Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 121(8): e2312621121, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38346191

ABSTRACT

One of the hallmarks of type 1 but also type 2 diabetes is pancreatic islet inflammation, associated with altered pancreatic islet function and structure, if unresolved. IL-1ß is a proinflammatory cytokine which detrimentally affects ß-cell function. In the course of diabetes, complement components, including the central complement protein C3, are deregulated. Previously, we reported high C3 expression in human pancreatic islets, with upregulation after IL-1ß treatment. In the current investigation, using primary human and rodent material and CRISPR/Cas9 gene-edited ß-cells deficient in C3, or producing only cytosolic C3 from a noncanonical in-frame start codon, we report a protective effect of C3 against IL-1ß-induced ß-cell death, that is attributed to the cytosolic fraction of C3. Further investigation revealed that intracellular C3 alleviates IL-1ß-induced ß-cell death, by interaction with and inhibition of Fyn-related kinase (FRK), which is involved in the response of ß-cells to cytokines. Furthermore, these data were supported by increased ß-cell death in vivo in a ß-cell-specific C3 knockout mouse. Our data indicate that a functional, cytoprotective association exists between FRK and cytosolic C3.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Islets of Langerhans , Mice , Animals , Humans , Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Cell Death , Cytokines/metabolism , Mice, Knockout
2.
Front Immunol ; 12: 726801, 2021.
Article in English | MEDLINE | ID: mdl-34539665

ABSTRACT

Neisseria gonorrhoeae is the etiological agent of gonorrhea, the second most common bacterial sexually transmitted infection worldwide. Reproductive sequelae of gonorrhea include infertility, ectopic pregnancy and chronic pelvic pain. Most antibiotics currently in clinical use have been rendered ineffective due to the rapid spread of antimicrobial resistance among gonococci. The developmental pipeline of new antibiotics is sparse and novel therapeutic approaches are urgently needed. Previously, we utilized the ability of N. gonorrhoeae to bind the complement inhibitor C4b-binding protein (C4BP) to evade killing by human complement to design a chimeric protein that linked the two N-terminal gonococcal binding domains of C4BP with the Fc domain of IgM. The resulting molecule, C4BP-IgM, enhanced complement-mediated killing of gonococci. Here we show that C4BP-IgM induced membrane perturbation through complement deposition and membrane attack complex pore insertion facilitates the access of antibiotics to their intracellular targets. Consequently, bacteria become more susceptible to killing by antibiotics. Remarkably, C4BP-IgM restored susceptibility to azithromycin of two azithromycin-resistant clinical gonococcal strains because of overexpression of the MtrC-MtrD-MtrE efflux pump. Our data show that complement activation can potentiate activity of antibiotics and suggest a role for C4BP-IgM as an adjuvant for antibiotic treatment of drug-resistant gonorrhea.


Subject(s)
Anti-Bacterial Agents/pharmacology , Complement Activation , Complement C4b-Binding Protein/administration & dosage , Drug Resistance, Bacterial/drug effects , Immunoglobulin M/administration & dosage , Neisseria gonorrhoeae/drug effects , Azithromycin/pharmacology , Ciprofloxacin/pharmacology , Complement C4b-Binding Protein/genetics , Humans , Immunoglobulin M/genetics , Neisseria gonorrhoeae/growth & development , Recombinant Fusion Proteins/administration & dosage , Spectinomycin/pharmacology
3.
Front Immunol ; 11: 2122, 2020.
Article in English | MEDLINE | ID: mdl-32983170

