Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
1.
Microb Pathog ; 187: 106518, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38160988

RESUMO

In vivo studies identifying a role of TLR2 in septic arthritis models are lacking. TNF-α played as the most important proinflammatory cytokine, and connected directly to the pathogenesis of bacterial arthritis. IL-1ß is another central mediator cytokine in arthritis. It is therefore reasonable to question the role of neutralization of endogenous TNF-α and IL-1ß along with TLR2 and associated downstream signaling as crucial mediators in the S. aureus -induced inflammatory arthritis. In reaction to an injury or a pathogen encounter, innate immune cells serve as the initial line of defense. TLR2 mediated entry of S. aureus into macrophage cells initiates an array of inflammatory cascades. After macrophage cell gets activated at the site inflammation, they generate elevated number of cytokines which includes TNF-α, IL-1ß. This cytokines signals through STAT1/STAT3 mediated pathways. Thus, aim of this study was to discover how This bone damage could be altered by altering the STAT/STAT3/SOCS3 ratio by blocking TLR2, a particular S. aureus binding site, in conjunction with the use of IL-1 and TNF- antibodies for neutralizing endogenous IL-1ß and TNF-α. Additionally, the role of local macrophages in therapy of arthritis was investigated in synovial and Splenic tissue. To comprehend the inflammatory milieu within the system, ROS and other antioxidant enzymes, along with the expression of mTOR in macrophage cells, were also taken into consideration. The detrimental impact of bacterial burden on synovial joints was reduced by simultaneously inhibiting TLR2, TNF-α, and IL-1ß. Lowered IFN-γ decreases its sensitivity to STAT1 and lowered IL-6 reduces STAT3 expressions. Whereas, elevated IL-10 enhances SOSC3 expression, which thereby able to limits STAT1/STAT3 inter-conversion. As a result, NF-κB activity was downregulated.


Assuntos
Artrite Infecciosa , Staphylococcus aureus Resistente à Meticilina , Humanos , Fator de Necrose Tumoral alfa/metabolismo , Receptor 2 Toll-Like/metabolismo , Staphylococcus aureus/metabolismo , Staphylococcus aureus Resistente à Meticilina/metabolismo , Citocinas/metabolismo , NF-kappa B/metabolismo , Macrófagos/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas/metabolismo , Fator de Transcrição STAT3/metabolismo
2.
Microb Pathog ; 183: 106294, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37567327

RESUMO

The CXCL8/CXCR1 axis in conjoint with the free radicals and anti-oxidants dictates the severity of inflammation caused by the bacteria, Staphylococcus aureus. S.aureus mediated inflammatory processes is regulated by NF-κB and its product, iNOS. The objective of this study was to examine the effects of inhibition of NF-κB and iNOS on CXCL8/CXCR1, alteration in M1/M2 polarization of macrophages and associated inflammatory responses during S.aureus infection in vitro. For this, the murine peritoneal macrophages were pretreated with NF-κB inhibitor, Pyrrolidine dithiocarbamate (PDTC) and iNOS inhibitor, L-N-monomethyl arginine (LNMMA), either alone or in combination, followed by time-dependent S.aureus infection. The chemotactic migrations of macrophages were determined by the agarose spot assay. The iNOS, NF-κB and CXCR1 protein expressions were evaluated. The ROS level (superoxide, H2O2, NO) and antioxidant activities (SOD, CAT, GSH, arginase) were measured. The intra-macrophage phagoctyic activity had been analyzed by confocal microscopy. S.aureus activated macrophages showed increased iNOS expression that symbolizes M1 characterization of macrophages. The results suggest that the combination treatment of LNMMA + PDTC was effective in diminution of CXCL8 production and CXCR1 expression through downregulation of NF-κB and iNOS signaling pathway. Consequently, there was decrement in macrophage migration, reduced ROS generation, elevated antioxidant enzyme activity as well as bacterial phagocytosis at 90 min post bacterial infection. The increased arginase activity further proves the switch from pro-inflammatory M1 to anti-inflammatory M2 polarization of macrophages. Concludingly, the combination of PDTC + LNMMA could resolve S.aureus mediated inflammation through mitigation of CXCL8/CXCR1 pathway switching from M1 to M2 polarization.


Assuntos
Macrófagos Peritoneais , Infecções Estafilocócicas , Camundongos , Animais , Macrófagos Peritoneais/microbiologia , Staphylococcus aureus/metabolismo , ômega-N-Metilarginina/metabolismo , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Espécies Reativas de Oxigênio/metabolismo , NF-kappa B/metabolismo , Peróxido de Hidrogênio/metabolismo , Arginase/metabolismo , Citocinas/metabolismo , Infecções Estafilocócicas/microbiologia , Receptores de Interleucina-8A/metabolismo , Inflamação/metabolismo
3.
Scand J Immunol ; 97(6): e13252, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36597222

