1
|
Cavaillon JM, Pitton C, Fitting C. Endotoxin tolerance is not a LPS-specific phenomenon: partial mimicry with IL-1, IL-10 and TGFβ. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/096805199400100105] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Administration of non-lethal doses of lipopolysaccharide (LPS) to experimental animals and humans results for a short period of time in a state of hyporesponsiveness to a second LPS challenge. This phenomenon, known as endotoxin tolerance, has been reproduced in vitro using human monocytes, rendered endotoxin-tolerant following a first incubation with LPS. A further activation by LPS was manifested by a dramatically reduced production of tumor necrosis factor α (TNFα). We report this low responsiveness of LPS pretreated monocytes as an endotoxin non-specific phenomenon. Indeed, TNFα release upon further activation with either killed Gram-positive bacteria (Staphylococci, Streptococci) or zymosan was also significantly diminished. This was not the case when phorbol myristate acetate (PMA) was used as a second triggering agent, suggesting that the monocyte hyporesponsiveness due to LPS does not affect all activation pathways, particularly that of protein kinase C. On the other hand, both PMA and zymosan pretreatment could reduce a further activation of monocytes by LPS. We investigated whether the first signal(s) delivered by LPS, could be mimicked by some of the LPS-induced cytokines. Preincubation of monocytes with either interleukin-1 (IL-1), IL-10 or transforming growth factor β (TGFβ) lower the TNFα production upon further activation with LPS. None of the cytokines alone was as efficient as the LPS molecule, but high levels of tolerization were obtained with combination of IL-1, IL-10 and TGFβ. Neither IL-6, IL-8 nor TNFα led to hyporeactive cells. Our data indicate that endotoxin tolerance is not an LPS-specific phenomenon and that more than one cytokine can contribute to render human monocytes hyporeactive to further activation by LPS.
Collapse
Affiliation(s)
| | - C. Pitton
- Unité d'Immuno-Allergie, Institut Pasteur, Paris, France
| | - C. Fitting
- Unité d'Immuno-Allergie, Institut Pasteur, Paris, France
| |
Collapse
|
2
|
Hirohashi N, Lei MG, Morrison DC. LPS pretreatment of mouse peritoneal macrophages differentially modulates TNFα and iNOS expression. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519990050050101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Many studies have established that pretreatment of mouse macrophages with LPS will alter subsequent responsiveness of these cells to stimulation either with LPS or other stimuli. Incubation of C3Heb/FeJ mouse macrophages with low sub-stimulatory (e.g. 0.10 ng/ml) LPS for 6 h, followed by activation with 100 ng/ml of LPS results in up-regulation of LPS-dependent TNFα production and suppression of the ability of these cells to secrete nitric oxide (NO). To assess whether these two responses might be co-ordinately regulated, immunocytochemical analyses of LPS-pretreated macrophages were carried out using anti-TNFα and anti-iNOS antibodies, both with and without a period of LPS pretreatment. As anticipated, the detection of individual iNOS-expressing macrophages in LPS-stimulated cultures accurately reflected threshold doses of LPS required for detection of NO in culture supernatants in terms of frequency of percentage of the total population positive for expression of iNOS protein. Pretreatment with sub-stimulatory doses of LPS significantly reduced the frequency of these iNOS-expressing macrophages responsive to subsequent LPS stimulation, supporting the concept of iNOS down-regulation at the pretranslational level. In contrast, the detection of TNFα-expressing macrophages in LPS-stimulated cultures did not correlate directly with the detection of TNFα in culture supernatants. Further, pretreatment with sub-stimulatory doses of LPS did not always correlate with the frequency of TNFα-expressing cells in response to LPS treatment. These results support the concept that different regulatory mechanisms may be operative in differential regulation of macrophage TNFα and NO responsiveness by pretreatment of macrophages with sub-stimulatory doses of LPS.
Collapse
Affiliation(s)
| | - Mei-Guey Lei
- University of Kansas Medical Center, Kansas City, Kansas, USA
| | | |
Collapse
|
3
|
Durando M, Ashton S, Makhlouf M, Simmons-Wagner R, Halushka P, Cook J. Endotoxin-induced desensitization of THP-1 cells is not associated with altered G protein binding or content. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/096805199700400203] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In rats endotoxin tolerance is characterized by decreased endotoxin-stimulated peritoneal macrophage arachidonic acid metabolism and decreased GTP binding protein function. The hypothesis that THP-1 cells can be altered in a similar manner by pretreatment with endotoxin was tested. These studies examined endotoxin's ability to stimulate eicosanoid and tumor necrosis factor α (TNFα) in control and desensitized THP 1 cells. Additionally, membrane GTPγ 35S binding and Western blot analyses with specific antisera to G i1,2α, Gi3a, Gαcommon, and the β subunit of G in control and endotoxin-desensitized THP-1 cells were assessed. Endotoxin (10 μg/ml) stimulated thromboxane (Tx) B2 production in THP-1 cells. Pretreatment with pertussis toxin (PT), resulted in significant inhibition of TxB2 production at concentrations not inhibited by equimolar concentrations of PT-B protomer. The latter observations suggest a role of PT-sensitive G protein in endotoxin activation of THP-1 cells. Pre-exposure to endotoxin (1 μg/ml) for 18 h desensitized THP-1 cells to endotoxin-stimulated TxB2 production and endotoxin-stimulated TNFα. To determine if endotoxin pretreatment affects G protein function, THP-1 cell membranes were isolated from endotoxin pretreated and control cells for equilibrium binding with GTPγ35S, a nonhydrolyzable analog of GTP. Neither the total number of binding sites (Bmax) nor the dissociation constant (Kd) for GTPγ35S in desensitized THP-1 cells were significantly different from those of control cells. PT-catalyzed ADP-ribosylation of G proteins in control and LPS-desensitized THP-1 cells demonstrated no difference in the quantity of G protein labelled versus desensitized cells. Immunoblots also showed no difference between control and desensitized cells in the membrane content of specific heterotrimeric G proteins. The data demonstrate that pre-exposure to endotoxin desensitizes the cells subsequent endotoxin stimulation of mediator production. However, unlike the in vivo rat model, this is not associated with a decrease in G protein binding or content.
Collapse
Affiliation(s)
- M. Durando
- Departments of Physiology, Division of Clinical Pharmacology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - S.H. Ashton
- Departments of Physiology, Division of Clinical Pharmacology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - M.A. Makhlouf
- Departments of Physiology, Division of Clinical Pharmacology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - R. Simmons-Wagner
- Departments of Physiology, Division of Clinical Pharmacology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - P.V. Halushka
- Pharmacology and Medicine, Division of Clinical Pharmacology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - J.A. Cook
- Departments of Physiology, Division of Clinical Pharmacology, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
4
|
Cavaillon JM, Adrie C, Fitting C, Adib-Conquy M. Endotoxin tolerance: is there a clinical relevance? ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519030090020501] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Beeson (1946) first defined endotoxin tolerance as a reduced endotoxin-induced fever following repeated injections of typhoid vaccine. Freudenberg and Galanos (1988) demonstrated that endotoxin tolerance that can protect against a lethal challenge of lipopolysaccharide (LPS) involves the participation of macrophages. Evans and Zuckerman (1991) reported a role for glucocorticoids in endotoxin tolerance. Prostaglandins, interleukin-(IL-)10, and transforming growth factor-β are other players of in vivo endotoxin tolerance. Dramatic reduction of plasma tumor necrosis factor (TNF) (Mathison et al. 1990) and other cytokines in response to LPS parallels endotoxin tolerance. The reduced capacity to produce TNF and other cytokines can be mimicked in vitro by pretreatment of monocytes or macrophages with LPS. It is not a specific phenomenon and can be induced by other agents or events. Cross-tolerance between LPS, TLR2 specific ligands, IL-1 and TNF has been regularly reported. A similar loss of LPS-reactivity has been repeatedly reported in leukocytes of septic patients and in patients with non-infectious systemic inflammation response syndrome (SIRS; e.g . surgery, trauma, cardiac arrest and resuscitation, etc.). Studies on cellular signaling within leukocytes from septic and SIRS patients reveal numerous alterations of the activation pathways reminiscent of those observed in endotoxin-tolerant cells. While endotoxin tolerance prevents severity of infections and ischemia-reperfusion damage, it has been suggested that the immune dysregulation observed in SIRS patients was associated with an enhanced sensitivity to nosocomial infections. In conclusion, in vitro and in vivo endotoxin tolerance, either experimental or due to clinical status, are similar but not identical.
Collapse
Affiliation(s)
- Jean-Marc Cavaillon
- UP Cytokines & Inflammation, Institut Pasteur, Paris, France, , Intensive Care Unit, Hôpital Delafontaine, St Denis, France
| | - Christophe Adrie
- UP Cytokines & Inflammation, Institut Pasteur, Paris, France, Intensive Care Unit, Hôpital Delafontaine, St Denis, France
| | - Catherine Fitting
- UP Cytokines & Inflammation, Institut Pasteur, Paris, France, Intensive Care Unit, Hôpital Delafontaine, St Denis, France
| | - Minou Adib-Conquy
- UP Cytokines & Inflammation, Institut Pasteur, Paris, France, Intensive Care Unit, Hôpital Delafontaine, St Denis, France
| |
Collapse
|
5
|
Flach R, Schade F. Peritoneal macrophages from endotoxin-tolerant mice produce an inhibitor of tumor necrosis factor α synthesis and protect against endotoxin shock. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/096805199700400401] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In the present study it was found that development of early endotoxin-tolerance is associated with the capacity of mouse peritoneal macrophages (MPM) to produce an activity interfering with the synthesis of tumor necrosis factor α. Peritoneal macrophages from LPS-tolerant mice (tMPM), treated with LPS in vitro produced less TNFα, IL-10 and TGFβ than LPS-treated macrophages from normal mice (nMPM). The supernatants of LPS-activated tMPM contained activities which suppressed formation of TNF in nMPM and RAW 264.7 cells as determined by bioassay, ELISA and PCR. Supernatants of nMPM and unstimulated tMPM were devoid of the inhibitory activity. The inhibitor did not interfere with the bioactivity of TNFα in WEHI cells. It also suppressed PMA/IFN-γ induced TNF synthesis in macrophage cultures. The transfer of macrophages isolated from endotoxin-tolerant mice into normal mice protected against endotoxin shock, whereas macrophages from normal mice increased susceptibility to endotoxin.
Collapse
Affiliation(s)
- R. Flach
- Klinische Forschergruppe Schock und Multiorganversagen, Department for Surgery, University Hospital Essen, Essen, Germany
| | - F.U. Schade
- Klinische Forschergruppe Schock und Multiorganversagen, Department for Surgery, University Hospital Essen, Essen, Germany
| |
Collapse
|
6
|
Nikola Kimmings A, Pajkrt D, Zaaijer K, Moojen TM, Meenan JK, ten Cate JW, van Deventer SJ. Factors involved in early in vitro endotoxin hyporesponsiveness in human endotoxemia. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/096805199600300401] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Sepsis and endotoxemia induce a state of early tolerance to the biological effects of endotoxin. Relatively little is known about the mechanisms or mediators involved in this endotoxin hyporesponsiveness. In this study endotoxin-induced TNF production of whole blood obtained at different time points from endotoxin challenged volunteers, and endotoxin- and PHA-induced TNF production by healthy PBMNCs in the presence of serum from endotoxin challenged volunteers were compared. Endotoxin-induced TNF production by whole blood was found to be significantly reduced at 2 h after in vivo endotoxin administration, returning to baseline levels at t = 6 h. In addition, a reduction of TNF produced by healthy PBMNCs in the presence of post-endotoxin serum was observed. No correlation was found with the concentration of any of the well described anti-inflammatory mediators investigated nor with plasma levels of endotoxin-binding proteins.
