Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
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
2.
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
3.
ERJ Open Res ; 5(3)2019 Jul.
Article in English | MEDLINE | ID: mdl-31304177

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is characterised by unpredictable disease course and poor survival. After the introduction of novel antifibrotic drugs, the prognosis of patients with IPF is probably changing. FinnishIPF, a nationwide registry of carefully characterised patients, was initiated in Finland in 2011. For the data analysis, we included 453 incident IPF patients diagnosed during 2011-2015. In this study, we describe the demographics and prognosis of these real-life patients. The median overall survival time of registered IPF patients was 4.5 years. The transplant-free survival at 1, 2, 3, 4 and 5 years was 95%, 83%, 70%, 58% and 45%, respectively. Smoking did not have any effect on survival. 117 (26%) patients received pirfenidone or nintedanib. Patients who received ≥6 months of treatment had better survival compared with those who did not receive treatment but this difference disappeared after age adjustment. The transplantation rate was 3%. Although IPF is diagnosed in Finland at a older age, the prognosis is better than expected due to a relatively well preserved lung function at diagnosis. Age and pulmonary function were identified as independent predictors of survival in the entire IPF patient population as well as in patients who had received antifibrotic treatment.

4.
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
5.
Sci Rep ; 8(1): 10070, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29968778

ABSTRACT

Malignant mesothelioma is an aggressive cancer with poor prognosis. It is characterized by prominent extracellular matrix, mesenchymal tumor cell phenotypes and chemoresistance. In this study, the ability of pirfenidone to alter mesothelioma cell proliferation and migration as well as mesothelioma tumor microenvironment was evaluated. Pirfenidone is an anti-fibrotic drug used in the treatment of idiopathic pulmonary fibrosis and has also anti-proliferative activities. Mesothelioma cell proliferation was decreased by pirfenidone alone or in combination with cisplatin. Pirfenidone also decreased significantly Transwell migration/invasion and 3D collagen invasion. This was associated with increased BMP pathway activity, decreased GREM1 expression and downregulation of MAPK/ERK and AKT/mTOR signaling. The canonical Smad-mediated TGF-ß signaling was not affected by pirfenidone. However, pirfenidone blocked TGF-ß induced upregulation of ERK and AKT pathways. Treatment of mice harboring mesothelioma xenografts with pirfenidone alone did not reduce tumor proliferation in vivo. However, pirfenidone modified the tumor microenvironment by reducing the expression of extracellular matrix associated genes. In addition, GREM1 expression was downregulated by pirfenidone in vivo. By reducing two major upregulated pathways in mesothelioma and by targeting tumor cells and the microenvironment pirfenidone may present a novel anti-fibrotic and anti-cancer adjuvant therapy for mesothelioma.


Subject(s)
Lung Neoplasms/drug therapy , Mesothelioma/metabolism , Pyridones/pharmacology , Tumor Microenvironment/drug effects , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Collagen/metabolism , Extracellular Matrix/metabolism , Female , Fibroblasts/metabolism , Fibrosis , Humans , Lung Neoplasms/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mesothelioma/drug therapy , Mesothelioma, Malignant , Mice , Mice, Inbred BALB C , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Pyridones/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
6.
Int J Pharm ; 544(1): 121-128, 2018 Jun 10.
Article in English | MEDLINE | ID: mdl-29655797

ABSTRACT

The aim of this work was to study the antifibrotic effect of pulmonary administration of tilorone to lung fibrosis. L-leucine coated tilorone particles were prepared and their aerosolization properties were analyzed using two dry powder inhalers (Easyhaler and Twister). In addition, the biological activity and cell monolayer permeation was tested. The antifibrotic effect of tilorone delivered by oropharyngeal aspiration was studied in vivo using a silica-induced model of pulmonary fibrosis in mice in a preventive setting. When delivered from the Easyhaler in an inhalation simulator, the emitted dose and fine particle fraction were independent from the pressure applied and showed dose repeatability. However, with Twister the aerosolization was pressure-dependent indicating poor compatibility between the device and the formulation. The formulation showed more consistent permeation through a differentiated Calu-3 cell monolayer compared to pristine tilorone. Tilorone decreased the histological fibrosis score in vivo in systemic and local administration, but only systemic administration decreased the mRNA expression of type I collagen. The difference was hypothesized to result from 40-fold higher drug concentration in tissue samples in the systemic administration group. These results show that tilorone can be formulated as inhalable dry powder and has potential as an oral and inhalable antifibrotic drug.


