Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Front Immunol ; 14: 1221108, 2023.
Article in English | MEDLINE | ID: mdl-37828992

ABSTRACT

Staphylococcus aureus pathology is caused by a plethora of virulence factors able to combat multiple host defence mechanisms. Fibrinogen (Fg), a critical component in the host coagulation cascade, plays an important role in the pathogenesis of this bacterium, as it is the target of numerous staphylococcal virulence proteins. Amongst its secreted virulence factors, coagulase (Coa) and Extracellular fibrinogen-binding protein (Efb) share common Fg binding motives and have been described to form a Fg shield around staphylococcal cells, thereby allowing efficient bacterial spreading, phagocytosis escape and evasion of host immune system responses. Targeting these proteins with monoclonal antibodies thus represents a new therapeutic option against S. aureus. To this end, here we report the selection and characterization of fully human, sequence-defined, monoclonal antibodies selected against the C-terminal of coagulase. Given the functional homology between Coa and Efb, we also investigated if the generated antibodies bound the two virulence factors. Thirteen unique antibodies were isolated from naïve antibodies gene libraries by antibody phage display. As anticipated, most of the selected antibodies showed cross-recognition of these two proteins and among them, four were able to block the interaction between Coa/Efb and Fg. Furthermore, our monoclonal antibodies could interact with the two main Fg binding repeats present at the C-terminal of Coa and distinguish them, suggesting the presence of two functionally different Fg-binding epitopes.


Subject(s)
Coagulase , Staphylococcal Infections , Humans , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Bacterial Proteins , Coagulase/immunology , Fibrinogen/chemistry , Fibrinogen/metabolism , Phagocytosis , Staphylococcus aureus , Virulence Factors/metabolism , Binding Sites, Antibody
2.
Transl Vis Sci Technol ; 11(10): 37, 2022 10 03.
Article in English | MEDLINE | ID: mdl-36301553

ABSTRACT

Purpose: Complement alternative pathway (AP) dysregulation has been implicated in geographic atrophy, an advanced form of age-related macular degeneration. Danicopan is an investigational, first-in-class inhibitor of factor D, an essential AP activation enzyme. We assessed danicopan distribution to the posterior segment of the eye after oral dosing. Methods: Tissue distribution of drug-derived radioactivity was evaluated using whole-body autoradiography following oral administration of [14C]-danicopan to pigmented and albino rats. Pharmacokinetics and ocular tissue distribution were studied in pigmented and albino rabbits following single and multiple oral dosing of danicopan. The melanin binding property was characterized in vitro. Results: Radioactivity was distributed widely in rats and became nonquantifiable in most tissues 24 hours postdose except in the pigmented rat uvea (quantifiable 672 hours postdose). Danicopan binding to melanin was established in vitro. After single dosing, the maximum concentration (Cmax) and area under the curve (AUC) in neural retina and plasma were similar in both rabbit types. After multiple dosing, AUC in neural retina was 3.4-fold higher versus plasma in pigmented rabbits. Drug levels in choroid/Bruch's membrane (BrM)/retinal pigment epithelium (RPE) were similar to plasma in albino rabbits but higher in pigmented rabbits: Cmax and AUC were 2.9- and 23.8-fold higher versus plasma after single dosing and 5.8- and 62.7-fold higher after multiple dosing. In pigmented rabbits, ocular tissue exposures slowly declined over time but remained quantifiable 240 hours postdose. Conclusions: The results demonstrate that danicopan crosses the blood-retina barrier and binds melanin reversibly, leading to a higher and more sustained exposure in melanin-containing ocular tissues (choroid/BrM/RPE) and in the neural retina as compared to in plasma after repeated oral dosing in pigmented animals. Translational Relevance: These findings suggest that oral danicopan possesses potential for treating geographic atrophy because AP dysregulation in the posterior segment of the eye is reported to be involved in the disease pathogenesis.


Subject(s)
Albinism , Geographic Atrophy , Animals , Albinism/metabolism , Complement Factor D/metabolism , Geographic Atrophy/metabolism , Melanins/metabolism , Retina , Rats
3.
Article in English | MEDLINE | ID: mdl-31041195

