Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Immunol Cell Biol ; 101(5): 412-427, 2023 05.
Article in English | MEDLINE | ID: mdl-36862017

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, fibrotic interstitial lung disease of unknown etiology. The accumulation of macrophages is associated with disease pathogenesis. The unfolded protein response (UPR) has been linked to macrophage activation in pulmonary fibrosis. To date, the impact of activating transcription factor 6 alpha (ATF6α), one of the UPR mediators, on the composition and function of pulmonary macrophage subpopulations during lung injury and fibrogenesis is not fully understood. We began by examining the expression of Atf6α in IPF patients' lung single-cell RNA sequencing dataset, archived surgical lung specimens, and CD14+ circulating monocytes. To assess the impact of ATF6α on pulmonary macrophage composition and pro-fibrotic function during tissue remodeling, we conducted an in vivo myeloid-specific deletion of Atf6α. Flow cytometric assessments of pulmonary macrophages were carried out in C57BL/6 and myeloid specific ATF6α-deficient mice in the context of bleomycin-induced lung injury. Our results demonstrated that Atf6α mRNA was expressed in pro-fibrotic macrophages found in the lung of a patient with IPF and in CD14+ circulating monocytes obtained from blood of a patient with IPF. After bleomycin administration, the myeloid-specific deletion of Atf6α altered the pulmonary macrophage composition, expanding CD11b+ subpopulations with dual polarized CD38+ CD206+ expressing macrophages. Compositional changes were associated with an aggravation of fibrogenesis including increased myofibroblast and collagen deposition. A further mechanistic ex vivo investigation revealed that ATF6α was required for CHOP induction and the death of bone marrow-derived macrophages. Overall, our findings suggest a detrimental role for the ATF6α-deficient CD11b+ macrophages which had altered function during lung injury and fibrosis.


Subject(s)
Idiopathic Pulmonary Fibrosis , Lung Injury , Mice , Animals , Lung Injury/metabolism , Activating Transcription Factor 6/metabolism , Mice, Inbred C57BL , Macrophages/metabolism , Lung/pathology , Idiopathic Pulmonary Fibrosis/pathology , Fibrosis , Bleomycin/adverse effects , Bleomycin/metabolism
2.
Am J Respir Cell Mol Biol ; 64(2): 235-246, 2021 02.
Article in English | MEDLINE | ID: mdl-33253593

ABSTRACT

Pulmonary fibrosis is a progressive lung disease characterized by myofibroblast accumulation and excessive extracellular matrix deposition. We sought to investigate the role of FKBP13 (13-kD FK506-binding protein), an endoplasmic reticulum-resident molecular chaperone, in various forms of pulmonary fibrosis. We first characterized the gene and protein expression of FKBP13 in lung biopsy specimens from 24 patients with idiopathic pulmonary fibrosis and 17 control subjects. FKBP13 expression was found to be elevated in the fibrotic regions of idiopathic pulmonary fibrosis lung tissues and correlated with declining forced vital capacity and dyspnea severity. FKBP13 expression was also increased in lung biopsy specimens of patients with hypersensitivity pneumonitis, rheumatoid arthritis, and sarcoidosis-associated interstitial lung disease. We next evaluated the role of this protein using FKBP13-/- mice in a bleomycin model of pulmonary fibrosis. Animals were assessed for lung function and histopathology at different stages of lung injury including the inflammatory (Day 7), fibrotic (Day 21), and resolution (Day 50) phases. FKBP13-/- mice showed increased infiltration of inflammatory cells and cytokines at Day 7, increased lung elastance and fibrosis at Day 21, and impaired resolution of fibrosis at Day 50. These changes were associated with an increased number of cells that stained positive for TUNEL and cleaved caspase 3 in the FKBP13-/- lungs, indicating a heightened cellular sensitivity to bleomycin. Our findings suggest that FKBP13 is a potential biomarker for severity of interstitial lung diseases and that it has a biologically relevant role in protecting mice against bleomycin-induced injury, inflammation, and fibrosis.