Subject(s)
Dry Powder Inhalers , Nanoparticles/administration & dosage , Pulmonary Fibrosis/drug therapy , Tilorone/administration & dosage , Administration, Inhalation , Animals , Cell Line , Humans , Leucine/administration & dosage , Leucine/chemistry , Leucine/therapeutic use , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Mice, Inbred C57BL , Microscopy, Electron, Scanning , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Nanoparticles/ultrastructure , Powders , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Silicon Dioxide , Tilorone/chemistry , Tilorone/pharmacokinetics , Tilorone/therapeutic use
7.
Oncotarget ; 8(58): 98280-98297, 2017 Nov 17.
Article in English | MEDLINE | ID: mdl-29228689

ABSTRACT

Malignant mesothelioma originates from mesothelial cells and is a cancer type that aggressively invades into the surrounding tissue, has poor prognosis and no effective treatment. Gremlin-1 is a cysteine knot protein that functions by inhibiting BMP-pathway activity during development. BMP-independent functions have also been described for gremlin-1. We have previously shown high gremlin-1 expression in mesothelioma tumor tissue. Here, we investigated the functions of gremlin-1 in mesothelioma cell migration and invasive growth. Gremlin-1 promoted mesothelioma cell sprouting and invasion into three dimensional collagen and Matrigel matrices. The expression level of gremlin-1 was linked to changes in the expression of SNAI2, integrins, matrix metalloproteinases (MMP) and TGF-ß family signaling - all previously associated with a mesenchymal invasive phenotype. Small molecule inhibitors of MMPs completely blocked mesothelioma cell invasive growth. In addition, inhibitors of TGF-ß receptors significantly reduced invasive growth. This was associated with reduced expression of MMP2 but not SNAI2, indicating that gremlin-1 has both TGF-ß pathway dependent and independent mechanisms of action. Results of in vivo mesothelioma xenograft experiments indicated that gremlin-1 overexpressing tumors were more vascular and had a tendency to send metastases. This suggests that by inducing a mesenchymal invasive cell phenotype together with enhanced tumor vascularization, gremlin-1 drives mesothelioma invasion and metastasis. These data identify gremlin-1 as a potential therapeutic target in mesothelioma.

8.
Matrix Biol ; 59: 109-120, 2017 05.
Article in English | MEDLINE | ID: mdl-27645114

ABSTRACT

Latent transforming growth factor beta binding protein 4 (LTBP4) belongs to the fibrillin/LTBP family of proteins and plays an important role as a structural component of extracellular matrix (ECM) and local regulator of TGFß signaling. We have previously reported that Ltbp4S knock out mice (Ltbp4S-/-) develop centrilobular emphysema reminiscent of late stage COPD, which could be partially rescued by inactivating the antioxidant protein Sestrin 2 (Sesn2). More recent studies showed that Sesn2 knock out mice upregulate Pdgfrß-controlled alveolar maintenance programs that protect against cigarette smoke induced pulmonary emphysema. Based on this, we hypothesized that the emphysema of Ltbp4S-/- mice is primarily caused by defective Pdgfrß signaling. Here we show that LTBP4 induces Pdgfrß signaling by inhibiting the antioxidant Nrf2/Keap1 pathway in a TGFß-dependent manner. Overall, our data identified Ltbp4 as a major player in lung remodeling and injury repair.


Subject(s)
Extracellular Matrix/metabolism , Latent TGF-beta Binding Proteins/genetics , NF-E2-Related Factor 2/genetics , Pulmonary Emphysema/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics , Transforming Growth Factor beta/genetics , Animals , Cell Line , Epithelial Cells/cytology , Epithelial Cells/metabolism , Extracellular Matrix/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Latent TGF-beta Binding Proteins/deficiency , Lung/metabolism , Lung/pathology , Mice , Mice, Knockout , Mink , NF-E2-Related Factor 2/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Peroxidases , Plasmids/chemistry , Plasmids/metabolism , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/pathology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Tropoelastin/deficiency , Tropoelastin/genetics
9.
PLoS One ; 11(7): e0159010, 2016.
Article in English | MEDLINE | ID: mdl-27428020

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is characterized by activation and injury of epithelial cells, the accumulation of connective tissue and changes in the inflammatory microenvironment. The bone morphogenetic protein (BMP) inhibitor protein gremlin-1 is associated with the progression of fibrosis both in human and mouse lung. We generated a transgenic mouse model expressing gremlin-1 in type II lung epithelial cells using the surfactant protein C (SPC) promoter and the Cre-LoxP system. Gremlin-1 protein expression was detected specifically in the lung after birth and did not result in any signs of respiratory insufficiency. Exposure to silicon dioxide resulted in reduced amounts of lymphocyte aggregates in transgenic lungs while no alteration in the fibrotic response was observed. Microarray gene expression profiling and analyses of bronchoalveolar lavage fluid cytokines indicated a reduced lymphocytic response and a downregulation of interferon-induced gene program. Consistent with reduced Th1 response, there was a downregulation of the mRNA and protein expression of the anti-fibrotic chemokine CXCL10, which has been linked to IPF. In human IPF patient samples we also established a strong negative correlation in the mRNA expression levels of gremlin-1 and CXCL10. Our results suggest that in addition to regulation of epithelial-mesenchymal crosstalk during tissue injury, gremlin-1 modulates inflammatory cell recruitment and anti-fibrotic chemokine production in the lung.


