31 results on '"Sascha A Kristian"'
Search Results
2. Generation and Improvement of Effector Function of a Novel Broadly Reactive and Protective Monoclonal Antibody against Pneumococcal Surface Protein A of Streptococcus pneumoniae.
- Author
-
Sascha A Kristian, Takayuki Ota, Sarah S Bubeck, Rebecca Cho, Brian C Groff, Tsuguo Kubota, Giuseppe Destito, Cécile Martin, John Laudenslager, Lilia Koriazova, Tomoyuki Tahara, and Yutaka Kanda
- Subjects
Medicine ,Science - Abstract
A proof-of-concept study evaluating the potential of Streptococcus pneumoniae Pneumococcal Surface Protein A (PspA) as a passive immunization target was conducted. We describe the generation and isolation of several broadly reactive mouse anti-PspA monoclonal antibodies (mAbs). MAb 140H1 displayed (i) 98% strain coverage, (ii) activity in complement deposition and opsonophagocytic killing (OPK) assays, which are thought to predict the in vivo efficacy of anti-pneumococcal mAbs, (iii) efficacy in mouse sepsis models both alone and in combination with standard-of-care antibiotics, and (iv) therapeutic activity in a mouse pneumonia model. Moreover, we demonstrate that antibody engineering can significantly enhance anti-PspA mAb effector function. We believe that PspA has promising potential as a target for the therapy of invasive pneumococcal disease by mAbs, which could be used alone or in conjunction with standard-of-care antibiotics.
- Published
- 2016
- Full Text
- View/download PDF
3. Beta-Lactamase Repressor BlaI Modulates Staphylococcus aureus Cathelicidin Antimicrobial Peptide Resistance and Virulence.
- Author
-
Morgan A Pence, Nina M Haste, Hiruy S Meharena, Joshua Olson, Richard L Gallo, Victor Nizet, and Sascha A Kristian
- Subjects
Medicine ,Science - Abstract
BlaI is a repressor of BlaZ, the beta-lactamase responsible for penicillin resistance in Staphylococcus aureus. Through screening a transposon library in S. aureus Newman for susceptibility to cathelicidin antimicrobial peptide, we discovered BlaI as a novel cathelicidin resistance factor. Additionally, through integrational mutagenesis in S. aureus Newman and MRSA Sanger 252 strains, we confirmed the role of BlaI in resistance to human and murine cathelidicin and showed that it contributes to virulence in human whole blood and murine infection models. We further demonstrated that BlaI could be a target for innate immune-based antimicrobial therapies; by removing BlaI through subinhibitory concentrations of 6-aminopenicillanic acid, we were able to sensitize S. aureus to LL-37 killing.
- Published
- 2015
- Full Text
- View/download PDF
4. AGI-134: a fully synthetic α-Gal glycolipid that converts tumors into in situ autologous vaccines, induces anti-tumor immunity and is synergistic with an anti-PD-1 antibody in mouse melanoma models
- Author
-
Stephen M. Shaw, Jenny Middleton, Kim Wigglesworth, Amber Charlemagne, Oliver Schulz, Melanie S. Glossop, Giles F. Whalen, Robert Old, Mike Westby, Chris Pickford, Rinat Tabakman, Irit Carmi-Levy, Abi Vainstein, Ella Sorani, Arik A. Zur, and Sascha A. Kristian
- Subjects
Immunotherapy ,alpha-Gal ,anti-Gal ,Melanoma ,anti-PD-1 ,Checkpoint inhibition ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 ,Cytology ,QH573-671 - Abstract
Abstract Background Treatments that generate T cell-mediated immunity to a patient’s unique neoantigens are the current holy grail of cancer immunotherapy. In particular, treatments that do not require cumbersome and individualized ex vivo processing or manufacturing processes are especially sought after. Here we report that AGI-134, a glycolipid-like small molecule, can be used for coating tumor cells with the xenoantigen Galα1-3Galβ1-4GlcNAc (α-Gal) in situ leading to opsonization with pre-existing natural anti-α-Gal antibodies (in short anti-Gal), which triggers immune cascades resulting in T cell mediated anti-tumor immunity. Methods Various immunological effects of coating tumor cells with α-Gal via AGI-134 in vitro were measured by flow cytometry: (1) opsonization with anti-Gal and complement, (2) antibody-dependent cell-mediated cytotoxicity (ADCC) by NK cells, and (3) phagocytosis and antigen cross-presentation by antigen presenting cells (APCs). A viability kit was used to test AGI-134 mediated complement dependent cytotoxicity (CDC) in cancer cells. The anti-tumoral activity of AGI-134 alone or in combination with an anti-programmed death-1 (anti-PD-1) antibody was tested in melanoma models in anti-Gal expressing galactosyltransferase knockout (α1,3GT−/−) mice. CDC and phagocytosis data were analyzed by one-way ANOVA, ADCC results by paired t-test, distal tumor growth by Mantel–Cox test, C5a data by Mann–Whitney test, and single tumor regression by repeated measures analysis. Results In vitro, α-Gal labelling of tumor cells via AGI-134 incorporation into the cell membrane leads to anti-Gal binding and complement activation. Through the effects of complement and ADCC, tumor cells are lysed and tumor antigen uptake by APCs increased. Antigen associated with lysed cells is cross-presented by CD8α+ dendritic cells leading to activation of antigen-specific CD8+ T cells. In B16-F10 or JB/RH melanoma models in α1,3GT−/− mice, intratumoral AGI-134 administration leads to primary tumor regression and has a robust abscopal effect, i.e., it protects from the development of distal, uninjected lesions. Combinations of AGI-134 and anti-PD-1 antibody shows a synergistic benefit in protection from secondary tumor growth. Conclusions We have identified AGI-134 as an immunotherapeutic drug candidate, which could be an excellent combination partner for anti-PD-1 therapy, by facilitating tumor antigen processing and increasing the repertoire of tumor-specific T cells prior to anti-PD-1 treatment.
- Published
- 2019
- Full Text
- View/download PDF
5. AGI-134: a fully synthetic α-Gal glycolipid that converts tumors into in situ autologous vaccines, induces anti-tumor immunity and is synergistic with an anti-PD-1 antibody in mouse melanoma models
- Author
-
Abi Vainstein, Arik A. Zur, Ella Sorani, Giles F. Whalen, Jenny Middleton, Chris Pickford, Melanie S. Glossop, Robert Old, Oliver Schulz, Sascha A. Kristian, Irit Carmi-Levy, Amber Charlemagne, Mike Westby, Rinat Tabakman, Kim Wigglesworth, and Stephen M. Shaw
- Subjects
Cancer Research ,medicine.medical_treatment ,T cell ,Abscopal effect ,lcsh:RC254-282 ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigen ,Cancer immunotherapy ,Checkpoint inhibition ,Cancer vaccine ,Genetics ,medicine ,lcsh:QH573-671 ,Antigen-presenting cell ,Melanoma ,030304 developmental biology ,0303 health sciences ,biology ,alpha-Gal ,lcsh:Cytology ,Chemistry ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Tumor antigen ,Complement-dependent cytotoxicity ,anti-Gal ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,AGI-134 ,Cancer research ,biology.protein ,anti-PD-1 ,Immunotherapy ,Antibody ,Primary Research ,Intratumoral injection - Abstract
BackgroundTreatments that generate T cell-mediated immunity to a patient’s unique neoantigens are the current holy grail of cancer immunotherapy. In particular, treatments that do not require cumbersome and individualized ex vivo processing or manufacturing processes are especially sought after. Here we report that AGI-134, a glycolipid-like small molecule, can be used for coating tumor cells with the xenoantigen Galα1-3Galβ1-4GlcNAc (α-Gal) in situ leading to opsonization with pre-existing natural anti-α-Gal antibodies (in short anti-Gal), which triggers immune cascades resulting in T cell mediated anti-tumor immunity.MethodsVarious immunological effects of coating tumor cells with α-Gal via AGI-134 in vitro were measured by flow cytometry: (1) opsonization with anti-Gal and complement, (2) antibody-dependent cell-mediated cytotoxicity (ADCC) by NK cells, and (3) phagocytosis and antigen cross-presentation by antigen presenting cells (APCs). A viability kit was used to test AGI-134 mediated complement dependent cytotoxicity (CDC) in cancer cells. The anti-tumoral activity of AGI-134 alone or in combination with an anti-programmed death-1 (anti-PD-1) antibody was tested in melanoma models in anti-Gal expressing galactosyltransferase knockout (α1,3GT−/−) mice. CDC and phagocytosis data were analyzed by one-way ANOVA, ADCC results by paired t-test, distal tumor growth by Mantel–Cox test, C5a data by Mann–Whitney test, and single tumor regression by repeated measures analysis.ResultsIn vitro, α-Gal labelling of tumor cells via AGI-134 incorporation into the cell membrane leads to anti-Gal binding and complement activation. Through the effects of complement and ADCC, tumor cells are lysed and tumor antigen uptake by APCs increased. Antigen associated with lysed cells is cross-presented by CD8α+ dendritic cells leading to activation of antigen-specific CD8+ T cells. In B16-F10 or JB/RH melanoma models in α1,3GT−/−mice, intratumoral AGI-134 administration leads to primary tumor regression and has a robust abscopal effect, i.e., it protects from the development of distal, uninjected lesions. Combinations of AGI-134 and anti-PD-1 antibody shows a synergistic benefit in protection from secondary tumor growth.ConclusionsWe have identified AGI-134 as an immunotherapeutic drug candidate, which could be an excellent combination partner for anti-PD-1 therapy, by facilitating tumor antigen processing and increasing the repertoire of tumor-specific T cells prior to anti-PD-1 treatment.
