Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Free Radic Biol Med ; 210: 183-194, 2024 01.
Article in English | MEDLINE | ID: mdl-37979892

ABSTRACT

OBJECTIVE: Pulmonary hypertension (PH) is a progressive disease with vascular remodeling as a critical structural alteration. We have previously shown that metabolic reprogramming is an early initiating mechanism in animal models of PH. This metabolic dysregulation has been linked to remodeling the mitochondrial network to favor fission. However, whether the mitochondrial fission/fusion balance underlies the metabolic reprogramming found early in PH development is unknown. METHODS: Utilizing a rat early model of PH, in conjunction with cultured pulmonary endothelial cells (PECs), we utilized metabolic flux assays, Seahorse Bioassays, measurements of electron transport chain (ETC) complex activity, fluorescent microscopy, and molecular approaches to investigate the link between the disruption of mitochondrial dynamics and the early metabolic changes that occur in PH. RESULTS: We observed increased fusion mediators, including Mfn1, Mfn2, and Opa1, and unchanged fission mediators, including Drp1 and Fis1, in a two-week monocrotaline-induced PH animal model (early-stage PH). We were able to establish a connection between increases in fusion mediator Mfn1 and metabolic reprogramming. Using an adenoviral expression system to enhance Mfn1 levels in pulmonary endothelial cells and utilizing 13C-glucose labeled substrate, we found increased production of 13C lactate and decreased TCA cycle metabolites, revealing a Warburg phenotype. The use of a 13C5-glutamine substrate showed evidence that hyperfusion also induces oxidative carboxylation. The increase in glycolysis was linked to increased hypoxia-inducible factor 1α (HIF-1α) protein levels secondary to the disruption of cellular bioenergetics and higher levels of mitochondrial reactive oxygen species (mt-ROS). The elevation in mt-ROS correlated with attenuated ETC complexes I and III activities. Utilizing a mitochondrial-targeted antioxidant to suppress mt-ROS, limited HIF-1α protein levels, which reduced cellular glycolysis and reestablished mitochondrial membrane potential. CONCLUSIONS: Our data connects mitochondrial fusion-mediated mt-ROS to the Warburg phenotype in early-stage PH development.


Subject(s)
Hypertension, Pulmonary , Mitochondrial Dynamics , Rats , Animals , Mitochondrial Dynamics/genetics , Reactive Oxygen Species/metabolism , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Electron Transport , Endothelial Cells/metabolism , Lung/metabolism , Hypertension, Pulmonary/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism
2.
J Biol Chem ; 299(4): 103067, 2023 04.
Article in English | MEDLINE | ID: mdl-36841483

ABSTRACT

Mitochondrial fission and a Warburg phenotype of increased cellular glycolysis are involved in the pathogenesis of pulmonary hypertension (PH). The purpose of this study was to determine whether increases in mitochondrial fission are involved in a glycolytic switch in pulmonary arterial endothelial cells (PAECs). Mitochondrial fission is increased in PAEC isolated from a sheep model of PH induced by pulmonary overcirculation (Shunt PAEC). In Shunt PAEC we identified increases in the S616 phosphorylation responsible for dynamin-related protein 1 (Drp1) activation, the mitochondrial redistribution of Drp1, and increased cellular glycolysis. Reducing mitochondrial fission attenuated cellular glycolysis in Shunt PAEC. In addition, we observed nitration-mediated activation of the small GTPase RhoA in Shunt PAEC, and utilizing a nitration-shielding peptide, NipR1 attenuated RhoA nitration and reversed the Warburg phenotype. Thus, our data identify a novel link between RhoA, mitochondrial fission, and cellular glycolysis and suggest that targeting RhoA nitration could have therapeutic benefits for treating PH.


Subject(s)
Dynamins , Glycolysis , Hypertension, Pulmonary , Mitochondrial Dynamics , Monomeric GTP-Binding Proteins , rhoA GTP-Binding Protein , Animals , Dynamins/metabolism , Endothelial Cells/metabolism , Hypertension, Pulmonary/metabolism , Mitochondria/metabolism , Mitochondrial Dynamics/genetics , Monomeric GTP-Binding Proteins/metabolism , Sheep , Disease Models, Animal
3.
J Clin Invest ; 131(6)2021 03 15.
Article in English | MEDLINE | ID: mdl-33555272

