Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Apoptosis ; 24(5-6): 499-510, 2019 06.
Article in English | MEDLINE | ID: mdl-30850922

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a deadly and progressive fibrotic lung disease, but the precise etiology remains elusive. IPF is characterized by the presence of apoptosis-resistant (myo)fibroblasts that relentlessly produce a collagen-rich extracellular matrix (ECM). Recent studies showed that an anti-cancer chemotherapy drug cisplatin is implicated in the development of pulmonary fibrosis, suggesting that the treatment of cancer patients with cisplatin may alter fibroblast viability. To address this possibility, we investigated the cisplatin-induced cell death mechanism in lung fibroblasts derived from IPF and non-IPF patients in response to a collagen matrix. IPF fibroblasts showed enhanced resistance to cisplatin-induced cell death compared to non-IPF fibroblasts in a time- and dose-dependent manner. Molecular study showed that the expression of γH2AX, PUMA and caspase-3/7 activity was abnormally reduced in IPF fibroblasts, suggesting that DNA damage-induced apoptosis caused by cisplatin was suppressed in IPF fibroblasts. Our study further revealed that DNA repair protein XRCC1 activity was aberrantly increased as a result of CK2 hyper-activation in cisplatin-treated IPF fibroblasts, and this alteration protected IPF fibroblasts from cisplatin-induced cell death. Our results showed that IPF fibroblasts residing in a collagen rich matrix are resistance to cisplatin-induced cell death due to the aberrantly high CK2/XRCC1-dependent DNA repair activity. This finding suggests that pulmonary fibrosis may develop and worsen due to the presence of apoptosis-resistant lung fibroblasts in cisplatin-treated cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Casein Kinase II/metabolism , Cell Death/drug effects , Cisplatin/pharmacology , Fibroblasts/drug effects , Idiopathic Pulmonary Fibrosis/pathology , X-ray Repair Cross Complementing Protein 1/metabolism , Apoptosis/drug effects , Casein Kinase II/antagonists & inhibitors , Casein Kinase II/genetics , Cell Survival/drug effects , Cells, Cultured , DNA Damage , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression , Humans , RNA, Small Interfering , X-ray Repair Cross Complementing Protein 1/genetics
2.
Mol Carcinog ; 58(3): 321-333, 2019 03.
Article in English | MEDLINE | ID: mdl-30365189

ABSTRACT

Although members of the hyaluronan (HA)-CD44/HA-mediated motility receptor (RHAMM) signaling pathway have been shown to be overexpressed in lung cancer, their role in lung tumorigenesis is unclear. In the present study, we first determined levels of HA and its receptors CD44 and RHAMM in human non-small cell lung cancer (NSCLC) cells and stromal cells as well as mouse lung tumors. Subsequently, we examined the role of HA-CD44/RHAMM signaling pathway in mediating the proliferation and survival of NSCLC cells and the cross-talk between NSCLC cells and normal human lung fibroblasts (NHLFs)/lung cancer-associated fibroblasts (LCAFs). The highest levels of HA and CD44 were observed in NHLFs/LCAFs followed by NSCLC cells, whereas THP-1 monocytes/macrophages showed negligible levels of both HA and CD44. Simultaneous silencing of HA synthase 2 (HAS2) and HAS3 or CD44 and RHAMM suppressed cell proliferation and survival as well as the EGFR/AKT/ERK signaling pathway. Exogenous HA partially rescued the defect in cell proliferation and survival. Moreover, conditioned media (CM) generated by NHLFs/LCAFs enhanced the proliferation of NSCLC cells in a HA-dependent manner as treatment of NHLFs and LCAFs with HAS2 siRNA, 4-methylumbelliferone, an inhibitor of HASs, LY2228820, an inhibitor of p38MAPK, or treatment of A549 cells with CD44 blocking antibody suppressed the effects of the CM. Upon incubation in CM generated by A549 cells or THP-1 macrophages, NHLFs/LCAFs secreted higher concentrations of HA. Overall, our findings indicate that targeting the HA-CD44/RHAMM signaling pathway could be a promising approach for the prevention and therapy of lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation , Culture Media, Conditioned/pharmacology , Extracellular Matrix Proteins/metabolism , Fibroblasts/pathology , Hyaluronan Receptors/metabolism , Hyaluronic Acid/metabolism , Animals , Apoptosis , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice
3.
Int J Nanomedicine ; 12: 6089-6105, 2017.
Article in English | MEDLINE | ID: mdl-28860768

