Multifunctional lipid-based nanocarriers with antibacterial and anti‐inflammatory activities for treating MRSA bacteremia in mice

Abstract Background Bacteremia-induced sepsis is a leading cause of mortality in intensive care units. To control a bacterial infection, an immune response is required, but this response might contribute to organ failure. Kidneys are one of the main organs affected by bacteremia. Combination therapi...

Full description

Bibliographic Details
Main Authors: Chia-Chih Liao, Huang-Ping Yu, Shih-Chun Yang, Ahmed Alalaiwe, You-Shan Dai, Fu-Chao Liu, Jia-You Fang
Format: Article
Language:English
Published: BMC 2021-02-01
Series:Journal of Nanobiotechnology
Subjects:
Online Access:https://doi.org/10.1186/s12951-021-00789-5
_version_ 1798034122940088320
author Chia-Chih Liao
Huang-Ping Yu
Shih-Chun Yang
Ahmed Alalaiwe
You-Shan Dai
Fu-Chao Liu
Jia-You Fang
author_facet Chia-Chih Liao
Huang-Ping Yu
Shih-Chun Yang
Ahmed Alalaiwe
You-Shan Dai
Fu-Chao Liu
Jia-You Fang
author_sort Chia-Chih Liao
collection DOAJ
description Abstract Background Bacteremia-induced sepsis is a leading cause of mortality in intensive care units. To control a bacterial infection, an immune response is required, but this response might contribute to organ failure. Kidneys are one of the main organs affected by bacteremia. Combination therapies with antibacterial and anti-inflammatory effects may be beneficial in treating bacteremia. This study aimed to develop nanostructured lipid carriers (NLCs) loaded with ciprofloxacin and rolipram that exert a combination of anti-methicillin-resistant Staphylococcus aureus (MRSA) and anti-inflammatory effects. Retinol was incorporated into the nanoparticles to transport retinol-binding protein 4 (RBP4) to the kidneys, which abundantly express RBP receptors. The NLCs were fabricated by high-shear homogenization and sonication, and neutrophils were used as a model to assess their anti-inflammatory effects. Mice were injected with MRSA to establish a model of bacteremia with organ injury. Results The mean nanoparticle size and zeta potential of the NLCs were 171 nm and − 39 mV, respectively. Ciprofloxacin (0.05%, w/v) and rolipram (0.02%) achieved encapsulation percentages of 88% and 96%, respectively, in the nanosystems. The minimum bactericidal concentration of free ciprofloxacin against MRSA increased from 1.95 to 15.63 µg/ml when combined with rolipram, indicating a possible drug-drug interaction that reduced the antibacterial effect. Nanoparticle inclusion promoted the anti-MRSA activity of ciprofloxacin according to time-kill curves. The NLCs were found to be largely internalized into neutrophils and exhibited superior superoxide anion inhibition than free drugs. Retinol incorporation into the nanocarriers facilitated their efficient targeting to the kidneys. The NLCs significantly mitigated MRSA burden and elastase distribution in the organs of MRSA-infected animals, and the greatest inhibition was observed in the kidneys. Bacterial clearance and neutrophil infiltration suppression attenuated the bacteremia-induced cytokine overexpression, leading to an improvement in the survival rate from 22% to 67%. Conclusions The dual role of our NLCs endowed them with greater efficacy in treating MRSA bacteremia than that of free drugs.
first_indexed 2024-04-11T20:39:55Z
format Article
id doaj.art-7c18706c95e346a29fb1d3241bd049bb
institution Directory Open Access Journal
issn 1477-3155
language English
last_indexed 2024-04-11T20:39:55Z
publishDate 2021-02-01
publisher BMC
record_format Article
series Journal of Nanobiotechnology
spelling doaj.art-7c18706c95e346a29fb1d3241bd049bb2022-12-22T04:04:15ZengBMCJournal of Nanobiotechnology1477-31552021-02-0119111810.1186/s12951-021-00789-5Multifunctional lipid-based nanocarriers with antibacterial and anti‐inflammatory activities for treating MRSA bacteremia in miceChia-Chih Liao0Huang-Ping Yu1Shih-Chun Yang2Ahmed Alalaiwe3You-Shan Dai4Fu-Chao Liu5Jia-You Fang6Department of Anesthesiology, Chang Gung Memorial HospitalDepartment of Anesthesiology, Chang Gung Memorial HospitalDepartment of Cosmetic Science, Providence UniversityDepartment of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz UniversityPharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung UniversityDepartment of Anesthesiology, Chang Gung Memorial HospitalDepartment of Anesthesiology, Chang Gung Memorial HospitalAbstract Background Bacteremia-induced sepsis is a leading cause of mortality in intensive care units. To control a bacterial infection, an immune response is