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

Country/Region as subject
Publication year range
1.
Acta Oncol ; 58(1): 38-44, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30375909

ABSTRACT

BACKGROUND: Malignant pleural mesothelioma (MPM) is a rare occupational cancer with a poor prognosis. Even with a multimodality treatment approach, the treatment outcomes remain unsatisfactory. The use of asbestos has been banned in most developed countries, but MPM continues to be a significant occupational disease also in these countries. Aim of this study is to identify modern epidemiology and assess equality in care. METHODS: Our study cohort consists of 1010 patients diagnosed with MPM in Finland during 2000-2012. The data were collected from the Finnish Cancer Registry, the National Workers' Compensation Center Registry and the National Registry of Causes of Death, Statistics Finland. RESULTS: Women were diagnosed a mean of 4.5 years later than males (p = .001), but survival did not differ (overall median survival 9.7 months). A workers' compensation claim was more common in males (OR 11.0 [95% CI 7.5-16.2]) and in regions with a major asbestos industry (OR 1.7 [95% CI 1.3-2.2]). One-year and three-year survivals did not differ regionally. Patients without chemotherapy treatment had an inferior survival (RR 1.8 [95% CI 1.5-2.0]). The initial survival benefit gained with pemetrexed was diluted at 51 months. CONCLUSIONS: MPM is a disease with a poor prognosis, although chemotherapy appears to improve survival time. Significant gender and regional variation exists among patients, with notable differences in diagnostic and treatment practices. Long-term outcomes with pemetrexed remain indeterminate. IMPACT: Emphasize centralized consult services for the diagnosis, treatment and support that patients receive for MPM, facilitating equal outcomes and compensation.


Subject(s)
Lung Neoplasms/epidemiology , Mesothelioma/epidemiology , Pleural Neoplasms/epidemiology , Adult , Aged , Aged, 80 and over , Female , Finland/epidemiology , Humans , Incidence , Male , Mesothelioma, Malignant , Middle Aged , Registries , Sex Distribution
2.
Exp Cell Res ; 332(1): 102-15, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25557874

ABSTRACT

Activin-A and activin-B, members of the TGF-ß superfamily, are regulators of reproductive functions, inflammation and wound healing. These dimeric molecules regulate various cellular activities such as proliferation, migration and suvival. Malignant mesothelioma is an asbestos exposure related tumor affecting mainly pleura and it usually has a dismal prognosis. Here, we demonstrate that both activin-A and -B are abundantly expressed in mesothelioma tumor tissue as well as in cultured primary and established mesothelioma cells. Migratory and invasive mesothelioma cells were also found to have attenuated activation of the Smad2/3 pathway in response to activins. Migration and invasive growth of the cells in three-dimentional matrix was prevented by inhibition of activin activity using a soluble activin receptor 2B (sActR2B-Fc). This was associated with decreased ERK activity. Furthermore, migration and invasive growth was significantly inhibited by blocking ERK phosphorylation. Mesothelioma tumors are locally invasive and our results clearly suggest that acivins have a tumor-promoting function in mesothelioma through increasing expression and switching from canonical Smad3 pathway to non-canonical ERK pathway signaling. Blocking activin activity offers a new therapeutic approach for inhibition of mesothelioma invasive growth.


Subject(s)
Activins/metabolism , Cell Movement , Extracellular Signal-Regulated MAP Kinases/metabolism , Lung Neoplasms/metabolism , Mesothelioma/metabolism , Smad3 Protein/metabolism , Activin Receptors, Type I/metabolism , Activin Receptors, Type II/metabolism , Activins/genetics , Aged , Enzyme Activation , Female , Gene Expression , Humans , Lung Neoplasms/pathology , MAP Kinase Signaling System , Male , Mesothelioma/pathology , Mesothelioma, Malignant , Middle Aged , Neoplasm Invasiveness
3.
Proc Natl Acad Sci U S A ; 110(8): 2852-7, 2013 Feb 19.
Article in English | MEDLINE | ID: mdl-23382201

ABSTRACT

Elastic fiber assembly requires deposition of elastin monomers onto microfibrils, the mechanism of which is incompletely understood. Here we show that latent TGF-ß binding protein 4 (LTBP-4) potentiates formation of elastic fibers through interacting with fibulin-5, a tropoelastin-binding protein necessary for elastogenesis. Decreased expression of LTBP-4 in human dermal fibroblast cells by siRNA treatment abolished the linear deposition of fibulin-5 and tropoelastin on microfibrils. It is notable that the addition of recombinant LTBP-4 to cell culture medium promoted elastin deposition on microfibrils without changing the expression of elastic fiber components. This elastogenic property of LTBP-4 is independent of bound TGF-ß because TGF-ß-free recombinant LTBP-4 was as potent an elastogenic inducer as TGF-ß-bound recombinant LTBP-4. Without LTBP-4, fibulin-5 and tropoelastin deposition was discontinuous and punctate in vitro and in vivo. These data suggest a unique function for LTBP-4 during elastic fibrogenesis, making it a potential therapeutic target for elastic fiber regeneration.