ABSTRACT

T cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematologic malignancy with inferior outcome compared with that of B cell ALL. Here, we show that Runt-related transcription factor 2 (RUNX2) was upregulated in high-risk T-ALL with KMT2A rearrangements (KMT2A-R) or an immature immunophenotype. In KMT2A-R cells, we identified RUNX2 as a direct target of the KMT2A chimeras, where it reciprocally bound the KMT2A promoter, establishing a regulatory feed-forward mechanism. Notably, RUNX2 was required for survival of immature and KMT2A-R T-ALL cells in vitro and in vivo. We report direct transcriptional regulation of CXCR4 signaling by RUNX2, thereby promoting chemotaxis, adhesion, and homing to medullary and extramedullary sites. RUNX2 enabled these energy-demanding processes by increasing metabolic activity in T-ALL cells through positive regulation of both glycolysis and oxidative phosphorylation. Concurrently, RUNX2 upregulation increased mitochondrial dynamics and biogenesis in T-ALL cells. Finally, as a proof of concept, we demonstrate that immature and KMT2A-R T-ALL cells were vulnerable to pharmacological targeting of the interaction between RUNX2 and its cofactor CBFß. In conclusion, we show that RUNX2 acts as a dependency factor in high-risk subtypes of human T-ALL through concomitant regulation of tumor metabolism and leukemic cell migration.


Subject(s)
Core Binding Factor Alpha 1 Subunit/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Animals , Cell Line, Tumor , Chemotaxis, Leukocyte , Child , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor beta Subunit/metabolism , Disease Progression , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Gene Rearrangement , Hematopoiesis , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , In Vitro Techniques , Mice , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Organelle Biogenesis , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, CXCR4/metabolism , Signal Transduction
4.
Redox Biol ; 36: 101679, 2020 09.
Article in English | MEDLINE | ID: mdl-32818797

ABSTRACT

The autophagic pathway involves the encapsulation of substrates in double-membraned vesicles, which are subsequently delivered to the lysosome for enzymatic degradation and recycling of metabolic precursors. Autophagy is a major cellular defense against oxidative stress, or related conditions that cause accumulation of damaged proteins or organelles. Selective forms of autophagy can maintain organelle populations or remove aggregated proteins. Dysregulation of redox homeostasis under pathological conditions results in excessive generation of reactive oxygen species (ROS), leading to oxidative stress and the associated oxidative damage of cellular components. Accumulating evidence indicates that autophagy is necessary to maintain redox homeostasis. ROS activates autophagy, which facilitates cellular adaptation and diminishes oxidative damage by degrading and recycling intracellular damaged macromolecules and dysfunctional organelles. The cellular responses triggered by oxidative stress include the altered regulation of signaling pathways that culminate in the regulation of autophagy. Current research suggests a central role for autophagy as a mammalian oxidative stress response and its interrelationship to other stress defense systems. Altered autophagy phenotypes have been observed in lung diseases such as chronic obstructive lung disease, acute lung injury, cystic fibrosis, idiopathic pulmonary fibrosis, and pulmonary arterial hypertension, and asthma. Understanding the mechanisms by which ROS regulate autophagy will provide novel therapeutic targets for lung diseases. This review highlights our current understanding on the interplay between ROS and autophagy in the development of pulmonary disease.


Subject(s)
Autophagy , Lung Diseases , Animals , Oxidation-Reduction , Oxidative Stress , Reactive Oxygen Species
5.
Cancer Sci ; 110(6): 1931-1946, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30974024

ABSTRACT

Activating mutations in cytokine receptors and transcriptional regulators govern aberrant signal transduction in T-cell lineage acute lymphoblastic leukemia (T-ALL). However, the roles played by suppressors of cytokine signaling remain incompletely understood. We examined the regulatory roles of suppressor of cytokine signaling 5 (SOCS5) in T-ALL cellular signaling networks and leukemia progression. We found that SOCS5 was differentially expressed in primary T-ALL and its expression levels were lowered in HOXA-deregulated leukemia harboring KMT2A gene rearrangements. Here, we report that SOCS5 expression is epigenetically regulated by DNA methyltransferase-3A-mediated DNA methylation and methyl CpG binding protein-2-mediated histone deacetylation. We show that SOCS5 negatively regulates T-ALL cell growth and cell cycle progression but has no effect on apoptotic cell death. Mechanistically, SOCS5 silencing induces activation of JAK-STAT signaling, and negatively regulates interleukin-7 and interleukin-4 receptors. Using a human T-ALL murine xenograft model, we show that genetic inactivation of SOCS5 accelerates leukemia engraftment and progression, and leukemia burden. We postulate that SOCS5 is epigenetically deregulated in T-ALL and serves as an important regulator of T-ALL cell proliferation and leukemic progression. Our results link aberrant downregulation of SOCS5 expression to the enhanced activation of the JAK-STAT and cytokine receptor-signaling cascade in T-ALL.


