Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.263
Filter
1.
PLoS Pathog ; 20(8): e1012437, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39102432

ABSTRACT

The ability of Staphylococcus aureus (S. aureus) to survive within macrophages is a critical strategy for immune evasion, contributing to the pathogenesis and progression of osteomyelitis. However, the underlying mechanisms remain poorly characterized. This study discovered that inhibiting the MEK1/2 pathway reduced bacterial load and mitigated bone destruction in a mouse model of S. aureus osteomyelitis. Histological staining revealed increased phosphorylated MEK1/2 levels in bone marrow macrophages surrounding abscess in the mouse model of S. aureus osteomyelitis. Activation of MEK1/2 pathway and its roles in impairing macrophage bactericidal function were confirmed in primary mouse bone marrow-derived macrophages (BMDMs). Transcriptome analysis and in vitro experiments demonstrated that S. aureus activates the MEK1/2 pathway through EGFR signaling. Moreover, we found that excessive activation of EGFR-MEK1/2 cascade downregulates mitochondrial reactive oxygen species (mtROS) levels by suppressing Chek2 expression, thereby impairing macrophage bactericidal function. Furthermore, pharmacological inhibition of EGFR signaling prevented upregulation of phosphorylated MEK1/2 and restored Chek2 expression in macrophages, significantly enhancing S. aureus clearance and improving bone microstructure in vivo. These findings highlight the critical role of the EGFR-MEK1/2 cascade in host immune defense against S. aureus, suggesting that S. aureus may reduce mtROS levels by overactivating the EGFR-MEK1/2 cascade, thereby suppressing macrophage bactericidal function. Therefore, combining EGFR-MEK1/2 pathway blockade with antibiotics could represent an effective therapeutic approach for the treatment of S. aureus osteomyelitis.


Subject(s)
ErbB Receptors , MAP Kinase Kinase 1 , Macrophages , Osteomyelitis , Staphylococcal Infections , Staphylococcus aureus , Animals , Osteomyelitis/microbiology , Osteomyelitis/immunology , Osteomyelitis/metabolism , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Mice , Staphylococcus aureus/immunology , ErbB Receptors/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , MAP Kinase Signaling System/physiology , Mice, Inbred C57BL , Disease Models, Animal , Signal Transduction
2.
FASEB J ; 38(16): e23881, 2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39166718

ABSTRACT

During infection, the host employs nutritional immunity to restrict access to iron. Staphylococcus lugdunensis has been recognized for its ability to utilize host-derived heme to overcome iron restriction. However, the mechanism behind this process involves the release of hemoglobin from erythrocytes, and the hemolytic factors of S. lugdunensis remain poorly understood. S. lugdunensis encodes four phenol-soluble modulins (PSMs), short peptides with hemolytic activity. The peptides SLUSH A, SLUSH B, and SLUSH C are ß-type PSMs, and OrfX is an α-type PSM. Our study shows the SLUSH locus to be essential for the hemolytic phenotype of S. lugdunensis. All four peptides individually exhibited hemolytic activity against human and sheep erythrocytes, but synergism with sphingomyelinase was observed exclusively against sheep erythrocytes. Furthermore, our findings demonstrate that SLUSH is crucial for allowing the utilization of erythrocytes as the sole source of nutritional iron and confirm the transcriptional regulation of SLUSH by Agr. Additionally, our study reveals that SLUSH peptides stimulate the human immune system. Our analysis identifies SLUSH as a pivotal hemolytic factor of S. lugdunensis and demonstrates its concerted action with heme acquisition systems to overcome iron limitation in the presence of host erythrocytes.


Subject(s)
Erythrocytes , Hemolysis , Iron , Staphylococcus lugdunensis , Erythrocytes/metabolism , Staphylococcus lugdunensis/metabolism , Humans , Iron/metabolism , Animals , Sheep , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Peptides/metabolism , Peptides/chemistry , Staphylococcal Infections/microbiology , Staphylococcal Infections/metabolism , Bacterial Toxins
3.
Nat Commun ; 15(1): 5583, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961085

ABSTRACT

The function of many bacterial processes depends on the formation of functional membrane microdomains (FMMs), which resemble the lipid rafts of eukaryotic cells. However, the mechanism and the biological function of these membrane microdomains remain unclear. Here, we show that FMMs in the pathogen methicillin-resistant Staphylococcus aureus (MRSA) are dedicated to confining and stabilizing proteins unfolded due to cellular stress. The FMM scaffold protein flotillin forms a clamp-shaped oligomer that holds unfolded proteins, stabilizing them and favoring their correct folding. This process does not impose a direct energy cost on the cell and is crucial to survival of ATP-depleted bacteria, and thus to pathogenesis. Consequently, FMM disassembling causes the accumulation of unfolded proteins, which compromise MRSA viability during infection and cause penicillin re-sensitization due to PBP2a unfolding. Thus, our results indicate that FMMs mediate ATP-independent stabilization of unfolded proteins, which is essential for bacterial viability during infection.