Subject(s)
Extracellular Matrix Proteins/metabolism , Latent TGF-beta Binding Proteins/physiology , Recombinant Proteins/metabolism , Animals , HEK293 Cells , Humans , Latent TGF-beta Binding Proteins/metabolism , Mice , Mice, Knockout , Protein Binding , RNA Interference
4.
Graefes Arch Clin Exp Ophthalmol ; 252(6): 881-8, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24218041

ABSTRACT

PURPOSE: To explore factors related to pathogenesis of rhegmatogenous retinal detachment (RRD) and development of proliferative vitreoretinopathy (PVR), vitreous levels of angiopoietin-1 and -2 (Ang-1 and -2), previously undefined in RRD, transforming growth factor-(TGF) ß1, vascular endothelial growth factor (VEGF), erythropoietin (EPO) and proteolytic mediators of extracellular matrix remodelling (MMP-2 and -9) were compared in eyes with RRD and eyes with idiopathic macular hole or pucker. METHODS: Vitreous samples were collected from 117 eyes with RRD (study group) and 40 eyes with macular hole or pucker (control group). Growth factors were measured by ELISA and matrix metalloproteinases (MMPs) by gelatin zymography. RESULTS: The mean vitreous concentrations of Ang-2, MMP-2, and MMP-9 were higher (all p < 0.01), whereas concentration of VEGF was lower (p = 0.01) in eyes with RRD relative to controls. Logistic regression analysis identified Ang-2 concentration as a novel marker of RRD (p = 0.0001, OR 48.7). Ang-1, EPO, and total TGF-ß1 levels were not significantly different between the groups. However, TGF-ß1 and MMP-2 were increased in eyes with total RRD compared to those with local RRD (p ≤ 0.05). In eyes with PVR, no differences were observed in any studied marker as compared with non-PVR eyes. CONCLUSIONS: Current results reveal Ang-2 as a key factor upregulated in RRD. It may co-operate with fibrosis-associated factors and contribute to vascular complications such as breakdown of blood-eye barrier and PVR development.


Subject(s)
Angiopoietin-2/metabolism , Biomarkers/metabolism , Retinal Detachment/metabolism , Vitreous Body/metabolism , Aged , Angiopoietin-1/metabolism , Enzyme-Linked Immunosorbent Assay , Erythropoietin/metabolism , Female , Humans , Male , Matrix Metalloproteinases/metabolism , Middle Aged , Prospective Studies , Retinal Detachment/diagnosis , Transforming Growth Factor beta1/metabolism , Vascular Endothelial Growth Factor A/metabolism
5.
BMC Pulm Med ; 14: 170, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25361680

ABSTRACT

BACKGROUND: Activins are members of the TGF-ß superfamily of growth factors. First, we identified by expression array screening that activin-B and follistatin are upregulated in human idiopathic pulmonary fibrosis (IPF). Next, we wanted to clarify their specific role in lung fibrosis formation. METHODS: We used specific antibodies for activin-A and -B subunits and follistatin to measure and localize their levels in idiopathic pulmonary fibrosis and control lung biopsies. To inhibit activin signaling, we used soluble activin type IIB receptor fused to the Fc portion of human IgG1 (sActRIIB-Fc) in two different mouse models of pulmonary fibrosis. RESULTS: Activin-B and follistatin mRNA levels were elevated in the human IPF lung. Immunoreactivity to activin-A, -B and follistatin localized predominantly to the hyperplastic, activated alveolar epithelium, but was also seen in inflammatory cells. Mice treated with sActRIIB-Fc showed increased skeletal muscle mass and a clear reduction in alveolar cell counts in bronchoalveolar lavage fluid, but no significant antifibrotic effect in the lung was observed. CONCLUSIONS: The upregulation of activin-B and follistatin in IPF is a novel finding. Our results indicate that activin inhibition is not an efficient tool for antifibrotic therapy, but could be useful in reducing alveolar cellular response to injury. Activin-B and follistatin levels may be useful as biomarkers of IPF.


Subject(s)
Activins/metabolism , Follistatin/metabolism , Inhibin-beta Subunits/genetics , Pulmonary Fibrosis/metabolism , RNA, Messenger/metabolism , Activins/drug effects , Activins/genetics , Animals , Bronchoalveolar Lavage Fluid/cytology , Cell Count , Disease Models, Animal , Follistatin/genetics , Humans , Immunity, Cellular/drug effects , Male , Mice , Mice, Inbred C57BL , Protein Biosynthesis , Pulmonary Alveoli/chemistry , Pulmonary Alveoli/immunology , Quadriceps Muscle/anatomy & histology , Quadriceps Muscle/drug effects , Recombinant Fusion Proteins/pharmacology , Respiratory Mucosa/chemistry , Respiratory Mucosa/immunology , Signal Transduction , Up-Regulation/drug effects
6.
Am J Respir Cell Mol Biol ; 48(4): 448-55, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23258233