RESUMO

Septic arthritis is a joint disease caused by Staphylococcus aureus. Different macrophage populations contribute in various ways to control blood-borne infections and induce inflammatory responses. Macrophage tissue-resident niche is necessary for the suppression of chronic inflammation and may contribute to the pathogenesis of septic arthritis. Thus, to obtain a resolution of the disease and restoration of synovial homeostasis, it needs the activation of macrophages that further regulate the inflammatory consequences. The aim of this study was to find out the mechanism by which neutralization of transforming growth factor-beta (TGF-ß) and/or interleukin (IL)-6 after induction of septic arthritis could alter the specific macrophage responses in spleen and synovial joints via different cytokines (osteoprotegerin (OPG), osteopontin (OPN), IL-10, IL-12 and CXCL8) cross-talking, and how the response could be modulated by reactive oxygen species vs antioxidant enzyme activities. Dual neutralization of TGF-ß and IL-6 is notably effective in eliciting splenic and synovial tissue-resident macrophage responses. Synovial macrophage-derived IL-10 can elicit protection against septic arthritis via regulating receptor-activated nuclear factor Kappa-B ligand (RANKL)/OPG interaction. They also reduced oxidative stress by increasing the activity of antioxidant enzymes including SOD and catalase. Histopathological analysis revealed that dual neutralization of TGF-ß and IL-6 prevented bone destruction and osteoclastic activity in septic arthritis by promoting the differential functional response of the splenic and synovial macrophages. Additionally, the macrophage-derived IL-10 can elicit protection against S. aureus-induced septic arthritis via regulating RANKL/OPG interaction. Further studies on STAT3 and STAT4 are needed for the understanding of such cross-talking in resident macrophages of arthritic mice.


Assuntos
Artrite Infecciosa , Interleucina-10 , Animais , Camundongos , Staphylococcus aureus , Fator de Crescimento Transformador beta , Interleucina-6 , Baço/patologia , Antioxidantes , Inflamação , Artrite Infecciosa/patologia , Macrófagos/patologia
4.
Cell Mol Neurobiol ; 44(1): 2, 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-38099973

RESUMO

Coronavirus disease 2019 (COVID-19) is primarily a respiratory disease causing a worldwide pandemic in the year of 2019. SARS-CoV-2 is an enveloped, positive-stranded RNA virus that could invade the host through spike protein and exhibits multi-organ effects. The Brain was considered to be a potential target for SARS-CoV-2 infection. Although neuropsychiatric symptoms and cognitive impairments were observed in COVID-19 patients even after recovery the mechanism of action is not well documented. In this review, the contribution of microglia in response to SARS-CoV-2 infection was discussed aiming to design a therapeutic regimen for the management of neuroinflammation and psycho-behavioral alterations. Priming of microglia facilitates the hyper-activation state when it interacts with SARS-CoV-2 known as the 'second hit'. Moreover, the microgliosis produces reactive free radicals and pro-inflammatory cytokines like IL-1ß, IFN-γ, and IL-6 which ultimately contribute to a 'cytokine storm', thereby increasing the occurrence of cognitive and neurological dysfunction. It was reported that elevated CCL11 may be responsible for psychiatric disorders and ROS/RNS-induced oxidative stress could promote major depressive disorder (MDD) and phenotypic switching. Additionally, during SARS-CoV-2 infection microglia-CD8+ T cell interaction may have a significant role in neuronal cell death. This cytokine-mediated cellular cross-talking plays a crucial role in pro-inflammatory and anti-inflammatory balance within the COVID-19 patient's brain. Therefore, all these aspects will be taken into consideration for developing novel therapeutic strategies to combat SARS-CoV-2-induced neuroinflammation.


Assuntos
COVID-19 , Transtorno Depressivo Maior , Humanos , SARS-CoV-2 , Microglia , Doenças Neuroinflamatórias , Citocinas
5.
J Appl Toxicol ; 43(10): 1549-1572, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37177863

RESUMO

Multi-organ dysfunction is one of the major reasons behind the high mortality of sepsis throughout the world. With the pathophysiology of sepsis remaining largely unknown, the uncontrolled reactive oxygen species (ROS) production along with the decreased antioxidants contributes to the progression toward septic shock. Being the effector cells of the innate immunity system, macrophages secrete both pro-inflammatory and anti-inflammatory mediators during inflammation. Lipopolysaccharide (LPS) binding to toll-like receptor 4 (TLR4) releases TNF-α, which initiates pro-inflammatory events through tumor necrosis factor receptor 1 (TNFR1) signaling. However, it is counteracted by the anti-inflammatory interleukin 10 (IL-10) causing decreased oxidative stress. Our study thus aimed to assess the effects of exogenous IL-10 treatment post-neutralization of TLR4 and TNFR1 (by anti-TLR4 antibody and anti-TNFR1 antibody, respectively) in an in vivo murine model of LPS-sepsis. We have also examined the tissue-specific antioxidant status in the spleen, liver, and lungs along with the serum cytokine levels in adult male Swiss albino mice to determine the functional association with the disease. The results showed that administration of recombinant IL-10 post-neutralization of the receptors was beneficial in shifting the macrophage polarization to the anti-inflammatory M2 phenotype. IL-10 treatment significantly downregulated the free radicals production resulting in diminished lipid peroxidase (LPO) levels. The increased antioxidant activities of superoxide dismutase (SOD), catalase (CAT), and glutathione reductase (GRX ) conferred protection against LPS-induced sepsis. Western blot data further confirmed diminished expressions of TLR4 and TNFR1 along with suppressed stress-activated protein kinases/Jun amino-terminal kinases (SAPK/JNK) and increased SOD and CAT expressions, which altogether indicated that neutralization of TLR4 and TNFR1 along with IL-10 posttreatment might be a potential therapeutic measure for the treatment of sepsis.


