respiratory illness in infants and young children, but its pathogenesis is not fully understood. The transcription factor Nrf2 protects lungs from oxidative injury and inflammation via antioxidant response element (ARE)-mediated gene induction. OBJECTIVES: The current study was designed to determine the role of Nrf2-mediated cytoprotective mechanisms in murine airway RSV disease. METHODS: Nrf2-deficient (Nrf2(-/-)) and wild-type (Nrf2(+/+)) mice were intranasally instilled with RSV or vehicle. In a separate study, Nrf2(+/+) and Nrf2(-/-) mice were treated orally with sulforaphane (an Nrf2-ARE inducer) or phosphate-buffered saline before RSV infection. MEASUREMENTS AND MAIN RESULTS: RSV-induced bronchopulmonary inflammation, epithelial injury, and mucus cell metaplasia as well as nasal epithelial injury were significantly greater in Nrf2(-/-) mice than in Nrf2(+/+) mice. Compared with Nrf2(+/+) mice, significantly attenuated viral clearance and IFN-gamma, body weight loss, heightened protein/lipid oxidation, and AP-1/NF-kappaB activity along with suppressed antioxidant induction was found in Nrf2(-/-) mice in response to RSV. Sulforaphane pretreatment significantly limited lung RSV replication and virus-induced inflammation in Nrf2(+/+) but not in Nrf2(-/-) mice. CONCLUSIONS: The results of this study support an association of oxidant stress with RSV pathogenesis and a key role for the Nrf2-ARE pathway in host defense against RSV.


Subject(s)
NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/metabolism , Animals , Anticarcinogenic Agents/administration & dosage , Bronchoalveolar Lavage Fluid , Buffers , Disease Models, Animal , Drug Therapy, Combination , Isothiocyanates , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphates/administration & dosage , Sodium Chloride/administration & dosage , Sulfoxides , Thiocyanates/administration & dosage
11.
Curr Protoc Pharmacol ; Chapter 5: Unit 5.46, 2008 Mar.
Article in English | MEDLINE | ID: mdl-22294226

ABSTRACT

Pulmonary fibrosis is a component of many interstitial lung diseases, including idiopathic pulmonary fibrosis, a chronic, progressive disease for which there is currently no effective therapy. Bleomycin has been widely used in rodents to model pulmonary fibrosis for the study of mechanisms involved in fibrogenesis and for evaluation of potential therapies. Bleomycin induces DNA strand breaks, resulting in pulmonary inflammation, injury, and subsequent interstitial fibrosis. This unit describes methods for delivering bleomycin, either directly into the lung or systemically, to create models of pulmonary fibrosis in rodents. Also described is a rapid and easy procedure for measuring lung collagen content to quantify the severity of fibrosis.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Disease Models, Animal , Pulmonary Fibrosis/chemically induced , Animals , Antibiotics, Antineoplastic/administration & dosage , Azo Compounds , Biological Assay/methods , Bleomycin/administration & dosage , Collagen/analysis , Coloring Agents , Dose-Response Relationship, Drug , Lung/chemistry , Mice , Rats , Specimen Handling/methods
12.
Antioxid Redox Signal ; 10(2): 321-32, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17999635

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic progressive disorder in which excessive deposition of extracellular matrix leads to irreversible scarring of interstitial lung tissue. The etiology of IPF remains unknown, but growing evidence suggests that disequilibrium in oxidant/antioxidant balance contributes significantly. IPF is currently regarded as a fibroproliferative disorder triggered by repeated alveolar epithelial cell injury. Oxidative stress plays a role in many processes involved in alveolar epithelial cell injury and fibrogenesis. Here we review the role of oxidative stress in IPF, and other forms of pulmonary fibrosis, with particular attention to antioxidant defenses regulated by the redox-sensitive transcription factor nuclear factor, erythroid derived 2, like (Nrf2). Nrf2 binds specific antioxidant response elements (AREs) in the promoter of antioxidant enzyme and defense protein genes and regulates their expression in many tissue types. Nrf2 protects from several phenotypes in which enhanced oxidative burden contributes to disease pathogenesis, including cancer, acute lung injury, and pulmonary fibrosis. We suggest that promoter polymorphisms in human NRF2 may contribute to IPF susceptibility, although this hypothesis has not been tested. Pulmonary fibrosis is a highly complex disease and involves multiple genes and processes, and new therapies for cellular and molecular targets involved in pathogenic mechanisms are needed.


Subject(s)
Antioxidants/pharmacology , NF-E2-Related Factor 2/physiology , Oxidative Stress , Pulmonary Fibrosis/physiopathology , Humans , Lung/pathology , Lung/physiopathology , Models, Biological , Oxidants/toxicity , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/prevention & control , Reactive Oxygen Species/metabolism
13.
Am J Pathol ; 167(5): 1221-9, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16251407

