Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Am J Respir Cell Mol Biol ; 68(2): 176-185, 2023 02.
Article in English | MEDLINE | ID: mdl-36174229

ABSTRACT

Tissue availability remains an important limitation of single-cell genomic technologies for investigating cellular heterogeneity in human health and disease. BAL represents a minimally invasive approach to assessing an individual's lung cellular environment for diagnosis and research. However, the lack of high-quality, healthy lung reference data is a major obstacle to using single-cell approaches to study a plethora of lung diseases. Here, we performed single-cell RNA sequencing on over 40,000 cells isolated from the BAL of four healthy volunteers. Of the six cell types or lineages we identified, macrophages were consistently the most numerous across individuals. Our analysis confirmed the expression of marker genes defining cell types despite background signals because of the ambient RNA found in many single-cell studies. We assessed the variability of gene expression across macrophages and defined a distinct subpopulation of cells expressing a set of genes associated with Macrophage Inflammatory Protein 1 (MIP-1). RNA in situ hybridization and reanalysis of published lung single-cell data validated the presence of this macrophage subpopulation. Thus, our study characterizes lung macrophage heterogeneity in healthy individuals and provides a valuable resource for future studies to understand the lung environment in health and disease.


Subject(s)
Macrophage Inflammatory Proteins , Macrophages , Humans , Macrophage Inflammatory Proteins/genetics , Bronchoalveolar Lavage Fluid , Healthy Volunteers , RNA
2.
iScience ; 25(7): 104669, 2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35856022

ABSTRACT

Intestinal dysbiosis is prominent in systemic sclerosis (SSc), but it remains unknown how it contributes to microvascular injury and fibrosis that are hallmarks of this disease. Trimethylamine (TMA) is generated by the gut microbiome and in the host converted by flavin-containing monooxygenase (FMO3) into trimethylamine N-oxide (TMAO), which has been implicated in chronic cardiovascular and metabolic diseases. Using cell culture systems and patient biopsies, we now show that TMAO reprograms skin fibroblasts, vascular endothelial cells, and adipocytic progenitor cells into myofibroblasts via the putative TMAO receptor protein R-like endoplasmic reticulum kinase (PERK). Remarkably, FMO3 was detected in skin fibroblasts and its expression stimulated by TGF-ß1. Moreover, FMO3 was elevated in SSc skin biopsies and in SSc fibroblasts. A meta-organismal pathway thus might in SSc link gut microbiome to vascular remodeling and fibrosis via stromal cell reprogramming, implicating the FMO3-TMAO-PERK axis in pathogenesis, and as a promising target for therapy.

3.
Int J Mol Sci ; 22(13)2021 Jun 25.
Article in English | MEDLINE | ID: mdl-34202229

ABSTRACT

Alveolar epithelial cell (AEC) mitochondrial (mt) DNA damage and fibrotic monocyte-derived alveolar macrophages (Mo-AMs) are implicated in the pathobiology of pulmonary fibrosis. We showed that sirtuin 3 (SIRT3), a mitochondrial protein regulating cell fate and aging, is deficient in the AECs of idiopathic pulmonary fibrosis (IPF) patients and that asbestos- and bleomycin-induced lung fibrosis is augmented in Sirt3 knockout (Sirt3-/-) mice associated with AEC mtDNA damage and intrinsic apoptosis. We determined whether whole body transgenic SIRT3 overexpression (Sirt3Tg) protects mice from asbestos-induced pulmonary fibrosis by mitigating lung mtDNA damage and Mo-AM recruitment. Crocidolite asbestos (100 µg/50 µL) or control was instilled intratracheally in C57Bl6 (Wild-Type) mice or Sirt3Tg mice, and at 21 d lung fibrosis (histology, fibrosis score, Sircol assay) and lung Mo-AMs (flow cytometry) were assessed. Compared to controls, Sirt3Tg mice were protected from asbestos-induced pulmonary fibrosis and had diminished lung mtDNA damage and Mo-AM recruitment. Further, pharmacologic SIRT3 inducers (i.e., resveratrol, viniferin, and honokiol) each diminish oxidant-induced AEC mtDNA damage in vitro and, in the case of honokiol, protection occurs in a SIRT3-dependent manner. We reason that SIRT3 preservation of AEC mtDNA is a novel therapeutic focus for managing patients with IPF and other types of pulmonary fibrosis.


Subject(s)
Asbestos/adverse effects , DNA Damage , Gene Expression , Idiopathic Pulmonary Fibrosis/etiology , Mitochondria/genetics , Monocytes/metabolism , Sirtuin 3/genetics , Animals , Biomarkers , DNA, Mitochondrial , Disease Models, Animal , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Immunohistochemistry , Mice , Mice, Transgenic , Mitochondria/metabolism , Monocytes/immunology , Monocytes/pathology , Oxidative Stress , Sirtuin 3/metabolism
4.
Biomater Sci ; 9(16): 5497-5507, 2021 Aug 21.
Article in English | MEDLINE | ID: mdl-34075946

ABSTRACT

Mitochondria are crucial regulators of the intrinsic pathway of cancer cell death. The high sensitivity of cancer cells to mitochondrial dysfunction offers opportunities for emerging targets in cancer therapy. Herein, magnetic nano-transducers, which convert external magnetic fields into physical stress, are designed to induce mitochondrial dysfunction to remotely kill cancer cells. Spindle-shaped iron oxide nanoparticles were synthesized to maximize cellular internalization and magnetic transduction. The magneto-mechanical transduction of nano-transducers in mitochondria enhances cancer cell apoptosis by promoting a mitochondrial quality control mechanism, referred to as mitophagy. In the liver cancer animal model, nano-transducers are infused into the local liver tumor via the hepatic artery. After treatment with a magnetic field, in vivo mitophagy-mediated cancer cell death was also confirmed by mitophagy markers, mitochondrial DNA damage assay, and TUNEL staining of tissues. This study is expected to contribute to the development of nanoparticle-mediated mitochondria-targeting cancer therapy and biological tools, such as magneto-genetics.