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease with a poor prognosis and very few therapeutic options. On the molecular level, patients with IPF have increased amounts of the bone morphogenetic protein (BMP) inhibitor gremlin in their lungs, which results in decreased BMP signaling, and an increase in transforming growth factor-ß signaling. Based on these findings, we hypothesized that restoration of the impaired BMP signaling would offer a novel strategy for the prevention of fibrosis progression or for the treatment of pulmonary fibrosis. We used reporter cell lines and high-throughput screening of a chemical compound library as an approach to finding molecules that increase BMP signaling in lung epithelial cells, without increasing transforming growth factor-ß signaling. The most promising candidate drug was analyzed further by studying its effects on BMP target gene expression, Smad protein phosphorylation, and a mouse model of silica-induced pulmonary fibrosis. The most promising drug candidate, tilorone, induced BMP signaling in the reporter cells and increased the expression of BMP-7 and a BMP target gene, Id3, in lung epithelial A549 cells. In a mouse model of pulmonary fibrosis, tilorone decreased lung hydroxyproline content and the expression of collagen genes Col1A1 and Col3A1. Mice treated with tilorone showed markedly decreased histological changes, compared with untreated mice. These findings indicate that tilorone has biologically significant antifibrotic properties.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Morphogenetic Protein 7/biosynthesis , Idiopathic Pulmonary Fibrosis/drug therapy , Tilorone/pharmacology , Animals , Cell Line, Tumor , Collagen Type I/biosynthesis , Collagen Type I, alpha 1 Chain , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Inhibitor of Differentiation Proteins/metabolism , Mice , Neoplasm Proteins/metabolism , Signal Transduction/drug effects
7.
J Cell Physiol ; 227(6): 2605-12, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21882188

ABSTRACT

Transforming growth factor-ß (TGF-ß) is a diverse cytokine regulating growth, apoptosis, differentiation, adhesion, invasion, and extracellular matrix production. Dysregulation of TGF-ß is associated with fibrotic disorders and epithelial-mesenchymal transition, and has been linked with idiopathic pulmonary fibrosis (IPF). Cysteine-rich protein 1 (CRP1) is a small LIM-domain containing protein involved in smooth muscle differentiation. Here, we show that TGF-ß1 increases the expression of CRP1 protein and that CRP1 levels increase in a biphasic fashion. A rapid transient (15-45 min) increase in CRP1 is followed by a subsequent, sustained increase in CRP1 a few hours afterwards that lasts several days. We find that TGF-ß1 regulates the expression of CRP1 through Smad and non-conventional p38 MAPK signaling pathways in a transcription-independent manner and that the induction occurs concomitant with an increase in myofibroblast differentiation. Using CRP1 silencing by shRNA, we identify CRP1 as a novel factor mediating cell contractility. Furthermore, we localize CRP1 to fibroblastic foci in IPF lungs and find that CRP1 is significantly more expressed in IPF as compared to control lung tissue. The results show that CRP1 is a novel TGF-ß1 regulated protein that is expressed in fibrotic lesions and may be relevant in the IPF disease.


Subject(s)
Carrier Proteins/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , LIM Domain Proteins/metabolism , Lung/metabolism , Myofibroblasts/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Carrier Proteins/genetics , Case-Control Studies , Cell Differentiation , Cell Line, Tumor , Cell Shape , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , LIM Domain Proteins/genetics , Lung/pathology , Mice , Myofibroblasts/pathology , NIH 3T3 Cells , RNA Interference , Signal Transduction , Smad2 Protein/genetics , Smad2 Protein/metabolism , Time Factors , Transfection , Up-Regulation , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Circulation ; 124(10): 1138-50, 2011 Sep 06.
Article in English | MEDLINE | ID: mdl-21844074

ABSTRACT

BACKGROUND: Ischemia/reperfusion injury may have deleterious short- and long-term consequences for cardiac allografts. The underlying mechanisms involve microvascular dysfunction that may culminate in primary graft failure or untreatable chronic rejection. METHODS AND RESULTS: Here, we report that rat cardiac allograft ischemia/reperfusion injury resulted in profound microvascular dysfunction that was prevented by donor treatment with peroral single-dose simvastatin, a 3-hydroxy-3-methylglutaryl coenzyme A reductase and Rho GTPase inhibitor, 2 hours before graft procurement. During allograft preservation, donor simvastatin treatment inhibited microvascular endothelial cell and pericyte RhoA/Rho-associated protein kinase activation and endothelial cell-endothelial cell gap formation; decreased intragraft mRNA levels of hypoxia-inducible factor-1α, inducible nitric oxide synthase, and endothelin-1; and increased heme oxygenase-1. Donor, but not recipient, simvastatin treatment prevented ischemia/reperfusion injury-induced vascular leakage, leukocyte infiltration, the no-reflow phenomenon, and myocardial injury. The beneficial effects of simvastatin on vascular stability and the no-reflow phenomenon were abolished by concomitant nitric oxide synthase inhibition with N-nitro-l-arginine methyl ester and RhoA activation by geranylgeranyl pyrophosphate supplementation, respectively. In the chronic rejection model, donor simvastatin treatment inhibited cardiac allograft inflammation, transforming growth factor-ß1 signaling, and myocardial fibrosis. In vitro, simvastatin inhibited transforming growth factor-ß1-induced microvascular endothelial-to-mesenchymal transition. CONCLUSIONS: Our results demonstrate that donor simvastatin treatment prevents microvascular endothelial cell and pericyte dysfunction, ischemia/reperfusion injury, and chronic rejection and suggest a novel, clinically feasible strategy to protect cardiac allografts.


