36 results on '"Rajaram MV"'
Search Results
2. Deletion of PPARγ in lung macrophages provides an immunoprotective response against M. tuberculosis infection in mice.
- Author
-
Guirado E, Rajaram MV, Chawla A, Daigle J, La Perle KM, Arnett E, Turner J, and Schlesinger LS
- Subjects
- Animals, Bacterial Load, Cells, Cultured, Cytokines immunology, Cytokines metabolism, Disease Models, Animal, Host-Pathogen Interactions, Inflammation Mediators immunology, Inflammation Mediators metabolism, Lung immunology, Lung microbiology, Macrophages, Alveolar immunology, Macrophages, Alveolar microbiology, Mice, Inbred BALB C, Mice, Knockout, Mycobacterium tuberculosis growth & development, Mycobacterium tuberculosis immunology, Mycobacterium tuberculosis metabolism, PPAR gamma deficiency, PPAR gamma immunology, Signal Transduction, Tuberculosis, Pulmonary genetics, Tuberculosis, Pulmonary immunology, Tuberculosis, Pulmonary microbiology, Virulence, Gene Deletion, Lung metabolism, Macrophages, Alveolar metabolism, Mycobacterium tuberculosis pathogenicity, PPAR gamma genetics, Tuberculosis, Pulmonary prevention & control
- Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is a nuclear transcription factor belonging to the superfamily of ligand-activated nuclear receptors. It is activated by diverse endogenous lipid metabolites as well as by exogenous ligands such as the thiazolidinediones. It regulates cellular metabolism, proliferation, differentiation, and inflammation, the latter in part through trans-repression of pro-inflammatory cytokines. PPARγ is highly expressed in alternatively activated alveolar macrophages (AMs), a primary host cell for airborne Mycobacterium tuberculosis (M.tb). Our previous in vitro study identified the importance of PPARγ activation through the mannose receptor (CD206) on human macrophages in enabling M. tb growth. The aim of the current study was to investigate the role of PPARγ in vivo during M. tb infection using a macrophage-specific PPARγ knock out mouse model with special emphasis on the lung environment. Our data show that the absence of PPARγ in lung macrophages reduces the growth of virulent M. tb, enhances pro-inflammatory cytokines and reduces granulomatous infiltration. These findings demonstrate that PPARγ activation, which down-regulates macrophage pro-inflammatory responses, impacts the lung's response to M. tb infection, thereby supporting PPARγ's role in tuberculosis (TB) pathogenesis., (Copyright © 2018. Published by Elsevier Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
3. Human Cystic Fibrosis Macrophages Have Defective Calcium-Dependent Protein Kinase C Activation of the NADPH Oxidase, an Effect Augmented by Burkholderia cenocepacia .
- Author
-
Assani K, Shrestha CL, Robledo-Avila F, Rajaram MV, Partida-Sanchez S, Schlesinger LS, and Kopp BT
- Subjects
- Animals, Calcium metabolism, Cells, Cultured, Down-Regulation, Humans, Mice, Phosphorylation, Reactive Oxygen Species metabolism, Burkholderia Infections immunology, Burkholderia cenocepacia immunology, Cystic Fibrosis immunology, Macrophages immunology, NADPH Oxidases metabolism, Protein Kinase C metabolism, Respiratory Burst
- Abstract
Macrophage intracellular pathogen killing is defective in cystic fibrosis (CF), despite abundant production of reactive oxygen species (ROS) in lung tissue. Burkholderia species can cause serious infection in CF and themselves affect key oxidase components in murine non-CF cells. However, it is unknown whether human CF macrophages have an independent defect in the oxidative burst and whether Burkholderia contributes to this defect in terms of assembly of the NADPH oxidase complex and subsequent ROS production. In this article, we analyze CF and non-CF human monocyte-derived macrophages (MDMs) for ROS production, NADPH assembly capacity, protein kinase C expression, and calcium release in response to PMA and CF pathogens. CF MDMs demonstrate a nearly 60% reduction in superoxide production after PMA stimulation compared with non-CF MDMs. Although CF MDMs generally have increased total NADPH component protein expression, they demonstrate decreased expression of the calcium-dependent protein kinase C conventional subclass α/β leading to reduced phosphorylation of NADPH oxidase components p47
phox and p40phox in comparison with non-CF MDMs. Ingestion of B. cenocepacia independently contributes to and worsens the overall oxidative burst deficits in CF MDMs compared with non-CF MDMs. Together, these results provide evidence for inherent deficits in the CF macrophage oxidative burst caused by decreased phosphorylation of NADPH oxidase cytosolic components that are augmented by Burkholderia These findings implicate a critical role for defective macrophage oxidative responses in persistent bacterial infections in CF and create new opportunities for boosting the macrophage immune response to limit infection., (Copyright © 2017 by The American Association of Immunologists, Inc.)- Published
- 2017
- Full Text
- View/download PDF
4. Mouse Liver Sinusoidal Endothelium Eliminates HIV-Like Particles from Blood at a Rate of 100 Million per Minute by a Second-Order Kinetic Process.
- Author
-
Mates JM, Yao Z, Cheplowitz AM, Suer O, Phillips GS, Kwiek JJ, Rajaram MV, Kim J, Robinson JM, Ganesan LP, and Anderson CL
- Abstract
We crafted human immunodeficiency virus (HIV)-like particles of diameter about 140 nm, which expressed two major HIV-1 proteins, namely, env and gag gene products, and used this reagent to simulate the rate of decay of HIV from the blood stream of BALB/c male mice. We found that most (~90%) of the particles were eliminated (cleared) from the blood by the liver sinusoidal endothelial cells (LSECs), the remainder from Kupffer cells; suggesting that LSECs are the major liver scavengers for HIV clearance from blood. Decay was rapid with kinetics suggesting second order with respect to particles, which infers dimerization of a putative receptor on LSEC. The number of HIV-like particles required for saturating the clearance mechanism was approximated. The capacity for elimination of blood-borne HIV-like particles by the sinusoid was 112 million particles per minute. Assuming that the sinusoid endothelial cells were about the size of glass-adherent macrophages, then elimination capacity was more than 540 particles per hour per endothelial cell.
- Published
- 2017
- Full Text
- View/download PDF
5. CD36-Mediated Uptake of Surfactant Lipids by Human Macrophages Promotes Intracellular Growth of Mycobacterium tuberculosis.
- Author
-
Dodd CE, Pyle CJ, Glowinski R, Rajaram MV, and Schlesinger LS
- Subjects
- 1,2-Dipalmitoylphosphatidylcholine metabolism, CD36 Antigens genetics, Cells, Cultured, Humans, Immunity, Innate, Lipid Metabolism, Lung pathology, Macrophages, Alveolar microbiology, Mycobacterium tuberculosis growth & development, Phagocytosis, Pulmonary Surfactant-Associated Protein A metabolism, Pulmonary Surfactants metabolism, RNA, Small Interfering genetics, CD36 Antigens metabolism, Intracellular Space microbiology, Macrophages, Alveolar immunology, Mycobacterium tuberculosis immunology, Tuberculosis, Pulmonary immunology
- Abstract
Mycobacterium tuberculosis imposes a large global health burden as the airborne agent of tuberculosis. Mycobacterium tuberculosis has been flourishing in human populations for millennia and is therefore highly adapted to the lung environment. Alveolar macrophages, a major host cell niche for M. tuberculosis, are not only phagocytose inhaled microbes and particulate matter but are also crucial in catabolizing lung surfactant, a lipid-protein complex that lines the alveolar spaces. Because macrophage host defense properties can be regulated by surfactant and M. tuberculosis can use host lipids as a carbon source during infection, we sought to determine the receptor(s) involved in surfactant lipid uptake by human macrophages and whether the presence of those lipids within macrophages prior to infection with M. tuberculosis enhances bacterial growth. We show that preformed scavenger receptor CD36 is redistributed to the cell membrane following exposure to surfactant lipids and surfactant protein A. Subsequently, surfactant lipids and/or surfactant protein A enhance CD36 transcript and protein levels. We show that CD36 participates in surfactant lipid uptake by human macrophages, as CD36 knockdown reduces uptake of dipalmitoylphosphatidylcholine, the most prevalent surfactant lipid species. Finally, exposing human macrophages to surfactant lipids prior to infection augments M. tuberculosis growth in a CD36-dependent manner. Thus, we provide evidence that CD36 mediates surfactant lipid uptake by human macrophages and that M. tuberculosis exploits this function for growth., Competing Interests: The authors have no financial conflicts of interest., (Copyright © 2016 by The American Association of Immunologists, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
6. Blood-Borne Lipopolysaccharide Is Rapidly Eliminated by Liver Sinusoidal Endothelial Cells via High-Density Lipoprotein.
