Natural latex serum: characterization and biocompatibility assessment using Galleria mellonella as an alternative in vivo model

Author(s):  
Giovana Sant’Ana Pegorin Brasil ◽  
Patrícia Pimentel de Barros ◽  
Matheus Carlos Romeiro Miranda ◽  
Natan Roberto de Barros ◽  
Juliana Campos Junqueira ◽  
...  
2021 ◽  
Author(s):  
Jess Vergis ◽  
S V S Malik ◽  
Richa Pathak ◽  
Manesh Kumar ◽  
Nitin V Kurkure ◽  
...  

Abstract High throughput in vivo laboratory models is need for screening and identification of effective therapeutic agents to overcome microbial drug-resistance. This study was undertaken to evaluate in vivo antimicrobial efficacy of short-chain antimicrobial peptide- Cecropin A (1–7)-Melittin (CAMA) against three multi- drug resistant enteroaggregative Escherichia coli (MDR-EAEC) field isolates in a Galleria mellonella larval model. The minimum inhibitory concentration (MIC; 2.0 mg/L) and minimum bactericidal concentration (MBC; 4.0 mg/L) of CAMA were determined by microdilution assay. CAMA was found to be stable at high temperatures, physiological concentration of cationic salts and proteases; safe with sheep erythrocytes, secondary cell lines and commensal lactobacilli at lower MICs; and exhibited membrane permeabilisation. In vitro time-kill assay revealed concentration- and time-dependent clearance of MDR-EAEC in CAMA-treated groups at 30 min. CAMA- treated G. mellonella larvae exhibited an increased survival rate, reduced MDR-EAEC counts, immunomodulatory effect and proved non-toxic which concurred with histopathological findings. CAMA exhibited either an equal or better efficacy than the tested antibiotic control, meropenem. This study highlights the possibility of G. mellonella larvae as an excellent in vivo model for investigating the host-pathogen interaction, including the efficacy of antimicrobials against MDR-EAEC strains.


Antibiotics ◽  
2021 ◽  
Vol 10 (1) ◽  
pp. 56
Author(s):  
Dalila Mil-Homens ◽  
Maria Martins ◽  
José Barbosa ◽  
Gabriel Serafim ◽  
Maria J. Sarmento ◽  
...  

Klebsiella pneumoniae, one of the most common pathogens found in hospital-acquired infections, is often resistant to multiple antibiotics. In fact, multidrug-resistant (MDR) K. pneumoniae producing KPC or OXA-48-like carbapenemases are recognized as a serious global health threat. In this sense, we evaluated the virulence of K. pneumoniae KPC(+) or OXA-48(+) aiming at potential antimicrobial therapeutics. K. pneumoniae carbapenemase (KPC) and the expanded-spectrum oxacillinase OXA-48 isolates were obtained from patients treated in medical care units in Lisbon, Portugal. The virulence potential of the K. pneumonia clinical isolates was tested using the Galleria mellonella model. For that, G. mellonella larvae were inoculated using patients KPC(+) and OXA-48(+) isolates. Using this in vivo model, the KPC(+) K. pneumoniae isolates showed to be, on average, more virulent than OXA-48(+). Virulence was found attenuated when a low bacterial inoculum (one magnitude lower) was tested. In addition, we also report the use of a synthetic polycationic oligomer (L-OEI-h) as a potential antimicrobial agent to fight infectious diseases caused by MDR bacteria. L-OEI-h has a broad-spectrum antibacterial activity and exerts a significantly bactericidal activity within the first 5-30 min treatment, causing lysis of the cytoplasmic membrane. Importantly, the polycationic oligomer showed low toxicity against in vitro models and no visible cytotoxicity (measured by survival and health index) was noted on the in vivo model (G. mellonella), thus L-OEI-h is foreseen as a promising polymer therapeutic for the treatment of MDR K. pneumoniae infections.


2019 ◽  
Vol 6 (Supplement_2) ◽  
pp. S896-S897
Author(s):  
Faris S Alnezary ◽  
Tasnuva Rashid ◽  
Khurshida Begum ◽  
Travis J Carlson ◽  
Anne J Gonzales-Luna ◽  
...  

