Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Int Immunopharmacol ; 133: 112153, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38678669

ABSTRACT

LPS induced sepsis is a complex process involving various immune cells and signaling molecules. Dysregulation of macrophage polarization and ROS production contributed to the pathogenesis of sepsis. PGP is a transmembrane transporter responsible for the efflux of a number of drugs and also expressed in murine macrophages. Natural products have been shown to decrease inflammation and expression of efflux transporters. However, no treatment is currently available to treat LPS induced sepsis. Verapamil and Tangeretin also reported to attenuate lipopolysaccharide-induced inflammation. However, the effects of verapamil or tangeretin on lipopolysaccharide (LPS)-induced sepsis and its detailed anti-inflammatory mechanism have not been reported. Here, we have determined that verapamil and tangeretin protects against LPS-induced sepsis by suppressing M1 macrophages populations and also through the inhibition of P-glycoprotein expression via downregulating STAT1/STAT3 and upregulating SOCS3 expression in macrophages. An hour before LPS (10 mg/kg) was administered; mice were given intraperitoneal injections of either verapamil (5 mg/kg) or tangeretin (5 mg/kg). The peritoneal macrophages from different experimental groups of mice were isolated. Hepatic, pulmonary and splenic morphometric analyses revealed that verapamil and tangeretin decreased the infiltration of neutrophils into the tissues. Verapamil and tangeritin also enhanced the activity of SOD, CAT, GRX and GSH level in all the tissues tested. verapamil or tangeretin pre-treated mice shifted M1 macrophages to M2 type possibly through the inhibition of P-glycoprotein expression via downregulating STAT1/STAT3 and upregulating SOCS3 expression. Hence, both these drugs have shown protective effects in sepsis via suppressing iNOS, COX-2, oxidative stress and NF-κB signaling in macrophages. Therefore, in our study we can summarize that mice were treated with either Vera or Tan before LPS administration cause an elevated IL-10 by the macrophages which enhances the SOCS3 expression, and thereby able to limits STAT1/STAT3 inter-conversion in the macrophages. As a result, NF-κB activity is also getting down regulated and ultimately mitigating the adverse effect of inflammation caused by LPS in resident macrophages. Whether verapamil or tangeretin offers such protection possibly through the inhibition of P-glycoprotein expression in macrophages needs clarification with the bio availability of these drugs under PGP inhibited conditions is a limitation of this study.


Subject(s)
Flavones , Lipopolysaccharides , STAT1 Transcription Factor , STAT3 Transcription Factor , Suppressor of Cytokine Signaling 3 Protein , Verapamil , Animals , Verapamil/pharmacology , STAT1 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism , Suppressor of Cytokine Signaling 3 Protein/genetics , Flavones/pharmacology , Flavones/therapeutic use , Mice , STAT3 Transcription Factor/metabolism , Male , Sepsis/drug therapy , Sepsis/immunology , Sepsis/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Down-Regulation/drug effects , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/immunology , Cells, Cultured , Macrophages/drug effects , Macrophages/metabolism , Macrophages/immunology , Signal Transduction/drug effects , Up-Regulation/drug effects
2.
Immunobiology ; 229(1): 152767, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38103391