ABSTRACT

The signal transducer and activator of transcription (Stat)-1 mediates growth arrest and apoptosis. We postulated that lung fibrosis characterized by excessive proliferation of lung fibroblasts would be enhanced in Stat1-deficient (Stat1-/-) mice. Two weeks after bleomycin aspiration (3 U/kg), Stat1-/- mice exhibited a more severe fibroproliferative response and significantly elevated total lung collagen compared to wild-type mice. Growth factors [epidermal growth factor (EGF) or platelet-derived growth factor (PDGF)] enhanced [3H]thymidine uptake in lung fibroblasts isolated from Stat1-/- mice compared to wild-type mice. Interferon (IFN)-gamma, which signals growth arrest via Stat1, inhibited EGF- or PDGF-stimulated mitogenesis in wild-type fibroblasts but enhanced [3H]thymidine uptake in Stat1-/- fibroblasts. Moreover, IFN-gamma treatment in the absence of growth factors induced a concentration-dependent increase in [3H]thymidine uptake in Stat1-/- but not wild-type fibroblasts. Mitogen-activated protein kinase (ERK-1/2) phosphorylation in response to PDGF or EGF did not differ among Stat1-/- and wild-type fibroblasts. However, Stat3 phosphorylation induced by PDGF, EGF, or IFN-gamma increased twofold in Stat1-/- fibroblasts compared to wild-type fibroblasts. Our findings indicate that Stat1-/- mice are more susceptible to bleomycin-induced lung fibrosis than wild-type mice due to 1) enhanced fibroblast proliferation in response to growth factors (EGF and PDGF), 2) stimulation of fibroblast growth by a Stat1-independent IFN-gamma signaling pathway, and 3) increased activation of Stat3.


Subject(s)
Lung/pathology , Pulmonary Fibrosis/pathology , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/physiology , Animals , Bleomycin/toxicity , Blotting, Western , Cell Proliferation , Cells, Cultured , Collagen/analysis , Disease Models, Animal , Epidermal Growth Factor/pharmacology , Fibroblasts/pathology , Growth Inhibitors/pharmacology , Hydroxyproline/analysis , Interferon-gamma/pharmacology , Lung/drug effects , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Platelet-Derived Growth Factor/pharmacology , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/physiopathology , STAT3 Transcription Factor/metabolism , Thymidine/metabolism
14.
Free Radic Biol Med ; 35(8): 845-55, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-14556849

ABSTRACT

Vanadium compounds present in air pollution particulate matter activate signal transduction pathways in pulmonary cell types leading to pathological outcomes including aberrant cell proliferation, apoptosis, and cytokine expression. Vanadium has been proposed to activate transcription factors via the generation of hydrogen peroxide (H2O2). We investigated the mechanisms through which vanadium pentoxide (V2O5), the major form of vanadium released from the industrial burning of fuel oil, activated the signal transducer and activator of transcription (STAT)-1. V2O5-induced STAT-1 activation was blocked by catalase and N-acetyl-L-cysteine (NAC), suggesting vanadium-induced generation of H2O2. Surprisingly, however, V2O5 did not increase H2O2 levels released by rat lung myofibroblasts into cell culture supernatants. Instead, these quiescent myofibroblasts spontaneously released micromolar concentrations of H2O2, and the addition of V2O5 reduced H2O2 levels in cell culture supernatants within minutes. V2O5 suppressed H2O2 for as long as 24 h. Differences in the temporal activation of STAT-1 and p38 MAPK were observed following V2O5 or H2O2 treatment, and STAT-1 activation by V2O5 or H2O2 was attenuated by an inhibitor of the EGF receptor tyrosine kinase (AG1478) or p38 MAPK (SB203580). The phosphorylation of p38 MAPK by V2O5 was inhibited by NAC and catalase, yet the EGF receptor inhibitor AG1478 had no effect on V2O5-induced p38 MAPK activation. Collectively, our findings support the novel hypothesis that H2O2 spontaneously generated by myofibroblasts fuels vanadium-induced activation of STAT-1. Moreover, p38 MAPK and EGF receptor activation are required for V2O5-induced STAT-1 activation.


Subject(s)
DNA-Binding Proteins/metabolism , Hydrogen Peroxide/metabolism , Lung/drug effects , Mitogen-Activated Protein Kinases/metabolism , Trans-Activators/metabolism , Vanadium Compounds/pharmacology , Acetylcysteine/pharmacology , Animals , Carbon/adverse effects , Catalase/pharmacology , Enzyme Inhibitors/pharmacology , ErbB Receptors/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Lung/cytology , Lung/metabolism , Phosphorylation/drug effects , Rats , STAT1 Transcription Factor , p38 Mitogen-Activated Protein Kinases
15.
Am J Respir Cell Mol Biol ; 27(4): 413-8, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12356574

ABSTRACT

Epidemiologic studies have suggested that exposure to airborne particulate matter (PM) can exacerbate allergic airway responses; however, the mechanism(s) are not well understood. We and others have recently shown that development of airway hyperresponsiveness (AHR) may be a complement-mediated process. In the present study, we examined the role of complement factor 3 (C3) in the development of PM-induced AHR and airway inflammation by comparing responses between C3-deficient (C3(-/-)) and wild-type mice. Mice were exposed to 0.5 mg of ambient particulate collected in urban Baltimore. Forty-eight hours later, airway responsiveness to intravenous acetylcholine was assessed and bronchoalveolar lavage was conducted. PM exposure of wild-type mice resulted in significant increases in AHR, whereas it did not significantly increase airway reactivity in C3(-/-) mice. Interestingly, PM induced similar inflammatory responses in both wild-type and C3(-/-) mice. Immunohistochemical staining demonstrated marked C3 deposition in the airway epithelium and connective tissue of wild-type mice after PM exposure. These results suggest that exposure to PM may induce AHR through activation of complement factor 3 in the airways.


Subject(s)
Air Pollution , Bronchi/immunology , Bronchi/metabolism , Complement C3/metabolism , Complement C3/physiology , Air Pollution/analysis , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Eosinophils/pathology , Epithelial Cells/pathology , Immunohistochemistry , Inflammation , Lung/pathology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Neutrophils/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...