Subject(s)
Epigenesis, Genetic , Janus Kinases/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , STAT Transcription Factors/genetics , Suppressor of Cytokine Signaling Proteins/genetics , Animals , Cell Line , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Disease Progression , Gene Expression Profiling , Humans , Janus Kinases/metabolism , Jurkat Cells , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , RNAi Therapeutics/methods , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , STAT Transcription Factors/metabolism , Signal Transduction/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Survival Analysis , Xenograft Model Antitumor Assays/methods
6.
Antioxid Redox Signal ; 31(12): 819-842, 2019 10 20.
Article in English | MEDLINE | ID: mdl-30623676

ABSTRACT

Significance: Oxidative stress in the cell is characterized by excessive generation of reactive oxygen species (ROS). Superoxide (O2-) and hydrogen peroxide (H2O2) are the main ROS involved in the regulation of cellular metabolism. As our fundamental understanding of the underlying causes of lung disease has increased it has become evident that oxidative stress plays a critical role. Recent Advances: A number of cells in the lung both produce, and respond to, ROS. These include vascular endothelial and smooth muscle cells, fibroblasts, and epithelial cells as well as the cells involved in the inflammatory response, including macrophages, neutrophils, eosinophils. The redox system is involved in multiple aspects of cell metabolism and cell homeostasis. Critical Issues: Dysregulation of the cellular redox system has consequential effects on cell signaling pathways that are intimately involved in disease progression. The lung is exposed to biomechanical forces (fluid shear stress, cyclic stretch, and pressure) due to the passage of blood through the pulmonary vessels and the distension of the lungs during the breathing cycle. Cells within the lung respond to these forces by activating signal transduction pathways that alter their redox state with both physiologic and pathologic consequences. Future Directions: Here, we will discuss the intimate relationship between biomechanical forces and redox signaling and its role in the development of pulmonary disease. An understanding of the molecular mechanisms induced by biomechanical forces in the pulmonary vasculature is necessary for the development of new therapeutic strategies.


Subject(s)
Lung Diseases/physiopathology , Reactive Oxygen Species/metabolism , Vascular Diseases/physiopathology , Animals , Biomechanical Phenomena , Humans , Lung Diseases/metabolism , Mitochondria/metabolism , Oxidative Stress , Signal Transduction , Vascular Diseases/metabolism
7.
Immunity ; 37(2): 223-34, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22921120

ABSTRACT

Autophagy is a fundamental biological process of the eukaryotic cell contributing to diverse cellular and physiological functions including cell-autonomous defense against intracellular pathogens. Here, we screened the Rab family of membrane trafficking regulators for effects on autophagic elimination of Mycobacterium tuberculosis var. bovis BCG and found that Rab8b and its downstream interacting partner, innate immunity regulator TBK-1, are required for autophagic elimination of mycobacteria in macrophages. TBK-1 was necessary for autophagic maturation. TBK-1 coordinated assembly and function of the autophagic machinery and phosphorylated the autophagic adaptor p62 (sequestosome 1) on Ser-403, a residue essential for its role in autophagic clearance. A key proinflammatory cytokine, IL-1ß, induced autophagy leading to autophagic killing of mycobacteria in macrophages, and this IL-1ß activity was dependent on TBK-1. Thus, TBK-1 is a key regulator of immunological autophagy and is responsible for the maturation of autophagosomes into lytic bactericidal organelles.