Subject(s)
Enzyme Inhibitors/therapeutic use , Graft Rejection/prevention & control , Heart Transplantation , Microvessels/drug effects , Primary Graft Dysfunction/prevention & control , Reperfusion Injury/prevention & control , Simvastatin/therapeutic use , Animals , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelin-1/biosynthesis , Gap Junctions/drug effects , Gap Junctions/enzymology , Heme Oxygenase-1/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Major Histocompatibility Complex/drug effects , Male , Microvessels/enzymology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/biosynthesis , No-Reflow Phenomenon/prevention & control , Polyisoprenyl Phosphates/pharmacology , Primary Graft Dysfunction/enzymology , Rats , Rats, Inbred WF , rho-Associated Kinases/antagonists & inhibitors
9.
J Cell Biochem ; 113(7): 2234-47, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22573546

ABSTRACT

The inhalation of asbestos fibers is considered to be highly harmful, and lead to fibrotic and/or malignant disease. Epithelial-to-mesenchymal transition (EMT) is a common pathogenic mechanism in asbestos associated fibrotic (asbestosis) and malignant lung diseases. The characterization of molecular pathways contributing to EMT may provide new possibilities for prognostic and therapeutic applications. The role of asbestos as an inducer of EMT has not been previously characterized. We exposed cultured human lung epithelial cells to crocidolite asbestos and analyzed alterations in the expression of epithelial and mesenchymal marker proteins and cell morphology. Asbestos was found to induce downregulation of E-cadherin protein levels in A549 lung carcinoma cells in 2-dimensional (2D) and 3D cultures. Similar findings were made in primary small airway epithelial cells cultured in 3D conditions where the cells retained alveolar type II cell phenotype. A549 cells also exhibited loss of cell-cell contacts, actin reorganization and expression of α-smooth muscle actin (α-SMA) in 2D cultures. These phenotypic changes were not associated with increased transforming growth factor (TGF)-ß signaling activity. MAPK/Erk signaling pathway was found to mediate asbestos-induced downregulation of E-cadherin and alterations in cell morphology. Our results suggest that asbestos can induce epithelial plasticity, which can be interfered by blocking the MAPK/Erk kinase activity.


Subject(s)
Alveolar Epithelial Cells/drug effects , Asbestos, Crocidolite/toxicity , Epithelial Cells/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System/drug effects , Pulmonary Alveoli/cytology , Actins/biosynthesis , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Cadherins/biosynthesis , Cell Line, Tumor , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/antagonists & inhibitors , Pulmonary Alveoli/metabolism , Pulmonary Fibrosis , Transforming Growth Factor beta/biosynthesis
10.
Cell Tissue Res ; 348(3): 491-503, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22434388

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive disease of unknown cause. The pathogenesis of the disease is characterized by fibroblast accumulation and excessive transforming growth factor-ß (TGF-ß) activation. Although TGF-ß activation is a complex process involving various protein interactions, little is known of the specific routes of TGF-ß storage and activation in human lung. Here, we have systematically analyzed the expression of specific proteins involved in extracellular matrix targeting and activation of TGF-ß. Latent TGF-ß-binding protein (LTBP)-1 was found to be significantly upregulated in IPF patient lungs. LTBP-1 expression was especially high in the fibroblastic foci, in which P-Smad2 immunoreactivity, indicative of TGF-ß signaling activity, was less prominent. In cultured primary lung fibroblasts and epithelial cells, short-interfering-RNA-mediated downregulation of LTBP-1 resulted in either increased or decreased TGF-ß signaling activity, respectively, suggesting that LTBP-1-mediated TGF-ß activation is dependent on the cellular context in the lung. Furthermore, LTBP-1 was shown to colocalize with fibronectin, fibrillin-1 and fibrillin-2 proteins in the IPF lung. Fibrillin-2, a developmental gene expressed only in blood vessels in normal adult lung, was found specifically upregulated in IPF fibroblastic foci. The TGF-ß-activating integrin ß8 subunit was expressed at low levels in both control and IPF lungs. Alterations in extracellular matrix composition, such as high levels of the TGF-ß storage protein LTBP-1 and the re-appearance of fibrillin-2, probably modulate TGF-ß availability and activation in different pulmonary compartments in the fibrotic lung.