ABSTRACT

The two coagulases, von Willebrand factor binding protein (vWbp) and Coagulase (Coa), are critical virulence factors in several animal models of invasive Staphylococcus aureus (S. aureus) infections. These proteins are part of an intricate system of proteins that S. aureus uses to assemble a fibrinogen (Fg)/fibrin protective shield surrounding itself. This shield allows the microorganism to evade clearance by the host phagocytic cells. The coagulases can non-proteolytically activate the zymogen prothrombin to convert Fg to fibrin and promote the Fg/fibrin shield formation. The coagulases also bind directly to Fg and the interaction between Coa and Fg has been previously characterized in some detail. However, the mechanism(s) by which vWbp interacts with Fg remains unclear. Here, we show that vWbp and Coa have distinct interactions with Fg, despite being structurally similar. Coa binds with a significantly higher affinity to soluble Fg than to Fg coated on a plastic surface, whereas vWbp demonstrates no preference between the two forms of Fg. The two coagulases appear to target different sites on Fg, as they do not compete with each other in binding to Fg. Similar to Coa, both the N- and C-terminal halves of vWbp (vWbp-N, vWbp-C, respectively) harbor Fg-binding activities. The higher affinity Fg-binding activity resides in vWbp-N; whereas, the C-terminal region of Coa encompasses the major Fg-binding activity. Peptides constituting the previously identified Coa/Efb1 Fg-binding motif fail to inhibit vWbp-C from binding to Fg, indicating that vWbp-C lacks a functional homolog to this motif. Interestingly, the N-terminal prothrombin-binding domains of both coagulases recognize the Fg ß-chain, but they appear to interact with different sequence motifs in the host protein. Collectively, our data provide insight into the complex interactions between Fg and the S. aureus coagulases.


Subject(s)
Carrier Proteins/metabolism , Coagulase/metabolism , Fibrinogen/metabolism , Staphylococcus aureus/enzymology , Virulence Factors/metabolism , Binding Sites , Humans , Protein Binding , Protein Interaction Mapping
4.
mSphere ; 3(2)2018.
Article in English | MEDLINE | ID: mdl-29564394

ABSTRACT

The fibronectin-binding protein A (FnBPA) is a cell surface-associated protein of Staphylococcus aureus which mediates adherence to the host extracellular matrix and is important for bacterial virulence. Previously, substantial sequence diversity was found among strains in the fibrinogen-binding A domain of this protein, and 7 different isotypes were described. The effect of this sequence diversity on the human antibody response, in terms of both antibody production and antibody function, remains unclear. In this study, we identify five different FnBPA A domain isotypes based on the sequence results of 22 clinical S. aureus isolates, obtained from the same number of patients suffering from bacteremia. Using a bead-based Luminex technique, we measure the patients' total immunoglobulin G (IgG) against the 7 FnBPA isotypes at the onset and during the time course of bacteremia (median of 10 serum samples per patient over a median of 35 days). A significant increase in IgG against the FnBPA A domain, including the isotype carried by the infecting strain, is observed in only three out of 22 patients (14%) after the onset of bacteremia. Using a Luminex-based FnBPA-fibrinogen-binding assay, we find that preincubation of recombinant FnBPA isotypes with IgG from diverse patients does not interfere with binding to fibrinogen. This observation is confirmed using an alternative Luminex-based assay and enzyme-linked immunosorbent assay (ELISA). IMPORTANCE Despite the many in vitro and murine in vivo studies involving FnBPA, the actual presence of this virulence factor during human infection is less well established. Furthermore, it is currently unknown to what extent sequence variation in such a virulence factor affects the human antibody response and the ability of antibodies to interfere with FnBPA function. This study sheds new light on these issues. First, the uniform presence of a patient's IgG against FnBPA indicates the presence and importance of this virulence factor during S. aureus pathogenesis. Second, the absence of an increase in antibody production in most patients following bacteremia indicates the complexity of S. aureus-host interactions, possibly involving immune evasion or lack of expression of FnBPA during invasive infection. Finally, we provide new insights into the inability of human antibodies to interfere with FnBPA-fibrinogen binding. These observations should be taken into account during the development of novel vaccination approaches.

5.
Semin Thromb Hemost ; 42(4): 408-21, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27056151

ABSTRACT

Fibrinogen not only plays a pivotal role in hemostasis but also serves key roles in antimicrobial host defense. As a rapidly assembled provisional matrix protein, fibrin(ogen) can function as an early line of host protection by limiting bacterial growth, suppressing dissemination of microbes to distant sites, and mediating host bacterial killing. Fibrinogen-mediated host antimicrobial activity occurs predominantly through two general mechanisms, namely, fibrin matrices functioning as a protective barrier and fibrin(ogen) directly or indirectly driving host protective immune function. The potential of fibrin to limit bacterial infection and disease has been countered by numerous bacterial species evolving and maintaining virulence factors that engage hemostatic system components within vertebrate hosts. Bacterial factors have been isolated that simply bind fibrinogen or fibrin, promote fibrin polymer formation, or promote fibrin dissolution. Staphylococcus aureus is an opportunistic gram-positive bacterium, the causative agent of a wide range of human infectious diseases, and a prime example of a pathogen exquisitely sensitive to host fibrinogen. Indeed, current data suggest fibrinogen serves as a context-dependent determinant of host defense or pathogen virulence in Staphylococcus infection whose ultimate contribution is dictated by the expression of S. aureus virulence factors, the path of infection, and the tissue microenvironment.