Subject(s)
Autophagy/immunology , Macrophages/immunology , Protein Serine-Threonine Kinases/immunology , rab GTP-Binding Proteins/immunology , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autophagy-Related Protein 7 , Green Fluorescent Proteins , HeLa Cells , Heat-Shock Proteins/immunology , Heat-Shock Proteins/metabolism , Humans , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Microscopy, Confocal , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , Mycobacterium bovis/immunology , Phagosomes/drug effects , Phagosomes/immunology , Phagosomes/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering , Sequestosome-1 Protein , Serine/immunology , Serine/metabolism , Tuberculosis/immunology , rab GTP-Binding Proteins/genetics
8.
EMBO J ; 30(23): 4701-11, 2011 Nov 08.
Article in English | MEDLINE | ID: mdl-22068051

ABSTRACT

Autophagy controls the quality and quantity of the eukaryotic cytoplasm while performing two evolutionarily highly conserved functions: cell-autonomous provision of energy and nutrients by cytosol autodigestion during starvation, and removal of defunct organelles and large aggregates exceeding the capacity of other cellular degradative systems. In contrast to these autodigestive processes, autophagy in yeast has additional, biogenesis functions. However, no equivalent biosynthetic roles have been described for autophagy in mammals. Here, we show that in mammalian cells, autophagy has a hitherto unappreciated positive contribution to the biogenesis and secretion of the proinflammatory cytokine IL-1ß via an export pathway that depends on Atg5, inflammasome, at least one of the two mammalian Golgi reassembly stacking protein (GRASP) paralogues, GRASP55 (GORASP2) and Rab8a. This process, which is a type of unconventional secretion, expands the functional manifestations of autophagy beyond autodigestive and quality control roles in mammals. It enables a subset of cytosolic proteins devoid of signal peptide sequences, and thus unable to access the conventional pathway through the ER, to enter an autophagy-based secretory pathway facilitating their exit from the cytoplasm.


Subject(s)
Autophagy/physiology , Carrier Proteins/metabolism , Interleukin-1beta , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Autophagy/drug effects , Autophagy-Related Protein 5 , Cells, Cultured , HMGB1 Protein/metabolism , Inflammasomes/metabolism , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins , Luminescent Proteins/metabolism , Macrolides/pharmacology , Macrophages/metabolism , Mice , Nigericin/pharmacology , Protein Processing, Post-Translational/drug effects , Protein Transport/drug effects , Secretory Pathway/drug effects , Secretory Pathway/physiology
9.
Nat Cell Biol ; 12(12): 1154-65, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21102437

ABSTRACT

IRGM, a human immunity-related GTPase, confers autophagic defence against intracellular pathogens by an unknown mechanism. Here, we report an unexpected mode of IRGM action. IRGM demonstrated differential affinity for the mitochondrial lipid cardiolipin, translocated to mitochondria, affected mitochondrial fission and induced autophagy. Mitochondrial fission was necessary for autophagic control of intracellular mycobacteria by IRGM. IRGM influenced mitochondrial membrane polarization and cell death. Overexpression of IRGMd, but not IRGMb splice isoforms, caused mitochondrial depolarization and autophagy-independent, but Bax/Bak-dependent, cell death. By acting on mitochondria, IRGM confers autophagic protection or cell death, explaining IRGM action both in defence against tuberculosis and in the damaging inflammation caused by Crohn's disease.


Subject(s)
Autophagy , GTP-Binding Proteins/metabolism , Mitochondria/metabolism , Animals , Cardiolipins/metabolism , Cell Line , Dynamins , GTP Phosphohydrolases/metabolism , GTP-Binding Proteins/analysis , Humans , Mice , Microtubule-Associated Proteins/metabolism , Mitochondria/chemistry , Mitochondrial Proteins/metabolism , Protein Isoforms/metabolism
10.
Immunol Rev ; 227(1): 189-202, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19120485

ABSTRACT

Autophagy is a physiologically and immunologically controlled intracellular homeostatic pathway that sequesters and degrades cytoplasmic targets including macromolecular aggregates, cellular organelles such as mitochondria, and whole microbes or their products. Recent advances show that autophagy plays a role in innate immunity in several ways: (i) direct elimination of intracellular microbes by digestion in autolysosomes, (ii) delivery of cytosolic microbial products to pattern recognition receptors (PRRs) in a process referred to as topological inversion, and (iii) as an anti-microbial effector of Toll-like receptors and other PRR signaling. Autophagy eliminates pathogens in vitro and in vivo but, when aberrant due to mutations, contributes to human inflammatory disorders such as Crohn's disease. In this review, we examine these relationships and propose that autophagy is one of the most ancient innate immune defenses that has possibly evolved at the time of alpha-protobacteria-pre-eukaryote relationships, leading up to modern eukaryotic cell-mitochondrial symbiosis, and that during the metazoan evolution, additional layers of immunological regulation have been superimposed and integrated with this primordial innate immunity mechanism.