- Author
-
Yao Z, Mates JM, Cheplowitz AM, Hammer LP, Maiseyeu A, Phillips GS, Wewers MD, Rajaram MV, Robinson JM, Anderson CL, and Ganesan LP
- Subjects
- Acute-Phase Proteins immunology, Acute-Phase Proteins metabolism, Animals, Carrier Proteins immunology, Carrier Proteins metabolism, Endothelial Cells immunology, Gram-Negative Bacterial Infections immunology, Half-Life, Inflammation immunology, Inflammation prevention & control, Kinetics, Kupffer Cells immunology, Lipopolysaccharides immunology, Lipoproteins, HDL immunology, Liver immunology, Membrane Glycoproteins immunology, Membrane Glycoproteins metabolism, Mice, Sepsis immunology, Endothelial Cells physiology, Lipopolysaccharides blood, Lipopolysaccharides metabolism, Lipoproteins, HDL metabolism, Liver cytology
- Abstract
During Gram-negative bacterial infections, excessive LPS induces inflammation and sepsis via action on immune cells. However, the bulk of LPS can be cleared from circulation by the liver. Liver clearance is thought to be a slow process mediated exclusively by phagocytic resident macrophages, Kupffer cells (KC). However, we discovered that LPS disappears rapidly from the circulation, with a half-life of 2-4 min in mice, and liver eliminates about three quarters of LPS from blood circulation. Using microscopic techniques, we found that ∼75% of fluor-tagged LPS in liver became associated with liver sinusoidal endothelial cells (LSEC) and only ∼25% with KC. Notably, the ratio of LSEC-KC-associated LPS remained unchanged 45 min after infusion, indicating that LSEC independently processes the LPS. Most interestingly, results of kinetic analysis of LPS bioactivity, using modified limulus amebocyte lysate assay, suggest that recombinant factor C, an LPS binding protein, competitively inhibits high-density lipoprotein (HDL)-mediated LPS association with LSEC early in the process. Supporting the previous notion, 3 min postinfusion, 75% of infused fluorescently tagged LPS-HDL complex associates with LSEC, suggesting that HDL facilitates LPS clearance. These results lead us to propose a new paradigm of LSEC and HDL in clearing LPS with a potential to avoid inflammation during sepsis., (Copyright © 2016 by The American Association of Immunologists, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
7. Cardiac Electrical and Structural Changes During Bacterial Infection: An Instructive Model to Study Cardiac Dysfunction in Sepsis.
- Author
-
Makara MA, Hoang KV, Ganesan LP, Crouser ED, Gunn JS, Turner J, Schlesinger LS, Mohler PJ, and Rajaram MV
- Subjects
- Animals, Apoptosis, Cytokines metabolism, Disease Models, Animal, Electrocardiography, Heat Shock Transcription Factors metabolism, Interleukin-1beta metabolism, Interleukin-8 metabolism, Mice, MicroRNAs metabolism, Myocardium metabolism, Myocytes, Cardiac pathology, Sepsis metabolism, Sepsis pathology, Superoxide Dismutase metabolism, Tularemia metabolism, Tularemia pathology, Tumor Necrosis Factor-alpha metabolism, Heart physiopathology, Myocardium pathology, Sepsis physiopathology, Tularemia physiopathology
- Abstract
Background: Sepsis patients with cardiac dysfunction have significantly higher mortality. Although several pathways are associated with myocardial damage in sepsis, the precise cause(s) remains unclear and treatment options are limited. This study was designed to develop a new model to investigate the early events of cardiac damage during sepsis progression., Methods and Results: Francisella tularensis subspecies novicida (Ft.n) is a Gram-negative intracellular pathogen causing severe sepsis syndrome in mice. BALB/c mice (N=12) were sham treated or infected with Ft.n through the intranasal route. Serial electrocardiograms were recorded at multiple time points until 96 hours. Hearts were then harvested for histology and gene expression studies. Similar to septic patients, we illustrate both cardiac electrical and structural phenotypes in our murine Ft.n infection model, including prominent R' wave formation, prolonged QRS intervals, and significant left ventricular dysfunction. Notably, in infected animals, we detected numerous microlesions in the myocardium, previously observed following nosocomial Streptococcus infection and in sepsis patients. We show that Ft.n-mediated microlesions are attributed to cardiomyocyte apoptosis, increased immune cell infiltration, and expression of inflammatory mediators (tumor necrosis factor, interleukin [IL]-1β, IL-8, and superoxide dismutase 2). Finally, we identify increased expression of microRNA-155 and rapid degradation of heat shock factor 1 following cardiac Ft.n infection as a primary cause of myocardial inflammation and apoptosis., Conclusions: We have developed and characterized an Ft.n infection model to understand the pathogenesis of cardiac dysregulation in sepsis. Our findings illustrate novel in vivo phenotypes underlying cardiac dysfunction during Ft.n infection with significant translational impact on our understanding of sepsis pathophysiology., (© 2016 The Authors. Published on behalf of the American Heart Association, Inc., by Wiley Blackwell.)
- Published
- 2016
- Full Text
- View/download PDF
8. Targeting CBLB as a potential therapeutic approach for disseminated candidiasis.
- Author
-
Xiao Y, Tang J, Guo H, Zhao Y, Tang R, Ouyang S, Zeng Q, Rappleye CA, Rajaram MV, Schlesinger LS, Tao L, Brown GD, Langdon WY, Li BT, and Zhang J
- Subjects
- Adaptor Proteins, Signal Transducing immunology, Animals, Blotting, Western, Candida albicans, Candidiasis, Invasive immunology, Cytokines immunology, Dendritic Cells immunology, Enzyme-Linked Immunosorbent Assay, Gene Knockdown Techniques, Humans, Immunity, Innate immunology, Immunoprecipitation, Lectins, C-Type metabolism, Leukocytes, Mononuclear immunology, Macrophages immunology, Mice, Mice, Knockout, Microscopy, Confocal, Mutagenesis, Site-Directed, Neutrophils immunology, Phagocytosis genetics, Phagocytosis immunology, Proto-Oncogene Proteins c-cbl immunology, Syk Kinase metabolism, Ubiquitination genetics, Ubiquitination immunology, Adaptor Proteins, Signal Transducing genetics, Candidiasis, Invasive genetics, Immunity, Innate genetics, Inflammasomes immunology, Proto-Oncogene Proteins c-cbl genetics, Reactive Oxygen Species immunology
- Abstract
Disseminated candidiasis has become one of the leading causes of hospital-acquired blood stream infections with high mobility and mortality. However, the molecular basis of host defense against disseminated candidiasis remains elusive, and treatment options are limited. Here we report that the E3 ubiquitin ligase CBLB directs polyubiquitination of dectin-1 and dectin-2, two key pattern-recognition receptors for sensing Candida albicans, and their downstream kinase SYK, thus inhibiting dectin-1- and dectin-2-mediated innate immune responses. CBLB deficiency or inactivation protects mice from systemic infection with a lethal dose of C. albicans, and deficiency of dectin-1, dectin-2, or both in Cblb(-/-) mice abrogates this protection. Notably, silencing the Cblb gene in vivo protects mice from lethal systemic C. albicans infection. Our data reveal that CBLB is crucial for homeostatic control of innate immune responses mediated by dectin-1 and dectin-2. Our data also indicate that CBLB represents a potential therapeutic target for protection from disseminated candidiasis., Competing Interests: Statement The authors declare no competing financial interests.
- Published
- 2016
- Full Text
- View/download PDF
9. Correction: Fine Tuning Inflammation at the Front Door: Macrophage Complement Receptor 3-mediates Phagocytosis and Immune Suppression for Francisella tularensis.
- Author
-
Dai S, Rajaram MV, Curry HM, Leander R, and Schlesinger LS
- Published
- 2016
- Full Text
- View/download PDF
10. Scavenger receptor B1, the HDL receptor, is expressed abundantly in liver sinusoidal endothelial cells.
- Author
-
Ganesan LP, Mates JM, Cheplowitz AM, Avila CL, Zimmerer JM, Yao Z, Maiseyeu A, Rajaram MV, Robinson JM, and Anderson CL
- Subjects
- Animals, Biological Transport, COS Cells, Cell Line, Cell Separation, Chlorocebus aethiops, Endothelial Cells cytology, Hepatocytes cytology, Liver cytology, Macrophages cytology, Macrophages metabolism, Male, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Microscopy, Confocal, Microtomy, Organ Specificity, RNA, Messenger metabolism, Receptors, IgG genetics, Receptors, IgG metabolism, Scavenger Receptors, Class B metabolism, beta Catenin genetics, beta Catenin metabolism, Cholesterol metabolism, Endothelial Cells metabolism, Hepatocytes metabolism, Liver metabolism, RNA, Messenger genetics, Scavenger Receptors, Class B genetics
- Abstract
Cholesterol from peripheral tissue, carried by HDL, is metabolized in the liver after uptake by the HDL receptor, SR-B1. Hepatocytes have long been considered the only liver cells expressing SR-B1; however, in this study we describe two disparate immunofluorescence (IF) experiments that suggest otherwise. Using high-resolution confocal microscopy employing ultrathin (120 nm) sections of mouse liver, improving z-axis resolution, we identified the liver sinusoidal endothelial cells (LSEC), marked by FcγRIIb, as the cell within the liver expressing abundant SR-B1. In contrast, the hepatocyte, identified with β-catenin, expressed considerably weaker levels, although optical resolution of SR-B1 was inadequate. Thus, we moved to a different IF strategy, first separating dissociated liver cells by gradient centrifugation into two portions, hepatocytes (parenchymal cells) and LSEC (non-parenchymal cells). Characterizing both portions for the cellular expression of SR-B1 by flow cytometry, we found that LSEC expressed considerable amounts of SR-B1 while in hepatocytes SR-B1 expression was barely perceptible. Assessing mRNA of SR-B1 by real time PCR we found messenger expression in LSEC to be about 5 times higher than in hepatocytes.
- Published
- 2016
- Full Text
- View/download PDF
11. γ-Tilmanocept, a New Radiopharmaceutical Tracer for Cancer Sentinel Lymph Nodes, Binds to the Mannose Receptor (CD206).
- Author
-
Azad AK, Rajaram MV, Metz WL, Cope FO, Blue MS, Vera DR, and Schlesinger LS
- Subjects
- Carbocyanines chemistry, Cells, Cultured, Dextrans chemistry, Flow Cytometry, HEK293 Cells, Humans, Immunohistochemistry, Lectins, C-Type genetics, Lymph Nodes metabolism, Macrophages metabolism, Mannans chemistry, Mannose Receptor, Mannose-Binding Lectins genetics, Microscopy, Confocal, Molecular Structure, Neoplasms diagnosis, Neoplasms metabolism, Protein Binding, RNA Interference, Radiopharmaceuticals metabolism, Receptors, Cell Surface genetics, Technetium Tc 99m Pentetate chemistry, Technetium Tc 99m Pentetate metabolism, Dextrans metabolism, Lectins, C-Type metabolism, Mannans metabolism, Mannose-Binding Lectins metabolism, Receptors, Cell Surface metabolism, Sentinel Lymph Node Biopsy methods, Technetium Tc 99m Pentetate analogs & derivatives
- Abstract
γ-Tilmanocept ((99m)Tc-labeled-tilmanocept or [(99m)Tc]-tilmanocept) is the first mannose-containing, receptor-directed, radiolabeled tracer for the highly sensitive imaging of sentinel lymph nodes in solid tumor staging. To elucidate the mannose-binding receptor that retains tilmanocept in this microenvironment, human macrophages were used that have high expression of the C-type lectin mannose receptor (MR; CD206). Cy3-labeled tilmanocept exhibited high specificity binding to macrophages that was nearly abolished in competitive inhibition experiments. Furthermore, Cy3-tilmanocept binding was markedly reduced on macrophages deficient in the MR by small interfering RNA treatment and was increased on MR-transfected HEK 293 cells. Finally, confocal microscopy revealed colocalization of Cy3-tilmanocept with the macrophage membrane MR and binding of labeled tilmanocept to MR(+) cells (macrophages and/or dendritic cells) in human sentinel lymph node tissues. Together these data provide strong evidence that CD206 is a major binding receptor for γ-tilmanocept. Identification of CD206 as the γ-tilmanocept-binding receptor enables opportunities for designing receptor-targeted advanced imaging agents and therapeutics for cancer and other diseases., (Copyright © 2015 by The American Association of Immunologists, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
12. Role for NOD2 in Mycobacterium tuberculosis-induced iNOS expression and NO production in human macrophages.
- Author
-
Landes MB, Rajaram MV, Nguyen H, and Schlesinger LS
- Subjects
- Acetylmuramyl-Alanyl-Isoglutamine analogs & derivatives, Acetylmuramyl-Alanyl-Isoglutamine pharmacology, Gene Expression Regulation, Host-Pathogen Interactions immunology, Humans, Macrophages drug effects, Macrophages metabolism, Macrophages microbiology, Mycobacterium bovis immunology, Mycobacterium tuberculosis drug effects, NF-kappa B genetics, Nitric Oxide Synthase Type II genetics, Nod2 Signaling Adaptor Protein genetics, Primary Cell Culture, Signal Transduction, Macrophages immunology, Mycobacterium tuberculosis immunology, NF-kappa B immunology, Nitric Oxide biosynthesis, Nitric Oxide Synthase Type II immunology, Nod2 Signaling Adaptor Protein immunology
- Abstract
M.tb, which causes TB, is a host-adapted intracellular pathogen of macrophages. Macrophage intracellular PRRs, such as NOD proteins, regulate proinflammatory cytokine production in response to various pathogenic organisms. We demonstrated previously that NOD2 plays an important role in controlling the inflammatory response and viability of M.tb and Mycobacterium bovis BCG in human macrophages. Various inflammatory mediators, such as cytokines, ROS, and RNS, such as NO, can mediate this control. iNOS (or NOS2) is a key enzyme for NO production and M.tb control during infection of mouse macrophages; however, the role of NO during infection of human macrophages remains unclear, in part, as a result of the low amounts of NO produced in these cells. Here, we tested the hypothesis that activation of NOD2 by its ligands (MDP and GMDP, the latter from M.tb) plays an important role in the expression and activity of iNOS and NO production in human macrophages. We demonstrate that M.tb or M. bovis BCG infection enhances iNOS expression in human macrophages. The M.tb-induced iNOS expression and NO production are dependent on NOD2 expression during M.tb infection. Finally, NF-κB activation is required for NOD2-dependent expression of iNOS in human macrophages. Our data provide evidence for a new molecular pathway that links activation of NOD2, an important intracellular PRR, and iNOS expression and activity during M.tb infection of human macrophages., (© Society for Leukocyte Biology.)
- Published
- 2015
- Full Text
- View/download PDF
13. Acetalated dextran encapsulated AR-12 as a host-directed therapy to control Salmonella infection.
- Author
-
Hoang KV, Borteh HM, Rajaram MV, Peine KJ, Curry H, Collier MA, Homsy ML, Bachelder EM, Gunn JS, Schlesinger LS, and Ainslie KM
- Subjects
- Acetals chemistry, Anti-Bacterial Agents pharmacology, Autophagy drug effects, Blotting, Western, Cell Survival drug effects, Cells, Cultured, Drug Compounding, Humans, Macrophages drug effects, Macrophages microbiology, Microscopy, Electron, Scanning, Microscopy, Fluorescence, Microtubule-Associated Proteins metabolism, Pyrazoles pharmacology, Salmonella Infections drug therapy, Salmonella Infections microbiology, Sulfonamides pharmacology, Surface Properties, Anti-Bacterial Agents administration & dosage, Dextrans chemistry, Drug Carriers chemistry, Pyrazoles administration & dosage, Salmonella typhimurium drug effects, Sulfonamides administration & dosage
- Abstract
AR-12 has been evaluated in clinical trials as an anti-cancer agent but also has demonstrated host-directed, broad-spectrum clearance of bacteria. We have previously shown that AR-12 has activity in vitro against Salmonella enterica serovar Typhimurium and Francisella species by inducing autophagy and other host immune pathways. AR-12 treatment of S. Typhimurium-infected mice resulted in a 10-fold reduction in bacterial load in the liver and spleen and an increased survival time. However, AR-12 treatment did not protect mice from death, likely due poor formulation. In the current study, AR-12 was encapsulated in a microparticulate carrier formulated from the novel degradable biopolymer acetalated dextran (Ace-DEX) and subsequently evaluated for its activity in human monocyte-derived macrophages (hMDMs). Our results show that hMDMs efficiently internalized Ace-DEX microparticles (MPs), and that encapsulation significantly reduced host cell cytotoxicity compared to unencapsulated AR-12. Efficient macrophage internalization of AR-12 loaded MPs (AR-12/MPs) was further demonstrated by autophagosome formation that was comparable to free AR-12 and resulted in enhanced clearance of intracellular Salmonella. Taken together, these studies provide support that Ace-DEX encapsulated AR-12 may be a promising new therapeutic agent to control intracellular bacterial pathogens of macrophages by targeting delivery and reducing drug toxicity., (Copyright © 2014 Elsevier B.V. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
14. Macrophage immunoregulatory pathways in tuberculosis.
- Author
-
Rajaram MV, Ni B, Dodd CE, and Schlesinger LS
- Subjects
- Cytokines genetics, Cytokines immunology, Gene Expression Regulation, Host-Pathogen Interactions, Humans, Immunity, Innate, Lung microbiology, Lung pathology, Macrophage Activation, Macrophages, Alveolar microbiology, Macrophages, Alveolar pathology, MicroRNAs genetics, MicroRNAs immunology, Receptors, Pattern Recognition genetics, Receptors, Pattern Recognition immunology, Tuberculosis, Pulmonary genetics, Tuberculosis, Pulmonary microbiology, Tuberculosis, Pulmonary pathology, Lung immunology, Macrophages, Alveolar immunology, Mycobacterium tuberculosis immunology, Tuberculosis, Pulmonary immunology
- Abstract
Macrophages, the major host cells harboring Mycobacterium tuberculosis (M.tb), are a heterogeneous cell type depending on their tissue of origin and host they are derived from. Significant discord in macrophage responses to M.tb exists due to differences in M.tb strains and the various types of macrophages used to study tuberculosis (TB). This review will summarize current concepts regarding macrophage responses to M.tb infection, while pointing out relevant differences in experimental outcomes due to the use of divergent model systems. A brief description of the lung environment is included since there is increasing evidence that the alveolar macrophage (AM) has immunoregulatory properties that can delay optimal protective host immune responses. In this context, this review focuses on selected macrophage immunoregulatory pattern recognition receptors (PRRs), cytokines, negative regulators of inflammation, lipid mediators and microRNAs (miRNAs)., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
15. Chemoproteomics reveals Toll-like receptor fatty acylation.
- Author
-
Chesarino NM, Hach JC, Chen JL, Zaro BW, Rajaram MV, Turner J, Schlesinger LS, Pratt MR, Hang HC, and Yount JS
- Subjects
- Acylation, Animals, B7-2 Antigen genetics, B7-2 Antigen metabolism, Cell Membrane metabolism, Cells, Cultured, Click Chemistry, Computational Biology, Dendritic Cells metabolism, Fibroblasts metabolism, HEK293 Cells, Humans, Interferon-alpha metabolism, Ligands, Mice, NF-kappa B genetics, NF-kappa B metabolism, Protein Processing, Post-Translational, Proteome genetics, Proteome metabolism, Toll-Like Receptor 2 genetics, Lipoylation, Proteomics methods, Toll-Like Receptor 2 metabolism
- Abstract
Background: Palmitoylation is a 16-carbon lipid post-translational modification that increases protein hydrophobicity. This form of protein fatty acylation is emerging as a critical regulatory modification for multiple aspects of cellular interactions and signaling. Despite recent advances in the development of chemical tools for the rapid identification and visualization of palmitoylated proteins, the palmitoyl proteome has not been fully defined. Here we sought to identify and compare the palmitoylated proteins in murine fibroblasts and dendritic cells., Results: A total of 563 putative palmitoylation substrates were identified, more than 200 of which have not been previously suggested to be palmitoylated in past proteomic studies. Here we validate the palmitoylation of several new proteins including Toll-like receptors (TLRs) 2, 5 and 10, CD80, CD86, and NEDD4. Palmitoylation of TLR2, which was uniquely identified in dendritic cells, was mapped to a transmembrane domain-proximal cysteine. Inhibition of TLR2 S-palmitoylation pharmacologically or by cysteine mutagenesis led to decreased cell surface expression and a decreased inflammatory response to microbial ligands., Conclusions: This work identifies many fatty acylated proteins involved in fundamental cellular processes as well as cell type-specific functions, highlighting the value of examining the palmitoyl proteomes of multiple cell types. S-palmitoylation of TLR2 is a previously unknown immunoregulatory mechanism that represents an entirely novel avenue for modulation of TLR2 inflammatory activity.
- Published
- 2014
- Full Text
- View/download PDF
16. Mycobacterium tuberculosis decreases human macrophage IFN-γ responsiveness through miR-132 and miR-26a.
- Author
-
Ni B, Rajaram MV, Lafuse WP, Landes MB, and Schlesinger LS
- Subjects
- 3' Untranslated Regions, Binding Sites, Cell Line, Gene Expression Profiling, Gene Expression Regulation drug effects, HLA-DR Antigens genetics, HLA-DR Antigens immunology, Humans, Interferon-gamma pharmacology, Macrophages drug effects, RNA Interference, RNA, Messenger genetics, Receptors, IgG genetics, Receptors, IgG metabolism, Reproducibility of Results, p300-CBP Transcription Factors genetics, p300-CBP Transcription Factors metabolism, Macrophages immunology, Macrophages metabolism, MicroRNAs genetics, Mycobacterium tuberculosis immunology, Tuberculosis genetics, Tuberculosis immunology
- Abstract
IFN-γ-activated macrophages play an essential role in controlling intracellular pathogens; however, macrophages also serve as the cellular home for the intracellular pathogen Mycobacterium tuberculosis. Based on previous evidence that M. tuberculosis can modulate host microRNA (miRNA) expression, we examined the miRNA expression profile of M. tuberculosis-infected primary human macrophages. We identified 31 differentially expressed miRNAs in primary human macrophages during M. tuberculosis infection by NanoString and confirmed our findings by quantitative real-time RT-PCR. In addition, we determined a role for two miRNAs upregulated upon M. tuberculosis infection, miR-132 and miR-26a, as negative regulators of transcriptional coactivator p300, a component of the IFN-γ signaling cascade. Knockdown expression of miR-132 and miR-26a increased p300 protein levels and improved transcriptional, translational, and functional responses to IFN-γ in human macrophages. Collectively, these data validate p300 as a target of miR-132 and miR-26a, and demonstrate a mechanism by which M. tuberculosis can limit macrophage responses to IFN-γ by altering host miRNA expression., (Copyright © 2014 by The American Association of Immunologists, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
17. Molecular composition of the alveolar lining fluid in the aging lung.
- Author
-
Moliva JI, Rajaram MV, Sidiki S, Sasindran SJ, Guirado E, Pan XJ, Wang SH, Ross P Jr, Lafuse WP, Schlesinger LS, Turner J, and Torrelles JB
- Subjects
- Adult, Animals, Female, Humans, Hydrolases metabolism, Male, Mice, Aging metabolism, Lung metabolism, Oxidative Stress, Pulmonary Alveoli metabolism, Pulmonary Surfactant-Associated Protein A metabolism
- Abstract
As we age, there is an increased risk for the development of pulmonary diseases, including infections, but few studies have considered changes in lung surfactant and components of the innate immune system as contributing factors to the increased susceptibility of the elderly to succumb to infections. We and others have demonstrated that human alveolar lining fluid (ALF) components, such as surfactant protein (SP)-A, SP-D, complement protein C3, and alveolar hydrolases, play a significant innate immune role in controlling microbial infections. However, there is a lack of information regarding the effect of increasing age on the level and function of ALF components in the lung. Here we addressed this gap in knowledge by determining the levels of ALF components in the aging lung that are important in controlling infection. Our findings demonstrate that pro-inflammatory cytokines, surfactant proteins and lipids, and complement components are significantly altered in the aged lung in both mice and humans. Further, we show that the aging lung is a relatively oxidized environment. Our study provides new information on how the pulmonary environment in old age can potentially modify mucosal immune responses, thereby impacting pulmonary infections and other pulmonary diseases in the elderly population.
- Published
- 2014
- Full Text
- View/download PDF
18. N-glycolylated peptidoglycan contributes to the immunogenicity but not pathogenicity of Mycobacterium tuberculosis.
- Author
-
Hansen JM, Golchin SA, Veyrier FJ, Domenech P, Boneca IG, Azad AK, Rajaram MV, Schlesinger LS, Divangahi M, Reed MB, and Behr MA
- Subjects
- Acetylmuramyl-Alanyl-Isoglutamine metabolism, Animals, Bacterial Load, Cells, Cultured, Disease Models, Animal, Gene Deletion, Humans, Macrophages microbiology, Mice, Mice, Inbred C57BL, Mixed Function Oxygenases genetics, Mixed Function Oxygenases metabolism, Mycobacterium tuberculosis chemistry, Mycobacterium tuberculosis enzymology, Peptidoglycan chemistry, Protein Processing, Post-Translational, Survival Analysis, Tuberculosis, Pulmonary immunology, Tuberculosis, Pulmonary microbiology, Virulence, Acetylmuramyl-Alanyl-Isoglutamine immunology, Mycobacterium tuberculosis immunology, Mycobacterium tuberculosis pathogenicity, Peptidoglycan immunology
- Abstract
Mycobacteria produce an unusual, glycolylated form of muramyl dipeptide (MDP) that is more potent and efficacious at inducing NOD2-mediated host responses. We tested the importance of this modified form of MDP in Mycobacterium tuberculosis by disrupting the gene, namH, responsible for this modification. In vitro, the namH mutant did not produce N-glycolylated muropeptides, but there was no alteration in colony morphology, growth kinetics, cellular morphology, or mycolic acid profile. Ex vivo, the namH mutant survived and replicated normally in murine and human macrophages, yet induced diminished production of tumor necrosis factor α. In vivo, namH disruption did not affect the bacterial burden during infection of C57BL/6 mice or cellular recruitment to the lungs but modestly prolonged survival after infection in Rag1(-/-) mice. These results indicate that the modified MDP is an important contributor to the unusual immunogenicity of mycobacteria but has a limited role in the pathogenesis of M. tuberculosis infection.
- Published
- 2014
- Full Text
- View/download PDF
19. Estrogen modulation of endosome-associated toll-like receptor 8: an IFNα-independent mechanism of sex-bias in systemic lupus erythematosus.
- Author
-
Young NA, Wu LC, Burd CJ, Friedman AK, Kaffenberger BH, Rajaram MV, Schlesinger LS, James H, Shupnik MA, and Jarjour WN
- Subjects
- Animals, Binding Sites, Cell Line, Tumor, Cells, Cultured, Endosomes immunology, Endosomes metabolism, Estrogen Receptor alpha genetics, Female, Gene Expression Regulation, Humans, Imidazoles pharmacology, Immunologic Factors pharmacology, Interferon-alpha immunology, Leukocytes, Mononuclear immunology, Leukocytes, Mononuclear metabolism, Lupus Erythematosus, Systemic genetics, Lupus Erythematosus, Systemic pathology, Male, Mice, Mice, Inbred C57BL, Protein Binding, Sex Factors, Signal Transduction, Toll-Like Receptor 8 agonists, Toll-Like Receptor 8 genetics, Endosomes drug effects, Estradiol pharmacology, Estrogen Receptor alpha immunology, Leukocytes, Mononuclear drug effects, Lupus Erythematosus, Systemic immunology, Toll-Like Receptor 8 immunology
- Abstract
Females of child-bearing age are more resistant to infectious disease and have an increased risk of systemic lupus erythematosus (SLE). We hypothesized that estrogen-induced gene expression could establish an immunoactivated state which would render enhanced defense against infection, but may be deleterious in autoimmune development. Using peripheral blood mononuclear cells (PBMCs), we demonstrate enhanced responses with immunogen stimulation in the presence of 17β-estradiol (E2) and gene array analyses reveal toll-like receptor 8 (TLR8) as an E2-responsive candidate gene. TLR8 expression levels are up-regulated in SLE and PBMCs stimulated with TLR8 agonist display a female sex-biased, E2-sensitive response. Moreover, we identify a putative ERα-binding region near the TLR8 locus and blocking ERα expression significantly decreases E2-mediated TLR8 induction. Our findings characterize TLR8 as a novel estrogen target gene that can lower the inflammatory threshold and implicate an IFNα-independent inflammatory mechanism that could contribute to higher SLE incidence in women., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
20. Exploitation of the Macrophage Mannose Receptor (CD206) in Infectious Disease Diagnostics and Therapeutics.
- Author
-
Azad AK, Rajaram MV, and Schlesinger LS
- Abstract
The macrophage mannose receptor (MR, CD206) is a C-type lectin expressed predominantly by most tissue macrophages, dendritic cells and specific lymphatic or endothelial cells. It functions in endocytosis and phagocytosis, and plays an important role in immune homeostasis by scavenging unwanted mannoglycoproteins. More attention is being paid to its particularly high expression in tissue pathology sites during disease such the tumor microenvironment. The MR recognizes a variety of microorganisms by their mannan-coated cell wall, which is exploited by adapted intracellular pathogens such as Mycobacterium tuberculosis , for their own survival. Despite the continued development of drug delivery technologies, the targeting of agents to immune cells, especially macrophages, for effective diagnosis and treatment of chronic infectious diseases has not been addressed adequately. In this regard, strategies that optimize MR-mediated uptake by macrophages in target tissues during infection are becoming an attractive approach. We review important progress in this area.
- Published
- 2014
- Full Text
- View/download PDF
21. Novel estrogen target gene ZAS3 is overexpressed in systemic lupus erythematosus.
- Author
-
Young NA, Friedman AK, Kaffenberger B, Rajaram MV, Birmingham DJ, Rovin BH, Hebert LA, Schlesinger LS, Wu LC, and Jarjour WN
- Subjects
- Animals, Cells, Cultured, DNA-Binding Proteins metabolism, Dose-Response Relationship, Drug, Estrogen Receptor alpha genetics, Estrogen Receptor alpha metabolism, Estrogens pharmacology, Female, Gene Expression Regulation drug effects, Humans, Lupus Erythematosus, Systemic metabolism, Mice, Mice, Inbred C57BL, Transcription Factors metabolism, Up-Regulation drug effects, Up-Regulation genetics, DNA-Binding Proteins genetics, Estradiol pharmacology, Lupus Erythematosus, Systemic genetics, Transcription Factors genetics
- Abstract
Systemic lupus erythematosus (SLE) is a prototypic, inflammatory autoimmune disease characterized by significant gender bias. Previous studies have established a role for hormones in SLE pathogenesis, including the sex hormone estrogen. Estrogen regulates gene expression by translocating estrogen receptors (ER) α and β into the nucleus where they induce transcription by binding to estrogen response elements (EREs) of target genes. The ZAS3 locus encodes a signaling and transcriptional molecule involved in regulating inflammatory responses. We show that ZAS3 is significantly up-regulated in SLE patients at both the protein and mRNA levels in peripheral blood mononuclear cells (PBMCs). Furthermore, estrogen stimulates the expression of ZAS3 in vitro in several leukocyte and breast cancer cell lines of both human and murine origin. In vivo estrogen treatment mediates induction of tissue specific ZAS3 expression in several lymphoid organs in mice. Estrogen stimulation also significantly up-regulates ZAS3 expression in primary PBMCs, while treatment with testosterone has no effect. Mechanistically, estrogen induces differential ERα binding to putative EREs within the ZAS3 gene and ERα knockdown with siRNA prevents estrogen induced ZAS3 up-regulation. In contrast, siRNA targeting IFNα has no effect. These data demonstrate that ZAS3 expression is directly regulated by estrogen and that ZAS3 is overexpressed in lupus. Since ZAS3 has been shown to regulate inflammatory pathways, its up-regulation by estrogen could play a critical role in female-biased autoimmune disorders., (Copyright © 2012 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
22. Type A Francisella tularensis acid phosphatases contribute to pathogenesis.
- Author
-
Mohapatra NP, Soni S, Rajaram MV, Strandberg KL, and Gunn JS
- Subjects
- Acid Phosphatase deficiency, Acid Phosphatase genetics, Animals, Female, Francisella tularensis genetics, Gene Deletion, Humans, Macrophages cytology, Macrophages metabolism, Macrophages microbiology, Mice, Mice, Inbred BALB C, Monocytes cytology, Monocytes microbiology, NADPH Oxidases metabolism, Neutrophils cytology, Neutrophils microbiology, Phagosomes microbiology, Protein Transport, Reactive Oxygen Species metabolism, Acid Phosphatase metabolism, Francisella tularensis enzymology, Francisella tularensis physiology
- Abstract
Different Francisella spp. produce five or six predicted acid phosphatases (AcpA, AcpB, AcpC, AcpD, HapA and HapB). The genes encoding the histidine acid phosphatases (hapA, hapB) and acpD of F. tularensis subsp. Schu S4 strain are truncated or disrupted. However, deletion of HapA (FTT1064) in F. tularensis Schu S4 resulted in a 33% reduction in acid phosphatase activity and loss of the four functional acid phosphatases in F. tularensis Schu S4 (ΔABCH) resulted in a>99% reduction in acid phosphatase activity compared to the wild type strain. All single, double and triple mutants tested, demonstrated a moderate decrease in mouse virulence and survival and growth within human and murine phagocytes, whereas the ΔABCH mutant showed >3.5-fold decrease in intramacrophage survival and 100% attenuation of virulence in mouse. While the Schu S4 ΔABCH strain was attenuated in the mouse model, it showed only limited protection against wild type challenge. F. tularensis Schu S4 failed to stimulate reactive oxygen species production in phagocytes, whereas infection by the ΔABCH strain stimulated 5- and 56-fold increase in reactive oxygen species production in neutrophils and human monocyte-derived macrophages, respectively. The ΔABCH mutant but not the wild type strain strongly co-localized with p47 (phox) and replicated in macrophages isolated from p47 (phox) knockout mice. Thus, F. tularensis Schu S4 acid phosphatases, including the truncated HapA, play a major role in intramacrophage survival and virulence of this human pathogen.
- Published
- 2013
- Full Text
- View/download PDF
23. Fine tuning inflammation at the front door: macrophage complement receptor 3-mediates phagocytosis and immune suppression for Francisella tularensis.
- Author
-
Dai S, Rajaram MV, Curry HM, Leander R, and Schlesinger LS
- Subjects
- Animals, CHO Cells immunology, CHO Cells metabolism, Cricetinae, Cricetulus, Gene Silencing, Humans, Immune Evasion, Immunologic Factors metabolism, Inflammation microbiology, Macrophages immunology, Macrophages metabolism, Macrophages microbiology, Opsonin Proteins metabolism, RNA, Small Interfering genetics, Transfection, Tularemia metabolism, Francisella tularensis immunology, Immune Tolerance, Inflammation immunology, Macrophage-1 Antigen metabolism, Phagocytosis immunology, Tularemia immunology
- Abstract
Complement receptor 3 (CR3, CD11b/CD18) is a major macrophage phagocytic receptor. The biochemical pathways through which CR3 regulates immunologic responses have not been fully characterized. Francisella tularensis is a remarkably infectious, facultative intracellular pathogen of macrophages that causes tularemia. Early evasion of the host immune response contributes to the virulence of F. tularensis and CR3 is an important receptor for its phagocytosis. Here we confirm that efficient attachment and uptake of the highly virulent Type A F. tularensis spp. tularensis strain Schu S4 by human monocyte-derived macrophages (hMDMs) requires complement C3 opsonization and CR3. However, despite a>40-fold increase in uptake following C3 opsonization, Schu S4 induces limited pro-inflammatory cytokine production compared with non-opsonized Schu S4 and the low virulent F. novicida. This suggests that engagement of CR3 by opsonized Schu S4 contributes specifically to the immune suppression during and shortly following phagocytosis which we demonstrate by CD11b siRNA knockdown in hMDMs. This immune suppression is concomitant with early inhibition of ERK1/2, p38 MAPK and NF-κB activation. Furthermore, TLR2 siRNA knockdown shows that pro-inflammatory cytokine production and MAPK activation in response to non-opsonized Schu S4 depends on TLR2 signaling providing evidence that CR3-TLR2 crosstalk mediates immune suppression for opsonized Schu S4. Deletion of the CD11b cytoplasmic tail reverses the CR3-mediated decrease in ERK and p38 activation during opsonized Schu-S4 infection. The CR3-mediated signaling pathway involved in this immune suppression includes Lyn kinase and Akt activation, and increased MKP-1, which limits TLR2-mediated pro-inflammatory responses. These data indicate that while the highly virulent F. tularensis uses CR3 for efficient uptake, optimal engagement of this receptor down-regulates TLR2-dependent pro-inflammatory responses by inhibiting MAPK activation through outside-in signaling. CR3-linked immune suppression is an important mechanism involved in the pathogenesis of F. tularensis infection.
- Published
- 2013
- Full Text
- View/download PDF
24. Pulmonary surfactant protein A and surfactant lipids upregulate IRAK-M, a negative regulator of TLR-mediated inflammation in human macrophages.
- Author
-
Nguyen HA, Rajaram MV, Meyer DA, and Schlesinger LS
- Subjects
- Cells, Cultured, Cytokines biosynthesis, Gene Knockdown Techniques, Humans, Inflammation Mediators metabolism, Interleukin-1 Receptor-Associated Kinases genetics, Signal Transduction, Up-Regulation, Inflammation metabolism, Interleukin-1 Receptor-Associated Kinases biosynthesis, Macrophages metabolism, Pulmonary Surfactant-Associated Protein A metabolism, Pulmonary Surfactants metabolism, Toll-Like Receptors metabolism
- Abstract
Alveolar macrophages (AMs) are exposed to frequent challenges from inhaled particulates and microbes and function as a first line of defense with a highly regulated immune response because of their unique biology as prototypic alternatively activated macrophages. Lung collectins, particularly surfactant protein A (SP-A), contribute to this activation state by fine-tuning the macrophage inflammatory response. During short-term (10 min-2 h) exposure, SP-A's regulation of human macrophage responses occurs through decreased activity of kinases required for proinflammatory cytokine production. However, AMs are continuously exposed to surfactant, and the biochemical pathways underlying long-term reduction of proinflammatory cytokine activity are not known. We investigated the molecular mechanism(s) underlying SP-A- and surfactant lipid-mediated suppression of proinflammatory cytokine production in response to Toll-like receptor (TLR) 4 (TLR4) activation over longer time periods. We found that exposure of human macrophages to SP-A for 6-24 h upregulates expression of IL-1 receptor-associated kinase M (IRAK-M), a negative regulator of TLR-mediated NF-κB activation. Exposure to Survanta, a natural bovine lung extract lacking SP-A, also enhances IRAK-M expression, but at lower magnitude and for a shorter duration than SP-A. Surfactant-mediated upregulation of IRAK-M in macrophages suppresses TLR4-mediated TNF-α and IL-6 production in response to LPS, and IRAK-M knockdown by small interfering RNA reverses this suppression. In contrast to TNF-α and IL-6, the surfactant components upregulate LPS-mediated immunoregulatory IL-10 production, an effect reversed by IRAK-M knockdown. In conclusion, these data identify an important signaling regulator in human macrophages that is used by surfactant to control the long-term alveolar inflammatory response, i.e., enhanced IRAK-M activity.
- Published
- 2012
- Full Text
- View/download PDF
25. MiR-155 induction by microbes/microbial ligands requires NF-κB-dependent de novo protein synthesis.
- Author
-
Cremer TJ, Fatehchand K, Shah P, Gillette D, Patel H, Marsh RL, Besecker BY, Rajaram MV, Cormet-Boyaka E, Kanneganti TD, Schlesinger LS, Butchar JP, and Tridandapani S
- Subjects
- Cells, Cultured, Humans, Protein Biosynthesis, Francisella tularensis immunology, MicroRNAs biosynthesis, Monocytes immunology, NF-kappa B metabolism
- Abstract
MiR-155 regulates numerous aspects of innate and adaptive immune function. This miR is induced in response to Toll-like receptor ligands, cytokines, and microbial infection. We have previously shown that miR-155 is induced in monocytes/macrophages infected with Francisella tularensis and suppresses expression of the inositol phosphatase SHIP to enhance activation of the PI3K/Akt pathway, which in turn promotes favorable responses for the host. Here we examined how miR-155 expression is regulated during infection. First, our data demonstrate that miR-155 can be induced through soluble factors of bacterial origin and not the host. Second, miR-155 induction is not a direct effect of infection and it requires NF-κB signaling to up-regulate fos/jun transcription factors. Finally, we demonstrate that the requirement for NF-κB-dependent de novo protein synthesis is globally shared by microbial ligands and live bacteria. This study provides new insight into the complex regulation of miR-155 during microbial infection.
- Published
- 2012
- Full Text
- View/download PDF
26. Mycobacterium tuberculosis lipomannan blocks TNF biosynthesis by regulating macrophage MAPK-activated protein kinase 2 (MK2) and microRNA miR-125b.
- Author
-
Rajaram MV, Ni B, Morris JD, Brooks MN, Carlson TK, Bakthavachalu B, Schoenberg DR, Torrelles JB, and Schlesinger LS
- Subjects
- Humans, In Vitro Techniques, Lipopolysaccharides pharmacology, MAP Kinase Signaling System immunology, Macrophages drug effects, Macrophages metabolism, Macrophages microbiology, MicroRNAs genetics, Mycobacterium tuberculosis pathogenicity, Phosphorylation, Promoter Regions, Genetic, Proto-Oncogene Proteins c-akt metabolism, RNA Stability, Toll-Like Receptor 2 metabolism, Tumor Necrosis Factor-alpha genetics, Virulence immunology, p38 Mitogen-Activated Protein Kinases metabolism, Lipopolysaccharides immunology, MAP Kinase Kinase 2 metabolism, Macrophages immunology, MicroRNAs metabolism, Mycobacterium tuberculosis immunology, Tumor Necrosis Factor-alpha biosynthesis
- Abstract
Contact of Mycobacterium tuberculosis (M.tb) with the immune system requires interactions between microbial surface molecules and host pattern recognition receptors. Major M.tb-exposed cell envelope molecules, such as lipomannan (LM), contain subtle structural variations that affect the nature of the immune response. Here we show that LM from virulent M.tb (TB-LM), but not from avirulent Myocobacterium smegmatis (SmegLM), is a potent inhibitor of TNF biosynthesis in human macrophages. This difference in response is not because of variation in Toll-like receptor 2-dependent activation of the signaling kinase MAPK p38. Rather, TB-LM stimulation leads to destabilization of TNF mRNA transcripts and subsequent failure to produce TNF protein. In contrast, SmegLM enhances MAPK-activated protein kinase 2 phosphorylation, which is critical for maintaining TNF mRNA stability in part by contributing microRNAs (miRNAs). In this context, human miRNA miR-125b binds to the 3' UTR region of TNF mRNA and destabilizes the transcript, whereas miR-155 enhances TNF production by increasing TNF mRNA half-life and limiting expression of SHIP1, a negative regulator of the PI3K/Akt pathway. We show that macrophages incubated with TB-LM and live M.tb induce high miR-125b expression and low miR-155 expression with correspondingly low TNF production. In contrast, SmegLM and live M. smegmatis induce high miR-155 expression and low miR-125b expression with high TNF production. Thus, we identify a unique cellular mechanism underlying the ability of a major M.tb cell wall component, TB-LM, to block TNF biosynthesis in human macrophages, thereby allowing M.tb to subvert host immunity and potentially increase its virulence.
- Published
- 2011
- Full Text
- View/download PDF
27. NOD2 controls the nature of the inflammatory response and subsequent fate of Mycobacterium tuberculosis and M. bovis BCG in human macrophages.
- Author
-
Brooks MN, Rajaram MV, Azad AK, Amer AO, Valdivia-Arenas MA, Park JH, Núñez G, and Schlesinger LS
- Subjects
- Animals, Blotting, Western, Cell Differentiation, Cells, Cultured, Enzyme-Linked Immunosorbent Assay, Humans, Immunoprecipitation, Inflammation immunology, Interleukin-1beta metabolism, Intracellular Signaling Peptides and Proteins genetics, Macrophages metabolism, Mice, Mice, Inbred C57BL, Microscopy, Confocal, Nod2 Signaling Adaptor Protein genetics, Protein-Tyrosine Kinases genetics, RNA, Small Interfering genetics, Receptor-Interacting Protein Serine-Threonine Kinase 2 genetics, Reverse Transcriptase Polymerase Chain Reaction, Signal Transduction, Syk Kinase, Toll-Like Receptor 2 genetics, Tumor Necrosis Factor-alpha metabolism, Macrophages immunology, Macrophages microbiology, Mycobacterium bovis immunology, Mycobacterium tuberculosis immunology, Nod2 Signaling Adaptor Protein metabolism
- Abstract
Mycobacterium tuberculosis (M.tb), which causes tuberculosis, is a host-adapted intracellular pathogen of macrophages. Intracellular pattern recognition receptors in macrophages such as nucleotide-binding oligomerization domain (NOD) proteins regulate pro-inflammatory cytokine production. NOD2-mediated signalling pathways in response to M.tb have been studied primarily in mouse models and cell lines but not in primary human macrophages. Thus we sought to determine the role of NOD2 in regulating cytokine production and growth of virulent M.tb and attenuated Mycobacterium bovis BCG (BCG) in human macrophages. We examined NOD2 expression during monocyte differentiation and observed a marked increase in NOD2 transcript and protein following 2-3 days in culture. Pre-treatment of human monocyte-derived and alveolar macrophages with the NOD2 ligand muramyl dipeptide enhanced production of TNF-α and IL-1β in response to M.tb and BCG in a RIP2-dependent fashion. The NOD2-mediated cytokine response was significantly reduced following knock-down of NOD2 expression by using small interfering RNA (siRNA) in human macrophages. Finally, NOD2 controlled the growth of both M.tb and BCG in human macrophages, whereas controlling only BCG growth in murine macrophages. Together, our results provide evidence that NOD2 is an important intracellular receptor in regulating the host response to M.tb and BCG infection in human macrophages., (© 2010 Blackwell Publishing Ltd.)
- Published
- 2011
- Full Text
- View/download PDF
28. Mycobacterium tuberculosis activates human macrophage peroxisome proliferator-activated receptor gamma linking mannose receptor recognition to regulation of immune responses.
- Author
-
Rajaram MV, Brooks MN, Morris JD, Torrelles JB, Azad AK, and Schlesinger LS
- Subjects
- Anilides pharmacology, Blotting, Western, Cells, Cultured, Cyclooxygenase 2 metabolism, Enzyme Activation drug effects, Host-Pathogen Interactions immunology, Humans, Interleukin-8 metabolism, Lectins, C-Type genetics, Lipopolysaccharides immunology, Lipopolysaccharides pharmacology, Macrophages cytology, Macrophages microbiology, Mannose Receptor, Mannose-Binding Lectins genetics, Models, Immunological, Mycobacterium tuberculosis physiology, NF-kappa B metabolism, PPAR gamma antagonists & inhibitors, PPAR gamma genetics, Phospholipases A2, Cytosolic metabolism, RNA Interference, Receptors, Cell Surface genetics, Signal Transduction drug effects, Signal Transduction immunology, Tumor Necrosis Factor-alpha metabolism, p38 Mitogen-Activated Protein Kinases metabolism, Lectins, C-Type metabolism, Macrophages metabolism, Mannose-Binding Lectins metabolism, Mycobacterium tuberculosis immunology, PPAR gamma metabolism, Receptors, Cell Surface metabolism
- Abstract
Mycobacterium tuberculosis enhances its survival in macrophages by suppressing immune responses in part through its complex cell wall structures. Peroxisome proliferator-activated receptor gamma (PPARgamma), a nuclear receptor superfamily member, is a transcriptional factor that regulates inflammation and has high expression in alternatively activated alveolar macrophages and macrophage-derived foam cells, both cell types relevant to tuberculosis pathogenesis. In this study, we show that virulent M. tuberculosis and its cell wall mannose-capped lipoarabinomannan induce PPARgamma expression through a macrophage mannose receptor-dependent pathway. When activated, PPARgamma promotes IL-8 and cyclooxygenase 2 expression, a process modulated by a PPARgamma agonist or antagonist. Upstream, MAPK-p38 mediates cytosolic phospholipase A(2) activation, which is required for PPARgamma ligand production. The induced IL-8 response mediated by mannose-capped lipoarabinomannan and the mannose receptor is independent of TLR2 and NF-kappaB activation. In contrast, the attenuated Mycobacterium bovis bacillus Calmette-Guérin induces less PPARgamma and preferentially uses the NF-kappaB-mediated pathway to induce IL-8 production. Finally, PPARgamma knockdown in human macrophages enhances TNF production and controls the intracellular growth of M. tuberculosis. These data identify a new molecular pathway that links engagement of the mannose receptor, an important pattern recognition receptor for M. tuberculosis, with PPARgamma activation, which regulates the macrophage inflammatory response, thereby playing a role in tuberculosis pathogenesis.
- Published
- 2010
- Full Text
- View/download PDF
29. Francisella acid phosphatases inactivate the NADPH oxidase in human phagocytes.
- Author
-
Mohapatra NP, Soni S, Rajaram MV, Dang PM, Reilly TJ, El-Benna J, Clay CD, Schlesinger LS, and Gunn JS
- Subjects
- Acid Phosphatase genetics, Animals, Cells, Cultured, Francisella tularensis growth & development, Humans, Intracellular Fluid enzymology, Intracellular Fluid immunology, Intracellular Fluid microbiology, Isoenzymes genetics, Isoenzymes physiology, Macrophages enzymology, Macrophages immunology, Macrophages microbiology, Mice, Mice, Inbred C57BL, Mice, Knockout, NADPH Oxidases biosynthesis, Neutrophils enzymology, Neutrophils immunology, Neutrophils microbiology, Phagocytes immunology, Phosphorylation immunology, Reactive Oxygen Species metabolism, Respiratory Burst immunology, Acid Phosphatase physiology, Francisella tularensis enzymology, Francisella tularensis immunology, NADPH Oxidases antagonists & inhibitors, NADPH Oxidases metabolism, Phagocytes enzymology, Phagocytes microbiology
- Abstract
Francisella tularensis contains four putative acid phosphatases that are conserved in Francisella novicida. An F. novicida quadruple mutant (AcpA, AcpB, AcpC, and Hap [DeltaABCH]) is unable to escape the phagosome or survive in macrophages and is attenuated in the mouse model. We explored whether reduced survival of the DeltaABCH mutant within phagocytes is related to the oxidative response by human neutrophils and macrophages. F. novicida and F. tularensis subspecies failed to stimulate reactive oxygen species production in the phagocytes, whereas the F. novicida DeltaABCH strain stimulated a significant level of reactive oxygen species. The DeltaABCH mutant, but not the wild-type strain, strongly colocalized with p47(phox) and replicated in phagocytes only in the presence of an NADPH oxidase inhibitor or within macrophages isolated from p47(phox) knockout mice. Finally, purified AcpA strongly dephosphorylated p47(phox) and p40(phox), but not p67(phox), in vitro. Thus, Francisella acid phosphatases play a major role in intramacrophage survival and virulence by regulating the generation of the oxidative burst in human phagocytes.
- Published
- 2010
- Full Text
- View/download PDF
30. Mannose receptor-dependent delay in phagosome maturation by Mycobacterium avium glycopeptidolipids.
- Author
-
Sweet L, Singh PP, Azad AK, Rajaram MV, Schlesinger LS, and Schorey JS
- Subjects
- Animals, CHO Cells, Cricetinae, Cricetulus, Humans, Macrophages metabolism, Mannose Receptor, Mice, Mice, Knockout, Lectins, C-Type metabolism, Lipids pharmacology, Lipids physiology, Mannose-Binding Lectins metabolism, Mycobacterium avium metabolism, Phagosomes physiology, Receptors, Cell Surface metabolism
- Abstract
The ability of pathogenic mycobacteria to block phagosome-lysosome fusion is critical for its pathogenesis. The molecules expressed by mycobacteria that inhibit phagosome maturation and the mechanism of this inhibition have been extensively studied. Recent work has indicated that mannosylated lipoarabinomannan (ManLAM) isolated from Mycobacterium tuberculosis can function to delay phagosome-lysosome fusion and that this delay requires the interaction of ManLAM with the mannose receptor (MR). However, the molecules expressed by other pathogenic mycobacteria that function to inhibit phagosome maturation have not been well described. In the present study, we show that phagosomes containing silica beads coated with glycopeptidolipids (GPLs), a major surface component of Mycobacterium avium, showed limited acidification and delayed recruitment of late endosomal/lysosomal markers compared to those of phosphatidylcholine-coated beads. The carbohydrate component of the GPLs was required, as beads coated only with the lipopeptide core failed to delay phagosome-lysosome fusion. Moreover, the ability of GPLs to delay phagosome maturation was dependent on the macrophage expression of the MR. Using CHO cells expressing the MR, we confirmed that the GPLs bind this receptor. Finally, human monocyte-derived macrophages knocked down for MR expression showed increased M. avium phagosome-lysosome fusion relative to control cells. Together, the data indicate that GPLs can function to delay phagosome-lysosome fusion and suggest that GPLs, like ManLAM, work through the MR to mediate this activity.
- Published
- 2010
- Full Text
- View/download PDF
31. Akt and SHIP modulate Francisella escape from the phagosome and induction of the Fas-mediated death pathway.
- Author
-
Rajaram MV, Butchar JP, Parsa KV, Cremer TJ, Amer A, Schlesinger LS, and Tridandapani S
- Subjects
- Animals, Bone Marrow Cells cytology, Cell Death, Cell Line, Cytosol metabolism, Inositol Polyphosphate 5-Phosphatases, Lysosomes metabolism, Macrophages cytology, Mice, Mice, Inbred C57BL, Phagocytosis, Francisella metabolism, Phagosomes metabolism, Phosphoric Monoester Hydrolases metabolism, Proto-Oncogene Proteins c-akt metabolism, fas Receptor metabolism
- Abstract
Francisella tularensis infects macrophages and escapes phago-lysosomal fusion to replicate within the host cytosol, resulting in host cell apoptosis. Here we show that the Fas-mediated death pathway is activated in infected cells and correlates with escape of the bacterium from the phagosome and the bacterial burden. Our studies also demonstrate that constitutive activation of Akt, or deletion of SHIP, promotes phago-lysosomal fusion and limits bacterial burden in the host cytosol, and the subsequent induction of Fas expression and cell death. Finally, we show that phagosomal escape/intracellular bacterial burden regulate activation of the transcription factors sp1/sp3, leading to Fas expression and cell death. These data identify for the first time host cell signaling pathways that regulate the phagosomal escape of Francisella, leading to the induction of Fas and subsequent host cell death.
- Published
- 2009
- Full Text
- View/download PDF
32. Francisella gains a survival advantage within mononuclear phagocytes by suppressing the host IFNgamma response.
- Author
-
Parsa KV, Butchar JP, Rajaram MV, Cremer TJ, Gunn JS, Schlesinger LS, and Tridandapani S
- Subjects
- Animals, Cell Line, Cell Proliferation drug effects, Francisella tularensis drug effects, Humans, Interferon-gamma pharmacology, Leukocytes, Mononuclear drug effects, Macrophages drug effects, Macrophages enzymology, Macrophages microbiology, Mice, Microbial Viability drug effects, Models, Immunological, Monocytes drug effects, Monocytes immunology, Monocytes microbiology, Nitric Oxide biosynthesis, Nitric Oxide Synthase Type II metabolism, Phagocytes drug effects, Phagosomes drug effects, Phagosomes microbiology, Phosphorylation drug effects, STAT1 Transcription Factor metabolism, Suppressor of Cytokine Signaling 3 Protein, Suppressor of Cytokine Signaling Proteins metabolism, Tularemia immunology, Tularemia microbiology, Francisella tularensis cytology, Interferon-gamma immunology, Leukocytes, Mononuclear immunology, Leukocytes, Mononuclear microbiology, Microbial Viability immunology, Phagocytes immunology, Phagocytes microbiology
- Abstract
Tularemia is a zoonotic disease caused by the Gram-negative intracellular pathogen Francisella tularensis. These bacteria evade phagolysosomal fusion, escape from the phagosome and replicate in the host cell cytoplasm. IFNgamma has been shown to suppress the intra-macrophage growth of Francisella through both nitric oxide-dependent and -independent pathways. Since Francisella is known to subvert host immune responses, we hypothesized that this pathogen could interfere with IFNgamma signaling. Here, we report that infection with Francisella suppresses IFNgamma-induced STAT1 expression and phosphorylation in both human and murine mononuclear phagocytes. This suppressive effect of Francisella is independent of phagosomal escape or replication and is mediated by a heat-stable and constitutively expressed bacterial factor. An analysis of the molecular mechanism of STAT1 inhibition indicated that expression of SOCS3, an established negative regulator of IFNgamma signaling, is highly up-regulated during infection and suppresses STAT1 phosphorylation. Functional analyses revealed that this interference with IFNgamma signaling is accompanied by the suppression of IP-10 production and iNOS induction resulting in increased intracellular bacterial survival. Importantly, the suppressive effect on IFNgamma-mediated host cell protection is most effective when IFNgamma is added post infection, suggesting that the bacteria establish a permissive environment within the host cell.
- Published
- 2008
- Full Text
- View/download PDF
33. The tyrosine kinase Syk promotes phagocytosis of Francisella through the activation of Erk.
- Author
-
Parsa KV, Butchar JP, Rajaram MV, Cremer TJ, and Tridandapani S
- Subjects
- Animals, Cell Line, Enzyme Activation drug effects, Francisella drug effects, Gram-Negative Bacterial Infections enzymology, Gram-Negative Bacterial Infections microbiology, Humans, Intracellular Signaling Peptides and Proteins antagonists & inhibitors, Macrophages cytology, Macrophages drug effects, Macrophages enzymology, Macrophages microbiology, Mice, Mitogen-Activated Protein Kinase 1 antagonists & inhibitors, Mitogen-Activated Protein Kinase 3 antagonists & inhibitors, Phosphatidylinositol 3-Kinases metabolism, Phosphorylation drug effects, Protein Kinase Inhibitors pharmacology, Protein-Tyrosine Kinases antagonists & inhibitors, Proto-Oncogene Proteins c-akt metabolism, Syk Kinase, Francisella cytology, Intracellular Signaling Peptides and Proteins metabolism, Mitogen-Activated Protein Kinase 1 metabolism, Mitogen-Activated Protein Kinase 3 metabolism, Phagocytosis drug effects, Protein-Tyrosine Kinases metabolism
- Abstract
Francisella tularensis is a highly infectious, Gram-negative intra-cellular pathogen that can cause the zoonotic disease tularemia. Although the receptors critical for internalization of Francisella by macrophages are beginning to be defined, the identity of the downstream signaling pathways essential for the engulfment are not yet identified. In this study we have tested the role of Syk in the phagocytosis of Francisella. We report that Syk is activated during Francisella infection and is critical for the uptake of the organisms. Pharmacologic inhibition of Syk almost completely abrogated uptake, whereas the overexpression of Syk significantly enhanced uptake. However, Syk appears to be dispensable during initial host-pathogen contact. Further analyses of the molecular mechanism of Syk influence on Francisella uptake revealed that the MAPK Erk but not the phosphatidylinositol 3 kinase (PI3K)/Akt pathway is the downstream effector of Syk. Thus, the inhibition of Erk in Syk-overexpressing cells or the inhibition of Syk in Erk-overexpressing cells led to a significant attenuation of uptake. Collectively, these data identify Syk and Erk as key players in the phagocytosis of Francisella.
- Published
- 2008
- Full Text
- View/download PDF
34. Francisella tularensis induces IL-23 production in human monocytes.
- Author
-
Butchar JP, Rajaram MV, Ganesan LP, Parsa KV, Clay CD, Schlesinger LS, and Tridandapani S
- Subjects
- Animals, Blotting, Western, Cells, Cultured, Cytokines genetics, Cytokines metabolism, Enzyme-Linked Immunosorbent Assay, Francisella tularensis pathogenicity, Humans, Interleukin-23 genetics, Mice, Monocytes microbiology, NF-kappa B metabolism, Oligonucleotide Array Sequence Analysis, Phagosomes immunology, Phosphatidylinositol 3-Kinases metabolism, Transcription, Genetic, Up-Regulation, Francisella tularensis immunology, Interleukin-23 metabolism, Monocytes immunology
- Abstract
Francisella tularensis, the causative agent of tularemia, is phagocytosed by immune cells such as monocytes and macrophages. Instead of being destroyed in the phagolysosome, the bacterium escapes the phagosome and replicates within the host cytosol. Recent studies indicate that phagosomal escape may have a major impact on the nature of the inflammatory cytokine response to infection. To better understand the host cell response to Francisella infection, we exposed human peripheral blood monocytes to Francisella novicida and analyzed transcriptional changes using high-density oligonucleotide microarrays. Results showed a nearly 300-fold up-regulation of transcripts for the p19 subunit of IL-23, and a nearly 18-fold up-regulation for the p40 subunit of IL-12. IL-23 is formed by the heterodimerization of p19 and p40, and is an important cytokine of the innate immune response. Up-regulation of p19 and p40 was confirmed at the protein level by Western blotting and ELISA analyses, and was found to be largely dependent on PI3K and NF-kappaB activity. Studies using medium from infected monocytes with or without a p19 blocking Ab showed that the secreted IL-23 induced IFN-gamma production from NK cells, suggesting a potential biologically important role for IL-23 in host defense. Finally, infection of human monocytes by the highly virulent Francisella SCHU S4 strain likewise led to IL-23 production, suggesting that the IL-23 response may be relevant during tularemia.
- Published
- 2007
- Full Text
- View/download PDF
35. Akt/Protein kinase B modulates macrophage inflammatory response to Francisella infection and confers a survival advantage in mice.
- Author
-
Rajaram MV, Ganesan LP, Parsa KV, Butchar JP, Gunn JS, and Tridandapani S
- Subjects
- Animals, Cytokines genetics, Enzyme Activation, Genes, Bacterial physiology, Macrophages immunology, Mice, NF-kappa B metabolism, Phagosomes immunology, Phagosomes microbiology, Proto-Oncogene Proteins c-akt antagonists & inhibitors, Survival Analysis, Transcription, Genetic, Cytokines metabolism, Francisella genetics, Gram-Negative Bacterial Infections enzymology, Macrophages enzymology, Macrophages microbiology, Proto-Oncogene Proteins c-akt metabolism
- Abstract
The Gram-negative bacterium Francisella novicida infects primarily monocytes/macrophages and is highly virulent in mice. Macrophages respond by producing inflammatory cytokines that confer immunity against the infection. However, the molecular details of host cell response to Francisella infection are poorly understood. In this study, we demonstrate that F. novicida infection of murine macrophages induces the activation of Akt. Inhibition of Akt significantly decreases proinflammatory cytokine production in infected macrophages, whereas production of the anti-inflammatory cytokine IL-10 is enhanced. Analysis of the mechanism of Akt influence on cytokine response demonstrated that Akt promotes NF-kappaB activation. We have extended these findings to show that Akt activation may be regulated by bacterial genes associated with phagosomal escape. Infection with mglA mutants of F. novicida elicited sustained activation of Akt in comparison to cells infected with wild-type F. novicida. Concomitantly, there was significantly higher proinflammatory cytokine production and lower IL-10 production in cells infected with the mglA mutant. Finally, transgenic animals expressing constitutively active Akt displayed a survival advantage over their wild-type littermates when challenged with lethal doses of F. novicida. Together, these observations indicate that Akt promotes proinflammatory cytokine production by F. novicida-infected macrophages through its influence on NF-kappaB, thereby contributing to immunity against F. novicida infection.
- Published
- 2006
- Full Text
- View/download PDF
36. Macrophage pro-inflammatory response to Francisella novicida infection is regulated by SHIP.
- Author
-
Parsa KV, Ganesan LP, Rajaram MV, Gavrilin MA, Balagopal A, Mohapatra NP, Wewers MD, Schlesinger LS, Gunn JS, and Tridandapani S
- Subjects
- Animals, Cells, Cultured, Cytokines metabolism, Down-Regulation, Inositol Polyphosphate 5-Phosphatases, Interleukin-10 biosynthesis, Mice, NF-kappa B metabolism, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism, Francisella, Gram-Negative Bacterial Infections metabolism, Gram-Negative Bacterial Infections pathology, Inflammation Mediators metabolism, Macrophages metabolism, Phosphoric Monoester Hydrolases metabolism
- Abstract
Francisella tularensis, a Gram-negative facultative intracellular pathogen infecting principally macrophages and monocytes, is the etiological agent of tularemia. Macrophage responses to F. tularensis infection include the production of pro-inflammatory cytokines such as interleukin (IL)-12, which is critical for immunity against infection. Molecular mechanisms regulating production of these inflammatory mediators are poorly understood. Herein we report that the SH2 domain-containing inositol phosphatase (SHIP) is phosphorylated upon infection of primary murine macrophages with the genetically related F. novicida, and negatively regulates F. novicida-induced cytokine production. Analyses of the molecular details revealed that in addition to activating the MAP kinases, F. novicida infection also activated the phosphatidylinositol 3-kinase (PI3K)/Akt pathway in these cells. Interestingly, SHIP-deficient macrophages displayed enhanced Akt activation upon F. novicida infection, suggesting elevated PI3K-dependent activation pathways in absence of SHIP. Inhibition of PI3K/Akt resulted in suppression of F. novicida-induced cytokine production through the inhibition of NFkappaB. Consistently, macrophages lacking SHIP displayed enhanced NFkappaB-driven gene transcription, whereas overexpression of SHIP led to decreased NFkappaB activation. Thus, we propose that SHIP negatively regulates F. novicida-induced inflammatory cytokine response by antagonizing the PI3K/Akt pathway and suppressing NFkappaB-mediated gene transcription. A detailed analysis of phosphoinositide signaling may provide valuable clues for better understanding the pathogenesis of tularemia.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.