- Published
- 2019
- Full Text
- View/download PDF
6. Topical nicotinic receptor activation improves wound bacterial infection outcomes and TLR2-mediated inflammation in diabetic mouse wounds
- Author
-
Mari Kishibe, Sascha A. Kristian, James Sinacore, Tina M. Griffin, Katherine A. Radek, and Melissa Goslawski
- Subjects
0301 basic medicine ,Inflammation ,Dermatology ,Pharmacology ,Receptors, Nicotinic ,Article ,Pathogenesis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Mice, Inbred NOD ,medicine ,Animals ,Nicotinic Agonists ,Receptor ,Acetylcholine receptor ,Wound Healing ,integumentary system ,business.industry ,Bacterial Infections ,Toll-Like Receptor 2 ,TLR2 ,Disease Models, Animal ,030104 developmental biology ,Nicotinic agonist ,030220 oncology & carcinogenesis ,Wound Infection ,Cholinergic ,Surgery ,medicine.symptom ,Inflammation Mediators ,Wound healing ,business - Abstract
The cholinergic anti-inflammatory pathway can directly affect skin antibacterial responses via nicotinic acetylcholine receptors (nAChRs). In particular, α7 nAChR (CHRNA7) present in the epidermis modulates the host response to wounding and/or wound bacterial infection. While physiologic inflammation is required to initiate normal wound repair and can be triggered by Toll-like receptor (TLR) activation, chronic inflammation is frequently observed in diabetic wounds and can occur, in part, via excessive TLR2 activation or production. Consequently, this can delay physiologic wound healing responses and increase diabetic host susceptibility to bacterial infection. In this study, we demonstrate that topical nAChR activation diminishes bacterial survival and systemic dissemination in a mouse model of diabetic wound infection, while reducing wound TLR2 production, relative to control mice. We further determined that the antimicrobial peptide activity of diabetic mouse wounds is increased compared to control mice, but this effect is lost following topical nAChR activation. Finally, we observed that human diabetic wounds exhibit impaired α7 nAChR (CHRNA7) abundance and localization relative to human control (nondiabetic) skin. These findings suggest that topical administration of nAChR agonists may improve wound healing and infection outcomes in diabetic wounds by dampening TLR2-mediated inflammation and antimicrobial peptide response, and that the diabetic microenvironment may promote aberrant CHRNA7 production/activation that likely contributes to diabetic wound pathogenesis.
- Published
- 2018
7. Retargeting pre-existing human antibodies to a bacterial pathogen with an alpha-Gal conjugated aptamer
- Author
-
Charles Edward Selkirk Roberts, Sascha A. Kristian, Emma Leire, Bradley Hall, Kary B. Mullis, Robert Old, Uri Galili, John Iacomini, Victor Nizet, John H. Hwang, and Mike Westby
- Subjects
Aptamer ,Phagocytosis ,Molecular Sequence Data ,Article ,Epitope ,Acetylglucosamine ,Microbiology ,Epitopes ,Immune system ,Streptococcal Infections ,Drug Discovery ,Animals ,Humans ,Genetics (clinical) ,Base Sequence ,biology ,Streptococcus ,Aptamers, Nucleotide ,Antibodies, Bacterial ,In vitro ,Anti-Bacterial Agents ,Antibody opsonization ,Immunization ,biology.protein ,Molecular Medicine ,Antibody - Abstract
The ever-increasing threat of multi-drug resistant bacterial infections has spurred renewed interest in alternative approaches to classical antibiotic therapy. In contrast to other mammals, humans do not express the galactose-α-1,3-galactosyl-β-1,4-N-acetyl-glucosamine (α-Gal) epitope. As a result of exposure of humans to α-Gal in the environment, a large proportion of circulating antibodies are specific for the trisaccharide. In this study, we examine whether these anti-Gal antibodies can be recruited and redirected to exert anti-bacterial activity. We show that a specific DNA aptamer conjugated to an α-Gal epitope at its 5′ end, herein termed an alphamer, can bind to group A Streptococcus (GAS) bacteria by recognition of a conserved region of the surface-anchored M protein. The anti-GAS alphamer was shown to recruit anti-Gal antibodies to the streptococcal surface in an α-Gal-specific manner, elicit uptake and killing of the bacteria by human phagocytes, and slow growth of invasive GAS in human whole blood. These studies provide a first in vitro proof of concept that alphamers have the potential to redirect pre-existing antibodies to bacteria in a specific manner and trigger an immediate antibacterial immune response. Further validation of this novel therapeutic approach of applying α-Gal technology in in vivo models of bacterial infection is warranted. • α-Gal-tagged aptamers lead to GAS opsonization with anti-Gal antibodies. • α-Gal-tagged aptamers confer phagocytosis and killing of GAS cells by human phagocytes. • α-Gal-tagged aptamers reduces replication of GAS in human blood. • α-Gal-tagged aptamers may have the potential to be used as novel passive immunization drugs.
- Published
- 2015
- Full Text
- View/download PDF
8. Correction: Generation and Improvement of Effector Function of a Novel Broadly Reactive and Protective Monoclonal Antibody against Pneumococcal Surface Protein A of Streptococcus pneumoniae
- Author
-
Lilia Koriazova, Cécile Martin, Tsuguo Kubota, Takayuki Ota, Yutaka Kanda, Sarah Sunrise Bubeck, Tomoyuki Tahara, Sascha A. Kristian, Brian C. Groff, Rebecca Cho, Giuseppe Destito, and John Laudenslager
- Subjects
0301 basic medicine ,Multidisciplinary ,biology ,medicine.drug_class ,Effector ,lcsh:R ,lcsh:Medicine ,medicine.disease ,Monoclonal antibody ,medicine.disease_cause ,Immunoglobulin G ,Microbiology ,Complement system ,03 medical and health sciences ,Pneumococcal infections ,030104 developmental biology ,Epitope mapping ,Streptococcus pneumoniae ,Immunology ,medicine ,biology.protein ,lcsh:Q ,Antibody ,lcsh:Science - Abstract
A proof-of-concept study evaluating the potential of Streptococcus pneumoniae Pneumococcal Surface Protein A (PspA) as a passive immunization target was conducted. We describe the generation and isolation of several broadly reactive mouse anti-PspA monoclonal antibodies (mAbs). MAb 140H1 displayed (i) 98% strain coverage, (ii) activity in complement deposition and opsonophagocytic killing (OPK) assays, which are thought to predict the in vivo efficacy of anti-pneumococcal mAbs, (iii) efficacy in mouse sepsis models both alone and in combination with standard-of-care antibiotics, and (iv) therapeutic activity in a mouse pneumonia model. Moreover, we demonstrate that antibody engineering can significantly enhance anti-PspA mAb effector function. We believe that PspA has promising potential as a target for the therapy of invasive pneumococcal disease by mAbs, which could be used alone or in conjunction with standard-of-care antibiotics.
- Published
- 2017
9. Intratumoral administration of the alpha-Gal glycolipid AGI-134 to induce tumor regression in a mouse model of melanoma
- Author
-
Jenny Middleton, Ella Sorani, Abi Vainstein Haras, Rinat Tabakman, Kim Wigglesworth, Sascha A. Kristian, Irit Carmi Levy, and Stephen M. Shaw
- Subjects
Cancer Research ,biology ,business.industry ,Melanoma ,medicine.disease ,Tumor antigen ,Complement system ,Lesion ,Ovalbumin ,Immune system ,Oncology ,Knockout mouse ,medicine ,Cancer research ,biology.protein ,medicine.symptom ,Antibody ,business - Abstract
68 Background: AGI-134 is a fully synthetic alpha-Gal glycolipid for intratumoral (i.t.) treatment of solid tumors to induce a patient-specific anti-tumor immune response. AGI-134 recruits pre-existing anti-Gal antibodies to the injected lesion, leading to complement activation and enhanced tumor antigen processing. Using the B16.F10 murine melanoma model, we have previously demonstrated that AGI-134 evokes a robust abscopal anti-tumor effect, with treatment of a primary lesion protecting mice from the growth of distant lesions. In the current study, we investigated the response of injected tumors to AGI-134 administration. Methods: Tumors were induced by s.c. injection of B16.F10 or ovalbumin expressing B16 (B16.OVA) cells into the flank of α1,3galactosyltransferase knock out mice. After reaching a treatable size, the tumors were injected twice, 24 hrs apart, with PBS or 1-1.25 mg AGI-134, and tumor growth monitored for up to 32 days. In addition, to measure the activation of complement after treatment with AGI-134, tumors were processed 2 hours after treatment and the i.t. concentration of complement fragment C5a determined by ELISA. Results: Almost 50% of AGI-134-treated B16.F10 tumors fully regressed vs. 24% in the PBS controls. Moreover, AGI-134 treatment had a significant survival benefit, with 23% of AGI-134-treated mice dying or requiring euthanisia due to tumor mass by Day 27 post treatment vs. 43% in the PBS groups (p < 0.05, Mantel-Cox test). In the B16.OVA model, 67% of the AGI-134 treated tumors fully regressed vs. 0% in PBS-treated mice in two independent assays. Comparison of C5a levels in B16.F10 tumors 2 hrs after AGI-134 or PBS treatment demonstrated that AGI-134 induced significant complement activation within injected tumors. Conclusions: Having previously shown that AGI-134 induces an abscopal effect that protects mice from the development of secondary tumors, we now show that AGI-134 also induces regression of injected tumors: AGI-134-treatment induces the activation of complement within the tumor and leads to complete regression of the tumor in significantly more AGI-134-treated mice than PBS-treated. Clinical trials with AGI-134 are planned for 2018.
- Published
- 2018
- Full Text
- View/download PDF
10. Biofilm Formation Induces C3a Release and ProtectsStaphylococcus epidermidisfrom IgG and Complement Deposition and from Neutrophil‐Dependent Killing
- Author
-
Ursula Sauder, Sascha A. Kristian, Regine Landmann, Friedrich Götz, Timo A. Birkenstock, and Dietrich Mack
- Subjects
Neutrophils ,Colony Count, Microbial ,Virulence ,Enzyme-Linked Immunosorbent Assay ,chemical and pharmacologic phenomena ,Biology ,Virulence factor ,Immunoglobulin G ,Microbiology ,Mice ,Catheters, Indwelling ,Staphylococcus epidermidis ,Animals ,Humans ,Immunology and Allergy ,Microbial Viability ,Biofilm ,Complement C3 ,Opsonin Proteins ,Staphylococcal Infections ,Flow Cytometry ,biology.organism_classification ,Complement system ,Mice, Inbred C57BL ,Antibody opsonization ,Microscopy, Electron ,Infectious Diseases ,Biofilms ,biology.protein ,Ex vivo - Abstract
BACKGROUND: Biofilm formation is considered to be an important virulence factor of the opportunistic pathogen Staphylococcus epidermidis. We hypothesized that biofilm formation could interfere with the deposition of immunoglobulins and complement on the bacterial surface, leading to diminished activation of the complement system and protection from killing by human phagocytes. METHODS: The killing of biofilm-encased and planktonically grown wild-type (wt) S. epidermidis and the killing of an isogenic biofilm-negative ica mutant (ica(-)) by human polymorphonuclear neutrophils (PMNs) were compared. C3a induction and deposition of C3b and immunoglobulin G (IgG) on the bacteria after opsonization with human serum were assessed by enzyme-linked immunosorbent assay, flow cytometry, and electron microscopy. The virulence of the bacterial strains was compared in a mouse model of catheter-associated infection. RESULTS: Biofilm-embedded wt S. epidermidis was killed less well by human PMNs and induced more C3a than planktonically grown wt and ica(-) S. epidermidis. However, the deposition of C3b and IgG on the bacterial surface was diminished in biofilm-encased staphylococci. wt S. epidermidis was more virulent in implant-associated infections and was killed more slowly than ica(-) in ex vivo assays of killing by PMNs. CONCLUSIONS: The results indicate that prevention of C3b and IgG deposition on the bacterial surface contributes to the biofilm-mediated protection of S. epidermidis from killing by PMNs.
- Published
- 2008
- Full Text
- View/download PDF
11. Impairment of innate immune killing mechanisms by bacteriostatic antibiotics
- Author
-
Xavier Lauth, Richard L. Gallo, Anjuli M. Timmer, Victor Nizet, Yechiel Shai, Sascha A. Kristian, George Y. Liu, Yosef Rosenfeld, and Neta Sal-Man
- Subjects
Staphylococcus aureus ,medicine.drug_class ,medicine.medical_treatment ,Antimicrobial peptides ,Antibiotics ,Drug resistance ,Biology ,medicine.disease_cause ,Biochemistry ,Membrane Potentials ,Microbiology ,Cathelicidin ,Mice ,Anti-Infective Agents ,Cathelicidins ,Drug Resistance, Bacterial ,Escherichia coli ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,Complement Inactivator Proteins ,Mice, Inbred BALB C ,Innate immune system ,Chloramphenicol ,biology.organism_classification ,Erythromycin ,Immunology ,Female ,Bacteria ,Antimicrobial Cationic Peptides ,Biotechnology ,medicine.drug - Abstract
Antibiotics are designed to support host defense in controlling infection. Here we describe a paradoxical inhibitory effect of bacteriostatic antibiotics on key mediators of mammalian innate immunity. When growth of species including Escherichia coli and Staphylococcus aureus is suppressed by chloramphenicol or erythromycin, the susceptibility of the bacteria to cathelicidin antimicrobial peptides or serum complement was markedly diminished. Survival of the bacteria in human whole blood, human wound fluid, or a mouse wound infection model was in turn increased after antibiotic-induced bacteriostasis. These findings provide a further rationale against the indiscriminate use of antibiotics.
- Published
- 2007
- Full Text
- View/download PDF
12. Beta-Lactamase Repressor BlaI Modulates Staphylococcus aureus Cathelicidin Antimicrobial Peptide Resistance and Virulence
- Author
-
Richard L. Gallo, Nina M. Haste, Joshua Olson, Victor Nizet, Hiruy S. Meharena, Sascha A. Kristian, Morgan A. Pence, and Cloeckaert, Axel
- Subjects
medicine.medical_treatment ,Penicillanic Acid ,lcsh:Medicine ,medicine.disease_cause ,Cathelicidin ,Mice ,2.1 Biological and endogenous factors ,Aetiology ,lcsh:Science ,Multidisciplinary ,Bacterial ,Staphylococcal Infections ,Antimicrobial ,3. Good health ,Infectious Diseases ,5.1 Pharmaceuticals ,Staphylococcus aureus ,Development of treatments and therapeutic interventions ,Infection ,Research Article ,medicine.drug ,General Science & Technology ,Penicillin Resistance ,Virulence ,Repressor ,Biology ,Staphylococcal infections ,beta-Lactamases ,Microbiology ,Vaccine Related ,Clinical Research ,Cathelicidins ,Biodefense ,Genetics ,medicine ,Animals ,Humans ,Prevention ,lcsh:R ,Gene Expression Regulation, Bacterial ,medicine.disease ,Methicillin-resistant Staphylococcus aureus ,Virology ,Penicillin ,Emerging Infectious Diseases ,Gene Expression Regulation ,DNA Transposable Elements ,lcsh:Q ,Antimicrobial Resistance ,Antimicrobial Cationic Peptides - Abstract
BlaI is a repressor of BlaZ, the beta-lactamase responsible for penicillin resistance in Staphylococcus aureus. Through screening a transposon library in S. aureus Newman for susceptibility to cathelicidin antimicrobial peptide, we discovered BlaI as a novel cathelicidin resistance factor. Additionally, through integrational mutagenesis in S. aureus Newman and MRSA Sanger 252 strains, we confirmed the role of BlaI in resistance to human and murine cathelidicin and showed that it contributes to virulence in human whole blood and murine infection models. We further demonstrated that BlaI could be a target for innate immune-based antimicrobial therapies; by removing BlaI through subinhibitory concentrations of 6-aminopenicillanic acid, we were able to sensitize S. aureus to LL-37 killing.
- Published
- 2015
13. The contribution of serum opacity factor to group A streptococcal epithelial cell invasion
- Author
-
Arthur Jeng, Bernard Beall, Sascha A. Kristian, Vivekanand Datta, Victor Nizet, and Anjuli M. Timmer
- Subjects
Operon ,Mutant ,Lactococcus lactis ,General Medicine ,Biology ,biology.organism_classification ,Molecular biology ,Microbiology ,Fibronectin ,Complementation ,Fibronectin binding ,biology.protein ,Heterologous expression ,Intracellular - Abstract
Serum opacity factor (SOF) is a bifunctional cell surface protein expressed by 40–50% of group A streptococcal (GAS) strains comprised of a C-terminal domain that binds fibronectin and an N-terminal domain that mediates opacification of mammalian sera. SOF is co-transcribed in a two-gene operon with another fibronectin-binding protein, SfbX. We compared the ability of an SOF(+) wild-type (WT) serotype M49 GAS strain and isogenic mutants lacking SOF or SfbX to invade cultured Hep-2 human pharyngeal epithelial cells. Elimination of SOF led to a significant decrease in Hep-2 intracellular invasion while loss of SfbX had minimal effect. The hypoinvasive phenotype of the SOF(−) mutant could be restored upon complementation with the sof gene on a plasmid vector, and heterologous expression of sof49 in M1 GAS or Lactococcus lactis conferred marked increases in Hep-2 cell invasion. Studies using a mutant sof49 in which the fibronectin-binding domain had been deleted in-frame confirmed that the C-terminal domain of SOF contributes to cellular invasion independent of the fibronectin-binding domain.
- Published
- 2006
- Full Text
- View/download PDF
14. Neutrophil chemotaxis by pathogen-associated molecular patterns - formylated peptides are crucial but not the sole neutrophil attractants produced by Staphylococcus aureus
- Author
-
Sascha A. Kristian, Kok P. M. van Kessel, Peter S. Margolis, Jos A. G. van Strijp, Michael Otto, Andreas Peschel, Erwin Bohn, Manuela Dürr, Joaquim Trias, Regine Landmann, and Gianluca Matteoli
- Subjects
Hydroxymethyl and Formyl Transferases ,Staphylococcus aureus ,Neutrophils ,Immunology ,In Vitro Techniques ,Biology ,Pertussis toxin ,Microbiology ,Mice ,chemistry.chemical_compound ,Virology ,Animals ,Humans ,CD11b Antigen ,Formyl peptide receptor ,Innate immune system ,Chemotactic Factors ,Pathogen-associated molecular pattern ,Chemotaxis ,Staphylococcal Infections ,N-Formylmethionine leucyl-phenylalanine ,biology.organism_classification ,Receptors, Formyl Peptide ,Immunity, Innate ,In vitro ,Mice, Inbred C57BL ,N-Formylmethionine Leucyl-Phenylalanine ,Chemotaxis, Leukocyte ,chemistry ,Mutation ,Calcium ,Female ,Peptides ,Bacteria - Abstract
Summary The chemotactic migration of phagocytes to sites of infection, guided by gradients of microbial molecules, plays a key role in the first line of host defence. Bac- teria are distinguished from eukaryotes by initiation of protein synthesis with formyl methionine. Syn- thetic formylated peptides (FPs) have been shown to be chemotactic for phagocytes, leading to the con- cept of FPs as pathogen-associated molecular pat- terns (PAMPs). However, it remains unclear whether FPs are major chemoattractants released by bacteria and whether further chemoattractants are produced. A Staphylococcus aureus mutant whose formyltrans- ferase gene was inactivated ( ∆ ∆ ∆ fmt ) produced no FPs and the in vitro and in vivo ability of ∆ fmt culture supernatants to recruit neutrophils was considerably reduced compared with those of the parental strain. However, some chemotactic activity was retained, indicating that bacteria produce also unknown, non- FP chemoattractants. The activity of these novel PAMPs was sensitive to pertussis toxin but insensi- tive to the formyl peptide receptor inhibitor CHIPS. ∆ fmt culture supernatants caused reduced calcium ion fluxes and reduced CD11b upregulation in neutro- phils compared with wild-type supernatants. These data demonstrate an important role of FPs in innate immunity against bacterial infections and indicate that host chemotaxis receptors recognize a larger set of bacterial molecules than previously thought.
- Published
- 2006
- Full Text
- View/download PDF
15. DNase Expression Allows the Pathogen Group A Streptococcus to Escape Killing in Neutrophil Extracellular Traps
- Author
-
James R. Feramisco, Ramy K. Aziz, John T. Buchanan, Malak Kotb, George Y. Liu, Amelia J. Simpson, Victor Nizet, and Sascha A. Kristian
- Subjects
Blood Bactericidal Activity ,Extracellular Traps ,Neutrophils ,Streptococcus pyogenes ,Virulence ,Biology ,Neutrophil Activation ,General Biochemistry, Genetics and Molecular Biology ,Microbiology ,Mice ,Animals ,Humans ,Fasciitis, Necrotizing ,Pathogen ,Deoxyribonucleases ,Neutrophil clearance ,Innate immune system ,Agricultural and Biological Sciences(all) ,Biochemistry, Genetics and Molecular Biology(all) ,Genetic Complementation Test ,Chemotaxis ,DNA ,Neutrophil extracellular traps ,Actins ,Disease Models, Animal ,Mutagenesis, Site-Directed ,Cattle ,Transformation, Bacterial ,Heterologous expression ,General Agricultural and Biological Sciences - Abstract
SummaryThe innate immune response plays a crucial role in satisfactory host resolution of bacterial infection. In response to chemotactic signals, neutrophils are early responding cells that migrate in large numbers to sites of infection. The recent discovery of secreted neutrophil extracellular traps (NETs) composed of DNA and histones opened a novel dimension in our understanding of the microbial killing capacity of these specialized leukocytes. M1 serotype strains of the pathogen Group A Streptococcus (GAS) are associated with invasive infections including necrotizing fasciitis (NF) and express a potent DNase (Sda1). Here we apply a molecular genetic approach of allelic replacement mutagenesis, single gene complementation, and heterologous expression to demonstrate that DNase Sda1 is both necessary and sufficient to promote GAS neutrophil resistance and virulence in a murine model of NF. Live fluorescent microscopic cell imaging and histopathological analysis are used to establish for the first time a direct linkage between NET degradation and bacterial pathogenicity. Inhibition of GAS DNase activity with G-actin enhanced neutrophil clearance of the pathogen in vitro and reduced virulence in vivo. The results demonstrate a significant role for NETs in neutrophil-mediated innate immunity, and at the same time identify a novel therapeutic target against invasive GAS infection.
- Published
- 2006
- Full Text
- View/download PDF
16. Lack of Wall Teichoic Acids inStaphylococcus aureusLeads to Reduced Interactions with Endothelial Cells and to Attenuated Virulence in a Rabbit Model of Endocarditis
- Author
-
Andreas Peschel, Christopher Weidenmaier, Yan-Qiong Xiong, Arnold S. Bayer, Klaus Dietz, Sascha A. Kristian, and Michael R. Yeaman
- Subjects
Staphylococcus aureus ,Endothelium ,Neutrophils ,Mutant ,Virulence ,Biology ,medicine.disease_cause ,Microbiology ,chemistry.chemical_compound ,Phagocytosis ,Cell Wall ,Cell Adhesion ,medicine ,Animals ,Humans ,Immunology and Allergy ,Teichoic acid ,Endothelial Cells ,Endocarditis, Bacterial ,Staphylococcal Infections ,In vitro ,Teichoic Acids ,Endothelial stem cell ,Infectious Diseases ,medicine.anatomical_structure ,chemistry ,Mutation ,Rabbits ,Staphylococcus - Abstract
Wall teichoic acids (WTAs) are major surface components of gram-positive bacteria that have recently been shown to play a key role in nasal colonization by Staphylococcus aureus. In the present study, we assessed the impact that WTAs have on endovascular infections by using a WTA-deficient S. aureus mutant ( Delta tagO). There were no significant differences detected between the isogenic parental strain (SA113) and the Delta tagO mutant in polymorphonuclear leukocyte-mediated opsonophagocytosis; killing by a prototypic platelet microbicidal protein; or binding to platelets, fibronectin, or fibrinogen. However, compared with the parental strain, the Delta tagO mutant adhered considerably less well to human endothelial cells, especially under flow conditions (70.3% reduction; P
- Published
- 2005
- Full Text
- View/download PDF
17. The ability of biofilm formation does not influence virulence of Staphylococcus aureus and host response in a mouse tissue cage infection model
- Author
-
Regine Landmann, Sarah E. Cramton, Friedrich Götz, Fabrizia Ferracin, Birgid Neumeister, Sascha A. Kristian, Andreas Peschel, and Thomas Golda
- Subjects
Staphylococcus aureus ,Chemokine CXCL2 ,Mutant ,Colony Count, Microbial ,Virulence ,Cell Count ,Inflammation ,Biology ,medicine.disease_cause ,Microbiology ,Bacterial Adhesion ,Bacterial genetics ,Mice ,In vivo ,medicine ,Animals ,Phagocytes ,Tumor Necrosis Factor-alpha ,Polysaccharides, Bacterial ,Biofilm ,Staphylococcal Infections ,Mice, Inbred C57BL ,Bacterial adhesin ,Infectious Diseases ,Biofilms ,Diffusion Chambers, Culture ,Chemokines ,medicine.symptom - Abstract
The virulence of Staphylococcus aureus Sa113 (SA113) and an isogenic ica deletion mutant (ica-), deficient in the production of polysaccharide intercellular adhesin (PIA), which is crucial for biofilm formation, was compared in a mouse tissue cage infection model. The minimal infective doses for the induction of persistent tissue infections in C57BL/6 mice were 10(3) CFU for both SA113 and the ica- mutant. Bacterial growth, initial adherence to surfaces within the implants and the course of inflammation including growth-dependent host TNF and MIP-2 release, influx of phagocytes and an accumulation of dead leukocytes were similar as well. Since SA113 expressed PIA in vivo, we could demonstrate that PIA and the lack of biofilm formation did not influence the capacity of S. aureus to induce persistent infections and did not modulate host responses in the mouse tissue cage model.