Collapse
Affiliation(s)
- A. Nikola Kimmings
- Department of Surgery, Center for Hemostasis, Thrombosis, Atherosclerosis and Inflammation Research
| | - Dasja Pajkrt
- Laboratory of Experimental Internal Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Krista Zaaijer
- Center for Hemostasis, Thrombosis, Atherosclerosis and Inflammation Research
| | - Thybout M. Moojen
- Department of Surgery, Laboratory of Experimental Internal Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - John K. Meenan
- Laboratory of Experimental Internal Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Jan W. ten Cate
- Center for Hemostasis, Thrombosis, Atherosclerosis and Inflammation Research
| | - Sander J.H. van Deventer
- Laboratory of Experimental Internal Medicine, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Silverstein R, Johnson W, Bucklin S, Johnson D. The protein kinase C activator PMA modulates LPS lethality in normal mice and protects against LPS lethality in D-galactosamine-sensitized mice. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/096805199600300104] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Pretreatment for 5 h with 10 μg phorbol-12-myristate-13-acetate (PMA), a well established activator of protein kinase C (PKC) in many kinds of cells including macrophages, was found to either (a) delay, or (b) potentiate, lethal endotoxin shock in mice, depending upon the dose of LPS. The latter occurred despite a marked attenuation (>90%) of the TNFα response to LPS. In mice sensitized with D-galactosamine the same PMA pretreatment offered protection against challenge from either LPS or TNFα. This protection, coupled with the ability of PMA to reduce serum TNFα while increasing serum corticosterone in response to LPS, adds in vivo support for a possible role for PKC activation in early endotoxin tolerance. A phorbol ester (4α-phorbol) that is not a PKC activator was found ineffective. PMA produced a prompt and profound decrease in body temperature which reached a nadir at 3 h. However, the protective effect produced by PMA was not dependent upon a decrease in body temperature per se, but was dependent upon administration of PMA at the same time or prior to LPS.
Collapse
Affiliation(s)
- R. Silverstein
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - W.M. Johnson
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - S.E. Bucklin
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - D.C. Johnson
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, Kansas, USA, Department of Physiology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
8
|
Ussher JE, van Wilgenburg B, Hannaway RF, Ruustal K, Phalora P, Kurioka A, Hansen TH, Willberg CB, Phillips RE, Klenerman P. TLR signaling in human antigen-presenting cells regulates MR1-dependent activation of MAIT cells. Eur J Immunol 2016; 46:1600-14. [PMID: 27105778 PMCID: PMC5297987 DOI: 10.1002/eji.201545969] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 02/27/2016] [Accepted: 04/18/2016] [Indexed: 12/14/2022]
Abstract
Mucosal-associated invariant T (MAIT) cells are an abundant innate-like T lymphocyte population that are enriched in liver and mucosal tissues. They are restricted by MR1, which presents antigens derived from a metabolic precursor of riboflavin synthesis, a pathway present in many microbial species, including commensals. Therefore, MR1-mediated MAIT cell activation must be tightly regulated to prevent inappropriate activation and immunopathology. Using an in vitro model of MR1-mediated activation of primary human MAIT cells, we investigated the mechanisms by which it is regulated. Uptake of intact bacteria by antigen presenting cells (APCs) into acidified endolysosomal compartments was required for efficient MR1-mediated MAIT cell activation, while stimulation with soluble ligand was inefficient. Consistent with this, little MR1 was seen at the surface of human monocytic (THP1) and B-cell lines. Activation with a TLR ligand increased the amount of MR1 at the surface of THP1 but not B-cell lines, suggesting differential regulation in different cell types. APC activation and NF-κB signaling were critical for MR1-mediated MAIT cell activation. In primary cells, however, prolonged TLR signaling led to downregulation of MR1-mediated MAIT cell activation. Overall, MR1-mediated MAIT cell activation is a tightly regulated process, dependent on integration of innate signals by APCs.
Collapse
Affiliation(s)
- James E Ussher
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, Oxfordshire, United Kingdom.,Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Bonnie van Wilgenburg
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, Oxfordshire, United Kingdom
| | - Rachel F Hannaway
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Kerstin Ruustal
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, Oxfordshire, United Kingdom
| | - Prabhjeet Phalora
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, Oxfordshire, United Kingdom
| | - Ayako Kurioka
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, Oxfordshire, United Kingdom
| | - Ted H Hansen
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Christian B Willberg
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, Oxfordshire, United Kingdom.,Oxford NIHR Biomedical Research Centre, The John Radcliffe Hospital, Oxford, United Kingdom
| | - Rodney E Phillips
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, Oxfordshire, United Kingdom
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, Oxfordshire, United Kingdom.,Oxford NIHR Biomedical Research Centre, The John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
9
|
Gobejishvili L, Ghare S, Khan R, Cambon A, Barker DF, Barve S, McClain C, Hill D. Misoprostol modulates cytokine expression through a cAMP pathway: Potential therapeutic implication for liver disease. Clin Immunol 2015; 161:291-9. [PMID: 26408955 DOI: 10.1016/j.clim.2015.09.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 07/09/2015] [Accepted: 09/15/2015] [Indexed: 12/20/2022]
Abstract
Dysregulated cytokine metabolism plays a critical role in the pathogenesis of many forms of liver disease, including alcoholic and non-alcoholic liver disease. In this study we examined the efficacy of Misoprostol in modulating LPS-inducible TNFα and IL-10 expression in healthy human subjects and evaluated molecular mechanisms for Misoprostol modulation of cytokines in vitro. Healthy subjects were given 14day courses of Misoprostol at doses of 100, 200, and 300μg four times a day, in random order. Baseline and LPS-inducible cytokine levels were examined ex vivo in whole blood at the beginning and the end of the study. Additionally, in vitro studies were performed using primary human PBMCs and the murine macrophage cell line, RAW 264.7, to investigate underlying mechanisms of misoprostol on cytokine production. Administration of Misoprostol reduced LPS inducible TNF production by 29%, while increasing IL-10 production by 79% in human subjects with no significant dose effect on ex vivo cytokine activity; In vitro, the effect of Misoprostol was largely mediated by increased cAMP levels and consequent changes in CRE and NFκB activity, which are critical for regulating IL-10 and TNF expression. Additionally, chromatin immunoprecipitation (ChIP) studies demonstrated that Misoprostol treatment led to changes in transcription factor and RNA Polymerase II binding, resulting in changes in mRNA levels. In summary, Misoprostol was effective at beneficially modulating TNF and IL-10 levels both in vivo and in vitro; these studies suggest a potential rationale for Misoprostol use in ALD, NASH and other liver diseases where inflammation plays an etiologic role.
Collapse
Affiliation(s)
- Leila Gobejishvili
- Department of Internal Medicine, University of Louisville, United States; Department of Pharmacology & Toxicology, University of Louisville, United States.
| | - Smita Ghare
- Department of Internal Medicine, University of Louisville, United States
| | - Rehan Khan
- RK Gastroenterology & Hepatology, Coppell, TX, United States
| | - Alexander Cambon
- Department of Bioinformatics & Biostatistics, University of Louisville, United States
| | - David F Barker
- Department of Internal Medicine, University of Louisville, United States
| | - Shirish Barve
- Department of Internal Medicine, University of Louisville, United States; Department of Pharmacology & Toxicology, University of Louisville, United States
| | - Craig McClain
- Department of Internal Medicine, University of Louisville, United States; Department of Pharmacology & Toxicology, University of Louisville, United States; VA Medical Center, Louisville, KY, United States
| | - Daniell Hill
- Department of Internal Medicine, University of Louisville, United States; VA Medical Center, Louisville, KY, United States
| |
Collapse
|
10
|
de Vallière C, Wang Y, Eloranta JJ, Vidal S, Clay I, Spalinger MR, Tcymbarevich I, Terhalle A, Ludwig MG, Suply T, Fried M, Kullak-Ublick GA, Frey-Wagner I, Scharl M, Seuwen K, Wagner CA, Rogler G. G Protein-coupled pH-sensing Receptor OGR1 Is a Regulator of Intestinal Inflammation. Inflamm Bowel Dis 2015; 21:1269-81. [PMID: 25856770 PMCID: PMC4450952 DOI: 10.1097/mib.0000000000000375] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 01/27/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND A novel family of proton-sensing G protein-coupled receptors, including OGR1, GPR4, and TDAG8, was identified to be important for physiological pH homeostasis and inflammation. Thus, we determined the function of proton-sensing OGR1 in the intestinal mucosa. MTEHODS OGR1 expression in colonic tissues was investigated in controls and patients with IBD. Expression of OGR1 upon cell activation was studied in the Mono Mac 6 (MM6) cell line and primary human and murine monocytes by real-time PCR. Ogr1 knockout mice were crossbred with Il-10 deficient mice and studied for more than 200 days. Microarray profiling was performed using Ogr1 and Ogr1 (WT) residential peritoneal macrophages. RESULTS Patients with IBD expressed higher levels of OGR1 in the mucosa than non-IBD controls. Treatment of MM6 cells with TNF, led to significant upregulation of OGR1 expression, which could be reversed by the presence of NF-κB inhibitors. Kaplan-Meier survival analysis showed a significantly delayed onset and progression of rectal prolapse in female Ogr1/Il-10 mice. These mice displayed significantly less rectal prolapses. Upregulation of gene expression, mediated by OGR1, in response to extracellular acidification in mouse macrophages was enriched for inflammation and immune response, actin cytoskeleton, and cell-adhesion gene pathways. CONCLUSIONS OGR1 expression is induced in cells of human macrophage lineage and primary human monocytes by TNF. NF-κB inhibition reverses the induction of OGR1 expression by TNF. OGR1 deficiency protects from spontaneous inflammation in the Il-10 knockout model. Our data indicate a pathophysiological role for pH-sensing receptor OGR1 during the pathogenesis of mucosal inflammation.