required, but this response might contribute to organ failure. Kidneys are one of the main organs affected by bacteremia. Combination therapies with antibacterial and anti-inflammatory effects may be beneficial in treating bacteremia. This study aimed to develop nanostructured lipid carriers (NLCs) loaded with ciprofloxacin and rolipram that exert a combination of anti-methicillin-resistant Staphylococcus aureus (MRSA) and anti-inflammatory effects. Retinol was incorporated into the nanoparticles to transport retinol-binding protein 4 (RBP4) to the kidneys, which abundantly express RBP receptors. The NLCs were fabricated by high-shear homogenization and sonication, and neutrophils were used as a model to assess their anti-inflammatory effects. Mice were injected with MRSA to establish a model of bacteremia with organ injury. Results The mean nanoparticle size and zeta potential of the NLCs were 171 nm and − 39 mV, respectively. Ciprofloxacin (0.05%, w/v) and rolipram (0.02%) achieved encapsulation percentages of 88% and 96%, respectively, in the nanosystems. The minimum bactericidal concentration of free ciprofloxacin against MRSA increased from 1.95 to 15.63 µg/ml when combined with rolipram, indicating a possible drug-drug interaction that reduced the antibacterial effect. Nanoparticle inclusion promoted the anti-MRSA activity of ciprofloxacin according to time-kill curves. The NLCs were found to be largely internalized into neutrophils and exhibited superior superoxide anion inhibition than free drugs. Retinol incorporation into the nanocarriers facilitated their efficient targeting to the kidneys. The NLCs significantly mitigated MRSA burden and elastase distribution in the organs of MRSA-infected animals, and the greatest inhibition was observed in the kidneys. Bacterial clearance and neutrophil infiltration suppression attenuated the bacteremia-induced cytokine overexpression, leading to an improvement in the survival rate from 22% to 67%. Conclusions The dual role of our NLCs endowed them with greater efficacy in treating MRSA bacteremia than that of free drugs.https://doi.org/10.1186/s12951-021-00789-5Nanostructured lipid carriersCiprofloxacinRolipramBacteremiaMethicillin‐resistant Staphylococcus aureusSepsis
spellingShingle Chia-Chih Liao
Huang-Ping Yu
Shih-Chun Yang
Ahmed Alalaiwe
You-Shan Dai
Fu-Chao Liu
Jia-You Fang
Multifunctional lipid-based nanocarriers with antibacterial and anti‐inflammatory activities for treating MRSA bacteremia in mice
Journal of Nanobiotechnology
Nanostructured lipid carriers
Ciprofloxacin
Rolipram
Bacteremia
Methicillin‐resistant Staphylococcus aureus
Sepsis
title Multifunctional lipid-based nanocarriers with antibacterial and anti‐inflammatory activities for treating MRSA bacteremia in mice
title_full Multifunctional lipid-based nanocarriers with antibacterial and anti‐inflammatory activities for treating MRSA bacteremia in mice
title_fullStr Multifunctional lipid-based nanocarriers with antibacterial and anti‐inflammatory activities for treating MRSA bacteremia in mice
title_full_unstemmed Multifunctional lipid-based nanocarriers with antibacterial and anti‐inflammatory activities for treating MRSA bacteremia in mice
title_short Multifunctional lipid-based nanocarriers with antibacterial and anti‐inflammatory activities for treating MRSA bacteremia in mice
title_sort multifunctional lipid based nanocarriers with antibacterial and anti inflammatory activities for treating mrsa bacteremia in mice
topic Nanostructured lipid carriers
Ciprofloxacin
Rolipram
Bacteremia
Methicillin‐resistant Staphylococcus aureus
Sepsis
url https://doi.org/10.1186/s12951-021-00789-5
work_keys_str_mv AT chiachihliao multifunctionallipidbasednanocarrierswithantibacterialandantiinflammatoryactivitiesfortreatingmrsabacteremiainmice
AT huangpingyu multifunctionallipidbasednanocarrierswithantibacterialandantiinflammatoryactivitiesfortreatingmrsabacteremiainmice
AT shihchunyang multifunctionallipidbasednanocarrierswithantibacterialandantiinflammatoryactivitiesfortreatingmrsabacteremiainmice
AT ahmedalalaiwe multifunctionallipidbasednanocarrierswithantibacterialandantiinflammatoryactivitiesfortreatingmrsabacteremiainmice
AT youshandai multifunctionallipidbasednanocarrierswithantibacterialandantiinflammatoryactivitiesfortreatingmrsabacteremiainmice
AT fuchaoliu multifunctionallipidbasednanocarrierswithantibacterialandantiinflammatoryactivitiesfortreatingmrsabacteremiainmice
AT jiayoufang multifunctionallipidbasednanocarrierswithantibacterialandantiinflammatoryactivitiesfortreatingmrsabacteremiainmice