Subject(s)
Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , Lung/pathology , Transforming Growth Factor beta1/metabolism , Adult , Aged , Case-Control Studies , Cells, Cultured , Down-Regulation/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibrillin-1 , Fibrillin-2 , Fibrillins , Fibroblasts/metabolism , Fibroblasts/pathology , Fibronectins/metabolism , Humans , Integrins/metabolism , Latent TGF-beta Binding Proteins/genetics , Latent TGF-beta Binding Proteins/metabolism , Microfilament Proteins/metabolism , Middle Aged , Phosphorylation , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Smad2 Protein/metabolism , Up-Regulation/genetics
11.
Am J Respir Cell Mol Biol ; 42(5): 626-32, 2010 May.
Article in English | MEDLINE | ID: mdl-19597127

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) (histopathology of usual interstitial pneumonia [UIP]) is a progressive disease with poor prognosis. Characteristic features of IPF/UIP include fibroblastic foci, which are patchy lesions of focal, disarranged myofibroblasts. GATA-6 is a transcription factor linked with cell differentiation. Its role in the development of IPF has not previously been investigated. We hypothesized that GATA-6 participates in the differentiation of fibroblasts into myofibroblasts in IPF/UIP lungs. The expression patterns of GATA-6, the mesenchymal marker alpha-smooth muscle actin (alpha-SMA), and markers for proliferation (Ki67) and apoptosis (caspase-3) were analyzed in human IPF/UIP tissue samples. The effects of GATA-6 overexpression and silencing were studied in cell cultures. The results show that the alpha-SMA-positive fibroblastic foci in IPF/UIP lungs are positive for GATA-6, but negative for Ki67 and caspase-3. Cultured human IPF/UIP fibroblasts expressed GATA-6 mRNA, whereas cells from the normal adult lung did not. In cultured A549 lung epithelial cells, the induction of GATA-6 by transforming growth factor-beta1 resulted in simultaneous expression of alpha-SMA and decrease of E-cadherin. The inhibition of GATA-6 expression in fibroblasts showed that GATA-6 mediates the alpha-SMA-inducing signal of transforming growth factor-beta1. In conclusion, the hallmark of IPF/UIP histopathology, the fibroblast focus, consists of differentiated, quiescent cells that prominently express GATA-6.


Subject(s)
Fibroblasts/metabolism , Fibroblasts/pathology , GATA6 Transcription Factor/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Actins/metabolism , Adult , Apoptosis/drug effects , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA-Binding Proteins/metabolism , Epithelium/drug effects , Epithelium/metabolism , Epithelium/pathology , Fibroblasts/drug effects , GATA6 Transcription Factor/genetics , Gene Expression Regulation/drug effects , Humans , Lung/drug effects , Lung/metabolism , Lung/pathology , Mesoderm/drug effects , Mesoderm/metabolism , Mesoderm/pathology , Protein Transport/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta1/pharmacology
12.
J Cell Physiol ; 223(3): 727-36, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20175115

ABSTRACT

Transforming growth factor (TGF)-beta is secreted and targeted into the extracellular matrix (ECM) in association with one of the latent TGF-beta binding proteins (LTBPs). Activation of these latent complexes is an important regulatory step in TGF-beta signaling. LTBPs target the growth factor into the ECM and expose it to activating mechanisms. Disruption of LTBP-4 gene causes severe developmental abnormalities in both humans and mice. Transcripts for two N-terminally distinct LTBP-4 variants, LTBP-4S (short) and -4L (long), have been identified. In the current work, we have characterized differences in the expression, processing, and ECM targeting of these LTBP-4 variants. Heart and skeletal muscle displayed expression of both variants, while liver expressed mainly LTBP-4L and lung as well as small intestine LTBP-4S. This tissue-specific expression pattern was found to originate from control of transcription by two independent promoters. Furthermore, LTBP-4S and -4L proteins were secreted and processed differently. During secretion, LTBP-4L was complexed with TGF-beta1, whereas the majority of LTBP-4S was secreted in a free form. In addition, LTBP-4S was incorporated into the ECM, while full-length LTBP-4L was not readily detectable in the ECM. These data suggest that LTBP-4 functions are modified by tissue-specific expression of the two N-terminally distinct variants, which in addition exhibit significant differences in cellular processing and targeting, that is, this provides a basis for understanding molecular diversity in LTBP-4 structure and function.


