Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Anal Biochem ; 547: 66-76, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29470948

ABSTRACT

Phosphatidylinositol (3,4,5) trisphosphate (PIP3) is a biologically active membrane phospholipid that is essential for the growth and survival of all eukaryotic cells. We describe a new method that directly measures PIP3 and describe the HPLC separation and measurement of the positional isomers of phosphatidylinositol bisphosphate, PI(3,5)P2, PI(3,4)P2 and PI(4,5)P2. Mass spectrometric analyses were performed online using ultra-high performance liquid chromatography (UHPLC)-electrospray ionization tandem mass spectrometry (LC-ESI-MS/MS) in the negative multiple-reaction monitoring (MRM) modes. Rapid separation of PIP3 from PI, phosphatidylinositol phosphate (PIP) and PIP2 was accomplished by C18 reverse phase chromatography with the addition of the ion pairing reagents diisopropylethanolamine (DiiPEA) and ethylenediamine tetraacetic acid tetrasodium salt dihydrate (EDTA) to the samples and mobile phase with a total run time, including equilibration, of 12 min. Importantly, these chromatography conditions result in no carryover of PIP, PIP2, and PIP3 between samples. To validate the new method, U87MG cancer cells were serum starved and treated with PDGF to stimulate PIP3 biosynthesis in the presence or absence of the phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002. Results generated with the LC/MS method were in excellent agreement with results generated using [33P] phosphate radiolabeled U87MG cells and anion exchange chromatography analysis, a well validated method for measuring PIP3. To demonstrate the usefulness of the new method, we generated reproducible IC50 data for several well-characterized PI3K small molecule inhibitors using a U87MG cell-based assay as well as showing PIP3 can be measured from additional cancer cell lines. Together, our results demonstrate this novel method is sensitive, reproducible and can be used to directly measure PIP3 without radiolabeling or complex lipid derivatization.


Subject(s)
Phosphatidylinositol Phosphates/analysis , Phosphatidylinositol Phosphates/metabolism , Spectrometry, Mass, Electrospray Ionization/methods , Cell Line, Tumor , Chromatography, Liquid/methods , Humans
2.
Mol Microbiol ; 73(4): 680-94, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19656291

ABSTRACT

The Escherichia coli NsrR protein is a nitric oxide-sensitive repressor of transcription. The NsrR-binding site is predicted to comprise two copies of an 11 bp motif arranged as an inverted repeat with 1 bp spacing. By mutagenesis we confirmed that both 11 bp motifs are required for maximal NsrR repression of the ytfE promoter. We used chromatin immunoprecipitation and microarray analysis (ChIP-chip) to show that NsrR binds to 62 sites close to the 5' ends of genes. Analysis of the ChIP-chip data suggested that a single 11 bp motif (with the consensus sequence AANATGCATTT) can function as an NsrR-binding site in vivo. NsrR binds to sites in the promoter regions of the fliAZY, fliLMNOPQR and mqsR-ygiT transcription units, which encode proteins involved in motility and biofilm development. Reporter fusion assays confirmed that NsrR negatively regulates the fliA and fliL promoters. A mutation in the predicted 11 bp NsrR-binding site in the fliA promoter impaired repression by NsrR and prevented detectable binding in vivo. Assays on soft-agar confirmed that NsrR is a negative regulator of motility in E. coli K12 and in a uropathogenic strain; surface attachment assays revealed decreased levels of attached growth in the absence of NsrR.


Subject(s)
DNA-Binding Proteins/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/genetics , Transcription Factors/metabolism , Base Sequence , Binding Sites , Chromatin Immunoprecipitation , DNA Mutational Analysis , DNA, Bacterial/genetics , DNA-Binding Proteins/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial , Molecular Sequence Data , Mutagenesis , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics
3.
EMBO Mol Med ; 12(6): e11164, 2020 06 08.
Article in English | MEDLINE | ID: mdl-32302470