Collapse
Affiliation(s)
- Cheryl de Vallière
- Division of Gastroenterology and Hepatology, University Hospital Zürich, Zürich, Switzerland
| | - Yu Wang
- Division of Gastroenterology and Hepatology, University Hospital Zürich, Zürich, Switzerland
- Institute of Physiology, University of Zürich, Zürich, Switzerland
| | - Jyrki J. Eloranta
- Department of Clinical Pharmacology and Toxicology, University Hospital Zürich, Zürich, Switzerland; and
| | - Solange Vidal
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Ieuan Clay
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Marianne R. Spalinger
- Division of Gastroenterology and Hepatology, University Hospital Zürich, Zürich, Switzerland
| | - Irina Tcymbarevich
- Division of Gastroenterology and Hepatology, University Hospital Zürich, Zürich, Switzerland
| | - Anne Terhalle
- Division of Gastroenterology and Hepatology, University Hospital Zürich, Zürich, Switzerland
| | | | - Thomas Suply
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Michael Fried
- Division of Gastroenterology and Hepatology, University Hospital Zürich, Zürich, Switzerland
| | - Gerd A. Kullak-Ublick
- Department of Clinical Pharmacology and Toxicology, University Hospital Zürich, Zürich, Switzerland; and
| | - Isabelle Frey-Wagner
- Division of Gastroenterology and Hepatology, University Hospital Zürich, Zürich, Switzerland
| | - Michael Scharl
- Division of Gastroenterology and Hepatology, University Hospital Zürich, Zürich, Switzerland
| | - Klaus Seuwen
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | | | - Gerhard Rogler
- Division of Gastroenterology and Hepatology, University Hospital Zürich, Zürich, Switzerland
| |
Collapse
|
11
|
Lee KH, Biswas A, Liu YJ, Kobayashi KS. Proteasomal degradation of Nod2 protein mediates tolerance to bacterial cell wall components. J Biol Chem 2012; 287:39800-11. [PMID: 23019338 DOI: 10.1074/jbc.m112.410027] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The innate immune system serves as the first line of defense by detecting microbes and initiating inflammatory responses. Although both Toll-like receptor (TLR) and nucleotide binding domain and leucine-rich repeat (NLR) proteins are important for this process, their excessive activation is hazardous to hosts; thus, tight regulation is required. Endotoxin tolerance is refractory to repeated lipopolysaccharide (LPS) stimulation and serves as a host defense mechanism against septic shock caused by an excessive TLR4 response during gram-negative bacterial infection. Gram-positive bacteria as well as their cell wall components also induce shock. However, the mechanism underlying tolerance is not understood. Here, we show that activation of Nod2 by its ligand, muramyl dipeptide (MDP) in the bacterial cell wall, induces rapid degradation of Nod2, which confers MDP tolerance in vitro and in vivo. Nod2 is constitutively associated with a chaperone protein, Hsp90, which is required for Nod2 stability and protects Nod2 from degradation. Upon MDP stimulation, Hsp90 rapidly dissociates from Nod2, which subsequently undergoes ubiquitination and proteasomal degradation. The SOCS-3 protein induced by Nod2 activation further facilitates this degradation process. Therefore, Nod2 protein stability is a key factor in determining responsiveness to MDP stimulation. This indicates that TLRs and NLRs induce a tolerant state through distinct molecular mechanisms that protect the host from septic shock.
Collapse
Affiliation(s)
- Kyoung-Hee Lee
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | | | | | | |
Collapse
|
12
|
Yang Q, Calvano SE, Lowry SF, Androulakis IP. A dual negative regulation model of Toll-like receptor 4 signaling for endotoxin preconditioning in human endotoxemia. Math Biosci 2011; 232:151-63. [PMID: 21624378 DOI: 10.1016/j.mbs.2011.05.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Revised: 05/10/2011] [Accepted: 05/16/2011] [Indexed: 12/17/2022]
Abstract
We discuss a model illustrating how the outcome of repeated endotoxin administration experiments can emerge as a natural consequence of the tightly regulated signaling pathways and also highlight the importance of a dual negative feedback regulation including PI3K/Akt and IRAK-M (IRAK3). We identify the relative time scales of the onset and the magnitude of the stimulus as key determinants of outcome in repeated administration experiments. The results of our simulations involve potentiated response, tolerance, and protective tolerance. Moreover, the knockout of negative regulators shows that IRAK-M is a necessary and sufficient factor for generation of endotoxin tolerance (ET). The effects of the knockout of IRAK-M gene or administration of PI3K inhibitor do yield predictions that have been verified experimentally. Finally, the pretreatment with PI3K inhibitor reveals the interaction between these two negative regulations.
Collapse
Affiliation(s)
- Qian Yang
- Chemical Engineering, Rutgers University, Piscataway, NJ 08854, USA.
| | | | | | | |
Collapse
|
13
|
Gobejishvili L, Avila DV, Barker DF, Ghare S, Henderson D, Brock GN, Kirpich IA, Joshi-Barve S, Mokshagundam SPL, McClain CJ, Barve S. S-adenosylmethionine decreases lipopolysaccharide-induced phosphodiesterase 4B2 and attenuates tumor necrosis factor expression via cAMP/protein kinase A pathway. J Pharmacol Exp Ther 2011; 337:433-43. [PMID: 21266552 DOI: 10.1124/jpet.110.174268] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
S-Adenosylmethionine (SAM) treatment has anti-inflammatory, cytoprotective effects against endotoxin-induced organ injury. An important component of the anti-inflammatory action of SAM involves down-regulation of the lipopolysaccharide (LPS)-induced transcriptional induction of tumor necrosis factor-α (TNF) expression by monocytes/macrophages. We examined the effect of SAM on expression and activity of LPS-induced up-regulation of phosphodiesterase 4 (PDE4), which regulates cellular cAMP levels and TNF expression. LPS treatment of RAW 264.7, a mouse macrophage cell line, led to the induction of Pde4b2 mRNA expression with no effect on Pde4a or Pde4d. SAM pretreatment led to a significant decrease in LPS-induced up-regulation of Pde4b2 expression in both RAW 264.7 cells and primary human CD14(+) monocytes. Of note, the decreased Pde4b2 mRNA expression correlated with the SAM-dependent increase in the transcriptionally repressive histone H3 lysine 9 trimethylation on the Pde4b2 intronic promoter region. The SAM-mediated decrease in LPS-inducible Pde4b2 up-regulation resulted in an increase in cellular cAMP levels and activation of cAMP-dependent protein kinase A (PKA), which plays an inhibitory role in LPS-induced TNF production. In addition, SAM did not affect LPS-inducible inhibitor of nuclear factor-κB degradation or nuclear factor-κB (NF-κB)-p65 translocation into the nucleus but rather inhibited NF-κB transcriptional activity. These results demonstrate for the first time that inhibition of LPS-induced PDE4B2 up-regulation and increased cAMP-dependent PKA activation are significant mechanisms contributing to the anti-TNF effect of SAM. Moreover, these data also suggest that SAM may be used as an effective PDE4B inhibitor in the treatment of chronic inflammatory disorders in which TNF expression plays a significant pathogenic role.
Collapse
Affiliation(s)
- Leila Gobejishvili
- Department of Internal Medicine, University of Louisville Medical Center, 505 S. Hancock St., Louisville, KY 40202, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Nahid MA, Pauley KM, Satoh M, Chan EKL. miR-146a is critical for endotoxin-induced tolerance: IMPLICATION IN INNATE IMMUNITY. J Biol Chem 2009; 284:34590-9. [PMID: 19840932 PMCID: PMC2787321 DOI: 10.1074/jbc.m109.056317] [Citation(s) in RCA: 325] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The human toll-like receptor 4 (TLR4) pathway is activated in response to lipopolysaccharide (LPS), and subsequent signal transductions lead to the production of cytokines such as tumor necrosis factor-α (TNF-α) by innate immune cells. Defects in innate immune response may contribute to the overproduction of TNF-α leading to systemic inflammation and diseases. Thus, the innate immune response needs to be tightly regulated by elaborate mechanisms to control its onset and termination. LPS tolerance is a state of hyporesponsiveness to subsequent LPS challenge and is achieved by monocytic cells after prolonged exposure to LPS. In this report, kinetics of endotoxin-responsive microRNAs expression analysis revealed a unique pattern of gradual increase for miR-146a starting 4 h after LPS stimulation in THP-1 cells and continued up to 35-fold over 24 h. Conversely, TNF-α increased up to 4 h and then decreased gradually implicating a negative correlation with miR-146a progression. The characteristic up-regulation of miR-146a toward subsequent LPS challenge in THP-1 cells was studied. Strikingly, microRNA expression analysis during the tolerized state of THP-1 cells showed only miR-146a overexpression suggesting its important role in LPS tolerance. In addition, LPS tolerance was dependent on a LPS-priming dose and associated miR-146a up-regulation. LPS-tolerized cells were observed to regain responsiveness in TNF-α production 22 h after LPS removal correlating with a decrease in miR-146a level. Transfection of miR-146a into THP-1 cells mimicked LPS priming, whereas transfection of miR-146a inhibitor largely abolished LPS tolerance. Thus our studies demonstrated that miR-146a is critical for the in vitro monocytic cell-based endotoxin tolerance.
Collapse
Affiliation(s)
- Md A Nahid
- Department of Oral Biology, University of Florida, Gainesville, Florida 32610, USA
| | | | | | | |
Collapse
|
15
|
Downregulation of CD40 ligand response in monocytes from sepsis patients. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2008; 15:1851-8. [PMID: 18945879 DOI: 10.1128/cvi.00184-08] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
It has been suggested that a defective adaptive immune response contributes to septic immunosuppression. Here, the response of monocytes to CD40 ligand (CD40L) for patients with sepsis due to infection with gram-negative organisms has been analyzed. Compared to cells from controls, monocytes from septic patients showed significantly reduced production of tumor necrosis factor alpha, interleukin-1beta (IL-1beta), and IL-12 and were unable to acquire high levels of CD80 and CD86 molecules. These alterations were observed at the onset of sepsis and persisted at day 7. However, the ability of monocytes to respond to CD40L stimulation was partially but significantly restored in cells from patients who recovered from sepsis. In addition, costimulation of autologous CD4+ T lymphocytes by CD40L-activated monocytes from septic patients failed to induce cell proliferation and gamma interferon production. Finally, the ability of CD40L to rescue monocytes from apoptosis was severely impaired. We conclude that downregulation of the CD40L response may be an appropriate model for the monocyte alteration observed during septic immunosuppression and may help in the development of novel therapeutic strategies.
Collapse
|
16
|
Gobejishvili L, Barve S, Joshi-Barve S, McClain C. Enhanced PDE4B expression augments LPS-inducible TNF expression in ethanol-primed monocytes: relevance to alcoholic liver disease. Am J Physiol Gastrointest Liver Physiol 2008; 295:G718-24. [PMID: 18687753 PMCID: PMC2575909 DOI: 10.1152/ajpgi.90232.2008] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Increased plasma and hepatic TNF-alpha expression is well documented in patients with alcoholic hepatitis and is implicated in the pathogenesis of alcoholic liver disease. We have previously shown that monocytes from patients with alcoholic hepatitis show increased constitutive and LPS-induced NF-kappaB activation and TNF-alpha production. Our recent studies showed that chronic ethanol exposure significantly decreased cellular cAMP levels in both LPS-stimulated and unstimulated monocytes and Kupffer cells, leading to an increase in LPS-inducible TNF-alpha production by affecting NF-kappaB activation and induction of TNF mRNA expression. Accordingly, the mechanisms underlying this ethanol-induced decrease in cellular cAMP leading to an increase in TNF expression were examined in monocytes/macrophages. In this study, chronic ethanol exposure was observed to significantly increase LPS-inducible expression of cAMP-specific phosphodiesterase (PDE)4B that degrades cellular cAMP. Increased PDE4B expression was associated with enhanced NF-kappaB activation and transcriptional activity and subsequent priming of monocytes/macrophages leading to enhanced LPS-inducible TNF-alpha production. Selective inhibition of PDE4 by rolipram abrogated LPS-mediated TNF-alpha expression at both protein and mRNA levels in control and ethanol-treated cells. Notably, PDE4 inhibition did not affect LPS-inducible NF-kappaB activation but significantly decreased NF-kappaB transcriptional activity. These findings strongly support the pathogenic role of PDE4B in the ethanol-mediated priming of monocytes/macrophages and increased LPS-inducible TNF production and the subsequent development of alcoholic liver disease (ALD). Since enhanced TNF expression plays a significant role in the evolution of clinical and experimental ALD, its downregulation via selective PDE4B inhibitors could constitute a novel therapeutic approach in the treatment of ALD.