Subject(s)
Lung Diseases, Interstitial/metabolism , Lung Diseases, Interstitial/pathology , Tacrolimus Binding Proteins/metabolism , Up-Regulation/physiology , Animals , Biomarkers/metabolism , Biopsy/methods , Bleomycin/adverse effects , Cytokines/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Female , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Inflammation/metabolism , Inflammation/pathology , Lung , Male , Mice , Mice, Inbred C57BL , Middle Aged , Severity of Illness Index , Up-Regulation/drug effects
3.
J Cell Mol Med ; 24(6): 3481-3491, 2020 03.
Article in English | MEDLINE | ID: mdl-32052562

ABSTRACT

Patients on peritoneal dialysis are at risk of developing peritoneal fibrosis and angiogenesis, which can lead to dysfunction of the peritoneal membrane. Recent evidence has identified cross-talk between transforming growth factor beta (TGFB) and the WNT/ß-catenin pathway to induce fibrosis and angiogenesis. Limited evidence exists describing the role of non-canonical WNT signalling in peritoneal membrane injury. Non-canonical WNT5A is suggested to have different effects depending on the receptor environment. WNT5A has been implicated in antagonizing canonical WNT/ß-catenin signalling in the presence of receptor tyrosine kinase-like orphan receptor (Ror2). We co-expressed TGFB and WNT5A using adenovirus and examined its role in the development of peritoneal fibrosis and angiogenesis. Treatment of mouse peritoneum with AdWNT5A decreased the submesothelial thickening and angiogenesis induced by AdTGFB. WNT5A appeared to block WNT/ß-catenin signalling by inhibiting phosphorylation of glycogen synthase kinase 3 beta (GSK3B) and reducing levels of total ß-catenin and target proteins. To examine the function of Ror2, we silenced Ror2 in a human mesothelial cell line. We treated cells with AdWNT5A and observed a significant increase in fibronectin compared with AdWNT5A alone. We also analysed fibronectin and vascular endothelial growth factor (VEGF) in a TGFB model of mesothelial cell injury. Both fibronectin and VEGF were significantly increased in response to Ror2 silencing when cells were exposed to TGFB. Our results suggest that WNT5A inhibits peritoneal injury and this is associated with a decrease in WNT/ß-catenin signalling. In human mesothelial cells, Ror2 is involved in regulating levels of fibronectin and VEGF.


Subject(s)
Peritoneum/metabolism , Peritoneum/pathology , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Wnt-5a Protein/metabolism , Animals , Epithelial-Mesenchymal Transition , Epithelium/metabolism , Fibronectins/metabolism , Fibrosis , Humans , Membranes , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway
4.
Chest ; 157(5): 1207-1220, 2020 05.
Article in English | MEDLINE | ID: mdl-31778676

ABSTRACT

Chronic lung disease accounts for a significant global burden with respect to death, disability, and health-care costs. Due to the heterogeneous nature and limited treatment options for these diseases, it is imperative that the cellular and molecular mechanisms underlying the disease pathophysiology are further understood. The lung is a complex organ with a diverse cell population, and each cell type will likely have different roles in disease initiation, progression, and resolution. The effectiveness of a given therapeutic agent may depend on the net effect on each of these cell types. Over the past decade, it has been established that endoplasmic reticulum stress and the unfolded protein response are involved in the development of several chronic lung diseases. These conserved cellular pathways are important for maintaining cellular proteostasis, but their aberrant activation can result in pathology. This review discusses the current understanding of endoplasmic reticulum stress and the unfolded protein response at the cellular level in the development and progression of various chronic lung diseases. We highlight the need for increased understanding of the specific cellular contributions of unfolded protein response activation to these pathologies and suggest that the development of cell-specific targeted therapies is likely required to further decrease disease progression and to promote resolution of chronic lung disease.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Lung Diseases/drug therapy , Lung Diseases/physiopathology , Unfolded Protein Response , Chronic Disease , Disease Progression , Humans , Protein Unfolding
5.
Am J Physiol Renal Physiol ; 317(1): F152-F162, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31141397

ABSTRACT

Glomerulonephritis (GN) is a common cause of end-stage kidney disease and is characterized by glomerular inflammation, hematuria, proteinuria, and progressive renal dysfunction. Transforming growth factor (TGF)-ß is involved in glomerulosclerosis and interstitial fibrosis. TGF-ß activates multiple signaling pathways, including the canonical SMAD pathway. We evaluated the role of SMAD signaling in renal injury and proteinuria in a murine model of GN. SMAD3+/+ or SMAD3-/- mice received anti-glomerular basement membrane antibodies to induce GN. We confirmed previous reports that demonstrated that SMAD3 is an important mediator of glomerulosclerosis and renal interstitial fibrosis. Proteinuria was highly SMAD3 dependent. We found differential effects of SMAD3 deletion on podocytes and glomerular endothelial cells. GN led to podocyte injury, including foot process effacement and loss of podocyte-specific markers. Interestingly, these changes were not SMAD3 dependent. Furthermore, there were significant changes to glomerular endothelial cells, including loss of fenestrations, swelling, and basement membrane reduplication, which were SMAD3 dependent. Despite ongoing markers of podocyte injury in SMAD3-/- mice, proteinuria was transient. Renal injury in the setting of GN involves TGF-ß and SMAD3 signaling. Cell populations within the glomerulus respond differently to SMAD3 deletion. Proteinuria correlated more with endothelial cell changes as opposed to podocyte injury in this model.