Subject(s)
Blood Bactericidal Activity , Fibrin/metabolism , Fibrinogen/metabolism , Hemostasis , Staphylococcal Infections/blood , Staphylococcus aureus/pathogenicity , Animals , Humans , Staphylococcus aureus/metabolism
6.
Microbiology (Reading) ; 162(7): 1185-1194, 2016 07.
Article in English | MEDLINE | ID: mdl-27112346

ABSTRACT

Staphylococcus aureus has developed many mechanisms to escape from human immune responses. To resist phagocytic clearance, S. aureus expresses a polysaccharide capsule, which effectively masks the bacterial surface and surface-associated proteins, such as opsonins, from recognition by phagocytic cells. Additionally, secretion of the extracellular fibrinogen binding protein (Efb) potently blocks phagocytic uptake of the pathogen. Efb creates a fibrinogen shield surrounding the bacteria by simultaneously binding complement C3b and fibrinogen at the bacterial surface. By means of neutrophil phagocytosis assays with fluorescently labelled encapsulated serotype 5 (CP5) and serotype 8 (CP8) strains we compare the immune-modulating function of these shielding mechanisms. The data indicate that, in highly encapsulated S. aureus strains, the polysaccharide capsule is able to prevent phagocytic uptake at plasma concentrations <10 %, but loses its protective ability at higher concentrations of plasma. Interestingly, Efb shows a strong inhibitory effect on both capsule-negative and encapsulated strains at all tested plasma concentrations. Furthermore, the results suggest that both shielding mechanisms can exist simultaneously and collaborate to provide optimal protection against phagocytosis at a broad range of plasma concentrations. As opsonizing antibodies will be shielded from recognition by either mechanism, incorporating both capsular polysaccharides and Efb in future vaccines could be of great importance.


Subject(s)
Bacterial Capsules/metabolism , Bacterial Proteins/metabolism , Fibrinogen/metabolism , Neutrophils/immunology , Phagocytosis/immunology , Polysaccharides, Bacterial/metabolism , Staphylococcus aureus/immunology , Antibodies, Bacterial/immunology , Complement C3b/metabolism , Fluorescent Dyes , Humans , Immunomodulation/immunology , Microscopy, Confocal , Opsonin Proteins/metabolism , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/metabolism
7.
mBio ; 7(1): e01885-15, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26733070

ABSTRACT

UNLABELLED: Coagulase (Coa) and Efb, secreted Staphylococcus aureus proteins, are important virulence factors in staphylococcal infections. Coa interacts with fibrinogen (Fg) and induces the formation of fibrin(ogen) clots through activation of prothrombin. Efb attracts Fg to the bacterial surface and forms a shield to protect the bacteria from phagocytic clearance. This communication describes the use of an array of synthetic peptides to identify variants of a linear Fg binding motif present in Coa and Efb which are responsible for the Fg binding activities of these proteins. This motif represents the first Fg binding motif identified for any microbial protein. We initially located the Fg binding sites to Coa's C-terminal disordered segment containing tandem repeats by using recombinant fragments of Coa in enzyme-linked immunosorbent assay-type binding experiments. Sequence analyses revealed that this Coa region contained shorter segments with sequences similar to the Fg binding segments in Efb. An alanine scanning approach allowed us to identify the residues in Coa and Efb that are critical for Fg binding and to define the Fg binding motifs in the two proteins. In these motifs, the residues required for Fg binding are largely conserved, and they therefore constitute variants of a common Fg binding motif which binds to Fg with high affinity. Defining a specific motif also allowed us to identify a functional Fg binding register for the Coa repeats that is different from the repeat unit previously proposed. IMPORTANCE: Staphylococcus aureus infections are a major health problem that affects an estimated 50 million people globally and causes the death of about 20,000 Americans each year. A number of experimental vaccines have been developed during the past years. However, these vaccines have all failed in clinical trials. The ability of S. aureus to form an Fg shield surrounding and protecting bacterial cells from clearance may explain why the vaccines are failing. Furthermore, S. aureus coagulase can induce the formation of a fibrin(ogen) shield in experimental abscess models which surrounds and protects bacteria in the microcolony from clearance. In this study, we identified for the first time a microbial Fg binding motif. Variants of this motif are present in coagulase and Efb. Our results provide a molecular basis for the rational design of inhibitors that could potentially prevent the formation of the obstructing Fg shield.


