51
|
Hoonakker ME, Verhagen LM, Pupo E, de Haan A, Metz B, Hendriksen CFM, Han WGH, Sloots A. Vaccine-Mediated Activation of Human TLR4 Is Affected by Modulation of Culture Conditions during Whole-Cell Pertussis Vaccine Preparation. PLoS One 2016; 11:e0161428. [PMID: 27548265 PMCID: PMC4993483 DOI: 10.1371/journal.pone.0161428] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 08/05/2016] [Indexed: 12/01/2022] Open
Abstract
The potency of whole-cell pertussis (wP) vaccines is still determined by an intracerebral mouse protection test. To allow development of suitable in vitro alternatives to this test, insight into relevant parameters to monitor the consistency of vaccine quality is essential. To this end, a panel of experimental wP vaccines of varying quality was prepared by sulfate-mediated suppression of the BvgASR master virulence regulatory system of Bordetella pertussis during cultivation. This system regulates the transcription of a range of virulence proteins, many of which are considered important for the induction of effective host immunity. The protein compositions and in vivo potencies of the vaccines were BvgASR dependent, with the vaccine containing the highest amount of virulence proteins having the highest in vivo potency. Here, the capacities of these vaccines to stimulate human Toll-like receptors (hTLR) 2 and 4 and the role these receptors play in wP vaccine-mediated activation of antigen-presenting cells in vitro were studied. Prolonged BvgASR suppression was associated with a decreased capacity of vaccines to activate hTLR4. In contrast, no significant differences in hTLR2 activation were observed. Similarly, vaccine-induced activation of MonoMac-6 and monocyte-derived dendritic cells was strongest with the highest potency vaccine. Blocking of TLR2 and TLR4 showed that differences in antigen-presenting cell activation could be largely attributed to vaccine-dependent variation in hTLR4 signalling. Interestingly, this BvgASR-dependent decrease in hTLR4 activation coincided with a reduction in GlcN-modified lipopolysaccharides in these vaccines. Accordingly, expression of the lgmA-C genes, required for this glucosamine modification, was significantly reduced in bacteria exposed to sulfate. Together, these findings demonstrate that the BvgASR status of bacteria during wP vaccine preparation is critical for their hTLR4 activation capacity and suggest that including such parameters to assess consistency of newly produced vaccines could bring in vitro testing of vaccine quality a step closer.
Collapse
Affiliation(s)
- Marieke E. Hoonakker
- Institute for Translational Vaccinology (Intravacc), Bilthoven, The Netherlands
- Department of Animals in Science and Society, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
- * E-mail:
| | - Lisa M. Verhagen
- Institute for Translational Vaccinology (Intravacc), Bilthoven, The Netherlands
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Elder Pupo
- Institute for Translational Vaccinology (Intravacc), Bilthoven, The Netherlands
| | - Alex de Haan
- Institute for Translational Vaccinology (Intravacc), Bilthoven, The Netherlands
| | - Bernard Metz
- Institute for Translational Vaccinology (Intravacc), Bilthoven, The Netherlands
| | - Coenraad F. M. Hendriksen
- Institute for Translational Vaccinology (Intravacc), Bilthoven, The Netherlands
- Department of Animals in Science and Society, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Wanda G. H. Han
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Arjen Sloots
- Institute for Translational Vaccinology (Intravacc), Bilthoven, The Netherlands
| |
Collapse
|
52
|
Steven S, Jurk K, Kopp M, Kröller-Schön S, Mikhed Y, Schwierczek K, Roohani S, Kashani F, Oelze M, Klein T, Tokalov S, Danckwardt S, Strand S, Wenzel P, Münzel T, Daiber A. Glucagon-like peptide-1 receptor signalling reduces microvascular thrombosis, nitro-oxidative stress and platelet activation in endotoxaemic mice. Br J Pharmacol 2016; 174:1620-1632. [PMID: 27435156 DOI: 10.1111/bph.13549] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 06/01/2016] [Accepted: 07/08/2016] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND AND PURPOSE Excessive inflammation in sepsis causes microvascular thrombosis and thrombocytopenia associated with organ dysfunction and high mortality. The present studies aimed to investigate whether inhibition of dipeptidyl peptidase-4 (DPP-4) and supplementation with glucagon-like peptide-1 (GLP-1) receptor agonists improved endotoxaemia-associated microvascular thrombosis via immunomodulatory effects. EXPERIMENTAL APPROACH Endotoxaemia was induced in C57BL/6J mice by a single injection of LPS (17.5 mg kg-1 for survival and 10 mg kg-1 for all other studies). For survival studies, treatment was started 6 h after LPS injection. For all other studies, drugs were injected 48 h before LPS treatment. KEY RESULTS Mice treated with LPS alone showed severe thrombocytopenia, microvascular thrombosis in the pulmonary circulation (fluorescence imaging), increased LDH activity, endothelial dysfunction and increased markers of inflammation in aorta and whole blood (leukocyte-dependent oxidative burst, nitrosyl-iron haemoglobin, a marker of nitrosative stress, and expression of inducible NOS). Treatment with the DPP-4 inhibitor linagliptin or the GLP-1 receptor agonist liraglutide, as well as genetic deletion of DPP-4 (DPP4-/- mice) improved all these parameters. In GLP-1 receptor-deficient mice, both linagliptin and liraglutide lost their beneficial effects and improvement of prognosis. Incubation of platelets and cultured monocytes (containing GLP-1 receptor protein) with GLP-1 receptor agonists inhibited the monocytic oxidative burst and platelet activation, with a GLP-1 receptor-dependent elevation of cAMP levels and PKA activation. CONCLUSIONS AND IMPLICATIONS GLP-1 receptor activation in platelets by linagliptin and liraglutide strongly attenuated endotoxaemia-induced microvascular thrombosis and mortality by a cAMP/PKA-dependent mechanism, preventing systemic inflammation, vascular dysfunction and end organ damage. LINKED ARTICLES This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.
Collapse
Affiliation(s)
- Sebastian Steven
- Centre for Cardiology, Cardiology I, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Kerstin Jurk
- Center for Thrombosis and Hemostasis, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Maximilian Kopp
- Centre for Cardiology, Cardiology I, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Swenja Kröller-Schön
- Centre for Cardiology, Cardiology I, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Yuliya Mikhed
- Centre for Cardiology, Cardiology I, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Kathrin Schwierczek
- Center for Thrombosis and Hemostasis, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Siyer Roohani
- Centre for Cardiology, Cardiology I, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Fatemeh Kashani
- Centre for Cardiology, Cardiology I, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Matthias Oelze
- Centre for Cardiology, Cardiology I, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Thomas Klein
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Sergey Tokalov
- Center for Thrombosis and Hemostasis, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Sven Danckwardt
- Center for Thrombosis and Hemostasis, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany.,Institute of Clinical Chemistry and Laboratory Medicine, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Susanne Strand
- I. Department of Internal Medicine, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Philip Wenzel
- Centre for Cardiology, Cardiology I, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Thomas Münzel
- Centre for Cardiology, Cardiology I, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Andreas Daiber
- Centre for Cardiology, Cardiology I, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
53
|
Zhang L, Stuber F, Lippuner C, Schiff M, Stamer UM. Phorbol-12-myristate-13-acetate induces nociceptin in human Mono Mac 6 cells via multiple transduction signalling pathways. Br J Anaesth 2016; 117:250-7. [PMID: 27307289 DOI: 10.1093/bja/aew063] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2016] [Indexed: 12/16/2023] Open
Abstract
BACKGROUND Nociceptin in the peripheral circulation has been proposed to have an immunoregulatory role with regards to inflammation and pain. However, the mechanisms involved in its regulation are still not clear. The aim of this study was to investigate signalling pathways contributing to the regulation of the expression of nociceptin under inflammatory conditions. METHODS Mono Mac 6 cells (MM6) were cultured with or without phorbol-12-myristate-13-acetate (PMA). Prepronociceptin (ppNOC) mRNA was detected by RT-qPCR and extracellular nociceptin by fluorescent-enzyme immunoassay. Intracellular nociceptin and phosphorylated kinases were measured using flow cytometry. To evaluate the contribution of various signalling pathways to the regulation of ppNOC mRNA and nociceptin protein, cells were pre-treated with specific kinase inhibitors before co-culturing with PMA. RESULTS ppNOC mRNA was expressed in untreated MM6 at low concentrations. Exposure of cells to PMA upregulated ppNOC after nine h compared with controls without PMA (median normalized ratio with IQR: 0.18 (0.15-0.26) vs. 0 (0-0.02), P<0.01). Inhibition of mitogen-activated protein kinases specific for signal transduction reversed the PMA effects (all P<0.001). Induction of nociceptin protein concentrations in PMA stimulated MM6 was prevented predominantly by identity of ERK inhibitor (P<0.05). CONCLUSIONS Upregulation of nociceptin expression by PMA in MM6 cells involves several pathways. Underlying mechanisms involved in nociceptin expression may lead to new insights in the treatment of pain and inflammatory diseases.
Collapse
Affiliation(s)
- L Zhang
- Department of Anaesthesiology and Pain Medicine, Inselspital and Department of Clinical Research, University of Bern, Bern, Switzerland
| | - F Stuber
- Department of Anaesthesiology and Pain Medicine, Inselspital and Department of Clinical Research, University of Bern, Bern, Switzerland
| | - C Lippuner
- Department of Anaesthesiology and Pain Medicine, Inselspital and Department of Clinical Research, University of Bern, Bern, Switzerland
| | - M Schiff
- Department of Anaesthesiology and Pain Medicine, Inselspital and Department of Clinical Research, University of Bern, Bern, Switzerland
| | - U M Stamer
- Department of Anaesthesiology and Pain Medicine, Inselspital and Department of Clinical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
54
|
Stoppelkamp S, Würschum N, Stang K, Löder J, Avci-Adali M, Toliashvili L, Schlensak C, Wendel HP, Fennrich S. Speeding up pyrogenicity testing: Identification of suitable cell components and readout parameters for an accelerated monocyte activation test (MAT). Drug Test Anal 2016; 9:260-273. [DOI: 10.1002/dta.1973] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 02/23/2016] [Accepted: 02/24/2016] [Indexed: 01/28/2023]
Affiliation(s)
- Sandra Stoppelkamp
- University Hospital Tuebingen; Clinic for Thoracic, Cardiac and Vascular Surgery; Calwerstr. 7/1 72076 Tuebingen Germany
| | - Noriana Würschum
- University Hospital Tuebingen; Clinic for Thoracic, Cardiac and Vascular Surgery; Calwerstr. 7/1 72076 Tuebingen Germany
| | - Katharina Stang
- University Hospital Tuebingen; Clinic for Thoracic, Cardiac and Vascular Surgery; Calwerstr. 7/1 72076 Tuebingen Germany
| | - Jasmin Löder
- University Hospital Tuebingen; Clinic for Thoracic, Cardiac and Vascular Surgery; Calwerstr. 7/1 72076 Tuebingen Germany
| | - Meltem Avci-Adali
- University Hospital Tuebingen; Clinic for Thoracic, Cardiac and Vascular Surgery; Calwerstr. 7/1 72076 Tuebingen Germany
| | - Leila Toliashvili
- University Hospital Tuebingen; Clinic for Thoracic, Cardiac and Vascular Surgery; Calwerstr. 7/1 72076 Tuebingen Germany
| | - Christian Schlensak
- University Hospital Tuebingen; Clinic for Thoracic, Cardiac and Vascular Surgery; Calwerstr. 7/1 72076 Tuebingen Germany
| | - Hans Peter Wendel
- University Hospital Tuebingen; Clinic for Thoracic, Cardiac and Vascular Surgery; Calwerstr. 7/1 72076 Tuebingen Germany
| | - Stefan Fennrich
- University Hospital Tuebingen; Clinic for Thoracic, Cardiac and Vascular Surgery; Calwerstr. 7/1 72076 Tuebingen Germany
| |
Collapse
|
55
|
Siad S, Byrne S, Mukamolova G, Stover C. Intracellular localisation of Mycobacterium marinum in mast cells. World J Immunol 2016; 6:83-95. [DOI: 10.5411/wji.v6.i1.83] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 10/30/2015] [Accepted: 02/16/2016] [Indexed: 02/05/2023] Open
Abstract
AIM: To study the bacteriocidal or bacteriostatic role of mast cells during infection with Mycobacterium.
METHODS: Mycobacterium marinum (M. marinum) (BAA-535/M strain) was investigated for its ability to grow at a temperature relevant to the mammalian host. Primary mast cells were differentiated from bone marrows of mice, a human mast cell line (HMC-1) and a human monocytic cell line (MonoMac6) were maintained in culture. Mice were stimulated by intraperitoneal injection of heat-killed M. marinum to study cytochemically the degranulation of peritoneal mast cells. HMC-1 cells were stimulated with M. marinum to analyse mRNA expression for inflammatory reactant genes, while HMC-1 and primary mouse mast cells were infected with M. marinum to establish in parallel cell viability (lactate dehydrogenase release and cell counts) and viable mycobacterial counts. Flow cytometry was used to assess intracellular presence of fluorescein isothiocyanate labelled M. marinum after trypan blue quenching and to measure the extent of infection-induced apoptosis or necrosis in HMC-1. A GFP expressing recombinant M. marinum strain was used to assess intracellular location by fluorescence microscopy. Light microscopy of osmium tetroxide and Gram Twort stained sections of 0.5 μm and transmission electron microscopy were undertaken as sensitive methods.
RESULTS: Since its isolation, M. marinum has adapted to grow at 37 °C. This study found that M. marinum infects HMC-1 cells and primary murine mast cells, where they survive, replicate, and cause dose dependent cell damage over the analysis period of up to 120 h. Amikacin was an effective aminoglycoside antibiotic to eliminate extracellular or membrane attached M. marinum in order to adequately quantify the intracellular bacterial loads. In vivo, intraperitoneal injection of heat-killed M. marinum led to the release of mast cell granules in mice. HMC-1 cells stimulated with M. marinum showed a biphasic pattern of increased mRNA expression for LL-37 and COX-2/TNF-α during 24 h of stimulation. In HMC-1, M. marinum localised to the cytoplasm whereas in primary mast cells, M. marinum were found in vacuoles.
CONCLUSION: The effector role of mast cells in infection with M. marinum can be studied in vitro and in vivo.
Collapse
|
56
|
Witola WH, Matthews K, McHugh M. In vitro anthelmintic efficacy of inhibitors of phosphoethanolamine Methyltransferases in Haemonchus contortus. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2016; 6:44-53. [PMID: 27054063 PMCID: PMC4805780 DOI: 10.1016/j.ijpddr.2016.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 01/12/2016] [Accepted: 01/12/2016] [Indexed: 12/03/2022]
Abstract
The essential phosphobase methylation pathway for synthesis of phosphocholine is unique to nematodes, protozoa and plants, and thus an attractive antiparasitic molecular target. Herein, we screened compounds from the National Cancer Institute (Developmental Therapeutics Program Open Chemical Repository) for specific inhibitory activity against Haemonchus contortus phosphoethanolamine methyltransferases (HcPMT1 and HcPMT2), and tested candidate compounds for anthelmintic activity against adult and third-stage larvae of H. contortus. We identified compound NSC-641296 with IC50 values of 8.3 ± 1.1 μM and 5.1 ± 1.8 μM for inhibition of the catalytic activity of HcPMT1 alone and HcPMT1/HcPMT2 combination, respectively. Additionally we identified compound NSC-668394 with inhibitory IC50 values of 5.9 ± 0.9 μM and 2.8 ± 0.6 μM for HcPMT1 alone and HcPMT1/HcPMT2 combination, respectively. Of the two compounds, NSC-641296 depicted significant anthelmintic activity against third-stage larvae (IC50 = 15 ± 2.9 μM) and adult stages (IC50 = 7 ± 2.9 μM) of H. contortus, with optimal effective in vitro concentrations being 2-fold and 4-fold, respectively, lower than its cytotoxic IC50 (29 ± 2.1 μM) in a mammalian cell line. Additionally, we identified two compounds, NSC-158011 and NSC-323241, with low inhibitory activity against the combined activity of HcPMT1 and HcPMT2, but both compounds did not show any anthelmintic activity against H. contortus. The identification of NSC-641296 that specifically inhibits a unique biosynthetic pathway in H. contortus and has anthelmintic activity against both larval and adult stages of H. contortus, provides impetus for the development of urgently needed new efficacious anthelmintics to address the prevailing problem of anthelmintic-resistant H. contortus. NSC-641296 and NSC-668394 inhibit HcPMT1 and HcPMT2 enzymes in Haemonchus contortus. NSC-641296 has in vitro anthelmintic activity against larvae and adult H. contortus. H. contortus HcPMT1 and HcPMT2 are two unique targets for anthelmintic development.
Collapse
Affiliation(s)
- William H Witola
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois, Urbana-Champaign, USA.
| | - Kwame Matthews
- Department of Agricultural and Environmental Sciences, Tuskegee University, Tuskegee, AL, USA
| | - Mark McHugh
- Department of Agricultural and Environmental Sciences, Tuskegee University, Tuskegee, AL, USA
| |
Collapse
|
57
|
Aasebø E, Forthun RB, Berven F, Selheim F, Hernandez-Valladares M. Global Cell Proteome Profiling, Phospho-signaling and Quantitative Proteomics for Identification of New Biomarkers in Acute Myeloid Leukemia Patients. Curr Pharm Biotechnol 2016; 17:52-70. [PMID: 26306748 PMCID: PMC5388801 DOI: 10.2174/1389201016666150826115626] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 05/29/2015] [Accepted: 07/23/2015] [Indexed: 12/31/2022]
Abstract
The identification of protein biomarkers for acute myeloid leukemia (AML) that could find applications in AML diagnosis and prognosis, treatment and the selection for bone marrow transplant requires substantial comparative analyses of the proteomes from AML patients. In the past years, several studies have suggested some biomarkers for AML diagnosis or AML classification using methods for sample preparation with low proteome coverage and low resolution mass spectrometers. However, most of the studies did not follow up, confirm or validate their candidates with more patient samples. Current proteomics methods, new high resolution and fast mass spectrometers allow the identification and quantification of several thousands of proteins obtained from few tens of μg of AML cell lysate. Enrichment methods for posttranslational modifications (PTM), such as phosphorylation, can isolate several thousands of site-specific phosphorylated peptides from AML patient samples, which subsequently can be quantified with high confidence in new mass spectrometers. While recent reports aiming to propose proteomic or phosphoproteomic biomarkers on the studied AML patient samples have taken advantage of the technological progress, the access to large cohorts of AML patients to sample from and the availability of appropriate control samples still remain challenging.
Collapse
Affiliation(s)
| | | | | | | | - Maria Hernandez-Valladares
- Department of Biomedicine, Faculty of Medicine, Building for Basic Biology, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway.
| |
Collapse
|
58
|
Substrate elasticity regulates the behavior of human monocyte-derived macrophages. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2015; 45:301-9. [PMID: 26613613 DOI: 10.1007/s00249-015-1096-8] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 11/04/2015] [Indexed: 01/25/2023]
Abstract
Macrophages play a key role in atherosclerosis, cancer, and in the response to implanted medical devices. In each of these situations, the mechanical environment of a macrophage can vary from soft to stiff. However, how stiffness affects macrophage behavior remains uncertain. Using substrates of varying stiffness, we show macrophage phenotype and function depends on substrate stiffness. Notably, the cell area increases slightly from a sphere after 18 h on substrates mimicking healthy arterial stiffness (1-5 kPa), whereas macrophages on stiffer substrates (280 kPa-70 GPa) increased in area by nearly eight-fold. Macrophage migration is random regardless of substrate stiffness. The total average track speed was 7.8 ± 0.5 μm/h, with macrophages traveling fastest on the 280-kPa substrate (12.0 ± 0.5 μm/h) and slowest on the 3-kPa substrate (5.0 ± 0.4 μm/h). In addition F-actin organization in macrophages depends on substrate stiffness. On soft substrates, F-actin is spread uniformly throughout the cytoplasm, whereas on stiff substrates F-actin is functionalized into stress fibers. The proliferation rate of macrophages was faster on stiff substrates. Cells plated on the 280-kPa gel had a significantly shorter doubling time than those plated on the softer substrate. However, the ability of macrophages to phagocytose 1-μm particles did not depend on substrate stiffness. In conclusion, the results herein show macrophages are mechanosensitive; they respond to changes in stiffness by modifying their area, migration speed, actin organization, and proliferation rate. These results are important to understanding how macrophages respond in complex mechanical environments such as an atherosclerotic plaque.
Collapse
|
59
|
Riemann A, Wußling H, Loppnow H, Fu H, Reime S, Thews O. Acidosis differently modulates the inflammatory program in monocytes and macrophages. Biochim Biophys Acta Mol Basis Dis 2015; 1862:72-81. [PMID: 26499398 DOI: 10.1016/j.bbadis.2015.10.017] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 10/15/2015] [Accepted: 10/20/2015] [Indexed: 12/13/2022]
Abstract
Inflammation, ischemia or the microenvironment of solid tumors is often accompanied by a reduction of extracellular pH (acidosis) that stresses the cells and acts on cellular signaling and transcription. The effect of acidosis on the expression of various inflammatory markers, on functional parameters (migration, phagocytic activity) and on signaling pathways involved was studied in monocytic cells and macrophages. In monocytic cell lines acidosis led to a reduction in expression of most of the inflammatory mediators, namely IL-1ß, IL-6, TNF-α, MCP-1, COX-2 and osteopontin. In primary human monocytes MCP-1 and TNF-α were reduced but COX-2 and IL-6 were increased. In RAW264.7 macrophage cell line IL-1ß, COX-2 and iNOS expression was increased, whereas MCP-1 was reduced similar to the effect in monocytic cells. For primary human monocyte-derived macrophages the regulation of inflammatory markers by acidosis depended on activation state, except for the acidosis-induced downregulation of MCP-1 and TNF-α. Acidosis affected functional immune cell behavior when looking at phagocytic activity which was increased in a time-dependent manner, but cellular motility was not changed. Neither ERK1/2 nor CREB signaling was stimulated by the reduction of extracellular pH. However, p38 was activated by acidosis in RAW264.7 cells and this activation was critical for the induction of IL-1ß, COX-2 and iNOS expression. In conclusion, acidosis may impede the recruitment of immune cells, but fosters inflammation when macrophages are present by increasing the level of COX-2 and iNOS and by functionally forcing up the phagocytic activity.
Collapse
Affiliation(s)
- Anne Riemann
- Julius-Bernstein-Institute of Physiology, University Halle-Wittenberg, Germany.
| | - Hanna Wußling
- Julius-Bernstein-Institute of Physiology, University Halle-Wittenberg, Germany
| | - Harald Loppnow
- Dept. of Internal Medicine III, University Halle-Wittenberg, Germany
| | - Hang Fu
- Dept. of Internal Medicine III, University Halle-Wittenberg, Germany
| | - Sarah Reime
- Julius-Bernstein-Institute of Physiology, University Halle-Wittenberg, Germany
| | - Oliver Thews
- Julius-Bernstein-Institute of Physiology, University Halle-Wittenberg, Germany
| |
Collapse
|
60
|
Xiong Y, Murphy M, Manavalan TT, Pattabiraman G, Qiu F, Chang HH, Ho IC, Medvedev AE. Endotoxin Tolerance Inhibits Lyn and c-Src Phosphorylation and Association with Toll-Like Receptor 4 but Increases Expression and Activity of Protein Phosphatases. J Innate Immun 2015; 8:171-84. [PMID: 26457672 DOI: 10.1159/000440838] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 09/03/2015] [Indexed: 11/19/2022] Open
Abstract
Endotoxin tolerance protects the host by limiting excessive 'cytokine storm' during sepsis, but compromises the ability to counteract infections in septic shock survivors. It reprograms Toll-like receptor (TLR) 4 responses by attenuating the expression of proinflammatory cytokines without suppressing anti-inflammatory and antimicrobial mediators, but the mechanisms of reprogramming remain unclear. In this study, we demonstrate that the induction of endotoxin tolerance in human monocytes, THP-1 and MonoMac-6 cells inhibited lipopolysaccharide (LPS)-mediated phosphorylation of Lyn, c-Src and their recruitment to TLR4, but increased total protein phosphatase (PP) activity and the expression of protein tyrosine phosphatase (PTP) 1B, PP2A, PTP nonreceptor type (PTPN) 22 and mitogen-activated protein kinase phosphatase (MKP)-1. Chemical PP inhibitors, okadaic acid, dephostatin and cantharidic acid markedly decreased or completely abolished LPS tolerance, indicating the importance of phosphatases in endotoxin tolerization. Overexpression of PTPN22 decreased LPS-mediated nuclear factor (NF)-x03BA;B activation, p38 phosphorylation and CXCL8 gene expression, while PTPN22 ablation upregulated LPS-induced p65 NF-x03BA;B and p38 phosphorylation and the expression of TNF-α and pro-IL-1β mRNA, indicating PTPN22 as an inhibitor of TLR4 signaling. Thus, LPS tolerance interferes with TLR4 signaling by inhibiting Lyn and c-Src phosphorylation and their recruitment to TLR4, while increasing the phosphatase activity and expression of PP2A, PTPN22, PTP1B and MKP1.
Collapse
Affiliation(s)
- Yanbao Xiong
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Md., USA
| | | | | | | | | | | | | | | |
Collapse
|
61
|
Kleinschmidt TK, Haraldsson M, Basavarajappa D, Lundeberg E, Thulasingam M, Ekoff M, Fauland A, Lehmann C, Kahnt AS, Lindbom L, Haeggström JZ. Tandem Benzophenone Amino Pyridines, Potent and Selective Inhibitors of Human Leukotriene C4 Synthase. J Pharmacol Exp Ther 2015; 355:108-16. [PMID: 26283693 DOI: 10.1124/jpet.115.227157] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 08/12/2015] [Indexed: 03/08/2025] Open
Abstract
Cysteinyl leukotrienes (cys-LTs) are lipid mediators of inflammation. The enzyme catalyzing synthesis of cys-LTs, leukotriene C4 synthase (LTC4S), is considered an important drug target. Here we report the synthesis and characterization of three tandem benzophenone amino pyridines as inhibitors of LTC4S in vitro and in vivo. The inhibitors were characterized in vitro using recombinant human LTC4S, MonoMac 6 cells, and a panel of peripheral human immune cells. In vivo, the compounds were tested in the Zymosan A-induced peritonitis mouse model. The molecules, denoted TK04, TK04a, and TK05, were potent and selective inhibitors of LTC4S with IC50 values of 116, 124, and 95 nM, respectively. Molecular docking revealed binding in a hydrophobic crevice between two enzyme monomers and interaction with two catalytic residues, Arg104 and Arg31. The TK compounds potently inhibited cys-LT biosynthesis in immune cells. In coincubations of platelets and polymorphonuclear leukocytes, inhibition of LTC4S led to shunting of LTA4 toward anti-inflammatory lipoxin A4, which was significantly enhanced by simultaneous inhibition of LTA4H. Finally, we found that TK05 (6 mg⋅kg(-1)⋅body weight) reduces LTE4 levels in peritoneal lavage fluid by 88% and significantly decreases vascular permeability in vivo. Our findings indicate that the TK compounds are valuable experimental tools in eicosanoid research in vitro and in vivo. Their chemical structures may serve as leads for further inhibitor design. Novel drugs depleting cys-LT production could be beneficial for treatment of inflammatory diseases associated with overexpression of LTC4S.
Collapse
Affiliation(s)
- Thea K Kleinschmidt
- Department of Medical Biochemistry and Biophysics (T.K.K., D.B., M.T., A.F., J.Z.H.) and Chemical Biology Consortium Sweden, Science for Life Laboratory Stockholm, Department of Medical Biochemistry and Biophysics (M.H.), and Microvascular Physiology Research Group, Department of Physiology and Pharmacology (E.L., L.L.), Karolinska Institutet, Stockholm Sweden; Clinical Immunology and Allergy Unit, Department of Medicine, Karolinska University Hospital, Stockholm, Sweden (M.E.); and Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine and Pharmacology, Frankfurt/Main, Germany (C.L., A.S.K.)
| | - Martin Haraldsson
- Department of Medical Biochemistry and Biophysics (T.K.K., D.B., M.T., A.F., J.Z.H.) and Chemical Biology Consortium Sweden, Science for Life Laboratory Stockholm, Department of Medical Biochemistry and Biophysics (M.H.), and Microvascular Physiology Research Group, Department of Physiology and Pharmacology (E.L., L.L.), Karolinska Institutet, Stockholm Sweden; Clinical Immunology and Allergy Unit, Department of Medicine, Karolinska University Hospital, Stockholm, Sweden (M.E.); and Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine and Pharmacology, Frankfurt/Main, Germany (C.L., A.S.K.)
| | - Devaraj Basavarajappa
- Department of Medical Biochemistry and Biophysics (T.K.K., D.B., M.T., A.F., J.Z.H.) and Chemical Biology Consortium Sweden, Science for Life Laboratory Stockholm, Department of Medical Biochemistry and Biophysics (M.H.), and Microvascular Physiology Research Group, Department of Physiology and Pharmacology (E.L., L.L.), Karolinska Institutet, Stockholm Sweden; Clinical Immunology and Allergy Unit, Department of Medicine, Karolinska University Hospital, Stockholm, Sweden (M.E.); and Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine and Pharmacology, Frankfurt/Main, Germany (C.L., A.S.K.)
| | - Erik Lundeberg
- Department of Medical Biochemistry and Biophysics (T.K.K., D.B., M.T., A.F., J.Z.H.) and Chemical Biology Consortium Sweden, Science for Life Laboratory Stockholm, Department of Medical Biochemistry and Biophysics (M.H.), and Microvascular Physiology Research Group, Department of Physiology and Pharmacology (E.L., L.L.), Karolinska Institutet, Stockholm Sweden; Clinical Immunology and Allergy Unit, Department of Medicine, Karolinska University Hospital, Stockholm, Sweden (M.E.); and Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine and Pharmacology, Frankfurt/Main, Germany (C.L., A.S.K.)
| | - Madhuranayaki Thulasingam
- Department of Medical Biochemistry and Biophysics (T.K.K., D.B., M.T., A.F., J.Z.H.) and Chemical Biology Consortium Sweden, Science for Life Laboratory Stockholm, Department of Medical Biochemistry and Biophysics (M.H.), and Microvascular Physiology Research Group, Department of Physiology and Pharmacology (E.L., L.L.), Karolinska Institutet, Stockholm Sweden; Clinical Immunology and Allergy Unit, Department of Medicine, Karolinska University Hospital, Stockholm, Sweden (M.E.); and Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine and Pharmacology, Frankfurt/Main, Germany (C.L., A.S.K.)
| | - Maria Ekoff
- Department of Medical Biochemistry and Biophysics (T.K.K., D.B., M.T., A.F., J.Z.H.) and Chemical Biology Consortium Sweden, Science for Life Laboratory Stockholm, Department of Medical Biochemistry and Biophysics (M.H.), and Microvascular Physiology Research Group, Department of Physiology and Pharmacology (E.L., L.L.), Karolinska Institutet, Stockholm Sweden; Clinical Immunology and Allergy Unit, Department of Medicine, Karolinska University Hospital, Stockholm, Sweden (M.E.); and Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine and Pharmacology, Frankfurt/Main, Germany (C.L., A.S.K.)
| | - Alexander Fauland
- Department of Medical Biochemistry and Biophysics (T.K.K., D.B., M.T., A.F., J.Z.H.) and Chemical Biology Consortium Sweden, Science for Life Laboratory Stockholm, Department of Medical Biochemistry and Biophysics (M.H.), and Microvascular Physiology Research Group, Department of Physiology and Pharmacology (E.L., L.L.), Karolinska Institutet, Stockholm Sweden; Clinical Immunology and Allergy Unit, Department of Medicine, Karolinska University Hospital, Stockholm, Sweden (M.E.); and Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine and Pharmacology, Frankfurt/Main, Germany (C.L., A.S.K.)
| | - Christoph Lehmann
- Department of Medical Biochemistry and Biophysics (T.K.K., D.B., M.T., A.F., J.Z.H.) and Chemical Biology Consortium Sweden, Science for Life Laboratory Stockholm, Department of Medical Biochemistry and Biophysics (M.H.), and Microvascular Physiology Research Group, Department of Physiology and Pharmacology (E.L., L.L.), Karolinska Institutet, Stockholm Sweden; Clinical Immunology and Allergy Unit, Department of Medicine, Karolinska University Hospital, Stockholm, Sweden (M.E.); and Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine and Pharmacology, Frankfurt/Main, Germany (C.L., A.S.K.)
| | - Astrid S Kahnt
- Department of Medical Biochemistry and Biophysics (T.K.K., D.B., M.T., A.F., J.Z.H.) and Chemical Biology Consortium Sweden, Science for Life Laboratory Stockholm, Department of Medical Biochemistry and Biophysics (M.H.), and Microvascular Physiology Research Group, Department of Physiology and Pharmacology (E.L., L.L.), Karolinska Institutet, Stockholm Sweden; Clinical Immunology and Allergy Unit, Department of Medicine, Karolinska University Hospital, Stockholm, Sweden (M.E.); and Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine and Pharmacology, Frankfurt/Main, Germany (C.L., A.S.K.)
| | - Lennart Lindbom
- Department of Medical Biochemistry and Biophysics (T.K.K., D.B., M.T., A.F., J.Z.H.) and Chemical Biology Consortium Sweden, Science for Life Laboratory Stockholm, Department of Medical Biochemistry and Biophysics (M.H.), and Microvascular Physiology Research Group, Department of Physiology and Pharmacology (E.L., L.L.), Karolinska Institutet, Stockholm Sweden; Clinical Immunology and Allergy Unit, Department of Medicine, Karolinska University Hospital, Stockholm, Sweden (M.E.); and Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine and Pharmacology, Frankfurt/Main, Germany (C.L., A.S.K.)
| | - Jesper Z Haeggström
- Department of Medical Biochemistry and Biophysics (T.K.K., D.B., M.T., A.F., J.Z.H.) and Chemical Biology Consortium Sweden, Science for Life Laboratory Stockholm, Department of Medical Biochemistry and Biophysics (M.H.), and Microvascular Physiology Research Group, Department of Physiology and Pharmacology (E.L., L.L.), Karolinska Institutet, Stockholm Sweden; Clinical Immunology and Allergy Unit, Department of Medicine, Karolinska University Hospital, Stockholm, Sweden (M.E.); and Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine and Pharmacology, Frankfurt/Main, Germany (C.L., A.S.K.)
| |
Collapse
|
62
|
Skiba J, Karpowicz R, Szabó I, Therrien B, Kowalski K. Synthesis and anticancer activity studies of ferrocenyl-thymine-3,6-dihydro-2H-thiopyranes – A new class of metallocene-nucleobase derivatives. J Organomet Chem 2015. [DOI: 10.1016/j.jorganchem.2015.07.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
63
|
Murphy MB, Xiong Y, Pattabiraman G, Manavalan TT, Qiu F, Medvedev AE. Pellino-3 promotes endotoxin tolerance and acts as a negative regulator of TLR2 and TLR4 signaling. J Leukoc Biol 2015; 98:963-74. [PMID: 26310831 DOI: 10.1189/jlb.2vma0515-229rr] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 07/30/2015] [Indexed: 12/24/2022] Open
Abstract
Development of endotoxin tolerance in macrophages during sepsis reprograms Toll-like receptor 4 signaling to inhibit proinflammatory cytokines without suppressing anti-inflammatory and antimicrobial mediators and protects the host from excessive inflammation and tissue damage. However, endotoxin tolerance renders septic patients immunocompromised and unable to control secondary infections. Although previous studies have revealed the importance of several negative regulators of Toll-like receptor signaling in endotoxin tolerance, the role of Pellino proteins has not been addressed. The present report shows that the induction of endotoxin tolerance in vivo in mice and in vitro in human monocytes and THP-1 and MonoMac-6 macrophages increases the expression of Pellino-3. Overexpression of Pellino-3 in human embryonic kidney 293/Toll-like receptor 2 or 293/Toll-like receptor 4/myeloid differentiation factor-2 cells inhibited Toll-like receptor 2/4-mediated activation of nuclear factor-κB and induction of CXCL-8 mRNA, and Pellino-3 ablation increased these responses. Pellino-3-deficient THP-1 cells had elevated Toll-like receptor 2/4-driven tumor necrosis factor-α, interleukin-6 mRNA, and Toll-like receptor 4-driven CCL5 gene expression in response to Toll-like receptor agonists and heat-killed Escherichia coli and Staphylococcus aureus, cytokines controlled by the MyD88 and Toll-interleukin-1R domain-containing protein inducing interferon-β-mediated pathways, respectively. In addition, deficiency in Pellino-3 slightly increased phagocytosis of heat-killed bacteria. Transfected Pellino-3 inhibited nuclear factor-κB activation driven by overexpression of MyD88, TIR domain-containing adapter inducing interferon-β, interleukin-1R-associated kinase-1, and tumor necrosis factor receptor activator of nuclear factor-κB-binding kinase-1, TGF-β-activated kinase 1, and tumor necrosis factor receptor-associated factor-6, and inhibited interleukin-1R-associated kinase 1 modifications and tumor necrosis factor receptor activator of nuclear factor-κB-binding kinase 1 phosphorylation. Finally, Pellino-3 ablation in THP-1 decreased the extent of endotoxin tolerization. Thus, Pellino-3 is involved in endotoxin tolerance and functions as a negative regulator of Toll-like receptor 2/4 signaling.
Collapse
Affiliation(s)
- Michael B Murphy
- *Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut, USA; and Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yanbao Xiong
- *Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut, USA; and Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Goutham Pattabiraman
- *Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut, USA; and Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Tissa T Manavalan
- *Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut, USA; and Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Fu Qiu
- *Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut, USA; and Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Andrei E Medvedev
- *Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut, USA; and Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
64
|
White MJV, Roife D, Gomer RH. Galectin-3 Binding Protein Secreted by Breast Cancer Cells Inhibits Monocyte-Derived Fibrocyte Differentiation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 195:1858-67. [PMID: 26136428 PMCID: PMC4530092 DOI: 10.4049/jimmunol.1500365] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 06/02/2015] [Indexed: 12/26/2022]
Abstract
To metastasize, tumor cells often need to migrate through a layer of collagen-containing scar tissue which encapsulates the tumor. A key component of scar tissue and fibrosing diseases is the monocyte-derived fibrocyte, a collagen-secreting profibrotic cell. To test the hypothesis that invasive tumor cells may block the formation of the fibrous sheath, we determined whether tumor cells secrete factors that inhibit monocyte-derived fibrocyte differentiation. We found that the human metastatic breast cancer cell line MDA-MB-231 secretes activity that inhibits human monocyte-derived fibrocyte differentiation, whereas less aggressive breast cancer cell lines secrete less of this activity. Purification indicated that Galectin-3 binding protein (LGALS3BP) is the active factor. Recombinant LGALS3BP inhibits monocyte-derived fibrocyte differentiation, and immunodepletion of LGALS3BP from MDA-MB 231 conditioned media removes the monocyte-derived fibrocyte differentiation-inhibiting activity. LGALS3BP inhibits the differentiation of monocyte-derived fibrocytes from wild-type mouse spleen cells, but not from SIGN-R1(-/-) mouse spleen cells, suggesting that CD209/SIGN-R1 is required for the LGALS3BP effect. Galectin-3 and galectin-1, binding partners of LGALS3BP, potentiate monocyte-derived fibrocyte differentiation. In breast cancer biopsies, increased levels of tumor cell-associated LGALS3BP were observed in regions of the tumor that were invading the surrounding stroma. These findings suggest LGALS3BP and galectin-3 as new targets to treat metastatic cancer and fibrosing diseases.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/pharmacology
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Biomarkers, Tumor/pharmacology
- Breast Neoplasms/genetics
- Breast Neoplasms/immunology
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Carrier Proteins/pharmacology
- Cell Adhesion Molecules/metabolism
- Cell Differentiation/drug effects
- Cell Line, Tumor
- Culture Media, Conditioned
- Female
- Galectin 1/metabolism
- Galectin 3/metabolism
- Glycoproteins/genetics
- Glycoproteins/metabolism
- Glycoproteins/pharmacology
- Humans
- Lectins, C-Type/metabolism
- Mice
- Monocytes/cytology
- Monocytes/drug effects
- Monocytes/metabolism
- RNA, Messenger/genetics
- Receptors, Cell Surface/metabolism
- Recombinant Proteins/pharmacology
Collapse
Affiliation(s)
- Michael J V White
- Department of Biology, Texas A&M University, College Station, TX 77843; and
| | - David Roife
- Department of Surgical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030
| | - Richard H Gomer
- Department of Biology, Texas A&M University, College Station, TX 77843; and
| |
Collapse
|
65
|
Murphy M, Xiong Y, Pattabiraman G, Qiu F, Medvedev AE. Pellino-1 Positively Regulates Toll-like Receptor (TLR) 2 and TLR4 Signaling and Is Suppressed upon Induction of Endotoxin Tolerance. J Biol Chem 2015; 290:19218-32. [PMID: 26082489 DOI: 10.1074/jbc.m115.640128] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Indexed: 11/06/2022] Open
Abstract
Endotoxin tolerance reprograms Toll-like receptor (TLR) 4-mediated macrophage responses by attenuating induction of proinflammatory cytokines while retaining expression of anti-inflammatory and antimicrobial mediators. We previously demonstrated deficient TLR4-induced activation of IL-1 receptor-associated kinase (IRAK) 4, IRAK1, and TANK-binding kinase (TBK) 1 as critical hallmarks of endotoxin tolerance, but mechanisms remain unclear. In this study, we examined the role of the E3 ubiquitin ligase Pellino-1 in endotoxin tolerance and TLR signaling. LPS stimulation increased Pellino-1 mRNA and protein expression in macrophages from mice injected with saline and in medium-pretreated human monocytes, THP-1, and MonoMac-6 cells, whereas endotoxin tolerization abrogated LPS inducibility of Pellino-1. Overexpression of Pellino-1 in 293/TLR2 and 293/TLR4/MD2 cells enhanced TLR2- and TLR4-induced nuclear factor κB (NF-κB) and expression of IL-8 mRNA, whereas Pellino-1 knockdown reduced these responses. Pellino-1 ablation in THP-1 cells impaired induction of myeloid differentiation primary response protein (MyD88), and Toll-IL-1R domain-containing adapter inducing IFN-β (TRIF)-dependent cytokine genes in response to TLR4 and TLR2 agonists and heat-killed Escherichia coli and Staphylococcus aureus, whereas only weakly affecting phagocytosis of heat-killed bacteria. Co-expressed Pellino-1 potentiated NF-κB activation driven by transfected MyD88, TRIF, IRAK1, TBK1, TGF-β-activated kinase (TAK) 1, and TNFR-associated factor 6, whereas not affecting p65-induced responses. Mechanistically, Pellino-1 increased LPS-driven K63-linked polyubiquitination of IRAK1, TBK1, TAK1, and phosphorylation of TBK1 and IFN regulatory factor 3. These results reveal a novel mechanism by which endotoxin tolerance re-programs TLR4 signaling via suppression of Pellino-1, a positive regulator of MyD88- and TRIF-dependent signaling that promotes K63-linked polyubiquitination of IRAK1, TBK1, and TAK1.
Collapse
Affiliation(s)
- Michael Murphy
- From the Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut 06030 and
| | - Yanbao Xiong
- the Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Goutham Pattabiraman
- From the Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut 06030 and
| | - Fu Qiu
- the Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Andrei E Medvedev
- From the Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut 06030 and
| |
Collapse
|
66
|
Valenzuela NM, Trinh KR, Mulder A, Morrison SL, Reed EF. Monocyte recruitment by HLA IgG-activated endothelium: the relationship between IgG subclass and FcγRIIa polymorphisms. Am J Transplant 2015; 15:1502-18. [PMID: 25648976 PMCID: PMC4439339 DOI: 10.1111/ajt.13174] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Revised: 12/01/2014] [Accepted: 12/15/2014] [Indexed: 01/25/2023]
Abstract
It is currently unclear which donor specific HLA antibodies confer the highest risk of antibody-mediated rejection (AMR) and allograft loss. In this study, we hypothesized that two distinct features (HLA IgG subclass and Fcγ receptor [FcγR] polymorphisms) which vary from patient to patient, influence the process of monocyte trafficking to and macrophage accumulation in the allograft during AMR in an interrelated fashion. Here, we investigated the contribution of human IgG subclass and FcγR polymorphisms in monocyte recruitment in vitro by primary human aortic endothelium activated with chimeric anti-HLA I human IgG1 and IgG2. Both subclasses triggered monocyte adhesion to endothelial cells, via a two-step process. First, HLA I crosslinking by antibodies stimulated upregulation of P-selectin on endothelium irrespective of IgG subclass. P-selectin-induced monocyte adhesion was enhanced by secondary interactions of IgG with FcγRs, which was highly dependent upon subclass. IgG1 was more potent than IgG2 through differential engagement of FcγRs. Monocytes homozygous for FcγRIIa-H131 adhered more readily to HLA antibody-activated endothelium compared with FcγRIIa-R131 homozygous. Finally, direct modification of HLA I antibodies with immunomodulatory enzymes EndoS and IdeS dampened recruitment by eliminating antibody-FcγR binding, an approach that may have clinical utility in reducing AMR and other forms of antibody-induced inflammation.
Collapse
Affiliation(s)
- Nicole M. Valenzuela
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - K. Ryan Trinh
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA
| | - Arend Mulder
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Sherie L. Morrison
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA
| | - Elaine F. Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| |
Collapse
|
67
|
Knape T, Flesch D, Kuchler L, Sha LK, Giegerich AK, Labocha S, Ferreirós N, Schmid T, Wurglics M, Schubert-Zsilavecz M, Proschak E, Brüne B, Parnham MJ, von Knethen A. Identification and characterisation of a prototype for a new class of competitive PPARγ antagonists. Eur J Pharmacol 2015; 755:16-26. [DOI: 10.1016/j.ejphar.2015.02.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 02/18/2015] [Accepted: 02/19/2015] [Indexed: 01/12/2023]
|
68
|
Haile LA, Puig M, Kelley-Baker L, Verthelyi D. Detection of innate immune response modulating impurities in therapeutic proteins. PLoS One 2015; 10:e0125078. [PMID: 25901912 PMCID: PMC4406594 DOI: 10.1371/journal.pone.0125078] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 03/20/2015] [Indexed: 12/31/2022] Open
Abstract
Therapeutic proteins can contain multiple impurities, some of which are variants of the product, while others are derived from the cell substrate and the manufacturing process. Such impurities, even when present at trace levels, have the potential to activate innate immune cells in peripheral blood or embedded in tissues causing expression of cytokines and chemokines, increasing antigen uptake, facilitating processing and presentation by antigen presenting cells, and fostering product immunogenicity. Currently, while products are tested for host cell protein content, assays to control innate immune response modulating impurities (IIRMIs) in products are focused mainly on endotoxin and nucleic acids, however, depending on the cell substrate and the manufacturing process, numerous other IIRMI could be present. In these studies we assess two approaches that allow for the detection of a broader subset of IIRMIs. In the first, we use commercial cell lines transfected with Toll like receptors (TLR) to detect receptor-specific agonists. This method is sensitive to trace levels of IIRMI and provides information of the type of IIRMIs present but is limited by the availability of stably transfected cell lines and requires pre-existing knowledge of the IIRMIs likely to be present in the product. Alternatively, the use of a combination of macrophage cell lines of human and mouse origin allows for the detection of a broader spectrum of impurities, but does not identify the source of the activation. Importantly, for either system the lower limit of detection (LLOD) of impurities was similar to that of PBMC and it was not modified by the therapeutic protein tested, even in settings where the product had inherent immune modulatory properties. Together these data indicate that a cell-based assay approach could be used to screen products for the presence of IIRMIs and inform immunogenicity risk assessments, particularly in the context of comparability exercises.
Collapse
Affiliation(s)
- Lydia Asrat Haile
- Laboratory of Immunology, Division of Biotechnology Review and Research III, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Montserrat Puig
- Laboratory of Immunology, Division of Biotechnology Review and Research III, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Logan Kelley-Baker
- Laboratory of Immunology, Division of Biotechnology Review and Research III, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Daniela Verthelyi
- Laboratory of Immunology, Division of Biotechnology Review and Research III, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
- * E-mail:
| |
Collapse
|
69
|
Kawata K, Iwai A, Muramatsu D, Aoki S, Uchiyama H, Okabe M, Hayakawa S, Takaoka A, Miyazaki T. Stimulation of macrophages with the β-glucan produced by aureobasidium pullulans promotes the secretion of tumor necrosis factor-related apoptosis inducing ligand (TRAIL). PLoS One 2015; 10:e0124809. [PMID: 25875639 PMCID: PMC4395393 DOI: 10.1371/journal.pone.0124809] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 03/19/2015] [Indexed: 01/29/2023] Open
Abstract
A β-glucan produced by Aureobasidium pullulans (AP-PG) is consisting of a β-(1,3)-linked main chain with β-(1,6)-linked glucose side residues. Various β-glucans consisting of β-(1,3)-linked main chain including AP-PG are believed to exhibit anti-tumor activities, and actually, anti-tumor activities of AP-PG in mice have been demonstrated. In this study, we demonstrate that stimulation with AP-PG induces TRAIL expression in mouse and human macrophage-like cell lines. TRAIL is known to be a cytokine which specifically induces apoptosis in transformed cells, but not in untransformed cells. The expression of TRAIL mRNA after stimulation with AP-PG was increased in RAW264.7 cells, Mono Mac 6 cells, and macrophage-differentiated THP-1 cells. The mRNA expression of TNF-α and FasL is only weakly increased after stimulation with AP-PG. The induction activity of TRAIL by curdlan, a bacterial β-glucan, was very similar to that by AP-PG in RAW264.7 cells, but weaker in macrophage-differentiated THP-1 cells. Activation of caspases was found in HeLa cells after treatment with the supernatant of cultured medium from AP-PG-stimulated Mono Mac 6 cells, and was inhibited by the anti-TRAIL neutralizing antibody. These findings suggest that the stimulation with AP-PG effectively induces TRAIL in macrophages, and that it may be related to apoptosis induction of tumor cells.
Collapse
Affiliation(s)
- Koji Kawata
- Aureo Science Co., Ltd., Sapporo, Hokkaido, Japan
- Aureo Co., Ltd., Kimitsu, Chiba, Japan
| | - Atsushi Iwai
- Aureo Science Co., Ltd., Sapporo, Hokkaido, Japan
- Aureo Co., Ltd., Kimitsu, Chiba, Japan
- * E-mail:
| | | | - Shiho Aoki
- Aureo Science Co., Ltd., Sapporo, Hokkaido, Japan
| | | | | | - Sumio Hayakawa
- Division of Signaling in Cancer and Immunology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Akinori Takaoka
- Division of Signaling in Cancer and Immunology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Tadaaki Miyazaki
- Department of Probiotics Immunology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
70
|
Le QT, Lotfi-Emran S, Min HK, Schwartz LB. A simple, sensitive and safe method to determine the human α/β-tryptase genotype. PLoS One 2014; 9:e114944. [PMID: 25545679 PMCID: PMC4278853 DOI: 10.1371/journal.pone.0114944] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 11/16/2014] [Indexed: 11/18/2022] Open
Abstract
The human tryptase locus on chromosome 16 contains one gene encoding only β-tryptase and another encoding either β-tryptase or the homologous α-tryptase, providing α:β gene ratios of 0:4, 1:3 or 2:2 in the diploid genome, these genotypes being of potential clinical relevance in severe atopy. Using an EcoRV restriction site in α- but not β-tryptase, PCR products, spanning intron 1 to exon 5, were used to determine α/β-tryptase gene ratios using non-radioactive labels, including ethidium bromide labeling of all PCR products, and either digoxigenin-primer or DY682-primer labeling of only the final PCR cycle products. Sensitivity increased ∼60-fold with each final PCR cycle labeling technique. Ethidium bromide labeling underestimated amounts of α-tryptase, presumably because heteroduplexes of α/β-tryptase amplimers, formed during annealing, were EcoRV resistant. In contrast, both final PCR cycle labeling techniques precisely quantified these gene ratios, because only homoduplexes were labeled. Using the DY682-primer was most efficient, because PCR/EcoRV products could be analyzed directly in the gel; while digoxigenin-labeled products required transfer to a nitrocellulose membrane followed by immunoblotting. This technique for determining the α/β-tryptase genotype is sensitive, accurate, simple and safe, and should permit high-throughput screening to detect potential phenotype-genotype relations for α/β-tryptases, and for other closely related alleles.
Collapse
Affiliation(s)
- Quang Trong Le
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Sahar Lotfi-Emran
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Hae-Ki Min
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Lawrence B. Schwartz
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
- * E-mail:
| |
Collapse
|
71
|
Horváti K, Bacsa B, Kiss É, Gyulai G, Fodor K, Balka G, Rusvai M, Szabó E, Hudecz F, Bősze S. Nanoparticle Encapsulated Lipopeptide Conjugate of Antitubercular Drug Isoniazid: In Vitro Intracellular Activity and in Vivo Efficacy in a Guinea Pig Model of Tuberculosis. Bioconjug Chem 2014; 25:2260-8. [DOI: 10.1021/bc500476x] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
| | | | | | | | | | | | | | - Eleonóra Szabó
- Laboratory
of Bacteriology, Korányi National Institute for Tuberculosis and Respiratory Medicine, Budapest, 1122 Hungary
| | | | | |
Collapse
|
72
|
Chanput W, Mes JJ, Wichers HJ. THP-1 cell line: An in vitro cell model for immune modulation approach. Int Immunopharmacol 2014; 23:37-45. [DOI: 10.1016/j.intimp.2014.08.002] [Citation(s) in RCA: 891] [Impact Index Per Article: 81.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 08/02/2014] [Accepted: 08/04/2014] [Indexed: 01/06/2023]
|
73
|
Bordetella pertussis naturally occurring isolates with altered lipooligosaccharide structure fail to fully mature human dendritic cells. Infect Immun 2014; 83:227-38. [PMID: 25348634 DOI: 10.1128/iai.02197-14] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Bordetella pertussis is a Gram-negative bacterium and the causative agent of whooping cough. Despite high vaccination coverage, outbreaks are being increasingly reported worldwide. Possible explanations include adaptation of this pathogen, which may interfere with recognition by the innate immune system. Here, we describe innate immune recognition and responses to different B. pertussis clinical isolates. By using HEK-Blue cells transfected with different pattern recognition receptors, we found that 3 out of 19 clinical isolates failed to activate Toll-like receptor 4 (TLR4). These findings were confirmed by using the monocytic MM6 cell line. Although incubation with high concentrations of these 3 strains resulted in significant activation of the MM6 cells, it was found to occur mainly through interaction with TLR2 and not through TLR4. When using live bacteria, these 3 strains also failed to activate TLR4 on HEK-Blue cells, and activation of MM6 cells or human monocyte-derived dendritic cells was significantly lower than activation induced by the other 16 strains. Mass spectrum analysis of the lipid A moieties from these 3 strains indicated an altered structure of this molecule. Gene sequence analysis revealed mutations in genes involved in lipid A synthesis. Findings from this study indicate that B. pertussis isolates that do not activate TLR4 occur naturally and that this phenotype may give this bacterium an advantage in tempering the innate immune response and establishing infection. Knowledge on the strategies used by this pathogen in evading the host immune response is essential for the improvement of current vaccines or for the development of new ones.
Collapse
|
74
|
Friedrichs B, Neumann U, Schüller J, Peck MJ. Cigarette-smoke-induced priming of neutrophils from smokers and non-smokers for increased oxidative burst response is mediated by TNF-α. Toxicol In Vitro 2014; 28:1249-58. [PMID: 24997298 DOI: 10.1016/j.tiv.2014.06.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 06/10/2014] [Accepted: 06/23/2014] [Indexed: 10/25/2022]
Abstract
In vitro treatment of human peripheral blood neutrophils from smokers and non-smokers with an aqueous cigarette smoke (CS) extract resulted in a concentration-dependent increase in surface expression of CD11b and CD66b and a corresponding decrease of CD62L, together with a concentration-dependent release of MMP-8, MMP-9, and lactoferrin, indicating considerable activation and degranulation. However, the burst response to N-formyl-methionyl-leucyl-phenylalanine (fMLP) was unchanged in CS-stimulated neutrophils from both smokers and non-smokers. When supernatants from CS-treated monocytic MonoMac-6 (MM6) cells were used for activation of neutrophils, concentration-dependent changes in surface marker expression, granule protein release, and the oxidative burst response to fMLP were observed, again with no major differences between smokers and non-smokers. CS-treated MM6 cells released significant amounts of IL-8 and TNF-α into the culture supernatant. However, antibody blocking experiments showed that only TNF-α mediated the increased burst response in neutrophils. These data show that, in the presence of secondary cells, CS is able to prime neutrophils for an increased burst response to fMLP which is mediated by TNF-α, released from the secondary cells in response to CS. Following stimulation with priming agents, peripheral blood neutrophils from healthy smokers show an equal burst response compared to those from non-smokers.
Collapse
Affiliation(s)
- Bärbel Friedrichs
- Philip Morris International R&D, Philip Morris Research Laboratories GmbH, D-51149 Cologne, Germany
| | - Ute Neumann
- Philip Morris International R&D, Philip Morris Research Laboratories GmbH, D-51149 Cologne, Germany
| | - Jutta Schüller
- Philip Morris International R&D, Philip Morris Research Laboratories GmbH, D-51149 Cologne, Germany
| | - Michael J Peck
- Philip Morris International R&D, Philip Morris Products S.A., CH-2000 Neuchâtel, Switzerland.
| |
Collapse
|
75
|
Kiss É, Gyulai G, Pénzes CB, Idei M, Horváti K, Bacsa B, Bősze S. Tuneable surface modification of PLGA nanoparticles carrying new antitubercular drug candidate. Colloids Surf A Physicochem Eng Asp 2014. [DOI: 10.1016/j.colsurfa.2014.05.048] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
76
|
Ilinskaya AN, Man S, Patri AK, Clogston JD, Crist RM, Cachau RE, McNeil SE, Dobrovolskaia MA. Inhibition of phosphoinositol 3 kinase contributes to nanoparticle-mediated exaggeration of endotoxin-induced leukocyte procoagulant activity. Nanomedicine (Lond) 2014; 9:1311-26. [PMID: 24279459 PMCID: PMC4035470 DOI: 10.2217/nnm.13.137] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Indexed: 01/17/2023] Open
Abstract
AIM Disseminated intravascular coagulation is an increasing concern for certain types of engineered nanomaterials. Recent studies have shed some light on the nanoparticle physicochemical properties contributing to this toxicity; however, the mechanisms are poorly understood. Leukocyte procoagulant activity (PCA) is a key factor contributing to the initiation of this toxicity. We have previously reported on the exaggeration of endotoxin-induced PCA by cationic dendrimers. Herein, we report an effort to discern the mechanism. MATERIALS & METHODS Poly(amidoamine) dendrimers with various sizes and surface functionalities were studied in vitro by the recalcification test, flow cytometry and other relevant assays. RESULTS & CONCLUSION Cationic dendrimers exaggerated endotoxin-induced PCA, but their anionic or neutral counterparts did not; the cationic charge prompts this phenomenon, but different cationic surface chemistries do not influence it. Cationic dendrimers and endotoxin differentially affect the PCA complex. The inhibition of phosphoinositol 3 kinase by dendrimers contributes to the exaggeration of the endotoxin-induced PCA.
Collapse
Affiliation(s)
- Anna N Ilinskaya
- Nanotechnology Characterization Laboratory, Advanced Technology Program, SAIC-Frederick Inc., NCI-Frederick, 1050 Boyles Street, Building 469, Frederick, MD 21702, USA
| | - Sonny Man
- Nanotechnology Characterization Laboratory, Advanced Technology Program, SAIC-Frederick Inc., NCI-Frederick, 1050 Boyles Street, Building 469, Frederick, MD 21702, USA
| | - Anil K Patri
- Nanotechnology Characterization Laboratory, Advanced Technology Program, SAIC-Frederick Inc., NCI-Frederick, 1050 Boyles Street, Building 469, Frederick, MD 21702, USA
| | - Jeffrey D Clogston
- Nanotechnology Characterization Laboratory, Advanced Technology Program, SAIC-Frederick Inc., NCI-Frederick, 1050 Boyles Street, Building 469, Frederick, MD 21702, USA
| | - Rachael M Crist
- Nanotechnology Characterization Laboratory, Advanced Technology Program, SAIC-Frederick Inc., NCI-Frederick, 1050 Boyles Street, Building 469, Frederick, MD 21702, USA
| | - Raul E Cachau
- Advanced Biomedical Computing Center, SAIC-Frederick Inc., NCI-Frederick, Frederick, MD 21702, USA
| | - Scott E McNeil
- Nanotechnology Characterization Laboratory, Advanced Technology Program, SAIC-Frederick Inc., NCI-Frederick, 1050 Boyles Street, Building 469, Frederick, MD 21702, USA
| | - Marina A Dobrovolskaia
- Nanotechnology Characterization Laboratory, Advanced Technology Program, SAIC-Frederick Inc., NCI-Frederick, 1050 Boyles Street, Building 469, Frederick, MD 21702, USA
| |
Collapse
|
77
|
Pupo E, Hamstra HJ, Meiring H, van der Ley P. Lipopolysaccharide engineering in Neisseria meningitidis: structural analysis of different pentaacyl lipid A mutants and comparison of their modified agonist properties. J Biol Chem 2014; 289:8668-80. [PMID: 24492609 PMCID: PMC3961689 DOI: 10.1074/jbc.m114.554345] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Indexed: 12/20/2022] Open
Abstract
Engineering the lipopolysaccharide (LPS) biosynthetic pathway offers the potential to obtain modified derivatives with optimized adjuvant properties. Neisseria meningitidis strain H44/76 was modified by expression of the pagL gene encoding lipid A 3-O-deacylase from Bordetella bronchiseptica and by inactivation of the lgtB gene encoding the terminal oligosaccharide galactosyltransferase. Mass spectrometry analysis of purified mutant LPS was used for detailed compositional analysis of all present molecular species. This determined that the modified LPS was mainly pentaacylated, demonstrating high efficiency of conversion from the hexaacyl to the 3-O-deacylated form by heterologous lipid A 3-O-deacylase (PagL) expression. MS analyses also provided evidence for expression of only one major oligosaccharide glycoform, which lacked the terminal galactose residue as expected from inactivation of the lgtB gene. The immunomodulatory properties of PagL-deacylated LPS were compared with another pentaacyl form obtained from an lpxL1(-) mutant, which lacks the 2' secondary acyl chain. Although both LPS mutants displayed impaired capacity to induce production of the pro-inflammatory cytokine IL-6 in the monocytic cell line Mono Mac 6, induction of the Toll-interleukin-1 receptor domain-containing adaptor-inducing interferon-β-dependent chemokine interferon-γ-induced protein 10 was largely retained only for the lgtB(-)/pagL(+) mutant. Removal of remaining hexaacyl species exclusively present in lgtB(-)/pagL(+) LPS demonstrated that these minor species potentiate but do not determine the activity of this LPS. These results are the first to indicate a qualitatively different response of human innate cells to pentaacyl lpxL1(-) and pagL(+) LPS and show the importance of detailed structure-function analysis when working with modified lipid A structures. The pagL(+) LPS has significant potential as immune modulator in humans.
Collapse
Affiliation(s)
- Elder Pupo
- From the Institute for Translational Vaccinology and
| | - Hendrik-Jan Hamstra
- the National Institute for Public Health and the Environment, 3721 MA Bilthoven, The Netherlands
| | - Hugo Meiring
- From the Institute for Translational Vaccinology and
| | | |
Collapse
|
78
|
Wen B, Combes V, Bonhoure A, Weksler BB, Couraud PO, Grau GER. Endotoxin-induced monocytic microparticles have contrasting effects on endothelial inflammatory responses. PLoS One 2014; 9:e91597. [PMID: 24646764 PMCID: PMC3960107 DOI: 10.1371/journal.pone.0091597] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 02/11/2014] [Indexed: 12/31/2022] Open
Abstract
Septic shock is a severe disease state characterised by the body's life threatening response to infection. Complex interactions between endothelial cells and circulating monocytes are responsible for microvasculature dysfunction contributing to the pathogenesis of this syndrome. Here, we intended to determine whether microparticles derived from activated monocytes contribute towards inflammatory processes and notably vascular permeability. We found that endotoxin stimulation of human monocytes enhances the release of microparticles of varying phenotypes and mRNA contents. Elevated numbers of LPS-induced monocytic microparticles (mMP) expressed CD54 and contained higher levels of transcripts for pro-inflammatory cytokines such as TNF, IL-6 and IL-8. Using a prothrombin time assay, a greater reduction in plasma coagulation time was observed with LPS-induced mMP than with non-stimulated mMP. Co-incubation of mMP with the human brain endothelial cell line hCMEC/D3 triggered their time-dependent uptake and significantly enhanced endothelial microparticle release. Unexpectedly, mMP also modified signalling pathways by diminishing pSrc (tyr416) expression and promoted endothelial monolayer tightness, as demonstrated by endothelial impedance and permeability assays. Altogether, these data strongly suggest that LPS-induced mMP have contrasting effects on the intercellular communication network and display a dual potential: enhanced pro-inflammatory and procoagulant properties, together with protective function of the endothelium.
Collapse
Affiliation(s)
- Beryl Wen
- Vascular Immunology Unit, Sydney Medical School & Bosch Institute, University of Sydney, Camperdown, Australia
| | - Valery Combes
- Vascular Immunology Unit, Sydney Medical School & Bosch Institute, University of Sydney, Camperdown, Australia
| | - Amandine Bonhoure
- Vascular Immunology Unit, Sydney Medical School & Bosch Institute, University of Sydney, Camperdown, Australia
| | - Babette B Weksler
- Weill Medical College, Cornell University, New York, New York, United States of America
| | - Pierre-Olivier Couraud
- Institut Cochin, INSERM U1016, Paris, France; CNRS, UMR 8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Georges E R Grau
- Vascular Immunology Unit, Sydney Medical School & Bosch Institute, University of Sydney, Camperdown, Australia
| |
Collapse
|
79
|
Proteomic analysis of detergent resistant membrane domains during early interaction of macrophages with rough and smooth Brucella melitensis. PLoS One 2014; 9:e91706. [PMID: 24643124 PMCID: PMC3958395 DOI: 10.1371/journal.pone.0091706] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 02/13/2014] [Indexed: 12/20/2022] Open
Abstract
The plasma membrane contains discrete nanometer-sized domains that are resistant to non-ionic detergents, and which are called detergent resistant membrane domains (DRMDs) or lipid rafts. Exposure of host cells to pathogenic bacteria has been shown to induce the re-distribution of specific host proteins between DRMDs and detergent soluble membranes, which leads to the initiation of cell signaling that enable pathogens to access host cells. DRMDs have been shown to play a role in the invasion of Brucella into host macrophages and the formation of replicative phagosomes called Brucella-containing vacuoles (BCVs). In this study we sought to characterize changes to the protein expression profiles in DRMDs and to respective cellular pathways and networks of Mono Mac 6 cells in response to the adherence of rough VTRM1 and smooth 16 M B. melitensis strains. DRMDs were extracted from Mono Mac 6 cells exposed for 2 minutes at 4°C to Brucella (no infection occurs) and from unexposed control cells. Protein expression was determined using the non-gel based quantitative iTRAQ (Isobaric Tags for Relative and Absolute Quantitation) mass spectrometry technique. Using the identified iTRAQ proteins we performed enrichment analyses and probed constructed human biochemical networks for interactions and metabolic reactions. We identified 149 proteins, which either became enriched, depleted or whose amounts did not change in DRMDs upon Brucella exposure. Several of these proteins were distinctly enriched or depleted in DRMDs upon exposure to rough and smooth B. melitensis strains which results in the differential engagement of cellular pathways and networks immediately upon Brucella encounter. For some of the proteins such as myosin 9, small G protein signaling modulator 3, lysine-specific demethylase 5D, erlin-2, and voltage-dependent anion-selective channel protein 2, we observed extreme differential depletion or enrichment in DRMDs. The identified proteins and pathways could provide the basis for novel ways of treating or diagnosing Brucellosis.
Collapse
|
80
|
Antimicrobial treatment improves mycobacterial survival in nonpermissive growth conditions. Antimicrob Agents Chemother 2014; 58:2798-806. [PMID: 24590482 PMCID: PMC3993263 DOI: 10.1128/aac.02774-13] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Antimicrobials targeting cell wall biosynthesis are generally considered inactive against nonreplicating bacteria. Paradoxically, we found that under nonpermissive growth conditions, exposure of Mycobacterium bovis BCG bacilli to such antimicrobials enhanced their survival. We identified a transcriptional regulator, RaaS (for regulator of antimicrobial-assisted survival), encoded by bcg1279 (rv1219c) as being responsible for the observed phenomenon. Induction of this transcriptional regulator resulted in reduced expression of specific ATP-dependent efflux pumps and promoted long-term survival of mycobacteria, while its deletion accelerated bacterial death under nonpermissive growth conditions in vitro and during macrophage or mouse infection. These findings have implications for the design of antimicrobial drug combination therapies for persistent infectious diseases, such as tuberculosis.
Collapse
|
81
|
Prostaglandin E 2 Does Not Modulate CCR7 Expression and Functionality after Differentiation of Blood Monocytes into Macrophages. Int J Inflam 2013; 2013:918016. [PMID: 24298392 PMCID: PMC3835491 DOI: 10.1155/2013/918016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 09/11/2013] [Indexed: 11/17/2022] Open
Abstract
Previously, we demonstrated that prostaglandin E2 (PGE2) induces C-C chemokine receptor type 7 (CCR7) expression on human monocytes, which stimulates their subsequent migration in response to the CCR7 natural ligands CCL19 and CCL21. In this study, we determined whether PGE2 affects CCR7 expression on macrophages. Flow cytometric analysis and chemotaxis assays were performed on Mono Mac-1-derived macrophage (MDMM-1) as well as unpolarized monocyte-derived macrophages (MDMs) to determine the CCR7 expression and functionality in the presence of PGE2. Data revealed that a MDMM-1 exhibited markedly downregulated CCR7 expression and functionality that were partially restored by treatment with PGE2. In MDMs, we observed a drastic downregulation of CCR7 expression and functionality that were unaffected following PGE2 treatment. Our data indicate that monocyte differentiation induces the loss of CCR7 expression and that PGE2 is unable to modulate CCR7 expression and functionality as shown previously in monocytes.
Collapse
|
82
|
Urokinase-type plasminogen activator (uPA) modulates monocyte-to-macrophage differentiation and prevents Ox-LDL-induced macrophage apoptosis. Atherosclerosis 2013; 231:29-38. [DOI: 10.1016/j.atherosclerosis.2013.08.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 08/01/2013] [Accepted: 08/19/2013] [Indexed: 01/09/2023]
|
83
|
Lee J, Espéli M, Anderson C, Linterman M, Pocock J, Williams N, Roberts R, Viatte S, Fu B, Peshu N, Hien T, Phu N, Wesley E, Edwards C, Ahmad T, Mansfield J, Gearry R, Dunstan S, Williams T, Barton A, Vinuesa C, Parkes M, Lyons PA, Smith KG. Human SNP links differential outcomes in inflammatory and infectious disease to a FOXO3-regulated pathway. Cell 2013; 155:57-69. [PMID: 24035192 PMCID: PMC3790457 DOI: 10.1016/j.cell.2013.08.034] [Citation(s) in RCA: 189] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 07/08/2013] [Accepted: 08/19/2013] [Indexed: 12/19/2022]
Abstract
The clinical course and eventual outcome, or prognosis, of complex diseases varies enormously between affected individuals. This variability critically determines the impact a disease has on a patient's life but is very poorly understood. Here, we exploit existing genome-wide association study data to gain insight into the role of genetics in prognosis. We identify a noncoding polymorphism in FOXO3A (rs12212067: T > G) at which the minor (G) allele, despite not being associated with disease susceptibility, is associated with a milder course of Crohn's disease and rheumatoid arthritis and with increased risk of severe malaria. Minor allele carriage is shown to limit inflammatory responses in monocytes via a FOXO3-driven pathway, which through TGFβ1 reduces production of proinflammatory cytokines, including TNFα, and increases production of anti-inflammatory cytokines, including IL-10. Thus, we uncover a shared genetic contribution to prognosis in distinct diseases that operates via a FOXO3-driven pathway modulating inflammatory responses.
Collapse
Affiliation(s)
- James C. Lee
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
| | - Marion Espéli
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
| | - Carl A. Anderson
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Michelle A. Linterman
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
| | - Joanna M. Pocock
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
| | - Naomi J. Williams
- Department of Pathogens and Immunity, John Curtin School of Medical Research, Australian National University, Canberra ACT 2601, Australia
| | - Rebecca Roberts
- University of Otago, Department of Medicine, Christchurch 8011, New Zealand
| | - Sebastien Viatte
- Arthritis Research UK Epidemiology Unit, Manchester Academic Health Science Center, University of Manchester, Manchester M13 9PT, UK
| | - Bo Fu
- Arthritis Research UK Epidemiology Unit, Manchester Academic Health Science Center, University of Manchester, Manchester M13 9PT, UK
- Centre for Biostatistics, Institute of Population Health, University of Manchester, Manchester M13 9PL, UK
| | - Norbert Peshu
- Kenya Medical Research Institute/Wellcome Trust Research Program, Centre for Geographic Medicine Research, Kilifi P.O. Box 230-80108, Kenya
| | - Tran Tinh Hien
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, Hospital for Tropical Diseases, District 5 Ho Chi Minh City, Vietnam
| | - Nguyen Hoan Phu
- The Hospital for Tropical Diseases, District 5 Ho Chi Minh City, Vietnam
| | - Emma Wesley
- Peninsula College of Medicine and Dentistry, University of Exeter, Exeter EX2 5DW, UK
| | - Cathryn Edwards
- Department of Gastroenterology, Torbay Hospital, Torquay TQ2 7AA, UK
| | - Tariq Ahmad
- Peninsula College of Medicine and Dentistry, University of Exeter, Exeter EX2 5DW, UK
| | - John C. Mansfield
- Institute of Genetic Medicine, Newcastle University, Newcastle NE1 3BZ, UK
| | - Richard Gearry
- University of Otago, Department of Medicine, Christchurch 8011, New Zealand
| | - Sarah Dunstan
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, Hospital for Tropical Diseases, District 5 Ho Chi Minh City, Vietnam
- Centre for Tropical Medicine, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7LJ, UK
| | - Thomas N. Williams
- Kenya Medical Research Institute/Wellcome Trust Research Program, Centre for Geographic Medicine Research, Kilifi P.O. Box 230-80108, Kenya
- Department of Medicine, Imperial College, London SW7 2AZ, UK
| | - Anne Barton
- Arthritis Research UK Epidemiology Unit, Manchester Academic Health Science Center, University of Manchester, Manchester M13 9PT, UK
| | - Carola G. Vinuesa
- Department of Pathogens and Immunity, John Curtin School of Medical Research, Australian National University, Canberra ACT 2601, Australia
| | - Miles Parkes
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
| | - Paul A. Lyons
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
| | - Kenneth G.C. Smith
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
| |
Collapse
|
84
|
Misquith A, Fung HWM, Dowling QM, Guderian JA, Vedvick TS, Fox CB. In vitro evaluation of TLR4 agonist activity: formulation effects. Colloids Surf B Biointerfaces 2013; 113:312-9. [PMID: 24121074 DOI: 10.1016/j.colsurfb.2013.09.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 09/03/2013] [Accepted: 09/04/2013] [Indexed: 12/17/2022]
Abstract
Effective in vitro evaluation of vaccine adjuvants would allow higher throughput screening compared to in vivo studies. However, vaccine adjuvants comprise a wide range of structures and formulations ranging from soluble TLR agonists to complex lipid-based formulations. The effects of formulation parameters on in vitro bioactivity assays and the correlations with in vivo adjuvant activity is not well understood. In the present work, we employ the Limulus amebocyte lysate assay and a human macrophage cellular cytokine production assay to demonstrate the differences in in vitro bioactivity of four distinct formulations of the synthetic TLR4 agonist GLA: an aqueous nanosuspension (GLA-AF), an oil-in-water emulsion (GLA-SE), a liposome (GLA-LS), and an alum-adsorbed formulation (GLA-Alum). Furthermore, we demonstrate the importance of the localization of GLA on in vitro potency. By comparing to previous published reports on the in vivo bioactivity of these GLA-containing formulations, we conclude that the most potent activators of the in vitro systems may not be the most potent in vivo adjuvant formulations. Furthermore, we discuss the formulation considerations which should be taken into account when interpreting data from in vitro adjuvant activity assays.
Collapse
Affiliation(s)
- Ayesha Misquith
- Infectious Disease Research Institute (IDRI), 1124 Columbia Street, Ste 400, Seattle, WA 98104, USA
| | - H W Millie Fung
- Infectious Disease Research Institute (IDRI), 1124 Columbia Street, Ste 400, Seattle, WA 98104, USA
| | - Quinton M Dowling
- Infectious Disease Research Institute (IDRI), 1124 Columbia Street, Ste 400, Seattle, WA 98104, USA
| | - Jeffrey A Guderian
- Infectious Disease Research Institute (IDRI), 1124 Columbia Street, Ste 400, Seattle, WA 98104, USA
| | - Thomas S Vedvick
- Infectious Disease Research Institute (IDRI), 1124 Columbia Street, Ste 400, Seattle, WA 98104, USA
| | - Christopher B Fox
- Infectious Disease Research Institute (IDRI), 1124 Columbia Street, Ste 400, Seattle, WA 98104, USA.
| |
Collapse
|
85
|
de Groote ML, Kazemier HG, Huisman C, van der Gun BT, Faas MM, Rots MG. Upregulation of endogenous ICAM-1 reduces ovarian cancer cell growth in the absence of immune cells. Int J Cancer 2013; 134:280-90. [DOI: 10.1002/ijc.28375] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 05/25/2013] [Accepted: 06/12/2013] [Indexed: 11/08/2022]
Affiliation(s)
- Marloes L. de Groote
- Epigenetic Editing; Department of Pathology and Medical Biology; University Medical Center Groningen, University of Groningen; Groningen The Netherlands
| | - Hinke G. Kazemier
- Epigenetic Editing; Department of Pathology and Medical Biology; University Medical Center Groningen, University of Groningen; Groningen The Netherlands
| | - Christian Huisman
- Epigenetic Editing; Department of Pathology and Medical Biology; University Medical Center Groningen, University of Groningen; Groningen The Netherlands
| | - Bernardina T.F. van der Gun
- Epigenetic Editing; Department of Pathology and Medical Biology; University Medical Center Groningen, University of Groningen; Groningen The Netherlands
| | - Marijke M. Faas
- Immunoendocrinology; Department of Pathology and Medical Biology; University Medical Center Groningen, University of Groningen; Groningen The Netherlands
| | - Marianne G. Rots
- Epigenetic Editing; Department of Pathology and Medical Biology; University Medical Center Groningen, University of Groningen; Groningen The Netherlands
| |
Collapse
|
86
|
Miller PG, Al-Shahrour F, Hartwell KA, Chu LP, Järås M, Puram RV, Puissant A, Callahan KP, Ashton J, McConkey ME, Poveromo LP, Cowley GS, Kharas MG, Labelle M, Shterental S, Fujisaki J, Silberstein L, Alexe G, Al-Hajj MA, Shelton CA, Armstrong SA, Root DE, Scadden DT, Hynes RO, Mukherjee S, Stegmaier K, Jordan CT, Ebert BL. In Vivo RNAi screening identifies a leukemia-specific dependence on integrin beta 3 signaling. Cancer Cell 2013; 24:45-58. [PMID: 23770013 PMCID: PMC3746037 DOI: 10.1016/j.ccr.2013.05.004] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 02/19/2013] [Accepted: 05/02/2013] [Indexed: 01/05/2023]
Abstract
We used an in vivo small hairpin RNA (shRNA) screening approach to identify genes that are essential for MLL-AF9 acute myeloid leukemia (AML). We found that Integrin Beta 3 (Itgb3) is essential for murine leukemia cells in vivo and for human leukemia cells in xenotransplantation studies. In leukemia cells, Itgb3 knockdown impaired homing, downregulated LSC transcriptional programs, and induced differentiation via the intracellular kinase Syk. In contrast, loss of Itgb3 in normal hematopoietic stem and progenitor cells did not affect engraftment, reconstitution, or differentiation. Finally, using an Itgb3 knockout mouse model, we confirmed that Itgb3 is dispensable for normal hematopoiesis but is required for leukemogenesis. Our results establish the significance of the Itgb3 signaling pathway as a potential therapeutic target in AML.
Collapse
Affiliation(s)
- Peter G Miller
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
van de Waterbeemd B, Zomer G, Kaaijk P, Ruiterkamp N, Wijffels RH, van den Dobbelsteen GPJM, van der Pol LA. Improved production process for native outer membrane vesicle vaccine against Neisseria meningitidis. PLoS One 2013; 8:e65157. [PMID: 23741478 PMCID: PMC3669287 DOI: 10.1371/journal.pone.0065157] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 04/23/2013] [Indexed: 11/18/2022] Open
Abstract
An improved detergent-free process has been developed to produce vaccine based on native outer membrane vesicles (NOMV) against Neisseria meningitidis serogroup B. Performance was evaluated with the NonaMen vaccine concept, which provides broad coverage based on nine distinct PorA antigens. Scalable aseptic equipment was implemented, replacing undesirable steps like ultracentrifugation, inactivation with phenol, and the use of preservatives. The resulting process is more consistent and gives a higher yield than published reference processes, enabling NOMV production at commercial scale. Product quality met preliminary specifications for 9 consecutive batches, and an ongoing study confirmed real-time stability up to 12 months after production. As the NOMV had low endotoxic activity and induced high bactericidal titres in mice, they are expected to be safe and effective in humans. The production process is not limited to NonaMen and may be applicable for other N. meningitidis serogroups and other gram-negative pathogens. The current results therefore facilitate the late-stage development and clinical evaluation of NOMV vaccines.
Collapse
Affiliation(s)
- Bas van de Waterbeemd
- National Institute for Public Health and the Environment (RIVM), Vaccinology, Bilthoven, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
88
|
Ochs MJ, Ossipova E, Oliynyk G, Steinhilber D, Suess B, Jakobsson PJ. Mass Spectrometry-Based Proteomics Identifies UPF1 as a Critical Gene Expression Regulator in MonoMac 6 Cells. J Proteome Res 2013; 12:2622-9. [DOI: 10.1021/pr301193f] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Meike J. Ochs
- Institute of Pharmaceutical Chemistry/ZAFES, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438
Frankfurt/M., Germany
- Institute
of Molecular Biosciences, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt/M., Germany
| | - Elena Ossipova
- Department of Medicine, Rheumatology Unit, Karolinska Institute, Stockholm, Sweden
| | - Ganna Oliynyk
- Department of Medicine, Rheumatology Unit, Karolinska Institute, Stockholm, Sweden
- Department of Microbiology, Tumor
and Cell Biology, Karolinska Institute,
Stockholm, Sweden
| | - Dieter Steinhilber
- Institute of Pharmaceutical Chemistry/ZAFES, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438
Frankfurt/M., Germany
| | - Beatrix Suess
- Institute
of Molecular Biosciences, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt/M., Germany
- Department of
Biology, Technical University Darmstadt, Schnittspahnstraße 1064287 Darmstadt, Germany
| | - Per-Johan Jakobsson
- Department of Medicine, Rheumatology Unit, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
89
|
Wöbke TK, von Knethen A, Steinhilber D, Sorg BL. CD69 is a TGF-β/1α,25-dihydroxyvitamin D3 target gene in monocytes. PLoS One 2013; 8:e64635. [PMID: 23696902 PMCID: PMC3655964 DOI: 10.1371/journal.pone.0064635] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 04/16/2013] [Indexed: 01/09/2023] Open
Abstract
CD69 is a transmembrane lectin that can be expressed on most hematopoietic cells. In monocytes, it has been functionally linked to the 5-lipoxygenase pathway in which the leukotrienes, a class of highly potent inflammatory mediators, are produced. However, regarding CD69 gene expression and its regulatory mechanisms in monocytes, only scarce data are available. Here, we report that CD69 mRNA expression, analogous to that of 5-lipoxygenase, is induced by the physiologic stimuli transforming growth factor-β (TGF-β) and 1α,25-dihydroxyvitamin D3 (1α,25(OH)2D3) in monocytic cells. Comparison with T- and B-cell lines showed that the effect was specific for monocytes. CD69 expression levels were increased in a concentration-dependent manner, and kinetic analysis revealed a rapid onset of mRNA expression, indicating that CD69 is a primary TGF-β/1α,25(OH)2D3 target gene. PCR analysis of different regions of the CD69 mRNA revealed that de novo transcription was initiated and proximal and distal parts were induced concomitantly. In common with 5-lipoxygenase, no activation of 0.7 kb or ∼2.3 kb promoter fragments by TGF-β and 1α,25(OH)2D3 could be observed in transient reporter assays for CD69. Analysis of mRNA stability using a transcription inhibitor and a 3'UTR reporter construct showed that TGF-β and 1α,25(OH)2D3 do not influence CD69 mRNA stability. Functional knockdown of Smad3 clearly demonstrated that upregulation of CD69 mRNA, in contrast to 5-LO, depends on Smad3. Comparative studies with different inhibitors for mitogen activated protein kinases (MAPKs) revealed that MAPK signalling is involved in CD69 gene regulation, whereas 5-lipoxygenase gene expression was only partly affected. Mechanistically, we found evidence that CD69 gene upregulation depends on TAK1-mediated p38 activation. In summary, our data indicate that CD69 gene expression, conforming with 5-lipoxygenase, is regulated monocyte-specifically by the physiologic stimuli TGF-β and 1α,25(OH)2D3 on mRNA level, although different mechanisms account for the upregulation of each gene.
Collapse
MESH Headings
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Differentiation, T-Lymphocyte/genetics
- Antigens, Differentiation, T-Lymphocyte/metabolism
- Blotting, Western
- Cell Line
- Cell Line, Tumor
- Humans
- Lectins, C-Type/genetics
- Lectins, C-Type/metabolism
- Monocytes/drug effects
- Monocytes/metabolism
- Polymerase Chain Reaction
- Transforming Growth Factor beta/pharmacology
- Vitamin D/analogs & derivatives
- Vitamin D/pharmacology
Collapse
Affiliation(s)
- Thea K. Wöbke
- Institute of Pharmaceutical Chemistry/ZAFES (Zentrum für Arzneimittelforschung, Entwicklung und Sicherheit), Goethe-University, Frankfurt am Main, Germany
| | - Andreas von Knethen
- Institute of Biochemistry I-Pathobiochemistry, Faculty of Medicine, Goethe-University, Frankfurt am Main, Germany
| | - Dieter Steinhilber
- Institute of Pharmaceutical Chemistry/ZAFES (Zentrum für Arzneimittelforschung, Entwicklung und Sicherheit), Goethe-University, Frankfurt am Main, Germany
| | - Bernd L. Sorg
- Institute of Pharmaceutical Chemistry/ZAFES (Zentrum für Arzneimittelforschung, Entwicklung und Sicherheit), Goethe-University, Frankfurt am Main, Germany
| |
Collapse
|
90
|
Preclinical safety and immunogenicity evaluation of a nonavalent PorA native outer membrane vesicle vaccine against serogroup B meningococcal disease. Vaccine 2013; 31:1065-71. [DOI: 10.1016/j.vaccine.2012.12.031] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 11/29/2012] [Accepted: 12/12/2012] [Indexed: 11/17/2022]
|
91
|
Wilkinson A, Ballabio E, Geng H, North P, Tapia M, Kerry J, Biswas D, Roeder R, Allis C, Melnick A, de Bruijn M, Milne T. RUNX1 is a key target in t(4;11) leukemias that contributes to gene activation through an AF4-MLL complex interaction. Cell Rep 2013; 3:116-27. [PMID: 23352661 PMCID: PMC3607232 DOI: 10.1016/j.celrep.2012.12.016] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Revised: 11/08/2012] [Accepted: 12/26/2012] [Indexed: 12/22/2022] Open
Abstract
The Mixed Lineage Leukemia (MLL) protein is an important epigenetic regulator required for the maintenance of gene activation during development. MLL chromosomal translocations produce novel fusion proteins that cause aggressive leukemias in humans. Individual MLL fusion proteins have distinct leukemic phenotypes even when expressed in the same cell type, but how this distinction is delineated on a molecular level is poorly understood. Here, we highlight a unique molecular mechanism whereby the RUNX1 gene is directly activated by MLL-AF4 and the RUNX1 protein interacts with the product of the reciprocal AF4-MLL translocation. These results support a mechanism of transformation whereby two oncogenic fusion proteins cooperate by activating a target gene and then modulating the function of its downstream product.
Collapse
Affiliation(s)
- Adam C. Wilkinson
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Erica Ballabio
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Huimin Geng
- Departments of Medicine/Hematology and Oncology Division, Weill Medical College of Cornell University, New York, NY, 10065, USA
- Institute for Computational Biomedicine, Weill Medical College of Cornell University, New York, NY, 10065, USA
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Phillip North
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Marta Tapia
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Jon Kerry
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Debabrata Biswas
- Laboratory of Biochemistry and Molecular Biology, The Rockefeller University, New York, NY 10065, USA
| | - Robert G. Roeder
- Laboratory of Biochemistry and Molecular Biology, The Rockefeller University, New York, NY 10065, USA
| | - C. David Allis
- Laboratory of Chromatin Biology and Epigenetics, The Rockefeller University, New York, NY 10065, USA
| | - Ari Melnick
- Departments of Medicine/Hematology and Oncology Division, Weill Medical College of Cornell University, New York, NY, 10065, USA
- Department of Pharmacology, Weill Medical College of Cornell University, New York, NY, 10065, USA
| | - Marella F.T.R. de Bruijn
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Thomas A. Milne
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| |
Collapse
|
92
|
Ehlers C, Schirmer S, Kehlenbach RH, Hauber J, Chemnitz J. Post-transcriptional regulation of CD83 expression by AUF1 proteins. Nucleic Acids Res 2013; 41:206-19. [PMID: 23161671 PMCID: PMC3592417 DOI: 10.1093/nar/gks1069] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 09/27/2012] [Accepted: 10/11/2012] [Indexed: 12/31/2022] Open
Abstract
Mature dendritic cells (DC), activated lymphocytes, mononuclear cells and neutrophils express CD83, a surface protein apparently necessary for effective DC-mediated activation of naïve T-cells and T-helper cells, thymic T-cell maturation and the regulation of B-cell activation and homeostasis. Although a defined ligand of CD83 remains elusive, the multiple cellular subsets expressing CD83, as well as its numerous potential implications in immunological processes suggest that CD83 plays an important regulatory role in the mammalian immune system. Lately, nucleocytoplasmic translocation of CD83 mRNA was shown to be mediated by direct interaction between the shuttle protein HuR and a novel post-transcriptional regulatory element (PRE) located in the CD83 transcript's coding region. Interestingly, this interaction commits the CD83 mRNA to efficient nuclear export through the CRM1 protein translocation pathway. More recently, the cellular phosphoprotein and HuR ligand ANP32B (APRIL) was demonstrated to be directly involved in this intracellular transport process by linking the CD83 mRNA:HuR ribonucleoprotein (RNP) complex with the CRM1 export receptor. Casein kinase II regulates this process by phosphorylating ANP32B. Here, we identify another RNA binding protein, AUF1 (hnRNP D) that directly interacts with CD83 PRE. Unlike HuR:PRE binding, this interaction has no impact on intracellular trafficking of CD83 mRNA-containing complexes; but it does regulate translation of CD83 mRNA. Thus, our data shed more light on the complex process of post-transcriptional regulation of CD83 expression. Interfering with this process may provide a novel strategy for inhibiting CD83, and thereby cellular immune activation.
Collapse
Affiliation(s)
- Christina Ehlers
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Martinistrasse 52, D-20251 Hamburg and Zentrum für Biochemie und Molekulare Zellbiologie, Universität Göttingen, Humboldtallee 23, D-37073 Göttingen, Germany
| | - Susann Schirmer
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Martinistrasse 52, D-20251 Hamburg and Zentrum für Biochemie und Molekulare Zellbiologie, Universität Göttingen, Humboldtallee 23, D-37073 Göttingen, Germany
| | - Ralph H. Kehlenbach
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Martinistrasse 52, D-20251 Hamburg and Zentrum für Biochemie und Molekulare Zellbiologie, Universität Göttingen, Humboldtallee 23, D-37073 Göttingen, Germany
| | - Joachim Hauber
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Martinistrasse 52, D-20251 Hamburg and Zentrum für Biochemie und Molekulare Zellbiologie, Universität Göttingen, Humboldtallee 23, D-37073 Göttingen, Germany
| | - Jan Chemnitz
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Martinistrasse 52, D-20251 Hamburg and Zentrum für Biochemie und Molekulare Zellbiologie, Universität Göttingen, Humboldtallee 23, D-37073 Göttingen, Germany
| |
Collapse
|
93
|
Lajkó E, Szabó I, Andódy K, Pungor A, Mező G, Kőhidai L. Investigation on chemotactic drug targeting (chemotaxis and adhesion) inducer effect of GnRH-III derivatives in Tetrahymena and human leukemia cell line. J Pept Sci 2012. [PMID: 23208929 DOI: 10.1002/psc.2472] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
GnRH-III has been shown to exert a cytotoxic effect on the GnRH-R positive tumor cells. The chemotactic drug targeting (CDT) represents a new way for drug delivery approach based on selective chemoattractant guided targeting. The major goal of the present work was to develop and investigate various GnRH-III derivatives as potential targeting moieties for CDT. The cell physiological effects (chemotaxis, adhesion, and signaling) induced by three native GnRHs (hGnRH-I, cGnRH-II, and lGnRH-III) and nine GnRH-III derivatives were evaluated in two model cells (Tetrahymena pyriformis and Mono Mac 6 human monocytes). According to our results, the native GnRH-III elicited the highest chemoattractant and adhesion inducer activities of all synthesized peptides in micromolar concentrations in monocytes. With respect to chemoattraction, dimeric derivatives linked by a disulfide bridge ([GnRH-III(C)](2) ) proved to be efficient in both model cells; furthermore, acetylation of the linker region ([GnRH-III(Ac-C)](2) ) could slightly improve the chemotactic and adhesion effects in monocytes. The length of the peptide and the type of N-terminal amino acid could also determine the chemotactic and adhesion modulation potency of each fragment. The application of the chemoattractant GnRH-III derivatives was accompanied by a significant activation of phosphatidylinositol 3-kinase in both model cells. In summary, our work on low-level differentiated model cells of tumors has proved that GnRH-III and some of its synthetic derivatives are promising candidates to be applied in CDT: these compounds might act both as carrier, delivery unit, and antitumor agents.
Collapse
Affiliation(s)
- Eszter Lajkó
- Department of Genetics Cell and Immunobiology, Semmelweis University, Nagyvárad tér. 4, H-1089, Budapest, Hungary
| | | | | | | | | | | |
Collapse
|
94
|
Rodenburg GD, Fransen F, Bogaert D, Schipper K, Groenwold RHH, Hamstra HJ, Westerhuis BM, van de Beek D, van der Ley P, Sanders EAM, van der Ende A. Prevalence and clinical course in invasive infections with meningococcal endotoxin variants. PLoS One 2012; 7:e49295. [PMID: 23209568 PMCID: PMC3510230 DOI: 10.1371/journal.pone.0049295] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 10/07/2012] [Indexed: 11/20/2022] Open
Abstract
Background Meningococci produce a penta-acylated instead of hexa-acylated lipid A when their lpxL1 gene is inactivated. Meningococcal strains with such lipid A endotoxin variants have been found previously in adult meningitis patients, where they caused less blood coagulopathy because of decreased TLR4 activation. Methods A cohort of 448 isolates from patients with invasive meningococcal disease in the Netherlands were screened for the ability to induce IL-6 in monocytic cell Mono Mac 6 cells. The lpxL1 gene was sequenced of isolates, which show poor capacity to induce IL-6.. Clinical characteristics of patients were retrieved from hospital records. Results Of 448 patients, 29 (6.5%) were infected with meningococci expressing a lipid A variant strain. Lipid A variation was not associated with a specific serogroup or genotype. Infections with lipid A variants were associated with older age (19.3 vs. 5.9 (median) years, p = 0.007) and higher prevalence of underlying comorbidities (39% vs. 17%; p = 0.004) compared to wild-type strains. Patients infected with lipid A variant strains had less severe infections like meningitis or shock (OR 0.23; 95%CI 0.09–0.58) and were less often admitted to intensive care (OR 0.21; 95%CI 0.07–0.60) compared to wild-type strains, independent of age, underlying comorbidities or strain characteristics. Conclusions In adults with meningococcal disease lipid A variation is rather common. Infection with penta-acylated lipid A variant meningococci is associated with a less severe disease course.
Collapse
Affiliation(s)
- Gerwin D Rodenburg
- Department of Pediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Kunnen A, Dekker DC, van Pampus MG, Harmsen HJM, Aarnoudse JG, Abbas F, Faas MM. Cytokine production induced by non-encapsulated and encapsulated Porphyromonas gingivalis strains. Arch Oral Biol 2012; 57:1558-66. [PMID: 22902095 DOI: 10.1016/j.archoralbio.2012.07.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 07/26/2012] [Accepted: 07/27/2012] [Indexed: 01/21/2023]
Abstract
OBJECTIVE Although the exact reason is not known, encapsulated gram-negative Porphyromonas gingivalis strains are more virulent than non-encapsulated strains. Since difference in virulence properties may be due to difference in cytokine production following recognition of the bacteria or their products by the host inflammatory cells, we compared cytokine production following stimulation with bacteria or lipopolysaccharides (LPS) of a non-encapsulated and an encapsulated P. gingivalis strain (K(-) and K1). DESIGN Tumour necrosis factor-alpha (TNF-α) production following stimulation of the cell-line Mono Mac 6 with bacteria or LPS of both P. gingivalis strains was determined using flow cytometry. Furthermore, we investigated the effects of the two P. gingivalis strains or their LPS on TNF-α and Interleukin (IL-1β, IL-6, IL-12 and IL-10) production in whole blood using Luminex. In both experiments, Escherichia coli bacteria and LPS were used as a reference. RESULTS Both P. gingivalis strains induced lower cytokine production than E. coli with the exception of IL-6. P. gingivalis K1 bacteria elicited a higher overall cytokine production than P. gingivalis K(-). In contrast, P. gingivalis K1 LPS stimulation induced a lower cytokine production than P. gingivalis K(-) LPS. CONCLUSIONS Our findings suggest that the encapsulated P. gingivalis K1 bacteria induce higher cytokine production than the non-encapsulated P. gingivalis K(-). This was not due to its LPS. The stronger induction of cytokines may contribute to the higher virulence of P. gingivalis K1.
Collapse
Affiliation(s)
- Alina Kunnen
- Department of Periodontology, Center for Dentistry and Oral Hygiene, University Medical Center Groningen and University of Groningen, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
96
|
Saaby L, Nielsen CH. Triterpene acids from rose hip powder inhibit self-antigen- and LPS-induced cytokine production and CD4⁺ T-cell proliferation in human mononuclear cell cultures. Phytother Res 2012; 26:1142-7. [PMID: 22170858 DOI: 10.1002/ptr.3713] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Accepted: 10/21/2011] [Indexed: 12/30/2022]
Abstract
A triterpene acid mixture consisting of oleanolic, ursolic and betulinic acid isolated from a standardized rose hip powder (Rosa canina L.) has been shown to inhibit interleukin (IL)-6 release from Mono Mac 6 cells. The present study examined the effects of the triterpene acid mixture on the cytokine production and proliferation of CD4⁺ T cells and CD19⁺ B cells induced by a self-antigen, human thyroglobulin and by lipopolysaccharide in cultures of normal mononuclear cells. The triterpene acid mixture inhibited the production of tumor necrosis factor-α and IL-6 with estimated IC₅₀ values in the range 35-56 µg/mL, the Th1 cytokines interferon-γ and IL-2 (IC₅₀ values 10-20 µg/mL) and the antiinflammatory cytokine IL-10 (IC₅₀ values 18-21 µg/mL). Moreover, the mixture also inhibited CD4⁺ T-cell and CD19⁺ B-cell proliferation (IC₅₀ value 22 and 12 µg/mL, respectively). Together, these data demonstrate that oleanolic, ursolic and betulinic acid are active immunomodulatory constituents of the standardized rose hip powder. However, since the estimated IC₅₀ values are in the µg/mL range, it is questionable whether the content of the triterpene acids in the standardized rose hip powder, alone, can explain the reported clinical effects.
Collapse
Affiliation(s)
- Lasse Saaby
- Department of Rheumatology, Institute for Inflammation Research, Copenhagen University Hospital, Copenhagen, Denmark.
| | | |
Collapse
|
97
|
Castagna A, Polati R, Bossi AM, Girelli D. Monocyte/macrophage proteomics: recent findings and biomedical applications. Expert Rev Proteomics 2012; 9:201-15. [PMID: 22462790 DOI: 10.1586/epr.12.11] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Macrophages, originating from the migration and differentiation of circulating monocytes into virtually all tissues, are extremely flexible and plastic cells that play vital homeostatic roles, but also contribute to the pathophysiology of many human diseases. For these reasons, they are intensively studied by different approaches, recently including proteomics. Macrophage cells can be taken from a range of different sources, including blood monocytes and macrophages from tissues. Macrophages can also be generated by in vitro culture from blood monocytes, and cell lines derived from this lineage can be used. Similarly, many different proteomic techniques can be used, ranging from classic approaches based on 2D gel electrophoresis to more recent high-throughput gel-free techniques essentially based on mass spectrometry. Here, we review the application of such techniques to the study of monocytes/macrophages, and summarize some results potentially relevant to two paradigmatic conditions - atherosclerosis and disorders of iron metabolism.
Collapse
Affiliation(s)
- Annalisa Castagna
- Department of Medicine, Unit of Internal Medicine, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, Verona, Italy
| | | | | | | |
Collapse
|
98
|
Use of human MonoMac6 cells for development of in vitro assay predictive of adjuvant safety in vivo. Vaccine 2012; 30:4859-65. [DOI: 10.1016/j.vaccine.2012.05.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 04/09/2012] [Accepted: 05/01/2012] [Indexed: 11/19/2022]
|
99
|
Leurs U, Lajkó E, Mező G, Orbán E, Öhlschläger P, Marquardt A, Kőhidai L, Manea M. GnRH-III based multifunctional drug delivery systems containing daunorubicin and methotrexate. Eur J Med Chem 2012; 52:173-83. [DOI: 10.1016/j.ejmech.2012.03.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2012] [Revised: 03/05/2012] [Accepted: 03/06/2012] [Indexed: 11/16/2022]
|
100
|
Yuan C, Song G, Jiang G. The characterization and role of leukemia cell-derived dendritic cells in immunotherapy for leukemic diseases. Intractable Rare Dis Res 2012; 1:53-65. [PMID: 25343074 PMCID: PMC4204560 DOI: 10.5582/irdr.2012.v1.2.53] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2011] [Revised: 03/28/2012] [Accepted: 04/13/2012] [Indexed: 11/05/2022] Open
Abstract
Usually, an effective anti-leukemia immune response cannot be initiated effectively in patients with leukemia. This is probably related to immunosuppression due to chemotherapy, down-regulation of major histocompatibility complex (MHC) II molecules, and the lack of co-stimulatory molecules on dendritic cells (DC). In light of this problem, some methods had been used to induce leukemia cells to differentiate into mature DCs, causing them to present leukemia-associated antigens and activating naïve T cells. Furthermore, leukemia-derived DCs could be modified with tumor antigens or tumor-associated antigens to provide a new approach to anti-leukemia therapy. Numerous studies have indicated factors related to the induction and functioning of leukemia-derived DCs and the activation of cytotoxic T-lymphocytes (CTLs). These include the amount of purified DCs, cytokine profiles appropriate for inducing leukemia-derived DCs, effective methods of activating CTLs, reasonable approaches to DC vaccines, and the standardization of their clinical use. Determining these factors could lead to more effective leukemia treatment and benefit both mankind and scientific development. What follows in a review of advances in and practices of inducing leukemia-derived DCs and the feasibility of their clinical use.
Collapse
Affiliation(s)
- Changjin Yuan
- Key Laboratory for Tumor Immunology & Traditional Chinese Medicine Immunology, Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Department of Hemato-Oncology, Shandong Academy of Medical Sciences, Key Laboratory for Biotech-Drugs of the Ministry of Health, Key Laboratory for Modern Medicine and Technology of Shandong Province, Ji'nan, Shandong, China
| | - Guanhua Song
- Key Laboratory for Tumor Immunology & Traditional Chinese Medicine Immunology, Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Department of Hemato-Oncology, Shandong Academy of Medical Sciences, Key Laboratory for Biotech-Drugs of the Ministry of Health, Key Laboratory for Modern Medicine and Technology of Shandong Province, Ji'nan, Shandong, China
| | - Guosheng Jiang
- Key Laboratory for Tumor Immunology & Traditional Chinese Medicine Immunology, Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Department of Hemato-Oncology, Shandong Academy of Medical Sciences, Key Laboratory for Biotech-Drugs of the Ministry of Health, Key Laboratory for Modern Medicine and Technology of Shandong Province, Ji'nan, Shandong, China
- Address correspondence to: Prof. Guosheng Jiang, Department of Hemato-oncology, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jingshi Road 18877, Ji'nan 250062, Shandong, China. E-mail:
| |
Collapse
|