Glycogen Synthase Kinase-3β Inhibition with 9-ING-41 Attenuates the Progression of Pulmonary Fibrosis

Ann Jeffers, Wenyi Qin, Shuzi Owens, Kathleen B Koenig, Satoshi Komatsu, Francis J Giles, Daniel M Schmitt, Steven Idell, Torry A Tucker, Ann Jeffers, Wenyi Qin, Shuzi Owens, Kathleen B Koenig, Satoshi Komatsu, Francis J Giles, Daniel M Schmitt, Steven Idell, Torry A Tucker

Abstract

Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial lung disease with a median survival of 3 years after diagnosis. Although the etiology of IPF is unknown, it is characterized by extensive alveolar epithelial cell apoptosis and proliferation of myofibroblasts in the lungs. While the origins of these myofibroblast appear to be diverse, fibroblast differentiation contributes to expansion of myofibroblasts and to disease progression. We found that agents that contribute to neomatrix formation and remodeling in pulmonary fibrosis (PF); TGF-β, Factor Xa, thrombin, plasmin and uPA all induced fibroblast/myofibroblast differentiation. These same mediators enhanced GSK-3β activation via phosphorylation of tyrosine-216 (p-Y216). Inhibition of GSK-3β signaling with the novel inhibitor 9-ING-41 blocked the induction of myofibroblast markers; α-SMA and Col-1 and reduced morphological changes of myofibroblast differentiation. In in vivo studies, the progression of TGF-β and bleomycin mediated PF was significantly attenuated by 9-ING-41 administered at 7 and 14 days respectively after the establishment of injury. Specifically, 9-ING-41 treatment significantly improved lung function (compliance and lung volumes; p < 0.05) of TGF-β adenovirus treated mice compared to controls. Similar results were found in mice with bleomycin-induced PF. These studies clearly show that activation of the GSK-3β signaling pathway is critical for the induction of myofibroblast differentiation in lung fibroblasts ex vivo and pulmonary fibrosis in vivo. The results offer a strong premise supporting the continued investigation of the GSK-3β signaling pathway in the control of fibroblast-myofibroblast differentiation and fibrosing lung injury. These data provide a strong rationale for extension of clinical trials of 9-ING-41 to patients with IPF.

Conflict of interest statement

D.M. Schmitt and F.J. Giles are employees of Actuate Therapeutics Inc. and have equity interest in Actuate Therapeutics Inc. No potential conflicts of interest were disclosed by the other authors.

Figures

Figure 1
Figure 1
Lung tissue sections from TGF-β and bleomycin injured mice were stained for GSK-3β (red) and nuclei (blue) and imaged by confocal microscopy. GSK-3β expression was increased in TGF-β (A) and bleomycin-injured (B) mice compared to controls. Images are representative of 30 fields/slide and n = 4–6 samples/condition. Images were taken at 25X optical zoom. Bar indicates 100 µm.
Figure 2
Figure 2
Mediators implicated in pulmonary organization induce myofibroblast differentiation of normal and IPF fibroblasts. Serum starved human fibroblasts were treated with various mediators to induce myofibroblast differentiation (TGF-β, FXa, thrombin (THB), plasmin (PLN) and uPA; see Materials and Methods). Cell lysates and conditioned medias, collected after 48 h, were then resolved by SDS-PAGE and western blotted for α-SMA, total GSK-3β, tyrosine 216 phosphorylated GSK-3β (pTyr-GSK-3β) and collagen 1 (Col-1), in NF (A) and IPF cells (C). β-actin was the loading control. α-SMA and collagen 1 expression were then quantified by densitometric analyses. Plotted data are the mean ± SEM of n = 3 independent experiments. Collagen was most prominently induced by TGF-β and FXa. Images are representative of three independent experiments. NF (B) and IPF (D) cells were treated PBS, TGF-β, Xa, thrombin, plasmin and uPA for 24 h incubation. RNA was then collected, and qPCR analyses were then performed for α-SMA and collagen 1 expression. GAPDH was the loading control. Plotted data are the mean ± SEM of n = 3–4 independent experiments.
Figure 3
Figure 3
IPF fibroblasts demonstrate increased GSK-3β nuclear localization. Normal and IPF fibroblasts were seeded on glass coverslips. Serum-starved cells were then treated with TGF-β for 48 h. Cells were then fixed, permeabilized and immunostained for GSK-3β. GSK-3β (green) and nuclei (red) were then visualized by confocal microscopy. Images are representative of 10 fields/slide and n = 3 samples/condition. Images were taken at 40X optical zoom. The Mean Fluorescence Intensity (MFI) of nuclear GSK-3β was graphed as mean ± SEM. *Denotes p 