Assuntos
Antioxidantes , Sepse , Masculino , Camundongos , Animais , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Antioxidantes/metabolismo , Interleucina-10 , Receptor 4 Toll-Like/metabolismo , Lipopolissacarídeos/toxicidade , Sepse/tratamento farmacológico , Sepse/metabolismo , Macrófagos , Fator de Necrose Tumoral alfa/metabolismo , Anti-Inflamatórios/metabolismo , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico
6.
Inflammopharmacology ; 30(4): 1303-1322, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35704229

RESUMO

Microglial inflammatory responses play a central role in the pathogenesis of S. aureus induced brain infections. Upon activation, microglia produces free radicals (ROS/RNS) and disrupts the cellular antioxidant defense to combat invading microorganisms. Despite conventional antibiotic or steroid therapy, microglial over-activation could not be controlled. So, an attempt had been taken by using a natural antioxidant ascorbic acid along with ciprofloxacin to regulate microglial over-activation by involving TLR-2 and glucocorticoid receptor (GR) in an in-vitro cell culture-based study. Combinatorial treatment during TLR-2 neutralization effectively reduced the bacterial burden at 60 min compared to the GR blocking condition (p < 0.05). Moreover, the infection-induced H2O2, O2.-, and NO release in microglial cell culture was diminished possibly by enhancing SOD and catalase activities in the same condition (p < 0.05). The arginase activity was markedly increased after TLR-2 blocking in the combinatorial group compared to single treatments (p < 0.05). Experimental results indicated that combinatorial treatment may act through up-regulating GR expression by augmenting endogenous corticosterone levels. However, better bacterial clearance could further suppress the TLR-2 mediated pro-inflammatory NF-κB signaling. From Western blot analysis, it was concluded that ciprofloxacin-ascorbic acid combination in presence of anti-TLR-2 antibody exhibited 81.25% inhibition of TLR-2 expression while the inhibition for GR was 3.57% with respect to the infected group. Therefore, during TLR-2 blockade ascorbic acid combination might be responsible for the restoration of redox balance in microglia via modulating TLR-2/GR interaction. The combination treatment could play a major role in the neuroendocrine-immune regulation of S. aureus induced microglial activation.


Assuntos
Staphylococcus aureus Resistente à Meticilina , Staphylococcus aureus , Antioxidantes/metabolismo , Antioxidantes/farmacologia , Ácido Ascórbico/farmacologia , Ciprofloxacina/metabolismo , Ciprofloxacina/farmacologia , Peróxido de Hidrogênio/metabolismo , Staphylococcus aureus Resistente à Meticilina/metabolismo , Microglia , Estresse Oxidativo , Receptores de Glucocorticoides/metabolismo
7.
Cell Immunol ; 370: 104441, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34628221

RESUMO

Septic arthritis is a destructive joint disease caused by Staphylococcus aureus. Synovial inflammation involved Th17 proliferation and down regulation of Treg population, thus resolution of inflammation targeting IL-17 may be important to control arthritis. Endogenous inhibition of IL-17 to regulate arthritic inflammation correlating with Th17/Treg cells TLR2 and TNFRs are not done. The role of SOD, CAT and GRx in relation to ROS production during arthritis along with expression of TLR2, TNFR1/TNFR2 in Th17/Treg cells of mice treated with IL-17A Ab/ IL-2 were studied. Increased ROS, reduced antioxidant enzyme activity was found in Th17 cells of SA infected mice whereas Treg cells of IL-17A Ab/ IL-2 treated group showed opposite effects. Neutralization of IL-17 after arthritis cause decreased TNFR1 and increased TNFR2 expression in Treg cells. Thus, neutralization of IL-17 or IL-2 treatment regulates septic arthritis by enhancing anti-inflammatory properties of Treg via antioxidant balance and modulating TLR2/TNFR response.


Assuntos
Artrite Infecciosa/imunologia , Interleucina-17/antagonistas & inibidores , Interleucina-2/farmacologia , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Animais , Antioxidantes/metabolismo , Artrite Experimental/imunologia , Interleucina-17/imunologia , Masculino , Camundongos , Espécies Reativas de Oxigênio/imunologia , Receptores do Fator de Necrose Tumoral/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Células Th17/efeitos dos fármacos , Receptor 2 Toll-Like/imunologia
8.
Immunol Invest ; 50(6): 700-725, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32602757

RESUMO

Anti-cytokine therapy is widely acknowledged as an anti-inflammatory technique to treat varied infectious diseases. TNF-α and IL-1ß are major cytokines that regulate every aspect of the inflammatory process. However, the effects of single or dual cytokine neutralization on S. aureus mediated CXCL8 secretion and CXCR1 expression in murine peritoneal macrophages remained noninvestigated. Thus we aimed to explore the effects of kinetic-dose dependent neutralization of TNF-α and IL-1ß using specific anti-cytokine antibodies and its influential impact on the CXCL8/CXCR1 axis at different stages of S. aureus (30, 60, and 90 min) infection. The murine peritoneal macrophages were isolated and infected with viable S. aureus followed by subsequent addition of anti-TNF-α and anti-IL-1ß into the medium. The treated cells were centrifuged and lysate and supernatant collected for various experiments. The ROS generation was measured and cytokine production was estimated by ELISA. The expression of TNFR1, IL-1R, CXCR1, signaling molecules (NF-κB and JNK) were evaluated by Western blot. The role of single or dual cytokine neutralization on intracellular bacterial phagocytosis had also been analyzed by confocal microscopy. Dual cytokine neutralization significantly suppressed ROS, cytokines, CXCL8 secretion, and intracellular bacterial count compared to single cytokine neutralization and it was more apparent at 90 min post S. aureus infection. There was a drastic reduction in TNFR1, IL-1R, and CXCR1 expression on macrophage surface due to reduced expression of downstream signaling molecules, NF-κB and JNK. Hence dual cytokine neutralization was more effectual compared to single cytokine neutralization in the downregulation of S. aureus induced CXCR1 expression.


