Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
2.
Cell Chem Biol ; 28(6): 802-812.e6, 2021 06 17.
Article in English | MEDLINE | ID: mdl-33333026

ABSTRACT

The recent development of successful CAR (chimeric antigen receptor) T cell therapies has been accompanied by a need to better control potentially fatal toxicities that can arise from adverse immune reactions. Here we present a ligand-controlled CAR system, based on the IKZF3 ZF2 ß-hairpin IMiD-inducible degron, which allows for the reversible control of expression levels of type I membrane proteins, including CARs. Testing this system in an established mouse xenotransplantation model for acute lymphoblastic leukemia, we validate the ability of the CAR19-degron to target and kill CD19-positive cells displaying complete control/clearance of the tumor. We also demonstrate that the activity of CAR19-degron can be regulated in vivo when dosing a US Food and Drug Administration-approved drug, lenalidomide.


Subject(s)
Ikaros Transcription Factor/immunology , Immunologic Factors/pharmacology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Adolescent , Animals , Cell Line , Cell Proliferation/drug effects , Female , Humans , Ikaros Transcription Factor/chemistry , Immunologic Factors/chemistry , Male , Mice , Mice, Congenic , Mice, Inbred NOD , Mice, SCID , Middle Aged , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Receptors, Chimeric Antigen/genetics , Young Adult
3.
Nat Protoc ; 10(3): 508-16, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25719269

ABSTRACT

Transpeptidation catalyzed by sortase A allows the preparation of proteins that are site-specifically and homogeneously modified with a wide variety of functional groups, such as fluorophores, PEG moieties, lipids, glycans, bio-orthogonal reactive groups and affinity handles. This protocol describes immobilization of sortase A on a solid support (Sepharose beads). Immobilization of sortase A simplifies downstream purification of a protein of interest after labeling of its N or C terminus. Smaller batch and larger-scale continuous-flow reactions require only a limited amount of enzyme. The immobilized enzyme can be reused for multiple cycles of protein modification reactions. The described protocol also works with a Ca(2+)-independent variant of sortase A with increased catalytic activity. This heptamutant variant of sortase A (7M) was generated by combining previously published mutations, and this immobilized enzyme can be used for the modification of calcium-senstive substrates or in instances in which low temperatures are needed. Preparation of immobilized sortase A takes 1-2 d. Batch reactions take 3-12 h and flow reactions proceed at 0.5 ml h(-1), depending on the geometry of the reactor used.


Subject(s)
Aminoacyltransferases/metabolism , Bacterial Proteins/metabolism , Cysteine Endopeptidases/metabolism , Enzymes, Immobilized/metabolism , Peptidyl Transferases/metabolism , Protein Engineering/methods , Proteins/metabolism , Aminoacyltransferases/genetics , Bacterial Proteins/genetics , Catalysis , Cysteine Endopeptidases/genetics , Mutation/genetics , Sepharose
4.
PLoS One ; 9(10): e109883, 2014.
Article in English | MEDLINE | ID: mdl-25275512

ABSTRACT

Aerolysin is a secreted bacterial toxin that perforates the plasma membrane of a target cell with lethal consequences. Previously explored native and epitope-tagged forms of the toxin do not allow site-specific modification of the mature toxin with a probe of choice. We explore sortase-mediated transpeptidation reactions (sortagging) to install fluorophores and biotin at three distinct sites in aerolysin, without impairing binding of the toxin to the cell membrane and with minimal impact on toxicity. Using a version of aerolysin labeled with different fluorophores at two distinct sites we followed the fate of the C-terminal peptide independently from the N-terminal part of the toxin, and show its loss in the course of intoxication. Making use of the biotinylated version of aerolysin, we identify mesothelin, urokinase plasminogen activator surface receptor (uPAR, CD87), glypican-1, and CD59 glycoprotein as aerolysin receptors, all predicted or known to be modified with a glycosylphosphatidylinositol anchor. The sortase-mediated reactions reported here can be readily extended to other pore forming proteins.