Subject(s)
Chemokine CXCL10/immunology , Idiopathic Pulmonary Fibrosis/immunology , Intercellular Signaling Peptides and Proteins/genetics , Lung/immunology , Lymphocytes/immunology , Up-Regulation , Animals , Cell Line , Chemokine CXCL10/analysis , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Intercellular Signaling Peptides and Proteins/analysis , Interferons/immunology , Lung/pathology , Lymphocytes/pathology , Mice , Mice, Transgenic , Silicon Dioxide/immunology
10.
Mol Endocrinol ; 29(12): 1787-91, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26495869

ABSTRACT

Recently, several LH/human chorionic gonadotropin (hCG) receptor-independent activities for hCG have been described, including activation of the TGF-ß receptor (TGFßR) by hyperglycosylated hCG and stimulation of trophoblast invasion. Because the hCG concentrations used in these studies have been rather high, reflecting physiological hCG levels in pregnancy, even a minor contamination with growth factors, which act at very low concentrations, may be significant. Several commercial hCG preparations have been found to contain significant amounts of epidermal growth factor (EGF), which we also confirmed here. Furthermore, we found that some hCG preparations also contain significant amounts of TGF-ß1. These hCG preparations were able to activate ERK1/2 in JEG-3 choriocarcinoma cells or TGFßR in mink lung epithelial cells transfected with a reporter gene for TGFßR activation. No such activation was found with highly purified hCG or its free ß-subunit (hCGß), irrespective of whether they were hyperglycosylated or not. Taken together, our results suggest that the growth factor contaminations in the hCG preparations can cause activation of TGFßR and, at least in JEG-3 cells, MAPK signaling. This highlights the importance to carefully control for potential contaminations and that highly purified hCG preparations have to be used for biological studies.


Subject(s)
Chorionic Gonadotropin/isolation & purification , Chorionic Gonadotropin/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Cell Line, Tumor , Female , Humans , Pregnancy , Signal Transduction
11.
Dis Model Mech ; 8(4): 403-15, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25713297

ABSTRACT

Recent studies have revealed an important role for LTBP-4 in elastogenesis. Its mutational inactivation in humans causes autosomal recessive cutis laxa type 1C (ARCL1C), which is a severe disorder caused by defects of the elastic fiber network. Although the human gene involved in ARCL1C has been discovered based on similar elastic fiber abnormalities exhibited by mice lacking the short Ltbp-4 isoform (Ltbp4S(-/-)), the murine phenotype does not replicate ARCL1C. We therefore inactivated both Ltbp-4 isoforms in the mouse germline to model ARCL1C. Comparative analysis of Ltbp4S(-/-) and Ltbp4-null (Ltbp4(-/-)) mice identified Ltbp-4L as an important factor for elastogenesis and postnatal survival, and showed that it has distinct tissue expression patterns and specific molecular functions. We identified fibulin-4 as a previously unknown interaction partner of both Ltbp-4 isoforms and demonstrated that at least Ltbp-4L expression is essential for incorporation of fibulin-4 into the extracellular matrix (ECM). Overall, our results contribute to the current understanding of elastogenesis and provide an animal model of ARCL1C.


Subject(s)
Cutis Laxa/genetics , Cutis Laxa/pathology , Genes, Recessive , Latent TGF-beta Binding Proteins/genetics , Animals , Animals, Newborn , Aorta/abnormalities , Aorta/pathology , Cardiomegaly/complications , Cardiomegaly/pathology , Elastic Tissue/metabolism , Elastin/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Female , Gene Silencing , Glycosylation , Heart Ventricles/pathology , Humans , Latent TGF-beta Binding Proteins/chemistry , Latent TGF-beta Binding Proteins/deficiency , Latent TGF-beta Binding Proteins/metabolism , Lung/abnormalities , Lung/pathology , Mice, Inbred C57BL , Models, Biological , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/metabolism , Skin/pathology , Weight Loss
12.
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
13.
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
14.
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
15.
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
16.
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
17.
Acta Ophthalmol ; 91(6): 531-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23106921