Subject(s)
Mitophagy , Neoplasms , Animals , Apoptosis , Cell Line, Tumor , Humans , Magnetic Phenomena , Mitochondria , Neoplasms/therapy
5.
Int J Mol Sci ; 21(16)2020 Aug 05.
Article in English | MEDLINE | ID: mdl-32764262

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic disease for which novel approaches are urgently required. We reported increased sphingosine kinase 1 (SPHK1) in IPF lungs and that SPHK1 inhibition using genetic and pharmacologic approaches reduces murine bleomycin-induced pulmonary fibrosis. We determined whether PF543, a specific SPHK1 inhibitor post bleomycin or asbestos challenge mitigates lung fibrosis by reducing mitochondrial (mt) DNA damage and pro-fibrotic monocyte recruitment-both are implicated in the pathobiology of pulmonary fibrosis. Bleomycin (1.5 U/kg), crocidolite asbestos (100 µg/50 µL) or controls was intratracheally instilled in Wild-Type (C57Bl6) mice. PF543 (1 mg/kg) or vehicle was intraperitoneally injected once every two days from day 7-21 following bleomycin and day 14-21 or day 30-60 following asbestos. PF543 reduced bleomycin- and asbestos-induced pulmonary fibrosis at both time points as well as lung expression of profibrotic markers, lung mtDNA damage, and fibrogenic monocyte recruitment. In contrast to human lung fibroblasts, asbestos augmented lung epithelial cell (MLE) mtDNA damage and PF543 was protective. Post-exposure PF543 mitigates pulmonary fibrosis in part by reducing lung epithelial cell mtDNA damage and monocyte recruitment. We reason that SPHK1 signaling may be an innovative therapeutic target for managing patients with IPF and other forms of lung fibrosis.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Idiopathic Pulmonary Fibrosis/drug therapy , Methanol/analogs & derivatives , Pulmonary Fibrosis/drug therapy , Pyrrolidines/pharmacology , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Alveolar Epithelial Cells/drug effects , Animals , Asbestos/toxicity , Bleomycin/pharmacology , DNA Damage/drug effects , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/genetics , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Lung/drug effects , Lung/metabolism , Methanol/pharmacology , Mice , Mitochondria/drug effects , Mitochondria/genetics , Monocytes/drug effects , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Signal Transduction/drug effects , Sulfones
6.
Am J Physiol Lung Cell Mol Physiol ; 318(5): L1084-L1096, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32209025

ABSTRACT

Alveolar epithelial cell (AEC) apoptosis, arising from mitochondrial dysfunction and mitophagy defects, is important in mediating idiopathic pulmonary fibrosis (IPF). Our group established a role for the mitochondrial (mt) DNA base excision repair enzyme, 8-oxoguanine-DNA glycosylase 1 (mtOGG1), in preventing oxidant-induced AEC mtDNA damage and apoptosis and showed that OGG1-deficient mice have increased lung fibrosis. Herein, we determined whether mice overexpressing the mtOGG1 transgene (mtOgg1tg) are protected against lung fibrosis and whether AEC mtOGG1 preservation of mtDNA integrity mitigates phosphatase and tensin homolog-induced putative kinase 1 (PINK1) deficiency and apoptosis. Compared with wild type (WT), mtOgg1tg mice have diminished asbestos- and bleomycin-induced pulmonary fibrosis that was accompanied by reduced lung and AEC mtDNA damage and apoptosis. Asbestos and H2O2 promote the MLE-12 cell PINK1 deficiency, as assessed by reductions in the expression of PINK1 mRNA and mitochondrial protein expression. Compared with WT, Pink1-knockout (Pink1-KO) mice are more susceptible to asbestos-induced lung fibrosis and have increased lung and alveolar type II (AT2) cell mtDNA damage and apoptosis. AT2 cells from Pink1-KO mice and PINK1-silenced (siRNA) MLE-12 cells have increased mtDNA damage that is augmented by oxidative stress. Interestingly, mtOGG1 overexpression attenuates oxidant-induced MLE-12 cell mtDNA damage and apoptosis despite PINK1 silencing. mtDNA damage is increased in the lungs of patients with IPF as compared with controls. Collectively, these findings suggest that mtOGG1 maintenance of AEC mtDNA is crucial for preventing PINK1 deficiency that promotes apoptosis and lung fibrosis. Given the key role of AEC apoptosis in pulmonary fibrosis, strategies aimed at preserving AT2 cell mtDNA integrity may be an innovative target.