Subject(s)
Aeromonas hydrophila/metabolism , Bacterial Toxins/metabolism , CD59 Antigens/metabolism , Glypicans/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism , Aeromonas hydrophila/chemistry , Amino Acid Sequence , Animals , Bacterial Toxins/analysis , Biotin/chemistry , CD59 Antigens/analysis , Cell Line , Fluorescent Dyes/chemistry , Glypicans/analysis , Humans , Molecular Sequence Data , Optical Imaging , Pore Forming Cytotoxic Proteins/analysis , Receptors, Urokinase Plasminogen Activator/analysis
5.
J Biol Chem ; 289(35): 24005-18, 2014 Aug 29.
Article in English | MEDLINE | ID: mdl-25031321

ABSTRACT

A number of toxins, including exotoxin A (PE) of Pseudomonas aeruginosa, kill cells by inhibiting protein synthesis. PE kills by ADP-ribosylation of the translation elongation factor 2, but many of the host factors required for entry, membrane translocation, and intracellular transport remain to be elucidated. A genome-wide genetic screen in human KBM7 cells was performed to uncover host factors used by PE, several of which were confirmed by CRISPR/Cas9-gene editing in a different cell type. Several proteins not previously implicated in the PE intoxication pathway were identified, including GPR107, an orphan G-protein-coupled receptor. GPR107 localizes to the trans-Golgi network and is essential for retrograde transport. It is cleaved by the endoprotease furin, and a disulfide bond connects the two cleaved fragments. Compromising this association affects the function of GPR107. The N-terminal region of GPR107 is critical for its biological function. GPR107 might be one of the long-sought receptors that associates with G-proteins to regulate intracellular vesicular transport.


Subject(s)
ADP Ribose Transferases/toxicity , Bacterial Toxins/toxicity , Exotoxins/toxicity , Furin/metabolism , Receptors, G-Protein-Coupled/metabolism , Virulence Factors/toxicity , trans-Golgi Network/metabolism , ADP Ribose Transferases/genetics , Bacterial Toxins/genetics , Base Sequence , DNA Primers , Endocytosis , Exotoxins/genetics , Mutation , Polymerase Chain Reaction , Protein Transport , Proteolysis , Receptors, G-Protein-Coupled/physiology , Virulence Factors/genetics , Pseudomonas aeruginosa Exotoxin A
6.
Nat Protoc ; 8(9): 1787-99, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23989673

ABSTRACT

Methods for site-specific modification of proteins should be quantitative and versatile with respect to the nature and size of the biological or chemical targets involved. They should require minimal modification of the target, and the underlying reactions should be completed in a reasonable amount of time under physiological conditions. Sortase-mediated transpeptidation reactions meet these criteria and are compatible with other labeling methods. Here we describe the expression and purification conditions for two sortase A enzymes that have different recognition sequences. We also provide a protocol that allows the functionalization of any given protein at its C terminus, or, for select proteins, at an internal site. The target protein is engineered with a sortase-recognition motif (LPXTG) at the place where modification is desired. Upon recognition, sortase cleaves the protein between the threonine and glycine residues, facilitating the attachment of an exogenously added oligoglycine peptide modified with the functional group of choice (e.g., fluorophore, biotin, protein or lipid). Expression and purification of sortase takes ∼3 d, and sortase-mediated reactions take only a few minutes, but reaction times can be extended to increase yields.


Subject(s)
Aminoacyltransferases/chemistry , Bacterial Proteins/chemistry , Cysteine Endopeptidases/chemistry , Protein Engineering/methods , Amino Acid Sequence , Cell Wall/metabolism , Chromatography, High Pressure Liquid , Chromatography, Liquid , Mass Spectrometry
7.
Nat Protoc ; 8(9): 1800-7, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23989674

ABSTRACT

This protocol describes the use of sortase-mediated reactions to label the N terminus of any given protein of interest. The sortase recognition sequence, LPXTG (for Streptococcus aureus sortase A) or LPXTA (for Staphylococcus pyogenes sortase A), can be appended to a variety of probes such as fluorophores, biotin or even to other proteins. The protein to be labeled acts as a nucleophile by attacking the intermediate formed between the probe containing the LPXTG/A motif and the sortase enzyme. If sortase, the protein of interest and a suitably functionalized label are available, the reactions usually require less than 3 h.


