Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Sci Transl Med ; 15(686): eabl4414, 2023 03 08.
Article in English | MEDLINE | ID: mdl-36888695

ABSTRACT

Natural killer (NK) cells are cytotoxic lymphocytes that accumulate within the tumor microenvironment and are generally considered to be antitumorigenic. Using single-cell RNA sequencing and functional analysis of multiple triple-negative breast cancer (TNBC) and basal tumor samples, we observed a unique subcluster of Socs3highCD11b-CD27- immature NK cells that were present only in TNBC samples. These tumor-infiltrating NK cells expressed a reduced cytotoxic granzyme signature and, in mice, were responsible for activating cancer stem cells through Wnt signaling. NK cell-mediated activation of these cancer stem cells subsequently enhanced tumor progression in mice, whereas depletion of NK cells or Wnt ligand secretion from NK cells by LGK-974 decreased tumor progression. In addition, NK cell depletion or inhibition of their function improved anti-programmed cell death ligand 1 (PD-L1) antibody or chemotherapy response in mice with TNBC. Furthermore, tumor samples from patients with TNBC and non-TNBC revealed that increased numbers of CD56bright NK cells were present in TNBC tumors and were correlated to poor overall survival in patients with TNBC. Together, our findings identify a population of protumorigenic NK cells that may be exploited for both diagnostic and therapeutic strategies to improve outcomes for patients with TNBC.


Subject(s)
Antineoplastic Agents , Triple Negative Breast Neoplasms , Humans , Animals , Mice , Triple Negative Breast Neoplasms/drug therapy , Antineoplastic Agents/therapeutic use , Killer Cells, Natural , B7-H1 Antigen/metabolism , Tumor Microenvironment
2.
Cancer Res ; 82(20): 3718-3733, 2022 10 17.
Article in English | MEDLINE | ID: mdl-36007109

ABSTRACT

SIGNIFICANCE: Dll1+ breast cancer cells activate Notch signaling in cancer-associated fibroblasts that increases Wnt ligand secretion and leads to ß-catenin-driven radioresistance and metastasis, opening new therapeutic avenues for breast cancer.


Subject(s)
Breast Neoplasms , Cancer-Associated Fibroblasts , Breast Neoplasms/pathology , Breast Neoplasms/radiotherapy , Cancer-Associated Fibroblasts/pathology , Female , Humans , Intercellular Signaling Peptides and Proteins , Ligands , Receptors, Notch , beta Catenin
4.
Methods Mol Biol ; 2429: 485-500, 2022.
Article in English | MEDLINE | ID: mdl-35507183

ABSTRACT

Evidence is emerging that cancer cells are arranged as a hierarchy that spans from stem cells to lineage-restricted progenitor cells. The recent development of spheroid cultures with several tissue type has provided new opportunities to assess cancer stem cell (CSC) activity by allowing them to propagate under conditions that resemble the microenvironment for growth of tumors. One tissue type widely used for stem cell investigations is mammary tissue, and the sphere formation assay has been used in both normal mammary tissue and in breast cancer. Here, we describe detailed experimental methodology for generating and propagating spheres from normal mammary tissue and primary breast tumors of mice, patient derived xenografts (PDXs) and breast cancer cell lines. We further describe how these sphere cultures can be employed for coculture assays to assess the effect of tumor microenvironment (TME) on self-renewal ability of CSCs in breast cancer.


Subject(s)
Breast Neoplasms , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Mice , Neoplastic Stem Cells/metabolism , Tumor Microenvironment
6.
Nat Commun ; 12(1): 432, 2021 01 18.
Article in English | MEDLINE | ID: mdl-33462238