Abstract Background Antimicrobials disrupt the gut microbiota by reducing gut microbiome diversity and quantity. Galleria mellonella provides an invertebrate model that is inexpensive, easy to maintain, and does not require specialized equipment. This study investigated the feasibility of using G. mellonella as an in vivo model to evaluate the effect of different antimicrobials on gut microbiota. Methods To determine baseline gut microbiota composition, the gut contents of G. mellonella were extracted and genomic DNA underwent shotgun meta-genomic sequencing. To determine the effect of infection and antibiotic use, 30 larvae were injected (left proleg) with ~1 × 105 colony-forming unit (cfu) of methicillin-resistant Staphylococcus aureus (MRSA) and were randomized 1:1:1 to treatment with vancomycin (20 mg/kg) or a natural antimicrobial (Nigella sativa seed oil, 70 mg/kg; NS oil), or a combination. The larvae were kept at 37°C post-infection and monitored daily for 72 hours for activity, extent of cocoon formation/growth, melanization, and survival. Two larvae from each group were randomly selected and homogenized with PBS as controls. After 24 hours of incubation, gut contents were extracted and plated for MRSA and Enterococcus cfu counts. Results Metagenomics analysis showed the gut microbiota composition of G. mellonella larvae was dominated by a subset of closely-related Enterococcus species. After 24 hours of exposure, mean Enterococcus counts were 4 × 103 cfu in the vancomycin arm and 6.2 × 104 cfu in the NS oil arm. Mean MRSA counts were 3.3 × 105 cfu in vancomycin arm and 1.5 × 104 cfu in NS oil arm. The combination of vancomycin and NS oil had higher Enterococcus counts than the vancomycin alone arm (6.3 × 104 cfu vs. 4 × 103 cfu, respectively), suggesting that NS oil may have a role in protecting the gut microbiota. Conclusion This study provides preliminary evidence to support the potential use of G. mellonella to assess the in vivo effect of a natural and synthetic antimicrobial on the gut microbiota. Disclosures All authors: No reported disclosures.


2017 ◽  
Vol 4 (suppl_1) ◽  
pp. S379-S379
Author(s):  
Farzad Moussavi ◽  
Sarath Nath ◽  
Daniel Abraham ◽  
David Landman ◽  
John Quale

Abstract Background Options for treatment of infections due to KPC-producing K. pneumoniae are limited, and combination therapy is often recommended. In this report, the in vitro and in vivo activity of potential therapeutic agents and combinations was assessed against four KPC-producing K. pneumoniae isolates. Methods Using clinically-relevant concentrations, time-kill experiments and the Galleria mellonella model of infection were used to examine the activity of polymyxin B, ceftazidime-avibactam, meropenem, rifampin, and amikacin alone and in combination. Four isolates of KPC-producing K. pneumoniae were studied, including two isolates that were resistant to polymyxin B and had ceftazidime-avibactam MICs of 8 µg/mL. The other two K. pneumoniae isolates were susceptible to polymyxin B and had lower MICs of ceftazidime-avibactam. Results Two isolates that were resistant to polymyxin B and with ceftazidime-avibactam MICs of 8 µg/mL were also resistant to amikacin and meropenem. When ceftazidime-avibactam was combined with either amikacin or meropenem, synergy was observed in vitro, and these combinations were associated with improved survival with the in vivo model. The other two K. pneumoniae isolates were susceptible to polymyxin B and had lower MICs of ceftazidime-avibactam. At concentrations four times the MIC, ceftazidime-avibactam had bactericidal activity in vitro; at one fourth the MIC, synergy was observed when combined with meropenem. Improved survival rates were observed with therapy with ceftazidime-avibactam, particularly when combined with a second agent for one isolate. In the in vivo model, polymyxin B with or without rifampin or meropenem, was ineffective against polymyxin B resistant strains. Conclusion Pending clinical studies, combining ceftazidime-avibactam with another agent (e.g., a carbapenem) should be encouraged when treating serious infections due to these pathogens, especially for isolates with ceftazidime-avibactam MICs near the susceptibility breakpoint. Disclosures All authors: No reported disclosures.


Antibiotics ◽  
2021 ◽  
Vol 10 (12) ◽  
pp. 1545
Author(s):  
Magdalena Piatek ◽  
Gerard Sheehan ◽  
Kevin Kavanagh

Larvae of the greater wax moth, Galleria mellonella, are a convenient in vivo model for assessing the activity and toxicity of antimicrobial agents and for studying the immune response to pathogens and provide results similar to those from mammals. G. mellonella larvae are now widely used in academia and industry and their use can assist in the identification and evaluation of novel antimicrobial agents. Galleria larvae are inexpensive to purchase and house, easy to inoculate, generate results within 24–48 h and their use is not restricted by legal or ethical considerations. This review will highlight how Galleria larvae can be used to assess the efficacy of novel antimicrobial therapies (photodynamic therapy, phage therapy, metal-based drugs, triazole-amino acid hybrids) and for determining the in vivo toxicity of compounds (e.g., food preservatives, ionic liquids) and/or solvents (polysorbate 80). In addition, the disease development processes are associated with a variety of pathogens (e.g., Staphylococcus aureus, Listeria monocytogenes, Aspergillus fumigatus, Madurella mycotomatis) in mammals are also present in Galleria larvae thus providing a simple in vivo model for characterising disease progression. The use of Galleria larvae offers many advantages and can lead to an acceleration in the development of novel antimicrobials and may be a prerequisite to mammalian testing.


Chemosphere ◽  
2015 ◽  
Vol 139 ◽  
pp. 197-201 ◽  
Author(s):  
Julianne Megaw ◽  
Thomas P. Thompson ◽  
Ryan A. Lafferty ◽  
Brendan F. Gilmore

Sign in / Sign up

Export Citation Format

Share Document