ABSTRACT

The lipopolysaccharide, a microbial toxin, is one of the major causative agents of sepsis. P-gp expression and its functions are altered during inflammation. LPS has been known to impair the functions of P-gp, an efflux transporter. But the effect of LPS on P-gp expression in murine peritoneal macrophages is poorly understood. Molecular docking studies reveal that vitexin is a potent substrate and verapamil a potent inhibitor of P-gp. In the present experimental study, the curative potential of vitexin as a fruit component and verapamil treated as a control inhibitor of P-gp was examined in a murine LPS sepsis model. The effects of vitexin and verapamil on P-gp expression in macrophages correlating with changes in macrophage polarization and associated functional responses during LPS induced sepsis were studied. Peritoneal macrophages of LPS (10 mg/kg body weight) challenged mice exhibited elevated levels of H2O2, superoxide, and NO in parallel with lower antioxidant activity. LPS treatment increased P-gp expression through increased TLR4/expression. However, LPS challenged mice treated with vitexin (5 mg/kg body weight) + verapamil (5 mg/kg body weight) showed higher anti-oxidant enzyme activity (SOD, CAT and GRx) resulting in reduced oxidative stress. This combination treatment also elevated TNFR2, concomitant with down-regulation of TLR4, NF-κB and P-gp expression in murine peritoneal macrophages, resulting in a switch from M1 to M2 polarisation of macrophages and reduced inflammatory responses. In conclusion, combined vitexin and verapamil treatment could be used as a promising therapy to regulate P-gp expression and protection against LPS mediated sepsis and inflammatory damages.


Subject(s)
Apigenin , NF-kappa B , Sepsis , Mice , Animals , NF-kappa B/metabolism , Lipopolysaccharides/pharmacology , Toll-Like Receptor 4/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Receptors, Tumor Necrosis Factor, Type II/pharmacology , Verapamil/pharmacology , Hydrogen Peroxide/metabolism , Molecular Docking Simulation , Macrophages/metabolism , Glycoproteins/metabolism , Sepsis/drug therapy , ATP Binding Cassette Transporter, Subfamily B/pharmacology , Body Weight
3.
Microb Pathog ; 187: 106518, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38160988

ABSTRACT

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.


Subject(s)
Arthritis, Infectious , Methicillin-Resistant Staphylococcus aureus , Humans , Tumor Necrosis Factor-alpha/metabolism , Toll-Like Receptor 2/metabolism , Staphylococcus aureus/metabolism , Methicillin-Resistant Staphylococcus aureus/metabolism , Cytokines/metabolism , NF-kappa B/metabolism , Macrophages/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism , STAT3 Transcription Factor/metabolism
4.
Cell Mol Neurobiol ; 44(1): 2, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38099973

ABSTRACT

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.


Subject(s)
COVID-19 , Depressive Disorder, Major , Humans , SARS-CoV-2 , Microglia , Neuroinflammatory Diseases , Cytokines
5.
Open Life Sci ; 18(1): 20220718, 2023.
Article in English | MEDLINE | ID: mdl-37772260

ABSTRACT

It is essential to revisit the global biodiversity, search for ethnopharmacologically relevant plants, and unveil their untapped potential to overcome the complications associated while treating infections triggered by multiple antibiotic-resistant Staphylococcus aureus. Catharanthus roseus (L.) G. Don of the Apocynaceae family is a medicinal plant used for remedial purposes against infectious diseases from ancient times. In this study, we intended to evaluate the mechanism by which the ethanolic extract of C. roseus root (EECRR) causes the reversal of ampicillin resistance in S. aureus. To achieve this goal, we have stained EECRR-treated S. aureus with acridine orange, analysed DNA damage by comet assay, and studied the alteration of plasmid band pattern and expression of penicillin-binding protein 2a (PBP2a) protein. Experiments revealed better S. aureus killing efficiency of EECRR at its minimum inhibitory concentration (MIC) doses due to DNA damage and reducing plasmid band intensities along with a decline in the expression of PBP2a in EECRR-treated cells at half-MIC dose. EECRR proved to be an efficient growth inhibitor of S. aureus that reduces the expression of PBP2a. Therefore, EECRR can also render ampicillin-resistant S. aureus susceptible to the antibiotic.

6.
Microb Pathog ; 183: 106294, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37567327

ABSTRACT

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.