ABSTRACT

Recent progress in nanomedicine has shown a strong possibility of targeted therapy for obstinate chronic lung diseases including idiopathic pulmonary fibrosis (IPF). IPF is a fatal lung disease characterized by persistent fibrotic fibroblasts in response to type I collagen-rich extracellular matrix. As a pathological microenvironment is important in understanding the biological behavior of nanoparticles, in vitro cellular uptake of glycol chitosan nanoparticles (CNPs) in human lung fibroblasts was comparatively studied in the presence or absence of type I collagen matrix. Primary human lung fibroblasts from non-IPF and IPF patients (n=6/group) showed significantly increased cellular uptake of CNPs (>33.6-78.1 times) when they were cultured on collagen matrix. To elucidate the underlying mechanism of enhanced cellular delivery of CNPs in lung fibroblasts on collagen, cells were pretreated with chlorpromazine, genistein, and amiloride to inhibit clathrin-mediated endocytosis, caveolae-mediated endocytosis, and macropinocytosis, respectively. Amiloride pretreatment remarkably reduced the cellular uptake of CNPs, suggesting that lung fibroblasts mainly utilize the macropinocytosis-dependent mechanism when interacted with collagen. In addition, the internalization of CNPs was predominantly suppressed by a phosphoinositide 3-kinase (PI3K) inhibitor in IPF fibroblasts, indicating that enhanced PI3K activity associated with late-stage macropinocytosis can be particularly important for the enhanced cellular delivery of CNPs in IPF fibroblasts. Our study strongly supports the concept that a pathological microenvironment which surrounds lung fibroblasts has a significant impact on the intracellular delivery of nanoparticles. Based on the property of enhanced intracellular delivery of CNPs when fibroblasts are made to interact with a collagen-rich matrix, we suggest that CNPs may have great potential as a drug-carrier system for targeting fibrotic lung fibroblasts.


Subject(s)
Chitosan/administration & dosage , Fibroblasts/drug effects , Idiopathic Pulmonary Fibrosis/pathology , Lung/cytology , Nanoparticles/administration & dosage , Amiloride/pharmacology , Cells, Cultured , Chitosan/chemistry , Chitosan/pharmacokinetics , Chlorpromazine/pharmacology , Collagen Type I/metabolism , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Drug Delivery Systems/methods , Endocytosis/drug effects , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Genistein/pharmacology , Humans , Lung/metabolism , Nanoparticles/chemistry , Phosphatidylinositol 3-Kinases/metabolism
4.
J Clin Med ; 5(9)2016 Sep 20.
Article in English | MEDLINE | ID: mdl-27657144

ABSTRACT

Chronic lung diseases include a variety of obstinate and fatal diseases, including asthma, chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), idiopathic pulmonary fibrosis (IPF), and lung cancers. Pharmacotherapy is important for the treatment of chronic lung diseases, and current progress in nanoparticles offers great potential as an advanced strategy for drug delivery. Based on their biophysical properties, nanoparticles have shown improved pharmacokinetics of therapeutics and controlled drug delivery, gaining great attention. Herein, we will review the nanoparticle-based drug delivery system for the treatment of chronic lung diseases. Various types of nanoparticles will be introduced, and recent innovative efforts to utilize the nanoparticles as novel drug carriers for the effective treatment of chronic lung diseases will also be discussed.

5.
J Pathol ; 240(1): 25-37, 2016 09.
Article in English | MEDLINE | ID: mdl-27218286

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is an irreversible lethal lung disease with an unknown etiology. IPF patients' lung fibroblasts express inappropriately high Akt activity, protecting them in response to an apoptosis-inducing type I collagen matrix. FasL, a ligand for Fas, is known to be increased in the lung tissues of patients with IPF, implicated with the progression of IPF. Expression of Decoy Receptor3 (DcR3), which binds to FasL, thereby subsequently suppressing the FasL-Fas-dependent apoptotic pathway, is frequently altered in various human disease. However, the role of DcR3 in IPF fibroblasts in regulating their viability has not been examined. We found that enhanced DcR3 expression exists in the majority of IPF fibroblasts on collagen matrices, resulting in the protection of IPF fibroblasts from FasL-induced apoptosis. Abnormally high Akt activity suppresses GSK-3ß function, thereby accumulating the nuclear factor of activated T-cells cytoplasmic 1 (NFATc1) in the nucleus, increasing DcR3 expression in IPF fibroblasts. This alteration protects IPF cells from FasL-induced apoptosis on collagen. However, the inhibition of Akt or NFATc1 decreases DcR3 mRNA and protein levels, which sensitizes IPF fibroblasts to FasL-mediated apoptosis. Furthermore, enhanced DcR3 and NFATc1 expression is mainly present in myofibroblasts in the fibroblastic foci of lung tissues derived from IPF patients. Our results showed that when IPF cells interact with collagen matrix, aberrantly activated Akt increases DcR3 expression via GSK-3ß-NFATc1 and protects IPF cells from the FasL-dependent apoptotic pathway. These findings suggest that the inhibition of DcR3 function may be an effective approach for sensitizing IPF fibroblasts in response to FasL, limiting the progression of lung fibrosis. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Apoptosis/physiology , Fas Ligand Protein/metabolism , Fibroblasts/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , Receptors, Tumor Necrosis Factor, Member 6b/metabolism , Cell Nucleus/metabolism , Cell Proliferation , Collagen/metabolism , Fibroblasts/pathology , Humans , Idiopathic Pulmonary Fibrosis/pathology , Lung/pathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology
6.
Autophagy ; 12(3): 547-64, 2016.
Article in English | MEDLINE | ID: mdl-27046250