Subject(s)
Alveolar Epithelial Cells/drug effects , Asbestosis/genetics , DNA Glycosylases/genetics , Lung/drug effects , Mitochondria/drug effects , Protein Kinases/genetics , Pulmonary Fibrosis/genetics , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Asbestos/administration & dosage , Asbestosis/etiology , Asbestosis/metabolism , Asbestosis/pathology , Bleomycin/administration & dosage , DNA Damage , DNA Glycosylases/deficiency , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Female , Gene Expression Regulation , Hydrogen Peroxide/pharmacology , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/metabolism , Primary Cell Culture , Protein Kinases/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Titanium/administration & dosage
7.
Front Immunol ; 10: 1813, 2019.
Article in English | MEDLINE | ID: mdl-31417574

ABSTRACT

Macrophages are often viewed through the lens of their core functions, but recent transcriptomic studies reveal them to be largely distinct across tissue types. While these differences appear to be shaped by their local environment, the key signals that drive these transcriptional differences remain unclear. Since Wnt signaling plays established roles in cell fate decisions, and tissue patterning during development and tissue repair after injury, we consider evidence that Wnt signals both target and are affected by macrophage functions. We propose that the Wnt gradients present in developing and adult tissues effectively shape macrophage fates and phenotypes. We also highlight evidence that macrophages, through an ability to dispatch Wnt signals, may couple tissue debridement and matrix remodeling with stem cell activation and tissue repair.


Subject(s)
Cell Differentiation/immunology , Macrophages/immunology , Stem Cells/immunology , Wnt Signaling Pathway/immunology , Animals , Humans , beta Catenin/immunology
8.
Am J Physiol Lung Cell Mol Physiol ; 317(2): L175-L187, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31090437

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a pernicious lung disease characterized by alveolar epithelial apoptosis, dysregulated repair of epithelial injury, scar formation, and respiratory failure. In this study, we identified phospholipase D (PLD)-generated phosphatidic acid (PA) signaling in the development of pulmonary fibrosis (PF). Of the PLD isoenzymes, the protein expression of PLD2, but not PLD1, was upregulated in lung tissues from IPF patients and bleomycin challenged mice. Both PLD1 (Pld1-/-)- and PLD2 (Pld2-/-)-deficient mice were protected against bleomycin-induced lung inflammation and fibrosis, thereby establishing the role of PLD in fibrogenesis. The role of PLD1 and PLD2 in bleomycin-induced lung epithelial injury was investigated by infecting bronchial airway epithelial cells (Beas2B) with catalytically inactive mutants of PLD (hPLD1-K898R or mPld2-K758R) or downregulation of expression of PLD1 or PLD2 with siRNA. Bleomycin stimulated mitochondrial (mt) superoxide production, mtDNA damage, and apoptosis in Beas2B cells, which was attenuated by the catalytically inactive mutants of PLD or PLD2 siRNA. These results show a role for PLD1 and PLD2 in bleomycin-induced generation of mt reactive oxygen species, mt DNA damage, and apoptosis of lung epithelial cells in mice. Thus, PLD may be a novel therapeutic target in ameliorating experimental PF in mice.


Subject(s)
Bleomycin/pharmacology , Lung/drug effects , Mitochondria/drug effects , Phospholipase D/metabolism , Animals , DNA Damage/drug effects , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , Mice, Transgenic , Mitochondria/metabolism , Phospholipase D/drug effects , Reactive Oxygen Species/metabolism
9.
Sci Transl Med ; 10(457)2018 09 05.
Article in English | MEDLINE | ID: mdl-30185650

ABSTRACT

The elevation of carbon dioxide (CO2) in tissues and the bloodstream (hypercapnia) occurs in patients with severe lung diseases, including chronic obstructive pulmonary disease (COPD). Whereas hypercapnia has been recognized as a marker of COPD severity, a role for hypercapnia in disease pathogenesis remains unclear. We provide evidence that CO2 acts as a signaling molecule in mouse and human airway smooth muscle cells. High CO2 activated calcium-calpain signaling and consequent smooth muscle cell contraction in mouse airway smooth muscle cells. The signaling was mediated by caspase-7-induced down-regulation of the microRNA-133a (miR-133a) and consequent up-regulation of Ras homolog family member A and myosin light-chain phosphorylation. Exposure of wild-type, but not caspase-7-null, mice to hypercapnia increased airway contraction and resistance. Deletion of the Caspase-7 gene prevented hypercapnia-induced airway contractility, which was restored by lentiviral transfection of a miR-133a antagonist. In a cohort of patients with severe COPD, hypercapnic patients had higher airway resistance, which improved after correction of hypercapnia. Our data suggest a specific molecular mechanism by which the development of hypercapnia may drive COPD pathogenesis and progression.