ABSTRACT

Development of chemoresistance in breast cancer patients greatly increases mortality. Thus, understanding mechanisms underlying breast cancer resistance to chemotherapy is of paramount importance to overcome this clinical challenge. Although activated Notch receptors have been associated with chemoresistance in cancer, the specific Notch ligands and their molecular mechanisms leading to chemoresistance in breast cancer remain elusive. Using conditional knockout and reporter mouse models, we demonstrate that tumor cells expressing the Notch ligand Dll1 is important for tumor growth and metastasis and bear similarities to tumor-initiating cancer cells (TICs) in breast cancer. RNA-seq and ATAC-seq using reporter models and patient data demonstrated that NF-κB activation is downstream of Dll1 and is associated with a chemoresistant phenotype. Finally, pharmacological blocking of Dll1 or NF-κB pathway completely sensitizes Dll1+ tumors to chemotherapy, highlighting therapeutic avenues for chemotherapy resistant breast cancer patients in the near future.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Calcium-Binding Proteins/metabolism , Drug Resistance, Neoplasm/genetics , Membrane Proteins/metabolism , NF-kappa B p50 Subunit/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzamides/pharmacology , Benzamides/therapeutic use , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Datasets as Topic , Disease Models, Animal , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm/drug effects , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Knockout , NF-kappa B p50 Subunit/antagonists & inhibitors , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , RNA-Seq , Receptors, Notch/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
7.
Nat Cell Biol ; 22(5): 591-602, 2020 05.
Article in English | MEDLINE | ID: mdl-32284542

ABSTRACT

Triple-negative breast cancer (TNBC) is characterized by a high degree of immune infiltrate in the tumour microenvironment, which may influence the fate of TNBC cells. We reveal that loss of the tumour suppressive transcription factor Elf5 in TNBC cells activates intrinsic interferon-γ (IFN-γ) signalling, promoting tumour progression and metastasis. Mechanistically, we find that loss of the Elf5-regulated ubiquitin ligase FBXW7 ensures stabilization of its putative protein substrate IFN-γ receptor 1 (IFNGR1) at the protein level in TNBC. Elf5low tumours show enhanced IFN-γ signalling accompanied by an increase of immunosuppressive neutrophils within the tumour microenvironment and increased programmed death ligand 1 expression. Inactivation of either programmed death ligand 1 or IFNGR1 elicited a robust anti-tumour and/or anti-metastatic effect. A positive correlation between ELF5 and FBXW7 expression and a negative correlation between ELF5, FBXW7 and IFNGR1 expression in the tumours of patients with TNBC strongly suggest that this signalling axis could be exploited for patient stratification and immunotherapeutic treatment strategies for Elf5low patients with TNBC.


Subject(s)
Cell Proliferation/physiology , DNA-Binding Proteins/metabolism , F-Box-WD Repeat-Containing Protein 7/metabolism , Interferon-gamma/metabolism , Neoplasm Metastasis/pathology , Receptors, Interferon/metabolism , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/metabolism , Animals , Cell Line , Cell Line, Tumor , Female , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Signal Transduction/physiology , Tumor Microenvironment/physiology , Interferon gamma Receptor
8.
Genes Dev ; 34(21-22): 1422-1438, 2020 11 01.
Article in English | MEDLINE | ID: mdl-33872192

ABSTRACT

Breast cancer is the second leading cause of cancer-related death in women and is a complex disease with high intratumoral and intertumoral heterogeneity. Such heterogeneity is a major driving force behind failure of current therapies and development of resistance. Due to the limitations of conventional therapies and inevitable emergence of acquired drug resistance (chemo and endocrine) as well as radio resistance, it is essential to design novel therapeutic strategies to improve the prognosis for breast cancer patients. Deregulated Notch signaling within the breast tumor and its tumor microenvironment (TME) is linked to poor clinical outcomes in treatment of resistant breast cancer. Notch receptors and ligands are also important for normal mammary development, suggesting the potential for conserved signaling pathways between normal mammary gland development and breast cancer. In this review, we focus on mechanisms by which Notch receptors and ligands contribute to normal mammary gland development and breast tumor progression. We also discuss how complex interactions between cancer cells and the TME may reduce treatment efficacy and ultimately lead to acquired drug or radio resistance. Potential combinatorial approaches aimed at disrupting Notch- and TME-mediated resistance that may aid in achieving in an improved patient prognosis are also highlighted.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/physiopathology , Breast Neoplasms/therapy , Drug Resistance, Neoplasm/genetics , Receptors, Notch/metabolism , Signal Transduction/physiology , Antineoplastic Agents/pharmacology , Humans
9.
Arch Physiol Biochem ; 126(4): 348-355, 2020 Oct.
Article in English | MEDLINE | ID: mdl-30468085

ABSTRACT

Context: Alteration of redox signalling and RANK-L expression in FBMCs of mice exposed to different intensities of cold stress (15 °C, 8 °C and 4 °C) were studied.Objective: To understand the effects of varying intensities of cold stress on murine FBMCs and its impact on osteoclastogenesis.Materials and methods: FBMCs were isolated from mice exposed to different intensities of cold stress and used for immunoblotting and biochemical assays. Bone histometry was also done.Results: Different intensities of cold stress perturb redox signalling in FBMCs and alters bone histometry. Higher RANK-L expressions were noted in FBMCs of mice exposed to 8 °C and 4 °C as compared with 15 °C.Discussion and conclusion: Cold stress boosts free radical production in FBMC's, which might enhance RANK-L expression, an indicator of osteoclastogenesis. Thus, we speculate that stronger cold stress (8 °C and 4 °C) contributes to the development of early bone loss.