Subject(s)
Bacterial Proteins , Membrane Microdomains , Membrane Proteins , Methicillin-Resistant Staphylococcus aureus , Membrane Proteins/metabolism , Membrane Microdomains/metabolism , Methicillin-Resistant Staphylococcus aureus/metabolism , Bacterial Proteins/metabolism , Protein Unfolding , Adenosine Triphosphate/metabolism , Penicillin-Binding Proteins/metabolism , Penicillin-Binding Proteins/genetics , Penicillin-Binding Proteins/chemistry , Humans , Protein Stability , Staphylococcal Infections/microbiology , Staphylococcal Infections/metabolism , Animals , Mice
4.
FASEB J ; 38(14): e23801, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39018106

ABSTRACT

Intracellular pathogens including Staphylococcus aureus contribute to the non-healing phenotype of chronic wounds. Lactobacilli, well known as beneficial bacteria, are also reported to modulate the immune system, yet their role in cutaneous immunity remains largely unknown. We explored the therapeutic potential of bacteria-free postbiotics, bioactive lysates of lactobacilli, to reduce intracellular S. aureus colonization and promote healing. Fourteen postbiotics derived from various lactobacilli species were screened, and Latilactobacillus curvatus BGMK2-41 was selected for further analysis based on the most efficient ability to reduce intracellular infection by S. aureus diabetic foot ulcer clinical isolate and S. aureus USA300. Treatment of both infected keratinocytes in vitro and infected human skin ex vivo with BGMK2-41 postbiotic cleared S. aureus. Keratinocytes treated in vitro with BGMK2-41 upregulated expression of antimicrobial response genes, of which DEFB4, ANG, and RNASE7 were also found upregulated in treated ex vivo human skin together with CAMP exclusively upregulated ex vivo. Furthermore, BGMK2-41 postbiotic treatment has a multifaceted impact on the wound healing process. Treatment of keratinocytes stimulated cell migration and the expression of tight junction proteins, while in ex vivo human skin BGMK2-41 increased expression of anti-inflammatory cytokine IL-10, promoted re-epithelialization, and restored the epidermal barrier via upregulation of tight junction proteins. Together, this provides a potential therapeutic approach for persistent intracellular S. aureus infections.


Subject(s)
Keratinocytes , Lactobacillus , Staphylococcus aureus , Humans , Keratinocytes/microbiology , Keratinocytes/metabolism , Keratinocytes/drug effects , Skin/microbiology , Skin/metabolism , Wound Healing/drug effects , Probiotics/pharmacology , Staphylococcal Infections/microbiology , Staphylococcal Infections/drug therapy , Staphylococcal Infections/metabolism , Ribonucleases/metabolism
5.
J Cell Mol Med ; 28(14): e18550, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39042561

ABSTRACT

Endometritis is one of the important causes of infertility. Puerarin (PU) can inhibit oxidative stress and reduce inflammation; however, it is unclear whether PU has a protective effect on the endometritis. In our study, we used Staphylococcus aureus to induce mouse endometritis. The PU group (100 mg/kg PU) and the S. aureus + PU group received daily intraperitoneal injection of PU (25, 50 or 100 mg/kg PU). The results showed that S. aureus significantly increased the levels of MPO, TNF-α, IL-1ß and IL-6 in uterine tissue, and increased the expression of p-p65 and p-IκBα proteins in uterine tissue to induce endometritis in mice (p < 0.05). Furthermore, it has been found that S. aureus promotes the occurrence of ferroptosis by reducing GSH and ATP content, increasing MDA and iron content and reducing GPX4 and SLC7A11 protein expression levels (p < 0.05). S. aureus significantly increase the expression of NLRP3, ASC, caspase-1 and P2X7 proteins in uterine tissue (p < 0.05). However, PU obviously reduced the inflammatory response and reversed the changes of ferroptosis and the expression of P2X7 receptor/NLRP3 pathway associated proteins of the uterus induced by S. aureus (p < 0.05). Taken together, these findings emphasize the protective effect of PU on endometritis by regulating the P2X7 receptor/NLRP3 signalling pathway and inhibiting ferroptosis.


Subject(s)
Endometritis , Ferroptosis , Isoflavones , NLR Family, Pyrin Domain-Containing 3 Protein , Receptors, Purinergic P2X7 , Signal Transduction , Staphylococcal Infections , Staphylococcus aureus , Animals , Female , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Isoflavones/pharmacology , Isoflavones/therapeutic use , Ferroptosis/drug effects , Staphylococcus aureus/pathogenicity , Endometritis/metabolism , Endometritis/microbiology , Endometritis/drug therapy , Endometritis/pathology , Signal Transduction/drug effects , Mice , Receptors, Purinergic P2X7/metabolism , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Staphylococcal Infections/drug therapy , Inflammation/metabolism , Inflammation/pathology , Uterus/metabolism , Uterus/pathology , Uterus/drug effects , Uterus/microbiology , Oxidative Stress/drug effects
6.
Nat Commun ; 15(1): 5746, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982056