Subject(s)
Anti-Glomerular Basement Membrane Disease/metabolism , Kidney Glomerulus/metabolism , Smad3 Protein/metabolism , Animals , Anti-Glomerular Basement Membrane Disease/genetics , Anti-Glomerular Basement Membrane Disease/immunology , Anti-Glomerular Basement Membrane Disease/pathology , Autoantibodies , Cell Line , Disease Models, Animal , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Fibrosis , Kidney Glomerulus/immunology , Kidney Glomerulus/pathology , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Paracrine Communication , Podocytes/immunology , Podocytes/metabolism , Podocytes/pathology , Proteinuria/immunology , Proteinuria/metabolism , Signal Transduction , Smad3 Protein/deficiency , Smad3 Protein/genetics , Transforming Growth Factor beta/metabolism
6.
Immunol Cell Biol ; 97(2): 203-217, 2019 02.
Article in English | MEDLINE | ID: mdl-30298952

ABSTRACT

Although recent evidence has shown that IL-6 is involved in enhanced alternative activation of macrophages toward a profibrotic phenotype, the mechanisms leading to their increased secretory capacity are not fully understood. Here, we investigated the effect of IL-6 on endoplasmic reticulum (ER) expansion and alternative activation of macrophages in vitro. An essential mediator in this ER expansion process is the IRE1 pathway, which possesses a kinase and endoribonuclease domain to cleave XBP1 into a spliced bioactive molecule. To investigate the IRE1-XBP1 expansion pathway, IL-4/IL-13 and IL-4/IL-13/IL-6-mediated alternative programming of murine bone marrow-derived and human THP1 macrophages were assessed by arginase activity in cell lysates, CD206 and arginase-1 expression by flow cytometry, and secreted CCL18 by ELISA, respectively. Ultrastructural intracellular morphology and ER biogenesis were examined by transmission electron microscopy and immunofluorescence. Transcription profiling of 128 genes were assessed by NanoString and Pharmacological inhibition of the IRE1-XBP1 arm was achieved using STF-083010 and was verified by RT-PCR. The addition of IL-6 to the conventional alternative programming cocktail IL-4/IL-13 resulted in increased ER and mitochondrial expansion, profibrotic profiles and unfolded protein response-mediated induction of molecular chaperones. IRE1-XBP1 inhibition substantially reduced the IL-6-mediated hyperpolarization and normalized the above effects. In conclusion, the addition of IL-6 enhances ER expansion and the profibrotic capacity of IL-4/IL-13-mediated activation of macrophages. Therapeutic strategies targeting IL-6 or the IRE1-XBP1 axis may be beneficial to prevent the profibrotic capacity of macrophages.


Subject(s)
Endoplasmic Reticulum , Endoribonucleases/metabolism , Interleukin-3/metabolism , Interleukin-4/metabolism , Interleukin-6/metabolism , Macrophage-Activating Factors/metabolism , Macrophages/immunology , Macrophages/ultrastructure , Protein Serine-Threonine Kinases/metabolism , Animals , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/physiology , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/immunology , Humans , Interleukin-4/pharmacology , Interleukin-6/pharmacology , Macrophage Activation , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , THP-1 Cells
7.
Am J Physiol Renal Physiol ; 314(6): F1036-F1045, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29363326

ABSTRACT

The wingless-type mouse mammary tumor virus integration site family (WNT) signaling pathway is involved in wound healing and fibrosis. We evaluated the WNT signaling pathway in peritoneal membrane injury. We assessed WNT1 protein expression in the peritoneal effluents of 54 stable peritoneal dialysis (PD) patients and WNT-related gene expression in ex vivo mesothelial cell cultures from 21 PD patients. In a transforming growth factor-ß (TGF-ß)-mediated animal model of peritoneal fibrosis, we evaluated regulation of the WNT pathway and the effect of WNT inhibition on peritoneal fibrosis and angiogenesis. WNT1 and WNT2 gene expression were positively correlated with peritoneal membrane solute transport in PD patients. In the mouse peritoneum, TGF-ß-induced peritoneal fibrosis was associated with increased expression of WNT2 and WNT4. Peritoneal ß-catenin protein was significantly upregulated after infection with adenovirus expressing TGF-ß (AdTGF-ß) along with elements of the WNT signaling pathway. Treatment with a ß-catenin inhibitor (ICG-001) in mice with AdTGF-ß-induced peritoneal fibrosis resulted in attenuation of peritoneal angiogenesis and reduced vascular endothelial growth factor. Similar results were also observed with the WNT antagonist Dickkopf-related protein (DKK)-1. In addition to this, DKK-1 blocked epithelial-mesenchymal transition and increased levels of the cell adhesion protein E-cadherin. We provide evidence that WNT signaling is active in the setting of experimental peritoneal fibrosis and WNT1 correlates with patient peritoneal membrane solute transport in PD patients. Intervention in this pathway is a possible therapy for peritoneal membrane injury.