Figure 4

9-ING-41 blocks TGF-β mediated fibroblast…

Figure 4

9-ING-41 blocks TGF-β mediated fibroblast to myofibroblast transition. Fibroblasts (normal, A , B…

Figure 4
9-ING-41 blocks TGF-β mediated fibroblast to myofibroblast transition. Fibroblasts (normal, A,B and IPF, C,D) were treated with various doses of 9-ING-41 (10-0.5 µM) in serum free media. Cells were then treated with TGF-β for 48 h. Conditioned medias and cell lysates were then resolved by SDS-PAGE and immunoblotted for collagen (Col-1), α-SMA, GSK-3β, and tyrosine-216 phosphorylated GSK-3β. Under control conditions, TGF-β induced Col-1 and α-SMA protein in NF and IPF fibroblasts (A and C). 9-ING-41 (10 and 5 µM) significantly blocked TGF-β mediated induction of α-SMA and Col-1 in NF and IPF cells. Tyr-216 phosphorylation of GSK-3β (pGSKt) was likewise reduced by pretreatment with 9-ING-41 in both NF and IPF fibroblasts. Graphed data are the means of n = 3 independent experiments. Images are representative of 3–4 independent experiments. Total RNA was isolated from TGF-β treated cells in the presence or absence varying doses 9-ING-41 (10–0.5 µM). Changes in α-SMA and collagen 1 expression were then determined by qPCR analyses (B and D). GAPDH was used as the reference gene. Data are expressed as mean ± SEM. n = 3 independent experiments. *Denotes p < 0.05 compared to TGF-β control. Normal (E) and IPF (F) fibroblasts were treated with varying doses of TDZD-8 (40-5 µM) prior to the addition of TGF-β. Cell lysates and conditioned media were then resolved via SDS-PAGE and probed for changes α-SMA and collagen. β-actin was the loading control. TDZD-8 modestly reduced α-SMA and collagen induction by TGF-β at the highest dose (40 µM). Images are representative of two independent experiments.

Figure 5

9-ING-41 reverses established fibroblast to…

Figure 5

9-ING-41 reverses established fibroblast to myofibroblast transition. Serum-starved NF and IPF cells were…

Figure 5
9-ING-41 reverses established fibroblast to myofibroblast transition. Serum-starved NF and IPF cells were treated with TGF-β for 24 hours. Varying doses of 9-ING-41 (10-5 µM) were then added to the TGF-β–treated cells and allowed to incubate for 48 hours. Conditioned media and lysates from NF (A) and IPF (C) were resolved by SDS-PAGE and immunoblotted for collagen (Col)-1 and α-smooth muscle actin (α-SMA). Lysates were also immunoblotted for GSK-3β phosphorylation at tyrosine 216. β-Actin was used as loading control. For quantitative PCR (qPCR) analyses varying doses of 9-ING-41 (10 to 0.5 µM) were added to TGF-β–treated normal (B) and IPF (D) cells and then allowed to incubate for 24 hours. Total RNA was then isolated and transcribed into cDNA. α-SMA expression was determined by qPCR analyses. GAPDH served as the reference gene. Data are expressed as means ± SEM. n = 3 independent experiments. ∗p < 0.05 versus TGF-β treatment.