ABSTRACT

Candida albicans and Staphylococcus aureus are two commonly associated pathogens that cause nosocomial infections with high morbidity and mortality. Our prior and current work using a murine model of polymicrobial intra-abdominal infection (IAI) demonstrates that synergistic lethality is driven by Candida-induced upregulation of functional S. aureus α-toxin leading to polymicrobial sepsis and organ damage. In order to determine the candidal effector(s) mediating enhanced virulence, an unbiased screen of C. albicans transcription factor mutants was undertaken revealing that zcf13Δ/Δ fails to drive augmented α-toxin or lethal synergism during co-infection. A combination of transcriptional and phenotypic profiling approaches shows that ZCF13 regulates genes involved in pentose metabolism, including RBK1 and HGT7 that contribute to fungal ribose catabolism and uptake, respectively. Subsequent experiments reveal that ribose inhibits the staphylococcal agr quorum sensing system and concomitantly represses toxicity. Unlike wild-type C. albicans, zcf13Δ/Δ did not effectively utilize ribose during co-culture or co-infection leading to exogenous ribose accumulation and agr repression. Forced expression of RBK1 and HGT7 in the zcf13Δ/Δ mutant fully restores pathogenicity during co-infection. Collectively, our results detail the interwoven complexities of cross-kingdom interactions and highlight how intermicrobial metabolism impacts polymicrobial disease pathogenesis with devastating consequences for the host.


Subject(s)
Candida albicans , Candidiasis , Coinfection , Fungal Proteins , Staphylococcal Infections , Staphylococcus aureus , Candida albicans/metabolism , Candida albicans/pathogenicity , Candida albicans/genetics , Animals , Coinfection/microbiology , Staphylococcus aureus/pathogenicity , Staphylococcus aureus/metabolism , Staphylococcus aureus/genetics , Staphylococcal Infections/microbiology , Staphylococcal Infections/metabolism , Candidiasis/microbiology , Mice , Fungal Proteins/metabolism , Fungal Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Intraabdominal Infections/microbiology , Female , Transcription Factors/metabolism , Transcription Factors/genetics , Quorum Sensing/genetics , Virulence , Gene Expression Regulation, Fungal , Disease Models, Animal , Trans-Activators/metabolism , Trans-Activators/genetics
7.
Cell Rep ; 43(7): 114486, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38990718

ABSTRACT

Skin/soft tissue infections (SSTIs) caused by methicillin-resistant Staphylococcus aureus (MRSA) pose a major healthcare burden. Distinct inflammatory and resolution phases comprise the host immune response to SSTIs. Resolution is a myeloid PPARγ-dependent anti-inflammatory phase that is essential for the clearance of MRSA. However, the signals activating PPARγ to induce resolution remain unknown. Here, we demonstrate that myeloid glucose transporter 1 (GLUT-1) is essential for the onset of resolution. MRSA-challenged macrophages are unsuccessful in generating an oxidative burst or immune radicals in the absence of GLUT-1 due to a reduction in the cellular NADPH pool. This translates in vivo as a significant reduction in lipid peroxidation products required for the activation of PPARγ in MRSA-infected mice lacking myeloid GLUT-1. Chemical induction of PPARγ during infection circumvents this GLUT-1 requirement and improves resolution. Thus, GLUT-1-dependent oxidative burst is essential for the activation of PPARγ and subsequent resolution of SSTIs.


Subject(s)
Glucose Transporter Type 1 , Methicillin-Resistant Staphylococcus aureus , Soft Tissue Infections , Animals , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 1/genetics , Mice , Soft Tissue Infections/microbiology , Soft Tissue Infections/metabolism , Soft Tissue Infections/pathology , PPAR gamma/metabolism , Staphylococcal Skin Infections/microbiology , Staphylococcal Skin Infections/metabolism , Staphylococcal Skin Infections/pathology , Staphylococcal Skin Infections/drug therapy , Mice, Inbred C57BL , Macrophages/metabolism , Macrophages/microbiology , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology
8.
PLoS Pathog ; 20(7): e1012425, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39078849

ABSTRACT

Pathogenic bacteria's metabolic adaptation for survival and proliferation within hosts is a crucial aspect of bacterial pathogenesis. Here, we demonstrate that citrate, the first intermediate of the tricarboxylic acid (TCA) cycle, plays a key role as a regulator of gene expression in Staphylococcus aureus. We show that citrate activates the transcriptional regulator CcpE and thus modulates the expression of numerous genes involved in key cellular pathways such as central carbon metabolism, iron uptake and the synthesis and export of virulence factors. Citrate can also suppress the transcriptional regulatory activity of ferric uptake regulator. Moreover, we determined that accumulated intracellular citrate, partly through the activation of CcpE, decreases the pathogenic potential of S. aureus in animal infection models. Therefore, citrate plays a pivotal role in coordinating carbon metabolism, iron homeostasis, and bacterial pathogenicity at the transcriptional level in S. aureus, going beyond its established role as a TCA cycle intermediate.


