Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Semin Cardiothorac Vasc Anesth ; 23(2): 156-163, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30985250

ABSTRACT

In this article, we present the annual review of the literature relevant for the practice of cardiovascular critical care.


Subject(s)
Anesthesiology , Cardiovascular Surgical Procedures/methods , Critical Care/methods , Anesthesiologists , Humans , Intensive Care Units
3.
Expert Rev Mol Med ; 14: e12, 2012 May 24.
Article in English | MEDLINE | ID: mdl-22622048

ABSTRACT

Malignant mesothelioma is a rare, highly aggressive cancer arising from mesothelial cells that line the pleural cavities. Approximately 80% of mesothelioma cases can be directly attributed to asbestos exposure. Additional suspected causes or co-carcinogens include other mineral fibres, simian virus 40 (SV40) and radiation. A mesothelioma epidemic in Turkey has demonstrated a probable genetic predisposition to mineral fibre carcinogenesis and studies of human tissues and animal models of mesothelioma have demonstrated genetic and epigenetic events that contribute to the multistep process of mineral fibre carcinogenesis. Several growth factors and their receptors have a significant role in the oncogenesis, progression and resistance to therapy of mesothelioma. Epidermal growth factor (EGF), hepatocyte growth factor (HGF), vascular endothelial growth factor (VEGF) and insulin-like growth factor (IGF) have been shown as targets for therapy based on promising preclinical data. However, clinical trials of tyrosine kinase inhibitors in mesothelioma have been disappointing. Bcl-XL is an important antiapoptotic member of the Bcl-2 family and is overexpressed in several solid tumours, including mesothelioma. Reduction of Bcl-XL expression in mesothelioma induces apoptosis and engenders sensitisation to cytotoxic chemotherapeutic agents. Pharmacological inhibitors of antiapoptotic Bcl-2 family members continue to undergo refinement and have shown promise in mesothelioma.


Subject(s)
Mesothelioma/genetics , Mesothelioma/pathology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Humans , Mesothelioma/drug therapy , Mesothelioma/etiology , bcl-X Protein/antagonists & inhibitors , bcl-X Protein/genetics , bcl-X Protein/metabolism
4.
Mol Cancer ; 9: 110, 2010 May 18.
Article in English | MEDLINE | ID: mdl-20482802

ABSTRACT

BACKGROUND: Bortezomib, a proteasome-specific inhibitor, has emerged as a promising cancer therapeutic agent. However, development of resistance to bortezomib may pose a challenge to effective anticancer therapy. Therefore, characterization of cellular mechanisms involved in bortezomib resistance and development of effective strategies to overcome this resistance represent important steps in the advancement of bortezomib-mediated cancer therapy. RESULTS: The present study reports the development of I-45-BTZ-R, a bortezomib-resistant cell line, from the bortezomib-sensitive mesothelioma cell line I-45. I-45-BTZ-R cells showed no cross-resistance to the chemotherapeutic drugs cisplatin, 5-fluorouracil, and doxorubicin. Moreover, the bortezomib-adapted I-45-BTZ-R cells had decreased growth kinemics and did not over express proteasome subunit beta5 (PSMB5) as compared to parental I-45 cells. I-45-BTZ-R cells and parental I-45 cells showed similar inhibition of proteasome activity, but I-45-BTZ-R cells exhibited much less accumulation of ubiquitinated proteins following exposure to 40 nm bortezomib. Further studies revealed that relatively low doses of bortezomib did not induce an unfolded protein response (UPR) in the bortezomib-adapted cells, while higher doses induced UPR with concomitant cell death, as evidenced by higher expression of the mitochondrial chaperone protein Bip and the endoplasmic reticulum (ER) stress-related pro-apoptotic protein CHOP. In addition, bortezomib exposure did not induce the accumulation of the pro-apoptotic proteins p53, Mcl-1S, and noxa in the bortezomib-adapted cells. CONCLUSION: These results suggest that UPR evasion, together with reduced pro-apoptotic gene induction, accounts for bortezomib resistance in the bortezomib-adapted mesothelioma cell line I-45-BTZ-R.