Figure 6

9-ING-41-treated mice demonstrate reduced TGF-β…

Figure 6

9-ING-41-treated mice demonstrate reduced TGF-β mediated PF. Mice were intratracheally administered TGF-β adenovirus…

Figure 6
9-ING-41-treated mice demonstrate reduced TGF-β mediated PF. Mice were intratracheally administered TGF-β adenovirus to induce pulmonary fibrosis. After 7d, mice received daily intraperitoneal injections of 9-ING-41 (30 mg/kg) for the next 7d. At the completion of the 14d time course, lung compliance and volumes were determined. A, Significant decrements in lung compliance and volume were reversed by 9-ING-41 treatment. Data are expressed as a mean ± SEM. n = 6 mice/condition. *Indicates a p $Denotes p < 0.05 compared to GFP adenovirus 9-ING-41 treatment. B. Lung tissue sections from DMSO and 9-ING-41 treated mice were Trichrome stained to show areas of injury and collagen (blue). Images were taken at 20X optical zoom. Images are representative of 30 fields/slide/condition. n = 6–7 mice. Lung injury score data are expressed as means ± SEM. (C) Lung tissue sections from TGF-β adenoviral mice treated with vehicle and 9-ING-41 mice were immunostained to visualize collagen (Col-1) deposition (red) and nuclei (blue) by confocal microscopy. Images were taken at 25x optical zoom. Collagen injury score data are expressed as mean ± SEM. n = 6 mice/condition. Bar indicates 100 µm. (D) Lung tissue sections from GFP and TGF-β adenovirus infected mice were stained for apoptosis by fluorescent TUNEL stain (green) and nuclei (blue). Images were taken at 20X optical zoom. Images are representative of 15 fields/slide/condition and n = 5–6 samples/condition. Data are expressed as mean ± SEM. p denotes a p < 0.05. Bar indicates 50 µm.

Figure 7

9-ING-41-treated mice demonstrate reduced bleomycin…

Figure 7

9-ING-41-treated mice demonstrate reduced bleomycin mediated PF. ( A ) Anesthetized C57Bl/6 J…