Subject(s)
Autophagy/immunology , GTP-Binding Proteins/immunology , Immunity, Innate , Nod Signaling Adaptor Proteins/metabolism , Toll-Like Receptors/metabolism , Animals , Antigen Presentation/immunology , Autophagy/genetics , Crohn Disease/genetics , Crohn Disease/immunology , Cytokines/genetics , Cytokines/metabolism , Evolution, Molecular , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Humans , Infections/immunology , MAP Kinase Signaling System/immunology , Mitochondria/immunology , Nod Signaling Adaptor Proteins/immunology , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Toll-Like Receptors/immunology
11.
J Clin Invest ; 117(11): 3489-97, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17948127

ABSTRACT

Progressive pulmonary disease and infections with Pseudomonas aeruginosa remain an intractable problem in cystic fibrosis (CF). At the cellular level, CF is characterized by organellar hyperacidification, which results in altered protein and lipid glycosylation. Altered pH of the trans-Golgi network (TGN) may further disrupt the protein processing and packaging that occurs in this organelle. Here we measured activity of the major TGN endoprotease furin and demonstrated a marked upregulation in human CF cells. Increased furin activity was linked to elevated production in CF of the immunosuppressive and tissue remodeling cytokine TGF-beta and its downstream effects, including macrophage deactivation and augmented collagen secretion by epithelial cells. As furin is responsible for the proteolytic processing of a range of endogenous and exogenous substrates including growth factors and bacterial toxins, we determined that elevated furin-dependent activation of exotoxin A caused increased cell death in CF respiratory epithelial cells compared with genetically matched CF transmembrane conductance regulator-corrected cells. Thus elevated furin levels in CF respiratory epithelial cells contributes to bacterial toxin-induced cell death, fibrosis, and local immunosuppression. These data suggest that the use of furin inhibitors may represent a strategy for pharmacotherapy in CF.


Subject(s)
ADP Ribose Transferases/toxicity , Bacterial Toxins/toxicity , Cystic Fibrosis/metabolism , Exotoxins/toxicity , Furin/metabolism , Respiratory Mucosa/metabolism , Virulence Factors/toxicity , Cell Line , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Feedback, Physiological , Furin/antagonists & inhibitors , Furin/genetics , Humans , Macrophages/metabolism , Nitric Oxide Synthase Type II/metabolism , Respiratory Mucosa/cytology , Transforming Growth Factor beta/metabolism , trans-Golgi Network/enzymology , Pseudomonas aeruginosa Exotoxin A
12.
Am J Physiol Lung Cell Mol Physiol ; 293(3): L712-9, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17586695

ABSTRACT

The CFTR gene encodes a chloride channel with pleiotropic effects on cell physiology and metabolism. Here, we show that increasing cGMP levels to inhibit epithelial Na(+) channel in cystic fibrosis (CF) respiratory epithelial cells corrects several aspects of the downstream pathology in CF. Cell culture models, using a range of CF cell lines and primary cells, showed that complementary pharmacological approaches to increasing intracellular cGMP, by elevating guanyl cyclase activity though reduced nitric oxide, addition of cell-permeable cGMP analogs, or inhibition of phosphodiesterase 5 corrected multiple aspects of the CF pathological cascade. These included correction of defective protein glycosylation, bacterial adherence, and proinflammatory responses. Furthermore, pharmacological inhibition of phosphodiesterase 5 in tissues ex vivo or in animal models improved transepithelial currents across nasal mucosae from transgenic F508del Cftr(tm1Eur) mice and reduced neutrophil infiltration on bacterial aerosol challenge in Pseudomonas aeruginosa-susceptible DBA/2 mice. Our findings define phosphodiesterase 5 as a specific target for correcting a number of previously disconnected defects in the CF respiratory tract, now linked through this study. Our study suggests that phosphodiesterase 5 inhibition provides an opportunity for simultaneous and concerted correction of seemingly disparate complications in CF.