ABSTRACT

ULK1 (unc-51 like autophagy activating kinase 1), the key mediator of MTORC1 signaling to autophagy, regulates early stages of autophagosome formation in response to starvation or MTORC1 inhibition. How ULK1 regulates the autophagy induction process remains elusive. Here, we identify that ATG13, a binding partner of ULK1, mediates interaction of ULK1 with the ATG14-containing PIK3C3/VPS34 complex, the key machinery for initiation of autophagosome formation. The interaction enables ULK1 to phosphorylate ATG14 in a manner dependent upon autophagy inducing conditions, such as nutrient starvation or MTORC1 inhibition. The ATG14 phosphorylation mimics nutrient deprivation through stimulating the kinase activity of the class III phosphatidylinositol 3-kinase (PtdIns3K) complex and facilitates phagophore and autophagosome formation. By monitoring the ATG14 phosphorylation, we determined that the ULK1 activity requires BECN1/Beclin 1 but not the phosphatidylethanolamine (PE)-conjugation machinery and the PIK3C3 kinase activity. Monitoring the phosphorylation also allowed us to identify that ATG9A is required to suppress the ULK1 activity under nutrient-enriched conditions. Furthermore, we determined that ATG14 phosphorylation depends on ULK1 and dietary conditions in vivo. These results define a key molecular event for the starvation-induced activation of the ATG14-containing PtdIns3K complex by ULK1, and demonstrate hierarchical relations between the ULK1 activation and other autophagy proteins involved in phagophore formation.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Proteins/metabolism , Autophagy , Intracellular Signaling Peptides and Proteins/metabolism , Multiprotein Complexes/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Vesicular Transport Proteins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Vesicular Transport/chemistry , Amino Acid Sequence , Animals , Autophagosomes/metabolism , Autophagy-Related Proteins/chemistry , Cell Line , Class I Phosphatidylinositol 3-Kinases/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Phosphatidylethanolamines/metabolism , Phosphorylation , Phosphoserine/metabolism , Protein Binding , Up-Regulation , Vesicular Transport Proteins/chemistry
7.
Drug Dev Res ; 76(7): 343-53, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26303294

ABSTRACT

Preclinical Research Idiopathic Pulmonary Fibrosis (IPF) is the most severe fibrotic lung disease and characterized by the accumulation of (myo)fibroblasts and collagen within the alveolar wall resulting in obliteration of the gas-exchange surface. Although the detailed pathogenesis is not understood, recent studies have found that several microRNAs (miRNAs) are associated with the progression of lung diseases including IPF. IPF is a fibrotic disease and, most frequently found in an aged population. In this review, the functional roles of miRNAs that are deregulated in IPF progression are discussed together with how aging affects the miRNA signature, altering the fibroblast phenotype and promoting lung fibrosis. Finally, the possibility of targeting miRNAs as a therapeutic approach for the treatment of IPF is discussed.


Subject(s)
Aging/genetics , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/genetics , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Molecular Targeted Therapy/methods , Humans , Idiopathic Pulmonary Fibrosis/pathology , Models, Genetic
8.
Am J Physiol Lung Cell Mol Physiol ; 309(6): L552-61, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26186945

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic and fatal lung disease, and fibroblasts derived from patients with IPF are resistant to type I collagen matrix-induced cell death. The alteration of the PTEN-Akt axis permits IPF fibroblasts to maintain a pathological phenotype on collagen by suppressing autophagy. However, the precise underlying mechanism by which the Akt downstream molecule suppresses autophagic activity remains elusive. FoxO3a is a direct target of Akt and is implicated with the transcriptional activation of autophagy. Therefore, we investigated whether reduced FoxO3a expression causes abnormally low autophagy in IPF fibroblasts on collagen. We found that FoxO3a mRNA and protein levels are low in IPF fibroblasts, which subsequently suppresses the autophagosomal marker LC3B expression on collagen matrix. In contrast, the majority of control fibroblasts showed an increase in FoxO3a and LC3B expression at both the mRNA and protein levels. The luciferase assay confirmed that FoxO3a binds to the promoter region of LC3B and transcriptionally activates LC3B. The overexpression of wild-type FoxO3a increased LC3B mRNA and protein expression in IPF fibroblasts, whereas the dominant negative FoxO3a decreased the LC3B level in control fibroblasts. The inhibition of autophagic activity sensitized control fibroblasts to collagen matrix-induced cell death. In contrast, enhanced viability was found when autophagic function was inhibited in IPF fibroblasts. Our study showed that aberrantly low FoxO3a expression participates in reducing autophagic activity via transcriptional suppression of LC3B in IPF fibroblasts on collagen. This suggests that low autophagic activity by the alteration of FoxO3a may contribute to IPF progression.