Subject(s)
Fibroblasts/metabolism , Gene Expression Profiling , Gene Expression Regulation , Latent TGF-beta Binding Proteins/genetics , Animals , Base Sequence , Cells, Cultured , Conserved Sequence , Humans , Latent TGF-beta Binding Proteins/chemistry , Latent TGF-beta Binding Proteins/metabolism , Mice , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Processing, Post-Translational , Sequence Analysis, DNA , Transcription Initiation Site
13.
Am J Pathol ; 174(1): 21-33, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19056849

ABSTRACT

Recent evidence suggests that subsets of lung fibroblasts differentially contribute to fibrogenic progression. We have previously shown that a subset of rat lung fibroblasts with fibrogenic characteristics [Thy-1 (-) fibroblasts] responds to stimuli (bleomycin, interleukin-4, etc) with increased latent transforming growth factor (TGF)-beta activation, whereas non-fibrogenic Thy-1-expressing [Thy-1 (+)] fibroblasts do not. Activation of latent TGF-beta1 by interstitial lung fibroblasts is critical for fibrogenic responses. To better understand the susceptibility of fibrogenic fibroblasts to the stimulation of TGF-beta activation, we examined the role of latent TGF-beta-binding proteins (LTBPs), key regulators of TGF-beta bioavailability and activation, in TGF-beta1 activation by these fibroblasts. Treatment of fibroblasts with bleomycin up-regulated LTBP-4 mRNA, protein, and soluble LTBP-4-bound large latent TGF-beta1 complexes in Thy-1 (-) fibroblasts to significantly higher levels than in Thy-1 (+) fibroblasts. Bleomycin-induced TGF-beta1 activation required LTBP-4, since lung fibroblasts deficient in LTBP-4 did not activate TGF-beta1. Expression of LTBP-4 restored TGF-beta1 activation in response to bleomycin, but expression either of LTBP-4 lacking the TGF-beta-binding site or only the TGF-beta-binding domain did not. Bleomycin treatment of mice increased LTBP-4 expression in the lung. Thy-1 knockout mice had increased levels of both LTBP-4 expression and TGF-beta activation, as well as enhanced Smad3 phosphorylation compared with wild-type mice. Together, these data identify a critical role for LTBP-4 in the regulation of latent TGF-beta1 activation in bleomycin-induced lung fibrosis.


Subject(s)
Fibroblasts/metabolism , Latent TGF-beta Binding Proteins/metabolism , Pulmonary Fibrosis/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Blotting, Northern , Blotting, Western , Cells, Cultured , Enzyme Activation/drug effects , Enzyme Activation/physiology , Fibroblasts/drug effects , Humans , Immunoprecipitation , Lung/drug effects , Lung/metabolism , Lung/pathology , Mice , Mice, Knockout , Pulmonary Fibrosis/chemically induced , Rats , Thy-1 Antigens/genetics , Thy-1 Antigens/metabolism
14.
Sci Rep ; 10(1): 7787, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32385381

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a lung parenchymal disease of unknown cause usually occurring in older adults. It is a chronic and progressive condition with poor prognosis and diagnosis is largely clinical. Currently, there exist few biomarkers that can predict patient outcome or response to therapies. Together with lack of markers, the need for novel markers for the detection and monitoring of IPF, is paramount. We have performed label-free plasma proteomics of thirty six individuals, 17 of which had confirmed IPF. Proteomics data was analyzed by volcano plot, hierarchical clustering, Partial-least square discriminant analysis (PLS-DA) and Ingenuity pathway analysis. Univariate and multivariate statistical analysis overlap identified haptoglobin-related protein as a possible marker of IPF when compared to control samples (Area under the curve 0.851, ROC-analysis). LXR/RXR activation and complement activation pathways were enriched in t-test significant proteins and oxidative regulators, complement proteins and protease inhibitors were enriched in PLS-DA significant proteins. Our pilot study points towards aberrations in complement activation and oxidative damage in IPF patients and provides haptoglobin-related protein as a new candidate biomarker of IPF.


Subject(s)
Blood Proteins , Complement System Proteins/immunology , Haptoglobins/metabolism , Idiopathic Pulmonary Fibrosis/immunology , Idiopathic Pulmonary Fibrosis/metabolism , Oxidative Stress , Proteomics , Signal Transduction , Aged , Biomarkers , Case-Control Studies , Complement System Proteins/metabolism , Computational Biology/methods , Female , Humans , Idiopathic Pulmonary Fibrosis/pathology , Male , Proteome , Proteomics/methods , ROC Curve
15.
Clin Lung Cancer ; 21(3): e142-e150, 2020 05.
Article in English | MEDLINE | ID: mdl-31734071

ABSTRACT

BACKGROUND: Previous preclinical studies have shown that activin A is overexpressed in malignant pleural mesothelioma (MPM), associates with cancer cachexia, and is observed in in vitro resistance to platinum-based chemotherapy. We evaluated circulating activin levels and their endogenous antagonists' follistatin/follistatin-like 3 in intrathoracic tumors. MATERIALS AND METHODS: Patients suspected of thoracic malignancy were recruited prior to surgery. Serum samples were collected from 21 patients with MPM, 59 patients with non-small-cell lung cancer (NSCLC), and 22 patients with benign lung lesions. Circulating activin/follistatin levels were measured using enzyme-linked immunosorbent assay and compared with clinicopathologic parameters. RESULTS: Circulating activin A levels were elevated in patients with MPM when compared with patients with NSCLC or benign lung lesion samples (P < .0001). Also, follistatin and follistatin-like 3 levels were the highest in MPM, although with less difference compared with activin A. Receiver operating characteristic analysis for activin A for separating NSCLC from benign lung lesion showed an area under the curve of 0.856 (95% confidence interval, 0.77-0.94). Activin A levels were higher in patients with cachexia (P < .001). In patients with MPM, activin A levels correlated positively with computed tomography-based baseline tumor size (R = 0.549; P = .010) and the change in tumor size after chemotherapy (R = 0.743; P = .0006). Patients with partial response or stable disease had lower circulating activin A levels than the ones with progressive disease (P = .028). CONCLUSION: Activin A serum level could be used as a biomarker in differentiating malignant and benign lung tumors. Circulating activin A levels were elevated in MPM and associates with cancer cachexia and reduced chemotherapy response.