Subject(s)
3',5'-Cyclic-GMP Phosphodiesterases/antagonists & inhibitors , Cyclic GMP/metabolism , Cystic Fibrosis/drug therapy , Piperazines/therapeutic use , Respiratory System/pathology , Sulfones/therapeutic use , Animals , Cell Line , Cells, Cultured , Cyclic Nucleotide Phosphodiesterases, Type 5 , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Epithelial Cells/pathology , Epithelial Sodium Channels/metabolism , Guanylate Cyclase/metabolism , Humans , Hydrogen-Ion Concentration/drug effects , In Vitro Techniques , Ion Transport/drug effects , Mice , Mice, Transgenic , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Piperazines/pharmacology , Purines/pharmacology , Purines/therapeutic use , Respiratory System/drug effects , Signal Transduction/drug effects , Sildenafil Citrate , Sodium/metabolism , Sulfones/pharmacology , trans-Golgi Network/drug effects , trans-Golgi Network/metabolism
13.
Free Radic Res ; 41(2): 208-15, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17364947

ABSTRACT

Alginate is a critical virulence factor contributing to the poor clinical prognosis associated with the conversion of Pseudomonas aeruginosa to mucoid phenotypes in cystic fibrosis (CF). An important mechanism of action is its ability to scavenge host innate-immune reactive species. We have previously analyzed the bacterial response to nitrosative stress by S-nitrosoglutathione (GSNO), a physiological NO radical donor with diminished levels in the CF lung. GSNO substantially increased bacterial nitrosative and oxidative defenses and so we hypothesized a similar increase in alginate production would occur. However, in mucoid P. aeruginosa, there was decreased expression of the majority of alginate synthetic genes. This microarray data was confirmed both by RT-PCR and at the functional level by direct measurements of alginate production. Our data suggest that the lowered levels of innate-immune nitrosative mediators (such as GSNO) in the CF lung exacerbate the effects of mucoid P. aeruginosa, by failing to suppress alginate biosynthesis.


Subject(s)
Cystic Fibrosis/microbiology , Glycosaminoglycans/metabolism , Lung/microbiology , Nitric Oxide Donors/pharmacology , Nitric Oxide/metabolism , Pseudomonas aeruginosa/pathogenicity , S-Nitrosoglutathione/pharmacology , Alginates , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Cells, Cultured/drug effects , Cells, Cultured/enzymology , Cells, Cultured/microbiology , Cystic Fibrosis/pathology , Gene Expression Profiling , Gene Expression Regulation, Bacterial/drug effects , Glucuronic Acid/biosynthesis , Hexuronic Acids , Humans , Lung/pathology , Nitric Oxide Synthase Type II/deficiency , Nitric Oxide Synthase Type II/metabolism , Nitrosation , Oligonucleotide Array Sequence Analysis , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Virulence/drug effects
14.
Pediatr Pulmonol ; 41(8): 771-8, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16779853

ABSTRACT

Cystic fibrosis (CF) remains a fatal progressive disease in spite of the discovery and characterization of the CFTR gene. Transforming growth factor beta (TGF-beta) has been implicated in pathophysiology of CF. Previous reports have shown the trans-Golgi network (TGN) is hyperacdified in CF epithelial cells in culture and that this hyperacidification can be corrected with the membrane permeant weak base, chloroquine. In this study bioactive TGF-beta produced by CF and normal cells was measured using a reporter cell line with a TGF-beta responsive promoter linked to luciferase. Increased levels of TGF-beta were detected in the conditioned media from CF epithelial cells compared to their matched controls-(IB3-1 vs. S9; pCEP-R vs. pCEP, CuFi-4 vs. NuLi-1). Levels of TGF-beta were normalized with chloroquine indicating that the hyperacidification of the TGN of CF cells is responsible for the altered TGF-beta levels.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Bronchi/cytology , Chloroquine/pharmacology , Cystic Fibrosis/physiopathology , Epithelial Cells/metabolism , Lung/cytology , Transforming Growth Factor beta/blood , Cells, Cultured , Culture Media, Conditioned , Cystic Fibrosis/blood , Humans , trans-Golgi Network/metabolism
15.
EMBO Rep ; 7(5): 553-9, 2006 May.
Article in English | MEDLINE | ID: mdl-16612392

ABSTRACT

Endosomal hyperacidification in cystic fibrosis (CF) respiratory epithelial cells is secondary to a loss of sodium transport control owing to a defective form of the CF transmembrane conductance regulator CFTR. Here, we show that endosomal hyperacidification can be corrected by activating the signalling cascade controlling sodium channels through cyclic GMP. Nitric oxide (NO) donors corrected the endosomal hyperacidification in CF cells. Stimulation of CF cells with guanylate cyclase agonists corrected the pH in endosomes. Exposure of CF cells to an inhibitor of cGMP-specific phosphodiesterase PDE5, Sildenafil, normalized the endosomal pH. Treatment with Sildenafil reduced secretion by CF cells of the proinflammatory chemokine interleukin 8 following stimulation with Pseudomonas aeruginosa products. Thus, the endosomal hyperacidification and excessive proinflammatory response in CF are in part due to deficiencies in NO- and cGMP-regulated processes and can be pharmacologically reversed using PDE5 inhibitors.