Subject(s)
Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Drug Resistance, Neoplasm/genetics , Mesothelioma/metabolism , Pyrazines/pharmacology , Unfolded Protein Response/drug effects , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/drug effects , Apoptosis Regulatory Proteins/metabolism , Blotting, Western , Bortezomib , Cell Line, Tumor , Cell Separation , Flow Cytometry , Gene Expression/drug effects , Humans , Mesothelioma/genetics , Oligopeptides/genetics , Oligopeptides/metabolism , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Transfection
5.
Int J Cancer ; 123(1): 202-8, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18360826

ABSTRACT

Bcl-xL functions as a dominant regulator of apoptotic cell death and is implicated in chemotherapeutic resistance of malignant pleural mesothelioma (MPM). Mesothelioma cell lines demonstrate increasing levels of Bcl-xL as resistant clones are selected in vitro. Moreover, upon introduction of antisense oligonucleotides specific to Bcl-xL mRNA, MPM cells are sensitized to chemotherapeutic agents. Here we describe the therapeutic effects of a novel combination therapy, Bcl-xL antisense oligonucleotide (ASO 15999) and cisplatin, on mesothelioma cell lines in vitro and in vivo; in addition, efficacy of ASO 15999 in decreasing tumor load as well as its effect on survival in an animal model. Finally, we initiated preliminary toxicity studies involved with intraperitoneal (IP) injections of ASO 15999 into mice. This novel combination, with doses of cisplatin four times below established IC(50) levels, significantly decreased viability of MPM cell lines after 48 hr. The growth of established mouse flank human tumor xenografts was reduced with intra-tumor administration of ASO 15999. Local spread and development of IP xenografts was reduced with treatments of ASO alone, and survival of mice afflicted with these xenografts was prolonged after administration of ASO alone and ASO 15999 + cisplatin combination therapy. These findings suggest that ASO 15999 sensitizes MPM cell lines to the toxic effects of cisplatin. ASO 15999 induced reduction of Bcl-xL is effective in slowing the progression of human mesothelioma cell lines both in vitro and in vivo. More notably, the combination of Bcl-xL ASO and cisplatin extends survival in an orthotopic tumor xenograft model.


Subject(s)
Antineoplastic Agents/pharmacology , Mesothelioma/drug therapy , Oligonucleotides, Antisense/pharmacology , Pleural Neoplasms/drug therapy , bcl-X Protein/pharmacology , Analysis of Variance , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Cisplatin/administration & dosage , Humans , Inhibitory Concentration 50 , Injections, Intraperitoneal , Kaplan-Meier Estimate , Mice , Mice, SCID , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/metabolism , Oligonucleotides, Antisense/toxicity , Time Factors , Xenograft Model Antitumor Assays , bcl-X Protein/administration & dosage , bcl-X Protein/metabolism
6.
Biophys J ; 92(3): 977-88, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17085493

ABSTRACT

Phosphoglycerate mutases (PGMs) catalyze the isomerization of 2- and 3-phosphoglycerates and are essential for glucose metabolism in most organisms. This study reports the production, structure, and molecular dynamics analysis of Bacillus anthracis cofactor-independent PGM (iPGM). The three-dimensional structure of B. anthracis PGM is composed of two structural and functional domains, the phosphatase and transferase. The structural relationship between these two domains is different than in the B. stearothermophilus iPGM structure determined previously. However, the structures of the two domains of B. anthracis iPGM show a high degree of similarity to those in B. stearothermophilus iPGM. The novel domain arrangement in B. anthracis iPGM and the dynamic property of these domains is directly linked to the mechanism of enzyme catalysis, in which substrate binding is proposed to result in close association of the two domains. The structure of B. anthracis iPGM and the molecular dynamics of this structure provide unique insight into the mechanism of iPGM catalysis, in particular the roles of changes in coordination geometry of the enzyme's two bivalent metal ions and the regulation of this enzyme's activity by changes in intracellular pH during spore formation and germination in Bacillus species.


Subject(s)
Bacillus anthracis/enzymology , Models, Chemical , Models, Molecular , Phosphoglycerate Mutase/chemistry , Phosphoglycerate Mutase/ultrastructure , Catalysis , Cell Proliferation , Computer Simulation , Enzyme Activation , Isoenzymes/chemistry , Protein Conformation , Protein Structure, Tertiary , Spores, Bacterial
7.
Proteins ; 66(3): 547-58, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17115425

ABSTRACT

Streptococcus pneumoniae open reading frame SP1492 encodes a surface protein that contains a novel conserved domain similar to the repeated fragments of mucin-binding proteins from lactobacilli and lactococci. To investigate the functional role(s) of this protein and its potential adhesive properties, the surface-exposed region of SP1492 was expressed in Escherichia coli, purified to homogeneity, and partially characterized by biophysical and immunological methods. Circular dichroism and sedimentation measurements confirmed that SP1492 is an all-beta protein that exists in solution as a monomer. The SP1492 protein has been shown to be expressed by S. pneumoniae and was experimentally localized to its surface. The protein functional domain binds to mucins II and III from porcine stomach and to purified submaxillary bovine gland mucin. It appears to be one of the very few unambiguous pneumococcal adhesin molecules known to date. A hypothetical model constructed by ab initio techniques predicts a novel beta-sandwich protein structure.