ABSTRACT

PURPOSE: Angiogenesis in diabetic retinopathy (DR) is a multifactorial process regulated by hypoxia-induced growth factors and inflammatory cytokines. In addition to the angiogenic switch, the proteolytic processing and altered synthesis of the extracellular matrix are critical steps in this disease. This study was performed to evaluate the levels of matrix metalloproteinase-2 and matrix metalloproteinase-9 (MMP-2 and MMP-9), angiopoietin-1 and angiopoietin-2 (Ang-1 and Ang-2), vascular endothelial growth factor (VEGF), erythropoietin (EPO) and transforming growth factor-ß1 (totalTGFß1) in the vitreous of diabetic eyes undergoing vitrectomy compared with control eyes operated because of macular hole or pucker. METHODS: Prospective consecutive controlled observational study performed in the unit of vitreoretinal surgery in Finland during the years 2006-2008. Vitreous samples were collected before the start of the conventional 3-ppp vitrectomy. Vitreous MMP-2 and MMP-9, Ang-1 and Ang-2, VEGF, EPO and TGFß1 concentrations were measured from 69 patients with Type 1 or 2 diabetes and 40 controls. RESULTS: Comparison of eyes with DR with controls revealed that the mean vitreous concentrations of proMMP-2 (p = 0.0015), totalMMP-2 (p = 0.0011), proMMP-9 (p = 0.00001), totalMMP-9 (p < 0.00001), Ang-2 (p < 0.00001), VEGF (p < 0.00001), EPO (p < 0.00001) and totalTGFß1 (p = 0.000026) were significantly higher in the former group. A multivariate logistic regression analysis suggested intravitreal Ang-2 concentration being the key marker of PDR (p = 0.00025) (OR = 1507.9). CONCLUSION: The main new finding is that the intravitreal concentrations of Ang-2 correlated significantly with MMP-9, VEGF, EPO and TGFß1 levels in diabetic eyes undergoing vitrectomy. Thus, these factors could promote retinal angiogenesis synergistically.


Subject(s)
Diabetic Retinopathy/metabolism , Erythropoietin/metabolism , Matrix Metalloproteinase 9/metabolism , Transforming Growth Factor beta1/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vesicular Transport Proteins/metabolism , Vitrectomy , Aged , Angiopoietin-1/metabolism , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 2/complications , Diabetic Retinopathy/surgery , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Matrix Metalloproteinase 2/metabolism , Middle Aged , Prospective Studies , Retinal Neovascularization/metabolism , Retinal Perforations/metabolism , Tomography, Optical Coherence , Up-Regulation , Vitreous Body/metabolism
18.
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
19.
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
20.
Endocrinology ; 153(3): 1509-18, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22294741

ABSTRACT

Genetic studies have identified bone morphogenetic protein-15 (BMP15) as an essential regulator of female fertility in humans and in sheep. Oocyte-derived BMP15 is a noncovalently linked dimeric growth factor mediating its effects to ovarian somatic cells in a paracrine manner. Although receptor ectodomains capable of binding BMP15 have previously been reported, no cell surface receptor complex involved in BMP15 signaling has previously been characterized. Here we have expressed and purified recombinant human BMP15 noncovalent and covalent dimer variants. The biological effects of these BMP15 variants were assessed in cultured human granulosa-luteal cells or COV434 granulosa cell tumor cells using BMP-responsive transcriptional reporter assays and an inhibin B ELISA. Biochemical characterization of ligand-receptor interactions was performed with affinity-labeling experiments using [(125)I]iodinated BMP15 variants. Both ligand variants were shown to form homodimers and to stimulate Smad1/5/8 signaling and inhibin B production in human granulosa cells in a similar manner. [(125)I]Iodination of both ligands was achieved, but only the covalent dimer variant retained receptor binding capacity. The [(125)I]BMP15(S356C) variant bound preferentially to endogenous BMP receptor 1B (BMPR1B) and BMPR2 receptors on COV434 cells. Binding experiments in COS cells with overexpression of these receptors confirmed that the [(125)I]BMP15(S356C) variant binds to BMPR1B and BMPR2 forming the BMP15 signaling complex. The results provide the first direct evidence in any species on the identification of specific cell surface receptors for a member of the GDF9/BMP15 subfamily of oocyte growth factors. The fact that BMP15 uses preferentially BMPR1B as its type I receptor suggests an important role for the BMPR1B receptor in human female fertility. The result is well in line with the demonstration of ovarian failure in a recently reported human subject with a homozygous BMPR1B loss-of-function mutant.


Subject(s)
Bone Morphogenetic Protein 15/metabolism , Bone Morphogenetic Protein Receptors, Type I/metabolism , Cell Membrane/metabolism , Gene Expression Regulation , Granulosa Cells/cytology , Ovary/metabolism , Animals , COS Cells , Chlorocebus aethiops , Dimerization , Female , Genes, Reporter , Granulosa Cells/metabolism , Homozygote , Humans , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...