Figure 7
9-ING-41-treated mice demonstrate reduced bleomycin mediated PF. (A) Anesthetized C57Bl/6 J mice were intratracheally administered 0.8U bleomycin/kg. After 14d mice were administered vehicle control (DMSO) or 9-ING-41 (30 mg/kg) by daily intraperitoneal injection for the next 14d. At the completion of the 28d time-course lung compliance and volumes were determined with the Scireq flexivent and by chest CT imaging, respectively. 9-ING-41 treated mice demonstrated significantly better lung compliance and lung volumes than DMSO-treated controls. *Indicates a p < 0.05 compared to saline control. $Denotes p < 0.05 compared to bleomycin/saline treated mice. #Denotes a p < 0.05 compared to bleomycin/DMSO treated mice. (B) Lung tissue sections from DMSO and 9-ING-41 treated mice were Trichrome stained to show areas of injury and collagen (blue). Images were taken at 20X optical zoom. Images are representative of 30 fields/slide/condition and n = 6 mice. Lung injury score data are expressed as means ± SEM. (C) Lung tissue sections from vehicle and 9-ING-41 treated mice were immunostained to visualize collagen deposition (red) by confocal microscopy. Images were taken at 10x optical zoom. Bar indicates 100 µM. (D) Lung tissue sections from saline, bleomycin and bleomycin/9-ING-41 treated mice were stained for apoptosis by fluorescent TUNEL stain (green). Images were taken at 20X optical zoom. Images are representative of 15 fields/slide/condition. Data are expressed as mean ± SEM. p denotes a p < 0.05. n = 5–6 samples/condition. Bar indicates 50 µm.
All figures (7)
Figure 4
Figure 4
9-ING-41 blocks TGF-β mediated fibroblast to myofibroblast transition. Fibroblasts (normal, A,B and IPF, C,D) were treated with various doses of 9-ING-41 (10-0.5 µM) in serum free media. Cells were then treated with TGF-β for 48 h. Conditioned medias and cell lysates were then resolved by SDS-PAGE and immunoblotted for collagen (Col-1), α-SMA, GSK-3β, and tyrosine-216 phosphorylated GSK-3β. Under control conditions, TGF-β induced Col-1 and α-SMA protein in NF and IPF fibroblasts (A and C). 9-ING-41 (10 and 5 µM) significantly blocked TGF-β mediated induction of α-SMA and Col-1 in NF and IPF cells. Tyr-216 phosphorylation of GSK-3β (pGSKt) was likewise reduced by pretreatment with 9-ING-41 in both NF and IPF fibroblasts. Graphed data are the means of n = 3 independent experiments. Images are representative of 3–4 independent experiments. Total RNA was isolated from TGF-β treated cells in the presence or absence varying doses 9-ING-41 (10–0.5 µM). Changes in α-SMA and collagen 1 expression were then determined by qPCR analyses (B and D). GAPDH was used as the reference gene. Data are expressed as mean ± SEM. n = 3 independent experiments. *Denotes p < 0.05 compared to TGF-β control. Normal (E) and IPF (F) fibroblasts were treated with varying doses of TDZD-8 (40-5 µM) prior to the addition of TGF-β. Cell lysates and conditioned media were then resolved via SDS-PAGE and probed for changes α-SMA and collagen. β-actin was the loading control. TDZD-8 modestly reduced α-SMA and collagen induction by TGF-β at the highest dose (40 µM). Images are representative of two independent experiments.
Figure 5
Figure 5