Subject(s)
Macrophages, Peritoneal , Staphylococcal Infections , Mice , Animals , Macrophages, Peritoneal/microbiology , Staphylococcus aureus/metabolism , omega-N-Methylarginine/metabolism , Antioxidants/pharmacology , Antioxidants/metabolism , Reactive Oxygen Species/metabolism , NF-kappa B/metabolism , Hydrogen Peroxide/metabolism , Arginase/metabolism , Cytokines/metabolism , Staphylococcal Infections/microbiology , Receptors, Interleukin-8A/metabolism , Inflammation/metabolism
7.
Int Immunopharmacol ; 123: 110766, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37572502

ABSTRACT

Researches of recent past years have emphasized potential of antibiotics to improve septic arthritis but as multi-drug resistant strains like MRSA are emerging fast, new alternative therapeutic advances are high in demand. This study aims to figure out the role of neutrophils in regulating inflammatory responses of S. aureus induced septic arthritis while using TNF-α Ab or IL-1ß Ab along with antibiotic gentamicin or both in combination. In this study, role of anti-oxidant enzymes were investigated and correlated with generated ROS level. While expression of TLR2, TNFR2, MMP2, RANKL, SAPK/JNK in the spleen were evaluated through western blot. Serum activity of IL-8, IL-10, IL-12, OPG, OPN, CRP was assessed using ELISA. Flow cytometry study evaluated inflamed neutrophil population. Results have shown TNF-α neutralization along with gentamicin was able to reduce arthritic swelling prominently. While combination therapy effectively reduced blood neutrophil ROS activity, arginase activity, MPO activity along with spleen bacterial burden. Serum OPG, CRP, IL-10 level got reduced while serum OPN, IL-8 and IL-12 level enhanced in treatment groups, showing mitigation of inflammatory damage. Overall, it is a novel work that observed how antibiotic and antibody therapy enhanced neutrophil function positively to combat sepsis. This study may not be fully applicable in clinical trials as it is performed with animal model. Clinical trials include crystalline and inflammatory arthritides, trauma, neoplasm. Interdisciplinary collaboration between radiology, orthopaedic surgery and knowledge of animal system responses may give better idea to find proper therapeutic approach in future research works.


Subject(s)
Arthritis, Infectious , Methicillin-Resistant Staphylococcus aureus , Animals , Tumor Necrosis Factor-alpha , Interleukin-10/metabolism , Neutrophils/metabolism , Staphylococcus aureus/physiology , Cytokines/metabolism , Spleen/metabolism , Gentamicins/therapeutic use , Interleukin-8 , Reactive Oxygen Species , Arthritis, Infectious/drug therapy , Interleukin-12 , Anti-Bacterial Agents/therapeutic use
8.
Int Immunopharmacol ; 120: 110297, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37207443

ABSTRACT

Overexpression of Staphylococcus aureus mediated CXCL8/CXCR1 axis is a major cause of sepsis and severe inflammatory diseases. This chemokine acts conjointly with various pro-inflammatory and anti-inflammatory cytokines that govern the severity of inflammation. The effects of different combinations of exogenous cytokines on CXCR1 expression in macrophages remain undetermined. Exogenous cytokine and anti-inflammatory cytokine therapy had been used to modulate CXCL8 and CXCR1 expression in peritoneal macrophages. Male Swiss albino mice were inoculated with live S. aureus (106 cells/ mouse) for the development of infection. Exogenous cytokines (TNF-α, IL-12, IFN-γ and IL-10) were administered intraperitoneally (single or combination) 24 h post S. aureus infection. The mice were sacrificed and peritoneal macrophages were isolated three days post infection. CXCL8, IL-12, IL-10 secretion, ROS generation and the bacterial phagocytic process had been evaluated. Western blot was used to study the expressions of TNFR1, IL-1R, CXCR1 and NF-κB. TNF-α, IL-12 and IFN-γ treatments aggravated CXCL8 and CXCR1 expression in the macrophages of infected mice. TNF-α + IFN-γ treatment was a major inducer of nitric oxide release and mediated maximum bacterial killing. IL-12 + TNF-α treatment was most potent in increasing ROS, CXCL8/CXCR1 expression through increased levels of TNFR1, IL-1R and NF-κB activation. IL-10 reversed the effects of exogenous cytokines but also impaired the bacterial clearance phenomenon in peritoneal lavage. Treatment with IL-12 + TNF-α + IL-10 was most effective in ameliorating oxidative stress, reduced CXCL8 release and expression levels of TNFR1, IL-1R, and NF-κB. Concludingly, IL-12 + TNF-α + IL-10 treatment mitigated CXCL8/CXCR1 expression and inflammatory signalling via downregulation of TNFR1-IL-1R-NF-κB pathway in peritoneal macrophages and inflammatory sequelae during S. aureus infection.