ABSTRACT

Moraxella catarrhalis is a human-specific commensal of the respiratory tract and an opportunistic pathogen. It is one of the leading cause of otitis media in children and of acute exacerbations in patients with chronic obstructive pulmonary disease, resulting in significant morbidity and economic burden. Vaccines and new immunotherapeutic strategies to treat this emerging pathogen are needed. Complement is a key component of innate immunity that mediates the detection, response, and subsequent elimination of invading pathogens. Many pathogens including M. catarrhalis have evolved complement evasion mechanisms, which include the binding of human complement inhibitors such as C4b-binding protein (C4BP) and Factor H (FH). Inhibiting C4BP and FH acquisition by M. catarrhalis may provide a novel therapeutic avenue to treat infections. To achieve this, we created two chimeric proteins that combined the Moraxella-binding domains of C4BP and FH fused to human immunoglobulin Fcs: C4BP domains 1 and 2 and FH domains 6 and 7 fused to IgM and IgG Fc, respectively. As expected, FH6-7/IgG displaced FH from the bacterial surface while simultaneously activating complement via Fc-C1q interactions, together increasing pathogen elimination. C4BP1-2/IgM also increased serum killing of the bacteria through enhanced complement deposition, but did not displace C4BP from the surface of M. catarrhalis. These Fc fusion proteins could act as anti-infective immunotherapies. Many microbes bind the complement inhibitors C4BP and FH through the same domains as M. catarrhalis, therefore these Fc fusion proteins may be promising candidates as adjunctive therapy against many different drug-resistant pathogens.


Subject(s)
Complement C4b-Binding Protein/pharmacology , Complement Factor H/pharmacology , Immunoglobulin Fc Fragments/pharmacology , Moraxella catarrhalis/drug effects , Recombinant Fusion Proteins/pharmacology , Animals , Binding, Competitive , Blood Bactericidal Activity , CHO Cells , Complement C3b/analysis , Complement C3d/analysis , Complement C4b-Binding Protein/genetics , Complement C4b-Binding Protein/metabolism , Complement Factor H/genetics , Complement Factor H/metabolism , Cricetinae , Cricetulus , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Immunoglobulin G/pharmacology , Immunoglobulin M/genetics , Immunoglobulin M/metabolism , Immunoglobulin M/pharmacology , Moraxella catarrhalis/metabolism , Protein Binding , Protein Domains , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
4.
JCI Insight ; 4(23)2019 12 05.
Article in English | MEDLINE | ID: mdl-31661468

ABSTRACT

Gonorrhea is a sexually transmitted infection with 87 million new cases per year globally. Increasing antibiotic resistance has severely limited treatment options. A mechanism that Neisseria gonorrhoeae uses to evade complement attack is binding of the complement inhibitor C4b-binding protein (C4BP). We screened 107 porin B1a (PorB1a) and 83 PorB1b clinical isolates randomly selected from a Swedish strain collection over the last 10 years and noted that 96/107 (89.7%) PorB1a and 16/83 (19.3%) PorB1b bound C4BP; C4BP binding substantially correlated with the ability to evade complement-dependent killing (r = 0.78). We designed 2 chimeric proteins that fused C4BP domains to the backbone of IgG or IgM (C4BP-IgG; C4BP-IgM) with the aim of enhancing complement activation and killing of gonococci. Both proteins bound gonococci (KD C4BP-IgM = 2.4 nM; KD C4BP-IgG 980.7 nM), but only hexameric C4BP-IgM efficiently outcompeted heptameric C4BP from the bacterial surface, resulting in enhanced complement deposition and bacterial killing. Furthermore, C4BP-IgM substantially attenuated the duration and burden of colonization of 2 C4BP-binding gonococcal isolates but not a non-C4BP-binding strain in a mouse vaginal colonization model using human factor H/C4BP-transgenic mice. Our preclinical data present C4BP-IgM as an adjunct to conventional antimicrobials for the treatment of gonorrhea.


Subject(s)
Complement C4b-Binding Protein/therapeutic use , Gonorrhea/drug therapy , Histocompatibility Antigens/therapeutic use , Immunoglobulin M/therapeutic use , Neisseria gonorrhoeae/drug effects , Animals , Disease Models, Animal , Female , Gonorrhea/immunology , Humans , Immunoglobulin G , Mice, Inbred BALB C , Mice, Transgenic , Porins , Protein Domains
SELECTION OF CITATIONS
SEARCH DETAIL
...