9-ING-41 reverses established fibroblast to myofibroblast transition. Serum-starved NF and IPF cells were treated with TGF-β for 24 hours. Varying doses of 9-ING-41 (10-5 µM) were then added to the TGF-β–treated cells and allowed to incubate for 48 hours. Conditioned media and lysates from NF (A) and IPF (C) were resolved by SDS-PAGE and immunoblotted for collagen (Col)-1 and α-smooth muscle actin (α-SMA). Lysates were also immunoblotted for GSK-3β phosphorylation at tyrosine 216. β-Actin was used as loading control. For quantitative PCR (qPCR) analyses varying doses of 9-ING-41 (10 to 0.5 µM) were added to TGF-β–treated normal (B) and IPF (D) cells and then allowed to incubate for 24 hours. Total RNA was then isolated and transcribed into cDNA. α-SMA expression was determined by qPCR analyses. GAPDH served as the reference gene. Data are expressed as means ± SEM. n = 3 independent experiments. ∗p < 0.05 versus TGF-β treatment.
Figure 6
Figure 6
9-ING-41-treated mice demonstrate reduced TGF-β mediated PF. Mice were intratracheally administered TGF-β adenovirus to induce pulmonary fibrosis. After 7d, mice received daily intraperitoneal injections of 9-ING-41 (30 mg/kg) for the next 7d. At the completion of the 14d time course, lung compliance and volumes were determined. A, Significant decrements in lung compliance and volume were reversed by 9-ING-41 treatment. Data are expressed as a mean ± SEM. n = 6 mice/condition. *Indicates a p $Denotes p < 0.05 compared to GFP adenovirus 9-ING-41 treatment. B. Lung tissue sections from DMSO and 9-ING-41 treated mice were Trichrome stained to show areas of injury and collagen (blue). Images were taken at 20X optical zoom. Images are representative of 30 fields/slide/condition. n = 6–7 mice. Lung injury score data are expressed as means ± SEM. (C) Lung tissue sections from TGF-β adenoviral mice treated with vehicle and 9-ING-41 mice were immunostained to visualize collagen (Col-1) deposition (red) and nuclei (blue) by confocal microscopy. Images were taken at 25x optical zoom. Collagen injury score data are expressed as mean ± SEM. n = 6 mice/condition. Bar indicates 100 µm. (D) Lung tissue sections from GFP and TGF-β adenovirus infected mice were stained for apoptosis by fluorescent TUNEL stain (green) and nuclei (blue). Images were taken at 20X optical zoom. Images are representative of 15 fields/slide/condition and n = 5–6 samples/condition. Data are expressed as mean ± SEM. p denotes a p < 0.05. Bar indicates 50 µm.
Figure 7
Figure 7
9-ING-41-treated mice demonstrate reduced bleomycin mediated PF. (A) Anesthetized C57Bl/6 J mice were intratracheally administered 0.8U bleomycin/kg. After 14d mice were administered vehicle control (DMSO) or 9-ING-41 (30 mg/kg) by daily intraperitoneal injection for the next 14d. At the completion of the 28d time-course lung compliance and volumes were determined with the Scireq flexivent and by chest CT imaging, respectively. 9-ING-41 treated mice demonstrated significantly better lung compliance and lung volumes than DMSO-treated controls. *Indicates a p < 0.05 compared to saline control. $Denotes p < 0.05 compared to bleomycin/saline treated mice. #Denotes a p < 0.05 compared to bleomycin/DMSO treated mice. (B) Lung tissue sections from DMSO and 9-ING-41 treated mice were Trichrome stained to show areas of injury and collagen (blue). Images were taken at 20X optical zoom. Images are representative of 30 fields/slide/condition and n = 6 mice. Lung injury score data are expressed as means ± SEM. (C) Lung tissue sections from vehicle and 9-ING-41 treated mice were immunostained to visualize collagen deposition (red) by confocal microscopy. Images were taken at 10x optical zoom. Bar indicates 100 µM. (D) Lung tissue sections from saline, bleomycin and bleomycin/9-ING-41 treated mice were stained for apoptosis by fluorescent TUNEL stain (green). Images were taken at 20X optical zoom. Images are representative of 15 fields/slide/condition. Data are expressed as mean ± SEM. p denotes a p < 0.05. n = 5–6 samples/condition. Bar indicates 50 µm.