Subject(s)
Receptors, Tumor Necrosis Factor, Type I , Staphylococcal Infections , Animals , Male , Mice , Cytokines/metabolism , Interleukin-10 , Interleukin-12/therapeutic use , Macrophages, Peritoneal/metabolism , NF-kappa B , Reactive Oxygen Species , Receptors, Interleukin-8A/metabolism , Receptors, Tumor Necrosis Factor, Type I/therapeutic use , Receptors, Tumor Necrosis Factor, Type I/metabolism , Staphylococcal Infections/metabolism , Staphylococcus aureus/metabolism , Tumor Necrosis Factor-alpha
9.
J Appl Toxicol ; 43(10): 1549-1572, 2023 10.
Article in English | MEDLINE | ID: mdl-37177863

ABSTRACT

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.


Subject(s)
Antioxidants , Sepsis , Male , Mice , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Antioxidants/metabolism , Interleukin-10 , Toll-Like Receptor 4/metabolism , Lipopolysaccharides/toxicity , Sepsis/drug therapy , Sepsis/metabolism , Macrophages , Tumor Necrosis Factor-alpha/metabolism , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use
10.
Scand J Immunol ; 97(6): e13252, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36597222

ABSTRACT

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.


Subject(s)
Arthritis, Infectious , Interleukin-10 , Animals , Mice , Staphylococcus aureus , Transforming Growth Factor beta , Interleukin-6 , Spleen/pathology , Antioxidants , Inflammation , Arthritis, Infectious/pathology , Macrophages/pathology
11.
Int Immunopharmacol ; 115: 109654, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36621328

ABSTRACT

Rheumatoid arthritis (RA) primarily affecting the synovial tissue, has emerged as a major concern leading to the pressing need to develop effective treatment strategies. In the affected synovial tissue, resident macrophages play a pivotal role in the pathogenesis of RA. TNF-α and IL-1ß released from pro-inflammatory M1 synovial macrophages are the master regulators of chronic joint inflammation. In this study collagen-induced rheumatoid arthritis model was developed in mice and post isolation, macrophages were subjected to administration with neutralizing antibodies IL1R and TNFR1 either alone or in combination. Flow cytometric analysis followed by Western blots, ROS, and IL-1ß, TNF-α release assays were performed. Outcomes suggested that post-dual blockade of IL1R and TNFR1 arthritic synovial macrophages showed a shifting of the M1 towards the anti-inflammatory M2 phenotype. Moreover, the switch towards the M2 phenotype might be responsible for decreased levels of IL-1ß,TNF-α, and ROS and simultaneous elevation in the activity of antioxidant enzymes like SOD, CAT, and GPX content in the isolated macrophages. Simultaneous blocking of both IL1R and TNFR1 also showed a sharp reduction in the expression of NF-κB and SAPK-JNK. The elevated arginase and GRX activity further confirmed the polarization towards M2. Moreover, bioinformatics analysis was performed,and it was found that blocking TNFR1 with an antibody could hamper the binding of TNF to TNFR1 in the TNF-TNFR1 pathway. Thus, it may be inferred that dual blockade of IL1R and TNFR1 and a suitable antibody blocking of TNFR1 might be alternative therapeutic approaches for the regulation of RA-induced inflammation in the future.