Subject(s)
Carbon , Citric Acid , Gene Expression Regulation, Bacterial , Homeostasis , Iron , Staphylococcal Infections , Staphylococcus aureus , Staphylococcus aureus/metabolism , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Iron/metabolism , Carbon/metabolism , Citric Acid/metabolism , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Animals , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Citric Acid Cycle , Mice , Signal Transduction
9.
Bone ; 187: 117181, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38960295

ABSTRACT

Staphylococcus aureus osteomyelitis leads to extensive bone destruction. Osteoclasts are bone resorbing cells that are often increased in bone infected with S. aureus. The cytokine RANKL is essential for osteoclast formation under physiological conditions but in vitro evidence suggests that inflammatory cytokines may by-pass the requirement for RANKL. The goal of this study was to determine whether RANKL-dependent osteoclast formation is essential for the bone loss that occurs in a murine model of S. aureus osteomyelitis. To this end, humanized-RANKL mice were infected by direct inoculation of S. aureus into a unicortical defect in the femur. Mice were treated with vehicle or denosumab, a human monoclonal antibody that inhibits RANKL, both before and during a 14-day infection period. The severe cortical bone destruction caused by infection was completely prevented by denosumab administration even though the bacterial burden in the femur was not affected. Osteoclasts were abundant near the inoculation site in vehicle-treated mice but absent in denosumab-treated mice. In situ hybridization demonstrated that S. aureus infection potently stimulated RANKL expression in bone marrow stromal cells. The extensive reactive bone formation that occurs in this osteomyelitis model was also reduced by denosumab administration. Lastly, there was a notable lack of osteoblasts near the infection site suggesting that the normal coupling of bone formation to bone resorption was disrupted by S. aureus infection. These results demonstrate that RANKL-mediated osteoclast formation is required for the bone loss that occurs in S. aureus infection and suggest that disruption of the coupling of bone formation to bone resorption may also contribute to bone loss in this condition.


Subject(s)
Bone Resorption , Denosumab , Disease Models, Animal , Osteoclasts , Osteomyelitis , RANK Ligand , Staphylococcal Infections , Staphylococcus aureus , Animals , Osteomyelitis/microbiology , Osteomyelitis/pathology , Osteomyelitis/metabolism , RANK Ligand/metabolism , Osteoclasts/metabolism , Osteoclasts/pathology , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Mice , Bone Resorption/pathology , Bone Resorption/microbiology , Bone Resorption/metabolism , Denosumab/pharmacology , Humans , Femur/pathology , Femur/microbiology , Antibodies, Monoclonal, Humanized/pharmacology
10.
Cell Mol Life Sci ; 81(1): 300, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39001897

ABSTRACT

BACKGROUND: Age-associated impairments in innate immunity are believed to be a causative factor responsible for severe pathogenesis of Staphylococcus aureus (S. aureus) infection in the bone tissue. However, the basis for age-associated decline in innate immune response upon S. aureus infection remains poorly understood. RESULTS: Our transcriptional data (GEO: GSE166522) from a mouse model of S. aureus osteomyelitis show up-regulated CXCL9 and CXCL10 (CXCL9/10), which is further confirmed in vitro and in vivo by the present study. Notably, monocytes are a main source for CXCL9/10 production in bone marrow upon S. aureus challenge, but this response declines in middle-aged mice. Interestingly, conditional medium of bone marrow monocytes from middle-aged mice has a strikingly decreased effect on bactericidal functions of neutrophils and macrophages compares with that from young mice. We further show that activation of CXCL9/10-CXCR3 axis between monocytes and macrophages/neutrophils promotes the bactericidal function of the cells, whereas blocking the axis impairs such function. Importantly, treatment with either exogenous CXCL9 or CXCL10 in a middle-aged mice model enhances, while pharmacological inhibition of CXCR3 in young mice model impairs, bacterial clearance and bone marrow structure. CONCLUSIONS: These findings demonstrate that bone marrow monocytes act as a critical promotor of innate immune response via the CXLCL9/10-CXCR3 axis upon S. aureus infection, and that the increased susceptibility to S. aureus infection in skeleton in an aged host may be largely attributable to the declined induction of CXCR9/10 in monocytes.


Subject(s)
Chemokine CXCL10 , Chemokine CXCL9 , Disease Models, Animal , Immunity, Innate , Monocytes , Osteomyelitis , Staphylococcal Infections , Staphylococcus aureus , Animals , Osteomyelitis/microbiology , Osteomyelitis/immunology , Osteomyelitis/metabolism , Osteomyelitis/pathology , Monocytes/immunology , Monocytes/metabolism , Chemokine CXCL9/metabolism , Chemokine CXCL9/genetics , Staphylococcus aureus/immunology , Mice , Chemokine CXCL10/metabolism , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcal Infections/metabolism , Mice, Inbred C57BL , Receptors, CXCR3/metabolism , Receptors, CXCR3/genetics , Aging/immunology , Neutrophils/immunology , Neutrophils/metabolism , Macrophages/immunology , Macrophages/metabolism
11.
J Neuroinflammation ; 21(1): 179, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39044282