Subject(s)
Activins/blood , Biomarkers, Tumor/blood , Cachexia/diagnosis , Mesothelioma, Malignant/drug therapy , Platinum/adverse effects , Pleural Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cachexia/blood , Cachexia/chemically induced , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Female , Follow-Up Studies , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Male , Mesothelioma, Malignant/pathology , Middle Aged , Pleural Neoplasms/pathology , Prognosis , Prospective Studies , ROC Curve , Retrospective Studies
16.
Trends Cell Biol ; 14(12): 657-9, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15564041

ABSTRACT

Transforming growth factor (TGF)-betas are powerful cytokines that are secreted as inactive (latent) precursors into the extracellular space. To exert their pleiotropic functions, latent TGF-betas require activation. This requisite restricts TGF-beta signaling to tissues that express TGF-beta-activating proteins such as the adhesion molecule alphavbeta6 integrin. Recent work has uncovered the molecular mechanism by which alphavbeta6 integrin activates latent TGF-beta. Latent-TGF-beta-binding protein 1 has been identified as being the major component of this process, and the integrin-interacting region has been mapped to a poorly conserved sequence stretch called the hinge region.


Subject(s)
Antigens, Neoplasm/physiology , Integrins/physiology , Intracellular Signaling Peptides and Proteins/physiology , Transforming Growth Factor beta/physiology , Animals , Antigens, Neoplasm/metabolism , Humans , Integrins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Latent TGF-beta Binding Proteins , Models, Biological , Protein Isoforms/physiology , Stress, Mechanical , Transforming Growth Factor beta/metabolism
17.
J Cell Biol ; 167(1): 123-33, 2004 Oct 11.
Article in English | MEDLINE | ID: mdl-15466481

ABSTRACT

Disruption of latent TGF-beta binding protein (LTBP)-4 expression in the mouse leads to abnormal lung development and colorectal cancer. Lung fibroblasts from these mice produced decreased amounts of active TGF-beta, whereas secretion of latent TGF-beta was significantly increased. Expression and secretion of TGF-beta2 and -beta3 increased considerably. These results suggested that TGF-beta activation but not secretion would be severely impaired in LTBP-4 -/- fibroblasts. Microarrays revealed increased expression of bone morphogenic protein (BMP)-4 and decreased expression of its inhibitor gremlin. This finding was accompanied by enhanced expression of BMP-4 target genes, inhibitors of differentiation 1 and 2, and increased deposition of fibronectin-rich extracellular matrix. Accordingly, increased expression of BMP-4 and decreased expression of gremlin were observed in mouse lung. Transfection of LTBP-4 rescued the -/- fibroblast phenotype, while LTBP-1 was inefficient. Treatment with active TGF-beta1 rescued BMP-4 and gremlin expression to wild-type levels. Our results indicate that the lack of LTBP-4-mediated targeting and activation of TGF-beta1 leads to enhanced BMP-4 signaling in mouse lung.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Bone Morphogenetic Proteins/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Blotting, Northern , Bone Morphogenetic Protein 4 , Cell Differentiation , DNA, Complementary/metabolism , Electrophoresis, Polyacrylamide Gel , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/metabolism , Latent TGF-beta Binding Proteins , Lung/metabolism , Mice , Mice, Transgenic , Microscopy, Fluorescence , Oligonucleotide Array Sequence Analysis , Oligonucleotides/chemistry , Phenotype , RNA/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Up-Regulation
18.
Exp Cell Res ; 314(13): 2488-500, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18585707

ABSTRACT

Latent transforming growth factor (TGF)-beta binding proteins are extracellular matrix (ECM) proteins involved in the regulation of TGF-beta sequestration and activation. In this study, we have identified binding domains in LTBP-4, which mediate matrix targeting and cell adhesion. LTBP-4 was found to possess heparin binding activity, especially in its N-terminal region. The C-terminal domain of LTBP-4 supported fibroblast adhesion, a property reduced by soluble heparin. In addition, we found that LTBP-4 binds directly to fibronectin (FN), which was indispensable for the matrix assembly of LTBP-4. The FN binding sites were also located in the N-terminal region. Interestingly, heparin was able to reduce the binding of LTBP-4 to FN. In fibroblast cultures, LTBP-4 colocalized first with FN and subsequently with fibrillin-1, pointing to a role for FN in the early assembly of LTBP-4. In FN -/- fibroblasts, LTBP-mediated ECM targeting was disturbed, resulting in increased TGF-beta activity. These results revealed new molecular interactions which are evidently important for the ECM targeting, but which also are evidence of novel functions for LTBP-4 as an adhesion molecule.