Subject(s)
Arthritis, Rheumatoid , Receptors, Tumor Necrosis Factor, Type I , Animals , Mice , Antibodies/pharmacology , Inflammation/metabolism , Macrophages , Reactive Oxygen Species/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factor-alpha/metabolism , Synovial Membrane/cytology , Synovial Membrane/metabolism , Receptors, Interleukin-1/metabolism
12.
Mol Immunol ; 152: 183-206, 2022 12.
Article in English | MEDLINE | ID: mdl-36371814

ABSTRACT

Septic arthritis is an inflammatory joint disease caused by S. aureus. Hematogenous entry of the bacteria to the synovium produces pro-inflammatory cytokines TGF-ß and IL-6, which alter the Th17/Treg balance. Hence, targeting TGF-ß and IL-6 could be beneficial in ameliorating arthritis. Antibody neutralization of TGF-ß and IL-6 to modulate Th17/Treg homeostasis and RANKL/OPG ratio are not investigated so far in S. aureus-induced septic arthritis. Contribution of synovial lymphocyte-derived cytokines IL-10, IL-12, and CXCL-8; along with OPN, OPG, CRP, cellular ROS, antioxidant enzymes, and the expressions of RANKL, SAPK-JNK, MMP2, SOD, CAT, GPx, TGF-ß and IL-6 were studied in lymphocytes of blood, spleen and synovial tissues of mice treated with antibody against of TGF-ß and IL-6 after induction of septic arthritis. Dual neutralization of TGF-ß and IL-6 is effective in shifting the Th17 cell into immunosuppressive Treg cell of the arthritic mice and enhances the RANKL/OPG interaction leading to the down-regulation of osteoclastic activity and reduces the production of OPN, IL-12, CXCL-8, and CRP. Additionally, it reduces oxidative stress via enhancing the activities of antioxidant enzymes including SOD, catalase, and GPx in lymphocytes. Thus it can be concluded that dual endogenous neutralization of TGF-ß and IL-6 may be chosen as an alternative therapeutic approach for controlling the severity of septic arthritis through Treg-derived IL-10 that could ameliorate the inflammatory consequences of septic arthritis via influencing RANKL/OPG interaction in lymphocytes.


Subject(s)
Arthritis, Infectious , Interleukin-10 , Mice , Animals , Interleukin-10/metabolism , Staphylococcus aureus , Transforming Growth Factor beta , Interleukin-6 , Antioxidants/pharmacology , Arthritis, Infectious/microbiology , Th17 Cells , Cytokines/metabolism , Interleukin-12/pharmacology , Superoxide Dismutase
13.
Inflammopharmacology ; 30(4): 1303-1322, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35704229

ABSTRACT

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.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcus aureus , Antioxidants/metabolism , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Ciprofloxacin/metabolism , Ciprofloxacin/pharmacology , Hydrogen Peroxide/metabolism , Methicillin-Resistant Staphylococcus aureus/metabolism , Microglia , Oxidative Stress , Receptors, Glucocorticoid/metabolism
14.
Cell Immunol ; 370: 104441, 2021 12.
Article in English | MEDLINE | ID: mdl-34628221

ABSTRACT

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.


Subject(s)
Arthritis, Infectious/immunology , Interleukin-17/antagonists & inhibitors , Interleukin-2/pharmacology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Antioxidants/metabolism , Arthritis, Experimental/immunology , Interleukin-17/immunology , Male , Mice , Reactive Oxygen Species/immunology , Receptors, Tumor Necrosis Factor/immunology , T-Lymphocytes, Regulatory/drug effects , Th17 Cells/drug effects , Toll-Like Receptor 2/immunology
15.
Immunol Res ; 69(4): 334-351, 2021 08.
Article in English | MEDLINE | ID: mdl-34235623