- Published
- 2004
- Full Text
- View/download PDF
18. Alanylation of Teichoic Acids ProtectsStaphylococcus aureusagainst Toll‐like Receptor 2–Dependent Host Defense in a Mouse Tissue Cage Infection Model
- Author
-
Andreas Peschel, Regine Landmann, Xavier Lauth, Birgid Neumeister, Victor Nizet, Sascha A. Kristian, and Friedrich Goetz
- Subjects
Staphylococcus aureus ,Chemokine CXCL2 ,Virulence ,Receptors, Cell Surface ,Biology ,medicine.disease_cause ,Microbiology ,Leukocyte Count ,Mice ,chemistry.chemical_compound ,Bacteriocins ,Leukocytes ,medicine ,Animals ,Immunology and Allergy ,Inflammation ,Mice, Knockout ,Toll-like receptor ,Teichoic acid ,Membrane Glycoproteins ,Innate immune system ,Tumor Necrosis Factor-alpha ,Monokines ,Toll-Like Receptors ,Staphylococcal Infections ,Toll-Like Receptor 2 ,Anti-Bacterial Agents ,Mice, Inbred C57BL ,Teichoic Acids ,carbohydrates (lipids) ,Disease Models, Animal ,TLR2 ,Infectious Diseases ,chemistry ,Mutation ,bacteria ,Female ,Tumor necrosis factor alpha ,Cell envelope ,Peptides - Abstract
Staphylococcus aureus is inherently resistant to cationic antimicrobial peptides because of alanylation of cell envelope teichoic acids. To test the effect of alanylated teichoic acids on virulence and host defense mediated by Toll-like receptor 2 (TLR2), wild-type (wt) S. aureus ATCC35556 (S.a.113) and its isogenic mutant expressing unalanylated teichoic acids (dlt − ) were compared in a tissue cage infection model that used C57BL/6 wt and TLR2-deficient mice. The minimum infective doses (MID) to establish persistent infection with S.a.113 were 10 3 and 10 2 colony-forming units (cfu) in wt and TLR2 −/− mice, respectively. The corresponding MID for dlt − were 5×10 5 and 10 3 cfu in wt and TLR2 −/− mice, respectively. Both mouse strains showed bacterial-load-dependent inflammation with elevations in tumor necrosis factor, macrophage inflammatory protein 2, and leukocytes, with increasing proportions of dead cells. These findings indicate that alanylated teichoic acids contribute to virulence of S. aureus, and TLR2 mediates host defense, which partly targets alanylated teichoic acids.
- Published
- 2003
- Full Text
- View/download PDF
19. MprF-Mediated Lysinylation of Phospholipids in Staphylococcus aureus Leads to Protection against Oxygen-Independent Neutrophil Killing
- Author
-
Manuela Dürr, Andreas Peschel, Sascha A. Kristian, Jos A. G. van Strijp, and Birgid Neumeister
- Subjects
Staphylococcus aureus ,Neutrophils ,Membrane lipids ,Immunology ,Virulence ,Biology ,medicine.disease_cause ,Microbiology ,Defensins ,Bacterial Proteins ,medicine ,Humans ,Defensin ,Phospholipids ,Host Response and Inflammation ,Innate immune system ,integumentary system ,Lysine ,Phosphatidylglycerols ,respiratory system ,Aminoacyltransferases ,biology.organism_classification ,Oxygen ,Infectious Diseases ,Beta defensin ,Myeloperoxidase ,biology.protein ,Parasitology ,Bacteria ,Antimicrobial Cationic Peptides - Abstract
Staphylococcus aureus achieves resistance to defensins and similar cationic antimicrobial peptides (CAMPs) by modifying anionic membrane lipids via MprF with l -lysine, which leads to repulsion of these host defense molecules. S. aureus Δ mprF , which lacks the modification, was very efficiently killed by neutrophil defensins and CAMP-producing leukocytes, even when oxygen-dependent killing was disrupted, but was as susceptible as wild-type bacteria to inactivation by myeloperoxidase or human monocytes lacking defensins. These results demonstrate the impact and specificity of MprF-mediated CAMP resistance and underscore the role of defensin-like peptides in innate host defense.
- Published
- 2003
- Full Text
- View/download PDF
20. Abstract 616: The novel α-Gal-based immunotherapy AGI-134 invokes CD8+ T cell-mediated immunity by driving tumor cell destruction, phagocytosis and tumor-associated antigen cross-presentation via multiple antibody-mediated effector functions
- Author
-
Sascha A. Kristian, Jenny Middleton, Amber Charlemagne, Oliver Schulz, and Stephen M. Shaw
- Subjects
Antibody-dependent cell-mediated cytotoxicity ,Cancer Research ,biology ,Chemistry ,030204 cardiovascular system & hematology ,Molecular biology ,Complement-dependent cytotoxicity ,Epitope ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,Antigen ,Polyclonal antibodies ,biology.protein ,Cytotoxic T cell ,Antibody ,Antigen-presenting cell ,030215 immunology - Abstract
Background: AGI-134 is a fully synthetic α-Gal (Galα1-3Galβ1-4GlcNAc-R) glycolipid that is being developed for the treatment of solid tumors. The α-Gal epitope is not expressed in humans, who, as a result of constant antigenic stimulation by α-Gal-bearing commensal bacteria, develop high titer natural antibodies to α-Gal. We have previously demonstrated that AGI-134 recruits anti-Gal antibodies to tumor cells in vitro, activating complement and driving phagocytosis by antigen presenting cells. AGI-134 also confers systemic protection from distal lesion development in a mouse model of melanoma and synergizes with anti-PD-11. Here we present further data characterizing the multiple pathways activated by the anti-Gal subclasses to drive AGI-134-mediated anti-tumor immunity. Results: Using quantitative methods, we demonstrate that human anti-Gal is composed of a diverse repertoire of effector antibodies in a panel of serum samples, with IgM, IgG1 and IgG2 being the major subclasses. Polyclonal anti-Gal IgG purified from human IVIG was, like human serum, comprised mainly of IgG1 and IgG2. When AGI-134 treated cells were incubated with human serum, binding of all anti-Gal subclasses was detected by flow cytometry, demonstrating that all anti-Gal subclasses can interact with AGI-134 treated cells and are available to activate downstream effector functions. When human serum was depleted of IgG, AGI-134 stimulated complement dependent cytotoxicity (CDC) was still effective, indicating that anti-Gal IgM is primarily responsible for deposition of complement on AGI-134 treated cells. AGI-134 treated cells incubated with purified polyclonal anti-Gal IgG activated FcγRIIIA on a reporter cell line and promoted natural killer cell-mediated antibody-dependent cell-mediated cytotoxicity (ADCC). Opsonization of AGI-134 treated human cancer cells with human anti-Gal and complement stimulated their phagocytosis by human monocyte-derived macrophages. Ovalbumin-expressing cells treated with AGI-134 and then incubated with human serum to initiate CDC were specifically phagocytosed by murine CD8α+ dendritic cells and the immunodominant antigen of ovalbumin, SIINFEKL, was cross-presented to CD8+ T cells. In conclusion, AGI-134 stimulates adaptive anti-tumor immunity through immune activation and antigen cross-presentation, which is driven by the diverse repertoire of anti-Gal antibodies. 1. Shaw, S. et al. Abstract 4862: AGI-134: a fully synthetic alpha-Gal glycolipid that prevents the development of distal lesions and is synergistic with an anti-PD-1 antibody in a mouse melanoma model. [abstract]. AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4862. Citation Format: Jenny L. Middleton, Oliver Schulz, Amber Charlemagne, Sascha A. Kristian, Stephen Michael Shaw. The novel α-Gal-based immunotherapy AGI-134 invokes CD8+ T cell-mediated immunity by driving tumor cell destruction, phagocytosis and tumor-associated antigen cross-presentation via multiple antibody-mediated effector functions [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 616. doi:10.1158/1538-7445.AM2017-616
- Published
- 2017
- Full Text
- View/download PDF
21. DltABCD-mediated d-alanylation of teichoic acids in Group A Streptococcus confers innate immune resistance
- Author
-
Rita Kansal, Sascha A. Kristian, Vivekanand Datta, Richard L. Gallo, Victor Nizet, Iris Fedtke, Andreas Peschel, and Christopher Weidenmaier
- Subjects
Teichoic acid ,Innate immune system ,Streptococcus ,Mutant ,General Medicine ,Biology ,medicine.disease_cause ,Group A ,Phenotype ,Microbiology ,chemistry.chemical_compound ,chemistry ,medicine ,Lysozyme ,Gene - Abstract
Group A Streptococcus (GAS) is a major cause of both mucosal and invasive human infections. Epithelial and leukocyte production of cationic antimicrobial peptides (AMPs) is an important aspect of mammalian innate immune defense against bacterial infection. In this study, we identify a specific GAS phenotype that confers resistance to host AMPs. Inactivation of the dltA gene in an invasive serotype M1 GAS isolate led to loss of teichoic acid d -alanylation. Compared to the wild-type strain, the GAS dltA mutant was found to be more susceptible to AMP and lysozyme killing. Killing of the dltA mutant by human PMN, which produce AMPs in large amounts, was greatly accelerated. Thus, teichoic acid d -alanylation may contribute to the ability of invasive GAS to bypass mucosal defenses and produce systemic infection.
- Published
- 2006
- Full Text
- View/download PDF
22. Role of teichoic acids in Staphylococcus aureus nasal colonization, a major risk factor in nosocomial infections
- Author
-
Graeme J. Nicholson, Hubert Kalbacher, Tanya Chanturiya, Sascha A. Kristian, Andreas Peschel, Birgid Neumeister, John F. Kokai-Kun, James J. Mond, Matthias Gross, and Christopher Weidenmaier
- Subjects
Staphylococcus aureus ,Population ,Mucous membrane of nose ,medicine.disease_cause ,Staphylococcal infections ,Bacterial Adhesion ,General Biochemistry, Genetics and Molecular Biology ,Microbiology ,chemistry.chemical_compound ,Risk Factors ,otorhinolaryngologic diseases ,medicine ,Animals ,Humans ,Colonization ,Sigmodontinae ,Cotton rat ,education ,Cells, Cultured ,Cross Infection ,Teichoic acid ,education.field_of_study ,Molecular Structure ,biology ,Epithelial Cells ,General Medicine ,Staphylococcal Infections ,respiratory system ,biology.organism_classification ,medicine.disease ,digestive system diseases ,Rats ,Anterior nares ,Teichoic Acids ,Nasal Mucosa ,medicine.anatomical_structure ,chemistry - Abstract
Colonization of the anterior nares in approximately 37% of the population is a major risk factor for severe Staphylococcus aureus infections. Here we show that wall teichoic acid (WTA), a surface-exposed staphylococcal polymer, is essential for nasal colonization and mediates interaction with human nasal epithelial cells. WTA-deficient mutants were impaired in their adherence to nasal cells, and were completely unable to colonize cotton rat nares. This study describes the first essential factor for S. aureus nasal colonization.
- Published
- 2004
- Full Text
- View/download PDF
23. Abstract 4862: AGI-134: a fully synthetic alpha-Gal glycolipid that prevents the development of distal lesions and is synergistic with an anti-PD-1 antibody in a mouse melanoma model
- Author
-
Jenny Middleton, Mel Glossop, Sascha A. Kristian, Mike Westby, Stephen Shaw, Kim Wigglesworth, Giles F. Whalen, Robert Old, and Chris Pickford
- Subjects
Cancer Research ,biology ,Chemistry ,Melanoma ,medicine.disease ,Molecular biology ,Primary tumor ,Epitope ,Complement system ,Immune system ,Oncology ,Antigen ,medicine ,biology.protein ,Antibody ,Antigen-presenting cell - Abstract
Background: AGI-134 is a fully synthetic glycolipid, composed of an alpha-Gal (Galá1-3Galâ1-4GlcNAc-R) sugar epitope attached via a linker to a lipid tail. Natural antibodies to the alpha-Gal epitope are responsible for the hyperacute rejection of xenografts in humans. It is proposed that intratumorally administered AGI-134 will incorporate into the cell membranes of the tumor cells, presenting the alpha-Gal epitope for binding of anti-Gal antibodies to the tumor cells. This will initiate an immune response that attacks the injected tumor and, through uptake of immune-complexed tumor antigens by antigen presenting cells, will create a patient-specific, systemic anti-tumor response against distant metastases. Results: We demonstrate that AGI-134 incorporates into tumor cell membranes in vitro and that the exposed alpha-Gal epitope binds anti-Gal IgG and IgM antibodies from human serum to the tumor cell surface. Using flow cytometry and a complement-dependent cytotoxicity assay we show that tumor cell opsonization with anti-Gal antibodies leads to deposition of complement proteins C3b and C5b-9, which ultimately leads to tumor cell lysis. Furthermore, we demonstrate that AGI-134-labeled tumor cells opsonized with human serum proteins are phagocytosed by professional APCs. Using the B16-F10 melanoma model in anti-Gal producing á1,3-galactosyltransferase knockout (GT KO) mice we present data to demonstrate that AGI-134 injection into a primary tumor provides significant dose-dependent protection from the development of established distant lesions. Using GT KO mouse serum we demonstrate in vitro that deposition of complement on AGI-134-labeled mouse tumor cells is both alpha-Gal and anti-Gal dependent. In vivo, we demonstrate that the effect of AGI-134 is due to the alpha-Gal moiety by replacing it with human blood group antigens. The protection from secondary lesions conferred by AGI-134 is long lasting in the GT KO mouse melanoma model (monitored up to 90 days). Importantly, when sub-optimal concentrations of AGI-134 were tested in vivo in combination with an anti-PD-1 antibody (RMP1-14), a significant enhancement in efficacy over either of the agents administered alone was observed. Citation Format: Stephen Shaw, Sascha Kristian, Kim Wigglesworth, Jenny Middleton, Mel Glossop, Giles Whalen, Robert Old, Mike Westby, Chris Pickford. AGI-134: a fully synthetic alpha-Gal glycolipid that prevents the development of distal lesions and is synergistic with an anti-PD-1 antibody in a mouse melanoma model. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4862.
- Published
- 2016
- Full Text
- View/download PDF
24. AGI-134, a fully synthetic α-Gal-based cancer immunotherapy: Synergy with an anti-PD-1 antibody and pre-clinical pharmacokinetic and toxicity profiles
- Author
-
Jenny Middleton, Kim Wigglesworth, Stephen M. Shaw, Chris Pickford, Robert Old, Mike Westby, Giles F. Whalen, Uri Galili, Melanie S. Glossop, and Sascha A. Kristian
- Subjects
Cancer Research ,biology ,medicine.medical_treatment ,030204 cardiovascular system & hematology ,Molecular biology ,Epitope ,In vitro ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Oncology ,Antigen ,Cancer immunotherapy ,In vivo ,Cancer cell ,biology.protein ,medicine ,Antibody ,030215 immunology - Abstract
3083Background: Unlike virtually all non-primate mammals, humans lack α-Gal (Gal-α-1,3-Gal-β-1,4-GlcNAc) epitopes and produce antibodies against α-Gal-positive antigens. Anti-α-Gal (anti-Gal) antibodies are among the most abundant antibodies in humans and responsible for hyper-acute rejection of α-Gal-positive xenotransplants. Studies using rabbit-derived α-Gal glycolipids provided a preclinical proof of principle for α-Gal glycolipids as cancer immunotherapy, inducing a CD8+ T cell response to tumor associated antigens (TAAs) that correlated with protection from distal tumor growth in mice. In the current study we show that a fully synthetic, α-Gal glycolipid-like small molecule, AGI-134, displays potent anti-tumor activity by engaging the cellular and humoral immune system against TAAs. Methods: in vitro and in vivo studies. Results: AGI-134 inserts into the plasma membranes of mouse and human cancer cells in vitro. Anti-Gal IgM and IgG bind to the α-Gal labeled cancer cells and mediate an anti-cancer c...