Assuntos
Imunossupressores/farmacologia , Interleucina-1beta/antagonistas & inibidores , Interleucina-8/metabolismo , Infecções Estafilocócicas/tratamento farmacológico , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Carga Bacteriana/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/imunologia , Humanos , Imunossupressores/uso terapêutico , Interleucina-1beta/metabolismo , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/microbiologia , Masculino , Camundongos , Cultura Primária de Células , Receptores de Interleucina-8A/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/imunologia , Fator de Necrose Tumoral alfa/metabolismo
9.
Microb Pathog ; 145: 104227, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32360190

RESUMO

Microglial inflammation is the hallmark of S. aureus induced brain abscesses. Conventional antibiotic therapy could not regulate inflammation and the use of steroids in CNS infection remained controversial. To address this issue the effect of dexamethasone along with ciprofloxacin on microglial inflammation has been attempted both in glucocorticoid receptor (GR) opened and blocked condition. We have investigated the effects of ciprofloxacin (0.24 µg/ml, pre-treatment) and dexamethasone (150 nM, pre-treatment) in combination with murine microglia infected with S. aureus for 30, 60 and 90 min by either keeping GR opened or blocked with GR antagonist RU486. Alterations in cellular motility, intracellular killing assay, free radical production, antioxidant enzyme activities, corticosterone, and cytokine levels were determined. The expressions of TLR-2, GR, and other inflammatory markers were determined in terms of this combinatorial treatment. Combination treatment significantly (p < 0.05) reduced the bacterial burden of microglia only when GR remained open and effectively suppressed S. aureus induced oxidative stress by augmenting SOD and catalase enzyme activity and suppressing other pro-inflammatory markers at 90 min. Arginase activity, a critical determinant of microglial polarization was found to be higher after treatment at 60 and 90 min. This situation was reversed when this combination treatment was applied by keeping GR blocked using GR antagonist RU486. Therefore, it can be concluded that combination treatment of ciprofloxacin and dexamethasone could regulate S. aureus induced microglial activation, in the presence of functional GR via utilizing glucocorticoid (GC)-GR pathway and ultimately confers protection to the host from brain inflammation.


Assuntos
Ciprofloxacina , Dexametasona , Glucocorticoides , Microglia , Receptores de Glucocorticoides , Animais , Ciprofloxacina/farmacologia , Dexametasona/farmacologia , Glucocorticoides/farmacologia , Inflamação , Camundongos , Microglia/efeitos dos fármacos , Microglia/microbiologia , Staphylococcus aureus
10.
Microb Pathog ; 139: 103903, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31790794

RESUMO

The contribution of Th17 and Treg in the pathogenesis of septic arthritis is well known. The imbalance of Th17/Treg ratio, especially the skewed CD4+ T cell differentiation towards pathogenic Th17 lineage is a major reason that mediates bone damage through one of its prime cytokine member IL-17A. The neutralization of released IL-17A, as well as exogenous administration of IL-2 at a lower dose, was seen to be potent in dampening the inflammatory response in many cases. Interestingly the effect of IL-17A neutralization to limit IL-17 mediated inflammation and induction of Tregs by the administration of IL-2 has not been studied in experimental arthritis. So in this study, we have treated arthritic mice with IL-17A Ab and recombinant mouse IL-2 either alone or in combination at 3, 9 and 15 days post-infection. We have found a marked decrease in Th17 cell population and their related pro-inflammatory cytokine levels at 15DPI in arthritic mice after IL-17 neutralization. An increased Treg cell population was also observed in mice after application of rIL-2 with a significantly heightened TGF-ß level in serum and synovial joints compared to the untreated one. However, in the case of combination therapy of IL-17A Ab and rIL-2 we have observed a beneficial effect in ameliorating the disease outcome as the arthritic index was decreased maximally at 15DPI with a significant reduction of arthritis compared to individual treatment. Overall the inflammatory microenvironment was counterbalanced most effectively in combination treatment by lowering the Th17/Treg ratio and their related cytokines that resulted in reducing the immunopathogenesis of the destructive arthritis.


Assuntos
Anticorpos/administração & dosagem , Artrite Infecciosa/tratamento farmacológico , Interleucina-17/imunologia , Interleucina-2/administração & dosagem , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus aureus/efeitos dos fármacos , Células Th17/efeitos dos fármacos , Animais , Artrite Infecciosa/imunologia , Artrite Infecciosa/microbiologia , Diferenciação Celular , Humanos , Masculino , Camundongos , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/genética , Staphylococcus aureus/fisiologia , Células Th17/citologia , Células Th17/imunologia , Fator de Crescimento Transformador beta/imunologia
11.
Exp Mol Pathol ; 116: 104485, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32574668

