Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
MAbs ; 13(1): 1883239, 2021.
Article in English | MEDLINE | ID: mdl-33557673

ABSTRACT

Despite substantial technological advances in antibody library and display platform development, the number of approved biotherapeutics from displayed libraries remains limited. In vivo, 20-50% of peripheral B cells undergo a process of receptor editing, which modifies the variable and junctional regions of light chains to delete auto-reactive clones. However, in vitro antibody evolution relies primarily on interaction with antigen, with no in-built checkpoints to ensure that the selected antibodies have not acquired additional specificities or biophysical liabilities during the optimization process. We had previously observed an enrichment of positive charge in the complementarity-determining regions of an anti-IL-21 R antibody during affinity optimization, which correlated with more potent IL-21 neutralization, but poor in vivo pharmacokinetics (PK). There is an emerging body of data that has correlated antibody nonspecificity with poor PK in vivo, and established a series of screening assays that are predictive of this behavior. In this study we revisit the challenge of developing an anti-IL-21 R antibody that can effectively compete with IL-21 for its highly negatively charged paratope while maintaining favorable biophysical properties. In vitro deselection methods that included an excess of negatively charged membrane preparations, or deoxyribonucleic acid, during phage selection of optimization libraries were unsuccessful in avoiding enrichment of highly charged, nonspecific antibody variants. However, a combination of structure-guided rational library design, next-generation sequencing of library outputs and application of linear regression models resulted in the identification of an antibody that maintained high affinity for IL-21 R and exhibited a desirable stability and biophysical profile.


Subject(s)
Antibodies, Neutralizing/pharmacology , Drug Design , High-Throughput Nucleotide Sequencing , Interleukin-21 Receptor alpha Subunit/antagonists & inhibitors , Mutagenesis , Protein Engineering , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/metabolism , Antibody Specificity , Computer-Aided Design , Drug Stability , HEK293 Cells , Humans , Interleukin-21 Receptor alpha Subunit/immunology , Interleukin-21 Receptor alpha Subunit/metabolism , Protein Conformation , Protein Stability , Structure-Activity Relationship
2.
Elife ; 72018 01 04.
Article in English | MEDLINE | ID: mdl-29299998

ABSTRACT

Both TH2-dependent helminth killing and suppression of the TH2 effector response have been attributed to macrophages (MΦ) activated by IL-4 (M(IL-4)). To investigate how M(IL-4) contribute to diverse infection outcomes, the MΦ compartment of susceptible BALB/c mice and more resistant C57BL/6 mice was profiled during infection of the pleural cavity with the filarial nematode, Litomosoides sigmodontis. C57BL/6 mice exhibited a profoundly expanded resident MΦ (resMΦ) population, which was gradually replenished from the bone marrow in an age-dependent manner. Infection status did not alter the bone-marrow derived contribution to the resMΦ population, confirming local proliferation as the driver of resMΦ expansion. Significantly less resMΦ expansion was observed in the susceptible BALB/c strain, which instead exhibited an influx of monocytes that assumed an immunosuppressive PD-L2+ phenotype. Inhibition of monocyte recruitment enhanced nematode killing. Thus, the balance of monocytic vs. resident M(IL-4) numbers varies between inbred mouse strains and impacts infection outcome.


Subject(s)
Cell Movement , Cell Proliferation , Filariasis/immunology , Filariasis/pathology , Filarioidea/growth & development , Filarioidea/immunology , Macrophages/physiology , Animals , Disease Resistance , Disease Susceptibility , Macrophages/parasitology , Mice, Inbred BALB C , Mice, Inbred C57BL , Pleural Cavity/immunology , Pleural Cavity/parasitology
3.
PLoS Pathog ; 13(3): e1006233, 2017 03.
Article in English | MEDLINE | ID: mdl-28334040

ABSTRACT

Rapid reprogramming of the macrophage activation phenotype is considered important in the defense against consecutive infection with diverse infectious agents. However, in the setting of persistent, chronic infection the functional importance of macrophage-intrinsic adaptation to changing environments vs. recruitment of new macrophages remains unclear. Here we show that resident peritoneal macrophages expanded by infection with the nematode Heligmosomoides polygyrus bakeri altered their activation phenotype in response to infection with Salmonella enterica ser. Typhimurium in vitro and in vivo. The nematode-expanded resident F4/80high macrophages efficiently upregulated bacterial induced effector molecules (e.g. MHC-II, NOS2) similarly to newly recruited monocyte-derived macrophages. Nonetheless, recruitment of blood monocyte-derived macrophages to Salmonella infection occurred with equal magnitude in co-infected animals and caused displacement of the nematode-expanded, tissue resident-derived macrophages from the peritoneal cavity. Global gene expression analysis revealed that although nematode-expanded resident F4/80high macrophages made an anti-bacterial response, this was muted as compared to newly recruited F4/80low macrophages. However, the F4/80high macrophages adopted unique functional characteristics that included enhanced neutrophil-stimulating chemokine production. Thus, our data provide important evidence that plastic adaptation of MΦ activation does occur in vivo, but that cellular plasticity is outweighed by functional capabilities specific to the tissue origin of the cell.