Collapse
Affiliation(s)
- Leila Gobejishvili
- Departments of Internal Medicine and Pharmacology and Toxicology, University of Louisville Medical Center; Louisville Veterans Affairs Medical Center, Louisville, Kentucky
| | - Shirish Barve
- Departments of Internal Medicine and Pharmacology and Toxicology, University of Louisville Medical Center; Louisville Veterans Affairs Medical Center, Louisville, Kentucky
| | - Swati Joshi-Barve
- Departments of Internal Medicine and Pharmacology and Toxicology, University of Louisville Medical Center; Louisville Veterans Affairs Medical Center, Louisville, Kentucky
| | - Craig McClain
- Departments of Internal Medicine and Pharmacology and Toxicology, University of Louisville Medical Center; Louisville Veterans Affairs Medical Center, Louisville, Kentucky
| |
Collapse
|
17
|
Eswarappa SM, Basu N, Joy O, Chakravortty D. Folimycin (concanamycin A) inhibits LPS-induced nitric oxide production and reduces surface localization of TLR4 in murine macrophages. Innate Immun 2008; 14:13-24. [PMID: 18387916 DOI: 10.1177/1753425907087349] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Lipopolysaccharide (LPS) is a major cell wall component of Gram-negative bacteria and signals through a receptor complex which consists of TLR4, MD-2 and CD14. LPS signaling in macrophages induces the production of many pro-inflammatory molecules, including nitric oxide (NO). In this study, we have shown that folimycin, a macrolide antibiotic and a specific inhibitor of vacuolar ATPase (V-ATPase), inhibits LPS-induced NO production, but not TNFalpha production, in murine elicited peritoneal macrophages. However, folimycin did not affect interferon-gamma induced NO production. LPS-induced iNOS mRNA and protein expression and NF-kappaB activation were also inhibited by folimycin. Interestingly, folimycin-treated cells showed reduced surface expression of TLR4 molecules and dilated Golgi apparatus. These findings suggest that folimycin, by inhibiting V-ATPases, alters intra-Golgi pH, which in turn causes defective processing and reduced surface expression of TLR4 reducing the strength of LPS signaling in murine macrophages.
Collapse
Affiliation(s)
- Sandeepa M Eswarappa
- Centre for Infectious Disease Research and Biosafety Laboratories, Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | | | | | | |
Collapse
|
18
|
Song M, Pinsky MR, Kellum JA. Heat shock factor 1 inhibits nuclear factor-kappaB nuclear binding activity during endotoxin tolerance and heat shock. J Crit Care 2008; 23:406-15. [PMID: 18725048 DOI: 10.1016/j.jcrc.2007.09.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2007] [Revised: 08/15/2007] [Accepted: 09/24/2007] [Indexed: 11/29/2022]
Abstract
RATIONALE Sepsis, endotoxin tolerance, and heat shock (HS) all display down-regulation of innate immunity. We hypothesize that HS factor 1 (HSF-1) induces competitive inhibition of nuclear factor-kappaB (NF-kappaB)-induced signal transduction in both endotoxin tolerance and HS. OBJECTIVES We compared endotoxin tolerance and HS in RAW 264.7 cells. We transfected cells with an HS protein 70 (HSP70) plasmid to test whether HSP70 is the mediator of HS-induced NF-kappaB inhibition. We studied the effects of endotoxin stimulation and HS, both separately and together, on "wild-type" cells, cells transfected with the HSP70 plasmid, and cells transfected with vehicle. FINDINGS Heat shock protein 70 plasmid-transfected cells had increased HSP70 expression and demonstrated decreased nitric oxide (NO) release and inducible NO synthase messenger RNA expression in response to endotoxin compared with wild-type and empty plasmid-transfected cells. Heat shock completely abolished subsequent NO and inducible NO synthase messenger RNA expression in wild-type cells. Heat shock factor 1 reached maximum expression 60 to 90 minutes after HS. Heat shock protein 70-transfected cells still displayed endotoxin-induced NF-kappaB nuclear binding, whereas endotoxin tolerance, HS, and exposure to HSF-1, but not exposure to an unrelated promoter, inhibited NF-kappaB nuclear binding. CONCLUSIONS Endotoxin tolerance and HS appear to share a common immune suppressive effect, possibly through HSF-1-mediated competitive inhibition of NF-kappaB nuclear binding.
Collapse
Affiliation(s)
- Mingchen Song
- The Mechanisms and Novel Therapies for Resuscitation and Acute Illness Laboratories, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | |
Collapse
|
19
|
Desai VR, Ramkrishnan R, Chintalwar GJ, Sainis KB. G1-4A, an immunomodulatory polysaccharide from Tinospora cordifolia, modulates macrophage responses and protects mice against lipopolysaccharide induced endotoxic shock. Int Immunopharmacol 2007; 7:1375-86. [PMID: 17673153 DOI: 10.1016/j.intimp.2007.06.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2007] [Revised: 06/11/2007] [Accepted: 06/11/2007] [Indexed: 10/23/2022]
Abstract
Pro-inflammatory cytokines are known to be the mediators of endotoxic shock and several immunomodulatory herbs can modulate the expression of these cytokines. Therefore we have investigated the possibility of using an arabinogalactan polysaccharide, G1-4A, from the stem of Tinospora cordifolia, for protection against endotoxin induced sepsis. There was 100% protection against lipopolysaccharide (LPS) induced mortality in mice pretreated with G1-4A. To elucidate the mechanism of action, its effect on macrophages, the primary source of these pro-inflammatory molecules was evaluated. G1-4A was shown to bind to the murine macrophages leading to their activation and reciprocally inhibited binding of LPS to macrophages. Following treatment with G1-4A, there was a small increase in serum TNF-alpha and IL-1beta levels. However, challenge with LPS elicited significantly reduced levels of TNF-alpha in G1-4A pretreated mice as compared to the controls while the level of soluble TNFR was enhanced. An increase in serum IL-1beta, IL-6, IFN-gamma levels and decrease in that of IL-10 was observed following challenge with LPS in mice pretreated with G1-4A as compared to the controls. In addition, G1-4A also modulated the release of nitric oxide by murine macrophages. Similar phenomenon was observed in a human monocytic cell line, U937. Thus G1-4A appeared to induce tolerance against endotoxic shock by modulation of cytokines and nitric oxide.
Collapse
Affiliation(s)
- Veena R Desai
- Radiation Biology and Health Sciences Division, Bio-Medical Group, Mumbai-400 085, India
| | | | | | | |
Collapse
|
20
|
Sinistro A, Ciaprini C, Natoli S, Sussarello E, Carducci FC, Almerighi C, Capozzi M, Bolacchi F, Rocchi G, Bergamini A. Lipopolysaccharide desensitizes monocytes-macrophages to CD40 ligand stimulation. Immunology 2007; 122:362-70. [PMID: 17608691 PMCID: PMC2266011 DOI: 10.1111/j.1365-2567.2007.02648.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Polymicrobial sepsis induces the suppression of macrophage function as determined by a reduction of pro-inflammatory cytokine production upon re-exposure to lipopolysaccharide (LPS) in vitro. Here, we examined whether macrophages were refractory to only LPS or if they were unable to respond to other stimuli such as CD40 ligand (CD40L). Monocytic cells exposed in vitro to LPS showed a dose-dependent reduction of their ability to produce interleukin-12 and tumour necrosis factor-alpha upon subsequent CD40L stimulation, as compared to cells stimulated with CD40L alone. Similarly, LPS interfered with the up-regulation of CD40, CD80 and CD86 induced by CD40L in monocytic cells. The effect of LPS on the response of monocytes to CD40L was similar whether these cells were directly exposed to LPS or cocultured with LPS-pretreated cells, indicating that soluble factors released by LPS stimulation could mediate tolerance to CD40L. We also show that the functional alterations induced by LPS in monocytes can be reversed by indomethacin, thus suggesting a role for inducible cyclooxygenase in mediating the LPS-induced hyporesponsive state of monocytes to CD40L. In conclusion, we propose that in vitro CD40L tolerance may be an appropriate model of monocyte alteration observed during septic immunosuppression and may help in the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Anna Sinistro
- Department of Public Health and Cellular Biology, Chair of Infectious Diseases, University of Rome 'Tor Vergata', Rome, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Gobejishvili L, Barve S, Joshi-Barve S, Uriarte S, Song Z, McClain C. Chronic ethanol-mediated decrease in cAMP primes macrophages to enhanced LPS-inducible NF-kappaB activity and TNF expression: relevance to alcoholic liver disease. Am J Physiol Gastrointest Liver Physiol 2006; 291:G681-8. [PMID: 16751174 DOI: 10.1152/ajpgi.00098.2006] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Increased plasma and hepatic TNF-alpha activity has been implicated in the pathogenesis of alcoholic liver disease (ALD). We previously reported that monocytes from alcoholic patients show enhanced constitutive as well as LPS-inducible NF-kappaB activation and TNF-alpha production. Studies in monocytes have shown that cAMP plays an important role in regulating TNF-alpha expression, and elevation of cellular cAMP suppresses TNF-alpha production. The effects of chronic ethanol exposure on the cellular levels of cAMP as well as TNF expression in monocytes were examined in vitro and in rat primary hepatic Kupffer cells obtained from a clinically relevant enteral alcohol feeding model of ALD. Chronic ethanol exposure significantly decreased cellular cAMP levels in both LPS-stimulated and unstimulated monocytes. Consistent with the decrease in cAMP levels, ethanol led to an increase in LPS-inducible TNF-alpha production by affecting NF-kappaB activation and induction of TNF mRNA expression, without any change in TNF mRNA stability. Enhancement of cellular cAMP with dibutyryl cAMP abrogated LPS-mediated TNF-alpha expression in ethanol-treated cells. Importantly, cAMP did not affect LPS-inducible NF-kappaB activation but significantly decreased its transcriptional activity. Together, these data strongly suggest that ethanol can synergize with LPS to upregulate the induction of TNF gene expression and consequent TNF overproduction by decreasing the cellular cAMP levels in monocytes/macrophages. Furthermore, these data also support the notion that cAMP-elevating agents could constitute an effective therapeutic approach in attenuating or preventing the progression of liver disease in alcoholic patients.