Subject(s)
Bone Marrow Cells/cytology , Cold-Shock Response , Osteoclasts/cytology , Signal Transduction , Animals , Female , Mice , Nitric Oxide/biosynthesis , Osteoclasts/metabolism , Oxidation-Reduction
10.
FEBS Lett ; 594(6): 973-985, 2020 03.
Article in English | MEDLINE | ID: mdl-31794060

ABSTRACT

The ∆Np63 isoform of the p53-family transcription factor Trp63 is a key regulator of mammary epithelial stem cells that is involved in breast cancer development. To investigate the role of ∆Np63 at different stages of normal mammary gland development, we generated a ∆Np63-inducible conditional knockout (cKO) mouse model. We demonstrate that the deletion of ∆Np63 at puberty results in depletion of mammary stem cell-enriched basal cells, reduces expression of E-cadherin and ß-catenin, and leads to a closed ductal lumen. RNA-sequencing analysis reveals reduced expression of oxidative phosphorylation (OXPHOS)-associated proteins and desmosomal polarity proteins. Functional assays show reduced numbers of mitochondria in the mammary epithelial cells of ΔNp63 cKO compared to wild-type, supporting the reduced OXPHOS phenotype. These findings identify a novel role for ∆Np63 in cellular metabolism and mammary epithelial cell polarity.


Subject(s)
Cell Polarity , Mammary Glands, Animal/metabolism , Sexual Maturation , Stem Cells/metabolism , Trans-Activators/metabolism , Animals , Cadherins/genetics , Cadherins/metabolism , Female , Mice , Mice, Knockout , Trans-Activators/genetics , beta Catenin/genetics , beta Catenin/metabolism
12.
Microb Pathog ; 113: 460-471, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29162483

ABSTRACT

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.


Subject(s)
Macrophages, Peritoneal/immunology , Receptors, Interleukin-1/metabolism , Receptors, Interleukin-8A/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Staphylococcal Infections/immunology , Staphylococcus aureus/pathogenicity , Animals , Antibodies, Blocking , Colony Count, Microbial , Cytokines/metabolism , Gene Expression Regulation , Hydrogen Peroxide/metabolism , Interleukin-1beta/metabolism , Macrophages, Peritoneal/microbiology , Male , Mice , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Superoxides/metabolism , Tumor Necrosis Factor-alpha/metabolism
13.
Immunol Lett ; 190: 93-107, 2017 10.
Article in English | MEDLINE | ID: mdl-28736159

ABSTRACT

Macrophages are remarkably versatile in their ability to recognize and respond to a wide range of stimuli by expressing a variety of surface and intracellular receptors and triggering multiple signal transduction pathways. The onset of microbial infection is primarily determined by the initial contacts made by the microbes with the host macrophages. Although there prevail a relationship between the chemokine receptor and Toll like receptors during disease, particularly TLR-2 and CCR-2 signaling interdependence on each other has not been yet investigated during acute staphylococcal infection. Thus, the present study was aimed to trace possible interaction between CCR-2 and TLR-2 in peritoneal macrophages during acute Staphylococcus aureus infection. We found that neutralization of CCR-2 attenuates TLR-2 expression and restricts S. aureus burden but TLR-2 neutralization augments CCR-2 expression in macrophages, along with compromised host-derived reactive oxygen species production. S. aureus infection to CCR-2 intact but TLR-2 neutralized macrophages triggered production of IL-1ß, TNF-α, IL-6, IFN-γ, MCP-1 and expression of iNOS, TNFR-1 and GPx with concomitant decrease in IL-10 production. Further, study with NG-monomethyl-l-arginine (L-NMMA) [iNOS blocker] and buthionine sulfoximine (BSO) [GPx blocker] revealed that S. aureus infection enhanced TLR-2 expression in CCR-2 intact and TLR-2 neutralized macrophages possibly via iNOS and TNFR-1 up regulation and GPx down regulation. Overall, our data indicate that targeting CCR-2 with neutralizing antibody in the early phase of S. aureus infection could restrict excessive inflammation with less compromised bacterial killing. It certainly would be a therapeutic strategy in S. aureus induced inflammatory and infective diseases.


