SwePub
Sök i SwePub databas

  Utökad sökning

Träfflista för sökning "WFRF:(Maziarz Karolina) "

Sökning: WFRF:(Maziarz Karolina)

  • Resultat 1-6 av 6
Sortera/gruppera träfflistan
   
NumreringReferensOmslagsbildHitta
1.
  •  
2.
  • Bettoni, Serena, et al. (författare)
  • C4BP-IgM protein as a therapeutic approach to treat Neisseria gonorrhoeae infections
  • 2019
  • Ingår i: JCI Insight. - : American Society for Clinical Investigation (ASCI). - 2379-3708. ; 4:23
  • Tidskriftsartikel (refereegranskat)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.
  •  
3.
  • Bettoni, Serena, et al. (författare)
  • Serum Complement Activation by C4BP-IgM Fusion Protein Can Restore Susceptibility to Antibiotics in Neisseria gonorrhoeae
  • 2021
  • Ingår i: Frontiers in Immunology. - : Frontiers Media S.A.. - 1664-3224. ; 12
  • Tidskriftsartikel (refereegranskat)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.
  •  
4.
  • Bettoni, Serena, et al. (författare)
  • Serum complement activation by C4BP-IgM fusion protein can restore susceptibility to antibiotics in Neisseria gonorrhoeae
  • 2022
  • Ingår i: Molecular Immunology. - : Elsevier. - 0161-5890 .- 1872-9142. ; 141, s. 215-215
  • Tidskriftsartikel (övrigt vetenskapligt/konstnärligt)abstract
    • Background: The sexually transmitted infection gonorrhea is a common health problem worldwide causing critical reproductive sequelae such as infertility. An effective vaccine remains elusive and antibiotics used in clinics are becoming ineffective because of the rapid spread of resistance among Neisseria gonorrhoeae, the causative organism of gonorrhea. We previously created a human fusion protein called C4BP-IgM to mark and eliminate bacteria by activating host complement. C4BP-IgM links the two N-terminal domains of C4BP, which bind to gonococci, with the Fc domain of IgM to increase complement activation on and kill bacteria. We documented that C4BP-IgM enhances bactericidal activity of serum against clinical C4BP-binding gonococcal isolates from patients, and markedly attenuated the duration and burden of gonococcal infection in a mouse vaginal colonizationmodel 1. Here, we explore the activity of C4BP-IgM as an adjuvant to several antibiotics (spectinomycin, azithromycin, cefixime, ceftriaxone and ciprofloxacin) currently or previously used to treat gonorrhea.Materials and methods: Cooperative bactericidal activity between C4BP-IgM, complement and antibiotics was evaluated by monitoring survival and membrane alterations of a laboratory isolate and two clinical azithromycin-resistant gonococcal strains, which also resisted killing by normal human serum. Effect of complement and C4BP-IgM on uptake and intracellular activity of selected antibiotics was also assessed.Results: We found that human serum, as source of complement components, reduced MIC values of antibiotics against N. gonorrhoeae. Addition of C4BP-IgM at concentrations which only partially reduced survival, induced complete killing of bacteria when both serum and antibiotics were present. Bactericidal cooperation between complement and antimicrobials was revealed to be triggered by membrane damage induced by C4BP-IgM complement activation. Formation of membrane attack complex pores on bacteria facilitated uptake of antimicrobials, which in turn enhanced their intracellular concentration and activity. Remarkably, C4BP-IgM restored susceptibility to azithromycin of two azithromycin-resistant clinical gonococcal strains that overexpressed the MtrC-MtrD-MtrE efflux pump.Conclusion: We provide proof-of-principle for the use of C4BP-IgM fusion protein as an adjuvant to antibiotics, which could be re-purposed for clinical use pending the development of effective new treatments.
  •  
5.
  • Kulak, Klaudia, et al. (författare)
  • Intracellular C3 protects β-cells from IL-1β-driven cytotoxicity via interaction with Fyn-related kinase
  • 2024
  • Ingår i: Proceedings of the National Academy of Sciences of the United States of America. - 1091-6490. ; 121:8
  • Tidskriftsartikel (refereegranskat)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-1β is a proinflammatory cytokine which detrimentally affects β-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-1β treatment. In the current investigation, using primary human and rodent material and CRISPR/Cas9 gene-edited β-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-1β-induced β-cell death, that is attributed to the cytosolic fraction of C3. Further investigation revealed that intracellular C3 alleviates IL-1β-induced β-cell death, by interaction with and inhibition of Fyn-related kinase (FRK), which is involved in the response of β-cells to cytokines. Furthermore, these data were supported by increased β-cell death in vivo in a β-cell-specific C3 knockout mouse. Our data indicate that a functional, cytoprotective association exists between FRK and cytosolic C3.
  •  
6.
  • Laabei, Maisem, et al. (författare)
  • Antibacterial Fusion Proteins Enhance Moraxella catarrhalis Killing
  • 2020
  • Ingår i: Frontiers in Immunology. - : Frontiers Media SA. - 1664-3224. ; 11
  • Tidskriftsartikel (refereegranskat)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.
  •  
Skapa referenser, mejla, bekava och länka
  • Resultat 1-6 av 6

Kungliga biblioteket hanterar dina personuppgifter i enlighet med EU:s dataskyddsförordning (2018), GDPR. Läs mer om hur det funkar här.
Så här hanterar KB dina uppgifter vid användning av denna tjänst.

 
pil uppåt Stäng

Kopiera och spara länken för att återkomma till aktuell vy