Subject(s)
Macrophage Activation/immunology , Macrophages/immunology , Macrophages/microbiology , Salmonella Infections, Animal/microbiology , Strongylida Infections/microbiology , Animals , Coinfection , Flow Cytometry , Mice , Mice, Inbred C57BL , Nematospiroides dubius/immunology , Oligonucleotide Array Sequence Analysis , Salmonella Infections, Animal/immunology , Salmonella typhi/immunology , Strongylida Infections/immunology
4.
Nat Commun ; 7: 12651, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27582256

ABSTRACT

Fat-associated lymphoid clusters (FALC) are inducible structures that support rapid innate-like B-cell immune responses in the serous cavities. Little is known about the physiological cues that activate FALCs in the pleural cavity and more generally the mechanisms controlling B-cell activation in FALCs. Here we show, using separate models of pleural nematode infection with Litomosoides sigmodontis and Altenaria alternata induced acute lung inflammation, that inflammation of the pleural cavity rapidly activates mediastinal and pericardial FALCs. IL-33 produced by FALC stroma is crucial for pleural B1-cell activation and local IgM secretion. However, B1 cells are not the direct target of IL-33, which instead requires IL-5 for activation. Moreover, lung inflammation leads to increased IL-5 production by type 2 cytokine-producing innate lymphoid cells (ILC2) in the FALC. These findings reveal a link between inflammation, IL-33 release by FALC stromal cells, ILC2 activation and pleural B-cell activation in FALCs, resulting in local and antigen-specific IgM production.


Subject(s)
Alternaria/immunology , Filarioidea/immunology , Immunoglobulin M/immunology , Interleukin-33/immunology , Lymphoid Tissue/immunology , Pneumonia/immunology , Alternariosis/immunology , Alternariosis/parasitology , Animals , B-Lymphocytes/immunology , Cells, Cultured , Female , Filariasis/immunology , Filariasis/parasitology , Immunity, Innate/immunology , Interleukin-5/immunology , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout
5.
Immunobiology ; 220(7): 924-33, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25700973

ABSTRACT

Tissue resident macrophages have vital homeostatic roles in many tissues but their roles are less well defined in the heart. The present study aimed to identify the density, polarisation status and distribution of macrophages in the healthy murine heart and to investigate their ability to respond to immune challenge. Histological analysis of hearts from CSF-1 receptor (csf1-GFP; MacGreen) and CX3CR1 (Cx3cr1(GFP/+)) reporter mice revealed a sparse population of GFP positive macrophages that were evenly distributed throughout the left and right ventricular free walls and septum. F4/80+CD11b+ cardiac macrophages, sorted from myocardial homogenates, were able to phagocytose fluorescent beads in vitro and expressed markers typical of both 'M1' (IL-1ß, TNF and CCR2) and 'M2' activation (Ym1, Arg 1, RELMα and IL-10), suggesting no specific polarisation in healthy myocardium. Exposure to Th2 challenge by infection of mice with helminth parasites Schistosoma mansoni, or Heligmosomoides polygyrus, resulted in an increase in cardiac macrophage density, adoption of a stellate morphology and increased expression of Ym1, RELMα and CD206 (mannose receptor), indicative of 'M2' polarisation. This was dependent on recruitment of Ly6ChighCCR2+ monocytes and was accompanied by an increase in collagen content. In conclusion, in the healthy heart resident macrophages are relatively sparse and have a phagocytic role. Following Th2 challenge this population expands due to monocyte recruitment and adopts an 'M2' phenotype associated with increased tissue fibrosis.


Subject(s)
Heart/parasitology , Macrophages/immunology , Myocardium/immunology , Schistosomiasis mansoni/immunology , Strongylida Infections/immunology , Animals , Antigens, Differentiation/metabolism , CD11b Antigen/metabolism , CX3C Chemokine Receptor 1 , Green Fluorescent Proteins/genetics , Intercellular Signaling Peptides and Proteins/biosynthesis , Lectins/biosynthesis , Lectins, C-Type/biosynthesis , Mannose Receptor , Mannose-Binding Lectins/biosynthesis , Mice , Mice, Knockout , Nematospiroides dubius/immunology , Phagocytosis/immunology , Receptor, Macrophage Colony-Stimulating Factor/genetics , Receptors, Cell Surface/biosynthesis , Receptors, Chemokine/genetics , Schistosoma mansoni/immunology , Schistosomiasis mansoni/parasitology , Strongylida Infections/parasitology , Th2 Cells/immunology , beta-N-Acetylhexosaminidases/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...