Subject(s)
Cyclic GMP/deficiency , Cystic Fibrosis/metabolism , Endosomes/metabolism , Nitric Oxide/deficiency , Respiratory Mucosa/metabolism , Signal Transduction/physiology , 3',5'-Cyclic-GMP Phosphodiesterases/antagonists & inhibitors , 3',5'-Cyclic-GMP Phosphodiesterases/metabolism , Cells, Cultured , Cyclic GMP/physiology , Cystic Fibrosis/enzymology , Cystic Fibrosis/pathology , Endosomes/drug effects , Endosomes/enzymology , Endosomes/pathology , Humans , Hydrogen-Ion Concentration/drug effects , Nitric Oxide/physiology , Piperazines/pharmacology , Purines/pharmacology , Respiratory Mucosa/drug effects , Respiratory Mucosa/enzymology , Respiratory Mucosa/pathology , Signal Transduction/drug effects , Sildenafil Citrate , Sulfones/pharmacology
16.
Infect Immun ; 72(9): 5012-8, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15321993

ABSTRACT

The main cause of the high morbidity and mortality of cystic fibrosis (CF) is the progressive lung inflammation associated with Pseudomonas aeruginosa colonization. During the course of chronic CF infections, P. aeruginosa undergoes a conversion to a mucoid phenotype. The emergence of mucoid P. aeruginosa in CF is associated with increased inflammation, respiratory decline, and a poor prognosis. Here we show, by the use of microarray analysis, that upon P. aeruginosa conversion to mucoidy, there is a massive and preferential induction of genes encoding bacterial lipoproteins. Bacterial lipoproteins are potent agonists of Toll-like receptor 2 (TLR2) signaling. The expression of TLR2 in human respiratory epithelial cells was ascertained by Western blot analysis. Human respiratory epithelial cells responded in a TLR2-dependent manner to bacterial lipopeptides derived from Pseudomonas lipoproteins induced in mucoid strains. The TLR2 proinflammatory response was further augmented in CF cells. Thus, the excessive inflammation in CF is the result of a global induction in mucoid P. aeruginosa of lipoproteins that act as proinflammatory toxins (here termed lipotoxins) superimposed on the hyperexcitability of CF cells. Blocking the signaling cascade responding to bacterial lipotoxins may provide therapeutic benefits for CF patients.


Subject(s)
Cystic Fibrosis/physiopathology , Gene Expression Regulation, Bacterial , Inflammation/physiopathology , Lipoproteins/metabolism , Oligonucleotide Array Sequence Analysis , Pseudomonas aeruginosa/pathogenicity , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bronchi/cytology , Cell Line , Cells, Cultured , Cystic Fibrosis/microbiology , Epithelial Cells/microbiology , Gene Expression Profiling , Humans , Inflammation/microbiology , Lipoproteins/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Molecular Sequence Data , NF-kappa B/genetics , NF-kappa B/metabolism , Pseudomonas Infections/microbiology , Pseudomonas Infections/physiopathology , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/growth & development , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Toll-Like Receptor 2 , Toll-Like Receptors , Virulence
17.
J Bacteriol ; 186(12): 4046-50, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15175322

ABSTRACT

The type strain of Pseudomonas aeruginosa, PAO1, showed great upregulation of many nitrosative defense genes upon treatment with S-nitrosoglutathione, while the mucoid strain PAO578II showed no further upregulation above its constitutive upregulation of nor and fhp. NO* consumption however, showed that both strains mount functional, protein synthesis-dependent NO*-consumptive responses.


Subject(s)
Bacterial Proteins/metabolism , Heat-Shock Response , Oligonucleotide Array Sequence Analysis/methods , Pseudomonas aeruginosa/growth & development , Pseudomonas aeruginosa/physiology , S-Nitrosoglutathione/pharmacology , Bacterial Proteins/genetics , Gene Expression Profiling , Humans , Nitric Oxide/metabolism , Pseudomonas aeruginosa/genetics , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...