Subject(s)
Cell Communication , Extracellular Matrix/metabolism , Fibronectins/metabolism , Heparin/metabolism , Latent TGF-beta Binding Proteins/metabolism , Latent TGF-beta Binding Proteins/physiology , Animals , Binding Sites/physiology , CHO Cells , Cell Adhesion , Cells, Cultured , Cricetinae , Cricetulus , Fibroblasts/metabolism , Humans , Latent TGF-beta Binding Proteins/chemistry , Mice , Mink , Models, Biological , Protein Binding , Protein Structure, Tertiary/physiology
19.
Am J Respir Crit Care Med ; 177(3): 321-9, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-17975199

ABSTRACT

RATIONALE: Members of the transforming growth factor (TGF)-beta superfamily, including TGF-betas and bone morphogenetic proteins (BMPs), are essential for the maintenance of tissue homeostasis and regeneration after injury. We have observed that the BMP antagonist, gremlin, is highly up-regulated in idiopathic pulmonary fibrosis (IPF). OBJECTIVES: To investigate the role of gremlin in the regulation of BMP signaling in pulmonary fibrosis. METHODS: Progressive asbestos-induced fibrosis in the mouse was used as a model of human IPF. TGF-beta and BMP expression and signaling activities were measured from murine and human fibrotic lungs. The mechanism of gremlin induction was analyzed in cultured lung epithelial cells. In addition, the possible therapeutic role of gremlin inhibition was tested by administration of BMP-7 to mice after asbestos exposure. MEASUREMENTS AND MAIN RESULTS: Gremlin mRNA levels were up-regulated in the asbestos-exposed mouse lungs, which is in agreement with the human IPF biopsy data. Down-regulation of BMP signaling was demonstrated by reduced levels of Smad1/5/8 and enhanced Smad2 phosphorylation in asbestos-treated lungs. Accordingly, analyses of cultured human bronchial epithelial cells indicated that asbestos-induced gremlin expression could be prevented by inhibitors of the TGF-beta receptor and also by inhibitors of the mitogen-activated protein kinase kinase/extracellular signal-regulated protein kinase pathways. BMP-7 treatment significantly reduced hydroxyproline contents in the asbestos-treated mice. CONCLUSIONS: The TGF-beta and BMP signaling balance is important for lung regenerative events and is significantly perturbed in pulmonary fibrosis. Rescue of BMP signaling activity may represent a potential beneficial strategy for treating human pulmonary fibrosis.


Subject(s)
Bone Morphogenetic Proteins/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Pulmonary Fibrosis/physiopathology , Transforming Growth Factor beta/physiology , Animals , Asbestos/adverse effects , Bone Morphogenetic Protein 7 , Cell Line, Tumor , Disease Models, Animal , Down-Regulation , Humans , Mice , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Signal Transduction , Transforming Growth Factor beta1/physiology , Up-Regulation
20.
COPD ; 6(4): 234-41, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19811381

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is characterized by inflammatory immune response, emphysematous destruction of alveolar structures and obstruction in the small conducting airways. Transforming growth factor (TGF)-beta is involved in the maintenance of normal lung tissue homeostasis as a regulator of extracellular proteolysis, tissue repair and inflammatory functions. This study was undertaken to characterize TGF-beta signaling in pathologically distinct areas of COPD lungs. Using Smad2 phosporylation (P-Smad2) as an indicator of TGF-beta signaling activity we analyzed COPD patient tissues and controls by immunohistochemistry. Emphysematous lung showed significantly reduced P-Smad2 immunoreactivity both in the alveolar and bronchiolar epithelium, which is evidence of reduced TGF-beta signaling activity. On the contrary, in the thickened peribronchial areas there was an increase in the amount of P-Smad2 positive cells. Isolated COPD lung fibroblasts also displayed increased TGF-beta signaling and target gene expression suggesting that the fibroblasts are characteristic to the small airway disease phenotype. Our results indicate that TGF-beta signaling activity is differentially regulated in distinct areas of COPD lung and likely contributes to both emphysematous development and small airway obstruction.


Subject(s)
Pulmonary Disease, Chronic Obstructive/metabolism , Smad2 Protein/metabolism , Transforming Growth Factor beta/metabolism , Biopsy , Case-Control Studies , Female , Fibroblasts/metabolism , Gene Expression Regulation , Humans , Immunoenzyme Techniques , Male , Middle Aged , Phenotype , Phosphorylation , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/pathology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology , Statistics, Nonparametric , Transforming Growth Factor beta/physiology
SELECTION OF CITATIONS
SEARCH DETAIL