References

    1. Ghatak S, et al. Transforming growth factor beta1 (TGFbeta1)-induced CD44V6-NOX4 signaling in pathogenesis of idiopathic pulmonary fibrosis. The Journal of biological chemistry. 2017;292:10490–10519. doi: 10.1074/jbc.M116.752469.
    1. Rangarajan S, et al. Novel Mechanisms for the Antifibrotic Action of Nintedanib. American journal of respiratory cell and molecular biology. 2016;54:51–59. doi: 10.1165/rcmb.2014-0445OC.
    1. Kandhare AD, Mukherjee A, Ghosh P, Bodhankar SL. Efficacy of antioxidant in idiopathic pulmonary fibrosis: A systematic review and meta-analysis. EXCLI J. 2016;15:636–651.
    1. Hecker L, et al. Reversal of persistent fibrosis in aging by targeting Nox4-Nrf2 redox imbalance. Science translational medicine. 2014;6:231ra247. doi: 10.1126/scitranslmed.3008182.
    1. Doble BW, Woodgett JR. Role of glycogen synthase kinase-3 in cell fate and epithelial-mesenchymal transitions. Cells, tissues, organs. 2007;185:73–84. doi: 10.1159/000101306.
    1. Doble BW, Woodgett JR. GSK-3: tricks of the trade for a multi-tasking kinase. Journal of cell science. 2003;116:1175–1186. doi: 10.1242/jcs.00384.
    1. Schwabe RF, Brenner DA. Role of glycogen synthase kinase-3 in TNF-alpha-induced NF-kappaB activation and apoptosis in hepatocytes. American journal of physiology. Gastrointestinal and liver physiology. 2002;283:G204–211. doi: 10.1152/ajpgi.00016.2002.
    1. Hoeflich KP, et al. Requirement for glycogen synthase kinase-3beta in cell survival and NF-kappaB activation. Nature. 2000;406:86–90. doi: 10.1038/35017574.
    1. Hughes K, Nikolakaki E, Plyte SE, Totty NF, Woodgett JR. Modulation of the glycogen synthase kinase-3 family by tyrosine phosphorylation. EMBO J. 1993;12:803–808. doi: 10.1002/j.1460-2075.1993.tb05715.x.
    1. Woodgett JR. Molecular cloning and expression of glycogen synthase kinase-3/factor A. The EMBO journal. 1990;9:2431–2438. doi: 10.1002/j.1460-2075.1990.tb07419.x.
    1. Bhat RV, et al. Regulation and localization of tyrosine216 phosphorylation of glycogen synthase kinase-3beta in cellular and animal models of neuronal degeneration. Proceedings of the National Academy of Sciences of the United States of America. 2000;97:11074–11079. doi: 10.1073/pnas.190297597.
    1. Tucker, T. et al. Myocardin is Involved in Mesothelial-Mesenchymal Transition of Human Pleural Mesothelial Cells. American journal of respiratory cell and molecular biology (2019).
    1. Baarsma HA, et al. Glycogen synthase kinase-3 (GSK-3) regulates TGF-beta(1)-induced differentiation of pulmonary fibroblasts. British journal of pharmacology. 2013;169:590–603. doi: 10.1111/bph.12098.
    1. Shin HS, et al. Metformin ameliorates the Phenotype Transition of Peritoneal Mesothelial Cells and Peritoneal Fibrosis via a modulation of Oxidative Stress. Scientific reports. 2017;7:5690. doi: 10.1038/s41598-017-05836-6.
    1. Xia H, et al. Low alpha(2)beta(1) integrin function enhances the proliferation of fibroblasts from patients with idiopathic pulmonary fibrosis by activation of the beta-catenin pathway. The American journal of pathology. 2012;181:222–233. doi: 10.1016/j.ajpath.2012.03.034.
    1. Caraci F, et al. TGF-beta1 targets the GSK-3beta/beta-catenin pathway via ERK activation in the transition of human lung fibroblasts into myofibroblasts. Pharmacological research. 2008;57:274–282. doi: 10.1016/j.phrs.2008.02.001.
    1. Frame S, Cohen P, Biondi RM. A common phosphate binding site explains the unique substrate specificity of GSK3 and its inactivation by phosphorylation. Molecular cell. 2001;7:1321–1327. doi: 10.1016/S1097-2765(01)00253-2.
    1. Karmali R, et al. GSK-3beta inhibitor, 9-ING-41, reduces cell viability and halts proliferation of B-cell lymphoma cell lines as a single agent and in combination with novel agents. Oncotarget. 2017;8:114924–114934. doi: 10.18632/oncotarget.22414.