Subject(s)
Caspase 7/metabolism , Hypercapnia/metabolism , Hypercapnia/physiopathology , Muscle Contraction , Muscle, Smooth/physiopathology , Signal Transduction , rhoA GTP-Binding Protein/metabolism , Acetylcholine/pharmacology , Aged , Aged, 80 and over , Airway Resistance , Animals , Calcium/metabolism , Calpain/metabolism , Carbon Dioxide , Chronic Disease , Down-Regulation/drug effects , Enzyme Activation/drug effects , Female , Humans , Hypercapnia/genetics , MEF2 Transcription Factors/metabolism , Male , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Disease, Chronic Obstructive/physiopathology
10.
Oncotarget ; 8(35): 58152-58162, 2017 Aug 29.
Article in English | MEDLINE | ID: mdl-28938544

ABSTRACT

The oligosaccharides in human milk have various biological functions. However, the molecular mechanism(s) underlying the anti-angiogenic action of sialylated human milk oligosaccharides (HMOs) are still unclear. Here, we show that siallylactose (SL) found in human milk can inhibit the activation of vascular endothelial growth factor (VEGF)-mediated VEGF receptor-2 (VEGFR-2) by binding to its VEGF binding site (second and third IgG-like domains), thus blocking downstream signal activation. SL also inhibits growth of VEGF-stimulated endothelial cells. In endothelial cells treated with VEGF, SL diminished tube formation, migration, and the arrangement of actin filament. In addition, SL clearly suppressed VEGF-induced neovascularization in an in vivo Matrigel plug assay. Notably, SL prevented the growth of tumor cells, and angiogenesis on tumor tissues in in vivo mice models allotransplanted with Lewis lung carcinoma, melanoma, and colon carcinoma cells. Taken together, we have demonstrated that the sialylated milk oligosaccharide sialyllactose functions as an inhibitor of angiogenesis through suppression of VEGF-mediated VEGFR-2 activation in endothelial cells, Accordingly, it could be a novel candidate for the development of anti-angiogenic drugs without any side effects.

11.
Am J Physiol Cell Physiol ; 312(6): C749-C764, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28424170

ABSTRACT

Reactive oxygen species (ROS) derived from NADPH oxidase (NOX) and mitochondria play a critical role in growth factor-induced switch from a quiescent to an angiogenic phenotype in endothelial cells (ECs). However, how highly diffusible ROS produced from different sources can coordinate to stimulate VEGF signaling and drive the angiogenic process remains unknown. Using the cytosol- and mitochondria-targeted redox-sensitive RoGFP biosensors with real-time imaging, here we show that VEGF stimulation in human ECs rapidly increases cytosolic RoGFP oxidation within 1 min, followed by mitochondrial RoGFP oxidation within 5 min, which continues at least for 60 min. Silencing of Nox4 or Nox2 or overexpression of mitochondria-targeted catalase significantly inhibits VEGF-induced tyrosine phosphorylation of VEGF receptor type 2 (VEGFR2-pY), EC migration and proliferation at the similar extent. Exogenous hydrogen peroxide (H2O2) or overexpression of Nox4, which produces H2O2, increases mitochondrial ROS (mtROS), which is prevented by Nox2 siRNA, suggesting that Nox2 senses Nox4-derived H2O2 to promote mtROS production. Mechanistically, H2O2 increases S36 phosphorylation of p66Shc, a key mtROS regulator, which is inhibited by siNox2, but not by siNox4. Moreover, Nox2 or Nox4 knockdown or overexpression of S36 phosphorylation-defective mutant p66Shc(S36A) inhibits VEGF-induced mtROS, VEGFR2-pY, EC migration, and proliferation. In summary, Nox4-derived H2O2 in part activates Nox2 to increase mtROS via pSer36-p66Shc, thereby enhancing VEGFR2 signaling and angiogenesis in ECs. This may represent a novel feed-forward mechanism of ROS-induced ROS release orchestrated by the Nox4/Nox2/pSer36-p66Shc/mtROS axis, which drives sustained activation of angiogenesis signaling program.


Subject(s)
Feedback, Physiological , Hydrogen Peroxide/metabolism , Membrane Glycoproteins/genetics , Mitochondria/metabolism , NADPH Oxidases/genetics , Signal Transduction , Biosensing Techniques , Catalase/genetics , Catalase/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells , Humans , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/metabolism , Microscopy, Fluorescence , Mitochondria/drug effects , NADPH Oxidase 2 , NADPH Oxidase 4 , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , Oxidation-Reduction , Phosphorylation/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1/genetics , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism , Time-Lapse Imaging , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
12.
Am J Physiol Lung Cell Mol Physiol ; 313(1): L16-L26, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28428174

ABSTRACT

Alveolar epithelial cell (AEC) apoptosis and inadequate repair resulting from "exaggerated" lung aging and mitochondrial dysfunction are critical determinants promoting lung fibrosis. α-Klotho, which is an antiaging molecule that is expressed predominantly in the kidney and secreted in the blood, can protect lung epithelial cells against hyperoxia-induced apoptosis. We reasoned that Klotho protects AEC exposed to oxidative stress in part by maintaining mitochondrial DNA (mtDNA) integrity and mitigating apoptosis. We find that Klotho levels are decreased in both serum and alveolar type II (AT2) cells from asbestos-exposed mice. We show that oxidative stress reduces AEC Klotho mRNA and protein expression, whereas Klotho overexpression is protective while Klotho silencing augments AEC mtDNA damage. Compared with wild-type, Klotho heterozygous hypomorphic allele (kl/+) mice have increased asbestos-induced lung fibrosis due in part to increased AT2 cell mtDNA damage. Notably, we demonstrate that serum Klotho levels are reduced in wild-type but not mitochondrial catalase overexpressing (MCAT) mice 3 wk following exposure to asbestos and that EUK-134, a MnSOD/catalase mimetic, mitigates oxidant-induced reductions in AEC Klotho expression. Using pharmacologic and genetic silencing studies, we show that Klotho attenuates oxidant-induced AEC mtDNA damage and apoptosis via mechanisms dependent on AKT activation arising from upstream fibroblast growth factor receptor 1 activation. Our findings suggest that Klotho preserves AEC mtDNA integrity in the setting of oxidative stress necessary for preventing apoptosis and asbestos-induced lung fibrosis. We reason that strategies aimed at augmenting AEC Klotho levels may be an innovative approach for mitigating age-related lung diseases.


Subject(s)
Aging/metabolism , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Apoptosis/drug effects , DNA Damage , DNA, Mitochondrial/metabolism , Glucuronidase/metabolism , Oxidants/toxicity , Alveolar Epithelial Cells/drug effects , Animals , Apoptosis/genetics , Asbestos , Catalase/metabolism , Cell Line , DNA Damage/genetics , Female , Gene Expression Regulation/drug effects , Glucuronidase/deficiency , Glucuronidase/genetics , Klotho Proteins , Male , Mice , Mitochondria/metabolism , Organometallic Compounds/pharmacology , Oxidative Stress/drug effects , Protective Agents/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , RNA, Messenger/blood , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, IGF Type 1/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Salicylates/pharmacology , Signal Transduction/drug effects
13.
FASEB J ; 31(6): 2520-2532, 2017 06.
Article in English | MEDLINE | ID: mdl-28258190

ABSTRACT

Alveolar epithelial cell (AEC) mitochondrial dysfunction and apoptosis are important in idiopathic pulmonary fibrosis and asbestosis. Sirtuin 3 (SIRT3) detoxifies mitochondrial reactive oxygen species, in part, by deacetylating manganese superoxide dismutase (MnSOD) and mitochondrial 8-oxoguanine DNA glycosylase. We reasoned that SIRT3 deficiency occurs in fibrotic lungs and thereby augments AEC mtDNA damage and apoptosis. Human lungs were assessed by using immunohistochemistry for SIRT3 activity via acetylated MnSODK68 Murine AEC SIRT3 and cleaved caspase-9 (CC-9) expression were assayed by immunoblotting with or without SIRT3 enforced expression or silencing. mtDNA damage was measured by using quantitative PCR and apoptosis via ELISA. Pulmonary fibrosis after asbestos or bleomycin exposure was evaluated in 129SJ/wild-type and SIRT3-knockout mice (Sirt3-/- ) by using fibrosis scoring and lung collagen levels. Idiopathic pulmonary fibrosis lung alveolar type II cells have increased MnSODK68 acetylation compared with controls. Asbestos and H2O2 diminished AEC SIRT3 protein expression and increased mitochondrial protein acetylation, including MnSODK68 SIRT3 enforced expression reduced oxidant-induced AEC OGG1K338/341 acetylation, mtDNA damage, and apoptosis, whereas SIRT3 silencing promoted these effects. Asbestos- or bleomycin-induced lung fibrosis, AEC mtDNA damage, and apoptosis in wild-type mice were amplified in Sirt3-/- animals. These data suggest a novel role for SIRT3 deficiency in mediating AEC mtDNA damage, apoptosis, and lung fibrosis.-Jablonski, R. P., Kim, S.-J., Cheresh, P., Williams, D. B., Morales-Nebreda, L., Cheng, Y., Yeldandi, A., Bhorade, S., Pardo, A., Selman, M., Ridge, K., Gius, D., Budinger, G. R. S., Kamp, D. W. SIRT3 deficiency promotes lung fibrosis by augmenting alveolar epithelial cell mitochondrial DNA damage and apoptosis.


Subject(s)
Alveolar Epithelial Cells/pathology , Apoptosis/physiology , DNA, Mitochondrial/physiology , Pulmonary Fibrosis/etiology , Sirtuin 3/metabolism , A549 Cells , Animals , Antibiotics, Antineoplastic/toxicity , Asbestos/toxicity , Bleomycin/toxicity , DNA Damage , Humans , Mice , Mice, Knockout , Oxidants/toxicity , Pulmonary Fibrosis/metabolism , Sirtuin 3/genetics
14.
Free Radic Biol Med ; 101: 482-490, 2016 12.
Article in English | MEDLINE | ID: mdl-27840320

ABSTRACT

RATIONALE: Alveolar epithelial cell (AEC) injury and mitochondrial dysfunction are important in the development of lung fibrosis. Our group has shown that in the asbestos exposed lung, the generation of mitochondrial reactive oxygen species (ROS) in AEC mediate mitochondrial DNA (mtDNA) damage and apoptosis which are necessary for lung fibrosis. These data suggest that mitochondrial-targeted antioxidants should ameliorate asbestos-induced lung. OBJECTIVE: To determine whether transgenic mice that express mitochondrial-targeted catalase (MCAT) have reduced lung fibrosis following exposure to asbestos or bleomycin and, if so, whether this occurs in association with reduced AEC mtDNA damage and apoptosis. METHODS: Crocidolite asbestos (100µg/50µL), TiO2 (negative control), bleomycin (0.025 units/50µL), or PBS was instilled intratracheally in 8-10 week-old wild-type (WT - C57Bl/6J) or MCAT mice. The lungs were harvested at 21d. Lung fibrosis was quantified by collagen levels (Sircol) and lung fibrosis scores. AEC apoptosis was assessed by cleaved caspase-3 (CC-3)/Surfactant protein C (SFTPC) immunohistochemistry (IHC) and semi-quantitative analysis. AEC (primary AT2 cells from WT and MCAT mice and MLE-12 cells) mtDNA damage was assessed by a quantitative PCR-based assay, apoptosis was assessed by DNA fragmentation, and ROS production was assessed by a Mito-Sox assay. RESULTS: Compared to WT, crocidolite-exposed MCAT mice exhibit reduced pulmonary fibrosis as measured by lung collagen levels and lung fibrosis score. The protective effects in MCAT mice were accompanied by reduced AEC mtDNA damage and apoptosis. Similar findings were noted following bleomycin exposure. Euk-134, a mitochondrial SOD/catalase mimetic, attenuated MLE-12 cell DNA damage and apoptosis. Finally, compared to WT, asbestos-induced MCAT AT2 cell ROS production was reduced. CONCLUSIONS: Our finding that MCAT mice have reduced pulmonary fibrosis, AEC mtDNA damage and apoptosis following exposure to asbestos or bleomycin suggests an important role for AEC mitochondrial H2O2-induced mtDNA damage in promoting lung fibrosis. We reason that strategies aimed at limiting AEC mtDNA damage arising from excess mitochondrial H2O2 production may be a novel therapeutic target for mitigating pulmonary fibrosis.


Subject(s)
Catalase/genetics , DNA, Mitochondrial/drug effects , Epithelial Cells/drug effects , Mitochondria/drug effects , Mitochondrial Proteins/genetics , Pulmonary Alveoli/drug effects , Pulmonary Fibrosis/prevention & control , Administration, Inhalation , Animals , Asbestos , Bleomycin , Caspase 3/genetics , Caspase 3/metabolism , Catalase/metabolism , Collagen/antagonists & inhibitors , Collagen/genetics , Collagen/metabolism , DNA, Mitochondrial/chemistry , DNA, Mitochondrial/metabolism , Epithelial Cells/enzymology , Epithelial Cells/pathology , Gene Expression , Gene Expression Regulation , Intercellular Signaling Peptides and Proteins , Intubation, Intratracheal , Mice , Mice, Transgenic , Mitochondria/enzymology , Mitochondria/pathology , Mitochondrial Proteins/metabolism , Organometallic Compounds/pharmacology , Peptides/genetics , Peptides/metabolism , Pulmonary Alveoli/enzymology , Pulmonary Alveoli/pathology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Pulmonary Surfactant-Associated Protein C , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Salicylates/pharmacology , Transgenes
15.
Int J Mol Sci ; 16(9): 21486-519, 2015 Sep 07.
Article in English | MEDLINE | ID: mdl-26370974

ABSTRACT

Convincing evidence has emerged demonstrating that impairment of mitochondrial function is critically important in regulating alveolar epithelial cell (AEC) programmed cell death (apoptosis) that may contribute to aging-related lung diseases, such as idiopathic pulmonary fibrosis (IPF) and asbestosis (pulmonary fibrosis following asbestos exposure). The mammalian mitochondrial DNA (mtDNA) encodes for 13 proteins, including several essential for oxidative phosphorylation. We review the evidence implicating that oxidative stress-induced mtDNA damage promotes AEC apoptosis and pulmonary fibrosis. We focus on the emerging role for AEC mtDNA damage repair by 8-oxoguanine DNA glycosylase (OGG1) and mitochondrial aconitase (ACO-2) in maintaining mtDNA integrity which is important in preventing AEC apoptosis and asbestos-induced pulmonary fibrosis in a murine model. We then review recent studies linking the sirtuin (SIRT) family members, especially SIRT3, to mitochondrial integrity and mtDNA damage repair and aging. We present a conceptual model of how SIRTs modulate reactive oxygen species (ROS)-driven mitochondrial metabolism that may be important for their tumor suppressor function. The emerging insights into the pathobiology underlying AEC mtDNA damage and apoptosis is suggesting novel therapeutic targets that may prove useful for the management of age-related diseases, including pulmonary fibrosis and lung cancer.


Subject(s)
Alveolar Epithelial Cells/metabolism , Apoptosis/genetics , DNA, Mitochondrial , Pulmonary Fibrosis/genetics , Aging , Animals , DNA Damage , DNA Glycosylases/metabolism , DNA Repair , Disease Models, Animal , Guanine/analogs & derivatives , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Oxidative Stress/genetics , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Reactive Oxygen Species/metabolism , Sirtuin 3/genetics , Sirtuin 3/metabolism
16.
Am J Respir Cell Mol Biol ; 52(1): 25-36, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24918270

ABSTRACT

Asbestos causes asbestosis and malignancies by mechanisms that are not fully established. Alveolar epithelial cell (AEC) injury and repair are crucial determinants of the fibrogenic potential of noxious agents such as asbestos. We previously showed that mitochondrial reactive oxygen species mediate asbestos-induced AEC intrinsic apoptosis and that mitochondrial human 8-oxoguanine-DNA glycosylase 1 (OGG1), a DNA repair enzyme, prevents oxidant-induced AEC apoptosis. We reasoned that OGG1 deficiency augments asbestos-induced pulmonary fibrosis. Compared with intratracheal instillation of PBS (50 µl) or titanium dioxide (100 µg/50 µl), crocidolite or Libby amphibole asbestos (100 µg/50 µl) each augmented pulmonary fibrosis in wild-type C57BL/6J (WT) mice after 3 weeks as assessed by histology, fibrosis score, lung collagen via Sircol, and type 1 collagen expression; these effects persisted at 2 months. Compared with WT mice, Ogg1 homozygous knockout (Ogg1(-/-)) mice exhibit increased pulmonary fibrosis after crocidolite exposure and apoptosis in cells at the bronchoalveolar duct junctions as assessed via cleaved caspase-3 immunostaining. AEC involvement was verified by colocalization studies using surfactant protein C. Asbestos increased endoplasmic reticulum stress in the lungs of WT and Ogg1(-/-) mice. Compared with WT, alveolar type 2 cells isolated from Ogg1(-/-) mice have increased mtDNA damage, reduced mitochondrial aconitase expression, and increased P53 and cleaved caspase-9 expression, and these changes were enhanced 3 weeks after crocidolite exposure. These findings suggest an important role for AEC mtDNA integrity maintained by OGG1 in the pathogenesis of pulmonary fibrosis that may represent a novel therapeutic target.


Subject(s)
Alveolar Epithelial Cells/enzymology , Asbestos, Crocidolite/toxicity , DNA Glycosylases/metabolism , Pulmonary Fibrosis/enzymology , Alveolar Epithelial Cells/pathology , Animals , DNA Damage/genetics , DNA Glycosylases/genetics , DNA Glycosylases/immunology , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Humans , Mice , Mice, Knockout , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Time Factors
17.
Mol Cancer ; 13: 222, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25255877

ABSTRACT

BACKGROUND: The metastasis of hematogenous cancer cells is associated with abnormal glycosylation such as sialyl lewis antigens. Although the hepatitis B virus X protein (HBx) plays important role in liver disease, the precise function of HBx on aberrant glycosylation for metastasis remains unclear. METHODS: The human hepatocellular carcinoma tissues, HBx transgenic mice and HBx-transfected cells were used to check the correlation of expressions between HBx and Sialyl lewis antigen for cancer metastasis. To investigate whether expression levels of glycosyltransferases induced in HBx-transfected cells are specifically associated with sialyl lewis A (SLA) synthesis, which enhances metastasis by interaction of liver cancer cells with endothelial cells, ShRNA and siRNAs targeting specific glycosyltransferases were used. RESULTS: HBx expression in liver cancer region of HCC is associated with the specific synthesis of SLA. Furthermore, the SLA was specifically induced both in liver tissues from HBx-transgenic mice and in in vitro HBx-transfected cells. HBx increased transcription levels and activities of α2-3 sialyltransferases (ST3Gal III), α1-3/4 fucosyltransferases III and VII (FUT III and VII) genes, which were specific for SLA synthesis, allowing dramatic cell-cell adhesion for metastatic potential. Interestingly, HBx specifically induced expression of N-acetylglucosamine-ß1-3 galactosyltransferase V (ß1-3GalT 5) gene associated with the initial synthesis of sialyl lewis A, but not ß1-4GalT I. The ß1-3GalT 5 shRNA suppressed SLA expression by HBx, blocking the adhesion of HBx-transfected cells to the endothelial cells. Moreover, ß1-3GalT 5 silencing suppressed lung metastasis of HBx-transfected cells in in vivo lung metastasis system. CONCLUSION: HBx targets the specific glycosyltransferases for the SLA synthesis and this process regulates hematogenous cancer cell adhesion to endothelial cells for cancer metastasis.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Glycosyltransferases/metabolism , Liver Neoplasms/metabolism , Liver/metabolism , Liver/virology , Oligosaccharides/metabolism , Trans-Activators/metabolism , Adult , Animals , CA-19-9 Antigen , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Glycosylation , Hepatitis B virus/physiology , Human Umbilical Vein Endothelial Cells , Humans , Lewis X Antigen/metabolism , Liver/pathology , Liver Neoplasms/pathology , Liver Neoplasms/virology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Neoplasm Metastasis/pathology , Sialyl Lewis X Antigen , Viral Regulatory and Accessory Proteins
18.
PLoS One ; 9(5): e92786, 2014.
Article in English | MEDLINE | ID: mdl-24829158

ABSTRACT

Cisplatin (cis-diamminedichloroplatinum, CDDP) is a well-known chemotherapeutic agent for the treatment of several cancers. However, the precise mechanism underlying apoptosis of cancer cells induced by CDDP remains unclear. In this study, we show mechanistically that CDDP induces GM3-mediated apoptosis of HCT116 cells by inhibiting cell proliferation, and increasing DNA fragmentation and mitochondria-dependent apoptosis signals. CDDP induced apoptosis within cells through the generation of reactive oxygen species (ROS), regulated the ROS-mediated expression of Bax, Bcl-2, and p53, and induced the degradation of the poly (ADP-ribosyl) polymerase (PARP). We also checked expression levels of different gangliosides in HCT116 cells in the presence or absence of CDDP. Interestingly, among the gangliosides, CDDP augmented the expression of only GM3 synthase and its product GM3. Reduction of the GM3 synthase level through ectopic expression of GM3 small interfering RNA (siRNA) rescued HCT116 cells from CDDP-induced apoptosis. This was evidenced by inhibition of apoptotic signals by reducing ROS production through the regulation of 12-lipoxigenase activity. Furthermore, the apoptotic sensitivity to CDDP was remarkably increased in GM3 synthase-transfected HCT116 cells compared to that in controls. In addition, GM3 synthase-transfected cells treated with CDDP exhibited an increased accumulation of intracellular ROS. These results suggest the CDDP-induced oxidative apoptosis of HCT116 cells is mediated by GM3.


Subject(s)
Apoptosis/drug effects , Colon/drug effects , Epithelial Cells/drug effects , G(M3) Ganglioside/metabolism , Gene Expression Regulation, Neoplastic , Sialyltransferases/genetics , Antineoplastic Agents/pharmacology , Apoptosis/genetics , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 12-Lipoxygenase/metabolism , Cell Line, Tumor , Cell Proliferation , Cisplatin/pharmacology , Colon/metabolism , Colon/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Sialyltransferases/antagonists & inhibitors , Sialyltransferases/metabolism , Signal Transduction , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism
19.
J Biol Chem ; 289(9): 6165-76, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24429287

ABSTRACT

Mitochondria-targeted human 8-oxoguanine DNA glycosylase (mt-hOgg1) and aconitase-2 (Aco-2) each reduce oxidant-induced alveolar epithelial cell (AEC) apoptosis, but it is unclear whether protection occurs by preventing AEC mitochondrial DNA (mtDNA) damage. Using quantitative PCR-based measurements of mitochondrial and nuclear DNA damage, mtDNA damage was preferentially noted in AEC after exposure to oxidative stress (e.g. amosite asbestos (5-25 µg/cm(2)) or H2O2 (100-250 µM)) for 24 h. Overexpression of wild-type mt-hOgg1 or mt-long α/ß 317-323 hOgg1 mutant incapable of DNA repair (mt-hOgg1-Mut) each blocked A549 cell oxidant-induced mtDNA damage, mitochondrial p53 translocation, and intrinsic apoptosis as assessed by DNA fragmentation and cleaved caspase-9. In contrast, compared with controls, knockdown of Ogg1 (using Ogg1 shRNA in A549 cells or primary alveolar type 2 cells from ogg1(-/-) mice) augmented mtDNA lesions and intrinsic apoptosis at base line, and these effects were increased further after exposure to oxidative stress. Notably, overexpression of Aco-2 reduced oxidant-induced mtDNA lesions, mitochondrial p53 translocation, and apoptosis, whereas siRNA for Aco-2 (siAco-2) enhanced mtDNA damage, mitochondrial p53 translocation, and apoptosis. Finally, siAco-2 attenuated the protective effects of mt-hOgg1-Mut but not wild-type mt-hOgg1 against oxidant-induced mtDNA damage and apoptosis. Collectively, these data demonstrate a novel role for mt-hOgg1 and Aco-2 in preserving AEC mtDNA integrity, thereby preventing oxidant-induced mitochondrial dysfunction, p53 mitochondrial translocation, and intrinsic apoptosis. Furthermore, mt-hOgg1 chaperoning of Aco-2 in preventing oxidant-mediated mtDNA damage and apoptosis may afford an innovative target for the molecular events underlying oxidant-induced toxicity.


Subject(s)
Aconitate Hydratase/metabolism , DNA Damage , DNA Glycosylases/metabolism , DNA, Mitochondrial/metabolism , Epithelial Cells/enzymology , Mitochondria/enzymology , Oxidants/pharmacology , Oxidative Stress/drug effects , Pulmonary Alveoli/enzymology , Aconitate Hydratase/genetics , Animals , Apoptosis/drug effects , Asbestos, Amosite/toxicity , Cell Line, Tumor , DNA Glycosylases/genetics , DNA, Mitochondrial/genetics , Epithelial Cells/pathology , Humans , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/pathology , Mutation , Oxidants/adverse effects , Pulmonary Alveoli/pathology , Rats , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
20.
Int J Biochem Cell Biol ; 46: 32-8, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24120649

ABSTRACT

Vascular endothelial growth factor (VEGF) is well known as a significant angiogenic factor, and also functions as a proinflammatory cytokine, which induces adhesion of leukocyte to endothelial cells in inflammation reaction. In this study, we show that ganglioside GM3 inhibits the VEGF-induced expression of intracellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) through activation of nuclear factor-κB (NF-κB) via protein kinase B (AKT) signaling in human umbilical vein endothelial cells (HUVECs), relating with leukocyte recruitment to endothelial cells under inflammatory conditions. In addition, ganglioside GM3 significantly reduced the monocyte adhesion to HUVECs as determined by the monolayer cell adhesion assay. Furthermore, in VEGF-injected mice for the inflammatory condition, ganglioside GM3 markedly decreased the expression of ICAM-1 and VCAM-1 in vein tissues. These results suggest that ganglioside GM3 has an anti-inflammatory role by suppressing the expression of inflammatory-related molecules during in vitro and in vivo inflammation.


Subject(s)
Cell Adhesion/physiology , Cytokines/metabolism , Endothelial Cells/cytology , G(M3) Ganglioside/metabolism , Inflammation/metabolism , Monocytes/cytology , Animals , Cell Adhesion/drug effects , Cell Adhesion Molecules/metabolism , Cytokines/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Human Umbilical Vein Endothelial Cells , Humans , Inflammation/pathology , Intercellular Adhesion Molecule-1/biosynthesis , Mice , Mice, Inbred BALB C , Monocytes/metabolism , Signal Transduction , Vascular Cell Adhesion Molecule-1/biosynthesis , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...