RESUMO

Septic arthritis is a condition of bone disorder caused predominantly by Staphylococcus aureus. Following the bacterial entry activated immune cells specially macrophages and dendritic cells release pro-inflammatory mediators such as IL-6, TNF-α, IL-1ß etc., which not only create an inflammatory microenvironment but also play crucial roles in the proliferation of different CD+ T cell subsets. Among them, Th17 and Tregs are of major concern in recent times because of their potential roles in regulating the ongoing inflammation in many diseases including experimental arthritis. But the downstream signalling mechanism of these cells in regulating the severity of inflammation in case of septic arthritis is not known yet. So, here we have established a murine model of S. aureus induced septic arthritis and kept the animal upto 15 days post-infection. To examine the signalling mechanism, Th17 and Treg cells were isolated from blood, spleen and synovial joints of control and infected mice and observed the expression of JNK, NFκB and RANKL in the lysate of isolated Th17 and Tregs. We have also estimated the levels of serum IL-21 and TGF-ß. NFκB, JNK and RANKL expression was found to be higher at 3 and 15 days post-infection along with serum IL-21 levels. On the other hand, maximum TGF-ß level was observed at 9 days post-infection along with increased Treg population. In conclusion it was hypothesized that bone resorption is related with downstream signalling pathways of Th17 cells, which stimulate osteoclast generation via NFκB/JNK-RANKL axis and helps in the persistence of the disease.


Assuntos
Artrite Infecciosa/imunologia , Inflamação/imunologia , Infecções Estafilocócicas/imunologia , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Animais , Artrite Experimental/genética , Artrite Experimental/imunologia , Artrite Experimental/microbiologia , Artrite Experimental/patologia , Artrite Infecciosa/genética , Artrite Infecciosa/microbiologia , Artrite Infecciosa/patologia , Regulação da Expressão Gênica/genética , Humanos , Inflamação/genética , Inflamação/microbiologia , Inflamação/patologia , Interleucina-1beta/genética , Articulações/imunologia , Articulações/microbiologia , Articulações/patologia , MAP Quinase Quinase 4/genética , Camundongos , Osteoclastos/imunologia , Osteoclastos/microbiologia , Osteoclastos/patologia , Ligante RANK/genética , Transdução de Sinais/genética , Infecções Estafilocócicas/genética , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia , Staphylococcus aureus/patogenicidade , Linfócitos T Reguladores/microbiologia , Células Th17/microbiologia , Fator de Crescimento Transformador beta/genética , Fator de Necrose Tumoral alfa/genética
12.
Microb Pathog ; 115: 239-250, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29274459

RESUMO

Alarming increase of death due to S. aureus sepsis demands newer treatment strategies. Enhancement of antibiotic resistant S. aureus strains caused increased mortality. Only antibiotic treatment for Staphylococcal sepsis has been found insufficient to improve outcomes. In the innate immune response, phagocytosis mediated killing of pathogen and further triggering of intracellular signaling cascades by the PRRs culminates in the release of a variety of pro inflammatory cytokines, which orchestrate together in the early host response to infection. Increased production of inflammatory cytokines not only delineate pathogen burden but also affects host cell by triggering inflammation. Therefore, combinational therapy of Ascorbic acid is used along with antibiotics Ofloxacin (OFX) or Chloramphenicol (CHL) to kill S. aureus by mouse peritoneal macrophages. For this ROS like H2O2, superoxide anion and NO production was accessed, TLR2 and COX2 expression was monitored. Pro-inflammatory cytokines along with antioxidant levels were also analyzed. Ascorbic acid along with antibiotics OFX or CHL promoted bacterial clearance at early infection by increasing H2O2 and O2-.NO production has been found to decrease, providing protection against harmful per-oxynitril ion. Increase in TLR-2 expression resulted in enhanced phagocytosis and subsequently more killing. Treatment with Ascorbic acid decreased proinflammatory cytokines and inflammatory markers like iNOS and COX2. This combination increased antioxidant enzymes like SOD, Catalase, GSH as well as decreased LPO, thus balancing ROS and antioxidant status inside the cell. Thus in-vitro augmentation of bacterial clearance along with regulated inflammation as found by decrease in proinflammatory cytokines like TNF-α IFN-γ,IL-6 and inflammatory markers like COX2 may be considered as a novel and important therapeutic strategy.


Assuntos
Antibacterianos/farmacologia , Ácido Ascórbico/farmacologia , Cloranfenicol/farmacologia , Inflamação/imunologia , Macrófagos Peritoneais/imunologia , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/imunologia , Ofloxacino/farmacologia , Animais , Antioxidantes/análise , Ciclo-Oxigenase 2/biossíntese , Citocinas/imunologia , Citocinas/metabolismo , Quimioterapia Combinada , Peróxido de Hidrogênio/análise , Macrófagos Peritoneais/microbiologia , Masculino , Camundongos , Óxido Nítrico/biossíntese , Fagocitose/imunologia , Infecções Estafilocócicas/microbiologia , Superóxidos/análise , Receptor 2 Toll-Like/biossíntese
13.
Microb Pathog ; 105: 307-320, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28242423