Subject(s)
Amino Acid Motifs , Bacterial Proteins/metabolism , Coagulase/metabolism , Fibrinogen/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , Coagulase/chemistry , Coagulase/genetics , Conserved Sequence , DNA Mutational Analysis , Humans , Protein Binding
8.
Blood ; 126(17): 2047-58, 2015 Oct 22.
Article in English | MEDLINE | ID: mdl-26228483

ABSTRACT

Fibrin(ogen) is central to hemostasis and thrombosis and also contributes to multiple physiologic and pathologic processes beyond coagulation. However, the precise contribution of soluble fibrinogen vs insoluble fibrin matrices to vascular integrity, tissue repair, inflammation, and disease has been undefined and unapproachable. To establish the means to distinguish fibrinogen- and fibrin-dependent processes in vivo, Fib(AEK) mice were generated that carry normal levels of circulating fibrinogen but lack the capacity for fibrin polymer formation due to a germ-line mutation in the Aα chain thrombin cleavage site. Homozygous Fib(AEK) mice developed to term and exhibited postnatal survival superior to that of fibrinogen-deficient mice. Unlike fibrinogen-deficient mice, platelet-rich plasma from Fib(AEK) mice supported normal platelet aggregation in vitro, highlighting that fibrinogen(AEK) retains the functional capacity to support interactions with platelets. Thrombin failed to release fibrinopeptide-A from fibrinogen(AEK) and failed to induce polymer formation with Fib(AEK) plasma or purified fibrinogen(AEK) in 37°C mixtures regardless of incubation time. Fib(AEK) mice displayed both an absence of fibrin polymer formation following liver injury, as assessed by electron microscopy, and a failure to generate stable occlusive thrombi following FeCl3 injury of carotid arteries. Fib(AEK) mice exhibited a profound impediment in Staphylococcus aureus clearance following intraperitoneal infection similar to fibrinogen-deficient mice, yet Fib(AEK) mice displayed a significant infection dose-dependent survival advantage over fibrinogen-deficient mice following peritonitis challenge. Collectively, these findings establish for the first time that fibrin polymer is the molecular form critical for antimicrobial mechanisms while simultaneously highlighting biologically meaningful contributions and functions of the soluble molecule.


Subject(s)
Fibrin/metabolism , Fibrinogen/physiology , Host-Pathogen Interactions , Mutation/genetics , Peritonitis/etiology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Animals , Blood Coagulation Tests , Cells, Cultured , Flow Cytometry , Gene Expression Profiling , Hemostatics , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Peritonitis/pathology , Platelet Aggregation , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology
9.
J Biol Chem ; 289(3): 1505-18, 2014 Jan 17.
Article in English | MEDLINE | ID: mdl-24293366

ABSTRACT

Matrilin-1 is the prototypical member of the matrilin protein family and is highly expressed in cartilage. However, gene targeting of matrilin-1 in mouse did not lead to pronounced phenotypes. Here we used the zebrafish as an alternative model to study matrilin function in vivo. Matrilin-1 displays a multiphasic expression during zebrafish development. In an early phase, with peak expression at about 15 h post-fertilization, matrilin-1 is present throughout the zebrafish embryo with exception of the notochord. Later, when the skeleton develops, matrilin-1 is expressed mainly in cartilage. Morpholino knockdown of matrilin-1 results both in overall growth defects and in disturbances in the formation of the craniofacial cartilage, most prominently loss of collagen II deposition. In fish with mild phenotypes, certain cartilage extracellular matrix components were present, but the tissue did not show features characteristic for cartilage. The cells showed endoplasmic reticulum aberrations but no activation of XBP-1, a marker for endoplasmic reticulum stress. In severe phenotypes nearly all chondrocytes died. During the early expression phase the matrilin-1 knockdown had no effects on cell morphology, but increased cell death was observed. In addition, the broad deposition of collagen II was largely abolished. Interestingly, the early phenotype could be rescued by the co-injection of mRNA coding for the von Willebrand factor C domain of collagen IIα1a, indicating that the functional loss of this domain occurs as a consequence of matrilin-1 deficiency. The results show that matrilin-1 is indispensible for zebrafish cartilage formation and plays a role in the early collagen II-dependent developmental events.


Subject(s)
Cartilage/embryology , Collagen Type II/metabolism , Embryo, Nonmammalian/embryology , Embryonic Development/physiology , Gene Expression Regulation, Developmental/physiology , Matrilin Proteins/metabolism , Zebrafish/embryology , Animals , Animals, Genetically Modified , Cartilage/cytology , Collagen Type II/genetics , Embryo, Nonmammalian/cytology , Embryonic Development/drug effects , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Gene Expression Regulation, Developmental/drug effects , Gene Knockdown Techniques , Matrilin Proteins/genetics , Mice , Morpholinos/pharmacology , Zebrafish/genetics , Zebrafish Proteins
10.
PLoS Pathog ; 9(12): e1003816, 2013.
Article in English | MEDLINE | ID: mdl-24348255