ABSTRACT

BACKGROUND: Craniotomy is a common neurosurgery used to treat intracranial pathologies. Nearly 5% of the 14 million craniotomies performed worldwide each year become infected, most often with Staphylococcus aureus (S. aureus), which forms a biofilm on the surface of the resected bone segment to establish a chronic infection that is recalcitrant to antibiotics and immune-mediated clearance. Tumor necrosis factor (TNF), a prototypical proinflammatory cytokine, has been implicated in generating protective immunity to various infections. Although TNF is elevated during S. aureus craniotomy infection, its functional importance in regulating disease pathogenesis has not been explored. METHODS: A mouse model of S. aureus craniotomy infection was used to investigate the functional importance of TNF signaling using TNF, TNFR1, and TNFR2 knockout (KO) mice by quantifying bacterial burden, immune infiltrates, inflammatory mediators, and transcriptional changes by RNA-seq. Complementary experiments examined neutrophil extracellular trap formation, leukocyte apoptosis, phagocytosis, and bactericidal activity. RESULTS: TNF transiently regulated neutrophil and granulocytic myeloid-derived suppressor cell recruitment to the brain, subcutaneous galea, and bone flap as evident by significant reductions in both cell types between days 7 to 14 post-infection coinciding with significant decreases in several chemokines, which recovered to wild type levels by day 28. Despite these defects, bacterial burdens were similar in TNF KO and WT mice. RNA-seq revealed enhanced lymphotoxin-α (Lta) expression in TNF KO granulocytes. Since both TNF and LTα signal through TNFR1 and TNFR2, KO mice for each receptor were examined to assess potential redundancy; however, neither strain had any impact on S. aureus burden. In vitro studies revealed that TNF loss selectively altered macrophage responses to S. aureus since TNF KO macrophages displayed significant reductions in phagocytosis, apoptosis, IL-6 production, and bactericidal activity in response to live S. aureus, whereas granulocytes were not affected. CONCLUSION: These findings implicate TNF in modulating granulocyte recruitment during acute craniotomy infection via secondary effects on chemokine production and identify macrophages as a key cellular target of TNF action. However, the lack of changes in bacterial burden in TNF KO animals suggests the involvement of additional signals that dictate S. aureus pathogenesis during craniotomy infection.


Subject(s)
Craniotomy , Mice, Inbred C57BL , Mice, Knockout , Staphylococcal Infections , Staphylococcus aureus , Tumor Necrosis Factor-alpha , Animals , Mice , Staphylococcal Infections/metabolism , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Tumor Necrosis Factor-alpha/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type I/deficiency , Leukocytes/metabolism , Disease Models, Animal , Receptors, Tumor Necrosis Factor, Type II/metabolism
12.
Cell Rep ; 43(7): 114453, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38985677

ABSTRACT

Methicillin-resistant Staphylococcus aureus (MRSA) infection, a major cause of hospital- and community-acquired pneumonia, still has a high mortality rate. Extracellular vesicles (EVs), as crucial mediators of intercellular communication, have a significant impact on infectious diseases. However, the role of EVs from alveolar macrophages (AMs) in MRSA pneumonia remains unclear. We report that AMs phagocytose MRSA and release more EVs in mice with MRSA pneumonia. EVs from AMs harboring phagocytosed MRSA exhibit significant proinflammatory effects and induce necroptosis by delivering tumor necrosis factor α (TNF-α) and miR-146a-5p. Mechanically, the upregulated miR-146a-5p in these EVs enhances the phosphorylation of RIPK1, RIPK3, and MLKL by targeting TNF receptor-associated factor 6 (TRAF6), thereby promoting TNF-α-induced necroptosis. The combination of a TNF-α antagonist and an miR-146a-5p antagomir effectively improves the outcomes of mice with MRSA pneumonia. Overall, we reveal the pronecrotic effect of EVs from MRSA-infected AMs and provide a promising target for the prevention and treatment of MRSA pneumonia.


Subject(s)
Extracellular Vesicles , Macrophages, Alveolar , Methicillin-Resistant Staphylococcus aureus , MicroRNAs , Necroptosis , Animals , Extracellular Vesicles/metabolism , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/microbiology , Mice , MicroRNAs/metabolism , MicroRNAs/genetics , Phagocytosis , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha/metabolism , Staphylococcal Infections/microbiology , Staphylococcal Infections/immunology , Staphylococcal Infections/pathology , Staphylococcal Infections/metabolism , Male , Humans
13.
J Biochem Mol Toxicol ; 38(7): e23753, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38923626

ABSTRACT

Osteomyelitis is an invasive bone infection that can lead to severe pain and even disability, posing a challenge for orthopedic surgery. Naringin can reduce bone-related inflammatory conditions. This study aimed to elucidate the function and mechanism of naringin in a Staphylococcus aureus-induced mouse model of osteomyelitis. Femurs of S. aureus-infected mice were collected after naringin administration and subjected to microcomputed tomography to analyze cortical bone destruction and bone loss. Bacterial growth in femurs was also assessed. Proinflammatory cytokine levels in mouse femurs were measured using enzyme-linked immunosorbent assays. Pathological changes and bone resorption were analyzed using hematoxylin and eosin staining and tartrate-resistant acid phosphatase staining, respectively. Quantitative reverse transcription polymerase chain reaction and western blot analysis were used to quantify the messenger RNA and protein expression of osteogenic differentiation-associated genes in the femurs. The viability of human bone marrow-derived stem cells (hBMSCs) was determined using cell counting kit-8. Alizarin Red S staining and alkaline phosphatase staining were performed to assess the formation of mineralization nodules and bone formation in vitro. Notch signaling-related protein levels in femur tissues and hBMSCs were assessed using western blot analysis. Experimental results revealed that naringin alleviated S. aureus-induced cortical bone destruction and bone loss in mice by increasing the bone volume/total volume ratio. Naringin suppressed S. aureus-induced bacterial growth and inflammation in femurs. Moreover, it alleviated histopathological changes, inhibited bone resorption, and increased the expression of osteogenic markers in osteomyelitic mice. It increased the viability of hBMSCs and promoted their differentiation and bone mineralization in vitro. Furthermore, naringin activated Notch signaling by upregulating the protein levels of Notch1, Jagged1, and Hes1 in the femurs of model mice and S. aureus-stimulated hBMSCs. In conclusion, naringin reduces bacterial growth, inflammation, and bone resorption while upregulating the expression of osteogenic markers in S. aureus-infected mice and hBMSCs by activating Notch signaling.