ABSTRACT

The angiopoietin (Ang)-Tie pathway has been intensely pursued as candidate second-generation anti-angiogenic target. While much of the translational work has focused on the ligand Ang2, the clinical efficacy of Ang2-targeting drugs is limited and failed to improve patient survival. In turn, the orphan receptor Tie1 remains therapeutically unexplored, although its endothelial-specific genetic deletion has previously been shown to result in a strong reduction in metastatic growth. Here, we report a novel Tie1 function-blocking antibody (AB-Tie1-39), which suppressed postnatal retinal angiogenesis. During primary tumor growth, neoadjuvant administration of AB-Tie1-39 strongly impeded systemic metastasis. Furthermore, the administration of AB-Tie1-39 in a perioperative therapeutic window led to a significant survival advantage as compared to control-IgG-treated mice. Additional in vivo experimental metastasis and in vitro transmigration assays concurrently revealed that AB-Tie1-39 treatment suppressed tumor cell extravasation at secondary sites. Taken together, the data phenocopy previous genetic work in endothelial Tie1 KO mice and thereby validate AB-Tie1-39 as a Tie1 function-blocking antibody. The study establishes Tie1 as a therapeutic target for metastasis in a perioperative or neoadjuvant setting.


Subject(s)
Neoplasms , Receptor, TIE-1 , Angiopoietin-1 , Angiopoietin-2 , Animals , Gene Deletion , Humans , Mice , Neovascularization, Pathologic , Receptor, TIE-1/genetics , Receptor, TIE-2
4.
Mol Cancer Ther ; 18(4): 856-867, 2019 04.
Article in English | MEDLINE | ID: mdl-30787172

ABSTRACT

Inhibition of VEGFR signaling is an effective treatment for renal cell carcinoma, but resistance continues to be a major problem. Recently, the sphingosine phosphate (S1P) signaling pathway has been implicated in tumor growth, angiogenesis, and resistance to antiangiogenic therapy. S1P is a bioactive lipid that serves an essential role in developmental and pathologic angiogenesis via activation of the S1P receptor 1 (S1P1). S1P1 signaling counteracts VEGF signaling and is required for vascular stabilization. We used in vivo and in vitro angiogenesis models including a postnatal retinal angiogenesis model and a renal cell carcinoma murine tumor model to test whether simultaneous inhibition of S1P1 and VEGF leads to improved angiogenic inhibition. Here, we show that inhibition of S1P signaling reduces the endothelial cell barrier and leads to excessive angiogenic sprouting. Simultaneous inhibition of S1P and VEGF signaling further disrupts the tumor vascular beds, decreases tumor volume, and increases tumor cell death compared with monotherapies. These studies suggest that inhibition of angiogenesis at two stages of the multistep process may maximize the effects of antiangiogenic therapy. Together, these data suggest that combination of S1P1 and VEGFR-targeted therapy may be a useful therapeutic strategy for the treatment of renal cell carcinoma and other tumor types.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Sphingosine-1-Phosphate Receptors/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Angiogenesis Inhibitors/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Drug Therapy, Combination , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Humans , Kidney Neoplasms/blood supply , Kidney Neoplasms/pathology , Lysophospholipids/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Neovascularization, Pathologic/drug therapy , Sphingosine/analogs & derivatives , Sphingosine/antagonists & inhibitors , Sunitinib/pharmacology , Treatment Outcome , Tumor Burden/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Xenograft Model Antitumor Assays
5.
J Bacteriol ; 190(18): 6170-7, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18658270

ABSTRACT

Chromatin immunoprecipitation and microarray (ChIP-chip) analysis showed that the nitric oxide (NO)-sensitive repressor NsrR from Escherichia coli binds in vivo to the promoters of the tynA and feaB genes. These genes encode the first two enzymes of a pathway that is required for the catabolism of phenylethylamine (PEA) and its hydroxylated derivatives tyramine and dopamine. Deletion of nsrR caused small increases in the activities of the tynA and feaB promoters in cultures grown on PEA. Overexpression of nsrR severely retarded growth on PEA and caused a marked repression of the tynA and feaB promoters. Both the growth defect and the promoter repression were reversed in the presence of a source of NO. These results are consistent with NsrR mediating repression of the tynA and feaB genes by binding (in an NO-sensitive fashion) to the sites identified by ChIP-chip. E. coli was shown to use 3-nitrotyramine as a nitrogen source for growth, conditions which partially induce the tynA and feaB promoters. Mutation of tynA (but not feaB) prevented growth on 3-nitrotyramine. Growth yields, mutant phenotypes, and analyses of culture supernatants suggested that 3-nitrotyramine is oxidized to 4-hydroxy-3-nitrophenylacetate, with growth occurring at the expense of the amino group of 3-nitrotyramine. Accordingly, enzyme assays showed that 3-nitrotyramine and its oxidation product (4-hydroxy-3-nitrophenylacetaldehyde) could be oxidized by the enzymes encoded by tynA and feaB, respectively. The results suggest that an additional physiological role of the PEA catabolic pathway is to metabolize nitroaromatic compounds that may accumulate in cells exposed to NO.