RESUMO

Death due to sepsis by S. aureus is rapidly increasing because of their potent weaponries against macrophage mediated killing. Macrophages serve as intracellular reservoirs of S. aureus. Although significant resources have been invested during the last decade in new treatments for sepsis, only antibiotic therapy has failed to improve outcomes. Moreover the host pathogen interaction resulted in host cell death triggering inflammation. So, successful therapy requires amalgamation of therapies to delineate pathogen along with providing protection to host cell. With this idea, LNMMA, the iNOS inhibitor is used along with antibiotics Ofloxacin or Chloramphenicol on S. aureus infected mouse peritoneal macrophage. ROS like H2O2, O2- production has been measured. NO inhibition by iNOS inhibitor and antioxidant levels has been analysed. COX2, TLR2 and iNOS expression along with proinflammatory cytokine level was studied. It was found that the use of iNOS inhibitor LNMMA along with antibiotics not only enhances bacterial clearance but also decreases proinflammatory responses in Staphylococcus aureus infected macrophages. Inhibition of TLR2 as well as COX2 has also been found in combined treatment groups. The use of iNOS inhibitor LNMMA plus Ofloxacin or Chloramphenicol pretreatment enhanced bacterial clearance by increasing ROS. Decreases in NO protect the cell from harmful peroxynitril as well as inflammatory damage by changes in iNOS, COX2 activity along with reduced proinflammatory cytokines like TNFα, IFNγ, IL1-ß etc. Changes in antioxidant level has been found. This in-vitro realm of augmented bacterial clearance and regulated inflammation may be considered as a novel and important therapeutic intervention.


Assuntos
Cloranfenicol/farmacologia , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/microbiologia , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Ofloxacino/farmacologia , Staphylococcus aureus/efeitos dos fármacos , ômega-N-Metilarginina/farmacologia , Animais , Antibacterianos/farmacologia , Antioxidantes/metabolismo , Ciclo-Oxigenase 2/biossíntese , Citocinas/metabolismo , Modelos Animais de Doenças , Peróxido de Hidrogênio/metabolismo , Inflamação/tratamento farmacológico , Inflamação/enzimologia , Inflamação/imunologia , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Oxigênio/metabolismo , Distribuição Aleatória , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/imunologia , Staphylococcus aureus/patogenicidade , Superóxido Dismutase/metabolismo , Superóxidos/metabolismo , Receptor 2 Toll-Like/biossíntese
14.
Microb Pathog ; 113: 248-264, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29074430

RESUMO

Intravenous inoculation of Swiss mice with S. aureus leads to severe synovial joint tissue swelling along with prominent T lymphocyte infiltrate with associated inflammation in synovial tissue. Cytokines released from macrophages such as TNF-α, IL-1ß and IL-6 the main players that precede cartilage and bone destruction during septic arthritis (SA) followed by osteoclast differentiation and bone resorption. CD4+ naïve T cells upon cytokine driven activation, differentiate into lineages of helper (Th) and regulatory T cells (Treg) including inflammatory Th17 cell lineage. Acting as counterbalance, Tregs protect the host by releasing anti-inflammatory IL-10. A disturbed balance between Th17 and Treg cell development skews the pathways towards Th17 lineage, but how it actually induces SA is still unexplored. Therefore, this study has been attempted to demonstrate the Th17/Treg ratio in synovial tissue, spleen and peripheral blood by FACS and their derived cytokines from serum of arthritic mice. Here, we reported that the ratios of Th17/Treg as well as their related cytokine levels were increased at 3 days post-infection which was decreased during 9 DPI but heightened again at 15DPI resulting in persistence of the disease, though decreased again at 30 DPI even in animals with increased dose of infection. Bacterial colonies were present in synovial joints at 15 DPI in animals with increased infection but found to be absent at 30 DPI. Maintaining Th17/Treg balance by neutralizing functionally active Th17 and their related cytokines or adoptive transfer of fully active Tregs and/or their related cytokines may lead to a novel therapeutic strategy for combating Staphylococcal arthritis.


Assuntos
Artrite Infecciosa/imunologia , Artrite Infecciosa/microbiologia , Citocinas/metabolismo , Staphylococcus aureus/patogenicidade , Linfócitos T Reguladores/fisiologia , Células Th17/fisiologia , Animais , Artrite Infecciosa/sangue , Artrite Infecciosa/fisiopatologia , Reabsorção Óssea , Linfócitos T CD4-Positivos , Diferenciação Celular , Contagem de Colônia Microbiana , Modelos Animais de Doenças , Inflamação , Interleucina-10/metabolismo , Interleucina-17/metabolismo , Interleucina-23/metabolismo , Linfócitos , Macrófagos/imunologia , Macrófagos/microbiologia , Masculino , Camundongos , Osteoclastos , Baço/microbiologia , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Membrana Sinovial/microbiologia , Fator de Necrose Tumoral alfa/metabolismo
15.
Microb Pathog ; 113: 460-471, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29162483

RESUMO

Currently, very few studies are available on the expression of CXCR1 in mouse macrophages having both intact TNFR1 and IL-1R or their deficiency in relation to acute S. aureus infection. Peritoneal macrophages from mice neutralized singly for TNFR1or IL-1R, or for both TNFR1 and IL-1R were infected with S. aureus in vitro and their ability to secrete cytokines and reactive oxygen species (ROS) were determined. It was observed that the release of TNF-α and IL-1ß in response to S. aureus infection was decreased in macrophages when both TNFR1 and IL-1R were neutralized. The amount of H2O2, superoxide anion, nitric oxide release and bacterial CFU were significantly decreased in TNFR1 plus IL-1R blocked macrophages when compared with macrophages having intact receptors at 60 min of S. aureus infection. There was decrement of CXCL8 (IL-8) release and expression of CXCR1 in macrophages during dual receptor (TNFR1 plus IL-1R) blocking prior to stimulation with S. aureus. Expression of CXCR1 on murine peritoneal macrophages was evaluated by immunoblots from lysate at 60 min after S. aureus infection. It was observed that at 60 min after S. aureus infection in murine peritoneal macrophages, the expression of CXCR1 was increased significantly (p < 0.05) in comparison to the control groups. CXCR1 expression was decreased significantly (p < 0.05) in macrophages pre-incubated separately with anti-TNFR1 antibody (10 µg/ml) or IL-1R antagonist protein (240 ng/ml) at 60 min after S. aureus infection. However, blocking of both TNFR1 as well as IL-1R in macrophages downregulated the CXCR1expression in comparison to the groups either pre-incubated with anti-TNFR1 antibody or IRAP alone.