Subject(s)
Anti-Bacterial Agents , Anti-Inflammatory Agents , Flavanones , Osteomyelitis , Staphylococcal Infections , Staphylococcus aureus , Animals , Flavanones/pharmacology , Mice , Osteomyelitis/drug therapy , Osteomyelitis/microbiology , Osteomyelitis/metabolism , Osteomyelitis/pathology , Staphylococcal Infections/drug therapy , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Anti-Bacterial Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Humans , Male , Osteogenesis/drug effects , Femur/pathology , Femur/metabolism , Femur/microbiology , Femur/drug effects
14.
J Biol Chem ; 300(7): 107455, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38852884

ABSTRACT

Menstrual toxic shock syndrome (mTSS) is a rare but severe disorder associated with the use of menstrual products such as high-absorbency tampons and is caused by Staphylococcus aureus strains that produce the toxic shock syndrome toxin-1 (TSST-1) superantigen. Herein, we screened a library of 3920 small bioactive molecules for the ability to inhibit transcription of the TSST-1 gene without inhibiting the growth of S. aureus. The dominant positive regulator of TSST-1 is the SaeRS two-component system (TCS), and we identified phenazopyridine hydrochloride (PP-HCl) that repressed the production of TSST-1 by inhibiting the kinase function of SaeS. PP-HCl competed with ATP for binding of the kinase SaeS leading to decreased phosphorylation of SaeR and reduced expression of TSST-1 as well as several other secreted virulence factors known to be regulated by SaeRS. PP-HCl targets the virulence of S. aureus, and it also decreases the impact of TSST-1 on human lymphocytes without affecting the healthy vaginal microbiota. Our findings demonstrate the promising potential of PP-HCl as a therapeutic strategy against mTSS.


Subject(s)
Bacterial Proteins , Bacterial Toxins , Enterotoxins , Staphylococcus aureus , Superantigens , Superantigens/metabolism , Superantigens/genetics , Enterotoxins/metabolism , Staphylococcus aureus/drug effects , Staphylococcus aureus/metabolism , Humans , Bacterial Toxins/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/antagonists & inhibitors , Female , Shock, Septic/drug therapy , Shock, Septic/metabolism , Shock, Septic/microbiology , Gene Expression Regulation, Bacterial/drug effects , Protein Kinases/metabolism , Protein Kinases/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Staphylococcal Infections/drug therapy , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Virulence/drug effects , Lymphocytes/metabolism , Lymphocytes/drug effects , Menstrual Hygiene Products
15.
Nat Commun ; 15(1): 3666, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38693120

ABSTRACT

Respiratory viral infection increases host susceptibility to secondary bacterial infections, yet the precise dynamics within airway epithelia remain elusive. Here, we elucidate the pivotal role of CD47 in the airway epithelium during bacterial super-infection. We demonstrated that upon influenza virus infection, CD47 expression was upregulated and localized on the apical surface of ciliated cells within primary human nasal or bronchial epithelial cells. This induced CD47 exposure provided attachment sites for Staphylococcus aureus, thereby compromising the epithelial barrier integrity. Through bacterial adhesion assays and in vitro pull-down assays, we identified fibronectin-binding proteins (FnBP) of S. aureus as a key component that binds to CD47. Furthermore, we found that ciliated cell-specific CD47 deficiency or neutralizing antibody-mediated CD47 inactivation enhanced in vivo survival rates. These findings suggest that interfering with the interaction between airway epithelial CD47 and pathogenic bacterial FnBP holds promise for alleviating the adverse effects of super-infection.