Subject(s)
DNA-Binding Proteins/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial , Nitrophenols/metabolism , Phenylacetates/metabolism , Transcription Factors/metabolism , Tyramine/analogs & derivatives , DNA-Binding Proteins/genetics , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli Proteins/genetics , Nitric Oxide/metabolism , Nitric Oxide/pharmacology , Oxidation-Reduction , Phenethylamines/metabolism , Promoter Regions, Genetic/drug effects , Transcription Factors/genetics , Tyramine/metabolism
6.
Methods Enzymol ; 437: 211-33, 2008.
Article in English | MEDLINE | ID: mdl-18433631

ABSTRACT

NsrR is a nitric oxide-sensitive regulator of transcription. In Escherichia coli, NsrR is a repressor of the hmp gene encoding the flavohemoglobin that detoxifies nitric oxide. Three other transcription units (ytfE, ygbA, and hcp-hcr) are known to be subject to regulation by NsrR. This chapter describes experimental and statistical protocols used to identify NsrR-binding sites in the E. coli chromosome using chromatin immunoprecipitation and microarray analysis. The methods are applicable, with suitable modifications, to any regulatory protein and any organism.


Subject(s)
DNA-Binding Proteins/metabolism , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial/drug effects , Genome, Bacterial , Nitric Oxide/pharmacology , Transcription Factors/metabolism , Binding Sites , Chromatin Immunoprecipitation/methods , Chromosome Mapping , Cloning, Organism/methods , DNA-Binding Proteins/physiology , Electronic Data Processing/methods , Escherichia coli Proteins/physiology , Gene Expression Profiling/methods , Microbiological Techniques , Models, Biological , Oligonucleotide Array Sequence Analysis/methods , Protein Binding , Staining and Labeling/methods , Transcription Factors/physiology
7.
J Clin Invest ; 113(11): 1571-81, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15173883

ABSTRACT

Uncontrolled hepatic glucose production contributes significantly to hyperglycemia in patients with type 2 diabetes. Hyperglucagonemia is implicated in the etiology of this condition; however, effective therapies to block glucagon signaling and thereby regulate glucose metabolism do not exist. To determine the extent to which blocking glucagon action would reverse hyperglycemia, we targeted the glucagon receptor (GCGR) in rodent models of type 2 diabetes using 2'-methoxyethyl-modified phosphorothioate-antisense oligonucleotide (ASO) inhibitors. Treatment with GCGR ASOs decreased GCGR expression, normalized blood glucose, improved glucose tolerance, and preserved insulin secretion. Importantly, in addition to decreasing expression of cAMP-regulated genes in liver and preventing glucagon-mediated hepatic glucose production, GCGR inhibition increased serum concentrations of active glucagon-like peptide-1 (GLP-1) and insulin levels in pancreatic islets. Together, these studies identify a novel mechanism whereby GCGR inhibitors reverse the diabetes phenotype by the dual action of decreasing hepatic glucose production and improving pancreatic beta cell function.


Subject(s)
Diabetes Mellitus/metabolism , Liver/metabolism , Oligodeoxyribonucleotides, Antisense/metabolism , Peptides/metabolism , Receptors, Glucagon/genetics , Animals , Blood Glucose/metabolism , Glucagon-Like Peptide 1 , Mice , Oligodeoxyribonucleotides, Antisense/genetics , Rats
8.
Diabetes ; 54(6): 1846-53, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15919808

