Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
ACS Infect Dis ; 3(7): 479-491, 2017 07 14.
Article in English | MEDLINE | ID: mdl-28591513

ABSTRACT

Preventing the spread of infectious diseases remains an urgent priority worldwide, and this is driving the development of advanced nanotechnology to diagnose infections at the point of care. Herein, we report the creation of a library of novel nanobody capture ligands to detect p24, one of the earliest markers of HIV infection. We demonstrate that these nanobodies, one tenth the size of conventional antibodies, exhibit high sensitivity and broad specificity to global HIV-1 subtypes. Biophysical characterization indicates strong 690 pM binding constants and fast kinetic on-rates, 1 to 2 orders of magnitude better than monoclonal antibody comparators. A crystal structure of the lead nanobody and p24 was obtained and used alongside molecular dynamics simulations to elucidate the molecular basis of these enhanced performance characteristics. They indicate that binding occurs at C-terminal helices 10 and 11 of p24, a negatively charged region of p24 complemented by the positive surface of the nanobody binding interface involving CDR1, CDR2, and CDR3 loops. Our findings have broad implications on the design of novel antibodies and a wide range of advanced biomedical applications.


Subject(s)
Antibodies, Monoclonal/chemistry , HIV Antibodies/chemistry , HIV Core Protein p24/chemistry , HIV-1/chemistry , Single-Domain Antibodies/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibody Specificity , Binding Sites , Camelids, New World , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , HIV Antibodies/biosynthesis , HIV Antibodies/immunology , HIV Antibodies/isolation & purification , HIV Core Protein p24/genetics , HIV Core Protein p24/immunology , Humans , Kinetics , Molecular Dynamics Simulation , Peptide Library , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Single-Domain Antibodies/biosynthesis , Single-Domain Antibodies/immunology , Single-Domain Antibodies/isolation & purification , Static Electricity
2.
Stem Cells Dev ; 25(23): 1808-1817, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27733096

ABSTRACT

Hypertrophic differentiation occurs during in vitro chondrogenesis of mesenchymal stem cells (MSCs), decreasing the quality of the cartilage construct. Previously we identified WNT pathway antagonists Dickkopf 1 homolog (DKK1) and frizzled-related protein (FRZB) as key factors in blocking hypertrophic differentiation of human MSCs (hMSCs). In this study, we investigated the role of endogenously expressed DKK1 and FRZB in chondrogenesis of hMSC and chondrocyte redifferentiation and in preventing cell hypertrophy using three relevant human cell based systems, isolated hMSCs, isolated primary human chondrocytes (hChs), and cocultures of hMSCs with hChs for which we specifically designed neutralizing nano-antibodies. We selected and tested variable domain of single chain heavy chain only antibodies (VHH) for their ability to neutralize the function of DKK1 or FRZB. In the presence of DKK1 and FRZB neutralizing VHH, glycosaminoglycan and collagen type II staining were significantly reduced in monocultured hMSCs and monocultured chondrocytes. Furthermore, in cocultures, cells in pellets showed hypertrophic differentiation. In conclusion, endogenous expression of the WNT antagonists DKK1 and FRZB is necessary for multiple steps during chondrogenesis: first DKK1 and FRZB are indispensable for the initial steps of chondrogenic differentiation of hMSCs, second they are necessary for chondrocyte redifferentiation, and finally in preventing hypertrophic differentiation of articular chondrocytes.


Subject(s)
Cell Culture Techniques/methods , Chondrocytes/metabolism , Chondrogenesis , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Antibodies, Neutralizing/pharmacology , Biomarkers/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrogenesis/drug effects , Coculture Techniques , Glycoproteins/immunology , Humans , Hypertrophy , Intercellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins , Mesenchymal Stem Cells/drug effects , Middle Aged , Single-Domain Antibodies/immunology , Wnt Signaling Pathway/drug effects
3.
Sci Rep ; 6: 31621, 2016 08 22.
Article in English | MEDLINE | ID: mdl-27546726

