Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.669
Filtrar
1.
Cell Rep ; 43(7): 114453, 2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-38985677

RESUMO

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.


Assuntos
Vesículas Extracelulares , Macrófagos Alveolares , Staphylococcus aureus Resistente à Meticilina , MicroRNAs , Necroptose , Animais , Vesículas Extracelulares/metabolismo , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/microbiologia , Camundongos , MicroRNAs/metabolismo , MicroRNAs/genética , Fagocitose , Camundongos Endogâmicos C57BL , Fator de Necrose Tumoral alfa/metabolismo , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/patologia , Infecções Estafilocócicas/metabolismo , Masculino , Humanos
2.
PLoS Pathog ; 20(7): e1012394, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38991026

RESUMO

Staphylococcus aureus is a facultative intracellular pathogen of human macrophages, which facilitates chronic infection. The genotypes, pathways, and mutations influencing that phenotype remain incompletely explored. Here, we used two distinct strategies to ascertain S. aureus gene mutations affecting pathogenesis in macrophages. First, we analyzed isolates collected serially from chronic cystic fibrosis (CF) respiratory infections. We found that S. aureus strains evolved greater macrophage invasion capacity during chronic human infection. Bacterial genome-wide association studies (GWAS) identified 127 candidate genes for which mutation was significantly associated with macrophage pathogenesis in vivo. In parallel, we passaged laboratory S. aureus strains in vitro to select for increased infection of human THP-1 derived macrophages, which identified 15 candidate genes by whole-genome sequencing. Functional validation of candidate genes using isogenic transposon mutant knockouts and CRISPR interference (CRISPRi) knockdowns confirmed virulence contributions from 37 of 39 tested genes (95%) implicated by in vivo studies and 7 of 10 genes (70%) ascertained from in vitro selection, with one gene in common to the two strategies. Validated genes included 17 known virulence factors (39%) and 27 newly identified by our study (61%), some encoding functions not previously associated with macrophage pathogenesis. Most genes (80%) positively impacted macrophage invasion when disrupted, consistent with the phenotype readily arising from loss-of-function mutations in vivo. This work reveals genes and mechanisms that contribute to S. aureus infection of macrophages, highlights differences in mutations underlying convergent phenotypes arising from in vivo and in vitro systems, and supports the relevance of S. aureus macrophage pathogenesis during chronic respiratory infection in CF. Additional studies will be needed to illuminate the exact mechanisms by which implicated mutations affect their phenotypes.


Assuntos
Fibrose Cística , Estudo de Associação Genômica Ampla , Macrófagos , Infecções Estafilocócicas , Staphylococcus aureus , Humanos , Staphylococcus aureus/genética , Staphylococcus aureus/patogenicidade , Macrófagos/microbiologia , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/genética , Infecções Estafilocócicas/patologia , Fibrose Cística/microbiologia , Mutação , Virulência/genética , Fatores de Virulência/genética , Adaptação Fisiológica
3.
Colloids Surf B Biointerfaces ; 241: 114068, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38954940

RESUMO

Inspired by glycyrrhizin's strong pharmacological activities and the directed self-assembly into hydrogels, we created a novel carrier-free, injectable hydrogel (CAR@glycygel) by combining glycyrrhizin with carvacrol (CAR), without any other chemical crosslinkers, to promote wound healing on bacteria-infected skin. CAR appeared to readily dissolve and load into CAR@glycygel. CAR@glycygel had a dense, porous, sponge structure and strong antioxidant characteristics. In vitro, it showed better antibacterial ability than free CAR. For methicillin-resistant Staphylococcus aureus (MRSA), Staphylococcus aureus, and Escherichia coli, the diameter of inhibition zone values of CAR@glycygel were 3.80 ± 0.04, 3.31 ± 0.20 and 3.12 ± 0.24 times greater, respectively, than those of free CAR. The MICs for CAR@glycygel was 156.25 µg/mL while it was 1250.00 µg/mL for free CAR to these three bacteria. Its antibacterial mechanism appeared to involve destruction of the integrity of the bacterial cell wall and biomembrane, leading to a leakage of AKP and inhibition of biofilm formation. In vivo, CAR@glycygel effectively stopped bleeding. When applied to skin wounds on rats infected with MRSA, CAR@glycygel had strong bactericidal activity and improved wound healing. The wound healing rates for CAR@glycygel were 49.59 ± 15.78 %, 93.02 ± 3.09 % and 99.02 ± 0.55 % on day 3, day 7, and day 11, respectively, which were much better than blank control and positive control groups. Mechanisms of CAR@glycygel accelerating wound healing involved facilitating epidermis remolding, promoting the growth of hair follicles, stimulating collagen deposition, mitigating inflammation, and promoting angiogenesis. Overall, CAR@glycygel showed great potential as wound dressing for infected skin wounds.


Assuntos
Antibacterianos , Cimenos , Ácido Glicirrízico , Hidrogéis , Staphylococcus aureus Resistente à Meticilina , Testes de Sensibilidade Microbiana , Cicatrização , Cicatrização/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Hidrogéis/química , Hidrogéis/farmacologia , Animais , Ácido Glicirrízico/farmacologia , Ácido Glicirrízico/química , Antibacterianos/farmacologia , Antibacterianos/química , Cimenos/farmacologia , Cimenos/química , Ratos , Ratos Sprague-Dawley , Masculino , Escherichia coli/efeitos dos fármacos , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia
4.
Sci Transl Med ; 16(754): eadi6887, 2024 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-38959328

RESUMO

Virulent infectious agents such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and methicillin-resistant Staphylococcus aureus (MRSA) induce tissue damage that recruits neutrophils, monocyte, and macrophages, leading to T cell exhaustion, fibrosis, vascular leak, epithelial cell depletion, and fatal organ damage. Neutrophils, monocytes, and macrophages recruited to pathogen-infected lungs, including SARS-CoV-2-infected lungs, express phosphatidylinositol 3-kinase gamma (PI3Kγ), a signaling protein that coordinates both granulocyte and monocyte trafficking to diseased tissues and immune-suppressive, profibrotic transcription in myeloid cells. PI3Kγ deletion and inhibition with the clinical PI3Kγ inhibitor eganelisib promoted survival in models of infectious diseases, including SARS-CoV-2 and MRSA, by suppressing inflammation, vascular leak, organ damage, and cytokine storm. These results demonstrate essential roles for PI3Kγ in inflammatory lung disease and support the potential use of PI3Kγ inhibitors to suppress inflammation in severe infectious diseases.


Assuntos
COVID-19 , Classe Ib de Fosfatidilinositol 3-Quinase , Inflamação , SARS-CoV-2 , Animais , Humanos , Camundongos , Permeabilidade Capilar/efeitos dos fármacos , Classe Ib de Fosfatidilinositol 3-Quinase/metabolismo , COVID-19/patologia , Tratamento Farmacológico da COVID-19 , Síndrome da Liberação de Citocina/tratamento farmacológico , Inflamação/patologia , Pulmão/patologia , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Inibidores de Fosfoinositídeo-3 Quinase/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase/uso terapêutico , SARS-CoV-2/fisiologia , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/patologia
5.
Cell Mol Life Sci ; 81(1): 300, 2024 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-39001897

RESUMO

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.


Assuntos
Quimiocina CXCL10 , Quimiocina CXCL9 , Modelos Animais de Doenças , Imunidade Inata , Monócitos , Osteomielite , Infecções Estafilocócicas , Staphylococcus aureus , Animais , Osteomielite/microbiologia , Osteomielite/imunologia , Osteomielite/metabolismo , Osteomielite/patologia , Monócitos/imunologia , Monócitos/metabolismo , Quimiocina CXCL9/metabolismo , Quimiocina CXCL9/genética , Staphylococcus aureus/imunologia , Camundongos , Quimiocina CXCL10/metabolismo , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia , Infecções Estafilocócicas/metabolismo , Camundongos Endogâmicos C57BL , Receptores CXCR3/metabolismo , Receptores CXCR3/genética , Envelhecimento/imunologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo
6.
Bone ; 187: 117181, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38960295

RESUMO

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.


Assuntos
Reabsorção Óssea , Denosumab , Modelos Animais de Doenças , Osteoclastos , Osteomielite , Ligante RANK , Infecções Estafilocócicas , Staphylococcus aureus , Animais , Osteomielite/microbiologia , Osteomielite/patologia , Osteomielite/metabolismo , Ligante RANK/metabolismo , Osteoclastos/metabolismo , Osteoclastos/patologia , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia , Camundongos , Reabsorção Óssea/patologia , Reabsorção Óssea/microbiologia , Reabsorção Óssea/metabolismo , Denosumab/farmacologia , Humanos , Fêmur/patologia , Fêmur/microbiologia , Anticorpos Monoclonais Humanizados/farmacologia
7.
Infect Immun ; 92(8): e0013324, 2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-38953668

RESUMO

Staphylococcus aureus α-hemolysin (Hla) is a pore-forming toxin critical for the pathogenesis of skin and soft tissue infections, which causes the pathognomonic lesion of cutaneous necrosis (dermonecrosis) in mouse models. To determine the mechanism by which dermonecrosis develops during S. aureus skin infection, mice were given control serum, Hla-neutralizing antiserum, or an inhibitor of Hla receptor [A-disintegrin and metalloprotease 10 (ADAM10) inhibitor] followed by subcutaneous infection by S. aureus, and the lesions were evaluated using immunohistochemistry and immunofluorescence. Hla induced apoptosis in the vascular endothelium at 6 hours post-infection (hpi), followed by apoptosis in keratinocytes at 24 hpi. The loss of vascular endothelial (VE)-cadherin expression preceded the loss of epithelial-cadherin expression. Hla also induced hypoxia in the keratinocytes at 24 hpi following vascular injury. Treatment with Hla-neutralizing antibody or ADAM10 inhibitor attenuated early cleavage of VE-cadherin, cutaneous hypoxia, and dermonecrosis. These findings suggest that Hla-mediated vascular injury with cutaneous hypoxia underlies the pathogenesis of S. aureus-induced dermonecrosis.


Assuntos
Proteína ADAM10 , Toxinas Bacterianas , Caderinas , Proteínas Hemolisinas , Queratinócitos , Necrose , Staphylococcus aureus , Animais , Proteínas Hemolisinas/metabolismo , Proteínas Hemolisinas/toxicidade , Camundongos , Toxinas Bacterianas/toxicidade , Toxinas Bacterianas/metabolismo , Staphylococcus aureus/patogenicidade , Queratinócitos/microbiologia , Queratinócitos/metabolismo , Proteína ADAM10/metabolismo , Caderinas/metabolismo , Apoptose , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/metabolismo , Antígenos CD/metabolismo , Proteínas de Membrana/metabolismo , Infecções Cutâneas Estafilocócicas/microbiologia , Infecções Cutâneas Estafilocócicas/patologia , Infecções Cutâneas Estafilocócicas/imunologia , Pele/patologia , Pele/microbiologia , Feminino , Endotélio Vascular/patologia , Endotélio Vascular/microbiologia , Endotélio Vascular/metabolismo , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/patologia , Modelos Animais de Doenças
8.
J Biochem Mol Toxicol ; 38(7): e23753, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38923626

RESUMO

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.


Assuntos
Antibacterianos , Anti-Inflamatórios , Flavanonas , Osteomielite , Infecções Estafilocócicas , Staphylococcus aureus , Animais , Flavanonas/farmacologia , Camundongos , Osteomielite/tratamento farmacológico , Osteomielite/microbiologia , Osteomielite/metabolismo , Osteomielite/patologia , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia , Antibacterianos/farmacologia , Anti-Inflamatórios/farmacologia , Humanos , Masculino , Osteogênese/efeitos dos fármacos , Fêmur/patologia , Fêmur/metabolismo , Fêmur/microbiologia , Fêmur/efeitos dos fármacos
9.
Free Radic Biol Med ; 222: 122-129, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38848785

RESUMO

Osteomyelitis, a grave deep tissue infection primarily caused by Staphylococcus aureus, results in serious complications such as abscesses and sepsis. With the incidence from open fractures exceeding 30 % and prevalent antibiotic resistance due to extensive treatment regimens, there's an urgent need for innovative, antibiotic-free strategies. Photothermal therapy (PTT) and photodynamic therapy (PDT) renowned for generating localized reactive oxygen species (ROS), face limitations in penetration depth. To overcome this, our method combines the deep penetration attributes of medical microwaves (MW) with the synergistic effects of the ZnO/ZrO2 solid solution. Comprehensive in vitro and in vivo evaluations showcased the solid-solution's potent antibacterial efficacy and biocompatibility. The ZnO/ZrO2 solid solution, especially in a 7:3 M ratio, manifests superior microstructural characteristics, optimizing MW-assisted therapy. Our findings highlight the potential of this integrated strategy as a promising avenue in osteomyelitis management.


Assuntos
Micro-Ondas , Osteomielite , Staphylococcus aureus , Zircônio , Osteomielite/tratamento farmacológico , Osteomielite/microbiologia , Osteomielite/patologia , Osteomielite/terapia , Animais , Staphylococcus aureus/efeitos dos fármacos , Zircônio/química , Camundongos , Humanos , Catálise , Antibacterianos/farmacologia , Antibacterianos/química , Fotoquimioterapia/métodos , Zinco/química , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia , Oxigênio/química , Oxigênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Terapia Fototérmica/métodos , Testes de Sensibilidade Microbiana
10.
Acta Biomater ; 181: 161-175, 2024 06.
Artigo em Inglês | MEDLINE | ID: mdl-38679405

RESUMO

Diabetic wound management remains a significant challenge in clinical care due to bacterial infections, excessive inflammation, presence of excessive reactive oxygen species (ROS), and impaired angiogenesis. The use of multifunctional wound dressings has several advantages in diabetic wound healing. Moreover, the balance of macrophage polarization plays a crucial role in promoting skin regeneration. However, few studies have focused on the development of multifunctional wound dressings that can regulate the inflammatory microenvironment and promote diabetic wound healing. In this study, an extracellular matrix-inspired glycopeptide hydrogel composed of glucomannan and polypeptide was proposed for regulating the local microenvironment of diabetic wound sites. The hydrogel network, which was formed via Schiff base and hydrogen bonding interactions, effectively inhibited inflammation and promoted angiogenesis during wound healing. The hydrogels exhibited sufficient self-healing ability and had the potential to scavenge ROS and to activate the mannose receptor (MR), thereby inducing macrophage polarization toward the M2 phenotype. The experimental results confirm that the glycopeptide hydrogel is an effective tool for managing diabetic wounds by showing antibacterial, ROS scavenging, and anti-inflammatory effects, and promoting angiogenesis to facilitate wound repair and skin regeneration in vivo. STATEMENT OF SIGNIFICANCE: •The designed wound dressing combines the advantage of natural polysaccharide and polypeptide. •The hydrogel promotes M2-polarized macrophages, antibacterial, scavenges ROS, and angiogenesis. •The multifunctional glycopeptide hydrogel dressing could accelerating diabetic wound healing in vivo.


Assuntos
Glicopeptídeos , Hidrogéis , Staphylococcus aureus Resistente à Meticilina , Nanofibras , Cicatrização , Animais , Cicatrização/efeitos dos fármacos , Hidrogéis/química , Hidrogéis/farmacologia , Nanofibras/química , Camundongos , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Glicopeptídeos/farmacologia , Glicopeptídeos/química , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/patologia , Células RAW 264.7 , Masculino , Mananas/química , Mananas/farmacologia , Antibacterianos/farmacologia , Antibacterianos/química , Espécies Reativas de Oxigênio/metabolismo , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/patologia , Ratos Sprague-Dawley , Complicações do Diabetes/patologia
11.
Acta Biomater ; 179: 300-312, 2024 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-38518865

RESUMO

Deep tissue bacterial infections, especially methicillin-resistant Staphylococcus aureus (MRSA) infections, pose challenges to clinical therapy due to their low debridement efficiency and relapsing. Molybdenum disulfide (MoS2) is used in the antibacterial field as a classic photothermal agent (NIR-I) with good biocompatibility. However, due to its limited NIR-I tissue penetration ability and single treatment mode, MoS2 has poor therapeutic effects on deep tissue infection. Herein, we prepared a defect-type hybrid 2H-MoS2 nanozyme (MoWS2) using hydrothermal method fabricate the MoWS2 composite, which is a new antibacterial strategy involving photothermal and enzyme catalysis, and further enhances the activity of the nanozyme through overheating. The regulation of 2H-MoS2 defects through tungsten ion doping endows MoWS2 with better near-infrared two-region absorption (NIR-II) and enzyme catalytic performance. Antibacterial activity experiments in vitro have shown that MoWS2 can achieve efficient bactericidal activity and biofilm clearance through hyperthermia and reactive oxygen species (ROS). Deep MRSA infection experiments have shown that MoWS2 rapidly removes bacteria from subcutaneous infected tissues through photothermal therapy (PTT) and chemodynamic therapy (CDT), accelerates the dissipation of abscesses, and promotes the healing of infected wounds. Additionally, the versatile treatment mode of MoWS2 was further confirmed through tissue sectioning and immunofluorescence staining analysis. Overall, these results provide a feasible approach for achieving efficient treatment of deep tissue infections through tungsten ion doping to regulate defective 2H-MoS2. STATEMENT OF SIGNIFICANCE: The photothermal effect of MoS2 nanosheets in the NIR-I (650-900 nm) window in anti-MRSA therapy is considered to be highly reliable and efficient in PTA. However, most of the developed PPT therapies or antimicrobial systems based on PTT therapies developed with 1T-MoS2 have in vivo sterilization temperatures of more than 55°C, which have the risk of damaging the normal tissues of the skin. In this study, we prepared W@MoS2 with a good photothermal effect (36.9%) in the NIR-II window and good peroxidase-like activity. The combined effect of PTT and CDT has a stronger bactericidal effect while avoiding high-temperature damage, which makes the W@MoS2 material more advantageous in terms of antimicrobial effect.


Assuntos
Antibacterianos , Staphylococcus aureus Resistente à Meticilina , Molibdênio , Infecções Estafilocócicas , Cicatrização , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Cicatrização/efeitos dos fármacos , Molibdênio/química , Molibdênio/farmacologia , Animais , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/patologia , Antibacterianos/farmacologia , Antibacterianos/química , Camundongos , Dissulfetos/química , Dissulfetos/farmacologia , Raios Infravermelhos , Biofilmes/efeitos dos fármacos , Terapia Fototérmica
12.
Int J Biol Macromol ; 262(Pt 2): 130039, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38354917

RESUMO

There is mounting evidence that the uterine microbiota has an important role in the pathogenesis of endometritis, with invasion of pathogenic bacteria being a main cause of uterine microbial imbalance. However, mechanisms of uterine microbiota resistance to pathogen invasion remain unclear. In this study, an intrauterine infusion of Staphylococcus aureus was used as a bovine endometritis model; it significantly increased abundance of pathogenic bacteria (Streptococcus, Helccoccus, Fusobacterium, and Escherichia-Shigella) and significantly decreased abundance of probiotics (Allstipes, Bacteroides, Phascolarctobacterium, Romboutsia, and Prevotella). In addition, the metabolite aloe-emodin was positively correlated with Prevotella and based on combined analyses of omics and probiotics, the presence of its metabolite aloe-emodin in the uterus at least partially resisted Staphylococcus aureus invasion. Therefore, Aloe-emodin has potential for regulating microbial structure and preventing endometritis.


Assuntos
Emodina , Endometrite , Infecções Estafilocócicas , Feminino , Humanos , Animais , Bovinos , Endometrite/microbiologia , Endometrite/patologia , Staphylococcus aureus/metabolismo , Útero/patologia , Bactérias , Infecções Estafilocócicas/patologia
13.
ACS Biomater Sci Eng ; 10(1): 563-574, 2024 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-38108141

RESUMO

Cobalt-chromium-molybdenum (CoCrMo) alloys are routinely used in arthroplasty. CoCrMo wear particles and ions derived from arthroplasty implants lead to macrophage-driven adverse local tissue reactions, which have been linked to an increased risk of periprosthetic joint infection after revision arthroplasty. While metal-induced cytotoxicity is well characterized in human macrophages, direct effects on their functionality have remained elusive. Synchrotron radiation X-ray microtomography and X-ray fluorescence mapping indicated that peri-implant tissues harvested during aseptic revision of different arthroplasty implants are exposed to Co and Cr in situ. Confocal laser scanning microscopy revealed that macrophage influx is predominant in patient tissue. While in vitro exposure to Cr3+ had only minor effects on monocytes/macrophage phenotype, pathologic concentrations of Co2+ significantly impaired both, monocyte/macrophage phenotype and functionality. High concentrations of Co2+ led to a shift in macrophage subsets and loss of surface markers, including CD14 and CD16. Both Co2+ and Cr3+ impaired macrophage responses to Staphylococcus aureus infection, and particularly, Co2+-exposed macrophages showed decreased phagocytic activity. These findings demonstrate the immunosuppressive effects of locally elevated metal ions on the innate immune response and support further investigations, including studies exploring whether Co2+ and Cr3+ or CoCrMo alloys per se expose the patients to a higher risk of infections post-revision arthroplasty.


Assuntos
Cobalto , Infecções Estafilocócicas , Humanos , Cobalto/toxicidade , Cromo/toxicidade , Staphylococcus aureus , Macrófagos/patologia , Íons/farmacologia , Ligas , Infecções Estafilocócicas/patologia
14.
J Appl Biomed ; 21(4): 200-207, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38112459

RESUMO

The role of oxidants and proinflammatory cytokines in the pathogenesis of pneumonia caused by Staphylococcus aureus (S. aureus) has been demonstrated. The present study aims to investigate the protective effect of ethyl acetate extract (EtOAc) obtained from Usnea longissima (UL) against acute oxidative and inflammatory lung damage due to S. aureus infection in rats. Albino Wistar-type male rats were divided into three groups: Healthy (HG), S. aureus inoculated (SaG), and S. aureus inoculated + ULEtOAc administered (SUL). SaG (n = 6) and SUL (n = 6) group rats' left nostrils (excluding HG) were inoculated with 0.1 ml bacterial mixture. After 24 hours, ULEtOAc (50 mg/kg) was administered orally to the SUL group, and the same volume of normal saline was administered orally to the HG (n = 6) and SaG groups. This procedure was performed once a day for seven days. Levels of oxidant and antioxidant parameters such as malondialdehyde (MDA) and total glutathione (tGSH), as well as pro-inflammatory cytokine levels such as nuclear factor-kappa B (NF-κB), tumor necrosis factor-alpha (TNF-α), interleukin-one beta (IL-1ß), were measured in removed lung tissues. Tissues were also examined histopathologically. Biochemical results showed that ULEtOAc significantly suppressed the increase of MDA, NF-κB, TNF-α, and IL-1ß levels and the decrease of tGSH caused by S. aureus in lung tissue. S. aureus inoculation caused severe mononuclear cell infiltration in interstitial areas, severe lymphoid hyperplasia in bronchial-associated lymphoid tissue and severe alveolar edema, histopathologically. Treatment with ULEtOAc had an attenuating effect on these histopathological findings. Experimental results from this study suggest that ULEtOAc may be beneficial in treating S. aureus-induced oxidative and inflammatory lung damage.


Assuntos
Pneumonia , Infecções Estafilocócicas , Ratos , Masculino , Animais , Staphylococcus aureus/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , NF-kappa B/metabolismo , Pneumonia/tratamento farmacológico , Pneumonia/patologia , Glutationa/metabolismo , Glutationa/farmacologia , Ratos Wistar , Pulmão/metabolismo , Pulmão/patologia , Citocinas , Estresse Oxidativo , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/patologia
15.
Cell ; 186(24): 5201-5202, 2023 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-37995654

RESUMO

Itch exacerbates infection and inflammation-associated skin pathology. In this issue of Cell, Deng et al. identify a V8 protease released by Staphylococcus aureus triggering itch via neuronal protease-activated receptor 1. In so doing, they uncover profound consequences of microbial neurosensory modulation and the ensuing scratch-induced tissue damage that potentiates infection.


Assuntos
Prurido , Infecções Estafilocócicas , Staphylococcus aureus , Humanos , Inflamação/microbiologia , Peptídeo Hidrolases , Prurido/microbiologia , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia
16.
Cell ; 186(24): 5375-5393.e25, 2023 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-37995657

RESUMO

Itch is an unpleasant sensation that evokes a desire to scratch. The skin barrier is constantly exposed to microbes and their products. However, the role of microbes in itch generation is unknown. Here, we show that Staphylococcus aureus, a bacterial pathogen associated with itchy skin diseases, directly activates pruriceptor sensory neurons to drive itch. Epicutaneous S. aureus exposure causes robust itch and scratch-induced damage. By testing multiple isogenic bacterial mutants for virulence factors, we identify the S. aureus serine protease V8 as a critical mediator in evoking spontaneous itch and alloknesis. V8 cleaves proteinase-activated receptor 1 (PAR1) on mouse and human sensory neurons. Targeting PAR1 through genetic deficiency, small interfering RNA (siRNA) knockdown, or pharmacological blockade decreases itch and skin damage caused by V8 and S. aureus exposure. Thus, we identify a mechanism of action for a pruritogenic bacterial factor and demonstrate the potential of inhibiting V8-PAR1 signaling to treat itch.


Assuntos
Peptídeo Hidrolases , Prurido , Receptor PAR-1 , Infecções Estafilocócicas , Staphylococcus aureus , Animais , Humanos , Camundongos , Peptídeo Hidrolases/metabolismo , Prurido/microbiologia , Receptor PAR-1/metabolismo , Staphylococcus aureus/enzimologia , Staphylococcus aureus/patogenicidade , Staphylococcus aureus/fisiologia , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia
17.
BMJ Case Rep ; 16(6)2023 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-37339827

RESUMO

Staphylococcus aureus causes clinical diseases ranging from mild skin infections to devastating conditions such as septic shock, endocarditis and osteomyelitis. S. aureus is a common cause of community-acquired bacteraemia. Prolonged bacteraemia may cause metastatic infection, manifesting as endocarditis, osteomyelitis and abscesses. A man in his 20s presented with a short-duration of fever and odynophagia. CT of the neck suggested a retropharyngeal abscess. Retropharyngeal abscesses are typically polymicrobial and caused by resident oral cavity flora. In the hospital, he developed shortness of breath and hypoxia. CT of the chest showed peripheral, subpleural nodular opacities raising suspicion for septic pulmonary emboli. Blood cultures demonstrated the growth of methicillin-resistant S. aureus The patient completely recovered with antibiotic therapy alone. This is a unique and rare presentation case of metastatic S. aureus bacteraemia, manifesting as a retropharyngeal abscess without any evidence of infective endocarditis on transoesophageal echocardiography.


Assuntos
Bacteriemia , Infecções Comunitárias Adquiridas , Endocardite Bacteriana , Staphylococcus aureus Resistente à Meticilina , Osteomielite , Abscesso Retrofaríngeo , Infecções Estafilocócicas , Masculino , Humanos , Abscesso Retrofaríngeo/diagnóstico por imagem , Staphylococcus aureus , Infecções Estafilocócicas/diagnóstico , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/patologia , Bacteriemia/tratamento farmacológico , Infecções Comunitárias Adquiridas/diagnóstico , Infecções Comunitárias Adquiridas/tratamento farmacológico
18.
J Biochem Mol Toxicol ; 37(4): e23298, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36727417

RESUMO

Methicillin-resistant Staphylococcus aureus (MRSA) is a highly infectious Gram-positive pathogen known to cause severe diseases such as endocarditis, food poisoning, pneumonia, osteomyelitis, and septicemia. MRSA is a major public health issue. Among these, osteomyelitis is inflammation of the bone caused by the invasion of the bacterial pathogen in the bones. Its prominent symptoms include fever, pain, and redness of bones. In the case of children, it affects the long bones of arms and legs, whereas in the case of adults it affects the hip, feet, and spine. Bacterial osteomyelitis can trigger pathological remodeling of bones and hence causes substantial morbidity and mortality. The present study aims to evaluate the isoflavone genistein's (5,7-dihydroxy-3-(4-hydroxyphenyl)-4H-1-benzopyran-4-one,4',5,7 trihydroxyisoflavone) antimicrobial and anti-inflammatory effects against osteomyelitis induced by MRSA in male Wistar rats. Classification of the animals was into the following: sham (Group I), osteomyelitis (Group II, control), genistein (25 mg/kg body weight, Group III), and genistein (50 mg/kg body weight, Group IV). The rats did not receive any treatment for 4 weeks after bacterial inoculation. Genistein was then administered twice daily for 2 weeks. Bacterial growth, mean body weight bone infection status, and side effects of genistein treatment were assessed. Furthermore, lipid peroxidation, superoxide dismutase, glutathione (GSH) peroxidase, catalase, reduced GSH, tumor necrosis factor-α (TNF-α), and interleukin (IL)-6 were also determined. Two days after treatment, it was found that genistein significantly suppressed bacterial growth and reduced serum pro-inflammatory cytokines TNF-α and IL-6. Therefore, the study suggests that genistein could be a promising lead against MRSA-induced osteomyelitis.


Assuntos
Staphylococcus aureus Resistente à Meticilina , Osteomielite , Infecções Estafilocócicas , Ratos , Masculino , Animais , Staphylococcus aureus , Genisteína/farmacologia , Genisteína/uso terapêutico , Fator de Necrose Tumoral alfa , Ratos Wistar , Antibacterianos , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia , Osteomielite/etiologia , Osteomielite/microbiologia , Glutationa , Interleucina-6/farmacologia , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico
19.
Front Immunol ; 14: 1125635, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36761743

RESUMO

Atopic dermatitis (AD) is a common chronic inflammatory skin disease that significantly affects the patient's quality of life. A disrupted skin barrier, type 2 cytokine-dominated inflammation, and microbial dysbiosis with increased Staphylococcus aureus colonization are critical components of AD pathogenesis. Patients with AD exhibit decreased expression of antimicrobial peptides (AMPs) which is linked to increased colonization by Staphylococcus aureus. The skin microbiome itself is a source of several AMPs. These host- and microbiome-derived AMPs define the microbial landscape of the skin based on their differential antimicrobial activity against a range of skin microbes or their quorum sensing inhibitory properties. These are particularly important in preventing and limiting dysbiotic colonization with Staphylococcus aureus. In addition, AMPs are critical for immune homeostasis. In this article, we share our perspectives about the implications of microbial derived AMPs in AD patients and their potential effects on overlapping factors involved in AD. We argue and discuss the potential of bacterial AMPs as therapeutics in AD.


Assuntos
Dermatite Atópica , Infecções Estafilocócicas , Humanos , Peptídeos Antimicrobianos , Qualidade de Vida , Pele , Inflamação/patologia , Staphylococcus aureus , Infecções Estafilocócicas/patologia
20.
Molecules ; 28(3)2023 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-36770872

RESUMO

Cinnamaldehyde (CNM) is an essential-oil component with reported anti-infective, anti-inflammatory, and healing effects, making it an interesting compound for the treatment of wound infection. Herein, we evaluated the effects of topical administration of CNM in experimental wounds infected by Staphylococcus aureus. Swiss mice (n = 12/group) were randomly allocated into three groups (CON: animals with uninfected lesions; Sa: animals with untreated infected lesions; Sa + CNM: animals with infected wounds and treated with CNM). Excisional lesions (64 mm2) were induced at the dorsal area followed by the addition of S. aureus (80 µL of a 1.5 × 108 CFU/mL bacterial suspension). The wounds were treated with CNM (200 µg/wound/day) or vehicle (2% DMSO) for 10 days. Skin samples were taken on the 3rd or 10th treatment day for quantification of inflammatory mediators, bacterial load, immunophenotyping, and histological analysis. The treatment with CNM improved the healing process and attenuated the severity of skin lesions infected by S. aureus. These effects were associated with significant decreases in bacterial loads in CNM-treated wounds. The levels of neutrophils, TNF-α, IL-6, NO, and VEGF were decreased in the lesions treated with CNM. Taken together, these data provide further evidence of the effectiveness of CNM for the treatment of skin infections.


Assuntos
Infecções Estafilocócicas , Infecção dos Ferimentos , Camundongos , Animais , Staphylococcus aureus , Cicatrização , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/patologia , Infecção dos Ferimentos/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...