Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
JAMA Intern Med ; 183(12): 1397-1398, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37955905

ABSTRACT

This case report describes a man in his 80s with a medical history of essential hypertension who presented to the emergency department with sudden onset epigastric pain.


Subject(s)
Myocardial Infarction , Humans , Myocardial Infarction/diagnosis , Electrocardiography
2.
Am J Hematol ; 96(6): 698-707, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33761144

ABSTRACT

Myeloproliferative neoplasms (MPNs) are characterized by a pathologic expansion of myeloid lineages. Mutations in JAK2, CALR and MPL genes are known to be three prominent MPN disease drivers. Mutant CALR (mutCALR) is an oncoprotein that interacts with and activates the thrombopoietin receptor (MPL) and represents an attractive target for targeted therapy of CALR mutated MPN. We generated a transgenic murine model with conditional expression of the human mutant exon 9 (del52) from the murine endogenous Calr locus. These mice develop essential thrombocythemia like phenotype with marked thrombocytosis and megakaryocytosis. The disease exacerbates with age showing prominent signs of splenomegaly and anemia. The disease is transplantable and mutCALR stem cells show proliferative advantage when compared to wild type stem cells. Transcriptome profiling of hematopoietic stem cells revealed oncogenic and inflammatory gene expression signatures. To demonstrate the applicability of the transgenic animals for immunotherapy, we treated mice with monoclonal antibody raised against the human mutCALR. The antibody treatment lowered platelet and stem cell counts in mutant mice. Secretion of mutCALR did not constitute a significant antibody sink. This animal model not only recapitulates human MPN but also serves as a relevant model for testing immunotherapeutic strategies targeting epitopes of the human mutCALR.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Calreticulin/antagonists & inhibitors , Disease Models, Animal , Hematopoietic Stem Cells/metabolism , Molecular Targeted Therapy , Thrombocythemia, Essential/therapy , Animals , Antibodies, Monoclonal/immunology , Blood Platelets/immunology , Blood Platelets/metabolism , Calreticulin/genetics , Calreticulin/immunology , Calreticulin/physiology , Exons/genetics , Frameshift Mutation , Gene Knock-In Techniques , Immunotherapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Radiation Chimera , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Splenomegaly/etiology , Thrombocythemia, Essential/blood , Thrombocythemia, Essential/complications , Thrombocythemia, Essential/genetics , Transcriptome
3.
Cytokine ; 111: 389-397, 2018 11.
Article in English | MEDLINE | ID: mdl-30463053

ABSTRACT

Streptococcuspneumoniae is a major human pathogen at the extremes of age. The elderly are particularly vulnerable to S.pneumoniae, the most common causative agent of bacterial pneumonia in this population. Despite the availability of vaccines and antibiotics, mortality rates associated with pneumococcal pneumonia in this age group remain high. In light of globally increasing life-expectancy, a better understanding of the patho-mechanisms of elderly pneumococcal pneumonia, including alterations in innate immune responses, is needed to develop improved therapies. In this study we aimed at investigating how increased susceptibility to pneumococcal infection relates to inflammation kinetics in the aged mouse pneumonia model by determining pulmonary cytokine and chemokine levels and comparing these parameters to those measured in young adult mice. Firstly, we detected overall higher pulmonary cytokine and chemokine levels in aged mice. However, upon induction of pneumococcal pneumonia in aged mice, delayed production of certain analytes, such as IFN-γ, MIG (CXCL9), IP-10 (CXCL10), MCP-1 (CCL2), TARC (CCL17) and MDC (CCL22) became apparent. In addition, aged mice were unable to control excess inflammatory responses: while young mice showed peak inflammatory responses at 20 h and subsequent resolution by 48 h post intranasal challenge, in aged mice increasing cytokine and chemokine levels were measured. These findings highlight the importance of considering multiple time points when delineating inflammatory responses to S.pneumoniae in an age-related context. Finally, correlation between pulmonary bacterial burden and cytokine or chemokine levels in young mice suggested that appropriately controlled inflammatory responses support the host to fight pneumococcal infection.