Subject(s)
Aminoacyltransferases/chemistry , Bacterial Proteins/chemistry , Cysteine Endopeptidases/chemistry , Protein Engineering/methods , Chromatography, High Pressure Liquid , Kinetics , Staphylococcus aureus/enzymology , Staphylococcus aureus/genetics , Streptococcus pyogenes/enzymology , Streptococcus pyogenes/genetics , Substrate Specificity
8.
Nat Protoc ; 8(9): 1808-19, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23989675

ABSTRACT

Chimeric proteins, including bispecific antibodies, are biological tools with therapeutic applications. Genetic fusion and ligation methods allow the creation of N-to-C and C-to-N fused recombinant proteins, but not unnaturally linked N-to-N and C-to-C fusion proteins. This protocol describes a simple procedure for the production of such chimeric proteins, starting from correctly folded proteins and readily available peptides. By equipping the N terminus or C terminus of the proteins of interest with a set of click handles using sortase A, followed by a strain-promoted click reaction, unnatural N-to-N and C-to-C linked (hetero) fusion proteins are established. Examples of proteins that have been conjugated via this method include interleukin-2, interferon-α, ubiquitin, antibodies and several single-domain antibodies. If the peptides, sortase A and the proteins of interest are in hand, the unnaturally N-to-N and C-to-C fused proteins can be obtained in 3-4 d.


Subject(s)
Aminoacyltransferases/chemistry , Bacterial Proteins/chemistry , Click Chemistry/methods , Cysteine Endopeptidases/chemistry , Protein Engineering/methods , Carbon/chemistry , Chromatography, High Pressure Liquid , Nitrogen/chemistry , Peptide Fragments/biosynthesis , Protein Folding , Recombinant Fusion Proteins/chemistry , Staphylococcus aureus/enzymology
9.
ACS Synth Biol ; 2(9): 490-6, 2013 Sep 20.
Article in English | MEDLINE | ID: mdl-23713956

ABSTRACT

M13 bacteriophage has been used as a scaffold to organize materials for various applications. Building more complex multiphage devices requires precise control of interactions between the M13 capsid proteins. Toward this end, we engineered a loop structure onto the pIII capsid protein of M13 bacteriophage to enable sortase-mediated labeling reactions for C-terminal display. Combining this with N-terminal sortase-mediated labeling, we thus created a phage scaffold that can be labeled orthogonally on three capsid proteins: the body and both ends. We show that covalent attachment of different DNA oligonucleotides at the ends of the new phage structure enables formation of multiphage particles oriented in a specific order. These have potential as nanoscale scaffolds for multi-material devices.


Subject(s)
Bacteriophage M13/genetics , Bacteriophage M13/metabolism , Capsid Proteins/chemistry , DNA, Viral/metabolism , Amino Acid Motifs , Bacteriophage M13/chemistry , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/metabolism , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Nucleic Acid Hybridization/methods , Optical Imaging , Protein Engineering
10.
Proc Natl Acad Sci U S A ; 110(16): 6406-11, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23576732

ABSTRACT

Cells genetically deficient in sphingomyelin synthase-1 (SGMS1) or blocked in their synthesis pharmacologically through exposure to a serine palmitoyltransferase inhibitor (myriocin) show strongly reduced surface display of influenza virus glycoproteins hemagglutinin (HA) and neuraminidase (NA). The transport of HA to the cell surface was assessed by accessibility of HA on intact cells to exogenously added trypsin and to HA-specific antibodies. Rates of de novo synthesis of viral proteins in wild-type and SGMS1-deficient cells were equivalent, and HA negotiated the intracellular trafficking pathway through the Golgi normally. We engineered a strain of influenza virus to allow site-specific labeling of HA and NA using sortase. Accessibility of both HA and NA to sortase was blocked in SGMS1-deficient cells and in cells exposed to myriocin, with a corresponding inhibition of the release of virus particles from infected cells. Generation of influenza virus particles thus critically relies on a functional sphingomyelin biosynthetic pathway, required to drive influenza viral glycoproteins into lipid domains of a composition compatible with virus budding and release.


Subject(s)
Biosynthetic Pathways/physiology , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Sphingomyelins/biosynthesis , Animals , Biological Transport/physiology , Dogs , Fatty Acids, Monounsaturated/pharmacology , Fluorescent Antibody Technique , Hemagglutinin Glycoproteins, Influenza Virus/physiology , Host-Pathogen Interactions , Madin Darby Canine Kidney Cells , Polyethylene Glycols , Serine C-Palmitoyltransferase/antagonists & inhibitors , Transferases (Other Substituted Phosphate Groups)/deficiency , Trypsin
11.
Proc Natl Acad Sci U S A ; 110(4): 1428-33, 2013 Jan 22.
Article in English | MEDLINE | ID: mdl-23297227

ABSTRACT

A monoclonal antibody against the C-type lectin DEC205 (αDEC205) is an effective vehicle for delivery of antigens to dendritic cells through creation of covalent αDEC205-antigen adducts. These adducts can induce antigen-specific T-cell immune responses or tolerance. We exploit the transpeptidase activity of sortase to install modified peptides and protein-sized antigens onto the heavy chain of αDEC205, including linkers that contain nonnatural amino acids. We demonstrate stoichiometric site-specific labeling on a scale not easily achievable by genetic fusions (49 distinct fusions in this report). We conjugated a biotinylated version of a class I MHC-restricted epitope to unlabeled αDEC205 and monitored epitope generation upon binding of the adduct to dendritic cells. Our results show transfer of αDEC205 heavy chain to the cytoplasm, followed by proteasomal degradation. Introduction of a labile dipeptide linker at the N terminus of a T-cell epitope improves proteasome-dependent class I MHC-restricted peptide cross-presentation when delivered by αDEC205 in vitro and in vivo. We also conjugated αDEC205 with a linker-optimized peptide library of known CD8 T-cell epitopes from the mouse γ-herpes virus 68. Animals immunized with such conjugates displayed a 10-fold reduction in viral load.


Subject(s)
Aminoacyltransferases/metabolism , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Viral/immunology , Bacterial Proteins/metabolism , Cysteine Endopeptidases/metabolism , Epitopes, T-Lymphocyte/immunology , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Amino Acid Sequence , Animals , Antigen Presentation , Antigens, CD/chemistry , Antigens, CD/genetics , Antigens, Viral/chemistry , Antigens, Viral/genetics , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/genetics , Herpesviridae Infections/immunology , Herpesviridae Infections/prevention & control , Histocompatibility Antigens Class I/metabolism , Immunization , Immunoconjugates/genetics , Immunoconjugates/immunology , Immunoconjugates/metabolism , Lectins, C-Type/chemistry , Lectins, C-Type/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Minor Histocompatibility Antigens , Molecular Sequence Data , Proteasome Endopeptidase Complex/metabolism , Protein Engineering , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Rhadinovirus/genetics , Rhadinovirus/immunology , Tumor Virus Infections/immunology , Tumor Virus Infections/prevention & control , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Viral Vaccines/genetics , Viral Vaccines/immunology
12.
Bioconjug Chem ; 23(7): 1478-87, 2012 Jul 18.
Article in English | MEDLINE | ID: mdl-22759232

ABSTRACT

We exploit bacterial sortases to attach a variety of moieties to the capsid proteins of M13 bacteriophage. We show that pIII, pIX, and pVIII can be functionalized with entities ranging from small molecules (e.g., fluorophores, biotin) to correctly folded proteins (e.g., GFP, antibodies, streptavidin) in a site-specific manner, and with yields that surpass those of any reported using phage display technology. A case in point is modification of pVIII. While a phage vector limits the size of the insert into pVIII to a few amino acids, a phagemid system limits the number of copies actually displayed at the surface of M13. Using sortase-based reactions, a 100-fold increase in the efficiency of display of GFP onto pVIII is achieved. Taking advantage of orthogonal sortases, we can simultaneously target two distinct capsid proteins in the same phage particle and maintain excellent specificity of labeling. As demonstrated in this work, this is a simple and effective method for creating a variety of structures, thus expanding the use of M13 for materials science applications and as a biological tool.


Subject(s)
Aminoacyltransferases/metabolism , Bacterial Proteins/metabolism , Bacteriophage M13/metabolism , Capsid Proteins/metabolism , Cell Surface Display Techniques/methods , Cysteine Endopeptidases/metabolism , Animals , Bacteriophage M13/chemistry , Bacteriophage M13/genetics , Capsid Proteins/chemistry , Capsid Proteins/genetics , Mice , Mice, Inbred C57BL , Surface Properties
13.
Proc Natl Acad Sci U S A ; 109(6): 2072-7, 2012 Feb 07.
Article in English | MEDLINE | ID: mdl-22308317