Subject(s)
Autophagy , Fibroblasts/physiology , Forkhead Transcription Factors/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Cells, Cultured , Collagen/chemistry , Culture Media/chemistry , Disease Progression , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Gene Expression , Gene Silencing , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism
9.
World J Biol Chem ; 5(3): 346-54, 2014 Aug 26.
Article in English | MEDLINE | ID: mdl-25225602

ABSTRACT

The Forkhead box O (FoxO) family has recently been highlighted as an important transcriptional regulator of crucial proteins associated with the many diverse functions of cells. So far, FoxO1, FoxO3a, FoxO4 and FoxO6 proteins have been identified in humans. Although each FoxO family member has its own role, unlike the other FoxO families, FoxO3a has been extensively studied because of its rather unique and pivotal regulation of cell proliferation, apoptosis, metabolism, stress management and longevity. FoxO3a alteration is closely linked to the progression of several types of cancers, fibrosis and other types of diseases. In this review, we will examine the function of FoxO3a in disease progression and also explore FoxO3a's regulatory mechanisms. We will also discuss FoxO3a as a potential target for the treatment of several types of disease.

10.
Am J Physiol Lung Cell Mol Physiol ; 307(8): L632-42, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25172912

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a lethal and progressive lung disease characterized by persistent (myo)fibroblasts and the relentless accumulation of collagen matrix. Unlike normal lung fibroblasts, IPF lung fibroblasts have suppressed forkhead box O3a (FoxO3a) activity, which allows them to expand in this diseased environment. microRNA-96 (miR-96) has recently been found to directly bind to the 3'-untranslated region of FoxO3a mRNA, which subsequently inhibits its function. We examined whether aberrantly low FoxO3a expression is in part due to increased miR-96 levels in IPF fibroblasts on polymerized collagen, thereby causing IPF fibroblasts to maintain their pathological properties. miR-96 expression was upregulated in IPF fibroblasts compared with control fibroblasts when cultured on collagen. In contrast, FoxO3a mRNA levels were reduced in most IPF fibroblasts. However, when miR-96 function was inhibited, FoxO3a mRNA and protein expression were increased, suppressing IPF fibroblast proliferation and promoting their cell death in a dose-dependent fashion. Likewise, FoxO3a and its target proteins p21, p27, and Bim expression was also increased in the presence of a miR-96 inhibitor in IPF fibroblasts. However, when control fibroblasts were treated with miR-96 mimic, FoxO3a, p27, p21, and Bim mRNA and protein levels were decreased. In situ hybridization analysis further revealed the presence of enhanced miR-96 expression in cells within the fibroblastic foci of IPF lung tissue. Our results suggest that when IPF fibroblasts interact with collagen-rich matrix, pathologically altered miR-96 expression inhibits FoxO3a function, causing IPF fibroblasts to maintain their pathological phenotype, which may contribute to the progression of IPF.


Subject(s)
Collagen Type I/metabolism , Fibroblasts/metabolism , Forkhead Transcription Factors/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , MicroRNAs/genetics , Blotting, Western , Cell Proliferation , Cell Survival , Cells, Cultured , Fibroblasts/pathology , Forkhead Box Protein O3 , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/genetics , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , In Situ Hybridization , Lung/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
11.
PLoS One ; 9(4): e94616, 2014.
Article in English | MEDLINE | ID: mdl-24728102

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic, lethal interstitial lung disease in which the aberrant PTEN/Akt axis plays a major role in conferring a survival phenotype in response to the cell death inducing properties of type I collagen matrix. The underlying mechanism by which IPF fibroblasts become desensitized to polymerized collagen, thereby eluding collagen matrix-induced cell death has not been fully elucidated. We hypothesized that the pathologically altered PTEN/Akt axis suppresses autophagy via high mTOR kinase activity, which subsequently desensitizes IPF fibroblasts to collagen matrix induced cell death. We found that the autophagosome marker LC3-2 expression is suppressed, while mTOR activity remains high when IPF fibroblasts are cultured on collagen. However, LC3-2 expression increased in response to IPF fibroblast attachment to collagen in the presence of rapamycin. In addition, PTEN over-expression or Akt inhibition suppressed mTOR activity, thereby increasing LC3-2 expression in IPF fibroblasts. Furthermore, the treatment of IPF fibroblasts over-expressing PTEN or dominant negative Akt with autophagy inhibitors increased IPF fibroblast cell death. Enhanced p-mTOR expression along with low LC3-2 expression was also found in myofibroblasts within the fibroblastic foci from IPF patients. Our data show that the aberrant PTEN/Akt/mTOR axis desensitizes IPF fibroblasts from polymerized collagen driven stress by suppressing autophagic activity, which produces a viable IPF fibroblast phenotype on collagen. This suggests that the aberrantly regulated autophagic pathway may play an important role in maintaining a pathological IPF fibroblast phenotype in response to collagen rich environment.


