Your browser doesn't support javascript.
loading
Macrophage targeted iron oxide nanodecoys augment innate immunological and drug killings for more effective Mycobacterium Tuberculosis clearance.
Shen, Ling; Liao, Kangsheng; Yang, Enzhuo; Yang, Fen; Lin, Wensen; Wang, Jiajun; Fan, Shuhao; Huang, Xueqin; Chen, Lingming; Shen, Hongbo; Jin, Hua; Ruan, Yongdui; Liu, Xing; Zeng, Gucheng; Xu, Jun-Fa; Pi, Jiang.
Afiliação
  • Shen L; Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, USA. lshen@uic.edu.
  • Liao K; Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.
  • Yang E; The Marine Biomedical Research Institute of Guangdong Zhanjiang, The Marine Biomedical Research Institute of Guangdong Medical University, ZhanJiang, Guangdong, China.
  • Yang F; Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China.
  • Lin W; Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, USA.
  • Wang J; Clinic and Research Center of Tuberculosis, Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
  • Fan S; Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.
  • Huang X; The Marine Biomedical Research Institute of Guangdong Zhanjiang, The Marine Biomedical Research Institute of Guangdong Medical University, ZhanJiang, Guangdong, China.
  • Chen L; Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China.
  • Shen H; Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.
  • Jin H; Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China.
  • Ruan Y; Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.
  • Liu X; Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China.
  • Zeng G; Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.
  • Xu JF; Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China.
  • Pi J; Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.
J Nanobiotechnology ; 21(1): 369, 2023 Oct 10.
Article em En | MEDLINE | ID: mdl-37817142
ABSTRACT
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) infection, is still one of the top killers worldwide among infectious diseases. The escape of Mtb from immunological clearance and the low targeting effects of anti-TB drugs remain the substantial challenges for TB control. Iron is particularly required for Mtb growth but also toxic for Mtb in high dosages, which makes iron an ideal toxic decoy for the 'iron-tropic' Mtb. Here, a macrophage-targeted iron oxide nanoparticles (IONPs)-derived IONPs-PAA-PEG-MAN nanodecoy is designed to augment innate immunological and drug killings against intracellular Mtb. IONPs-PAA-PEG-MAN nanodecoy exhibits preferential uptake in macrophages to significantly increase drug uptake with sustained high drug contents in host cells. Moreover, it can serve as a specific nanodecoy for the 'iron-tropic' Mtb to realize the localization of Mtb contained phagosomes surrounding the drug encapsulated nanodecoys and co-localization of Mtb with the drug encapsulated nanodecoys in lysosomes, where the incorporated rifampicin (Rif) can be readily released under acidic lysosomal condition for enhanced Mtb killing. This drug encapsulated nanodecoy can also polarize Mtb infected macrophages into anti-mycobacterial M1 phenotype and enhance M1 macrophage associated pro-inflammatory cytokine (TNF-α) production to trigger innate immunological responses against Mtb. Collectively, Rif@IONPs-PAA-PEG-MAN nanodecoy can synergistically enhance the killing efficiency of intracellular Mtb in in vitro macrophages and ex vivo monocyte-derived macrophages, and also significantly reduce the mycobacterial burdens in the lung of infected mice with alleviated pathology. These results indicate that Rif@IONPs-PAA-PEG-MAN nanodecoy may have a potential for the development of more effective therapeutic strategy against TB by manipulating augmented innate immunity and drug killings.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Tuberculose / Mycobacterium tuberculosis Idioma: En Revista: J Nanobiotechnology Ano de publicação: 2023 Tipo de documento: Article País de afiliação: Estados Unidos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Tuberculose / Mycobacterium tuberculosis Idioma: En Revista: J Nanobiotechnology Ano de publicação: 2023 Tipo de documento: Article País de afiliação: Estados Unidos