ABSTRACT

Glucocorticoids (GCs) increase hepatic gluconeogenesis and play an important role in the regulation of hepatic glucose output. Whereas systemic GC inhibition can alleviate hyperglycemia in rodents and humans, it results in adrenal insufficiency and stimulation of the hypothalamic-pituitary-adrenal axis. In the present study, we used optimized antisense oligonucleotides (ASOs) to cause selective reduction of the glucocorticoid receptor (GCCR) in liver and white adipose tissue (WAT) and evaluated the resultant changes in glucose and lipid metabolism in several rodent models of diabetes. Treatment of ob/ob mice with GCCR ASOs for 4 weeks resulted in approximately 75 and approximately 40% reduction in GCCR mRNA expression in liver and WAT, respectively. This was accompanied by approximately 65% decrease in fed and approximately 30% decrease in fasted glucose levels, a 60% decrease in plasma insulin concentration, and approximately 20 and 35% decrease in plasma resistin and tumor necrosis factor-alpha levels, respectively. Furthermore, GCCR ASO reduced hepatic glucose production and inhibited hepatic gluconeogenesis in liver slices from basal and dexamethasone-treated animals. In db/db mice, a similar reduction in GCCR expression caused approximately 40% decrease in fed and fasted glucose levels and approximately 50% reduction in plasma triglycerides. In ZDF and high-fat diet-fed streptozotocin-treated (HFD-STZ) rats, GCCR ASO treatment caused approximately 60% reduction in GCCR expression in the liver and WAT, which was accompanied by a 40-70% decrease in fasted glucose levels and a robust reduction in plasma triglyceride, cholesterol, and free fatty acids. No change in circulating corticosterone levels was seen in any model after GCCR ASO treatment. To further demonstrate that GCCR ASO does not cause systemic GC antagonism, normal Sprague-Dawley rats were challenged with dexamethasone after treating with GCCR ASO. Dexamethasone increased the expression of GC-responsive genes such as PEPCK in the liver and decreased circulating lymphocytes. GCCR ASO treatment completely inhibited the increase in dexamethasone-induced PEPCK expression in the liver without causing any change in the dexamethasone-induced lymphopenia. These studies demonstrate that tissue-selective GCCR antagonism with ASOs may be a viable therapeutic strategy for the treatment of the metabolic syndrome.


Subject(s)
Adipose Tissue/metabolism , Diabetes Mellitus, Experimental/drug therapy , Liver/metabolism , Oligoribonucleotides, Antisense/pharmacology , Receptors, Glucocorticoid/metabolism , Animals , Dexamethasone/pharmacology , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/metabolism , Gene Expression/drug effects , Glucocorticoids/metabolism , Hyperglycemia/drug therapy , Hyperlipidemias/drug therapy , Lymphopenia/chemically induced , Lymphopenia/physiopathology , Mice , Mice, Obese , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Pro-Opiomelanocortin/metabolism , RNA, Messenger/metabolism , Rats
9.
Cancer Res ; 76(9): 2573-86, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27197264

ABSTRACT

Treatment of metastatic gastric cancer typically involves chemotherapy and monoclonal antibodies targeting HER2 (ERBB2) and VEGFR2 (KDR). However, reliable methods to identify patients who would benefit most from a combination of treatment modalities targeting the tumor stroma, including new immunotherapy approaches, are still lacking. Therefore, we integrated a mouse model of stromal activation and gastric cancer genomic information to identify gene expression signatures that may inform treatment strategies. We generated a mouse model in which VEGF-A is expressed via adenovirus, enabling a stromal response marked by immune infiltration and angiogenesis at the injection site, and identified distinct stromal gene expression signatures. With these data, we designed multiplexed IHC assays that were applied to human primary gastric tumors and classified each tumor to a dominant stromal phenotype representative of the vascular and immune diversity found in gastric cancer. We also refined the stromal gene signatures and explored their relation to the dominant patient phenotypes identified by recent large-scale studies of gastric cancer genomics (The Cancer Genome Atlas and Asian Cancer Research Group), revealing four distinct stromal phenotypes. Collectively, these findings suggest that a genomics-based systems approach focused on the tumor stroma can be used to discover putative predictive biomarkers of treatment response, especially to antiangiogenesis agents and immunotherapy, thus offering an opportunity to improve patient stratification. Cancer Res; 76(9); 2573-86. ©2016 AACR.