Subject(s)
Macrophages, Peritoneal/immunology , Receptors, CCR2/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Toll-Like Receptor 2/metabolism , Animals , Antibodies, Neutralizing/pharmacology , Bacteriolysis , Buthionine Sulfoximine/pharmacology , Cells, Cultured , Cytokines/metabolism , Host-Pathogen Interactions , Mice , Nitric Oxide Synthase Type II/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Toll-Like Receptor 2/immunology , omega-N-Methylarginine/pharmacology
14.
Microb Pathog ; 109: 131-150, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28552636

ABSTRACT

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.


Subject(s)
Bone Marrow Cells/immunology , Macrophages, Peritoneal/immunology , Receptors, Interleukin-8/metabolism , Spleen/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Animals , Bone Marrow Cells/microbiology , Catalase/metabolism , Cytokines/metabolism , Hot Temperature , Humans , Hydrogen Peroxide/metabolism , Inflammation , Interleukin-8/metabolism , Macrophages/immunology , Macrophages/microbiology , Macrophages, Peritoneal/microbiology , Mice , Neutrophils/immunology , Nitric Oxide/metabolism , Receptors, Interleukin-8/immunology , Spleen/microbiology , Superoxide Dismutase/metabolism , Superoxides/metabolism
15.
Innate Immun ; 23(4): 345-372, 2017 05.
Article in English | MEDLINE | ID: mdl-28409543

ABSTRACT

CCR-2 signaling regulates recruitment of monocytes from the bone marrow into the bloodstream and then to sites of infection. We sought to determine whether CCL-2/CCR-2 signaling is involved in the killing of Staphylococcus aureus by murine bone marrow cells (BMCs). The intermittent link of reactive oxygen species (ROS)-NF-κB/p38-MAPK-mediated CCL-2 production in CCR-2 signaling prompted us to determine whether neutralization of CCR-2 augments the response of murine fresh BMCs (FBMCs) after S. aureus infection. It was observed that anti-CCR-2 Ab-treated FBMCs released fewer ROS on encountering S. aureus infection than CCR-2 non-neutralized FBMCs, also correlating with reduced killing of S. aureus in CCR-2 neutralized FBMCs. Staphylococcal catalase and SOD were also found to play a role in protecting S. aureus from the ROS-mediated killing of FBMC. S. aureus infection of CCR-2 intact FBMCs pre-treated with either NF-κB or p-38-MAPK blocker induced less CCL-2, suggesting that NF-κB or p-38-MAPK is required for CCL-2 production by FBMCs. Moreover, blocking of CCR-2 along with NF-κB or p-38-MAPK resulted in elevated CCL-2 production and reduced CCR-2 expression. Inhibition of CCR-2 impairs the response of murine BMCs to S. aureus infection by attenuation ROS production and modulating the cytokine response.


Subject(s)
Bacteriolysis/drug effects , Bone Marrow Cells/physiology , Monocytes/physiology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Animals , Antibodies, Neutralizing/pharmacology , Bone Marrow Cells/drug effects , Cell Differentiation/drug effects , Cell Movement/drug effects , Cells, Cultured , Cytokines/metabolism , Humans , Male , Mice , Monocytes/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Receptors, CCR2/metabolism , Signal Transduction/drug effects , Staphylococcal Infections/drug therapy
16.
Inflammation ; 39(6): 2072-2093, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27682182

ABSTRACT

The administration of melatonin during acute bacterial infection was evaluated in this study. Mice pre-exposed to normal photoperiodic (NP), short photoperiodic (SP), and long photoperiodic (LP) day lengths were infected separately with live Staphylococcus aureus (5 × 106 cells/ml) or Escherichia coli (2.5 × 107 colony-forming units/ml) and treated with melatonin (10 mg/kg body weight). Behavioral studies were performed before bacterial infection and after melatonin administration. In mice pre-exposed to SP, exogenous melatonin administration resulted in better clearance of bacteria from blood and behavioral improvement. Reduced glutathione content and superoxide dismutase activities were increased, with concomitant decrease in lipid peroxidation content and catalase activities in the liver, brain, and spleen after exogenous melatonin administration. The overproduction of tumor necrosis factor-α, interferon-γ, and interleukin-6 during acute bacterial infection in mice exposed to different photoperiods was probably regulated by the administration of exogenous melatonin, by reducing neutrophil recruitment to spleen, expression of inducible nitric oxide synthase and cyclooxygenase-2 in hypothalamus, and C-reactive protein in the serum, and was also associated with improved behavioral response. Photoperiodic variations in inflammatory and oxidative stress markers might be correlated to serum melatonin and corticosterone levels. This study suggests that the administration of melatonin during SP exposure is protective in infection-induced inflammation than NP and LP exposure.


Subject(s)
Behavior, Animal/drug effects , Escherichia coli Infections/drug therapy , Inflammation/drug therapy , Melatonin/administration & dosage , Staphylococcal Infections/drug therapy , Animals , Escherichia coli Infections/pathology , Inflammation/etiology , Inflammation/microbiology , Melatonin/blood , Melatonin/pharmacology , Mice , Photoperiod , Staphylococcal Infections/pathology , Time Factors
17.
Microb Pathog ; 99: 148-161, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27554276

ABSTRACT

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.


Subject(s)
Arthritis, Infectious/pathology , Cytokines/metabolism , Matrix Metalloproteinase 2/metabolism , Staphylococcal Infections/pathology , Staphylococcus aureus/growth & development , Animals , Disease Models, Animal , Mice , Staphylococcus aureus/immunology
18.
Microb Pathog ; 97: 131-47, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27270212

ABSTRACT

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.


Subject(s)
Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/microbiology , Microbial Viability , Phagocytosis , Staphylococcus aureus/immunology , Staphylococcus aureus/pathogenicity , Toll-Like Receptor 2/metabolism , Animals , Antioxidants/analysis , Catalase/analysis , Cell Survival , Cytokines/analysis , Mice , Muramidase/analysis , Reactive Oxygen Species/analysis , Superoxide Dismutase/analysis , Virulence
19.
Immunol Res ; 64(1): 213-32, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26616292

ABSTRACT

C-C chemokine receptor-2 (CCR-2) is a cognate receptor for monocyte chemotactic protein-1 (MCP-1), and recent studies revealed that MCP-1-CCR-2 signaling is involved in several inflammatory diseases characterized by macrophage infiltration. Currently, there is no study on the involvement of CCR-2 in the killing of S. aureus by macrophages of Swiss albino mice, and its substantial role in host defense against S. aureus infection in murine macrophages is still unclear. Therefore, the present study was aimed to investigate the functional and interactive role of CCR-2 and MCP-1 in regulating peritoneal macrophage responses with respect to acute S. aureus infection. We found that phagocytosis of S. aureus can serve as an important stimulus for MCP-1 production by peritoneal macrophages, which is dependent directly or indirectly on cytokines, reactive oxygen species and nitric oxide. Neutralization of CCR-2 in macrophages leads to increased production of IL-10 and decreased production of IFN-γ and IL-6. In CCR-2 blocked macrophages, pretreatment with specific blocker of NF-κB or p38-MAPK causes elevation in MCP-1 level and subsequent downregulation of CCR-2 itself. We speculate that CCR-2 is involved in S. aureus-induced MCP-1 production via NF-κB or p38-MAPK signaling. We also hypothesized that unnaturally high level of MCP-1 that build up upon CCR-2 neutralization might allow promiscuous binding to one or more other chemokine receptors, a situation that would not occur in CCR-2 non-neutralized condition. This may be the plausible explanation for such observed Th-2 response in CCR-2 blocked macrophages infected with S. aureus in the present study.


Subject(s)
Chemokine CCL2/metabolism , Macrophages, Peritoneal/immunology , Receptors, CCR2/metabolism , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Animals , Antibodies, Blocking/pharmacology , Cells, Cultured , Cytokines/metabolism , Immunity, Innate/drug effects , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/microbiology , Male , Mice , NF-kappa B/metabolism , Phagocytosis/drug effects , Receptors, CCR2/immunology , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Microb Pathog ; 89: 114-27, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26416307

ABSTRACT

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.


Subject(s)
Antioxidants/metabolism , Catalase/metabolism , Microbial Viability , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Superoxide Dismutase/metabolism , Toll-Like Receptor 2/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Host-Pathogen Interactions , Mice , Oxidation-Reduction , Staphylococcus aureus/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...