Subject(s)
CD47 Antigen , Epithelial Cells , Staphylococcal Infections , Staphylococcus aureus , Superinfection , CD47 Antigen/metabolism , CD47 Antigen/genetics , Humans , Animals , Superinfection/microbiology , Mice , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Epithelial Cells/virology , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Influenza, Human/metabolism , Influenza, Human/immunology , Influenza, Human/virology , Bacterial Adhesion , Respiratory Mucosa/metabolism , Respiratory Mucosa/microbiology , Respiratory Mucosa/virology , Mice, Inbred C57BL , Bronchi/metabolism , Bronchi/cytology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Mice, Knockout , Influenza A Virus, H1N1 Subtype
16.
Cell Death Dis ; 15(5): 360, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38789414

ABSTRACT

Disseminated intravascular coagulation (DIC) is considered to be the most common and lethal complication of sepsis. NLR-family pyrin domain-containing-3 (NLRP3) inflammasome plays an important role in host defense against microbial pathogens, and its deregulation may cause coagulation cascade and should be strictly managed. Here, we identified the deubiquitinase YOD1, which played a vital role in regulating coagulation in a NLRP3 inflammasome-dependent manner in sepsis induced by methicillin-resistant Staphylococcus aureus (MRSA). YOD1 interacted with NLRP3 to remove K33-linked ubiquitination of NLRP3 based on its deubiquitinating enzyme activity and specifically inhibited expression of NLRP3 as well as activation of NLRP3 inflammasome. Deficiency of YOD1 expression enhanced NLRP3 inflammasome activation and coagulation both in vitro and in vivo. In addition, pharmacological inhibition of the NLRP3 effectively improved coagulation and alleviated organ injury in Yod1-/- mice infected with MRSA. Thus, our study reported that YOD1 is a key regulator of coagulation during MRSA infection, and provided YOD1 as a potential therapeutic target for the treatment of NLRP3 inflammasome-related diseases, especially MRSA sepsis-induced DIC.


Subject(s)
Disseminated Intravascular Coagulation , Inflammasomes , Methicillin-Resistant Staphylococcus aureus , NLR Family, Pyrin Domain-Containing 3 Protein , Sepsis , Ubiquitination , Animals , Humans , Male , Mice , Disseminated Intravascular Coagulation/metabolism , Disseminated Intravascular Coagulation/pathology , Disseminated Intravascular Coagulation/microbiology , HEK293 Cells , Inflammasomes/metabolism , Lysine/metabolism , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Sepsis/microbiology , Sepsis/complications , Sepsis/metabolism , Staphylococcal Infections/microbiology , Staphylococcal Infections/metabolism
17.
Int J Biol Sci ; 20(7): 2555-2575, 2024.
Article in English | MEDLINE | ID: mdl-38725861

ABSTRACT

Staphylococcus aureus (S. aureus) persistence in macrophages, potentially a reservoir for recurrence of chronic osteomyelitis, contributes to resistance and failure in treatment. As the mechanisms underlying survival of S. aureus in macrophages remain largely unknown, there has been no treatment approved. Here, in a mouse model of S. aureus osteomyelitis, we identified significantly up-regulated expression of SLC7A11 in both transcriptomes and translatomes of CD11b+F4/80+ macrophages, and validated a predominant distribution of SLC7A11 in F4/80+ cells around the S. aureus abscess. Importantly, pharmacological inhibition or genetic knockout of SLC7A11 promoted the bactericidal function of macrophages, reduced bacterial burden in the bone and improved bone structure in mice with S. aureus osteomyelitis. Mechanistically, aberrantly expressed SLC7A11 down-regulated the level of intracellular ROS and reduced lipid peroxidation, contributing to the impaired bactericidal function of macrophages. Interestingly, blocking SLC7A11 further activated expression of PD-L1 via the ROS-NF-κB axis, and a combination therapy of targeting both SLC7A11 and PD-L1 significantly enhanced the efficacy of clearing S. aureus in vitro and in vivo. Our findings suggest that targeting both SLC7A11 and PD-L1 is a promising therapeutic approach to reprogram the bactericidal function of macrophages and promote bacterial clearance in S. aureus osteomyelitis.


Subject(s)
Amino Acid Transport System y+ , Macrophages , Osteomyelitis , Staphylococcal Infections , Animals , Mice , Amino Acid Transport System y+/metabolism , Amino Acid Transport System y+/genetics , Macrophages/metabolism , Mice, Inbred C57BL , Osteomyelitis/metabolism , Osteomyelitis/microbiology , Reactive Oxygen Species/metabolism , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus
18.
Proc Natl Acad Sci U S A ; 121(22): e2402764121, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38771879

ABSTRACT

Staphylococcus aureus (S. aureus) can evade antibiotics and host immune defenses by persisting within infected cells. Here, we demonstrate that in infected host cells, S. aureus type VII secretion system (T7SS) extracellular protein B (EsxB) interacts with the stimulator of interferon genes (STING) protein and suppresses the inflammatory defense mechanism of macrophages during early infection. The binding of EsxB with STING disrupts the K48-linked ubiquitination of EsxB at lysine 33, thereby preventing EsxB degradation. Furthermore, EsxB-STING binding appears to interrupt the interaction of 2 vital regulatory proteins with STING: aspartate-histidine-histidine-cysteine domain-containing protein 3 (DHHC3) and TNF receptor-associated factor 6. This persistent dual suppression of STING interactions deregulates intracellular proinflammatory pathways in macrophages, inhibiting STING's palmitoylation at cysteine 91 and its K63-linked ubiquitination at lysine 83. These findings uncover an immune-evasion mechanism by S. aureus T7SS during intracellular macrophage infection, which has implications for developing effective immunomodulators to combat S. aureus infections.


Subject(s)
Bacterial Proteins , Macrophages , Membrane Proteins , Staphylococcal Infections , Staphylococcus aureus , Type VII Secretion Systems , Ubiquitination , Staphylococcus aureus/immunology , Membrane Proteins/metabolism , Membrane Proteins/immunology , Humans , Bacterial Proteins/metabolism , Bacterial Proteins/immunology , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Animals , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Infections/metabolism , Type VII Secretion Systems/metabolism , Type VII Secretion Systems/immunology , Type VII Secretion Systems/genetics , Mice , Immune Evasion , Host-Pathogen Interactions/immunology
19.
mSystems ; 9(5): e0017924, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38656122

ABSTRACT

The utilization of ATP within cells plays a fundamental role in cellular processes that are essential for the regulation of host-pathogen dynamics and the subsequent immune response. This study focuses on ATP-binding proteins to dissect the complex interplay between Staphylococcus aureus and human cells, particularly macrophages (THP-1) and keratinocytes (HaCaT), during an intracellular infection. A snapshot of the various protein activity and function is provided using a desthiobiotin-ATP probe, which targets ATP-interacting proteins. In S. aureus, we observe enrichment in pathways required for nutrient acquisition, biosynthesis and metabolism of amino acids, and energy metabolism when located inside human cells. Additionally, the direct profiling of the protein activity revealed specific adaptations of S. aureus to the keratinocytes and macrophages. Mapping the differentially activated proteins to biochemical pathways in the human cells with intracellular bacteria revealed cell-type-specific adaptations to bacterial challenges where THP-1 cells prioritized immune defenses, autophagic cell death, and inflammation. In contrast, HaCaT cells emphasized barrier integrity and immune activation. We also observe bacterial modulation of host processes and metabolic shifts. These findings offer valuable insights into the dynamics of S. aureus-host cell interactions, shedding light on modulating host immune responses to S. aureus, which could involve developing immunomodulatory therapies. IMPORTANCE: This study uses a chemoproteomic approach to target active ATP-interacting proteins and examines the dynamic proteomic interactions between Staphylococcus aureus and human cell lines THP-1 and HaCaT. It uncovers the distinct responses of macrophages and keratinocytes during bacterial infection. S. aureus demonstrated a tailored response to the intracellular environment of each cell type and adaptation during exposure to professional and non-professional phagocytes. It also highlights strategies employed by S. aureus to persist within host cells. This study offers significant insights into the human cell response to S. aureus infection, illuminating the complex proteomic shifts that underlie the defense mechanisms of macrophages and keratinocytes. Notably, the study underscores the nuanced interplay between the host's metabolic reprogramming and immune strategy, suggesting potential therapeutic targets for enhancing host defense and inhibiting bacterial survival. The findings enhance our understanding of host-pathogen interactions and can inform the development of targeted therapies against S. aureus infections.


Subject(s)
Adenosine Triphosphate , Host-Pathogen Interactions , Keratinocytes , Macrophages , Staphylococcus aureus , Humans , Staphylococcus aureus/metabolism , Adenosine Triphosphate/metabolism , Host-Pathogen Interactions/immunology , Macrophages/microbiology , Macrophages/metabolism , Macrophages/immunology , Keratinocytes/microbiology , Keratinocytes/metabolism , Keratinocytes/immunology , THP-1 Cells , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Proteomics/methods , Bacterial Proteins/metabolism , HaCaT Cells
20.
Int J Mol Sci ; 25(8)2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38673764

ABSTRACT

The exacerbation of pneumonia in children with human adenovirus type 3 (HAdV-3E) is secondary to a Staphylococcus aureus (S. aureus) infection. The influence of host-pathogen interactions on disease progression remains unclear. It is important to note that S. aureus infections following an HAdV-3E infection are frequently observed in clinical settings, yet the underlying susceptibility mechanisms are not fully understood. This study utilized an A549 cell model to investigate secondary infection with S. aureus following an HAdV-3E infection. The findings suggest that HAdV-3E exacerbates the S. aureus infection by intensifying lung epithelial cell damage. The results highlight the role of HAdV-3E in enhancing the interferon signaling pathway through RIG-I (DDX58), resulting in the increased expression of interferon-stimulating factors like MX1, RSAD2, and USP18. The increase in interferon-stimulating factors inhibits the NF-κB and MAPK/P38 pro-inflammatory signaling pathways. These findings reveal new mechanisms of action for HAdV-3E and S. aureus in secondary infections, enhancing our comprehension of pathogenesis.


Subject(s)
Adenovirus Infections, Human , Adenoviruses, Human , DEAD Box Protein 58 , Signal Transduction , Staphylococcal Infections , Staphylococcus aureus , Humans , A549 Cells , Adaptor Proteins, Signal Transducing/metabolism , Adenovirus Infections, Human/metabolism , Adenovirus Infections, Human/immunology , Adenovirus Infections, Human/virology , Adenoviruses, Human/physiology , Adenoviruses, Human/immunology , Coinfection/microbiology , DEAD Box Protein 58/metabolism , Host-Pathogen Interactions/immunology , Inflammation/metabolism , NF-kappa B/metabolism , Receptors, Immunologic/metabolism , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus/pathogenicity , Ubiquitin Thiolesterase
SELECTION OF CITATIONS
SEARCH DETAIL