Subject(s)
Autophagy , Collagen Type I/metabolism , Fibroblasts/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Autophagy/drug effects , Cell Line , Cell Proliferation , Cell Survival , Chloroquine/pharmacology , Collagen Type I/chemistry , Enzyme Activation , Humans , Phagosomes/metabolism , Protein Multimerization
12.
PLoS One ; 8(4): e61017, 2013.
Article in English | MEDLINE | ID: mdl-23580232

ABSTRACT

Idiopathic Pulmonary Fibrosis is a lethal fibrotic disease characterized by the unrelenting proliferation and persistence of fibroblasts in a type I collagen-rich matrix that result in an expanding reticular network of fibrotic tissue. However, the underlying mechanism responsible for the persistence of myofibroblasts in IPF remains unclear. During normal tissue repair, unwanted fibroblasts are eliminated during collagen-matrix contraction by a mechanism whereby high PTEN activity suppresses Akt. We have previously found that FoxO3a, a transcriptional activator of apoptosis-inducing proteins, is inactivated in IPF fibroblasts resulting from aberrantly high PI3K/Akt activity due to inappropriately low PTEN activity. Here we demonstrate that this low FoxO3a activity confers IPF fibroblasts with resistance to collagen-mediated apoptosis. We show that the mechanism by which low FoxO3a activity confers IPF fibroblasts with an apoptotic resistant phenotype involves suppression of Fas expression as a result of down regulation of cav-1 expression via a PTEN/Akt-dependent pathway. We demonstrate that PTEN over-expression or Akt inhibition increases FoxO3a expression in IPF fibroblasts, resulting in up-regulation of caveolin-1. We show that FoxO3a binds to the cav-1 promoter region and ectopic expression of FoxO3a transcriptionally increases cav-1 mRNA and protein expression. In turn, we show that overexpression of caveolin-1 increases Fas levels and caspase-3/7 activity and promotes IPF fibroblast apoptosis on polymerized type I collagen. We have found that the expression of caveolin-1, Fas and cleaved caspase-3 proteins in fibroblasts within the fibroblastic foci of IPF patient specimens is low. Our data indicate that the pathologically altered PTEN/Akt axis inactivates FoxO3a down-regulating cav-1 and Fas expression. This confers IPF fibroblasts with an apoptosis-resistant phenotype and may be responsible for IPF progression.


Subject(s)
Apoptosis , Caveolin 1/metabolism , Collagen Type I/metabolism , Forkhead Transcription Factors/deficiency , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , fas Receptor/metabolism , Apoptosis/genetics , Caspase 3/metabolism , Caveolin 1/genetics , Cell Line , Collagen Type I/chemistry , Fibroblasts/metabolism , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Humans , Lung/metabolism , Lung/pathology , PTEN Phosphohydrolase/metabolism , Phenotype , Protein Multimerization , Proto-Oncogene Proteins c-akt/metabolism , fas Receptor/genetics
13.
Translation (Austin) ; 1(1): e23934, 2013.
Article in English | MEDLINE | ID: mdl-26824013

ABSTRACT

Pulmonary fibrosis is a severe lung disease characterized by sustained propagation of lung fibroblasts and relentless accumulation of extracellular matrix (ECM). Idiopathic pulmonary fibrosis (IPF) is the most severe chronic form of pulmonary fibrosis and results both in the gradual exchange of normal lung parenchyma with fibrotic tissue and in the irreversible impairment of gas exchange in the lung. Despite the urgency for novel therapies in IPF treatment, there is no effective and proven medical therapy available. Molecular mechanisms underlying IPF pathogenesis include aberrant ECM signaling through the canonical integrin/PI3K/Akt/mTORC1 signal transduction pathway. One important and well-characterized downstream effector of this pathway is the cellular protein synthesis machinery. Here we will review the recent advances in our understanding of the function of ECM and integrin receptor signaling in development of IPF and will present evidence indicating that the dysregulation of the eIF4F-mediated translational apparatus is an important factor in the development and progression of IPF and other fibrotic disorders. We further discuss the perspectives and challenges to curbing this deadly disease by targeting aberrant translation.