Subject(s)
Stomach Neoplasms/classification , Stomach Neoplasms/genetics , Transcriptome/genetics , Tumor Microenvironment/genetics , Animals , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Computational Biology/methods , Disease Models, Animal , Gene Expression Profiling/methods , Heterografts , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Mice , Neovascularization, Pathologic/genetics , Oligonucleotide Array Sequence Analysis , Tissue Array Analysis , Vascular Endothelial Growth Factor A/metabolism
10.
DNA Seq ; 13(2): 77-83, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12180349

ABSTRACT

Nine thousand and eighty-eight base pairs of the chicken Hoxa 11 gene, including 8470 bases 5' of the translation start site were sequenced, and the characteristics of the upstream sequence investigated. Consistent with previous findings that middle repetitive elements are rare in the HoxA cluster, no repetitive elements were found other than simple oligonucleotide repeats. Multiple and pairwise alignments of the chicken upstream sequence with its human and mouse orthologs revealed multiple regions of 80% or higher homology across species. For the chicken, these regions were separated by sequences with no significant homology to human, mouse, or in most cases any other Genbank sequences. Selective clustering of transcription factor binding motifs was found to occur within the conserved homologous regions, suggesting evolutionary conservation of critical regulatory sequences. Of particular interest, seven conserved Cdx binding sites were found in the Hoxa 11 promoter, suggesting regulation by a non-clustered Caudal homeobox gene. Previous analysis of the mouse and human Hoxa 11 genes found a conserved antisense transcript, of unknown function. The chicken Hoxa 11 antisense strand included a conserved open reading frame capable of encoding 168 amino acids. Comparison of this region in mouse and chicken showed seven insertion/deletions, with each a multiple of three bases, thereby preserving open reading frame.


Subject(s)
DNA Transposable Elements , DNA, Antisense/physiology , Homeodomain Proteins/genetics , Oncogene Proteins/genetics , Phylogeny , Animals , Base Sequence , Binding Sites , Chickens , Conserved Sequence , Humans , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Proto-Oncogene Proteins , Repetitive Sequences, Nucleic Acid , Sequence Alignment , Sequence Analysis, DNA
11.
Mol Cancer Ther ; 10(10): 1796-806, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21846830

ABSTRACT

Replication protein A (RPA) is a single-strand DNA-binding protein with essential roles in DNA replication, recombination, and repair. It is necessary for the formation of the preincision complex that is required for proper incision of damaged DNA nucleotides during DNA repair. We have previously identified small molecule inhibitors (SMI) with the ability to disrupt RPA-binding activity to ssDNA. Further characterization of these RPA inhibitors was done using both lung and ovarian cancer cell lines. Lung cancer cell lines showed increased apoptotic cell death following treatment with the SMI MCI13E, with IC(50) values of approximately 5 µmol/L. The ovarian cancer cell line A2780 and the p53-null lung cancer cell line H1299 were particularly sensitive to MCI13E treatment, with IC(50) values less than 3 µmol/L. Furthermore, a cell-cycle effect was observed in lung cancer cell lines that resulted in a lengthening of either G(1) or S-phases of the cell cycle following single-agent treatment. Sequential treatment with MCI13E and cisplatin resulted in synergism. Overall, these data suggest that decreasing DNA-binding activity of RPA via a SMI may disrupt the role of RPA in cell-cycle regulation. Thus, SMIs of RPA hold the potential to be used as single-agent chemotherapeutics or in combination with current chemotherapeutic regimens to increase efficacy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Camphanes/chemistry , Camphanes/pharmacology , Cisplatin/pharmacology , DNA Replication/drug effects , Lung Neoplasms/drug therapy , Ovarian Neoplasms/drug therapy , Replication Protein A/antagonists & inhibitors , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , DNA Damage , DNA, Neoplasm/biosynthesis , DNA, Neoplasm/drug effects , DNA, Neoplasm/genetics , DNA, Single-Stranded/drug effects , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Drug Synergism , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Replication Protein A/chemistry , Replication Protein A/genetics , Replication Protein A/metabolism , Small Molecule Libraries
12.
J Bacteriol ; 188(3): 874-81, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16428390

ABSTRACT

Microarray studies of the Escherichia coli response to nitric oxide and nitrosative stress have suggested that additional transcriptional regulators of this response remain to be characterized. We identify here the product of the yjeB gene as a negative regulator of the transcription of the ytfE, hmpA and ygbA genes, all of which are known to be upregulated by nitrosative stress. Transcriptional fusions to the promoters of these genes were expressed constitutively in a yjeB mutant, indicating that all three are targets for repression by YjeB. An inverted repeat sequence that overlaps the -10 element of all three promoters is proposed to be a binding site for the YjeB protein. A similar inverted repeat sequence was identified in the tehA promoter, which is also known to be sensitive to nitrosative stress. The ytfE, hmpA, ygbA, and tehA promoters all caused derepression of a ytfE-lacZ transcriptional fusion when present in the cell in multiple copies, presumably by a repressor titration effect, suggesting the presence of functional YjeB binding sites in these promoters. However, YjeB regulation of tehA was weak, as judged by the activity of a tehA-lacZ fusion, perhaps because YjeB repression of tehA is masked by other regulatory mechanisms. Promoters regulated by YjeB could be derepressed by iron limitation, which is consistent with an iron requirement for YjeB activity. The YjeB protein is a member of the Rrf2 family of transcriptional repressors and shares three conserved cysteine residues with its closest relatives. We propose a regulatory model in which the YjeB repressor is directly sensitive to nitrosative stress. On the basis of similarity to the nitrite-responsive repressor NsrR from Nitrosomonas europaea, we propose that the yjeB gene of E. coli be renamed nsrR.


Subject(s)
Escherichia coli Proteins/genetics , Escherichia coli/physiology , Gene Expression Regulation, Bacterial , Nitric Oxide/pharmacology , Nitrite Reductases/metabolism , Transcription, Genetic/drug effects , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli Proteins/physiology , Genes, Regulator/genetics , Nitrite Reductases/genetics
13.
Muscle Nerve ; 33(5): 609-18, 2006 May.
Article in English | MEDLINE | ID: mdl-16421881

ABSTRACT

Transthyretin (TTR) amyloidosis, the most common form of hereditary systemic amyloidosis, is characterized clinically by adult-onset axonal neuropathy and restrictive cardiomyopathy. More than 85 mutations in transthyretin have been found to cause this hereditary disease. Since essentially all circulating TTR is of hepatic origin, orthotopic liver transplantation has been used as the only specific form of therapy. Unfortunately, in many patients amyloid deposition continues after orthotopic liver transplantation, indicating that mutant TTR is no longer required for progression of the disease after tissue deposits have been initiated. As a first step toward medical treatment of this disease, we have employed antisense oligonucleotides (ASOs) to inhibit hepatic expression of TTR. A transgenic mouse model carrying the human TTR Ile84Ser mutation was created and shown to express high levels of human mutant transthyretin. TTR ASOs suppressed hepatic TTR mRNA levels and serum TTR levels by as much as 80%. Suppression of hepatic synthesis of transthyretin may offer a medical treatment for transthyretin systemic amyloidosis.


Subject(s)
Amyloidosis, Familial/drug therapy , Amyloidosis, Familial/genetics , Gene Silencing , Oligonucleotides, Antisense/therapeutic use , Prealbumin/antagonists & inhibitors , Alanine Transaminase/blood , Amyloidosis, Familial/blood , Animals , Aspartate Aminotransferases/blood , Carcinoma, Hepatocellular/pathology , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression/drug effects , Humans , Immunohistochemistry/methods , Isoleucine/genetics , Liver/drug effects , Liver/enzymology , Mice , Mice, Transgenic , Mutation/physiology , Prealbumin/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Serine/genetics , Time Factors
14.
J Bacteriol ; 186(5): 1438-47, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14973013

ABSTRACT

Caulobacter crescentus has a dimorphic life cycle composed of a motile stage and a sessile stage. In the sessile stage, C. crescentus is often found tightly attached to a surface through its adhesive holdfast. In this study, we examined the contribution of growth and external structures to the attachment of C. crescentus to abiotic surfaces. We show that the holdfast is essential but not sufficient for optimal attachment. Rather, adhesion in C. crescentus is a complex developmental process. We found that the attachment of C. crescentus to surfaces is cell cycle regulated and that growth or energy or both are essential for this process. The initial stage of attachment occurs in swarmer cells and is facilitated by flagellar motility and pili. Our results suggest that strong attachment is mediated by the synthesis of a holdfast as the swarmer cell differentiates into a stalked cell.