Subject(s)
Bacterial Proteins/genetics , Carrier Proteins/genetics , Genome, Bacterial , Mucins/metabolism , Streptococcus pneumoniae/genetics , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cell Membrane/metabolism , Chromatography, Gel , Circular Dichroism , Cloning, Molecular , Computational Biology , Gene Expression Regulation, Bacterial , Genetic Engineering , Molecular Sequence Data , Open Reading Frames , Sequence Analysis, Protein
8.
J Mol Biol ; 358(4): 1165-78, 2006 May 12.
Article in English | MEDLINE | ID: mdl-16569416

ABSTRACT

Streptococcus pneumoniae hyaluronan lyase is a surface enzyme of this Gram-positive bacterium. The enzyme degrades several biologically important, information-rich linear polymeric glycans: hyaluronan, unsulfated chondroitin, and some chondroitin sulfates. This degradation facilitates spreading of bacteria throughout the host tissues and presumably provides energy and a carbon source for pneumococcal cells. Its beta-elimination catalytic mechanism is an acid/base process termed proton acceptance and donation leading to cleavage of beta-1,4 linkages of the substrates. The degradation of hyaluronan occurs in two stages, initial endolytic cuts are followed by processive exolytic cleavage of one disaccharide at a time. In contrast, the degradation of chondroitins is purely endolytic. Structural studies together with flexibility analyses of two streptococcal enzymes, from S.pneumoniae and Streptococcus agalactiae, allowed for insights into this enzyme's molecular mechanism. Here, two new X-ray crystal structures of the pneumococcal enzyme in novel conformations are reported. These new conformations, complemented by molecular dynamics simulation results, directly confirm the predicted domain motions presumed to facilitate the processive degradative process. One of these new structures resembles the S.agalactiae enzyme conformation, and provides evidence of a uniform mechanistic/dynamic behavior of this protein across different bacteria.


Subject(s)
Polysaccharide-Lyases/chemistry , Streptococcus pneumoniae/enzymology , Catalytic Domain , Crystallography, X-Ray , Models, Molecular , Polysaccharide-Lyases/metabolism , Protein Conformation , Species Specificity , Streptococcus agalactiae/enzymology , Thermodynamics
9.
OMICS ; 8(4): 341-56, 2004.
Article in English | MEDLINE | ID: mdl-15703481

ABSTRACT

Streptococcus pneumoniae open reading frame SP0082 encodes a surface protein that contains four copies of a novel conserved repeat domain that bears no significant sequence similarity to proteins of known function. Homologous sequences from other streptococci contain two to six of these repeats, designated the SSURE (streptococcal surface repeat) domain. To investigate the functional role(s) of this domain, the third SSURE repeat of SP0082 sequence has been expressed in Escherichia coli, purified to homogeneity and characterized by biochemical and immunological methods. The expressed protein fragment was found to bind to fibronectin, but not to collagen or submaxillary mucin. Anti-SSURE antibodies recognized the corresponding protein on the surface of pneumococcal cells. These data identify S. pneumoniae SP0082 protein and its homologs in other streptococci as fibronectin-binding surface adhesins. The SSURE domain is likely to contain a novel protein fold, which was tentatively modeled using ab initio modeling methods.


Subject(s)
Bacterial Adhesion , Computational Biology/methods , Fibronectins/chemistry , Streptococcus pneumoniae/metabolism , Streptococcus pneumoniae/pathogenicity , Adhesins, Bacterial/chemistry , Amino Acid Sequence , Animals , Bacterial Outer Membrane Proteins/chemistry , Bacterial Proteins/chemistry , Cell Membrane/metabolism , Cloning, Molecular , Collagen/chemistry , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Escherichia coli/metabolism , Fibronectins/metabolism , Flow Cytometry , Humans , Mice , Models, Molecular , Molecular Sequence Data , Phylogeny , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Species Specificity
10.
FEMS Immunol Med Microbiol ; 39(3): 265-73, 2003 Dec 05.
Article in English | MEDLINE | ID: mdl-14642312

ABSTRACT

Pneumococcal surface protein A (PspA) is an antigenic variable vaccine candidate of Streptococcus pneumoniae. Epitope similarities between PspA from the American vaccine candidate strain Rx1 and Norwegian clinical isolates were studied using PspA specific monoclonal antibodies (mAbs) made against clinical Norwegian strains. Using recombinant PspA/Rx1 fragments and immunoblotting the epitopes for mAbs were mapped to two regions of amino acids, 1-67 and 67-236. The discovered epitopes were visualized by modelling of the PspA:Fab part of mAb in three dimensions. Flow cytometric analysis showed that the epitopes for majority of mAbs were accessible for antibody binding on live pneumococci. Also, the epitopes for majority of the mAbs are widely expressed among clinical Norwegian isolates.