ABSTRACT

Upon contact with human plasma, bacteria are rapidly recognized by the complement system that labels their surface for uptake and clearance by phagocytic cells. Staphylococcus aureus secretes the 16 kD Extracellular fibrinogen binding protein (Efb) that binds two different plasma proteins using separate domains: the Efb N-terminus binds to fibrinogen, while the C-terminus binds complement C3. In this study, we show that Efb blocks phagocytosis of S. aureus by human neutrophils. In vitro, we demonstrate that Efb blocks phagocytosis in plasma and in human whole blood. Using a mouse peritonitis model we show that Efb effectively blocks phagocytosis in vivo, either as a purified protein or when produced endogenously by S. aureus. Mutational analysis revealed that Efb requires both its fibrinogen and complement binding residues for phagocytic escape. Using confocal and transmission electron microscopy we show that Efb attracts fibrinogen to the surface of complement-labeled S. aureus generating a 'capsule'-like shield. This thick layer of fibrinogen shields both surface-bound C3b and antibodies from recognition by phagocytic receptors. This information is critical for future vaccination attempts, since opsonizing antibodies may not function in the presence of Efb. Altogether we discover that Efb from S. aureus uniquely escapes phagocytosis by forming a bridge between a complement and coagulation protein.


Subject(s)
Bacterial Proteins/metabolism , Complement C3b/metabolism , Fibrinogen/metabolism , Immune Evasion , Phagocytosis/immunology , Staphylococcus aureus/immunology , Staphylococcus aureus/metabolism , Animals , Blood Coagulation Factors/metabolism , Cells, Cultured , Female , Humans , Mice , Mice, Inbred C57BL , Protein Binding , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism
11.
J Biol Chem ; 288(28): 20520-31, 2013 Jul 12.
Article in English | MEDLINE | ID: mdl-23720782

ABSTRACT

Members of a family of collagen-binding microbial surface components recognizing adhesive matrix molecules (MSCRAMMs) from Gram-positive bacteria are established virulence factors in several infectious diseases models. Here, we report that these adhesins also can bind C1q and act as inhibitors of the classical complement pathway. Molecular analyses of Cna from Staphylococcus aureus suggested that this prototype MSCRAMM bound to the collagenous domain of C1q and interfered with the interactions of C1r with C1q. As a result, C1r2C1s2 was displaced from C1q, and the C1 complex was deactivated. This novel function of the Cna-like MSCRAMMs represents a potential immune evasion strategy that could be used by numerous Gram-positive pathogens.


Subject(s)
Adhesins, Bacterial/immunology , Complement Activation/immunology , Complement Pathway, Classical/immunology , Gram-Positive Bacteria/immunology , Adhesins, Bacterial/chemistry , Adhesins, Bacterial/metabolism , Binding, Competitive/immunology , Collagen/immunology , Collagen/metabolism , Complement C1q/immunology , Complement C1q/metabolism , Complement C1r/immunology , Complement C1r/metabolism , Enzyme-Linked Immunosorbent Assay , Gram-Positive Bacteria/metabolism , History, 18th Century , Humans , Immunoblotting , Kinetics , Models, Molecular , Protein Binding/immunology , Protein Structure, Tertiary , Surface Plasmon Resonance
12.
J Biol Chem ; 286(11): 9865-74, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21247890

ABSTRACT

In addition to its pivotal role in hemostasis, fibrinogen (Fg) and provisional fibrin matrices play important roles in inflammation and regulate innate immune responses by interacting with leukocytes. Efb (the extracellular fibrinogen-binding protein) is a secreted Staphylococcus aureus protein that engages host Fg and complement C3. However, the molecular details underlying the Efb-Fg interaction and the biological relevance of this interaction have not been determined. In the present study, we characterize the interaction of Efb with Fg. We demonstrate that the Fg binding activity is located within the intrinsically disordered N-terminal half of Efb (Efb-N) and that the D fragment of Fg is the region that mediates Efb-N binding. More detailed studies of the Efb-N-Fg interactions using ELISA and surface plasmon resonance analyses revealed that Efb-N exhibits a much higher affinity for Fg than typically observed with Fg-binding MSCRAMMs (microbial surface components recognizing adhesive matrix molecules), and data obtained from ELISA analyses using truncated Efb-N constructs demonstrate that Efb-N contains two binding sites located within residues 30-67 and 68-98, respectively. Efb-N inhibits neutrophil adhesion to immobilized Fg by binding to Fg and blocking the interaction of the protein with the leukocyte integrin receptor, α(M)ß(2). A motif in the Fg γ chain previously shown to be central to the α(M)ß(2) interaction was shown to be functionally distinguishable from the Efb-N binding site, suggesting that the Fg-Efb interaction indirectly impedes Fg engagement by α(M)ß(2). Taken together, these studies provide insights into how Efb interacts with Fg and suggest that Efb may support bacterial virulence at least in part by impeding Fg-driven leukocyte adhesion events.