ABSTRACT

The Gram-negative bacterial lipopolysaccharide (LPS)-induced sepsis has emerged as major concern worldwide due to the pressing need to develop its effective treatment strategies which is not available yet. LPS is the major causative agent in the pathogenesis of septic shock. In macrophages, LPS interacts with cell surface TLR4 leading to reactive oxygen species (ROS), TNF-α, IL-1ß production, oxidative stress and markedly activated the MAPKs and NF-kB pathway. Post cell isolation, the macrophages were subjected to administration with neutralizing antibodies to TLR4 and TNFR1 either alone or in combination prior to LPS challenge. Subsequently, we performed flow cytometric analysis along with Western blots, reactive oxygen species production, and TNF-α, IL-1ß release. Outcomes suggested that the dual blockade of TLR4 and TNFR1 was indeed beneficial in shifting the LPS-induced M1 polarization towards M2. Both TLR4 and TNFR1 exhibited dependency during LPS stimulation. Furthermore, the switch towards the M2 phenotype might be responsible for the decreased levels of TNF-α, IL-1ß, NO, and superoxide anion and the simultaneous elevation in the activity level of anti-oxidant enzymes like SOD, CAT (catalase), and GSH content in the isolated peritoneal macrophages. Simultaneous blocking of both TLR4 and TNFR1 also showed reduced expression of NF-kB, JNK, and COX-2 by promoting TNFR2-mediated TNF-α signaling. The increased arginase activity further confirmed the polarization towards M2. Thus it may be inferred that dual blockade of TLR4 and TNFR1 might be an alternative therapeutic approach for regulating of sepsis in future.


Subject(s)
Antibodies, Neutralizing/pharmacology , Macrophages, Peritoneal/drug effects , Receptors, Immunologic/antagonists & inhibitors , Animals , Arginase/metabolism , Free Radicals/metabolism , Glutathione/metabolism , Interleukin-1beta/metabolism , Lipopolysaccharides , Macrophage Activation/drug effects , Macrophages, Peritoneal/metabolism , Male , Mice , Oxidoreductases/metabolism , Phenotype , Receptors, Immunologic/metabolism , Tumor Necrosis Factor-alpha/metabolism
16.
Int Immunopharmacol ; 97: 107695, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33962227

ABSTRACT

Staphylococcus aureus induced brain abscess is a critical health concern throughout the developing world. The conventional surgical intervention could not regulate the abscess-induced brain inflammation. Thus further study over the alternative therapeutic strategy for treating a brain abscess is of high priority. The resident glial cells recognize the invading S. aureus by their cell surface Toll-like receptor-2 (TLR-2). Glucocorticoid receptor (GR) was known for its immunosuppressive effects. In this study, an attempt had been taken to utilize the functional relationship or cross-talking between TLR-2 and GR during the pathogenesis of brain abscesses. Here, the combination of an antibiotic (i.e. ciprofloxacin) and dexamethasone was used to regulate the brain inflammation either in TLR-2 or GR blocking condition. We were also interested to figure out the possible impact of alternative therapy on behavioral impairments. The results indicated that combination treatment during TLR-2 blockade significantly reduced the bacterial burden and abscess area score in the infected brain. However, marked improvements were observed in anxiety, depression-like behavior, and motor co-ordination. The combination treatment after TLR-2 blocking effectively scavenged free radicals (H2O2, superoxide anion, and NO) through modulating antioxidant enzyme activities that ultimately control S. aureus induced glial reactivity possibly via up-regulating GR expression. The exogenous dexamethasone might regulate the GR expression in the brain by increasing the corticosterone concentration and the GC-GR mediated signaling. Therefore, this in-vivo study demonstrates the possible regulatory mechanism of bacterial brain abscess that involved TLR-2 and GR as a part of neuroendocrine-immune interaction.