Collapse
Affiliation(s)
- Leila Gobejishvili
- Dept. of Medicine, Pharmacology and Toxicology, Univ. of Louisville Medical Center, 550 S. Jackson St., ACB 3rd Floor, Louisville, KY 40292, USA.
| | | | | | | | | | | |
Collapse
|
22
|
Wolk K, Kunz S, Crompton NEA, Volk HD, Sabat R. Multiple mechanisms of reduced major histocompatibility complex class II expression in endotoxin tolerance. J Biol Chem 2003; 278:18030-6. [PMID: 12637533 DOI: 10.1074/jbc.m207714200] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Patients after polytrauma, burns, or septic shock frequently develop a life-threatening immunodeficiency. This state is associated with specific functional alterations of monocytic cells. We previously proposed endotoxin tolerance, the monocyte state after acute response to lipopolysaccharide, as a respective model system. One major feature in both the clinical situation and the in vitro model is the dramatic down-regulation of monocyte major histocompatibility complex (MHC) class II surface expression, which is associated with impaired antigen presentation capacity. This study focused on the mechanisms behind reduced MHC class II expression in endotoxin tolerance. Endotoxin priming provoked a decrease of monocyte intracellular MHC class II. It also led to a reduced expression of the chaperonic invariant chain and to an inhibited synthesis of the major lysosomal enzyme for final cleavage of the invariant chain going along with a relative accumulation of p10. The expression of HLA-DM necessary for loading MHC class II with antigenic peptide was also decreased. Additionally, reduced export of MHC class II alphabeta complexes to the cell surface was observed. The down-regulation of HLA-DR, invariant chain, and HLA-DM was regulated at the mRNA level and may be the consequence of reduced class II transactivator expression observed in this study. The simultaneous interference at different regulatory levels may explain the uniquely strong and long lasting MHC class II down-modulating effect of endotoxin priming compared with transforming growth factor-beta and interleukin-10. These results not only contribute to a better understanding of experimental endotoxin tolerance but may also give rise to new therapeutics for temporary immunodeficiency and, conversely, for MHC class II-dependent diseases such as autoimmunity and transplant rejection.
Collapse
Affiliation(s)
- Kerstin Wolk
- Institute of Medical Immunology, Medical School Charité, Humboldt University Berlin, D-10117 Berlin, Germany
| | | | | | | | | |
Collapse
|
23
|
Lewis JB, Randol TM, Lockwood PE, Wataha JC. Effect of subtoxic concentrations of metal ions on NFkappaB activation in THP-1 human monocytes. J Biomed Mater Res A 2003; 64:217-24. [PMID: 12522807 DOI: 10.1002/jbm.a.10352] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
THP-1 human monocytes and human peripheral blood monocytes have altered inflammatory cytokine secretion profiles after exposure to a variety of metal ions known to be released from biomaterials. Transcriptional regulation of these cytokines often involves activation of the transcription factor NFkappaB. The present study was designed to determine whether metal ion treatment of monocytes results in changes in levels of activated NFkappaB. THP-1 cells were grown in suspension in the presence of sublethal concentrations of ions of Ag(+), Co(2+), Cu(2+), Hg(2+), Ni(2+), and Pd(2+). After 24 h of exposure to metal ions, the cells were harvested, counted, and the nuclear proteins extracted. Electrophoretic mobility shift assays were performed using a (32)P-ATP end-labeled oligonucleotide consensus sequence for the NFkappaB transcription factor. DNA/protein complexes were quantified by phosphorimage analysis and compared by ANOVA (Tukey, alpha = 0.05). Exposure of THP-1 cells to 100 microM of Pd(2+) caused a significant increase in activated NFkappaB (p < 0.05) whereas treatment with 5 microM of Ag(+) resulted in significantly decreased levels of nuclear NFkappaB (p < 0.05). No other metal ions tested caused a significant change in basal levels of nuclear NFkappaB (Co(2+), Hg(2+), Ni(2+), and Cu(2+)). However, exposure to 50 microM of Cu(2+) resulted in a reproducible, though not significant, increase in nuclear NFkappaB levels. These results indicate that inflammatory responses to some metal ions may be influenced by NFkappaB-mediated transcriptional regulation.
Collapse
Affiliation(s)
- Jill B Lewis
- Oral Biology and Maxillofacial Pathology, Medical College of Georgia School of Dentistry, Augusta, Georgia 30912-1126, USA
| | | | | | | |
Collapse
|
24
|
Lewis JB, Wataha JC, Randol TM, McCloud VV, Lockwood PE. Metal ions alter lipopolysaccharide-induced NFκB binding in monocytes. ACTA ACUST UNITED AC 2003; 67:868-75. [PMID: 14613235 DOI: 10.1002/jbm.a.10169] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Metals are components of a variety of biomaterials used in orthopedic and dental appliances; however, their biocompatibility with the surrounding tissues is not completely understood. Monocytes are important immune cells that respond to inflammatory stimuli by rapidly producing a variety of inflammatory proteins. Regulation of this response often involves activation of the transcription factor NF kappa B. The current study was designed to determine whether monocyte activation of NF kappa B in response to bacterial lipopolysaccharide (LPS) is affected by pretreatment with metal ions. Concentrations of metal ions that affected cell number after 24 h of exposure were first determined. Then THP-1 human monocytes were cultured for 2 h in media containing metal ions at concentrations below levels that altered cell growth. Parallel cultures were treated with 10 microg/mL Escherichia coli LPS, and all samples were cultured an additional 2 h. Nuclear proteins were extracted and normalized amounts were incubated with [(32)P]-end-labeled NF kappa B consensus oligonucleotide. NF kappa B-DNA complexes were identified and quantified by electrophoretic mobility shift analysis. The extent of NF kappa B-DNA complex formation after metal ion pretreatment with or without LPS induction was compared to no treatment or LPS-only treated controls. Finally, LPS-induced IL1 beta secretion was measured from palladium-treated and control cells. Concentrations were identified for each metal ion (Ag(+), Co(2+), Cu(2+), Hg(2+), Ni(2+), and Pd(2+)) that did not reduce cell number after 24 h of exposure (ranging from 5 microM for Ag(+) and Hg(2+) to 200 microM for Ni(2+)). Exposures of 2 h at these concentrations did not alter cell morphology, staining with trypan blue, or cell number. LPS exposure had no effect on cell number with or without metal ions after 2 h. When metal treatment alone was assessed, none of the metal ions had a significant effect on NF kappa B-DNA binding. However, pretreatment with Co(2+), Ni(2+), Ag(1+), Hg(2+), and Pd(2+) significantly decreased NF kappa B-DNA binding by 40-70% versus LPS alone. Only Cu(2+) had no effect on LPS-induced NF kappa B-DNA complex formation. Pd(2+) lowered, but did not abolish, IL1 beta secretion at concentrations comparable to those that altered NF kappa B-DNA binding. These results suggest that many commonly used metals alter monocyte function at concentrations that are not overtly toxic, and that protein levels controlled in part by NF kappa B also may be altered.
Collapse
Affiliation(s)
- J B Lewis
- School of Dentistry, Oral Biology and Maxillofacial Pathology, Medical College of Georgia, Augusta, Georgia 30912, USA.
| | | | | | | | | |
Collapse
|
25
|
Reddy RC, Chen GH, Tekchandani PK, Standiford TJ. Sepsis-induced immunosuppression: from bad to worse. Immunol Res 2002; 24:273-87. [PMID: 11817325 DOI: 10.1385/ir:24:3:273] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The sepsis syndrome is characterized by the acute release of a variety of inflammatory mediators, which often result in detrimental effects to the host. The release of these mediators is regulated and counterbalanced by the coordinated expression of antiinflammatory molecules. It is the balance between the expression of pro- and antiinflammatory mediators that often determines the magnitude of early tissue injury and subsequent risk of infectious complications. As our understanding of the pathophysiology of sepsis continues to evolve, we have gained a greater appreciation for the effects that sepsis and similar states of overwhelming stress have on host antimicrobial immunity. A number of functional defects in leukocytes isolated from sepsis patients have been characterized. These defects include diminished expression of important cell surface antigens, dysregulated cytokine production, alterations in antigen-presenting ability, and accelerated apoptosis. Impaired leukocyte function has important clinical ramifications, as high mortality rates have been observed in patients displaying evidence of sepsis-induced immune deactivation. In this article, we review the current literature supporting evidence of dysregulation of host immunity occurring during sepsis syndrome, characterize the underlying pathophysiology, and describe novel therapeutic interventions directed at augmenting host immunity during sepsis.
Collapse
Affiliation(s)
- R C Reddy
- Department of Medicine, The University of Michigan Medical School, Ann Arbor, USA
| | | | | | | |
Collapse
|
26
|
Woitas RP, Petersen U, Moshage D, Brackmann HH, Matz B, Sauerbruch T, Spengler U. HCV-specific cytokine induction in monocytes of patients with different outcomes of hepatitis C. World J Gastroenterol 2002; 8:562-6. [PMID: 12046093 PMCID: PMC4656444 DOI: 10.3748/wjg.v8.i3.562] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2002] [Revised: 04/13/2002] [Accepted: 04/25/2002] [Indexed: 02/06/2023] Open
Abstract
AIM Cytokine release by macrophages critically determines the type of immune response to an antigen. Therefore, we studied hepatitis C virus (HCV)-specific induction of interleukins-1 beta, -10, -12 (IL-1 beta, IL-10, IL-12), and tumor necrosis factor-alpha (TNF-alpha) in monocytes. METHODS Intracellular cytokine expression was studied by flow cytometry in 23 patients with chronic hepatitis C, 14 anti-HCV seropositives without viremia and 11 controls after stimulation of peripheral blood mononuclear cells with recombinant core, NS3, NS4, NS5a and NS5b proteins. RESULTS Patients with HCV viremia revealed greater spontaneous expression of IL-1beta, TNF-alpha, and IL-10. Furthermore, greater than twofold higher IL-10 expression was induced by the HCV antigens in chronic hepatitis C than in the other two groups (P<0.05). In contrast, neither IL-12 nor TNF-alpha was induced preferentially. CONCLUSION In chronic hepatitis C antigen-specific cytokine induction in monocytes is apparently shifted towards predominant IL-10 induction - not counterbalanced by antiviral type 1 cytokines. This may contribute to persistent viral replication.
Collapse
Affiliation(s)
- Rainer P Woitas
- Department of Internal Medicine I, University of Bonn, D-53105 Bonn, Germany.
| | | | | | | | | | | | | |
Collapse
|
27
|
Lehner MD, Hartung T. Endotoxin tolerance-mechanisms and beneficial effects in bacterial infection. Rev Physiol Biochem Pharmacol 2002; 144:95-141. [PMID: 11987826 DOI: 10.1007/bfb0116586] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- M D Lehner
- Universität Konstanz, Biochemische Pharmakologie, Fach M655, 78457 Konstanz, Germany
| | | |
Collapse
|
28
|
Ekman P, Saarinen M, He Q, Gripenberg-Lerche C, Grönberg A, Arvilommi H, Granfors K. HLA-B27-transfected (Salmonella permissive) and HLA-A2-transfected (Salmonella nonpermissive) human monocytic U937 cells differ in their production of cytokines. Infect Immun 2002; 70:1609-14. [PMID: 11854251 PMCID: PMC127747 DOI: 10.1128/iai.70.3.1609-1614.2002] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The cytokine secretion of the Salmonella-permissive, HLA-B27-positive U937 cells was examined, as it was previously shown that these cells kill Salmonella less efficiently than controls. Salmonella-permissive U937 cells showed upregulated production of interleukin 10 and to a lesser extent tumor necrosis factor alpha. HLA-B27-associated modulation of cytokine responses may have importance in the pathogenesis of reactive arthritis.
Collapse
Affiliation(s)
- Päivi Ekman
- National Public Health Institute, Department in Turku, Turku, Finland
| | | | | | | | | | | | | |
Collapse
|
29
|
Faas MM, Moes H, Fijen JW, Muller Kobold AC, Tulleken JE, Zijlstra JG. Monocyte intracellular cytokine production during human endotoxaemia with or without a second in vitro LPS challenge: effect of RWJ-67657, a p38 MAP-kinase inhibitor, on LPS-hyporesponsiveness. Clin Exp Immunol 2002; 127:337-43. [PMID: 11876759 PMCID: PMC1906333 DOI: 10.1046/j.1365-2249.2002.01765.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
In the present study, we investigated the effect of RWJ-67657, a p38 MAP kinase inhibitor, upon in vivo LPS-induced monocyte cytokine production and upon monocyte LPS-hyporesponsiveness. Thirty minutes before a single injection of LPS (4 ng/kg BW), healthy male volunteers received a single oral dose of RWJ-67657 at increasing dosages (0-1400 mg). Blood samples (pre-medication, 3, 6 and 24 h after LPS) were immediately incubated with LPS (reflecting LPS-hyporesponsiveness) or without LPS (reflecting in vivo monocyte stimulation) for 4 h at 37 degrees C. Following red blood cells lysis and white blood cell permeabilization, cells were labelled with alpha-CD14-FITC and alpha-IL-1beta, alpha-IL-12 or alpha-TNFalpha (PE-labelled), fixed, and analysed using flow cytometry. In vivo LPS injection resulted in an increased percentage of circulating monocytes producing IL-1beta, TNFalpha and IL-12 only at 3 h after the LPS injection. This was dose-dependently inhibited by RWJ-67657 treatment. LPS-hyporesponsiveness to in vitro LPS treatment was most prominent at 3 and 6 h after the in vivo LPS injection; compared with pre-medication monocytes, at these intervals a reduced percentage of monocytes produced IL-1beta, TNFalpha or IL-12 after the in vitro LPS stimulus. At t = 6 h, this LPS-hyporesponsiveness could dose-dependently be inhibited by RWJ-67657 treatment of the volunteers. We therefore conclude that p38 MAP kinase inhibition with RWJ-67657 inhibited monocyte production of cytokines following in vivo LPS injection. Treatment with RWJ-67657 also reversed the LPS-hyporesponsiveness. Whether this result can be extended to the clinical situation remains to be elucidated. Patients with sepsis or an otherwise high risk for multi-organ failure are potential study groups.
Collapse
Affiliation(s)
- M M Faas
- Reproductive Immunology, Medical Biology Branch, Department of Pathology, University of Groningen, The Netherlands.
| | | | | | | | | | | |
Collapse
|
30
|
Exner R, Weingartmann G, Eliasen MM, Gerner C, Spittler A, Roth E, Oehler R. Glutamine deficiency renders human monocytic cells more susceptible to specific apoptosis triggers. Surgery 2002; 131:75-80. [PMID: 11812966 DOI: 10.1067/msy.2002.118318] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND After major trauma and sepsis, patients frequently show a decreased blood glutamine (Gln) level. Gln deprivation has been shown to induce apoptosis in intestinal epithelial cells. In this study, we investigated whether the Gln level also affects the susceptibility of monocytic cells to apoptosis. METHODS Human monocytic U937 cells were suspended in a Gln-free medium, exposed for 20 minutes to either tumor necrosis factor alpha, Fas ligand, heat shock, or UV irradiation and allowed to recover for 4 hours or 24 hours. Apoptosis was measured by annexin-V assay and confirmed by nuclear condensation. The activation of caspase-3 was determined by Western blot. RESULTS When induced by tumor necrosis factor alpha, Fas ligand, or heat shock, the apoptosis rate was significantly lower (50%-60%) in the presence of Gln than in the absence of Gln (P <.02). However, Gln had no effect on UV irradiation-induced apoptosis. Caspase-3 was activated by all inducers and was independent of Gln. CONCLUSIONS This study shows that glutamine deprivation increases the susceptibility of monocytic cells to some but not all inducers of apoptosis. Because Gln has no effect on caspase-3 activation, we hypothesize that the selective anti-apoptotic effect of Gln occurs downstream of caspase-3. These results suggest that Gln serves as a selective immunomodulating factor.
Collapse
Affiliation(s)
- Ruth Exner
- Surgical Research Laboratories, Department of Surgery and Institute of Tumor Biology-Cancer Research, University of Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
31
|
Koutsos EA, Klasing KC. The acute phase response in Japanese quail (Coturnix coturnix japonica). Comp Biochem Physiol C Toxicol Pharmacol 2001; 128:255-63. [PMID: 11239838 DOI: 10.1016/s1532-0456(00)00199-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Experiments were conducted to determine the effect of injection of lipopolysaccharide (LPS, from S. typhimurium) or muramyl dipeptide (MDP, N-acetylmuramyl-L-ala-isoglutamine) in Japanese quail. Doses of MDP between 0.3 and 10 mg/kg body wt. had no effect on body temperature. In contrast, doses of 1.0-22.5 mg LPS/kg body wt. caused significant increases in body temperature. None of the doses of LPS or MDP resulted in mortality. The febrile response to LPS was diminished following a second injection 48 h after the first, and was absent following a third injection. Plasma zinc, an indicator of the acute phase response, was significantly reduced by either LPS or MDP after the first injection (P<0.001), but not after the second or third injection. Splenic interleukin 1-beta (IL-1beta) mRNA expression was increased after the first and last injection of LPS (P<0.001), but only after the first injection of MDP (P<0.005). Hepatic IL-1beta mRNA expression was increased after the first, but not the third injection of LPS (P<0.001), while MDP had no effect. These data indicate that Japanese quail are less sensitive to MDP than LPS, and that quail demonstrate tolerance to LPS following repeated injections.
Collapse
Affiliation(s)
- E A Koutsos
- Department of Animal Science, One Shields Avenue, University of California, Davis, CA 95616, USA
| | | |
Collapse
|
32
|
Genois N, Robichaud GA, Tremblay MJ. Mono Mac 1: a new
in vitro
model system to study HIV‐1 infection in human cells of the mononuclear phagocyte series. J Leukoc Biol 2000. [DOI: 10.1189/jlb.68.6.854] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Nicolas Genois
- Centre de Recherche en Infectiologie, Centre Hospitalier Universitaire de Québec, Pavillon CHUL, and Département de Biologie Médicale, Faculté de Médecine, Université Laval, Ste‐Foy, Québec, Canada
| | - Gilles A Robichaud
- Centre de Recherche en Infectiologie, Centre Hospitalier Universitaire de Québec, Pavillon CHUL, and Département de Biologie Médicale, Faculté de Médecine, Université Laval, Ste‐Foy, Québec, Canada
| | - Michel J. Tremblay
- Centre de Recherche en Infectiologie, Centre Hospitalier Universitaire de Québec, Pavillon CHUL, and Département de Biologie Médicale, Faculté de Médecine, Université Laval, Ste‐Foy, Québec, Canada
| |
Collapse
|
33
|
Meja KK, Seldon PM, Nasuhara Y, Ito K, Barnes PJ, Lindsay MA, Giembycz MA. p38 MAP kinase and MKK-1 co-operate in the generation of GM-CSF from LPS-stimulated human monocytes by an NF-kappa B-independent mechanism. Br J Pharmacol 2000; 131:1143-53. [PMID: 11082122 PMCID: PMC1572440 DOI: 10.1038/sj.bjp.0703684] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
1. The extent to which the p38 mitogen-activated protein (MAP) kinase and MAP kinase kinase (MKK)-1-signalling pathways regulate the expression of granulocyte/macrophage colony-stimulating factor (GM-CSF) from LPS-stimulated human monocytes has been investigated and compared to the well studied cytokine tumour necrosis factor-alpha (TNF alpha). 2. Lipopolysaccharide (LPS) evoked a concentration-dependent generation of GM-CSF from human monocytes. Temporally, this effect was preceded by an increase in GM-CSF mRNA transcripts and abolished by actinomycin D and cycloheximide. 3. LPS-induced GM-CSF release and mRNA expression were associated with a rapid and time-dependent activation of p38 MAP kinase, ERK-1 and ERK-2. 4. The respective MKK-1 and p38 MAP kinase inhibitors, PD 098059 and SB 203580, maximally suppressed LPS-induced GM-CSF generation by >90%, indicating that both of these signalling cascades co-operate in the generation of this cytokine. 5. Electrophoretic mobility shift assays demonstrated that LPS increased nuclear factor kappa B (NF-kappa B) : DNA binding. SN50, an inhibitor of NF-kappa B translocation, abolished LPS-induced NF-kappaB : DNA binding and the elaboration of TNFalpha, a cytokine known to be regulated by NF-kappaB in monocytes. In contrast, SN50 failed to affect the release of GM-CSF from the same monocyte cultures. 6. Collectively, these results suggest that the generation of GM-CSF by LPS-stimulated human monocytes is regulated in a co-operative fashion by p38 MAP kinase- and MKK-1-dependent signalling pathways independently of the activation of NF-kappa B.
Collapse
Affiliation(s)
- Koremu K Meja
- Thoracic Medicine, National Heart and Lung Institute, Imperial College School of Medicine, Dovehouse Street, SW3 6LY
| | - Paul M Seldon
- Thoracic Medicine, National Heart and Lung Institute, Imperial College School of Medicine, Dovehouse Street, SW3 6LY
| | - Yasuyuki Nasuhara
- Thoracic Medicine, National Heart and Lung Institute, Imperial College School of Medicine, Dovehouse Street, SW3 6LY
| | - Kazuhiro Ito
- Thoracic Medicine, National Heart and Lung Institute, Imperial College School of Medicine, Dovehouse Street, SW3 6LY
| | - Peter J Barnes
- Thoracic Medicine, National Heart and Lung Institute, Imperial College School of Medicine, Dovehouse Street, SW3 6LY
| | - Mark A Lindsay
- Thoracic Medicine, National Heart and Lung Institute, Imperial College School of Medicine, Dovehouse Street, SW3 6LY
| | - Mark A Giembycz
- Thoracic Medicine, National Heart and Lung Institute, Imperial College School of Medicine, Dovehouse Street, SW3 6LY
- Author for correspondence:
| |
Collapse
|
34
|
Fujihara M, Wakamoto S, Ito T, Muroi M, Suzuki T, Ikeda H, Ikebuchi K. Lipopolysaccharide‐triggered desensitization of TNF‐α mRNA expression involves lack of phosphorylation of IκBα in a murine macrophage‐like cell line, P388D1. J Leukoc Biol 2000. [DOI: 10.1189/jlb.68.2.267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
| | - Shinobu Wakamoto
- Japanese Red Cross, Hokkaido Red Cross Blood Center, Sapporo, Japan
| | - Takatoshi Ito
- Japanese Red Cross, Hokkaido Red Cross Blood Center, Sapporo, Japan
| | - Masashi Muroi
- Division of Microbiology, National Institutes of Health Sciences, Tokyo, Japan; and
| | - Tsuneo Suzuki
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City
| | - Hisami Ikeda
- Japanese Red Cross, Hokkaido Red Cross Blood Center, Sapporo, Japan
| | - Kenji Ikebuchi
- Japanese Red Cross, Hokkaido Red Cross Blood Center, Sapporo, Japan
| |
Collapse
|
35
|
Abstract
Abstract
Endotoxin tolerance (ET) has been described as a temporary alteration in the lipopolysaccharide (LPS) response of monocytic cells after an initial LPS exposure with respect to the production of soluble immunomodulators. Apart from the LPS response, monocytic cells play an important role in initiation of the specific immune response as antigen-presenting cells. This study investigated the capacity of human blood monocytes to induce T-cell stimulation in ET. First, the expression of monocyte surface molecules, important for T-cell interaction, was analyzed by flow cytometry. In vitro priming of peripheral blood mononuclear cells with LPS clearly down-regulates major histocompatibility complex class II molecules and the costimulatory molecule CD86. Both changes were dependent on the endogenous interleukin (IL)-10 and less so on the transforming growth factor-β. In contrast, other accessory molecules on monocytes were only marginally down-regulated (CD58), were not significantly changed during ET (CD40), or even remained up-regulated after initial LPS priming (CD54, CD80). Second, an impact of these phenotypic alterations on the accessory function of monocytes was observed. This was manifested as diminished T-cell proliferation and interferon (IFN)-γ release in response to the presence of different recall antigens. Neutralizing IL-10 during LPS priming prevented the diminished T-cell IFN-γ production but had little effect on T-cell proliferation. These data confirm that ET is an appropriate model of the monocyte functional state in immunoparalysis, which is frequently observed in patients after septic shock, trauma, or major surgery.
Collapse
|
36
|
Abstract
Endotoxin tolerance (ET) has been described as a temporary alteration in the lipopolysaccharide (LPS) response of monocytic cells after an initial LPS exposure with respect to the production of soluble immunomodulators. Apart from the LPS response, monocytic cells play an important role in initiation of the specific immune response as antigen-presenting cells. This study investigated the capacity of human blood monocytes to induce T-cell stimulation in ET. First, the expression of monocyte surface molecules, important for T-cell interaction, was analyzed by flow cytometry. In vitro priming of peripheral blood mononuclear cells with LPS clearly down-regulates major histocompatibility complex class II molecules and the costimulatory molecule CD86. Both changes were dependent on the endogenous interleukin (IL)-10 and less so on the transforming growth factor-β. In contrast, other accessory molecules on monocytes were only marginally down-regulated (CD58), were not significantly changed during ET (CD40), or even remained up-regulated after initial LPS priming (CD54, CD80). Second, an impact of these phenotypic alterations on the accessory function of monocytes was observed. This was manifested as diminished T-cell proliferation and interferon (IFN)-γ release in response to the presence of different recall antigens. Neutralizing IL-10 during LPS priming prevented the diminished T-cell IFN-γ production but had little effect on T-cell proliferation. These data confirm that ET is an appropriate model of the monocyte functional state in immunoparalysis, which is frequently observed in patients after septic shock, trauma, or major surgery.
Collapse
|
37
|
Lush CW, Cepinskas G, Kvietys PR. LPS tolerance in human endothelial cells: reduced PMN adhesion, E-selectin expression, and NF-kappaB mobilization. Am J Physiol Heart Circ Physiol 2000; 278:H853-61. [PMID: 10710354 DOI: 10.1152/ajpheart.2000.278.3.h853] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cytokine release from inflammatory (CD14(+)) cells is reduced after repeated stimulation with lipopolysaccharide (LPS; LPS tolerance). However, it is not known whether LPS tolerance can be induced in CD14(-) cells. The aim of the present study was to determine whether endothelial cells [human umbilical vein endothelial cells (HUVEC)] could be rendered tolerant to LPS with respect to LPS-induced polymorphonuclear neutrophil (PMN) adhesion. LPS stimulation (0.5 microg/ml; 4 h) of naive HUVEC increased PMN adhesion. Pretreatment of HUVEC with LPS (0.5 microg/ml) for 24 h resulted in a reduction in the proadhesive effects of a subsequent LPS challenge. The initial LPS stimulation increased 1) mobilization of the nuclear transcription factor NF-kappaB to the nucleus and 2) surface levels of the adhesion molecules intercellular adhesion molecule-1 (ICAM-1) and E-selectin. In LPS-tolerant HUVEC, a second LPS challenge resulted in 1) less accumulation of NF-kappaB in the nucleus, 2) a reduction in E-selectin expression, and 3) unchanged ICAM-1 expression. LPS-tolerant cells were still capable of mobilizing NF-kappaB in response to stimulation with either interleukin-1beta or tumor necrosis factor-alpha, resulting in elevated E-selectin levels and increased PMN adhesion. These studies show for the first time that LPS tolerance can be induced in endothelial cells with respect to PMN adhesion. This tolerance is specific for LPS and is associated with an inability of LPS to mobilize NF-kappaB, resulting in less E-selectin expression.
Collapse
Affiliation(s)
- C W Lush
- Department of Physiology, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | | | | |
Collapse
|
38
|
Ammon C, Mondal K, Andreesen R, Krause SW. Differential expression of the transcription factor NF-kappaB during human mononuclear phagocyte differentiation to macrophages and dendritic cells. Biochem Biophys Res Commun 2000; 268:99-105. [PMID: 10652220 DOI: 10.1006/bbrc.1999.2083] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
An important role for the Rel/NF-kappaB family of transcription factors in the differentiation process of dendritic cells (DC) and macrophages (MAC) was recently suggested by a number of mouse knockout studies but only little information is available for defined populations of human cells. To investigate the role of individual NF-kappaB proteins [p50, p52, p65 (RelA), RelB] in the differentiation of monocyte-derived cell types we analyzed and compared the expression pattern and DNA binding activity of NF-kappaB members in human monocytes (MO), MO-derived MAC, and MO-derived DC. Constitutive expression of p65 and RelB mRNA was found in MO and no significant regulation was observed during differentiation of MO into MAC or immature DC. Only during lipopolysaccharide-induced terminal differentiation of DC was a marked increase in RelB mRNA detected. In DNA binding assays performed with nuclear extracts from blood MO, p50/p50 homodimers were mainly detected, whereas complexes containing p50/RelB and p50/p65 heterodimers were less abundant. DNA-bound protein complexes containing p50/RelB and p50/p65 increased and additional p65/p65 complexes appeared during differentiation of MO into either MAC or immature DC. A strong increase in complexes containing p50/RelB was observed during terminal differentiation of DC. Therefore, gradual differences in the DNA binding activities of different NF-kappaB homo- and heterodimers correlate with differentiation stages of MO, MAC, and DC and are probably important for the biological role of these cells.
Collapse
Affiliation(s)
- C Ammon
- Department of Hematology, University of Regensburg, Regensburg, D-93042, Germany
| | | | | | | |
Collapse
|
39
|
Ciacci-Woolwine F, McDermott PF, Mizel SB. Induction of cytokine synthesis by flagella from gram-negative bacteria may be dependent on the activation or differentiation state of human monocytes. Infect Immun 1999; 67:5176-85. [PMID: 10496893 PMCID: PMC96868 DOI: 10.1128/iai.67.10.5176-5185.1999] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We have previously demonstrated that salmonellae, but not Escherichia coli or Yersinia enterocolitica, stimulates tumor necrosis factor alpha (TNFalpha) production in the human promonocytic cell line U38. Subsequent analysis revealed that the TNFalpha-inducing activity of salmonellae was associated with flagellin, a major component of flagella from gram-negative bacteria. In the present study, we have explored the basis for the apparent specificity of action of Salmonella flagella on TNFalpha expression in U38 cells and have extended this analysis to normal human peripheral blood mononuclear cells (PBMC). Flagella from the enteropathogenic E. coli strain E2348/69, Y. enterocolitica JB580, and Pseudomonas aeruginosa PAO1, which did not induce significant levels of TNFalpha production in U38 cells, were as potent as Salmonella flagella in terms of TNFalpha and interleukin 1beta activation in PBMC. However, TNFalpha production in U38 cells was greatly enhanced when these cells were stimulated with flagella from E. coli, Y. enterocolitica, and P. aeruginosa in the presence of a costimulant, phorbol 13-myristate acetate. These findings are consistent with the hypothesis that the activation or differentiation state of a monocyte may have a substantial effect on the cell's responsiveness to flagellum stimulation of cytokine synthesis. Furthermore, these results indicate that cytokine induction in monocytes may be a general property of flagella from gram-negative bacteria.
Collapse
Affiliation(s)
- F Ciacci-Woolwine
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | | | | |
Collapse
|
40
|
Kastenbauer S, Ziegler-Heitbrock HW. NF-kappaB1 (p50) is upregulated in lipopolysaccharide tolerance and can block tumor necrosis factor gene expression. Infect Immun 1999; 67:1553-9. [PMID: 10084986 PMCID: PMC96496 DOI: 10.1128/iai.67.4.1553-1559.1999] [Citation(s) in RCA: 129] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Monocytes respond to lipopolysaccharide (LPS) stimulation with a rapid expression of the tumor necrosis factor (TNF) gene. Upon repeated LPS stimulation there is, however, little production of TNF mRNA and protein; i.e., the cells are tolerant to LPS. Analysis of NF-kappaB proteins in gel shift assays demonstrated that the DNA binding activity that is induced by LPS stimulation in tolerant cells consists mainly of p50-p50 homodimers. Since p50 can bind to DNA but lacks a transactivation domain, this may explain the blockade of TNF gene expression. We now show that in the monocytic cell line Mono Mac 6, this inability to respond can be largely ascribed to NF-kappaB, since a reporter construct directed by a trimeric NF-kappaB motif is strongly transactivated by LPS stimulation of naive cells whereas LPS-tolerant cells exhibit only low activity. Also, Western blot analyses of proteins extracted from purified nuclei showed mobilization of threefold-higher levels of p50 protein in tolerant compared to naive cells, while mobilization of p65 was unaltered. Overexpression of p50 in HEK 293 cells resulted in a strong reduction of p65-driven TNF promoter activity at the levels of both luciferase mRNA and protein. These data support the concept that an upregulation of p50 is instrumental in LPS tolerance in human monocytes.
Collapse
Affiliation(s)
- S Kastenbauer
- Institute for Immunology, University of Munich, 80336 Munich, Germany
| | | |
Collapse
|
41
|
Burger M, Almanasreh N, Bauermeister K, Knopf HP, Schollmeyer P, Dobos GJ. In vitro differentiation and characterization of human peritoneal macrophages from CAPD-peritonitis patients. Immunobiology 1999; 200:62-76. [PMID: 10084696 DOI: 10.1016/s0171-2985(99)80033-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Studies on human macrophages are restricted due to difficulties in isolating significant numbers of human macrophages. High numbers of monocytes/macrophages can be obtained from peritonitis effluents of patients treated with peritoneal dialysis. To determine whether these cells might be useful for functional studies, we characterized peritoneal macrophages (PM) immediately after isolation from the dialysate effluents and their subsequent differentiation. During a 10 days culture period they differentiated morphologically and phenotypically (FACS-analysis) from monocyte-like cells to macrophages. Reflecting the intraperitoneal inflammation we found protein- and mRNA-synthesis of IL-8 and monocyte-chemoattractant-protein-1 (MCP-1) to be upregulated in PM after isolation from the effluents. In contrast, TNF-alpha was downregulated and could not be stimulated by LPS and/or IFN-gamma, reflecting the phenomenon of desensitization. After 10 days in culture, cytokine production normalized to a constitutive level and the TNF-alpha responsiveness to LPS was restored. These data suggest the recovery of PM from the inflammatory prestimulation. Therefore PM harvested from peritoneal dialysis effluents might provide a useful tool for further studies on the role of human macrophages in inflammation.