Subject(s)
Bacterial Adhesion , Caulobacter crescentus/growth & development , Caulobacter crescentus/physiology , Gene Expression Regulation, Bacterial , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Caulobacter crescentus/genetics , Cell Cycle , Culture Media , Fimbriae, Bacterial/physiology , Flagella/physiology , Glass , Movement , Polystyrenes , Surface Properties
15.
J Bacteriol ; 185(4): 1432-42, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12562815

ABSTRACT

Adhesion to both abiotic and biotic surfaces by the gram-negative prothescate bacterium Caulobacter crescentus is mediated by a polar organelle called the "holdfast," which enables the bacterium to form stable monolayer biofilms. The holdfast, a complex polysaccharide composed in part of N-acetylglucosamine, localizes to the tip of the stalk (a thin cylindrical extension of the cell wall and membranes). We report here the isolation of adhesion mutants with transposon insertions in an uncharacterized gene cluster involved in holdfast biogenesis (hfs) as well as in previously identified polar development genes (podJ and pleC), and the holdfast attachment genes (hfa). Clean deletions of three of the four genes in the hfs gene cluster (hfsDAB) resulted in a severe holdfast biogenesis phenotype. These mutants do not bind to surfaces or to a fluorescently labeled lectin, specific for N-acetylglucosamine. Transmission electron microscopy indicated that the hfsDAB mutants fail to synthesize a holdfast at the stalk tip. The predicted hfs gene products have significant sequence similarity to proteins necessary for exopolysaccharide export in gram-negative bacteria. HfsA has sequence similarity to GumC from Xanthomonas campestris, which is involved in exopolysaccharide export in the periplasm. HfsD has sequence similarity to Wza from Escherichia coli, an outer membrane protein involved in secretion of polysaccharide through the outer membrane. HfsB is a novel protein involved in holdfast biogenesis. These data suggest that the hfs genes play an important role in holdfast export.


Subject(s)
Bacterial Adhesion , Bacterial Proteins/genetics , Caulobacter crescentus/genetics , Mutation , Organelles/metabolism , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Bacterial Adhesion/genetics , Bacterial Proteins/metabolism , Caulobacter crescentus/growth & development , Caulobacter crescentus/physiology , Culture Media , DNA Transposable Elements , Multigene Family , Mutagenesis, Insertional
16.
Mol Microbiol ; 49(6): 1671-83, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12950929

ABSTRACT

The differentiating bacterium Caulobacter crescentus produces two different cell types at each cell division, a motile swarmer cell and an adhesive stalked cell. The stalked cell harbours a stalk, a thin cylindrical extension of the cell surface. The tip of the stalk is decorated with a holdfast, an adhesive organelle composed at least in part of polysaccharides. The synthesis of the stalk and holdfast occur at the same pole during swarmer cell differentiation. Mutations in the hfaABDC gene cluster had been shown to disrupt the attachment of the holdfast to the tip of the stalk, but the role of individual genes was unknown. We used lacZ fusions of various DNA fragments from the hfaABDC region to show that these genes form an operon. In order to analyse the relative contribution of the different genes to holdfast attachment, mutations were constructed for each gene. hfaC was not required for holdfast attachment or binding to surfaces. The hfaA and hfaD mutants shed some holdfast material into the surrounding medium and were partially deficient in binding to surfaces. Unlike hfaA and hfaB mutants, hfaD mutants were still able to form rosettes efficiently. Cells with insertions in hfaB were unable to bind to surfaces, and lectin binding studies indicated that the hfaB mutants had the strongest holdfast shedding phenotype. We determined that HfaB and HfaD are membrane-associated proteins and that HfaB is a lipoprotein. Purification of stalks and cell bodies indicated that both HfaB and HfaD are enriched in the stalk as compared to the cell body. These results suggest that HfaB and HfaD, and probably HfaA, serve to anchor the holdfast to the tip of the stalk.


Subject(s)
Bacterial Adhesion , Bacterial Proteins/metabolism , Caulobacter crescentus/cytology , Caulobacter crescentus/metabolism , Membrane Proteins/metabolism , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Bacterial Proteins/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caulobacter crescentus/chemistry , Caulobacter crescentus/genetics , Cell Fractionation , Chromosome Mapping , Electrophoresis, Polyacrylamide Gel , Gene Expression Regulation, Bacterial , Genes, Reporter , Immunoblotting , Lipoproteins/genetics , Lipoproteins/metabolism , Membrane Proteins/genetics , Mutagenesis, Insertional , Operon , Promoter Regions, Genetic/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Wheat Germ Agglutinins/metabolism , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL