Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
iScience ; 25(3): 103968, 2022 Mar 18.
Article in English | MEDLINE | ID: mdl-35224469

ABSTRACT

As the emergence of SARS-CoV-2 variants brings the global pandemic to new levels, the performance of current rapid antigen tests against variants of concern and interest (VOC/I) is of significant public health concern. Here, we report assessment of the Abbot BinaxNOW COVID-19 Antigen Self-Test. Using genetically sequenced remnant clinical samples collected from individuals positive for SARS-CoV-2, we assessed the performance of BinaxNOW against the variants that currently pose public health threats. We measured the limit of detection of BinaxNOW against various VOC/I in a blinded manner. BinaxNOW successfully detected the Omicron (B.1.1.529), Mu (B.1.621), Delta (B.1.617.2), Lambda (C.37), Gamma (P.1), Alpha (B.1.1.7), Beta (B.1.351), Eta (B.1.525), and P.2 variants and at low viral concentrations. BinaxNOW also detected the Omicron variant in individual remnant clinical samples. Overall, these data indicate that this inexpensive and simple-to-use, FDA-authorized and broadly distributed rapid test can reliably detect Omicron, Delta, and other VOC/I.

2.
IEEE Open J Eng Med Biol ; 2: 286-290, 2021.
Article in English | MEDLINE | ID: mdl-35257097

ABSTRACT

Goal: Monitoring the genetic diversity and emerging mutations of SARS-CoV-2 is crucial for understanding the evolution of the virus and assuring the performance of diagnostic tests, vaccines, and therapies against COVID-19. SARS-CoV-2 is still adapting to humans and, as illustrated by B.1.1.7 (Alpha) and B.1.617.2 (Delta), lineage dynamics are fluid, and strain prevalence may change radically in a matter of months. The National Institutes of Health's Rapid Acceleration of Diagnostics (RADxSM) initiative created a Variant Task Force to assess the impact of emerging SARS-CoV-2 variants on in vitro diagnostic testing. Working in tandem with clinical laboratories, the FDA, and the CDC, the Variant Task Force uses both in silico modeling and in vitro testing to determine the effect of SARS-CoV-2 mutations on diagnostic molecular and antigen tests. Here, we offer an overview of the approach and activities of the RADx Variant Task Force to ensure test performance against emerging SARS-CoV-2 lineages.

3.
Sci Rep ; 7(1): 9718, 2017 09 08.
Article in English | MEDLINE | ID: mdl-28887440

ABSTRACT

Data-independent acquisition mass spectrometry promises higher performance in terms of quantification and reproducibility compared to data-dependent acquisition mass spectrometry methods. To enable high-accuracy quantification of Staphylococcus aureus proteins, we have developed a global ion library for data-independent acquisition approaches employing high-resolution time of flight or Orbitrap instruments for this human pathogen. We applied this ion library resource to investigate the time-resolved adaptation of S. aureus to the intracellular niche in human bronchial epithelial cells and in a murine pneumonia model. In epithelial cells, abundance changes for more than 400 S. aureus proteins were quantified, revealing, e.g., the precise temporal regulation of the SigB-dependent stress response and differential regulation of translation, fermentation, and amino acid biosynthesis. Using an in vivo murine pneumonia model, our data-independent acquisition quantification analysis revealed for the first time the in vivo proteome adaptation of S. aureus. From approximately 2.15 × 105 S. aureus cells, 578 proteins were identified. Increased abundance of proteins required for oxidative stress response, amino acid biosynthesis, and fermentation together with decreased abundance of ribosomal proteins and nucleotide reductase NrdEF was observed in post-infection samples compared to the pre-infection state.


Subject(s)
Bacterial Proteins/metabolism , Host-Pathogen Interactions , Proteome , Proteomics , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus/metabolism , Animals , Computational Biology/methods , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Humans , Ions/metabolism , Mice , Peptides , Proteomics/methods , Respiratory Mucosa/metabolism , Respiratory Mucosa/microbiology
4.
J Infect Dis ; 211(11): 1822-30, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25512625

ABSTRACT

BACKGROUND: Streptococcus pneumoniae is the most common cause of community-acquired pneumonia worldwide. During pneumococcal pneumonia, the human airway epithelium is exposed to large amounts of H2O2 as a product of host and pathogen oxidative metabolism. Airway cells are known to be highly vulnerable to oxidant damage, but the pathophysiology of oxidative stress induced by S. pneumoniae and the role of nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated antioxidant systems of the host are not well characterized. METHODS: For gluthation/gluthathion disulfide analysis BEAS-2B cells, primary broncho-epithelial cells (pBEC), explanted human lung tissue and mouse lungs were infected with different S. pneumoniae strains (D39, A66, R6x, H2O2/pneumolysin/LytA- deficient mutants of R6x). Cell death was proven by LDH assay and cell viability by IL-8 ELISA. The translocation of Nrf2 and the expression of catalase were shown via Western blot. The binding of Nrf2 at the catalase promoter was analyzed by ChIP. RESULTS: We observed a significant induction of oxidative stress induced by S. pneumoniae in vivo, ex vivo, and in vitro. Upon stimulation, the oxidant-responsive transcription factor Nrf2 was activated, and catalase was upregulated via Nrf2. The pneumococci-induced oxidative stress was independent of S. pneumoniae-derived H2O2 and pneumolysin but depended on the pneumococcal autolysin LytA. The Nrf2 inducer resveratrol, as opposed to catalase, reversed oxidative stress in lung epithelial cells. CONCLUSIONS: These observations indicate a H2O2-independent induction of oxidative stress in lung epithelial cells via the release of bacterial factors of S. pneumoniae. Resveratrol might be an option for prevention of acute lung injury and inflammatory responses observed in pneumococcal pneumonia.


Subject(s)
Oxidative Stress/drug effects , Oxidative Stress/physiology , Pneumonia, Pneumococcal/immunology , Stilbenes/pharmacology , Streptococcus pneumoniae/immunology , Animals , Antioxidants/pharmacology , Autolysis , Bacterial Proteins/metabolism , Cell Line , Cell Survival , Epithelial Cells/immunology , Glutathione/metabolism , Glutathione Disulfide/metabolism , Humans , Hydrogen Peroxide/metabolism , Interleukin-8/metabolism , Lung/immunology , Mice , NF-E2-Related Factor 2/immunology , NF-E2-Related Factor 2/metabolism , Pneumonia, Pneumococcal/microbiology , Pneumonia, Pneumococcal/physiopathology , Resveratrol , Streptolysins/metabolism
5.
Mol Microbiol ; 93(6): 1183-206, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25060741

ABSTRACT

Bacterial cell wall hydrolases are essential for peptidoglycan turnover and crucial to preserve cell shape. The d,d-carboxypeptidase DacA and l,d-carboxypeptidase DacB of Streptococcus pneumoniae function in a sequential manner. Here, we determined the structure of the surface-exposed lipoprotein DacB. The crystal structure of DacB, radically different to that of DacA, contains a mononuclear Zn(2+) catalytic centre located in the middle of a large and fully exposed groove. Two different conformations were found presenting a different arrangement of the active site topology. The critical residues for catalysis and substrate specificity were identified. Loss-of-function of DacA and DacB altered the cell shape and this was consistent with a modified peptidoglycan peptide composition in dac mutants. Contrary, an lgt mutant lacking lipoprotein diacylglyceryl transferase activity required for proper lipoprotein maturation retained l,d-carboxypeptidase activity and showed an intact murein sacculus. In addition we demonstrated pathophysiological effects of disabled DacA or DacB activities. Real-time bioimaging of intranasal infected mice indicated a substantial attenuation of ΔdacB and ΔdacAΔdacB pneumococci, while ΔdacA had no significant effect. In addition, uptake of these mutants by professional phagocytes was enhanced, while the adherence to lung epithelial cells was decreased. Thus, structural and functional studies suggest DacA and DacB as optimal drug targets.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Carboxypeptidases/chemistry , Carboxypeptidases/genetics , Pneumococcal Infections/veterinary , Streptococcus pneumoniae/enzymology , Animals , Bacterial Proteins/metabolism , Carboxypeptidases/metabolism , Catalytic Domain , Cell Wall/physiology , Gene Expression Regulation, Bacterial , Gene Expression Regulation, Enzymologic , Mice , Models, Molecular , Phenotype , Pneumococcal Infections/metabolism , Protein Structure, Secondary , Streptococcus pneumoniae/chemistry , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/pathogenicity
6.
J Vis Exp ; (84): e51174, 2014 Feb 23.
Article in English | MEDLINE | ID: mdl-24637643

ABSTRACT

Pneumonia is one of the major health care problems in developing and industrialized countries and is associated with considerable morbidity and mortality. Despite advances in knowledge of this illness, the availability of intensive care units (ICU), and the use of potent antimicrobial agents and effective vaccines, the mortality rates remain high(1). Streptococcus pneumoniae is the leading pathogen of community-acquired pneumonia (CAP) and one of the most common causes of bacteremia in humans. This pathogen is equipped with an armamentarium of surface-exposed adhesins and virulence factors contributing to pneumonia and invasive pneumococcal disease (IPD). The assessment of the in vivo role of bacterial fitness or virulence factors is of utmost importance to unravel S. pneumoniae pathogenicity mechanisms. Murine models of pneumonia, bacteremia, and meningitis are being used to determine the impact of pneumococcal factors at different stages of the infection. Here we describe a protocol to monitor in real-time pneumococcal dissemination in mice after intranasal or intraperitoneal infections with bioluminescent bacteria. The results show the multiplication and dissemination of pneumococci in the lower respiratory tract and blood, which can be visualized and evaluated using an imaging system and the accompanying analysis software.


Subject(s)
Luminescent Measurements/methods , Pneumonia, Pneumococcal/microbiology , Streptococcus pneumoniae/pathogenicity , Virulence Factors/analysis , Animals , Bacteremia/blood , Bacteremia/microbiology , Disease Models, Animal , Female , Lung/microbiology , Mice , Pneumonia, Pneumococcal/blood , Streptococcus pneumoniae/chemistry , Streptococcus pneumoniae/metabolism , Virulence Factors/metabolism
7.
J Proteome Res ; 13(2): 650-67, 2014 Feb 07.
Article in English | MEDLINE | ID: mdl-24387739

ABSTRACT

Surface proteins are important for the fitness and virulence of the Gram-positive pathogen Streptococcus pneumoniae. They are crucial for interaction of the pathogen with its human host during infection. Therefore, the analysis of the pneumococcal surface proteome is an important task that requires powerful tools. In this study, two different methods, an optimized biotinylation approach and shaving with trypsin beads, were applied to study the pneumococcal surface proteome and to identify surface-exposed protein domains, respectively. The identification of nearly 95% of the predicted lipoproteins and 75% of the predicted sortase substrates reflects the high coverage of the two classical surface protein classes accomplished in this study. Furthermore, the biotinylation approach was applied to study the impact of an impaired lipoprotein maturation pathway on the cell envelope proteome and exoproteome. Loss of the lipoprotein diacylglyceryl transferase Lgt leads to striking changes in the lipoprotein distribution. Many lipoproteins disappear from the surface proteome and accumulate in the exoproteome. Further insights into lipoprotein processing in pneumococci are provided by immunoblot analyses of bacterial lysates and corresponding supernatant fractions. Taken together, the first comprehensive overview of the pneumococcal surface and exoproteome is presented, and a model for lipoprotein processing in S. pneumoniae is proposed.


Subject(s)
Bacterial Proteins/biosynthesis , Lipoproteins/biosynthesis , Proteome , Streptococcus pneumoniae/metabolism , Bacterial Proteins/metabolism , Base Sequence , Biotin/metabolism , DNA Primers , Electrophoresis, Polyacrylamide Gel , Lipoproteins/metabolism , Polymerase Chain Reaction , Subcellular Fractions/metabolism , Trypsin/metabolism
8.
EMBO Mol Med ; 5(12): 1852-70, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24136784

ABSTRACT

The respiratory pathogen Streptococcus pneumoniae has evolved efficient mechanisms to resist oxidative stress conditions and to displace other bacteria in the nasopharynx. Here we characterize at physiological, functional and structural levels two novel surface-exposed thioredoxin-family lipoproteins, Etrx1 and Etrx2. The impact of both Etrx proteins and their redox partner methionine sulfoxide reductase SpMsrAB2 on pneumococcal pathogenesis was assessed in mouse virulence studies and phagocytosis assays. The results demonstrate that loss of function of either both Etrx proteins or SpMsrAB2 dramatically attenuated pneumococcal virulence in the acute mouse pneumonia model and that Etrx proteins compensate each other. The deficiency of Etrx proteins or SpMsrAB2 further enhanced bacterial uptake by macrophages, and accelerated pneumococcal killing by H2 O2 or free methionine sulfoxides (MetSO). Moreover, the absence of both Etrx redox pathways provokes an accumulation of oxidized SpMsrAB2 in vivo. Taken together our results reveal insights into the role of two extracellular electron pathways required for reduction of SpMsrAB2 and surface-exposed MetSO. Identification of this system and its target proteins paves the way for the design of novel antimicrobials.


Subject(s)
Bacterial Proteins/metabolism , Streptococcus pneumoniae/metabolism , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Catalytic Domain , Crystallography, X-Ray , Disease Models, Animal , Female , Hydrogen Peroxide/pharmacology , Macrophages/immunology , Macrophages/physiology , Methionine/analogs & derivatives , Methionine/pharmacology , Mice , Molecular Sequence Data , Oxidative Stress/drug effects , Phagocytosis , Pneumonia/immunology , Pneumonia/microbiology , Pneumonia/mortality , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Streptococcus pneumoniae/pathogenicity , Survival Analysis , Virulence
9.
J Biol Chem ; 288(22): 15614-27, 2013 May 31.
Article in English | MEDLINE | ID: mdl-23603906

ABSTRACT

Adherence of Streptococcus pneumoniae is directly mediated by interactions of adhesins with eukaryotic cellular receptors or indirectly by exploiting matrix and serum proteins as molecular bridges. Pneumococci engage vitronectin, the human adhesive glycoprotein and complement inhibitor, to facilitate attachment to epithelial cells of the mucosal cavity, thereby modulating host cell signaling. In this study, we identified PspC as a vitronectin-binding protein interacting with the C-terminal heparin-binding domain of vitronectin. PspC is a multifunctional surface-exposed choline-binding protein displaying various adhesive properties. Vitronectin binding required the R domains in the mature PspC protein, which are also essential for the interaction with the ectodomain of the polymeric immunoglobulin receptor and secretory IgA. Consequently, secretory IgA competitively inhibited binding of vitronectin to purified PspC and to PspC-expressing pneumococci. In contrast, Factor H, which binds to the N-terminal part of mature PspC molecules, did not interfere with the PspC-vitronectin interaction. Using a series of vitronectin peptides, the C-terminal heparin-binding domain was shown to be essential for the interaction of soluble vitronectin with PspC. Binding experiments with immobilized vitronectin suggested a region N-terminal to the identified heparin-binding domain as an additional binding region for PspC, suggesting that soluble, immobilized, as well as cellularly bound vitronectin possesses different conformations. Finally, vitronectin bound to PspC was functionally active and inhibited the deposition of the terminal complement complex. In conclusion, this study identifies and characterizes (on the molecular level) the interaction between the pneumococcal adhesin PspC and the human glycoprotein vitronectin.


Subject(s)
Bacterial Proteins/metabolism , Streptococcus pneumoniae/metabolism , Vitronectin/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Humans , Protein Binding , Protein Structure, Tertiary , Streptococcus pneumoniae/chemistry , Streptococcus pneumoniae/genetics , Vitronectin/chemistry , Vitronectin/genetics
10.
J Biol Chem ; 288(22): 15654-67, 2013 May 31.
Article in English | MEDLINE | ID: mdl-23603911

ABSTRACT

Streptococcus pneumoniae is a Gram-positive human pathogen with a complex lipoteichoic acid (pnLTA) structure. Because the current structural model for pnLTA shows substantial inconsistencies, we reinvestigated purified and, more importantly, O-deacylated pnLTA, which is most suitable for NMR spectroscopy and electrospray ionization-MS spectrometry. We analyzed pnLTA of nonencapsulated pneumococcal strains D39Δcps and TIGR4Δcps, respectively. The data obtained allowed us to (re)define (i) the position and linkage of the repeating unit, (ii) the putative α-GalpNAc substitution at the ribitiol 5-phosphate (Rib-ol-5-P), and (iii) the length of (i.e. the number of repeating units in) the pnLTA chain. We here also describe for the first time that the terminal sugar residues in the pnLTA (Forssman disaccharide; α-D-GalpNAc-(1→3)-ß-D-GalpNAc-(1→)), responsible for the cross-reactivity with anti-Forssman antigen antibodies, can be heterogeneous with respect to its degree of phosphorylcholine substitution in both O-6-positions. To assess the proinflammatory potency of pnLTA, we generated a (lipopeptide-free) Δlgt mutant of strain D39Δcps, isolated its pnLTA, and showed that it is capable of inducing IL-6 release in human mononuclear cells, independent of TLR2 activation. This finding was quite in contrast to LTA of the Staphylococcus aureus SA113Δlgt mutant, which did not activate human mononuclear cells in our experiments. Remarkably, this is also contrary to various other reports showing a proinflammatory potency of S. aureus LTA. Taken together, our study refines the structure of pnLTA and indicates that pneumococcal and S. aureus LTAs differ not only in their structure but also in their bioactivity.


Subject(s)
Adjuvants, Immunologic , Leukocytes, Mononuclear/immunology , Lipopolysaccharides , Models, Molecular , Streptococcus pneumoniae/immunology , Teichoic Acids , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/genetics , Adjuvants, Immunologic/metabolism , Adjuvants, Immunologic/pharmacology , Antibodies, Bacterial/immunology , Antibodies, Heterophile/immunology , Humans , Interleukin-6/immunology , Interleukin-6/metabolism , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides/chemistry , Lipopolysaccharides/genetics , Lipopolysaccharides/immunology , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Male , Mutation , Species Specificity , Staphylococcus aureus/chemistry , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology , Staphylococcus aureus/metabolism , Streptococcus pneumoniae/chemistry , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/metabolism , Teichoic Acids/chemistry , Teichoic Acids/genetics , Teichoic Acids/immunology , Teichoic Acids/metabolism , Teichoic Acids/pharmacology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 2/metabolism
11.
J Immunol ; 188(2): 811-7, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22156592

ABSTRACT

Streptococcus pneumoniae is the leading cause of community-acquired pneumonia. In this study, we examine an innate immune recognition pathway that senses pneumococcal infection, triggers type I IFN production, and regulates RANTES production. We found that human and murine alveolar macrophages as well as murine bone marrow macrophages, but not alveolar epithelial cells, produced type I IFNs upon infection with S. pneumoniae. This response was dependent on the pore-forming toxin pneumolysin and appeared to be mediated by a cytosolic DNA-sensing pathway involving the adapter molecule STING and the transcription factor IFN regulatory factor 3. Indeed, DNA was present in the cytosol during pneumococcal infection as indicated by the activation of the AIM2 inflammasome, which is known to sense microbial DNA. Type I IFNs produced by S. pneumoniae-infected macrophages positively regulated gene expression and RANTES production in macrophages and cocultured alveolar epithelial cells in vitro. Moreover, type I IFNs controlled RANTES production during pneumococcal pneumonia in vivo. In conclusion, we identified an immune sensing pathway detecting S. pneumoniae that triggers a type I IFN response and positively regulates RANTES production.


Subject(s)
Chemokine CCL5/biosynthesis , Interferon Regulatory Factor-3/physiology , Interferon Type I/biosynthesis , Macrophages, Alveolar/immunology , Membrane Proteins/physiology , Respiratory Mucosa/immunology , Streptococcus pneumoniae/immunology , Animals , Autocrine Communication/immunology , Bacterial Proteins/physiology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Coculture Techniques , Cytosol/immunology , Cytosol/metabolism , DNA, Bacterial/immunology , DNA, Bacterial/metabolism , Disease Models, Animal , Humans , Immunity, Innate , Interferon Type I/physiology , Lung/cytology , Lung/immunology , Lung/metabolism , Macrophages, Alveolar/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Paracrine Communication/immunology , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Streptolysins/physiology
12.
J Biol Chem ; 285(6): 4099-4109, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-19933271

ABSTRACT

Studies in yeast have shown that a deficiency in Atp12p prevents assembly of the extrinsic domain (F(1)) of complex V and renders cells unable to make ATP through oxidative phosphorylation. De Meirleir et al. (De Meirleir, L., Seneca, S., Lissens, W., De Clercq, I., Eyskens, F., Gerlo, E., Smet, J., and Van Coster, R. (2004) J. Med. Genet. 41, 120-124) have reported that a homozygous missense mutation in the gene for human Atp12p (HuAtp12p), which replaces Trp-94 with Arg, was linked to the death of a 14-month-old patient. We have investigated the impact of the pathogenic W94R mutation on Atp12p structure/function. Plasmid-borne wild type human Atp12p rescues the respiratory defect of a yeast ATP12 deletion mutant (Deltaatp12). The W94R mutation alters the protein at the most highly conserved position in the Pfam sequence and renders HuAtp12p insoluble in the background of Deltaatp12. In contrast, the yeast protein harboring the corresponding mutation, ScAtp12p(W103R), is soluble in the background of Deltaatp12 but not in the background of Deltaatp12Deltafmc1, a strain that also lacks Fmc1p. Fmc1p is a yeast mitochondrial protein not found in higher eukaryotes. Tryptophan 94 (human) or 103 (yeast) is located in a positively charged region of Atp12p, and hence its mutation to arginine does not alter significantly the electrostatic properties of the protein. Instead, we provide evidence that the primary effect of the substitution is on the dynamic properties of Atp12p.


Subject(s)
Chaperonins/genetics , Molecular Chaperones/genetics , Mutation , Proton-Translocating ATPases/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Amino Acid Substitution , Arginine/genetics , Arginine/metabolism , Blotting, Western , Cells, Cultured , Chaperonins/chemistry , Chaperonins/metabolism , Electron Transport/genetics , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Genetic Complementation Test , Humans , Microscopy, Electron , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Proton-Translocating ATPases , Models, Molecular , Molecular Chaperones/metabolism , Protein Conformation , Proton-Translocating ATPases/chemistry , Proton-Translocating ATPases/metabolism , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Solubility , Static Electricity , Tryptophan/genetics , Tryptophan/metabolism
13.
J Biol Chem ; 284(25): 17138-17146, 2009 Jun 19.
Article in English | MEDLINE | ID: mdl-19383603

ABSTRACT

Mitochondrial F(1)-ATPase contains a hexamer of alternating alpha and beta subunits. The assembly of this structure requires two specialized chaperones, Atp11p and Atp12p, that bind transiently to beta and alpha. In the absence of Atp11p and Atp12p, the hexamer is not formed, and alpha and beta precipitate as large insoluble aggregates. An early model for the mechanism of chaperone-mediated F(1) assembly (Wang, Z. G., Sheluho, D., Gatti, D. L., and Ackerman, S. H. (2000) EMBO J. 19, 1486-1493) hypothesized that the chaperones themselves look very much like the alpha and beta subunits, and proposed an exchange of Atp11p for alpha and of Atp12p for beta; the driving force for the exchange was expected to be a higher affinity of alpha and beta for each other than for the respective chaperone partners. One important feature of this model was the prediction that as long as Atp11p is bound to beta and Atp12p is bound to alpha, the two F(1) subunits cannot interact at either the catalytic site or the noncatalytic site interface. Here we present the structures of Atp11p from Candida glabrata and Atp12p from Paracoccus denitrificans, and we show that some features of the Wang model are correct, namely that binding of the chaperones to alpha and beta prevents further interactions between these F(1) subunits. However, Atp11p and Atp12p do not resemble alpha or beta, and it is instead the F(1) gamma subunit that initiates the release of the chaperones from alpha and beta and their further assembly into the mature complex.


Subject(s)
Mitochondrial Proton-Translocating ATPases/chemistry , Mitochondrial Proton-Translocating ATPases/metabolism , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Candida glabrata/genetics , Candida glabrata/metabolism , Crystallography, X-Ray , DNA Primers/genetics , Fungal Proteins/genetics , Fungal Proteins/metabolism , Mitochondrial Proton-Translocating ATPases/genetics , Models, Molecular , Molecular Chaperones/genetics , Molecular Sequence Data , Multiprotein Complexes , Mutagenesis, Site-Directed , Paracoccus denitrificans/genetics , Paracoccus denitrificans/metabolism , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Sequence Homology, Amino Acid
14.
J Neurosci ; 25(30): 7004-13, 2005 Jul 27.
Article in English | MEDLINE | ID: mdl-16049176

ABSTRACT

The myelin basic protein (MBP) gene encodes two families of proteins, the classic MBP constituents of myelin and the golli-MBPs, the function of which is less well understood. In this study, targeted ablation of the golli-MBPs, but not the classic MBPs, resulted in a distinct phenotype unlike that of knock-outs (KOs) of the classic MBPs or other myelin proteins. Although the golli KO animals did not display an overt dysmyelinating phenotype, they did exhibit delayed and/or hypomyelination in selected areas of the brain, such as the visual cortex and the optic nerve, as determined by Northern and Western blots and immunohistochemical analysis with myelin protein markers. Hypomyelination in some areas, such as the visual cortex, persisted into adulthood. Ultrastructural analysis of the KOs confirmed both the delay and hypomyelination and revealed abnormalities in myelin structure and in some oligodendrocytes. Abnormal visual-evoked potentials indicated that the hypomyelination in the visual cortex had functional consequences in the golli KO brain. Evidence that the abnormal myelination in these animals was a consequence of intrinsic problems with the oligodendrocyte was indicated by an impaired ability of oligodendrocytes to form myelin sheets in culture and by the presence of abnormal Ca2+ transients in purified cortical oligodendrocytes studied in vitro. The Ca2+ results reported in this study complement previous results implicating golli proteins in modulating intracellular signaling in T-cells. Together, all these findings suggest a role for golli proteins in oligodendrocyte differentiation, migration, and/or myelin elaboration in the brain.


Subject(s)
Myelin Sheath/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Oligodendroglia/pathology , Optic Nerve/pathology , Transcription Factors/genetics , Transcription Factors/physiology , Visual Cortex/pathology , Animals , Calcium/metabolism , Female , Homeostasis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Electron , Myelin Basic Protein , Nerve Fibers, Myelinated/pathology , Nerve Fibers, Myelinated/ultrastructure , Oligodendroglia/metabolism
15.
J Neuroimmunol ; 139(1-2): 44-50, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12799019

ABSTRACT

Increased golli MBP (golli) expression has been observed in the peripheral immune system of mice in the relapsing phase of EAE, raising the possibility that golli MBP expression in the periphery may contribute to relapses. Here we describe the generation of golli MBP-deficient mice and a comparison of the clinical course of EAE between heterozygous (golli(+/-)) and wild-type (golli(+/+)) mice. There was no difference between the two groups in incidence of disease, severity of the first episode of disease, or remission after the first episode. However, there was a significant reduction in relapses in golli(+/-) mice vs. controls, suggesting a role for golli proteins in the relapses in EAE.


Subject(s)
Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Multiple Sclerosis/genetics , Myelin Basic Protein/deficiency , Myelin Sheath/immunology , Nerve Tissue Proteins/deficiency , Transcription Factors/deficiency , Animals , Central Nervous System/pathology , Central Nervous System/physiopathology , Disease Models, Animal , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Gene Expression Regulation/immunology , Heterozygote , Immunity, Cellular/genetics , Male , Mice , Mice, Knockout , Multiple Sclerosis/immunology , Multiple Sclerosis/physiopathology , Myelin Basic Protein/genetics , Myelin Sheath/metabolism , Myelin Sheath/pathology , Nerve Tissue Proteins/genetics , Recurrence , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...