Subject(s)
Bacterial Proteins/immunology , Pneumococcal Vaccines/immunology , Streptococcus pneumoniae/immunology , Antibodies, Monoclonal , Bacterial Proteins/chemistry , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Flow Cytometry , Humans , Models, Molecular , Pneumococcal Infections/microbiology , Pneumococcal Infections/prevention & control , Recombinant Proteins , Streptococcus pneumoniae/chemistry
11.
J Mol Biol ; 328(4): 909-20, 2003 May 09.
Article in English | MEDLINE | ID: mdl-12729763

ABSTRACT

Phosphoglycerate mutases catalyze the isomerization of 2 and 3-phosphoglycerates, and are essential for glucose metabolism in most organisms. Here, we further characterize the 2,3-bisphosphoglycerate-independent phosphoglycerate mutase (iPGM) from Bacillus stearothermophilus by determination of a high-resolution (1.4A) crystal structure of the wild-type enzyme and the crystal structure of its S62A mutant. The mutant structure surprisingly showed the replacement of one of the two catalytically essential manganese ions with a water molecule, offering an additional possible explanation for its lack of catalytic activity. Crystal structures invariably show substrate phosphoglycerate to be entirely buried in a deep cleft between the two iPGM domains. Flexibility analyses were therefore employed to reveal the likely route of substrate access to the catalytic site through an aperture created in the enzyme's surface during certain stages of the catalytic process. Several conserved residues lining this aperture may contribute to orientation of the substrate as it enters. Factors responsible for the retention of glycerate within the phosphoenzyme structure in the proposed mechanism are identified by molecular modeling of the glycerate complex of the phosphoenzyme. Taken together, these results allow for a better understanding of the mechanism of action of iPGMs. Many of the results are relevant to a series of evolutionarily related enzymes. These studies will facilitate the development of iPGM inhibitors which, due to the demonstrated importance of this enzyme in many bacteria, would be of great potential clinical significance.


Subject(s)
Geobacillus stearothermophilus/enzymology , Phosphoglycerate Mutase/chemistry , Phosphoglycerate Mutase/metabolism , Alanine/chemistry , Catalysis , Catalytic Domain , Crystallography, X-Ray , Glycolysis , Microscopy, Electron , Models, Molecular , Protein Conformation , X-Ray Diffraction
12.
J Mol Biol ; 325(3): 411-20, 2003 Jan 17.
Article in English | MEDLINE | ID: mdl-12498792

ABSTRACT

Bacillus stearothermophilus phosphatase PhoE is a member of the cofactor-dependent phosphoglycerate mutase superfamily possessing broad specificity phosphatase activity. Its previous structural determination in complex with glycerol revealed probable bases for its efficient hydrolysis of both large, hydrophobic, and smaller, hydrophilic substrates. Here we report two further structures of PhoE complexes, to higher resolution of diffraction, which yield a better and thorough understanding of its catalytic mechanism. The environment of the phosphate ion in the catalytic site of the first complex strongly suggests an acid-base catalytic function for Glu83. It also reveals how the C-terminal tail ordering is linked to enzyme activation on phosphate binding by a different mechanism to that seen in Escherichia coli phosphoglycerate mutase. The second complex structure with an unusual doubly covalently bound trivanadate shows how covalent modification of the phosphorylable His10 is accompanied by small structural changes, presumably to catalytic advantage. When compared with structures of related proteins in the cofactor-dependent phosphoglycerate mutase superfamily, an additional phosphate ligand, Gln22, is observed in PhoE. Functional constraints lead to the corresponding residue being conserved as Gly in fructose-2,6-bisphosphatases and Thr/Ser/Cys in phosphoglycerate mutases. A number of sequence annotation errors in databases are highlighted by this analysis. B. stearothermophilus PhoE is evolutionarily related to a group of enzymes primarily present in Gram-positive bacilli. Even within this group substrate specificity is clearly variable highlighting the difficulties of computational functional annotation in the cofactor-dependent phosphoglycerate mutase superfamily.


Subject(s)
Geobacillus stearothermophilus/enzymology , Phosphates/metabolism , Phosphoric Monoester Hydrolases/chemistry , Protein Structure, Tertiary , Vanadates/metabolism , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , Enzyme Activation , Genes, Bacterial , Geobacillus stearothermophilus/chemistry , Glutamine/metabolism , Models, Molecular , Molecular Sequence Data , Multigene Family , Phosphoglycerate Mutase/chemistry , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...