Assuntos
Macrófagos Peritoneais/imunologia , Receptores de Interleucina-1/metabolismo , Receptores de Interleucina-8A/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/patogenicidade , Animais , Anticorpos Bloqueadores , Contagem de Colônia Microbiana , Citocinas/metabolismo , Regulação da Expressão Gênica , Peróxido de Hidrogênio/metabolismo , Interleucina-1beta/metabolismo , Macrófagos Peritoneais/microbiologia , Masculino , Camundongos , Óxido Nítrico/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Superóxidos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
16.
Microb Pathog ; 109: 131-150, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28552636

RESUMO

Literature reveals that interaction with live Staphylococcus aureus (S. aureus) or heat killed S. aureus (HKSA) promotes secretion of CXCL-8 or interleukin-8 (IL-8) from leukocytes, however, the expressions of CXCR1 in murine splenic (SPM), peritoneal macrophages (PM) and resident fresh bone marrow cells (FBMC) have not been identified. Currently, very few studies are available on the functional characterization of CXCR1 in mouse macrophage subtypes and its modulation in relation to acute S. aureus infection. SPM, PM and FBMCs were infected with viable S. aureus or stimulated with HKSA in presence and absence of anti-CXCR1 antibody in this study. We reported here that CXCR1 was not constitutively expressed by macrophage subtypes and the receptor was induced only after S. aureus stimulation. The CXCR1 band was found specific as we compared with human polymorphonuclear neutrophils (PMNs) as a positive control (data not shown). Although, we did not show that secreted IL-8 from S. aureus-infected macrophages promotes migration of PMNs. Blocking of cell surface CXCR1 decreases the macrophage's ability to clear staphylococcal infection, attenuates proinflammatory cytokine production and the increased catalase and decreased superoxide dismutase (SOD) enzymes of the bacteria might indicate their role in scavenging macrophage derived hydrogen peroxide (H2O2). The decreased levels of cytokines due to CXCR1 blockade before S. aureus infection appear to regulate the killing of bacteria by destroying H2O2 and nitric oxide (NO). Moreover, functional importance of macrophage subpopulation heterogeneity might be important in designing new effective approaches to limit S. aureus infection induced inflammation and cytotoxicity.


Assuntos
Células da Medula Óssea/imunologia , Macrófagos Peritoneais/imunologia , Receptores de Interleucina-8/metabolismo , Baço/imunologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/imunologia , Animais , Células da Medula Óssea/microbiologia , Catalase/metabolismo , Citocinas/metabolismo , Temperatura Alta , Humanos , Peróxido de Hidrogênio/metabolismo , Inflamação , Interleucina-8/metabolismo , Macrófagos/imunologia , Macrófagos/microbiologia , Macrófagos Peritoneais/microbiologia , Camundongos , Neutrófilos/imunologia , Óxido Nítrico/metabolismo , Receptores de Interleucina-8/imunologia , Baço/microbiologia , Superóxido Dismutase/metabolismo , Superóxidos/metabolismo
17.
Microb Pathog ; 97: 131-47, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27270212

RESUMO

Staphylococcus aureus with high virulence potential is contributing to a current public health crisis in both hospital and community settings. TLR-2 and generation of reactive oxygen species (ROS) by phagocytic cells is thought to be an important component of the host's immunity against S. aureus infection. However, response of S. aureus against modulation of host-derived ROS in absence of TLR-2 during acute staphylococcal infection is still remains unclear. Peritoneal macrophages were pretreated with either inhibitors of superoxide dismutase (SOD) or catalase in presence or absence of anti TLR-2 antibody and were infected with S. aureus strain AG-789. Bacteria were recovered after time dependent phagocytosis; intracellular killing, level and expression of SOD and catalase were measured. Phagocytosed bacteria from respective groups were further used for infection to fresh peritoneal macrophages as well as for in vivo infection. Levels of ROS, cytokine, lysozyme, antioxidant enzymes activity and TLR-2 expression were measured. Results revealed that more bacteria were escaped killing in SOD and catalase inhibitor pretreated TLR-2 neutralized macrophages, found to express more catalase and are antibiotic resistant. Infection of fresh macrophages with S. aureus, recovered from SOD and catalase inhibited TLR-2 neutralized macrophages induced lower ROS, lysozyme and cytokine production and caused increased bacterial count. Furthermore, bacterial antioxidants by modulating host-derived ROS could regulate the cell surface TLR-2 expression in murine peritoneal macrophages. So, in the early phase of infection, TLR-2 participates in the innate immune response and targeting bacterial antioxidants might be useful in the alleviation of Staphylococcus aureus infection.


Assuntos
Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/microbiologia , Viabilidade Microbiana , Fagocitose , Staphylococcus aureus/imunologia , Staphylococcus aureus/patogenicidade , Receptor 2 Toll-Like/metabolismo , Animais , Antioxidantes/análise , Catalase/análise , Sobrevivência Celular , Citocinas/análise , Camundongos , Muramidase/análise , Espécies Reativas de Oxigênio/análise , Superóxido Dismutase/análise , Virulência
18.
Microb Pathog ; 99: 148-161, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27554276

RESUMO

Matrix metalloproteinases (MMPs) are crucial players in Staphylococcus aureus mediated synovial tissue destruction in the pathogenesis of septic arthritis. Bacterial insult increases proteolytic matrix fragments by activated chondrocytes and synovial fibroblasts leading to induction of matrix metalloproteinases. Tissue destruction via MMPs induced by bacterial products, necrotic tissues and proinflammatory cytokines have been reported. Cytokines like TNF-α, IL-1ß released from host cells in response to S. aureus infection promote cartilage degradation by stimulating the production of MMPs. Antibiotic treatment can eradicate invading bacteria but elevated levels of cytokines and cytokines induced MMPs activation lead to progressive and devastating bone and cartilage destruction even after bacterial clearance. Like other MMPs, MMP-2 also contributes to extracellular matrix degradation in different types of arthritis. Release of certain pro inflammatory cytokines can also be regulated by MMP-2 activation leading to further tissue destruction. The role of MMP-2 in the pathogenesis of S. aureus infection induced septic arthritis and its influence on cytokines regulation needs further investigation. Whether neutralization of MMP-2 provides protection against Staphylococcus aureus infection induced septic arthritis in mice is an obvious question. Here we reported that neutralization of MMP-2 during S. aureus infection induced septic arthritis might be beneficial for preventing infection induced extracellular matrix destruction thereby decreasing bacterial burden in synovial tissues and regulating inflammatory cytokines in arthritic mice.


Assuntos
Artrite Infecciosa/patologia , Citocinas/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Infecções Estafilocócicas/patologia , Staphylococcus aureus/crescimento & desenvolvimento , Animais , Modelos Animais de Doenças , Camundongos , Staphylococcus aureus/imunologia
19.
Microb Pathog ; 89: 114-27, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26416307

RESUMO

Staphylococcus aureus is an important pathogen in bone disease and innate immune recognition receptor, TLR-2 is reported to be crucial for inflammatory bone loss. Role of TLR-2 in bacterial clearance and cytokine response to S. aureus infection in murine bone marrow macrophages has been reported but the role of host derived ROS in host-pathogen relationship still remains an obvious question. In the present study, blocking of SOD and catalase in TLR-2 neutralized fresh bone marrow cells (FBMC) with Diethyldithiocarbamic acid (DDC) and 3-Amino-1,2,4-triazole (ATZ), separately, during acute S. aureus infection, produces moderate level of ROS and limits inflammation as compared with only TLR-2 non-neutralized condition and leads to decreased bacterial count compared with only TLR-2 neutralized condition. In summary, host SOD and catalase modulates ROS generation, cytokine levels and TLR-2 expression in FBMCs during acute S. aureus infection which might be useful in the alleviation of S. aureus infection and bone loss.


Assuntos
Antioxidantes/metabolismo , Catalase/metabolismo , Viabilidade Microbiana , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/imunologia , Superóxido Dismutase/metabolismo , Receptor 2 Toll-Like/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Interações Hospedeiro-Patógeno , Camundongos , Oxirredução , Staphylococcus aureus/fisiologia
20.
Indian J Exp Biol ; 53(2): 82-92, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25757238

RESUMO

Toll-like receptors (TLR) are a family of pattern recognition receptors identifying pathogen associated molecular patterns (PAMPs). They play a critical role in the innate immune response during the initial interaction between the infecting microorganism and phagocytic cells. Here, we verified the presence of TLR-2 in spleen, lymph node and thymus of Swiss albino mice and their modulation after infection with Staphylococcus aureus and Lipopolysaccharide (LPS) challenge. It was seen that TLR-2 gene transcribed to its respective mRNA on S. aureus infection, in thymus, spleen and lymph node of mice but their levels and mode of expression varied. When challenged with LPS no prominent changes in the expression of TLR-2 receptor was observed but its expression increased gradually with time in the thymus, spleen and lymph node of S. aureus infected mice. TLR-2 expression was also found enhanced in infected splenic macrophages. By studying the serum cytokine profile the functionality of the receptor was measured. The results indicate the presence of TLR-2 in thymus, spleen and lymph node of Swiss albino strain of mice and that they are modulated by S. aureus.


Assuntos
Lipopolissacarídeos/imunologia , Linfonodos/imunologia , Baço/imunologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/imunologia , Timo/imunologia , Receptor 2 Toll-Like/imunologia , Animais , Antibacterianos/farmacologia , Western Blotting , Citocinas/sangue , Citocinas/imunologia , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/imunologia , Interações Hospedeiro-Patógeno/imunologia , Lipopolissacarídeos/farmacologia , Linfonodos/metabolismo , Linfonodos/microbiologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Masculino , Camundongos , Testes de Sensibilidade Microbiana , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/metabolismo , Baço/microbiologia , Infecções Estafilocócicas/sangue , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/fisiologia , Timo/metabolismo , Timo/microbiologia , Fatores de Tempo , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA