Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
ERJ Open Res ; 5(3)2019 Jul.
Article in English | MEDLINE | ID: mdl-31423451

ABSTRACT

Idiopathic pulmonary fibrosis (IPF), the scarring of lung parenchyma resulting in the loss of lung function, remains a fatal disease with a significant unmet medical need. Patients with severe IPF often develop acute exacerbations resulting in the rapid deterioration of lung function, requiring transplantation. Understanding the pathophysiological mechanisms contributing to IPF is key to develop novel therapeutic approaches for end-stage disease. We report here RNA-sequencing analyses of lung tissues from a cohort of patients with transplant-stage IPF (n=36), compared with acute lung injury (ALI) (n=11) and nondisease controls (n=19), that reveal a robust gene expression signature unique to end-stage IPF. In addition to extracellular matrix remodelling pathways, we identified pathways associated with T-cell infiltration/activation, tumour development, and cholesterol homeostasis, as well as novel alternatively spliced transcripts that are differentially regulated in the advanced IPF lung versus ALI or nondisease controls. Additionally, we show a subset of genes that are correlated with percent predicted forced vital capacity and could reflect disease severity. Our results establish a robust transcriptomic fingerprint of an advanced IPF lung that is distinct from previously reported microarray signatures of moderate, stable or progressive IPF and identifies hitherto unknown candidate targets and pathways for therapeutic intervention in late-stage IPF as well as biomarkers to characterise disease progression and enable patient stratification.

2.
Am J Physiol Lung Cell Mol Physiol ; 316(2): L348-L357, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30489156

ABSTRACT

The translation of novel pulmonary fibrosis therapies from preclinical models into the clinic represents a major challenge demonstrated by the high attrition rate of compounds that showed efficacy in preclinical models but demonstrated no significant beneficial effects in clinical trials. A precision-cut lung tissue slice (PCLS) contains all major cell types of the lung and preserves the original cell-cell and cell-matrix contacts. It represents a promising ex vivo model to study pulmonary fibrosis. In this study, using RNA sequencing, we demonstrated that transforming growth factor-ß1 (TGFß1) induced robust fibrotic responses in the rat PCLS model, as it changed the expression of genes functionally related to extracellular matrix remodeling, cell adhesion, epithelial-to-mesenchymal transition, and various immune responses. Nintedanib, pirfenidone, and sorafenib each reversed a subset of genes modulated by TGFß1, and of those genes we identified 229 whose expression was reversed by all three drugs. These genes define a molecular signature characterizing many aspects of pulmonary fibrosis pathology and its attenuation in the rat PCLS fibrosis model. A panel of 12 genes and three secreted biomarkers, including procollagen I, hyaluronic acid, and WNT1-inducible signaling pathway protein 1 were validated as efficacy end points for the evaluation of antifibrotic activity of experimental compounds. Finally, we showed that blockade of αV-integrins suppressed TGFß1-induced fibrotic responses in the rat PCLS fibrosis model. Overall, our results suggest that the TGFß1-induced rat PCLS fibrosis model may represent a valuable system for target validation and to determine the efficacy of experimental compounds.


Subject(s)
Fibrosis/drug therapy , Indoles/pharmacology , Lung/drug effects , Pyridones/pharmacology , Animals , Biomarkers/metabolism , Collagen Type I/drug effects , Collagen Type I/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism
3.
Am J Physiol Gastrointest Liver Physiol ; 316(1): G15-G24, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30406699

ABSTRACT

Precision-cut liver tissue slice (PCLS) contains all major cell types of the liver parenchyma and preserves the original cell-cell and cell-matrix contacts. It represents a promising ex vivo model to study liver fibrosis and test the antifibrotic effect of experimental compounds in a physiological environment. In this study using RNA sequencing, we demonstrated that various pathways functionally related to fibrotic mechanisms were dysregulated in PCLSs derived from rats subjected to bile duct ligation. The activin receptor-like kinase-5 (Alk5) inhibitor SB525334, nintedanib, and sorafenib each reversed a subset of genes dysregulated in fibrotic PCLSs, and of those genes we identified 608 genes whose expression was reversed by all three compounds. These genes define a molecular signature characterizing many aspects of liver fibrosis pathology and its attenuation in the model. A panel of 12 genes and 4 secreted biomarkers including procollagen I, hyaluronic acid (HA), insulin-like growth factor binding protein 5 (IGFBP5), and WNT1-inducible signaling pathway protein 1 (WISP1) were further validated as efficacy end points for the evaluation of antifibrotic activity of experimental compounds. Finally, we showed that blockade of αV-integrins with a small molecule inhibitor attenuated the fibrotic phenotype in the model. Overall, our results suggest that the rat fibrotic PCLS model may represent a valuable system for target validation and determining the efficacy of experimental compounds. NEW & NOTEWORTHY We investigated the antifibrotic activity of three compounds, the activin receptor-like kinase-5 (Alk5) inhibitor SB525334, nintedanib, and sorafenib, in a rat fibrotic precision-cut liver tissue slice model using RNA sequencing analysis. A panel of 12 genes and 4 secreted biomarkers including procollagen I, hyaluronic acid (HA), insulin-like growth factor binding protein 5 (IGFBP5), and WNT1-inducible signaling pathway protein 1 (WISP1) were then established as efficacy end points to validate the antifibrotic activity of the αV-integrin inhibitor CWHM12. This study demonstrated the value of the rat fibrotic PCLS model for the evaluation of antifibrotic drugs.


Subject(s)
Imidazoles/pharmacology , Indoles/pharmacology , Liver Cirrhosis/drug therapy , Liver/drug effects , Quinoxalines/pharmacology , Animals , Biomarkers/metabolism , Collagen Type I/drug effects , Collagen Type I/genetics , Liver/metabolism , Liver Cirrhosis/metabolism , Male , Rats, Sprague-Dawley
4.
Biotechnol Bioeng ; 115(9): 2377-2382, 2018 09.
Article in English | MEDLINE | ID: mdl-29777592

ABSTRACT

This study reports findings of an unusual cluster of mutations spanning 22 bp (base pairs) in a monoclonal antibody expression vector. It was identified by two orthogonal methods: mass spectrometry on expressed protein and next-generation sequencing (NGS) on the plasmid DNA. While the initial NGS analysis confirmed the designed sequence modification, intact mass analysis detected an additional mass of the antibody molecule expressed in CHO cells. The extra mass was eventually found to be associated with unmatched nucleotides in a distal region by checking full-length sequence alignment plots. Interestingly, the complementary sequence of the mutated sequence was a reverse sequence of the original sequence and flanked by two 10-bp reverse-complementary sequences, leading to an undesirable DNA recombination. The finding highlights the necessity of rigorous examination of expression vector design and early monitoring of molecule integrity at both DNA and protein levels to prevent clones from having sequence variants during cell line development.


Subject(s)
Antibodies/metabolism , Genetic Vectors , Immunologic Factors/metabolism , Mutation , Recombinant Proteins/metabolism , Animals , Antibodies/chemistry , Antibodies/genetics , CHO Cells , Cricetulus , High-Throughput Nucleotide Sequencing , Immunologic Factors/chemistry , Immunologic Factors/genetics , Mass Spectrometry , Plasmids , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombination, Genetic
6.
Nat Protoc ; 7(6): 1024-41, 2012 May 03.
Article in English | MEDLINE | ID: mdl-22555242

ABSTRACT

Copy number variation (CNV) is increasingly recognized as an important contributor to phenotypic variation in health and disease. Most methods for determining CNV rely on admixtures of cells in which information regarding genetic heterogeneity is lost. Here we present a protocol that allows for the genome-wide copy number analysis of single nuclei isolated from mixed populations of cells. Single-nucleus sequencing (SNS), combines flow sorting of single nuclei on the basis of DNA content and whole-genome amplification (WGA); this is followed by next-generation sequencing to quantize genomic intervals in a genome-wide manner. Multiplexing of single cells is discussed. In addition, we outline informatic approaches that correct for biases inherent in the WGA procedure and allow for accurate determination of copy number profiles. All together, the protocol takes ∼3 d from flow cytometry to sequence-ready DNA libraries.


Subject(s)
DNA Copy Number Variations , Genetic Techniques , Single-Cell Analysis/methods , Algorithms , Cell Nucleus/genetics , Flow Cytometry , Genetic Heterogeneity , Humans
7.
Science ; 334(6055): 525-8, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-22034435

ABSTRACT

Germline mutations of the breast cancer 1 (BRCA1) gene are a major cause of familial breast and ovarian cancer. The BRCA1 protein displays E3 ubiquitin ligase activity, and this enzymatic function is thought to be required for tumor suppression. To test this hypothesis, we generated mice that express an enzymatically defective Brca1. We found that this mutant Brca1 prevents tumor formation to the same degree as does wild-type Brca1 in three different genetically engineered mouse (GEM) models of cancer. In contrast, a mutation that ablates phosphoprotein recognition by the BRCA C terminus (BRCT) domains of BRCA1 elicits tumors in each of the three GEM models. Thus, BRCT phosphoprotein recognition, but not the E3 ligase activity, is required for BRCA1 tumor suppression.


Subject(s)
BRCA1 Protein/metabolism , Genes, BRCA1 , Mammary Neoplasms, Experimental/genetics , Pancreatic Neoplasms/genetics , Phosphoproteins/metabolism , Animals , BRCA1 Protein/chemistry , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Cells, Cultured , Disease Models, Animal , Embryonic Stem Cells/metabolism , Ligands , Mammary Neoplasms, Experimental/metabolism , Mice , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Pancreatic Neoplasms/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , RING Finger Domains , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism
8.
DNA Cell Biol ; 29(3): 149-60, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20184449

ABSTRACT

Endothelial nitric oxide synthase (eNOS) is inhibited by hydrogen peroxide (H(2)O(2)), but the mechanism has not been determined. Thus, the purpose of this study was to delineate the mechanism by which H(2)O(2) inhibits eNOS activity. Using mass spectroscopy, we found that the tetrathiolate cysteine residues 94 and 99 were susceptible to oxidation by H(2)O(2). Molecular modeling predicted that these cysteic acid modifications would disrupt the van der Waals interactions and the hydrogen bonding network mediated by the tetrathiolate cysteines 94 and 99 resulting in changes in quaternary structure, zinc release, and dimer collapse. Using recombinant human eNOS (heNOS) to test the predictions of the molecular modeling we found that H(2)O(2) caused disruption of the heNOS dimer and this was accompanied by zinc release and decreased NO generation. We also found that H(2)O(2) increased the oxidation of tetrahydrobiopterin (BH(4)) to dihydrobiopterin (BH(2)), whereas preincubation of heNOS with excess BH(4) prevented the destruction of zinc tetrathiolate and dimer collapse and preserved activity. Interestingly, we found that the dimmer-stabilizing effect of BH(4) is due to its ability to act as a catalase mimetic. Further, we confirmed that, in ovine aortic endothelial cells, H(2)O(2) could also induce dimer collapse and that increasing cellular BH(4) levels could maintain eNOS in its dimeric form and NO signaling when cells were challenged with H(2)O(2). This study links the inhibitory action of H(2)O(2) on heNOS through the destruction of zinc tetrathiolate metal-binding site and dimer collapse both in vitro and in vivo.


Subject(s)
Hydrogen Peroxide/pharmacology , Models, Molecular , Nitric Oxide Synthase Type III/chemistry , Nitric Oxide Synthase Type III/metabolism , Protein Multimerization/drug effects , Zinc/metabolism , Amino Acid Sequence , Binding Sites/drug effects , Biopterins/analogs & derivatives , Biopterins/metabolism , Biopterins/pharmacology , Cysteic Acid/metabolism , Cysteine/metabolism , Humans , Mass Spectrometry , Molecular Sequence Data , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type III/antagonists & inhibitors , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Protein Structure, Quaternary/drug effects
9.
Genome Res ; 19(9): 1593-605, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19581485

ABSTRACT

DNA methylation stabilizes developmentally programmed gene expression states. Aberrant methylation is associated with disease progression and is a common feature of cancer genomes. Presently, few methods enable quantitative, large-scale, single-base resolution mapping of DNA methylation states in desired regions of a complex mammalian genome. Here, we present an approach that combines array-based hybrid selection and massively parallel bisulfite sequencing to profile DNA methylation in genomic regions spanning hundreds of thousands of bases. This single molecule strategy enables methylation variable positions to be quantitatively examined with high sampling precision. Using bisulfite capture, we assessed methylation patterns across 324 randomly selected CpG islands (CGI) representing more than 25,000 CpG sites. A single lane of Illumina sequencing permitted methylation states to be definitively called for >90% of target sties. The accuracy of the hybrid-selection approach was verified using conventional bisulfite capillary sequencing of cloned PCR products amplified from a subset of the selected regions. This confirmed that even partially methylated states could be successfully called. A comparison of human primary and cancer cells revealed multiple differentially methylated regions. More than 25% of islands showed complex methylation patterns either with partial methylation states defining the entire CGI or with contrasting methylation states appearing in specific regional blocks within the island. We observed that transitions in methylation state often correlate with genomic landmarks, including transcriptional start sites and intron-exon junctions. Methylation, along with specific histone marks, was enriched in exonic regions, suggesting that chromatin states can foreshadow the content of mature mRNAs.


Subject(s)
CpG Islands/genetics , DNA Methylation , Oligonucleotide Array Sequence Analysis/methods , Sequence Analysis, DNA/methods , Sulfites/chemistry , Breast Neoplasms/genetics , Cell Line, Tumor , Female , Gene Expression Profiling , Genome, Human , Humans , Polymorphism, Single Nucleotide , Skin Neoplasms/genetics
10.
Nucleic Acids Res ; 37(12): e89, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19474344

ABSTRACT

Methylation of CpG islands associated with genes can affect the expression of the proximal gene, and methylation of non-associated CpG islands correlates to genomic instability. This epigenetic modification has been shown to be important in many pathologies, from development and disease to cancer. We report the development of a novel high-resolution microarray that detects the methylation status of over 25,000 CpG islands in the human genome. Experiments were performed to demonstrate low system noise in the methodology and that the array probes have a high signal to noise ratio. Methylation measurements between different cell lines were validated demonstrating the accuracy of measurement. We then identified alterations in CpG islands, both those associated with gene promoters, as well as non-promoter-associated islands in a set of breast and ovarian tumors. We demonstrate that this methodology accurately identifies methylation profiles in cancer and in principle it can differentiate any CpG methylation alterations and can be adapted to analyze other species.


Subject(s)
CpG Islands , DNA Methylation , Oligonucleotide Array Sequence Analysis/methods , Cell Line , Genes, Neoplasm , Genome, Human , Humans
11.
Am J Physiol Lung Cell Mol Physiol ; 297(2): L309-17, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19447893

ABSTRACT

Our previous studies have demonstrated that nitric oxide (NO) leads to nitric oxide synthase (NOS) uncoupling and an increase in NOS-derived superoxide. However, the cause of this uncoupling has not been adequately resolved. The pteridine cofactor tetrahydrobiopterin (BH(4)) is a critical determinant of endothelial NOS (eNOS) activity and coupling, and GTP cyclohydrolase I (GCH1) is the rate-limiting enzyme in its generation. Thus the initial purpose of this study was to determine whether decreases in BH(4) could underlie, at least in part, the NO-mediated uncoupling of eNOS we have observed both in vitro and in vivo. Initially we evaluated the effect of inhaled NO levels on GCH1 expression and BH(4) levels in the intact lamb. Contrary to our hypothesis, we found that there was a significant increase in both plasma BH4 levels and peripheral lung GCH1 protein levels. Furthermore, in vitro, we found that exposure to the NO donor spermine NONOate (SPNONO) led to an increase in GCH1 protein and BH(4) levels in both COS-7 and pulmonary arterial endothelial cells. However, SPNONO treatment also caused a significant increase in phospho-cAMP response element binding protein (CREB) levels, as detected by Western blot analysis, and significantly increased cAMP levels, as detected by enzyme immunoassay. Furthermore, utilizing GCH1 promoter fragments fused to a luciferase reporter gene, we found that GCH1 promoter activity was enhanced by SPNONO in a CREB-dependent manner, and electromobility shift assays revealed an NO-dependent increase in the nuclear binding of CREB. These data suggest that NO increases BH(4) levels through a cAMP/CREB-mediated increase in GCH1 transcription and that the eNOS uncoupling associated with exogenous NO does not involved reduced BH(4) levels.


