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.
Nat Commun ; 15(1): 4740, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38834545

ABSTRACT

Mitophagy is critical for mitochondrial quality control and function to clear damaged mitochondria. Here, we found that Burkholderia pseudomallei maneuvered host mitophagy for its intracellular survival through the type III secretion system needle tip protein BipD. We identified BipD, interacting with BTB-containing proteins KLHL9 and KLHL13 by binding to the Back and Kelch domains, recruited NEDD8 family RING E3 ligase CUL3 in response to B. pseudomallei infection. Although evidently not involved in regulation of infectious diseases, KLHL9/KLHL13/CUL3 E3 ligase complex was essential for BipD-dependent ubiquitination of mitochondria in mouse macrophages. Mechanistically, we discovered the inner mitochondrial membrane IMMT via host ubiquitome profiling as a substrate of KLHL9/KLHL13/CUL3 complex. Notably, K63-linked ubiquitination of IMMT K211 was required for initiating host mitophagy, thereby reducing mitochondrial ROS production. Here, we show a unique mechanism used by bacterial pathogens that hijacks host mitophagy for their survival.


Subject(s)
Bacterial Proteins , Burkholderia pseudomallei , Macrophages , Mitochondria , Mitophagy , Burkholderia pseudomallei/metabolism , Burkholderia pseudomallei/pathogenicity , Burkholderia pseudomallei/physiology , Burkholderia pseudomallei/genetics , Animals , Mice , Mitochondria/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Humans , Macrophages/microbiology , Macrophages/metabolism , Ubiquitination , Melioidosis/microbiology , Melioidosis/metabolism , Host-Pathogen Interactions , Reactive Oxygen Species/metabolism , Type III Secretion Systems/metabolism , Type III Secretion Systems/genetics , Mice, Inbred C57BL , Mitochondrial Membranes/metabolism , HEK293 Cells , RAW 264.7 Cells
2.
Int J Biol Macromol ; 258(Pt 2): 128922, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38141699

ABSTRACT

Burkholderia pseudomallei, an intracellular pathogen, is responsible for melioidosis, a zoonotic disease. Its pathogenesis involves several virulence factors, among which lipopolysaccharide (LPS) plays a crucial role. Our research reveals that the O antigen present within the LPS significantly regulates the host immune response. In a previous study, we obtained a B. pseudomallei mutant strain ΔwbiI. Here, the purification of LPS from ΔwbiI and a gas chromatography-mass spectrometry (GC-MS) analysis were conducted. The results confirmed the absence of specific sugar 6-deoxy-Talp, which is a typical component of the O antigen in the wild type B. pseudomallei. Our findings underscore the potent impact the O antigen exerts on the virulence of B. pseudomallei. The ΔwbiI strain displayed significantly increased invasiveness and cytotoxicity in vitro. This enhanced cytotoxicity seems to be related to the exposure of lipid A and an increased cell membrane hydrophobicity resulting from the deletion of the O antigen. Additionally, in mouse models, the ΔwbiI strain resulted in a heightened host lethality and an excessive inflammatory response in mice. These findings indicate that the O-antigenic polysaccharide moiety of B. pseudomallei plays a role in its pathogenicity in vitro and in vivo.


Subject(s)
Burkholderia pseudomallei , Mice , Animals , O Antigens/genetics , Lipopolysaccharides , Virulence , Mutation
3.
Int J Biol Macromol ; 225: 795-808, 2023 Jan 15.
Article in English | MEDLINE | ID: mdl-36402383

ABSTRACT

O antigen is the major component of lipopolysaccharide LPS. The chemical structure of the O antigen determines the LPS serospecificity of the bacteria, and the diversity of O antigen is the basis for serotyping Burkholderia pseudomallei. In this study, structural elucidation of type B O antigen obtained from a clinical B. pseudomallei strain was conducted, and the effects of different types of LPS on macrophage differentiation were investigated. The O antigen was found to be composed of repeating units of [→4)-α-L-Rhap(1 â†’ 4)-α-L-Rhap(1→2)-α-L-Rhap(1 â†’ 2)-α-L-Rhap(1 â†’ 3)-α-L-Rhap(1 â†’ 3)-α-L-Rhap(1 â†’ 4)-α-L-Rhap(1 â†’ 6)-α-D-Galp(1→]n, where some of the →4)-α-L-Rhap(1 â†’ units were substituted at O-3 by ß-D-Xylp(1 â†’ residues, and minor →3)-α-L-Rhap(1 â†’ units were substituted at O-2 by ß-D-Xylp(1 â†’ residues. Meahwhile, the →6)-α-D-Galp(1 â†’ units were substituted at O-3 by α-D-Galp(1 â†’ residues. Furthermore, both type A and type B O antigens of B. pseudomallei could polarize macrophages toward the M1 phenotype, but the core oligosaccharides had no such activity. Therefore, we deduced that this polarization relies on the O antigen of LPS and might be related to the ability of B. pseudomallei to survive and replicate within macrophages. Thus, the characterization of different types of O antigen structural motifs is essential for further clarifying the persistence/survival mechanisms and inflammatory potential of B. pseudomallei.


Subject(s)
Burkholderia pseudomallei , O Antigens , O Antigens/chemistry , Lipopolysaccharides/chemistry , Antigens, Bacterial , Oligosaccharides/chemistry
4.
Pathogens ; 13(1)2023 Dec 31.
Article in English | MEDLINE | ID: mdl-38251350

ABSTRACT

Melioidosis, a severe tropical illness caused by Burkholderia pseudomallei, poses significant treatment challenges due to limited therapeutic options and the absence of effective vaccines. The pathogen's intrinsic resistance to numerous antibiotics and propensity to induce sepsis during acute infections further complicate management strategies. Thus, exploring alternative methods for prevention and treatment is crucial. Monoclonal antibodies (mAbs) have emerged as a promising strategy for the prevention and treatment of infectious diseases. This study focused on generating three mAbs (13F1, 14G11, and 15D9) targeting hemolysin-coregulated protein 1 (Hcp1), a protein involved in the type VI secretion system cluster 1 (T6SS1) of B. pseudomallei. Notably, pretreatment with 13F1 mAb significantly reduced the intracellular survival of B. pseudomallei and inhibited the formation of macrophage-derived multinucleated giant cells (MNGCs). This protective effect was also observed in vivo. We identified a sequence of amino acids (Asp95-Leu114) within Hcp1 as the likely binding site for 13F1 mAb. In summary, our findings reveal that 13F1 mAb counteracts infection by targeting Hcp1, offering potential new targets and insights for melioidosis prevention.

SELECTION OF CITATIONS
SEARCH DETAIL
...