- Published
- 2016
- Full Text
- View/download PDF
25. Neutrophil antimicrobial defense against Staphylococcus aureus is mediated by phagolysosomal but not extracellular trap-associated cathelicidin
- Author
-
Sascha A. Kristian, Victor Nizet, Mathias Schmaler, Naja J. Jann, Andreas Peschel, Regine Landmann, Katherine A. Radek, and Richard L. Gallo
- Subjects
musculoskeletal diseases ,Lipopolysaccharides ,Staphylococcus aureus ,genetic structures ,Cell Survival ,Neutrophils ,medicine.medical_treatment ,Phagocytosis ,Immunology ,medicine.disease_cause ,Staphylococcal infections ,Cathelicidin ,Microbiology ,Mice ,Cathelicidins ,Extracellular ,medicine ,Immunology and Allergy ,Animals ,Humans ,Mice, Knockout ,NADPH oxidase ,biology ,musculoskeletal, neural, and ocular physiology ,Cell Biology ,Staphylococcal Infections ,medicine.disease ,Flow Cytometry ,nervous system diseases ,body regions ,Mice, Inbred C57BL ,Leukocytes: Tissue Interactions, Homeostasis and Host Defense ,biology.protein ,lipids (amino acids, peptides, and proteins) ,Lysosomes ,Intracellular ,Fluorescein-5-isothiocyanate ,Antimicrobial Cationic Peptides - Abstract
Study took advantage of the hyper-susceptible phenotype of S. aureus ΔdltA against cationic AMPs to investigate the impact of the murine cathelicidin CRAMP to identify its key site of action in neutrophils. Neutrophils kill invading pathogens by AMPs, including cathelicidins, ROS, and NETs. The human pathogen Staphylococcus aureus exhibits enhanced resistance to neutrophil AMPs, including the murine cathelicidin CRAMP, in part, as a result of alanylation of teichoic acids by the dlt operon. In this study, we took advantage of the hypersusceptible phenotype of S. aureus ΔdltA against cationic AMPs to study the impact of the murine cathelicidin CRAMP on staphylococcal killing and to identify its key site of action in murine neutrophils. We demonstrate that CRAMP remained intracellular during PMN exudation from blood and was secreted upon PMA stimulation. We show first evidence that CRAMP was recruited to phagolysosomes in infected neutrophils and exhibited intracellular activity against S. aureus. Later in infection, neutrophils produced NETs, and immunofluorescence revealed association of CRAMP with S. aureus in NETs, which similarly killed S. aureus wt and ΔdltA, indicating that CRAMP activity was reduced when associated with NETs. Indeed, the presence of DNA reduced the antimicrobial activity of CRAMP, and CRAMP localization in response to S. aureus was independent of the NADPH oxidase, whereas killing was partially dependent on a functional NADPH oxidase. Our study indicates that neutrophils use CRAMP in a timed and locally coordinated manner in defense against S. aureus.
- Published
- 2009
26. The CiaR Response Regulator in Group B Streptococcus Promotes Intracellular Survival and Resistance to Innate Immune Defenses ▿
- Author
-
Kelly S. Doran, Sascha A. Kristian, Darin Quach, Joshua D. Bryan, Nina M. van Sorge, and Daniel W. Shelver
- Subjects
Male ,Neutrophils ,Antimicrobial peptides ,Molecular Sequence Data ,Virulence ,Biology ,medicine.disease_cause ,Microbiology ,Neonatal meningitis ,Cell Line ,Streptococcus agalactiae ,Mice ,Gentamicin protection assay ,Bacterial Proteins ,Immunity ,Streptococcal Infections ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Molecular Biology ,reproductive and urinary physiology ,Cells, Cultured ,Molecular Biology of Pathogens ,Innate immune system ,Macrophages ,Endothelial Cells ,Gene Expression Regulation, Bacterial ,medicine.disease ,bacterial infections and mycoses ,Immunity, Innate ,Immunology ,Protein Kinases ,Sequence Alignment ,Intracellular - Abstract
Group B Streptococcus (GBS) is major cause of invasive disease in newborn infants and the leading cause of neonatal meningitis. To gain access to the central nervous system (CNS), GBS must not only subvert host defenses in the bloodstream but also invade and survive within brain microvascular endothelial cells (BMEC), the principal cell layer composing the blood-brain barrier (BBB). While several GBS determinants that contribute to the invasion of BMEC have been identified, little is known about the GBS factors that are required for intracellular survival and ultimate disease progression. In this study we sought to identify these factors by screening a random GBS mutant library in an in vitro survival assay. One mutant was identified which contained a disruption in a two-component regulatory system homologous to CiaR/CiaH, which is present in other streptococcal pathogens. Deletion of the putative response regulator, ciaR , in GBS resulted in a significant decrease in intracellular survival within neutrophils, murine macrophages, and human BMEC, which was linked to increased susceptibility to killing by antimicrobial peptides, lysozyme, and reactive oxygen species. Furthermore, competition experiments with mice showed that wild-type GBS had a significant survival advantage over the GBS Δ ciaR mutant in the bloodstream and brain. Microarray analysis comparing gene expression between wild-type and Δ ciaR mutant GBS bacteria revealed several CiaR-regulated genes that may contribute to stress tolerance and the subversion of host defenses by GBS. Our results identify the GBS CiaR response regulator as a crucial factor in GBS intracellular survival and invasive disease pathogenesis.
- Published
- 2008
27. The GraRS regulatory system controls Staphylococcus aureus susceptibility to antimicrobial host defenses
- Author
-
Andreas Peschel, Silvia Herbert, Sascha A. Kristian, Victor Nizet, Friedrich Götz, Dirk Kraus, and Arya Khosravi
- Subjects
Microbiology (medical) ,Staphylococcus aureus ,Neutrophils ,medicine.medical_treatment ,lcsh:QR1-502 ,Virulence ,Kidney ,Staphylococcal infections ,medicine.disease_cause ,Microbiology ,lcsh:Microbiology ,Cathelicidin ,Mice ,Cathelicidins ,Genes, Regulator ,Operon ,medicine ,Animals ,Humans ,Cells, Cultured ,Regulator gene ,Mice, Inbred BALB C ,Alanine ,biology ,Lysine ,Cell Membrane ,Cytochromes c ,Phosphatidylglycerols ,Staphylococcal Infections ,medicine.disease ,Antimicrobial ,Immunity, Innate ,Teichoic Acids ,Genes, Bacterial ,Myeloperoxidase ,biology.protein ,Female ,Gene Deletion ,Research Article ,Antimicrobial Cationic Peptides - Abstract
Background Modification of teichoic acids with D-alanine by the products of the dlt operon protects Gram-positive bacteria against major antimicrobial host defense molecules such as defensins, cathelicidins, myeloperoxidase or phospholipase. The gra RS regulatory genes have recently been implicated in the control of D-alanylation in Staphylococcus aureus. Results To determine the impact of the GraRS regulatory system on resistance to antimicrobial host defense mechanisms and virulence of S. aureus, we compared inactivation of S. aureus SA113 wild type and its isogenic gra RS deletion mutant by the human cathelicidin LL-37 or human neutrophil granulocytes in vitro, and the ability to cause infection in vivo. We show here that gra RS deletion considerably alters bacterial surface charge, increases susceptibility to killing by human neutrophils or the defense peptide LL-37, and attenuates virulence of S. aureus in a mouse infection model. Conclusion Our results indicate that S. aureus can regulate its surface properties in order to overcome innate host defenses.