ABSTRACT

Bispecific antibodies are of great interest due to their ability to simultaneously bind and engage different antigens or epitopes. Nevertheless, it remains a challenge to assemble, produce and/or purify them. Here we present an innovative dual anti-idiotypic purification process, which provides pure bispecific antibodies with native immunoglobulin format. Using this approach, a biparatopic IgG1 antibody targeting two distinct, HGF-competing, non-overlapping epitopes on the extracellular region of the MET receptor, was purified with camelid single-domain antibody fragments that bind specifically to the correct heavy chain/light chain pairings of each arm. The purity and functionality of the anti-MET biparatopic antibody was then confirmed by mass spectrometry and binding experiments, demonstrating its ability to simultaneously target the two epitopes recognized by the parental monoclonal antibodies. The improved MET-inhibitory activity of the biparatopic antibody compared to the parental monoclonal antibodies, was finally corroborated in cell-based assays and more importantly in a tumor xenograft mouse model. In conclusion, this approach is fast and specific, broadly applicable and results in the isolation of a pure, novel and native-format anti-MET biparatopic antibody that shows superior biological activity over the parental monospecific antibodies both in vitro and in vivo.


Subject(s)
Antibodies, Bispecific , Antineoplastic Agents, Immunological , Neoplasms, Experimental/drug therapy , Proto-Oncogene Proteins c-met/antagonists & inhibitors , A549 Cells , Animals , Antibodies, Bispecific/immunology , Antibodies, Bispecific/isolation & purification , Antibodies, Bispecific/pharmacology , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/isolation & purification , Antineoplastic Agents, Immunological/pharmacology , Humans , Immunoglobulin G/immunology , Immunoglobulin G/isolation & purification , Immunoglobulin G/pharmacology , Mice , Mice, Nude , Mice, SCID , Neoplasms, Experimental/immunology , Proto-Oncogene Proteins c-met/immunology , Xenograft Model Antitumor Assays
4.
Mol Imaging Biol ; 18(4): 535-44, 2016 08.
Article in English | MEDLINE | ID: mdl-26589824

ABSTRACT

PURPOSE: The aim of this work was to develop a CAIX-specific nanobody conjugated to IRDye800CW for molecular imaging of pre-invasive breast cancer. PROCEDURES: CAIX-specific nanobodies were selected using a modified phage display technology, conjugated site-specifically to IRDye800CW and evaluated in a xenograft breast cancer mouse model using ductal carcinoma in situ cells (DCIS). RESULTS: Specific anti-CAIX nanobodies were obtained. Administration of a CAIX-specific nanobody into mice with DCIS xenografts overexpressing CAIX showed after 2 h a mean tumor-to-normal tissue ratio (TNR) of 4.3 ± 0.6, compared to a TNR of 1.4 ± 0.2 in mice injected with the negative control nanobody R2-IR. In DCIS mice, a TNR of 1.8 ± 0.1 was obtained. Biodistribution studies demonstrated an uptake of 14.0 ± 1.1 %I.D./g in DCIS + CAIX tumors, 4.6 ± 0.8 %I.D./g in DCIS tumors, while 2.0 ± 0.2 %I.D./g was obtained with R2-IR. CONCLUSIONS: These results demonstrate the successful generation of a CAIX-specific nanobody-IRDye800CW conjugate that can be used for rapid imaging of (pre-)invasive breast cancer.


Subject(s)
Hypoxia/pathology , Mammary Neoplasms, Animal/pathology , Molecular Imaging/methods , Optical Imaging/methods , Single-Domain Antibodies/metabolism , Animals , Carbonic Anhydrase IX/metabolism , Carcinoma, Intraductal, Noninfiltrating/pathology , Cell Hypoxia , Cell Surface Display Techniques , DNA/metabolism , Female , HeLa Cells , Humans , Immunization , Immunohistochemistry , Mice , Neoplasm Invasiveness , Tissue Distribution , Xenograft Model Antitumor Assays
5.
Mol Cancer Ther ; 14(11): 2527-40, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26351325

ABSTRACT

Bone morphogenetic proteins (BMP) have important but distinct roles in tissue homeostasis and disease, including carcinogenesis and tumor progression. A large number of BMP inhibitors are available to study BMP function; however, as most of these antagonists are promiscuous, evaluating specific effects of individual BMPs is not feasible. Because the oncogenic role of the different BMPs varies for each neoplasm, highly selective BMP inhibitors are required. Here, we describe the generation of three types of llama-derived heavy chain variable domains (VHH) that selectively bind to either BMP4, to BMP2 and 4, or to BMP2, 4, 5, and 6. These generated VHHs have high affinity to their targets and are able to inhibit BMP signaling. Epitope binning and docking modeling have shed light into the basis for their BMP specificity. As opposed to the wide structural reach of natural inhibitors, these small molecules target the grooves and pockets of BMPs involved in receptor binding. In organoid experiments, specific inhibition of BMP4 does not affect the activation of normal stem cells. Furthermore, in vitro inhibition of cancer-derived BMP4 noncanonical signals results in an increase of chemosensitivity in a colorectal cancer cell line. Therefore, because of their high specificity and low off-target effects, these VHHs could represent a therapeutic alternative for BMP4(+) malignancies.


Subject(s)
Antibodies/pharmacology , Antibody Specificity/immunology , Bone Morphogenetic Proteins/antagonists & inhibitors , Camelids, New World/immunology , Neoplasms/drug therapy , Animals , Antibodies/immunology , Antibodies/metabolism , Antibody Affinity/immunology , Blotting, Western , Bone Morphogenetic Protein 2/chemistry , Bone Morphogenetic Protein 2/immunology , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 4/chemistry , Bone Morphogenetic Protein 4/immunology , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Proteins/immunology , Bone Morphogenetic Proteins/metabolism , Cell Line , HT29 Cells , Humans , Mice , Models, Molecular , Neoplasms/immunology , Neoplasms/metabolism , Protein Binding/immunology , Protein Structure, Tertiary
6.
FEBS J ; 282(18): 3618-31, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26147692

ABSTRACT

UNLABELLED: Accumulation and aggregation of the amyloid-ß (Aß) peptide is associated with Alzheimer's disease (AD). Aß is generated from the amyloid precursor protein by the successive action of two membrane-associated processing enzymes: ß-secretase or ß-site of amyloid precursor protein cleaving enzyme 1 (BACE1) and γ-secretase. Inhibition of one or both of these enzymes prevents Aß generation and the accompanying Aß accumulation. Antigen binding fragments from camelid heavy chain only antibodies (VHHs) were found to exert excellent enzyme inhibition activity. In the present study, we generated VHHs against BACE1 by active immunization of Lama glama with the recombinant BACE1 protein. Two classes of VHHs were selected from a VHH-phage display library by competitive elution with a peptide encoding the Swedish mutation variant of the BACE1 processing site. One VHH was found to inhibit the enzyme activity of BACE1 in vitro and in cell culture, whereas two other VHHs were found to stimulate BACE1 activity under the same conditions in vitro. Furthermore, an in vivo study with a transgenic AD mouse model, using intracisternal injection of the inhibitory VHH, led to acute reduction of the Aß load in the blood and brain. This inhibitory VHH may be considered as a candidate molecule for a therapy directed towards reduction of Aß load and prevention of AD progression. Both the inhibitory and stimulatory VHH may be useful for improving our understanding of the structure-function relationship of BACE1, as well as its role in AD progression. DATABASE: The GenBank sequence accession numbers are KR363186 for VHH B1a; KR363187 for VHH B3a; and KR363188 for VHH B5a.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/immunology , Amyloid beta-Protein Precursor/metabolism , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/immunology , Camelids, New World/immunology , Immunoglobulin Fragments/metabolism , Immunoglobulin Heavy Chains/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid Endopeptidases/genetics , Cell Line , Disease Models, Animal , Disease Progression , Female , Humans , Immunoglobulin Fragments/administration & dosage , Immunoglobulin Heavy Chains/administration & dosage , In Vitro Techniques , Mice , Mice, Transgenic , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Vaccination
7.
Eur J Nucl Med Mol Imaging ; 40(11): 1718-29, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23778558

ABSTRACT

PURPOSE: Molecular optical imaging using monoclonal antibodies is slow with low tumour to background ratio. We used anti-HER2 VHHs conjugated to IRDye 800CW to investigate their potential as probes for rapid optical molecular imaging of HER2-positive tumours by the determination of tumour accumulation and tumour to background levels. METHODS: Three anti-HER2 VHHs (11A4, 18C3, 22G12) were selected with phage display and produced in Escherichia coli. Binding affinities of these probes to SKBR3 cells were determined before and after site-specific conjugation to IRDye 800CW. To determine the potential of VHH-IR as imaging probes, serial optical imaging studies were carried out using human SKBR3 and human MDA-MB-231 xenograft breast cancer models. Performance of the anti-HER2 VHH-IR was compared to that of trastuzumab-IR and a non-HER2-specific VHH-IR. Image-guided surgery was performed during which SKBR3 tumour was removed under the guidance of the VHH-IR signal. RESULTS: Site-specific conjugation of IRDye 800CW to three anti-HER2 VHHs preserved high affinity binding with the following dissociation constants (KD): 11A4 1.9 ± 0.03, 18C3 14.3 ± 1.8 and 22G12 3.2 ± 0.5 nM. Based upon different criteria such as binding, production yield and tumour accumulation, 11A4 was selected for further studies. Comparison of 11A4-IR with trastuzumab-IR showed ∼20 times faster tumour accumulation of the anti-HER2 VHH, with a much higher contrast between tumour and background tissue (11A4-IR 2.5 ± 0.3, trastuzumab-IR 1.4 ± 0.4, 4 h post-injection). 11A4-IR was demonstrated to be a useful tool in image-guided surgery. CONCLUSION: VHH-IR led to a much faster tumour accumulation with high tumour to background ratios as compared to trastuzumab-IR allowing same-day imaging for clinical investigation as well as image-guided surgery.


Subject(s)
Benzenesulfonates/pharmacokinetics , Fluorescent Dyes/pharmacokinetics , Indoles/pharmacokinetics , Optical Imaging/methods , Receptor, ErbB-2/immunology , Single-Domain Antibodies/immunology , Surgery, Computer-Assisted/methods , Animals , Antibody Affinity , Benzenesulfonates/therapeutic use , Breast Neoplasms/pathology , Breast Neoplasms/surgery , Camelids, New World , Fluorescent Dyes/therapeutic use , Humans , Indoles/therapeutic use , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Single-Domain Antibodies/therapeutic use , Xenograft Model Antitumor Assays
8.
PLoS Pathog ; 9(3): e1003202, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23505368

ABSTRACT

The membrane proximal external region (MPER) of the HIV-1 glycoprotein gp41 is targeted by the broadly neutralizing antibodies 2F5 and 4E10. To date, no immunization regimen in animals or humans has produced HIV-1 neutralizing MPER-specific antibodies. We immunized llamas with gp41-MPER proteoliposomes and selected a MPER-specific single chain antibody (VHH), 2H10, whose epitope overlaps with that of mAb 2F5. Bi-2H10, a bivalent form of 2H10, which displayed an approximately 20-fold increased affinity compared to the monovalent 2H10, neutralized various sensitive and resistant HIV-1 strains, as well as SHIV strains in TZM-bl cells. X-ray and NMR analyses combined with mutagenesis and modeling revealed that 2H10 recognizes its gp41 epitope in a helical conformation. Notably, tryptophan 100 at the tip of the long CDR3 is not required for gp41 interaction but essential for neutralization. Thus bi-2H10 is an anti-MPER antibody generated by immunization that requires hydrophobic CDR3 determinants in addition to epitope recognition for neutralization similar to the mode of neutralization employed by mAbs 2F5 and 4E10.


Subject(s)
Antibodies, Neutralizing/immunology , Camelids, New World/immunology , Complementarity Determining Regions/immunology , HIV Antibodies/immunology , HIV Envelope Protein gp41/immunology , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Epitopes/immunology , Humans , Hydrophobic and Hydrophilic Interactions , Immunization , Molecular Sequence Data , Mutagenesis, Site-Directed , Neutralization Tests , Proteolipids/administration & dosage , Proteolipids/immunology , Single-Domain Antibodies , Surface Plasmon Resonance
9.
Eur J Pharm Sci ; 42(4): 332-9, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21220006

ABSTRACT

The 10 isozymes of the protein kinase C (PKC) family can have different roles on the same biological process, making isozyme specific analysis of function crucial. Currently, only few pharmacological compounds with moderate isozyme specific effects exist thus hampering research into individual PKC isozymes. The antigen binding regions of camelid single chain antibodies (VHHs) could provide a solution for obtaining PKC isozyme specific modulators. In the present study, we have successfully selected and characterized PKCɛ specific VHH antibodies from two immune VHH libraries using phage display. The VHHs were shown to exclusively bind to PKCɛ in ELISA and immunoprecipitation studies. Strikingly, five of the VHHs had an effect on PKCɛ kinase activity in vitro. VHHs A10, C1 and D1 increased PKCɛ kinase activity in a concentration-dependent manner (EC(50) values: 212-310nM), whereas E6 and G8 inhibited PKCɛ activity (IC(50) values: 103-233nM). None of these VHHs had an effect on the activity of the other novel PKC isozymes PKCδ and PKCθ. To our knowledge, these antibodies are the first described VHH activators and inhibitors for a protein kinase. Furthermore, the development of PKCɛ specific modulators is an important contribution to PKC research.


Subject(s)
Isoenzymes/immunology , Protein Kinase C-epsilon , Single-Chain Antibodies/immunology , Single-Chain Antibodies/isolation & purification , Amino Acid Sequence , Animals , Camelids, New World/immunology , Complementarity Determining Regions , Humans , Immunoglobulin Heavy Chains , Inhibitory Concentration 50 , Isoenzymes/antagonists & inhibitors , Peptide Library , Protein Binding , Protein Kinase C-epsilon/antagonists & inhibitors , Protein Kinase C-epsilon/immunology
10.
Shock ; 34(2): 125-32, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20016410

ABSTRACT

Staphylococcus aureus produces the superantigen toxic shock syndrome toxin 1 (TSST-1). When the bacterium invades the human circulation, this toxin can induce life-threatening gram-positive sepsis. Current sepsis treatment does not remove bacterial toxins. Variable domains of llama heavy-chain antibodies (VHH) against toxic shock syndrome toxin 1 ([alpha]-TSST-1 VHH) were previously found to be effective in vitro. We hypothesized that removing TSST-1 with [alpha]-TSST-1 VHH hemofiltration filters would ameliorate experimental sepsis in pigs. After assessing in vitro whether timely removing TSST-1 interrupted TSST-1-induced mononuclear cell TNF-[alpha] production, VHH-coated filters were applied in a porcine sepsis model. Clinical course, survival, plasma interferon [gamma], and TSST-1 levels were similar with and without VHH-coated filters as were TSST-1 concentrations before and after the VHH filter. Plasma TSST-1 levels were much lower than anticipated from the distribution of the amount of infused TSST-1, suggesting compartmentalization to space or adhesion to surface not accessible to hemofiltration or pheresis techniques. Removing TSST-1 from plasma was feasible in vitro. However, the [alpha]-TSST-1 VHH adsorption filter-based technique was ineffective in vivo, indicating that improvement of VHH-based hemofiltration is required. Sequestration likely prevented the adequate removal of TSST-1. The latter warrants further investigation of TSST-1 distribution and clearance in vivo.


Subject(s)
Immunoglobulin Heavy Chains/therapeutic use , Peptide Fragments/therapeutic use , Shock, Septic/prevention & control , Animals , Bacterial Toxins , Camelids, New World/immunology , Cells, Cultured , Enterotoxins , Female , Hemofiltration/methods , Humans , Leukocytes, Mononuclear/metabolism , Shock, Septic/immunology , Superantigens , Sus scrofa
11.
Biotechnol Bioeng ; 104(1): 143-51, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19475676

ABSTRACT

Toxic-shock syndrome is primarily caused by the Toxic-shock syndrome toxin 1 (TSST-1), which is secreted by the Gram-positive bacterium Staphylococcus aureus. The toxin belongs to a family of superantigens (SAgs) which exhibit several shared biological properties, including the induction of massive cytokine release and V(beta)-specific T-cell proliferation. In this study we explored the possibility to use monoclonal Variable domains of Llama Heavy-chain antibodies (VHH) in the immuno capturing of TSST-1 from plasma. Data is presented that the selected VHHs are highly specific for TSST-1 and can be efficiently produced in large amounts in yeast. In view of affinity chromatography, the VHHs are easily coupled to beads, and are able to deplete TSST-1 from plasma at very low, for example, pathologically relevant, concentrations. When spiked with 4 ng/mL TSST-1 more than 96% of TSST-1 was depleted from pig plasma. These data pave the way to further explore application of high-affinity columns in the specific immuno depletion of SAgs in experimental sepsis models and in sepsis in humans.


Subject(s)
Antigens, Bacterial/isolation & purification , Bacterial Toxins/isolation & purification , Chromatography, Affinity/methods , Enterotoxins/isolation & purification , Plasma/chemistry , Staphylococcus aureus/pathogenicity , Superantigens/isolation & purification , Animals , Antibodies, Monoclonal/immunology , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Camelids, New World , Enterotoxins/immunology , Enterotoxins/metabolism , Humans , Protein Binding , Sensitivity and Specificity , Superantigens/immunology
12.
Mol Immunol ; 46(10): 2022-8, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19359042

ABSTRACT

Therapeutic approaches aimed at targeting tumor surface markers using monoclonal antibodies provide a powerful strategy in cancer treatment. Here we report selection of single variable domains (VHH) of llama heavy chain antibodies, using a VHH-phage-display library. A reverse proteomic approach was used to identify the cognate proteins recognized by enriched VHH on HeLa cells. One of these VHH bound the integrin alpha 3 beta 1 (VLA-3) and was further characterized. Most interestingly, this VHH could inhibit VLA-3 mediated cell-matrix adhesion. Our approach provides a fast and efficient method to screen for novel cell surface markers on normal and tumor cells that may find diagnostic or therapeutic application in disease management or treatment.


Subject(s)
Antibodies/analysis , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Variable Region/immunology , Integrin alpha3beta1/immunology , Proteomics/methods , Adhesiveness , Amino Acid Sequence , Animals , Camelids, New World , Cell Adhesion , Cell Membrane/immunology , Enzyme-Linked Immunosorbent Assay , Extracellular Matrix/immunology , Fluorescent Antibody Technique , HeLa Cells , Humans , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Variable Region/chemistry , Immunoprecipitation , Integrin alpha3beta1/chemistry , Keratinocytes/cytology , Keratinocytes/immunology , Microscopy, Fluorescence , Molecular Sequence Data , Reproducibility of Results
13.
Biochem J ; 409(1): 193-8, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17910535

ABSTRACT

Incorporation of Trp (tryptophan) analogues into a protein may facilitate its structural analysis by spectroscopic techniques. Development of a biological system for the biosynthetic incorpor-ation of such analogues into proteins is of considerable importance. The Gram-negative Escherichia coli is the only prokaryotic expression host regularly used for the incorporation of Trp analogues into recombinant proteins. Here, we present the use of the versatile Gram-positive expression host Lactococcus lactis for the incorporation of Trp analogues. The availability of a tightly regulated expression system for this organism, the potential to secrete modified proteins into the growth medium and the construction of the trp-synthetase deletion strain PA1002 of L. lactis rendered this organism potentially an efficient tool for the incorporation of Trp analogues into recombinant proteins. The Trp analogues 7-azatryptophan, 5-fluorotryptophan and 5-hydroxytryptophan were incorporated with efficiencies of >97, >97 and 89% respectively. Interestingly, 5-methylTrp (5-methyltryptophan) could be incorporated with 92% efficiency. Successful biosynthetical incorporation of 5-methylTrp into recombinant proteins has not been reported previously.


Subject(s)
Biochemistry/methods , Lactococcus lactis/metabolism , Recombinant Proteins/chemistry , Tryptophan/analogs & derivatives , Tryptophan/chemistry , Bacterial Proteins/chemistry , Cloning, Molecular , Escherichia coli/metabolism , Gene Deletion , Gene Expression Regulation, Bacterial , Glycine/analogs & derivatives , Glycine/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Glyphosate
14.
Clin Vaccine Immunol ; 13(10): 1079-86, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16928888

ABSTRACT

Sepsis is a considerable health problem and a burden on the health care system. Endotoxin, or lipopolysaccharide (LPS), present in the outer membrane of gram-negative bacteria, is responsible for more than 50% of the sepsis cases and is, therefore, a legitimate target for therapeutic approaches against sepsis. In this study, we selected and characterized a llama single-chain antibody fragment (VHH) directed to Neisseria meningitidis LPS. The VHH, designated VHH 5G, showed affinity to purified LPS as well as to LPS on the surfaces of the bacteria. Epitope mapping using a panel of N. meningitidis mutants revealed that VHH 5G recognizes an epitope in the inner core of LPS, and as expected, the VHH proved to have broad specificity for LPS from different bacteria. Furthermore, this VHH blocked binding of LPS to target cells of the immune system, resulting in the inhibition of LPS signaling in whole blood. Moreover, it was found to remove LPS efficiently from aqueous solutions, including serum. The selected anti-LPS VHH is a leading candidate for therapies against LPS-mediated sepsis.


Subject(s)
Camelids, New World/immunology , Immunoglobulin Fab Fragments/physiology , Immunoglobulin Fab Fragments/therapeutic use , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/immunology , Signal Transduction/immunology , Animals , Binding Sites, Antibody , Binding, Competitive/immunology , Cells, Cultured , Escherichia coli/immunology , Humans , Immunoglobulin Fab Fragments/metabolism , Neisseria meningitidis/immunology , Protein Binding/immunology
15.
Methods ; 38(2): 144-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16414272

ABSTRACT

Mucosal immunization with subunit vaccines requires new types of antigen delivery vehicles and adjuvants for optimal immune responses. We have developed a non-living and non-genetically modified gram-positive bacterial delivery particle (GEM) that has built-in adjuvant activity and a high loading capacity for externally added heterologous antigens that are fused to a high affinity binding domain. This binding domain, the protein anchor (PA), is derived from the Lactococcus lactis AcmA cell-wall hydrolase, and contains three repeats of a LysM-type cell-wall binding motif. Antigens are produced as antigen-PA fusions by recombinant expression systems that secrete the hybrid proteins into the culture growth medium. GEM particles are then used as affinity beads to isolate the antigen-PA fusions from the complex growth media in a one step procedure after removal of the recombinant producer cells. This procedure is also highly suitable for making multivalent vaccines. The resulting vaccines are stable at room temperature, lack recombinant DNA, and mimic pathogens by their bacterial size, surface display of antigens and adjuvant activity of the bacterial components in the GEM particles. The GEM-based vaccines do not require additional adjuvant for eliciting high levels of specific antibodies in mucosal and systemic compartments.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigens/administration & dosage , Drug Delivery Systems/methods , Immunity, Mucosal/immunology , Lactococcus lactis/immunology , Vaccination/methods , Adjuvants, Immunologic/chemistry , Administration, Intranasal , Animals , Antibody Formation/immunology , Antigens/chemistry , Antigens/genetics , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Binding Sites/genetics , Gene Expression/genetics , Genetic Vectors/genetics , Hot Temperature , Immunoglobulin A/blood , Immunoglobulin A/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Lactococcus lactis/chemistry , Lactococcus lactis/genetics , Lung/immunology , Mice , Muramidase/genetics , Nose/immunology , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Streptococcus pneumoniae/immunology , Transformation, Genetic , Trichloroacetic Acid/chemistry
16.
J Biol Chem ; 278(26): 23874-81, 2003 Jun 27.
Article in English | MEDLINE | ID: mdl-12684515

ABSTRACT

The C-terminal region (cA) of the major autolysin AcmA of Lactococcus lactis contains three highly similar repeated regions of 45 amino acid residues (LysM domains), which are separated by nonhomologous sequences. The cA domain could be deleted without destroying the cell wall-hydrolyzing activity of the enzyme in vitro. This AcmA derivative was capable neither of binding to lactococcal cells nor of lysing these cells while separation of the producer cells was incomplete. The cA domain and a chimeric protein consisting of cA fused to the C terminus of MSA2, a malaria parasite surface antigen, bound to lactococcal cells specifically via cA. The fusion protein also bound to many other Gram-positive bacteria. By chemical treatment of purified cell walls of L. lactis and Bacillus subtilis, peptidoglycan was identified as the cell wall component interacting with cA. Immunofluorescence studies showed that binding is on specific locations on the surface of L. lactis, Enterococcus faecalis, Streptococcus thermophilus, B. subtilis, Lactobacillus sake, and Lactobacillus casei cells. Based on these studies, we propose that LysM-type repeats bind to peptidoglycan and that binding is hindered by other cell wall constituents, resulting in localized binding of AcmA. Lipoteichoic acid is a candidate hindering component. For L. lactis SK110, it is shown that lipoteichoic acids are not uniformly distributed over the cell surface and are mainly present at sites where no MSA2cA binding is observed.


Subject(s)
Cell Wall/chemistry , Gram-Positive Bacteria/chemistry , Peptidoglycan/chemistry , Bacillus subtilis/chemistry , Bacillus subtilis/ultrastructure , Binding Sites , Cell Wall/metabolism , Enterococcus faecalis/chemistry , Enterococcus faecalis/ultrastructure , Gram-Positive Bacteria/ultrastructure , Lactobacillus/chemistry , Lactobacillus/ultrastructure , Lactococcus lactis/chemistry , Lactococcus lactis/ultrastructure , Muramidase/metabolism , Peptidoglycan/metabolism , Protein Binding , Protein Structure, Tertiary , Repetitive Sequences, Nucleic Acid , Streptococcus/chemistry , Streptococcus/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...