14.
Am J Pathol ; 179(5): 2420-30, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21893017

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a prevalent, progressive, and incurable fibroproliferative lung disease. The phenotype of IPF fibroblasts is characterized by their ability to elude the proliferation-suppressive properties of polymerized type I collagen. The mechanism underlying this pathological response is incompletely understood but involves aberrant activation of the phosphatidylinositol 3-kinase-Akt signaling pathway owing to inappropriately low phosphatase and tensin homolog phosphatase activity. Akt can phosphorylate and inactivate the forkhead box O3a (FoxO3a) transcriptional factor, which, when transcriptionally active, increases the expression of the CDK inhibitor p27 and promotes cell cycle arrest. Herein, we demonstrate that IPF fibroblasts display high levels of inactive FoxO3a compared with nonfibrotic control fibroblasts because of their high Akt activity. We found that p27 levels are decreased in IPF compared with control fibroblasts cultured on polymerized collagen. Furthermore, overexpression of FoxO3a in IPF fibroblasts increases p27 levels and suppresses the ability of IPF fibroblasts to proliferate on polymerized collagen. In contrast, the expression of dominant-negative FoxO3a augmented control fibroblast proliferation. IHC examination of fibroblastic foci in IPF lung tissue demonstrates the presence of inactive FoxO3a in cells within fibroblastic foci. These data indicate that the ability of IPF fibroblasts to circumvent the proliferation-suppressive properties of polymerized collagen involves inactivation of FoxO3a by high Akt activity, resulting in down-regulation of p27.


Subject(s)
Collagen Type I/metabolism , Fibroblasts/pathology , Forkhead Transcription Factors/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Forkhead Box Protein O3 , Humans , PTEN Phosphohydrolase/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism
15.
J Biol Chem ; 286(37): 31953-65, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21784851

ABSTRACT

Human lung fibroblasts utilize integrins to attach and proliferate on type I collagen. ß1 integrin is the major integrin subunit for this attachment. Integrins coordinate cellular responses to cell-cell and cell-extracellular matrix interactions that regulate a variety of biological processes. Although ß1 integrin-mediated signaling pathways in lung fibroblasts have been studied, a detailed molecular mechanism regulating translational control of gene expression by 4EBP-1 is not understood. 4EBP-1 inhibits cap-dependent translation by binding to the eIF4E translation initiation factor. We found that when lung fibroblasts attach to collagen via ß1 integrin, high Src activity suppresses 4EBP-1 expression via PP2A, and the decrease of 4EBP-1 is due to protein degradation. The inhibition of Src activity dramatically increases PP2A and 4EBP-1 expression. Furthermore ectopic expression of PP2A, or PP2A silencing using PP2A siRNA confirmed that 4EBP-1 is regulated by PP2A. In addition, we found that 4EBP-1 inhibition by fibroblast attachment to collagen increases cap-dependent translation. Our study showed that when lung fibroblasts are attached to collagen matrix, the ß1 integrin/Src/PP2A-mediated 4EBP-1 regulatory pathway is activated. We suggest that ß1 integrin-mediated signaling pathway may be a crucial event in regulating fibroblast translational control machinery on collagen matrix.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Integrin beta1/metabolism , Lung/metabolism , Phosphoproteins/metabolism , Protein Phosphatase 2/metabolism , src-Family Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Adhesion/physiology , Cell Cycle Proteins , Cell Line , Extracellular Matrix/genetics , Fibroblasts/cytology , Gene Silencing , Humans , Integrin beta1/genetics , Lung/cytology , Phosphoproteins/genetics , Protein Biosynthesis/physiology , Protein Phosphatase 2/genetics , RNA Caps/genetics , RNA Caps/metabolism , Signal Transduction/physiology , src-Family Kinases/genetics
16.
J Biol Chem ; 285(19): 14195-209, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20223831

ABSTRACT

When cells attach to the extracellular matrix (ECM) a proliferation permissive signal is engaged. The mechanism involves activation of the integrin/PI3K/Akt signal pathway. FoxO3a is a transcriptional activator and inhibits cell proliferation via up-regulating the expression of the cell cycle inhibitor p27. Furthermore, it is known that activated Akt can suppress FoxO3a function. However, it is not known whether integrin interaction with the ECM regulates FoxO3a function. We examined whether the beta1-integrin-mediated signaling pathway promotes fibroblast proliferation via FoxO3a suppression. We found that when fibroblasts are attached to collagen, PTEN protein expression and activity are inhibited due to promotion of PTEN degradation. This decrease in PTEN function permits FoxO3a suppression via the PI3K/Akt pathway. In contrast, the inhibition of PI3K/Akt or reconstitution of PTEN restores FoxO3a expression on collagen. Furthermore, we found that the serine/threonine phosphatase PP2A also regulates FoxO3a. PP2A expression/activity is low when fibroblasts are attached to collagen, and PP2A overexpression augments FoxO3a levels. Thus the mechanism involves a coordinated decrease in PTEN and PP2A phosphatase activity and increase in PI3K/Akt activity. We show that beta1-integrin-ECM interaction decreases FoxO3a protein levels via caspase-3-mediated cleavage. Our novel finding indicates that during fibroblast interaction with ECM, activation of beta1-integrin/PI3K/Akt by inhibiting PTEN in combination with low PP2A phosphatase activity synergistically inhibits FoxO3a, promoting fibroblast proliferation.