ABSTRACT

The ability to induce humoral and cellular immunity via antigen delivery through the unbroken skin (epicutaneous immunization, EPI) has immediate relevance for vaccine development. However, it is unclear which adjuvants induce protective memory CD8 T-cell responses by this route, and the molecular and cellular requirements for priming through intact skin are not defined. We report that cholera toxin (CT) is superior to other adjuvants in its ability to prime memory CD8 T cells that control bacterial and viral challenges. Epicutaneous immunization with CT does not require engagement of classic toll-like receptor (TLR) and inflammasome pathways and, surprisingly, is independent of skin langerin-expressing cells (including Langerhans cells). However, CT adjuvanticity required type-I IFN sensitivity, participation of a Batf3-dependent dendritic cell (DC) population and engagement of CT with suitable gangliosides. Chemoenzymatic generation of CT-antigen fusion proteins led to efficient priming of the CD8 T-cell responses, paving the way for development of this immunization strategy as a therapeutic option.


Subject(s)
Adjuvants, Immunologic/metabolism , CD8-Positive T-Lymphocytes/immunology , Cholera Toxin/administration & dosage , Cholera Toxin/immunology , Signal Transduction/drug effects , Vaccination , Administration, Cutaneous , Animals , Antigens/immunology , Antigens, Surface/metabolism , Basic-Leucine Zipper Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/drug effects , Cells, Cultured , G(M1) Ganglioside/immunology , Immunologic Memory/drug effects , Interferon Type I/immunology , Lectins, C-Type/metabolism , Mannose-Binding Lectins/metabolism , Mice , Mice, Inbred C57BL , Oligodeoxyribonucleotides/pharmacology , Repressor Proteins/metabolism , Signal Transduction/immunology , Toll-Like Receptors/immunology
14.
J Cell Biol ; 195(5): 751-64, 2011 Nov 28.
Article in English | MEDLINE | ID: mdl-22123862

ABSTRACT

We describe a novel labeling strategy to site-specifically attach fluorophores, biotin, and proteins to the C terminus of the A1 subunit (CTA1) of cholera toxin (CTx) in an otherwise correctly assembled and active CTx complex. Using a biotinylated N-linked glycosylation reporter peptide attached to CTA1, we provide direct evidence that ~12% of the internalized CTA1 pool reaches the ER. We also explored the sortase labeling method to attach the catalytic subunit of diphtheria toxin as a toxic warhead to CTA1, thus converting CTx into a cytolethal toxin. This new toxin conjugate enabled us to conduct a genetic screen in human cells, which identified ST3GAL5, SLC35A2, B3GALT4, UGCG, and ELF4 as genes essential for CTx intoxication. The first four encode proteins involved in the synthesis of gangliosides, which are known receptors for CTx. Identification and isolation of the ST3GAL5 and SLC35A2 mutant clonal cells uncover a previously unappreciated differential contribution of gangliosides to intoxication by CTx.


Subject(s)
Cholera Toxin/toxicity , Diphtheria Toxin/chemistry , Protein Transport/genetics , Aminoacyltransferases/chemistry , Aminoacyltransferases/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , Cells, Cultured , Cholera/physiopathology , Cholera Toxin/chemistry , Cholera Toxin/genetics , Cloning, Molecular , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/genetics , Gangliosides/metabolism , Gangliosides/physiology , Genetic Engineering , Haploidy , Humans , Molecular Sequence Data , Monosaccharide Transport Proteins/chemistry , Monosaccharide Transport Proteins/metabolism , Monosaccharide Transport Proteins/physiology , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/toxicity , Sequence Analysis, Protein , Sialyltransferases/chemistry , Sialyltransferases/metabolism , Sialyltransferases/physiology
15.
Nat Biotechnol ; 29(6): 542-6, 2011 May 29.
Article in English | MEDLINE | ID: mdl-21623355

ABSTRACT

Insertional mutagenesis in a haploid background can disrupt gene function. We extend our earlier work by using a retroviral gene-trap vector to generate insertions in >98% of the genes expressed in a human cancer cell line that is haploid for all but one of its chromosomes. We apply phenotypic interrogation via tag sequencing (PhITSeq) to examine millions of mutant alleles through selection and parallel sequencing. Analysis of pools of cells, rather than individual clones enables rapid assessment of the spectrum of genes involved in the phenotypes under study. This facilitates comparative screens as illustrated here for the family of cytolethal distending toxins (CDTs). CDTs are virulence factors secreted by a variety of pathogenic Gram-negative bacteria responsible for tissue damage at distinct anatomical sites. We identify 743 mutations distributed over 12 human genes important for intoxication by four different CDTs. Although related CDTs may share host factors, they also exploit unique host factors to yield a profile characteristic for each CDT.


Subject(s)
Genetic Association Studies/methods , High-Throughput Nucleotide Sequencing/methods , Phenotype , Alleles , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Bacterial Toxins/toxicity , Cell Line, Tumor , Chromosomes, Human , Cloning, Molecular , Genome, Human , Gram-Negative Bacteria/pathogenicity , Haploidy , Humans , Mutagenesis, Insertional , Mutation , RNA Interference , Sequence Tagged Sites , Virulence Factors/analysis , Virulence Factors/metabolism
16.
FASEB J ; 25(8): 2650-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21525488

ABSTRACT

Cyclic peptides are highly valued tools in biomedical research. In many cases, they show higher receptor affinity, enhanced biological activity, and improved serum stability. Technical difficulties in producing cyclic peptides, especially larger ones, in appreciable yields have precluded a prolific use in biomedical research. Here, we describe a novel and efficient cyclization method that uses the peptidyl-transferase activity of the Staphylococcus aureus enzyme sortase A to cyclize linear synthetic precursor peptides. As a model, we used histatin 1, a 38-mer salivary peptide with motogenic activity. Chemical cyclization of histatin 1 resulted in ≤ 3% yields, whereas sortase-mediated cyclization provided a yield of >90%. The sortase-cyclized peptide displayed a maximum wound closure activity at 10 nM, whereas the linear peptide displayed maximal activity at 10 µM. Circular dichroism and NMR spectroscopic analysis of the linear and cyclic peptide in solution showed no evidence for conformational changes, suggesting that structural differences due to cyclization only became manifest when these peptides were located in the binding domain of the receptor. The sortase-based cyclization technology provides a general method for easy and efficient manufacturing of large cyclic peptides.


Subject(s)
Aminoacyltransferases/metabolism , Bacterial Proteins/metabolism , Cysteine Endopeptidases/metabolism , Histatins/biosynthesis , Peptides, Cyclic/biosynthesis , Amino Acid Sequence , Aminoacyltransferases/genetics , Bacterial Proteins/genetics , Cell Line , Circular Dichroism , Cysteine Endopeptidases/genetics , Histatins/chemistry , Histatins/genetics , Histatins/pharmacology , Humans , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Peptides, Cyclic/chemistry , Peptides, Cyclic/genetics , Peptides, Cyclic/pharmacology , Protein Conformation , Protein Engineering , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Staphylococcus aureus/enzymology , Staphylococcus aureus/genetics , Wound Healing/drug effects
17.
Science ; 326(5957): 1231-5, 2009 Nov 27.
Article in English | MEDLINE | ID: mdl-19965467

ABSTRACT

Loss-of-function genetic screens in model organisms have elucidated numerous biological processes, but the diploid genome of mammalian cells has precluded large-scale gene disruption. We used insertional mutagenesis to develop a screening method to generate null alleles in a human cell line haploid for all chromosomes except chromosome 8. Using this approach, we identified host factors essential for infection with influenza and genes encoding important elements of the biosynthetic pathway of diphthamide, which are required for the cytotoxic effects of diphtheria toxin and exotoxin A. We also identified genes needed for the action of cytolethal distending toxin, including a cell-surface protein that interacts with the toxin. This approach has both conceptual and practical parallels with genetic approaches in haploid yeast.


Subject(s)
Bacterial Toxins/metabolism , Genetic Testing , Haploidy , Host-Pathogen Interactions , Influenza A Virus, H1N1 Subtype/pathogenicity , ADP Ribose Transferases/metabolism , ADP Ribose Transferases/toxicity , Adenosine Diphosphate Ribose/metabolism , Amino Acid Sequence , Antigens, Bacterial/metabolism , Antigens, Bacterial/toxicity , Bacterial Toxins/toxicity , Biosynthetic Pathways , Carboxylic Ester Hydrolases , Cell Line, Tumor , Diphtheria Toxin/metabolism , Diphtheria Toxin/toxicity , Exotoxins/metabolism , Exotoxins/toxicity , Genes , Histidine/analogs & derivatives , Histidine/biosynthesis , Humans , Methyltransferases , Molecular Sequence Data , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , Mutagenesis, Insertional , N-Acylneuraminate Cytidylyltransferase/genetics , N-Acylneuraminate Cytidylyltransferase/metabolism , Peptide Elongation Factor 2/metabolism , Proteins/chemistry , Proteins/genetics , Proteins/metabolism , Virulence Factors/metabolism , Virulence Factors/toxicity , Pseudomonas aeruginosa Exotoxin A
18.
J Am Chem Soc ; 131(31): 10800-1, 2009 Aug 12.
Article in English | MEDLINE | ID: mdl-19610623

ABSTRACT

The unique reactivity of two sortase enzymes, SrtA(staph) from Staphylococcus aureus and SrtA(strep) from Streptococcus pyogenes, is exploited for site-specific labeling of a single polypeptide with different labels at its N and C termini. SrtA(strep) is used to label the protein's C terminus at an LPXTG site with a fluorescently labeled dialanine nucleophile. Selective N-terminal labeling of proteins containing N-terminal glycine residues is achieved using SrtA(staph) and LPXT derivatives. The generality of N-terminal labeling with SrtA(staph) is demonstrated by near-quantitative labeling of multiple protein substrates with excellent site specificity.


Subject(s)
Aminoacyltransferases/metabolism , Bacterial Proteins/metabolism , Cysteine Endopeptidases/metabolism , Fluorescent Dyes/chemistry , Peptides/chemistry , Alanine , Amino Acid Sequence , Binding Sites , Staphylococcus aureus/enzymology , Streptococcus pyogenes/enzymology , Substrate Specificity
19.
J Hum Genet ; 50(2): 99-105, 2005.
Article in English | MEDLINE | ID: mdl-15682271

ABSTRACT

The adrenoleukodystrophy protein (ALDP) is a half-ABC (ATP-binding cassette) transporter localized in the peroxisomal membrane. Dysfunction of this protein is the cause of the human genetic disorder X-linked adrenoleukodystrophy (X-ALD), which is characterized by accumulation of saturated, very-long-chain fatty acids (VLCFAs). This observation suggests that ALDP is involved in the metabolism of these compounds. Whether ALDP transports VLCFAs or their derivatives across the peroxisomal membrane or some cofactors essential for the efficient peroxisomal beta-oxidation of these fatty acids is still unknown. In this work, we used a protease-based approach to search for substrate-induced conformational alterations on ALDP. Our results suggest that ALDP is directly involved in the transport of long- and very-long-chain acyl-CoAs across the peroxisomal membrane.


Subject(s)
ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Acyl Coenzyme A/pharmacokinetics , Adrenoleukodystrophy/genetics , Adrenoleukodystrophy/physiopathology , Peptide Hydrolases/pharmacology , ATP Binding Cassette Transporter, Subfamily D, Member 1 , Humans , Oxidation-Reduction , Peroxisomes/physiology , Placenta/chemistry , Protein Conformation , Reverse Transcriptase Polymerase Chain Reaction
20.
Cell Biochem Biophys ; 41(3): 451-68, 2004.
Article in English | MEDLINE | ID: mdl-15509892

ABSTRACT

Peroxisomal matrix proteins are synthesized on free cytosolic ribosomes and posttranslationally imported into the organelle. Translocation of these newly synthesized proteins across the peroxisomal membrane requires the concerted action of many different proteins, the majority of which were already identified. However, not much is known regarding the mechanism of protein translocation across this membrane system. Here, we discuss recent mechanistic and structural data. These results point to a model in which proteins en route to the peroxisomal matrix are translocated across the organelle membrane by their own receptor in a process that occurs through a large membrane protein assembly.


Subject(s)
Biophysics/methods , Membrane Proteins/chemistry , Peroxisomes/metabolism , Protein Transport , Saccharomyces cerevisiae/metabolism , Cytoplasm/metabolism , Intracellular Membranes/metabolism , Membrane Transport Proteins/chemistry , Models, Biological , Organelles , Peroxisomal Targeting Signal 2 Receptor , Peroxisome-Targeting Signal 1 Receptor , Phenotype , Pichia/metabolism , Protein Binding , Receptors, Cytoplasmic and Nuclear/chemistry , Saccharomyces cerevisiae Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...