    1. Ugolkov A, et al. Combination Treatment with the GSK-3 Inhibitor 9-ING-41 and CCNU Cures Orthotopic Chemoresistant Glioblastoma in Patient-Derived Xenograft Models. Transl Oncol. 2017;10:669–678. doi: 10.1016/j.tranon.2017.06.003.
    1. Ugolkov AV, et al. 9-ING-41, a small-molecule glycogen synthase kinase-3 inhibitor, is active in neuroblastoma. Anti-cancer drugs. 2018;29:717–724.
    1. Wu X, et al. Targeting glycogen synthase kinase 3 for therapeutic benefit in lymphoma. Blood. 2019;134:363–373. doi: 10.1182/blood.2018874560.
    1. Sahin I, et al. Glycogen synthase kinase-3 beta inhibitors as novel cancer treatments and modulators of antitumor immune responses. Cancer Biol Ther. 2019;20:1047–1056. doi: 10.1080/15384047.2019.1595283.
    1. Boren J, et al. Inhibition of Glycogen Synthase Kinase 3beta Blocks Mesomesenchymal Transition and Attenuates Streptococcus pneumonia-Mediated Pleural Injury in Mice. The American journal of pathology. 2017;187:2461–2472. doi: 10.1016/j.ajpath.2017.07.007.
    1. Bergmann C, et al. Inhibition of glycogen synthase kinase 3beta induces dermal fibrosis by activation of the canonical Wnt pathway. Ann Rheum Dis. 2011;70:2191–2198. doi: 10.1136/ard.2010.147140.
    1. Lan A, Qi Y, Du J. Akt2 mediates TGF-beta1-induced epithelial to mesenchymal transition by deactivating GSK3beta/snail signaling pathway in renal tubular epithelial cells. Cellular physiology and biochemistry: international journal of experimental cellular physiology, biochemistry, and pharmacology. 2014;34:368–382. doi: 10.1159/000363006.
    1. Liu H, et al. SB216763, a selective small molecule inhibitor of glycogen synthase kinase-3, improves bleomycin-induced pulmonary fibrosis via activating autophagy. Acta Pharmaceutica Sinica B. 2013;3:226–233. doi: 10.1016/j.apsb.2013.05.004.
    1. Zhang X, Liu Y, Shao R, Li W. Cdc42-interacting protein 4 silencing relieves pulmonary fibrosis in STZ-induced diabetic mice via the Wnt/GSK-3beta/beta-catenin pathway. Exp Cell Res. 2017;359:284–290. doi: 10.1016/j.yexcr.2017.07.018.
    1. Hummler SC, et al. Targeting glycogen synthase kinase-3beta to prevent hyperoxia-induced lung injury in neonatal rats. American journal of respiratory cell and molecular biology. 2013;48:578–588. doi: 10.1165/rcmb.2012-0383OC.
    1. Sisson TH, et al. Inhibition of myocardin-related transcription factor/serum response factor signaling decreases lung fibrosis and promotes mesenchymal cell apoptosis. The American journal of pathology. 2015;185:969–986. doi: 10.1016/j.ajpath.2014.12.005.
    1. Kamata H, et al. KIF5A transports collagen vesicles of myofibroblasts during pleural fibrosis. Scientific reports. 2017;7:4556. doi: 10.1038/s41598-017-04437-7.
    1. Tucker TA, et al. Organizing empyema induced in mice by Streptococcus pneumoniae: effects of plasminogen activator inhibitor-1 deficiency. Clinical and translational medicine. 2016;5:17. doi: 10.1186/s40169-016-0097-2.
    1. Mackinnon AC, et al. Regulation of transforming growth factor-beta1-driven lung fibrosis by galectin-3. American journal of respiratory and critical care medicine. 2012;185:537–546. doi: 10.1164/rccm.201106-0965OC.
    1. Williams L, et al. Tissue factor pathway inhibitor attenuates the progression of malignant pleural mesothelioma in nude mice. American journal of respiratory cell and molecular biology. 2012;46:173–179. doi: 10.1165/rcmb.2011-0276OC.
    1. Tucker TA, et al. Plasminogen activator inhibitor-1 deficiency augments visceral mesothelial organization, intrapleural coagulation, and lung restriction in mice with carbon black/bleomycin-induced pleural injury. American journal of respiratory cell and molecular biology. 2014;50:316–327.