Subject(s)
Epithelial Cells/metabolism , Neovascularization, Pathologic , Peritoneal Fibrosis/metabolism , Peritoneum/blood supply , Peritoneum/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , Aged , Animals , Cells, Cultured , Disease Models, Animal , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition , Female , Humans , Male , Mice, Inbred C57BL , Middle Aged , Peritoneal Dialysis/adverse effects , Peritoneal Fibrosis/etiology , Peritoneal Fibrosis/genetics , Peritoneal Fibrosis/pathology , Peritoneum/pathology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Wnt Proteins/genetics , Wnt1 Protein/genetics , Wnt1 Protein/metabolism , Wnt4 Protein/genetics , Wnt4 Protein/metabolism , beta Catenin/metabolism
8.
Nephrol Dial Transplant ; 32(1): 50-61, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27190383

ABSTRACT

Background: For patients using peritoneal dialysis (PD), the peritoneal membrane can develop fibrosis and angiogenesis, leading to ultrafiltration failure, chronic hypervolemia and increased risk of technique failure and mortality. Matrix metalloproteinases (MMPs), and specifically the gelatinases (MMP2 and MMP9), may be involved in peritoneal membrane injury. Methods: From stable PD patients, mesothelial cells were assayed for MMP gene expression. MMP9 was overexpressed in mouse peritoneum by adenovirus, and MMP9 -/- mice were subjected to transforming growth factor ß (TGF-ß)-induced peritoneal fibrosis. Results: MMP9 mRNA expression correlated with peritoneal membrane solute transport properties. Overexpression of MMP9 in the mouse peritoneum induced submesothelial thickening and angiogenesis. MMP9 induced mesothelial cell transition to a myofibroblast phenotype measured by increased alpha smooth muscle actin and decreased E-cadherin expression. Angiogenesis was markedly reduced in MMP9 -/- mice treated with an adenovirus expressing active TGF-ß compared with wild-type mice. TGF-ß-mediated E-cadherin cleavage was MMP9 dependent, and E-cadherin cleavage led to ß-catenin-mediated signaling. A ß-catenin inhibitor blocked the angiogenic response induced by AdMMP9. Conclusions: Our data suggest that MMP9 is involved in peritoneal membrane injury possibly through cleavage of E-cadherin and induction of ß-catenin signaling. MMP9 is a potential biomarker for peritoneal membrane injury and is a therapeutic target to protect the peritoneal membrane in PD patients.


Subject(s)
Cadherins/metabolism , Hemodialysis Solutions/metabolism , Matrix Metalloproteinase 9/metabolism , Neovascularization, Pathologic/etiology , Peritoneal Dialysis/adverse effects , Peritoneal Fibrosis/etiology , beta Catenin/metabolism , Animals , Biological Transport , Cadherins/genetics , Humans , Matrix Metalloproteinase 9/genetics , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Peritoneal Fibrosis/metabolism , Peritoneal Fibrosis/pathology , Signal Transduction/drug effects , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , beta Catenin/genetics
9.
Kidney Res Clin Pract ; 35(3): 133-41, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27668155

ABSTRACT

Peritoneal fibrosis is one of the major complications occurring in long-term peritoneal dialysis patients as a result of injury. Peritoneal fibrosis is characterized by submesothelial thickening and fibrosis which is associated with a decline in peritoneal membrane function. The myofibroblast has been identified as the key player involved in the development and progression of peritoneal fibrosis. Activation of the myofibroblast is correlated with expansion of the extracellular matrix and changes in peritoneal membrane integrity. Over the years, epithelial to mesenchymal transition (EMT) has been accepted as the predominant source of the myofibroblast. Peritoneal mesothelial cells have been described to undergo EMT in response to injury. Several animal and in vitro studies support the role of EMT in peritoneal fibrosis; however, emerging evidence from genetic fate-mapping studies has demonstrated that myofibroblasts may be arising from resident fibroblasts and pericytes/perivascular fibroblasts. In this review, we will discuss hypotheses currently surrounding the origin of the myofibroblast and highlight the experimental systems predominantly being used to investigate this.

SELECTION OF CITATIONS
SEARCH DETAIL
...