Subject(s)
Cyclic AMP/metabolism , GTP Cyclohydrolase/genetics , Gene Expression Regulation, Enzymologic/physiology , Nitric Oxide/pharmacokinetics , Respiratory Mucosa/enzymology , Animals , Biopterins/analogs & derivatives , Biopterins/blood , COS Cells , CREB-Binding Protein/metabolism , Chlorocebus aethiops , GTP Cyclohydrolase/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphorylation/physiology , Promoter Regions, Genetic/physiology , Respiratory Mucosa/cytology , Sheep , Signal Transduction/physiology , Superoxides/metabolism , Transcriptional Activation/physiology , Transfection
12.
DNA Cell Biol ; 27(1): 25-33, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17941803

ABSTRACT

S-nitrosylation, or the replacement of the hydrogen atom in the thiol group of cysteine residues by a -NO moiety, is a physiologically important posttranslational modification. In our previous work we have shown that S-nitrosylation is involved in the disruption of the endothelial nitric oxide synthase (eNOS) dimer and that this involves the disruption of the zinc (Zn) tetrathiolate cluster due to the S-nitrosylation of Cysteine 98. However, human eNOS contains 28 other cysteine residues whose potential to undergo S-nitrosylation has not been determined. Thus, the goal of this study was to identify the cysteine residues within eNOS that are susceptible to S-nitrosylation in vitro. To accomplish this, we utilized a modified biotin switch assay. Our modification included the tryptic digestion of the S-nitrosylated eNOS protein to allow the isolation of S-nitrosylated peptides for further identification by mass spectrometry. Our data indicate that multiple cysteine residues are capable of undergoing S-nitrosylation in the presence of an excess of a nitrosylating agent. All these cysteine residues identified were found to be located on the surface of the protein according to the available X-ray structure of the oxygenase domain of eNOS. Among those identified were Cys 93 and 98, the residues involved in the formation of the eNOS dimer through a Zn tetrathiolate cluster. In addition, cysteine residues within the reductase domain were identified as undergoing S-nitrosylation. We identified cysteines 660, 801, and 1113 as capable of undergoing S-nitrosylation. These cysteines are located within regions known to bind flavin mononucleotide (FMN), flavin adenine dinucleotide (FAD), and nicotinamide adenine dinucleotide (NADPH) although from our studies their functional significance is unclear. Finally we identified cysteines 852, 975/990, and 1047/1049 as being susceptible to S-nitrosylation. These cysteines are located in regions of eNOS that have not been implicated in any known biochemical functions and the significance of their S-nitrosylation is not clear from this study. Thus, our data indicate that the eNOS protein can be S-nitrosylated at multiple sites other than within the Zn tetrathiolate cluster, suggesting that S-nitrosylation may regulate eNOS function in ways other than simply by inducing dimer collapse.


Subject(s)
Cysteine/chemistry , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type III/chemistry , Sulfhydryl Compounds/chemistry , Crystallography, X-Ray , Humans , Mass Spectrometry , Nitric Oxide Donors/chemistry , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Nitro Compounds/chemistry , Oxygenases/chemistry , Peptide Fragments/analysis , Peptide Fragments/metabolism , Protein Structure, Tertiary , S-Nitroso-N-Acetylpenicillamine/chemistry , S-Nitroso-N-Acetylpenicillamine/pharmacology , S-Nitrosoglutathione/chemistry , S-Nitrosoglutathione/pharmacology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Sulfhydryl Compounds/metabolism
13.
Cell Cycle ; 6(24): 3132-44, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-18073535

ABSTRACT

The establishment of a vertebrate body plan during embryogenesis is achieved through precise coordination of cell proliferation and morphogenetic cell movements. Here we show that nitric oxide (NO) suppresses cell division and facilitates cell movements during early development of Xenopus, such that inhibition of NO synthase (NOS) increases proliferation in the neuroectoderm and suppresses convergent extension in the axial mesoderm and neuroectoderm. NO controls cell division and cell movement through two separate signaling pathways. Both rely on RhoA-ROCK signaling but can be distinguished by the involvement of either guanylate cyclase or the planar cell polarity regulator Dishevelled. Through the cGMP-dependent pathway, NO suppresses cell division by negatively regulating RhoA and controlling the nuclear distribution of ROCK and p21WAF1. Through the cGMP-independent pathway, NO facilitates cell movement by regulating the intracellular distribution and level of Dishevelled and the activity of RhoA, thereby controlling the activity of ROCK and regulating actin cytoskeleton remodeling and cell polarization. Concurrent control by NO helps ensure that the crucial processes of cell proliferation and morphogenetic movements are coordinated during early development.


Subject(s)
Cell Movement , Cell Proliferation , Nitric Oxide/physiology , Xenopus laevis/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cyclic GMP/metabolism , Dishevelled Proteins , Mesoderm/metabolism , Nitric Oxide Synthase/metabolism , Phosphoproteins/metabolism , Signal Transduction , Xenopus laevis/embryology , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
14.
Clin Hemorheol Microcirc ; 37(1-2): 111-20, 2007.
Article in English | MEDLINE | ID: mdl-17641401

ABSTRACT

An increasing number of studies implicate oxidative stress in the development of endothelial dysfunction and the pathogenesis of cardiovascular disease. Further, this oxidative stress has been shown to be associated with alterations in both the endothelin-1 (ET-1) and nitric oxide (NO) signaling pathways such that bioavailable NO is decreased and ET-1 signaling is potentiated. However, recent data, from our groups and others, have shown that oxidative stress, ET-1, and NO are co-regulated in a complex fashion that appears to be dependent on the cellular levels of each species. Thus, when ROS levels are transiently elevated, NO signaling is potentiated through transcriptional, post-transcriptional, and post-translational mechanisms. However, in pediatric pulmonary hypertensive disorders, when reactive oxygen species (ROS) increases are sustained by ET-1 mediated activation of smooth muscle cell ET(A) subtype receptors, NOS gene expression and NO signaling are reduced. Further, increases in oxidative stress can stimulate both the expression of the ET-1 gene and the secretion of the ET-1 peptide. Thus, this manuscript will review the available data regarding the interaction of NO, ET-1, and ROS in the endothelial dysfunction of pediatric pulmonary hypertension. In addition, we will suggest avenues of both basic and clinical research that will be important to develop novel pulmonary hypertension treatment and prevention strategies.


Subject(s)
Endothelin-1/physiology , Hypertension, Pulmonary/etiology , Nitric Oxide/physiology , Child , Endothelin-1/metabolism , Endothelium, Vascular/physiopathology , Humans , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Nitric Oxide/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism
15.
Nitric Oxide ; 13(3): 176-87, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16081307

ABSTRACT

Oxidative addition of a nitric oxide (NO) molecule to the thiol group of cysteine residues is a physiologically important post-translational modification that has been implicated in several metabolic and pathophysiological events. Our previous studies have indicated that S-nitrosylation can result in the disruption of the endothelial NO synthase (eNOS) dimer. It has been suggested that for S-nitrosylation to occur, the cysteine residue must be flanked by hydrophilic residues either in the primary structure or in the spatial proximity through appropriate conformation. However, this hypothesis has not been confirmed. Thus, the objective of this study was to determine if the nature of the amino acid residues that flank the cysteine in the primary structure has a significant effect on the rate and/or specificity of S-nitrosylation. To accomplish this, we utilized several model peptides based on the eNOS protein sequence. Some of these peptides contained point mutations to allow for different combinations of amino acid properties (acidic, basic, and hydrophobic) around the cysteine residue. To ensure that the results obtained were not dependent on the nitrosylation procedure, several common S-nitrosylation techniques were used and S-nitrosylation followed by mass spectrometric detection. Our data indicated that all peptides independent of the amino acids surrounding the cysteine residue underwent rapid S-nitrosylation. Thus, there does not appear to be a profound effect of the primary sequence of adjacent amino acid residues on the rate of cysteine S-nitrosylation at least at the peptide levels. Finally, our studies using recombinant human eNOS confirm that Cys98 undergoes S-nitrosylation. Thus, our data validate the importance of Cys98 in regulating eNOS dimerization and activity, and the utility of mass spectroscopy to identify cysteine residues susceptible to S-nitrosoylation.


Subject(s)
Cysteine/chemistry , Nitric Oxide Donors/chemistry , Nitric Oxide Synthase Type III/chemistry , Peptides/chemistry , Sulfhydryl Reagents/chemistry , Sulfhydryl Reagents/pharmacology , Amino Acid Sequence , Cysteine/metabolism , Humans , Molecular Sequence Data , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Nitrogen Oxides/chemistry , Nitrogen Oxides/pharmacology , Peptides/genetics , S-Nitroso-N-Acetylpenicillamine/chemistry , S-Nitroso-N-Acetylpenicillamine/pharmacology , S-Nitrosoglutathione/chemistry , S-Nitrosoglutathione/pharmacology , Sodium Nitrite/chemistry , Sodium Nitrite/pharmacology , Spectrometry, Mass, Electrospray Ionization , Spermine/analogs & derivatives , Spermine/chemistry , Spermine/pharmacology , Sulfhydryl Reagents/metabolism
16.
Brain Res Dev Brain Res ; 156(2): 183-92, 2005 May 12.
Article in English | MEDLINE | ID: mdl-16099305

ABSTRACT

The effect of hypoxia-ischemia on the nitric oxide synthase (NOS) cofactor tetrahydrobiopterin (BH4) and changes in the enzyme dimer state have not previously been studied. Cell-based studies have demonstrated the regulation of nitric oxide (NO) synthesis by intracellular BH4 levels. Activation of NOS requires two NOS polypeptides to form a homodimer. Dimerization results in the creation of high-affinity binding sites for BH4 and L-arginine. Our previous studies have indicated that nNOS activity falls 2 h post-hypoxia-ischemia in the immature rodent model. Thus, the objective of this study was to determine whether changes in nNOS dimeric state could be responsible for the decrease in nNOS activity. Using the immature rat model of HI in conjunction with LT-PAGE and Western blot analysis, we determined the effect of HI on NOS dimer state in hippocampus and cortex and the effects of pharmacologic modulation of NO levels during HI on dimer formation. Using high-performance liquid chromatography (HPLC) and electrospray tandem mass spectrometry (MS-MS), we measured BH4 and L-arginine levels respectively after HI under the same conditions. We found minimal or no changes in either BH4 levels or NOS dimer state at 2 h, 24 h and 7 day recovery from HI on postnatal day 7. In contrast, L-arginine levels were transiently increased in the hypoxic ischemic hemisphere. Thus, our data suggest that the previously described decrease in NOS activity after HI is not associated with depletion of the cofactor BH4, L-arginine substrate or changes in the NOS enzyme dimer state.


Subject(s)
Biopterins/analogs & derivatives , Hypoxia-Ischemia, Brain/metabolism , Nitric Oxide Synthase/metabolism , Analysis of Variance , Animals , Animals, Newborn , Arginine/metabolism , Biopterins/metabolism , Blotting, Western/methods , Brain/drug effects , Brain/metabolism , Brain Chemistry/drug effects , Brain Chemistry/physiology , Chromatography, High Pressure Liquid/methods , Electrochemistry/methods , Enzyme Inhibitors/pharmacology , Functional Laterality/drug effects , Functional Laterality/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Male , NG-Nitroarginine Methyl Ester/pharmacology , Rats , Rats, Wistar , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...