Subject(s)
Brain Abscess/drug therapy , Ciprofloxacin/pharmacology , Dexamethasone/pharmacology , Neuroinflammatory Diseases/drug therapy , Staphylococcal Infections/drug therapy , Animals , Behavior, Animal/drug effects , Brain Abscess/complications , Brain Abscess/immunology , Brain Abscess/microbiology , Ciprofloxacin/therapeutic use , Dexamethasone/therapeutic use , Disease Models, Animal , Drug Therapy, Combination , Humans , Male , Mice , Neuroinflammatory Diseases/diagnosis , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/microbiology , Neurosecretory Systems/drug effects , Neurosecretory Systems/immunology , Receptors, Glucocorticoid/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Staphylococcal Infections/complications , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/immunology , Toll-Like Receptor 2/metabolism
17.
Immunol Invest ; 50(6): 700-725, 2021 Aug.
Article in English | MEDLINE | ID: mdl-32602757

ABSTRACT

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.


Subject(s)
Immunosuppressive Agents/pharmacology , Interleukin-1beta/antagonists & inhibitors , Interleukin-8/metabolism , Staphylococcal Infections/drug therapy , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Bacterial Load/drug effects , Cells, Cultured , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/immunology , Humans , Immunosuppressive Agents/therapeutic use , Interleukin-1beta/metabolism , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/microbiology , Male , Mice , Primary Cell Culture , Receptors, Interleukin-8A/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcus aureus/immunology , Tumor Necrosis Factor-alpha/metabolism
18.
Exp Mol Pathol ; 116: 104485, 2020 10.
Article in English | MEDLINE | ID: mdl-32574668

ABSTRACT

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.


Subject(s)
Arthritis, Infectious/immunology , Inflammation/immunology , Staphylococcal Infections/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/immunology , Arthritis, Experimental/microbiology , Arthritis, Experimental/pathology , Arthritis, Infectious/genetics , Arthritis, Infectious/microbiology , Arthritis, Infectious/pathology , Gene Expression Regulation/genetics , Humans , Inflammation/genetics , Inflammation/microbiology , Inflammation/pathology , Interleukin-1beta/genetics , Joints/immunology , Joints/microbiology , Joints/pathology , MAP Kinase Kinase 4/genetics , Mice , Osteoclasts/immunology , Osteoclasts/microbiology , Osteoclasts/pathology , RANK Ligand/genetics , Signal Transduction/genetics , Staphylococcal Infections/genetics , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/pathogenicity , T-Lymphocytes, Regulatory/microbiology , Th17 Cells/microbiology , Transforming Growth Factor beta/genetics , Tumor Necrosis Factor-alpha/genetics
19.
Microb Pathog ; 145: 104227, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32360190

ABSTRACT

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.


Subject(s)
Ciprofloxacin , Dexamethasone , Glucocorticoids , Microglia , Receptors, Glucocorticoid , Animals , Ciprofloxacin/pharmacology , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Inflammation , Mice , Microglia/drug effects , Microglia/microbiology , Staphylococcus aureus
20.
J Neuroimmunol ; 344: 577262, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32450430

ABSTRACT

Microglial inflammation plays a pivotal role in the pathogenesis of S. aureus induced brain abscesses. The objective of this study was to regulate microglial activation by the combinatorial treatment of ciprofloxacin either with dexamethasone or celecoxib via targeting M1 and M2 polarization. The antibiotic-immunomodulator combinations were applied either by opening both TLR-2 and GR or neutralizing each of them. Our results confirmed that dexamethasone along with ciprofloxacin attenuated bacterial burden along with ROS production more efficiently than celecoxib combination during TLR-2 neutralization. FACS data indicated microglial M1 to M2 switching that was responsible for the better resolution of microglial inflammation.


Subject(s)
Ciprofloxacin/administration & dosage , Dexamethasone/administration & dosage , Microglia/metabolism , Staphylococcal Infections/metabolism , Staphylococcus aureus , Toll-Like Receptor 2/metabolism , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Inflammatory Agents/administration & dosage , Cells, Cultured , Drug Therapy, Combination , Male , Mice , Microglia/drug effects , Staphylococcal Infections/drug therapy , Toll-Like Receptor 2/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...