Subject(s)
Bacterial Proteins/metabolism , Fibrinogen/metabolism , Neutrophils/metabolism , Staphylococcus aureus/metabolism , Adhesins, Bacterial/chemistry , Adhesins, Bacterial/metabolism , Amino Acid Motifs , Bacterial Proteins/chemistry , Binding Sites , Cell Adhesion , Fibrinogen/chemistry , HEK293 Cells , Humans , Neutrophils/chemistry , Protein Binding , Staphylococcus aureus/chemistry , Staphylococcus aureus/pathogenicity
13.
PLoS Pathog ; 4(11): e1000226, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19043557

ABSTRACT

The fibrinogen (Fg) binding MSCRAMM Clumping factor A (ClfA) from Staphylococcus aureus interacts with the C-terminal region of the fibrinogen (Fg) gamma-chain. ClfA is the major virulence factor responsible for the observed clumping of S. aureus in blood plasma and has been implicated as a virulence factor in a mouse model of septic arthritis and in rabbit and rat models of infective endocarditis. We report here a high-resolution crystal structure of the ClfA ligand binding segment in complex with a synthetic peptide mimicking the binding site in Fg. The residues in Fg required for binding to ClfA are identified from this structure and from complementing biochemical studies. Furthermore, the platelet integrin alpha(IIb)beta(3) and ClfA bind to the same segment in the Fg gamma-chain but the two cellular binding proteins recognize different residues in the common targeted Fg segment. Based on these differences, we have identified peptides that selectively antagonize the ClfA-Fg interaction. The ClfA-Fg binding mechanism is a variant of the "Dock, Lock and Latch" mechanism previously described for the Staphylococcus epidermidis SdrG-Fg interaction. The structural insights gained from analyzing the ClfANFg peptide complex and identifications of peptides that selectively recognize ClfA but not alpha(IIb)beta(3) may allow the design of novel anti-staphylococcal agents. Our results also suggest that different MSCRAMMs with similar structural organization may have originated from a common ancestor but have evolved to accommodate specific ligand structures.


Subject(s)
Coagulase/metabolism , Fibrinogen/metabolism , Models, Molecular , Staphylococcal Infections/drug therapy , Binding Sites , Crystallography, X-Ray , Drug Design , Peptides/chemical synthesis , Peptides/pharmacology , Protein Binding/drug effects , Staphylococcus aureus
14.
J Biol Chem ; 282(30): 22163-75, 2007 Jul 27.
Article in English | MEDLINE | ID: mdl-17502381

ABSTRACT

Matrilins are oligomeric extracellular matrix adaptor proteins mediating interactions between collagen fibrils and other matrix constituents. All four matrilins are expressed in cartilage and mutations in the human gene encoding matrilin-3 (MATN3) are associated with different forms of chondrodysplasia. Surprisingly, however, Matn3-null as well as Matn1- and Matn2-null mice do not show an overt skeletal phenotype, suggesting a dominant negative pathomechanism for the human disorders and redundancy/compensation among the family members in the knock-out situation. Here, we show that mice lacking both matrilin-1 and matrilin-3 develop an apparently normal skeleton, but exhibit biochemical and ultrastructural abnormalities of the knee joint cartilage. At the protein level, an altered SDS-PAGE band pattern and a clear up-regulation of the homotrimeric form of matrilin-4 were evident in newborn Matn1/Matn3 and Matn1 knock-out mice, but not in Matn3-null mice. The ultrastructure of the cartilage matrix after conventional chemical fixation was grossly normal; however, electron microscopy of high pressure frozen and freeze-substituted samples, revealed two consistent observations: 1) moderately increased collagen fibril diameters throughout the epiphysis and the growth plate in both single and double mutants; and 2) increased collagen volume density in Matn1(-/-)/Matn3(-/-) and Matn3(-/-) mice. Taken together, our results demonstrate that matrilin-1 and matrilin-3 modulate collagen fibrillogenesis in cartilage and provide evidence that biochemical compensation might exist between matrilins.


Subject(s)
Bone and Bones/diagnostic imaging , Cartilage, Articular/pathology , Collagen/metabolism , Extracellular Matrix Proteins/deficiency , Glycoproteins/deficiency , Animals , Animals, Newborn , Bone Development , Cartilage Oligomeric Matrix Protein , Crosses, Genetic , Homozygote , Matrilin Proteins , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Tomography, X-Ray Computed
15.
FEBS Lett ; 579(15): 3323-9, 2005 Jun 13.
Article in English | MEDLINE | ID: mdl-15943978

ABSTRACT

The matrilins form a four-member family of modular, multisubunit matrix proteins, which are expressed in cartilage but also in many other forms of extracellular matrix. They participate in the formation of fibrillar or filamentous structures and are often associated with collagens. It appears that they mediate interactions between collagen-containing fibrils and other matrix constituents, such as aggrecan. This adaptor function may be modulated by physiological proteolysis that causes the loss of single subunits and thereby a decrease in binding avidity. Attempts to study matrilin function by gene inactivation in mouse have been frustrating and so far not yielded pronounced phenotypes, presumably because of the extensive redundancy within the family allowing compensation by one family member for another. However, mutations in matrilin-3 in humans cause different forms of chondrodysplasias and perhaps also hand osteoarthritis. As loss of matrilin-3 is not critical in mouse, these phenotypes are likely to be caused by dominant negative effects.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Extracellular Matrix/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/classification , Adaptor Proteins, Signal Transducing/genetics , Animals , Disease Models, Animal , Humans , Protein Binding , Protein Processing, Post-Translational
16.
Int J Cancer ; 116(2): 243-52, 2005 Aug 20.
Article in English | MEDLINE | ID: mdl-15812828

ABSTRACT

Cancer metastasis is a multiple-step process that involves the regulated interaction of diverse cellular proteins. We recently reported that the expression of tumor-associated antigen L6 (TAL6) promoted the invasiveness of lung cancer cells and was inversely correlated with disease-free survival of squamous lung carcinoma patients. We now report that CD13 (aminopeptidase N) can associate with TAL6 and can enhance cancer cell migration. CD13 was shown by coimmunoprecipitation to associate in vitro with TAL6 on several cancer cell lines and to associate in vivo by antibody-mediated copatching immunofluorescence. CD13 was selectively expressed on highly invasive CL1-5 lung cancer cells as compared to poorly invasive CL1-0 lung cancer cells. The role of CD13 aminopeptidase activity in regulating cell motility was investigated with chemical inhibitors, specific antibodies and a catalytically inactive CD13 protein. Inhibition of CD13 aminopeptidase activity by nontoxic concentrations of leuhistin modestly decreased the migration of CL1-5 cells. In contrast, binding of CD13 by specific antibodies significantly reduced both the migration and the invasion of CL1-5 cells. Poorly invasive CL1-0 cells that stably expressed CD13 displayed significantly (p < or = 0.0005) enhanced cell migration (300% of control). Expression of an enzymatically inactive CD13 mutant on CL1-0 cells also significantly (p < or = 0.0005) enhanced cell migration (200% of control). Our results show that TAL6 and CD13 can form a complex on lung cancer cells, that these molecules can modulate cell migration and invasion and that the influence of CD13 on cell motility did not strictly depend on its aminopeptidase activity.


Subject(s)
Adenocarcinoma/immunology , Adenocarcinoma/physiopathology , Antigens, Surface/biosynthesis , Antigens, Surface/physiology , CD13 Antigens/biosynthesis , CD13 Antigens/physiology , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/physiopathology , Cell Movement , Lung Neoplasms/immunology , Lung Neoplasms/physiopathology , Neoplasm Metastasis/physiopathology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/physiology , Antibodies , Gene Expression Regulation, Neoplastic , Humans , Immunoprecipitation , Neoplasm Invasiveness , Neoplasm Metastasis/immunology
17.
Biochem J ; 386(Pt 2): 367-79, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15588228

ABSTRACT

We have cloned the cDNAs of the zebrafish (Danio rerio) members of the matrilin family of extracellular adaptor proteins. In contrast to mammals, no orthologue of matrilin-2 was found in zebrafish, either by RT (reverse-transcriptase) PCR using degenerated primers or by screening the databases (Ensembl and NCBI); however, two forms of matrilin-3, matrilin-3a and -3b, were present. The identity with the mammalian matrilins is from more than 70% for the VWA (von Willebrand factor A)-like domains to only 28% for the coiled-coil domains of matrilin-3a and -3b. In all zebrafish matrilins we found a greater variety of splice variants than in mammals, with splicing mainly affecting the number of EGF (epidermal growth factor)-like repeats. The exon-intron organization is nearly identical with that of mammals, and also the characteristic AT-AC intron interrupting the exons coding for the coiled-coil domain is conserved. In the matrilin-3b gene a unique exon codes for a proline- and serine/threonine-rich domain, possibly having mucin-like properties. The matrilin-1 and -3a genes were mapped to chromosome 19 and 20 respectively by the radiation hybrid method. The temporal and spatial expression of zebrafish matrilins is similar to that seen in the mouse. Zebrafish matrilin-4 is highly expressed as early as 24 hpf (h post fertilization), whereas the other matrilins show peak expression at 72 hpf. By immunostaining of whole mounts and sections, we found that matrilin-1 and -3a show predominantly skeletal staining, whereas matrilin-4 is more widespread, with the protein also being present in loose connective tissues and epithelia.