- Published
- 2008
- Full Text
- View/download PDF
28. Serum opacity factor promotes group A streptococcal epithelial cell invasion and virulence
- Author
-
Anjuli M. Timmer, Arthur Jeng, Victor Nizet, Vivekanand Datta, Bernard Beall, Sascha A. Kristian, Christine M. Gillen, and Mark J. Walker
- Subjects
Male ,Streptococcus pyogenes ,Mutant ,Virulence ,Biology ,medicine.disease_cause ,Microbiology ,Virulence factor ,Mice ,Bacterial Proteins ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Molecular Biology ,Intracellular Signaling Peptides and Proteins ,Epithelial Cells ,Fibronectins ,Complementation ,Fibronectin ,Fibronectin binding ,Mutation ,biology.protein ,Heterologous expression ,Carrier Proteins ,Peptide Hydrolases ,Protein Binding - Abstract
Serum opacity factor (SOF) is a bifunctional cell surface protein expressed by 40-50% of group A streptococcal (GAS) strains comprised of a C-terminal domain that binds fibronectin and an N-terminal domain that mediates opacification of mammalian sera. The sof gene was recently discovered to be cotranscribed in a two-gene operon with a gene encoding another fibronectin-binding protein, sfbX. We compared the ability of a SOF(+) wild-type serotype M49 GAS strain and isogenic mutants lacking SOF or SfbX to invade cultured HEp-2 human pharyngeal epithelial cells. Elimination of SOF led to a significant decrease in HEp-2 intracellular invasion while loss of SfbX had minimal effect. The hypoinvasive phenotype of the SOF(-) mutant could be restored upon complementation with the sof gene on a plasmid vector, and heterologous expression of sof49 in M1 GAS or Lactococcus lactis conferred marked increases in HEp-2 cell invasion. Studies using a mutant sof49 gene lacking the fibronectin-binding domain indicated that the N-terminal opacification domain of SOF contributes to HEp-2 invasion independent of the C-terminal fibronectin binding domain, findings corroborated by observations that a purified SOF N-terminal peptide could promote latex bead adherence to HEp-2 cells and inhibit GAS invasion of HEp-2 cells in a dose-dependent manner. Finally, the first in vivo studies to employ a single gene allelic replacement mutant of SOF demonstrate that this protein contributes to GAS virulence in a murine model of necrotizing skin infection.
- Published
- 2006
29. D-alanylation of teichoic acids promotes group a streptococcus antimicrobial peptide resistance, neutrophil survival, and epithelial cell invasion
- Author
-
Vivekanand Datta, Iris Fedtke, Andreas Peschel, Richard L. Gallo, Victor Nizet, Christopher Weidenmaier, Rita Kansal, and Sascha A. Kristian
- Subjects
Cell Survival ,Neutrophils ,Streptococcus pyogenes ,Virulence Factors ,Mutant ,Antimicrobial peptides ,Biology ,medicine.disease_cause ,Microbiology ,Bacterial Adhesion ,chemistry.chemical_compound ,Phagocytosis ,Streptococcal Infections ,Operon ,medicine ,Humans ,Molecular Biology ,Cells, Cultured ,Molecular Biology of Pathogens ,Teichoic acid ,Alanine ,Virulence ,Streptococcus ,Toxic shock syndrome ,Epithelial Cells ,Hydrogen-Ion Concentration ,medicine.disease ,Antimicrobial ,Cathelicidins ,Teichoic Acids ,chemistry ,Mutagenesis ,Muramidase ,Antimicrobial Cationic Peptides - Abstract
Group A streptococcus (GAS) is a leading cause of severe, invasive human infections, including necrotizing fasciitis and toxic shock syndrome. An important element of the mammalian innate defense system against invasive bacterial infections such as GAS is the production of antimicrobial peptides (AMPs) such as cathelicidins. In this study, we identify a specific GAS phenotype that confers resistance to host AMPs. Allelic replacement of the dltA gene encoding d -alanine- d -alanyl carrier protein ligase in an invasive serotype M1 GAS isolate led to loss of teichoic acid d -alanylation and an increase in net negative charge on the bacterial surface. Compared to the wild-type (WT) parent strain, the GAS Δ dltA mutant exhibited increased susceptibility to AMP and lysozyme killing and to acidic pH. While phagocytic uptake of WT and Δ dltA mutants by human neutrophils was equivalent, neutrophil-mediated killing of the Δ dltA strain was greatly accelerated. Furthermore, we observed the Δ dltA mutant to be diminished in its ability to adhere to and invade cultured human pharyngeal epithelial cells, a likely proximal step in the pathogenesis of invasive infection. Thus, teichoic acid d -alanylation may contribute in multiple ways to the propensity of invasive GAS to bypass mucosal defenses and produce systemic infection.
- Published
- 2005
30. Bacterial resistance to antimicrobial host defenses--an emerging target for novel antiinfective strategies?
- Author
-
Andreas Peschel, Christopher Weidenmaier, and Sascha A. Kristian
- Subjects
Pharmacology ,Teichoic acid ,Clinical Biochemistry ,Antimicrobial peptides ,Drug Resistance ,Pathogenic bacteria ,Drug Resistance, Microbial ,Biology ,Antimicrobial ,medicine.disease_cause ,Bacterial cell structure ,Microbiology ,Cathelicidins ,chemistry.chemical_compound ,Drug Delivery Systems ,chemistry ,Anti-Infective Agents ,Drug Discovery ,medicine ,Molecular Medicine ,Animals ,Humans ,Peptidoglycan ,Cell envelope - Abstract
Increasing bacterial resistance to virtually all available antibiotics causes an urgent need for new antimicrobial drugs, drug targets and therapeutic concepts. This review focuses on strategies to render bacteria highly susceptible to the antimicrobial arsenal of the immune system by targeting bacterial immune escape mechanisms that are conserved in a major number of pathogens. Virtually all innate molecules that inactivate bacteria, ranging from antimicrobial peptides such as defensins and cathelicidins to bacteriolytic enzymes such as lysozyme and group IIA phospholipase A2, are highly cationic in order to facilitate binding to the anionic bacterial cell envelopes. Bacteria have found ways to modulate their anionic cell wall polymers such as peptidoglycan, lipopolysaccharide, teichoic acid or phospholipids by introducing positively charged groups. Two of these mechanisms involving the transfer of D-alanine into teichoic acids and of L-lysine into phospholipids, respectively, have been identified and characterized in Staphylococcus aureus, a major human pathogen in community- and hospital-acquired infections. Inactivation of the responsible genes, dltABCD for alanylation of teichoic acids and mprF for lysinylation of phosphatidylglycerol, renders S. aureus highly susceptible to many human antimicrobial molecules and leads to profoundly attenuated virulence in several animal models. dltABCD- and mprF-related genes are found in the genomes of many bacterial pathogens indicating that the escape from human host defenses by modulation of the cell envelope is a general trait in pathogenic bacteria. This review suggests that inhibitors of DltABCD or MprF should have great potential in complementing or replacing the conventional antibiotic therapies.
- Published
- 2003
31. Staphylococcus aureus strains lacking D-alanine modifications of teichoic acids are highly susceptible to human neutrophil killing and are virulence attenuated in mice
- Author
-
Andreas Peschel, Birgid Neumeister, Marion Faigle, Christopher Weidenmaier, Sascha A. Kristian, Friedrich Götz, L. Vincent Collins, Jos A. G. van Strijp, and Kok P. M. van Kessel
- Subjects
Staphylococcus aureus ,alpha-Defensins ,Neutrophils ,Antimicrobial peptides ,Virulence ,Biology ,medicine.disease_cause ,Monocytes ,Microbiology ,chemistry.chemical_compound ,Mice ,Bacterial Proteins ,Phagocytosis ,medicine ,Immunology and Allergy ,Animals ,Humans ,Defensin ,Teichoic acid ,Alanine ,Membrane Transport Proteins ,Staphylococcal Infections ,Respiratory burst ,Anti-Bacterial Agents ,Teichoic Acids ,Disease Models, Animal ,Infectious Diseases ,chemistry ,Mutation ,Female ,Cell envelope ,Staphylococcus - Abstract
Staphylococcus aureus is resistant to alpha-defensins, antimicrobial peptides that play an important role in oxygen-independent killing of human neutrophils. The dlt operon mediates d-alanine incorporation into teichoic acids in the staphylococcal cell envelope and is a determinant of defensin resistance. By using S. aureus wild-type (WT) and Dlt- bacteria, the relative contributions of oxygen-dependent and -independent antimicrobial phagocyte components were analyzed. The Dlt- strain was efficiently killed by human neutrophils even in the absence of a functional respiratory burst, whereas the killing of the WT organism was strongly diminished when the respiratory burst was inhibited. Human monocytes, which do not produce defensins, inactivated the WT and Dlt- bacteria with similar efficiencies. In addition, mice injected with the Dlt- strain had significantly lower rates of sepsis and septic arthritis and fewer bacteria in the kidneys, compared with mice infected with the WT strain.
- Published
- 2001
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.