Subject(s)
Collagen Type I/metabolism , Forkhead Transcription Factors/physiology , Integrin beta1/metabolism , PTEN Phosphohydrolase/physiology , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Blotting, Western , Caspase 3/metabolism , Cell Adhesion , Cell Proliferation , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Extracellular Matrix/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Forkhead Box Protein O3 , Forkhead Transcription Factors/antagonists & inhibitors , Humans , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
17.
PLoS One ; 3(9): e3220, 2008 Sep 16.
Article in English | MEDLINE | ID: mdl-18795102

ABSTRACT

BACKGROUND: As a group, fibroproliferative disorders of the lung, liver, kidney, heart, vasculature and integument are common, progressive and refractory to therapy. They can emerge following toxic insults, but are frequently idiopathic. Their enigmatic propensity to resist therapy and progress to organ failure has focused attention on the myofibroblast-the primary effector of the fibroproliferative response. We have recently shown that aberrant beta 1 integrin signaling in fibrotic fibroblasts results in defective PTEN function, unrestrained Akt signaling and subsequent activation of the translation initiation machinery. How this pathological integrin signaling alters the gene expression pathway has not been elucidated. RESULTS: Using a systems approach to study this question in a prototype fibrotic disease, Idiopathic Pulmonary Fibrosis (IPF); here we show organized changes in the gene expression pathway of primary lung myofibroblasts that persist for up to 9 sub-cultivations in vitro. When comparing IPF and control myofibroblasts in a 3-dimensional type I collagen matrix, more genes differed at the level of ribosome recruitment than at the level of transcript abundance, indicating pathological translational control as a major characteristic of IPF myofibroblasts. To determine the effect of matrix state on translational control, myofibroblasts were permitted to contract the matrix. Ribosome recruitment in control myofibroblasts was relatively stable. In contrast, IPF cells manifested large alterations in the ribosome recruitment pattern. Pathological studies suggest an epithelial origin for IPF myofibroblasts through the epithelial to mesenchymal transition (EMT). In accord with this, we found systems-level indications for TGF-beta -driven EMT as one source of IPF myofibroblasts. CONCLUSIONS: These findings establish the power of systems level genome-wide analysis to provide mechanistic insights into fibrotic disorders such as IPF. Our data point to derangements of translational control downstream of aberrant beta 1 integrin signaling as a fundamental component of IPF pathobiology and indicates that TGF-beta -driven EMT is one source for IPF myofibroblasts.


Subject(s)
Genome, Human , Protein Biosynthesis , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/metabolism , Aged , Aged, 80 and over , Female , Fibroblasts/metabolism , Humans , Integrin beta1/metabolism , Male , Middle Aged , Models, Biological , Ribosomes/chemistry , Ribosomes/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism
18.
J Biol Chem ; 281(44): 33291-301, 2006 Nov 03.
Article in English | MEDLINE | ID: mdl-16963781

ABSTRACT

During tissue repair, excess fibroblasts are eliminated by apoptosis. This physiologic process limits fibrosis and restores normal anatomic patterns. Replicating physiologic apoptosis associated with tissue repair, fibroblasts incorporated into type I collagen matrices undergo apoptosis in response to collagen matrix contraction. In this in vitro model of wound repair, fibroblasts first attach to collagen via alpha2beta1 integrin. This provides a survival signal via activation of the phosphatidylinositol 3-kinase/Akt signal pathway. However, during subsequent collagen matrix contraction, the level of phosphorylated Akt progressively declines, triggering apoptosis. The mechanism underlying the fall in phosphorylated Akt is incompletely understood. Here we show that PTEN phosphatase becomes activated during collagen matrix contraction and is responsible for antagonizing phosphatidylinositol 3-kinase activity and promoting a decline in phosphorylated Akt and fibroblast apoptosis in response to collagen contraction. PTEN null fibroblasts displayed enhanced levels of phosphorylated Akt and were resistant to collagen matrix contraction-induced apoptosis. Reconstitution of PTEN in PTEN null cells conferred susceptibility to apoptosis in response to contraction of collagen matrices. Consistent with this, knockdown of PTEN in PTEN(+/+) embryonic fibroblasts by small interfering RNA augmented Akt activity and suppressed apoptosis in contractile collagen matrices. Furthermore, inhibition of Akt activity restored the sensitivity of PTEN null cells to collagen contraction-induced apoptosis, indicating that the mechanism by which PTEN alters fibroblast viability is through modulation of phosphorylated Akt levels. Our work suggests that collagen matrix contraction activates PTEN by a mechanism involving cytoskeletal disassembly. Our studies indicate a key role for PTEN in regulating fibroblast viability during tissue repair.