Subject(s)
Extracellular Matrix Proteins/genetics , Genetic Variation/genetics , Glycoproteins/genetics , Zebrafish/genetics , Alternative Splicing/genetics , Animals , Cartilage Oligomeric Matrix Protein , Cell Line , Cloning, Molecular/methods , Conserved Sequence/genetics , Exons/genetics , Extracellular Matrix Proteins/immunology , Gene Expression Regulation/genetics , Gene Expression Regulation, Developmental/genetics , Glycoproteins/immunology , Humans , Immune Sera/metabolism , Introns/genetics , Kidney/chemistry , Kidney/cytology , Kidney/embryology , Kidney/metabolism , Matrilin Proteins , Molecular Sequence Data , Peptides/genetics , Peptides/immunology , Radiation Hybrid Mapping/methods , Recombinant Proteins/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/immunology
18.
Biotechnol Appl Biochem ; 40(Pt 2): 167-72, 2004 Oct.
Article in English | MEDLINE | ID: mdl-14725509

ABSTRACT

Expression of recombinant proteins in mammalian cells is useful for obtaining products with normal post-translational modifications. We describe a simple and economical method for the production of milligram levels of proteins in murine fibroblasts. Retroviral or LIPOFECTAMINE (Gibco Laboratories) transduction was employed to generate stable murine-fibroblast producer cells. Confluent cultures of stable fibroblast clones were maintained for up to 1 month in 0.5% serum. Culture medium was collected every 2-3 days and polyhistidine-tagged proteins were purified by ammonium sulphate precipitation and Ni(2+)-nitrilotriacetic acid affinity chromatography. Highly pure, active, glycosylated recombinant proteins, including human beta-glucuronidase, mouse beta-glucuronidase, aminopeptidase N (CD13) and a single-chain antibody-enzyme fusion protein, were obtained with yields of 3-6 mg/l of culture medium. Fc-tagged proteins were also produced and purified in a single step by Protein A affinity chromatography with yields of 6-12 mg/l. The techniques described here allow simple and economical production of recombinant mammalian proteins with post-translational modifications.


Subject(s)
Cell Culture Techniques/methods , Cloning, Molecular/methods , Protein Engineering/methods , Recombinant Proteins/biosynthesis , Animals , BALB 3T3 Cells , Cell Line , Humans , Mice
19.
Clin Cancer Res ; 9(7): 2807-16, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12855661

ABSTRACT

Metastasis is a coordinated process that depends on the interaction of cancer cells with the tumor microenvironment. Members of the transmembrane-4 superfamily (TM4SF) of surface proteins have been implicated in the regulation of cancer cell metastasis, and the expression of several TM4SF members on tumor cells is inversely correlated with patient prognosis. The tumor-associated antigen L6 (TAL6), a distant member of the TM4SF, is expressed on most epithelial cell carcinomas and is a target for antibody-mediated therapy. We examined whether TAL6 may play a role in cancer metastasis by using an established series of human lung carcinoma cell lines (CL1-0 to CL1-5) that exhibit increasing invasiveness in vitro and in vivo. We found that TAL6 expression correlated with the in vitro invasiveness of CL lung carcinoma cells (r(2) = 0.98) and human carcinoma cells (r(2) = 0.69). Forced expression of TAL6 on CL1-0 lung carcinoma cells significantly increased their in vitro invasiveness and decreased the survival of SCID mice in an experimental metastasis model. Specific antibody against TAL6 (monoclonal antibody L6) significantly reduced the migration and invasiveness of CL1-5 lung carcinoma cells. The effects of monoclonal antibody L6 on CL1-5 invasion required clustering of TAL6 on the cell surface. Real-time reverse transcription-PCR of lung cancer specimens showed that increased expression of TAL6 was significantly associated with early postoperative relapse (P = 0.034) and shorter survival (P = 0.025) in squamous cell lung cancer patients. Thus, TAL6 appears to be involved in cancer invasion and metastasis.


Subject(s)
Antigens, Neoplasm/biosynthesis , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Aged , Animals , Antibodies, Monoclonal/chemistry , Antigens, Surface/biosynthesis , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/mortality , Cell Adhesion , Cell Line, Tumor , Cell Movement , Collagen/metabolism , Disease-Free Survival , Dose-Response Relationship, Drug , Female , Humans , Lung Neoplasms/mortality , Male , Mice , Mice, SCID , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/biosynthesis , RNA, Messenger/metabolism , Recurrence , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...