Subject(s)
Chemokines/metabolism , Cytokines/metabolism , Pneumococcal Infections/metabolism , Pneumonia, Pneumococcal/metabolism , Adult , Aged , Animals , Disease Models, Animal , Female , Humans , Inflammation/metabolism , Kinetics , Lung/metabolism , Mice , Mice, Inbred C57BL , Middle Aged , Streptococcus pneumoniae/pathogenicity , Young Adult
4.
Virulence ; 9(1): 1521-1538, 2018.
Article in English | MEDLINE | ID: mdl-30289054

ABSTRACT

Streptococcus pneumoniae isolates express up to three neuraminidases (sialidases), NanA, NanB and NanC, all of which cleave the terminal sialic acid of glycan-structures that decorate host cell surfaces. Most research has focused on the role of NanA with limited investigations evaluating the roles of all three neuraminidases in host-pathogen interactions. We generated two highly potent monoclonal antibodies (mAbs), one that blocks the enzymatic activity of NanA and one cross-neutralizing NanB and NanC. Total neuraminidase activity of clinical S. pneumoniae isolates could be inhibited by this mAb combination in enzymatic assays. To detect desialylation of cell surfaces by pneumococcal neuraminidases, primary human tracheal/bronchial mucocilial epithelial tissues were infected with S. pneumoniae and stained with peanut lectin. Simultaneous targeting of the neuraminidases was required to prevent desialylation, suggesting that inhibition of NanA alone is not sufficient to preserve terminal lung glycans. Importantly, we also found that all three neuraminidases increased the interaction of S. pneumoniae with human airway epithelial cells. Lectin-staining of lung tissues of mice pre-treated with mAbs before intranasal challenge with S. pneumoniae confirmed that both anti-NanA and anti-NanBC mAbs were required to effectively block desialylation of the respiratory epithelium in vivo. Despite this, no effect on survival, reduction in pulmonary bacterial load, or significant changes in cytokine responses were observed. This suggests that neuraminidases have no pivotal role in this murine pneumonia model that is induced by high bacterial challenge inocula and does not progress from colonization as it happens in the human host.


Subject(s)
Antibodies, Monoclonal/immunology , Bacterial Proteins/immunology , Neuraminidase/immunology , Pneumonia, Pneumococcal/immunology , Streptococcus pneumoniae/enzymology , A549 Cells , Animals , Antibodies, Bacterial/immunology , Disease Models, Animal , Epithelial Cells/microbiology , Female , Host-Pathogen Interactions , Humans , Lung/immunology , Lung/microbiology , Mice , Polysaccharides/metabolism , Trachea/cytology , Trachea/microbiology
5.
Article in English | MEDLINE | ID: mdl-29686149

ABSTRACT

Plasmid-encoded colistin resistance is emerging among extraintestinal pathogenic Escherichia coli strains, including those of the epidemic clone sequence type 131 (ST131)-H30. Mcr-1 transfers a phosphoethanolamine to the lipid A portion of lipopolysaccharide (LPS), conferring resistance to polymyxins. We investigated whether this modification changed the activity of the monoclonal antibody ASN-4, specific to the O25b side chain of ST131 LPS. We confirmed that, unlike colistin, ASN-4 retained its bactericidal and endotoxin-neutralizing activities and therefore offers a treatment option against extremely drug-resistant ST131 isolates.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Escherichia coli Infections/drug therapy , Escherichia coli Proteins/metabolism , Extraintestinal Pathogenic Escherichia coli/drug effects , Animals , Colistin/pharmacology , Drug Resistance, Multiple, Bacterial/genetics , Endotoxins/metabolism , Extraintestinal Pathogenic Escherichia coli/genetics , Female , Humans , Lipopolysaccharides/chemistry , Mice , Mice, Inbred BALB C
6.
Article in English | MEDLINE | ID: mdl-28874372

ABSTRACT

The multidrug-resistant H30 subclone of extraintestinal pathogenic Escherichia coli sequence type 131 (ST131-H30) has spread worldwide. This clone expresses a conserved lipopolysaccharide (LPS) O antigen, O25b. Previously, we described monoclonal antibodies (MAbs) specific to the O25b antigen and characterized them as diagnostic and therapeutic tools. In this study, evidence is provided that besides the previously shown complement-mediated bactericidal effect, an O25b-specific humanized MAb, A1124, also enhances opsonophagocytic uptake by the murine macrophage cell line RAW 264.7. Both phagocyte-dependent killing and phagocyte-independent killing, triggered by A1124, were confirmed in human whole blood. Furthermore, A1124 was shown to neutralize endotoxin activity of purified LPS of clinical isolates. This activity was demonstrated in vitro using both RAW 264.7 cells and a human Toll-like receptor 4 (TLR4) reporter cell line, as well as in a murine model of endotoxemia using purified LPS for challenge. Significant protective efficacy of A1124 at low doses (<1 mg/kg of body weight) was shown in murine and rat models of bacteremia. The contribution of the bactericidal and anti-inflammatory effects was dissected in the mouse bacteremia model through depletion of complement with cobra venom factor (CVF). Protective efficacy was lost in complement-depleted mice, suggesting the essential role of complement-mediated activities for protection in this model. These data suggest that A1124 exhibits different mechanisms of action, namely, direct complement-mediated and opsonophagocytic killing as well as endotoxin neutralization in various challenge models. Which of these activities are the most relevant in a clinical setting will need to be addressed by future translational studies.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Drug Resistance, Multiple, Bacterial/drug effects , Escherichia coli Infections/drug therapy , Extraintestinal Pathogenic Escherichia coli/drug effects , Animals , Blood/microbiology , Cell Line , Complement System Proteins/metabolism , Disease Models, Animal , Endotoxemia/drug therapy , Endotoxemia/microbiology , Endotoxins/metabolism , Extraintestinal Pathogenic Escherichia coli/isolation & purification , Female , Humans , Macrophages/microbiology , Mice, Inbred BALB C , O Antigens/immunology , Rats, Sprague-Dawley
7.
Virulence ; 8(7): 1203-1215, 2017 10 03.
Article in English | MEDLINE | ID: mdl-28103139

ABSTRACT

Klebsiella pneumoniae ST258 is a globally distributed multi-drug resistant pathogen responsible for severe invasive infections. In this study, the different virulence potential of K. pneumoniae ST258 isolates in endotoxin susceptible versus resistant animal models was shown. Furthermore, ST258 clinical isolates were found highly sensitive to the bactericidal effect of naive animal and human serum. These observations imply that LPS, released from the rapidly lysed bacteria, may contribute to the high mortality associated with ST258 bacteremia cases. A humanized version (mAb A1102) of a previously described murine mAb specific for the conserved LPS O-antigen, was tested for endotoxin neutralization. A1102 was able to neutralize TLR-4 activation by ST258-derived LPS in vitro with an efficacy exceeding that of polymyxin B by 3 orders of magnitude. Passive immunization with A1102 afforded a significant level of protection in a galactosamine-sensitized mouse model of endotoxemia, induced by ST258-derived LPS, or upon challenge with live bacteria. Efficacy was retained using an aglycosylated IgG, as well as upon complement depletion, suggesting that Fc-independent endotoxin neutralization may be the main protective mechanism in this model, in spite of the complement-dependent bactericidal and opsonic activities additionally observed for A1102 in vitro. Furthermore, rabbits that are naturally highly susceptible to endotoxin, were also significantly protected by low doses of A1102 when challenged with an ST258 strain. Given this unique mode of action and the high protective efficacy of this mAb, passive immunization, as prophylactic or adjunct therapeutic approach for the treatment of infections caused by ST258 isolates should be considered.


Subject(s)
Antibodies, Bacterial/immunology , Antibodies, Monoclonal/immunology , Endotoxins/immunology , Klebsiella Infections/drug therapy , Klebsiella pneumoniae/immunology , O Antigens/immunology , Animals , Antibodies, Bacterial/therapeutic use , Antibodies, Monoclonal/therapeutic use , Female , Humans , Immunization, Passive , Klebsiella Infections/immunology , Klebsiella Infections/microbiology , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/genetics , Male , Mice , Mice, Inbred BALB C , Neutralization Tests , Rabbits , Rats , Rats, Sprague-Dawley
8.
Int J Med Microbiol ; 306(2): 89-98, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26723873

ABSTRACT

Klebsiella pneumoniae ST258 is a globally disseminated, extremely drug resistant, nosocomial clone with limited treatment options. We show that the vast majority of ST258 isolates express modified d-galactan-I lipopolysaccharide O-antigen, termed hereinafter as D-galactan-III. The genetic determinant required for galactan-III synthesis was identified as a distinct operon adjacent to the rfb (wb) locus encoding D-galactan-I synthesis. The three genes within the operon encode predicted glycosyltransferases. Testing an isogenic transformant pair revealed that expression of D-galactan-III, in comparison to D-galactan-I, conferred improved survival in the presence of human serum. Eighty-three percent of the more than 200 ST258 draft genome sequences currently available carries the corresponding operon and hence these isolates are predicted to express galactan-III antigens. A D-galactan-III specific monoclonal antibody (mAb) was shown to bind to extracted LPS from a panel of ST258 isolates. The same mAb confirmed accessibility of galactan-III in surface staining of ST258 irrespective of the distinct capsular antigens expressed by both clades described previously. Based on these data, the galactan-III antigen may represent an attractive target for active and passive immunization approaches against K. pneumoniae ST258.


Subject(s)
Galactans/metabolism , Klebsiella pneumoniae/immunology , O Antigens/immunology , Animals , Antibodies, Monoclonal/immunology , Antigens, Surface/immunology , Cloning, Molecular , Epitopes/immunology , Female , Galactans/classification , Galactans/genetics , Galactans/immunology , Hybridomas , Klebsiella pneumoniae/classification , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/pathogenicity , Lipopolysaccharides/immunology , Magnetic Resonance Spectroscopy , Mice , Mice, Inbred BALB C , O Antigens/analysis , O Antigens/genetics , Operon/genetics , Virulence
9.
Antimicrob Agents Chemother ; 59(6): 3109-16, 2015.
Article in English | MEDLINE | ID: mdl-25779571

ABSTRACT

The Escherichia coli sequence type 131 (ST131)-O25b:H4 clone has spread worldwide and become responsible for a significant proportion of multidrug-resistant extraintestinal infections. We generated humanized monoclonal antibodies (MAbs) that target the lipopolysaccharide O25b antigen conserved within this lineage. These MAbs bound to the surface of live bacterial cells irrespective of the capsular type expressed. In a serum bactericidal assay in vitro, MAbs induced >95% bacterial killing in the presence of human serum as the complement source. Protective efficacy at low antibody doses was observed in a murine model of bacteremia. The mode of action in vivo was investigated by using aglycosylated derivatives of the protective MAbs. The significant binding to live E. coli cells and the in vitro and in vivo efficacy were corroborated in assays using bacteria grown in human serum to mimic relevant clinical conditions. Given the dry pipeline of novel antibiotics against multidrug-resistant Gram-negative pathogens, passive immunization with bactericidal antibodies offers a therapeutic alternative to control infections caused by E. coli ST131-O25b:H4.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Escherichia coli/drug effects , Escherichia coli/metabolism , O Antigens/metabolism , Animals , Drug Resistance, Multiple, Bacterial , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Female , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...