Subject(s)
Collagen/metabolism , Extracellular Matrix/metabolism , PTEN Phosphohydrolase/metabolism , Actins/metabolism , Apoptosis , Cell Line , Fibroblasts , Gene Expression Regulation , Humans , Lung/cytology , Lung/enzymology , Microtubules/metabolism , Mutation/genetics , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics
19.
J Biol Chem ; 280(28): 26630-9, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-15905178

ABSTRACT

A beta1 integrin phosphatidylinositol 3-kinase/Akt pathway regulates fibroblast survival in collagen matrices. When fibroblasts attach to collagen, Akt becomes phosphorylated, providing a survival signal. In contrast, in response to mechanical forces generated during collagen contraction, Akt is dephosphorylated and fibroblasts undergo apoptosis. The kinase(s) responsible for regulating Akt phosphorylation in response to matrix-derived mechanical signals are unclear. Integrin-linked kinase (ILK) is associated with the beta1 integrin in the focal adhesion complex and as such is a candidate kinase that may regulate Akt phosphorylation and fibroblast viability. Nevertheless, there is no direct evidence that matrix-derived mechanical forces regulate cell viability by modulating ILK activity. Here, we show that ILK activity decreased in response to collagen matrix contraction, which correlated with Akt dephosphorylation and induction of fibroblast apoptosis. In contrast, enforced activation of beta1 integrin by activating antibody preserved ILK and Akt activity during collagen matrix contraction, and this is associated with protection from collagen contraction-induced apoptosis. Knock-down of ILK by small, interfering RNA (siRNA) attenuated Akt phosphorylation in response to ligation of beta1 integrin by collagen or activating antibody and enhanced fibroblast apoptosis in response to collagen contraction. Kinase dead ILK attenuated Akt phosphorylation and enhanced fibroblast apoptosis, whereas hyperactive and wild type ILK augmented Akt phosphorylation and protected fibroblasts from apoptosis. Constitutively active Akt preserved Akt activity and rescued ILK siRNA-treated fibroblasts from collagen contraction-induced apoptosis. These data establish that matrix-derived mechanical forces sensed by beta1 integrin are capable of modulating ILK activity which regulates fibroblast viability via an Akt-dependent mechanism.


Subject(s)
Fibroblasts/cytology , Integrin beta1/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/metabolism , Apoptosis , Blotting, Western , Cell Adhesion , Cell Line , Cell Survival , Cells, Cultured , Collagen/chemistry , Fibroblasts/metabolism , Humans , Immunoprecipitation , In Situ Nick-End Labeling , Lung/cytology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Serine/chemistry , Signal Transduction , Time Factors , Transfection
20.
J Biol Chem ; 279(31): 33024-34, 2004 Jul 30.
Article in English | MEDLINE | ID: mdl-15166238

ABSTRACT

The beta(1) integrin, functioning as a mechanoreceptor, senses a mechanical stimulus generated during collagen matrix contraction and down-regulates the phosphatidylinositol 3-kinase (PI3K)/Akt survival signal triggering apoptosis. The identities of integrin-associated signal molecules in the focal adhesion complex that are responsible for propagating beta(1) integrin viability signals in response to collagen matrix contraction are not known. Here we show that in response to collagen contraction focal adhesion kinase (FAK) is dephosphorylated. In contrast, enforced activation of beta(1) integrin by anti-beta(1) integrin antibody, which protects fibroblasts from apoptosis, preserves FAK phosphorylation. We demonstrate that ligation of beta(1) integrin by type I collagen or by enforced activation of beta(1) integrin by antibody promotes phosphorylation of FAK, p85 subunit of PI3K, and serine 473 of Akt. Wortmannin inhibited Akt but not FAK phosphorylation in response to enforced activation of beta(1) integrin by antibody. Blocking FAK by pharmacologic inhibition or by dominant negative FAK attenuated phosphorylation of p85 subunit of PI3K and Akt. Dominant negative FAK augmented fibroblast apoptosis during collagen contraction, and this was associated with diminished Akt activity. Constitutively active FAK augmented levels of p85 subunit of PI3K and Akt phosphorylation, and fibroblasts were protected from apoptosis. Our data identify a novel role for FAK, functioning upstream of PI3K/Akt, in transducing a beta(1) integrin viability signal in collagen matrices.


Subject(s)
Collagen/chemistry , Fibroblasts/metabolism , Integrin beta1/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Adenoviridae/genetics , Androstadienes/pharmacology , Anoikis , Apoptosis , Blotting, Western , Cell Survival , Cells, Cultured , DNA, Complementary/metabolism , Down-Regulation , Enzyme Inhibitors/pharmacology , Fibrinogen/metabolism , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Genes, Dominant , Humans , In Situ Nick-End Labeling , Models, Biological , Phosphorylation , Precipitin Tests , Protein Structure, Tertiary , Retroviridae/genetics , Signal Transduction , Time Factors , Up-Regulation , Wortmannin
SELECTION OF CITATIONS
SEARCH DETAIL
...