    1. Tucker TA, et al. Lipoprotein receptor-related protein 1 regulates collagen 1 expression, proteolysis, and migration in human pleural mesothelial cells. American journal of respiratory cell and molecular biology. 2012;46:196–206. doi: 10.1165/rcmb.2011-0071OC.
    1. Idell S, Zwieb C, Kumar A, Koenig KB, Johnson AR. Pathways of fibrin turnover of human pleural mesothelial cells in vitro. American journal of respiratory cell and molecular biology. 1992;7:414–426. doi: 10.1165/ajrcmb/7.4.414.
    1. Jeffers A, et al. Thrombin down-regulates tissue factor pathway inhibitor expression in a PI3K/nuclear factor-kappaB-dependent manner in human pleural mesothelial cells. American journal of respiratory cell and molecular biology. 2015;52:674–682. doi: 10.1165/rcmb.2014-0084OC.
    1. Shetty S, Padijnayayveetil J, Tucker T, Stankowska D, Idell S. The fibrinolytic system and the regulation of lung epithelial cell proteolysis, signaling, and cellular viability. American journal of physiology. Lung cellular and molecular physiology. 2008;295:L967–975. doi: 10.1152/ajplung.90349.2008.
    1. Owens S, et al. Mesomesenchymal transition of pleural mesothelial cells is PI3K and NF-kappaB dependent. American journal of physiology. Lung cellular and molecular physiology. 2015;308:L1265–1273. doi: 10.1152/ajplung.00396.2014.
    1. Lee JM, et al. Involvement of Alveolar Epithelial Cell Necroptosis in Idiopathic Pulmonary Fibrosis Pathogenesis. American journal of respiratory cell and molecular biology. 2018;59:215–224. doi: 10.1165/rcmb.2017-0034OC.
    1. Thannickal VJ, Horowitz JC. Evolving concepts of apoptosis in idiopathic pulmonary fibrosis. Proceedings of the American Thoracic Society. 2006;3:350–356. doi: 10.1513/pats.200601-001TK.
    1. Fan M, Liu F, Yang Y, Ye Y, Huang G. Phosphorylation of glycogen synthase kinase-3beta induces epithelial mesenchymal transition in human peritoneal mesothelial cells. Zhong nan da xue xue bao. Yi xue ban = Journal of Central South University. Medical sciences. 2010;35:329–334.
    1. Jang YH, et al. Effects of dexamethasone on the TGF-beta1-induced epithelial-to-mesenchymal transition in human peritoneal mesothelial cells. Laboratory investigation; a journal of technical methods and pathology. 2013;93:194–206. doi: 10.1038/labinvest.2012.166.
    1. Luo L, et al. Small interfering RNA targeting ILK inhibits EMT in human peritoneal mesothelial cells through phosphorylation of GSK3beta. Molecular medicine reports. 2014;10:137–144. doi: 10.3892/mmr.2014.2162.
    1. Rusai K, Herzog R, Kuster L, Kratochwill K, Aufricht C. GSK-3beta inhibition protects mesothelial cells during experimental peritoneal dialysis through upregulation of the heat shock response. Cell stress & chaperones. 2013;18:569–579. doi: 10.1007/s12192-013-0410-6.
    1. Meares GP, Jope RS. Resolution of the nuclear localization mechanism of glycogen synthase kinase-3: functional effects in apoptosis. The Journal of biological chemistry. 2007;282:16989–17001. doi: 10.1074/jbc.M700610200.
    1. Bijur GN, Jope RS. Glycogen synthase kinase-3 beta is highly activated in nuclei and mitochondria. Neuroreport. 2003;14:2415–2419. doi: 10.1097/00001756-200312190-00025.
    1. Hilliard TS, et al. Glycogen synthase kinase 3beta inhibitors induce apoptosis in ovarian cancer cells and inhibit in-vivo tumor growth. Anti-cancer drugs. 2011;22:978–985.
    1. Fernandez IE, Eickelberg O. The impact of TGF-beta on lung fibrosis: from targeting to biomarkers. Proceedings of the American Thoracic Society. 2012;9:111–116. doi: 10.1513/pats.201203-023AW.
    1. Tatler AL, Jenkins G. TGF-beta activation and lung fibrosis. Proceedings of the American Thoracic Society. 2012;9:130–136. doi: 10.1513/pats.201201-003AW.
    1. Kalluri R, Neilson EG. Epithelial-mesenchymal transition and its implications for fibrosis. J Clin Invest. 2003;112:1776–1784. doi: 10.1172/JCI200320530.

Source: PubMed

3
Iratkozz fel