Collapse
Affiliation(s)
- M Burger
- Department of Nephrology, University Hospital Freiburg, Germany
| | | | | | | | | | | |
Collapse
|
42
|
Steinhauser ML, Hogaboam CM, Kunkel SL, Lukacs NW, Strieter RM, Standiford TJ. IL-10 Is a Major Mediator of Sepsis-Induced Impairment in Lung Antibacterial Host Defense. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.1.392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
To explore the mechanism of immunosuppression associated with sepsis, we developed a murine model of sepsis-induced Pseudomonas aeruginosa pneumonia. CD-1 mice underwent either cecal ligation and 26-gauge needle puncture (CLP) or sham surgery, followed by the intratracheal (i.t.) administration of P. aeruginosa or saline. Survival in mice undergoing CLP followed 24 h later by the i.t. administration of saline or P. aeruginosa was 58% and 10%, respectively, whereas 95% of animals undergoing sham surgery followed by P. aeruginosa administration survived. Increased mortality in the CLP/P. aeruginosa group was attributable to markedly impaired lung bacterial clearance and the early development of P. aeruginosa bacteremia. The i.t. administration of bacteria to CLP-, but not sham-, operated mice resulted in an impressive intrapulmonary accumulation of neutrophils. Furthermore, P. aeruginosa challenge in septic mice resulted in a relative shift toward enhanced lung IL-10 production concomitant with a trend toward decreased IL-12. The i.p., but not i.t., administration of IL-10 Abs given just before P. aeruginosa challenge in septic mice significantly improved both survival and clearance of bacteria from the lungs of septic animals administered P. aeruginosa. Finally, alveolar macrophages isolated from animals undergoing CLP displayed a marked impairment in the ability to ingest and kill P. aeruginosa ex vivo, and this defect was partially reversed by the in vivo neutralization of IL-10. Collectively, these observations indicate that the septic response substantially impairs lung innate immunity to P. aeruginosa, and this effect is mediated primarily by endogenously produced IL-10.
Collapse
Affiliation(s)
- Matthew L. Steinhauser
- †Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109-0360
| | - Cory M. Hogaboam
- †Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109-0360
| | - Steven L. Kunkel
- †Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109-0360
| | - Nicholas W. Lukacs
- †Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109-0360
| | - Robert M. Strieter
- *Divisions of Pulmonary and Critical Care Medicine, Department of Medicine, and
| | | |
Collapse
|
43
|
Febrile tolerance develops in response to repeated administration of bacterial lipopolysaccharide but not of interleukin-1β in rats. J Therm Biol 1998. [DOI: 10.1016/s0306-4565(98)00030-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
44
|
Fujihara M, Ikebuchi K, Maekawa TL, Wakamoto S, Ogiso C, Ito T, Takahashi TA, Suzuki T, Sekiguchi S. Lipopolysaccharide-Induced Desensitization of junB Gene Expression in a Mouse Macrophage-Like Cell Line, P388D1. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.7.3659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Abstract
Treatment of a mouse macrophage cell line, P388D1, for 1 h with bacterial LPS caused a transient increase in the level of junB mRNA expression. These cells became refractory in terms of the junB gene response to exposure to a second round of LPS or lipid A, but not to PMA. The LPS-induced desensitized state was not due to the shortening of the half-life of junB mRNA, but was suggested, by nuclear run-on analysis, to be caused by reduction of junB gene transcription. Pretreating cells with herbimycin A, a tyrosine kinase inhibitor, substantially inhibited LPS-induced expression of junB mRNA and decreased tyrosine phosphorylation of 38- to 42-kDa proteins, which comigrated with p38 and p42 mitogen-activated protein (MAP) kinases. Parallel to down-regulation of junB mRNA expression, activation of the p38 MAP kinase was markedly reduced in LPS-tolerant cells, whereas activation of p42 MAP kinase was relatively constant. The specific p38 MAP kinase inhibitor, SB202190, potently inhibited LPS-induced junB mRNA expression. These results suggest that the LPS-induced desensitization of junB gene expression occurs at or upstream of the level of gene transcription and may be involved in a defective LPS-induced p38 MAP kinase pathway.
Collapse
Affiliation(s)
- Mitsuhiro Fujihara
- *Japanese Red Cross, Hokkaido Red Cross Blood Center, Yamanote, Nishi-ku, Sapporo, Japan; and
| | - Kenji Ikebuchi
- *Japanese Red Cross, Hokkaido Red Cross Blood Center, Yamanote, Nishi-ku, Sapporo, Japan; and
| | | | - Shinobu Wakamoto
- *Japanese Red Cross, Hokkaido Red Cross Blood Center, Yamanote, Nishi-ku, Sapporo, Japan; and
| | - Chikayo Ogiso
- *Japanese Red Cross, Hokkaido Red Cross Blood Center, Yamanote, Nishi-ku, Sapporo, Japan; and
| | - Takatoshi Ito
- *Japanese Red Cross, Hokkaido Red Cross Blood Center, Yamanote, Nishi-ku, Sapporo, Japan; and
| | | | - Tsuneo Suzuki
- †Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Sadayoshi Sekiguchi
- *Japanese Red Cross, Hokkaido Red Cross Blood Center, Yamanote, Nishi-ku, Sapporo, Japan; and
| |
Collapse
|
45
|
Abstract
In humans or experimental animals, the repeated confrontation with lipopolysaccharides (LPS) from gram-negative bacteria, but not with muramyl dipeptide (MDP) from gram-positive bacteria, leads to attenuation of almost all pathophysiologic effects mediated by proinflammatory cytokines. Our experiments in guinea pigs and rats demonstrate that attenuation of the febrile response during the development of LPS tolerance is associated with a reduced production of cytokines rather than a decrease in responsiveness to cytokines. Cross-tolerance experiments demonstrate that different stimuli influencing LPS-induced tumor necrosis factor (TNF) release and nitric oxide (NO) synthesis can modify the development of tolerance. On the other hand, the lack of cross-tolerance between LPS and MDP indicates that MDP can activate the cytokine cascade and induce the febrile response in animals tolerant to LPS. This may indicate distinct receptors and signal pathways for LPS and MDP, leading to activation of the cytokine cascade. LPS tolerance has also been demonstrated in ex vivo and in vitro studies. In cultures of monocytes, diminished synthesis of TNF and NO reported after LPS restimulation could be prevented and reversed by interferon and granulocyte-macrophage colony-stimulating factor. These findings add an additional hypothesis in tolerance development.
Collapse
Affiliation(s)
- Eugen Zeisberger
- Physiologisches Institut, Klinikum der Justus Liebig Universität Giessen, Germany
| | - Joachim Roth
- Physiologisches Institut, Klinikum der Justus Liebig Universität Giessen, Germany
| |
Collapse
|
46
|
Shames BD, Meldrum DR, Selzman CH, Pulido EJ, Cain BS, Banerjee A, Harken AH, Meng X. Increased levels of myocardial IkappaB-alpha protein promote tolerance to endotoxin. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 275:H1084-91. [PMID: 9724317 DOI: 10.1152/ajpheart.1998.275.3.h1084] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Endotoxin [lipopolysaccharide (LPS)] causes tumor necrosis factor-alpha (TNF-alpha)-mediated myocardial contractile depression. Tolerance to the cardiac toxicity of LPS can be induced by a prior exposure to LPS or by pretreatment with glucocorticoids. The mechanisms by which the myocardium acquires tolerance to LPS remain unknown. LPS causes phosphorylation and degradation of inhibitory kappaB-alpha (IkappaB-alpha), releasing nuclear factor-kappaB (NF-kappaB) to activate TNF-alpha gene transcription. We hypothesized that LPS induces supranormal synthesis of myocardial IkappaB-alpha protein and thus renders the myocardium tolerant to subsequent LPS. Rats were challenged with LPS after pretreatment with LPS, dexamethasone, or saline. In saline-pretreated rats, LPS caused a rapid decrease in myocardial IkappaB-alpha protein levels, activation of NF-kappaB, and increased TNF-alpha production. These events were followed by myocardial contractile depression. After the initial decrease in myocardial IkappaB-alpha, IkappaB-alpha protein levels rebounded to a level greater than control levels by 24 h. Dexamethasone pretreatment similarly increased myocardial IkappaB-alpha protein levels. In rats pretreated with either LPS or dexamethasone, myocardial IkappaB-alpha protein levels remained similar to control levels after LPS challenge. The preserved level of myocardial IkappaB-alpha protein was associated with diminished NF-kappaB activation, attenuated myocardial TNF-alpha production, and improved cardiac contractility. We conclude that LPS and dexamethasone upregulate myocardial IkappaB-alpha protein expression and that an increased level of myocardial IkappaB-alpha protein may promote cardiac tolerance to LPS by inhibition of NF-kappaB intranuclear translocation and myocardial TNF-alpha production.
Collapse
Affiliation(s)
- B D Shames
- Department of Surgery, University of Colorado Health Sciences Center, Denver, Colorado 80262, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Neumeister B, Faigle M, Sommer M, Zähringer U, Stelter F, Menzel R, Schütt C, Northoff H. Low endotoxic potential of Legionella pneumophila lipopolysaccharide due to failure of interaction with the monocyte lipopolysaccharide receptor CD14. Infect Immun 1998; 66:4151-7. [PMID: 9712761 PMCID: PMC108499 DOI: 10.1128/iai.66.9.4151-4157.1998] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Legionella pneumophila, a gram-negative bacterium causing Legionnaires' disease and Pontiac fever, was shown to be highly reactive in in vitro gelation of Limulus lysate but not able to induce fever and the local Shwartzman reaction in rabbits and mice. We analyzed the capacity of purified L. pneumophila lipopolysaccharide (LPS-Lp) to induce activation of the human monocytic cell line Mono Mac 6, as revealed by secretion of proinflammatory cytokines and desensitization to subsequent LPS stimulation. We showed that despite normal reactivity of LPS-Lp in the Limulus amoebocyte lysate assay, induction of cytokine secretion in Mono Mac 6 cells and desensitization to an endotoxin challenge required LPS-Lp concentrations 1,000 times higher than for LPS of Salmonella enterica serovar Minnesota. Therefore, we examined the interaction of LPS-Lp with the LPS receptor CD14. We demonstrated that LPS-Lp did not bind to membrane-bound CD14 expressed on transfected CHO cells, nor did it react with soluble CD14. Our results suggest that the low endotoxic potential of LPS-Lp is due to a failure of interaction with the LPS receptor CD14.
Collapse
Affiliation(s)
- B Neumeister
- Abteilung Transfusionsmedizin der Universität Tübingen, AG Infektionsimmunologie, D-72076 Tübingen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Udalova IA, Knight JC, Vidal V, Nedospasov SA, Kwiatkowski D. Complex NF-kappaB interactions at the distal tumor necrosis factor promoter region in human monocytes. J Biol Chem 1998; 273:21178-86. [PMID: 9694874 DOI: 10.1074/jbc.273.33.21178] [Citation(s) in RCA: 106] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We describe a dense cluster of DNA-protein interactions located 600 nucleotides upstream of the transcriptional start site of the human tumor necrosis factor (TNF) gene. This area was identified as being of potential importance for lipopolysaccharide-inducible TNF expression in the human monocyte cell line Mono Mac 6, based on reporter gene analysis of point mutations at a number of nuclear factor kappaB (NF-kappaB)-like motifs within the human TNF promoter region. The area contains two NF-kappaB sites, which are here shown by DNase I and methylation interference footprinting to flank a novel binding site. UV cross-linking studies reveal that the novel site can also bind NF-kappaB as well as an unknown protein(s) of approximately 40 kDa. We show that these three adjacent kappaB-binding sites differ markedly in their relative affinities for p50/p50, p65/p65, and p65/p50, yet this 39-nucleotide segment of DNA appears capable of binding up to three NF-kappaB heterodimers simultaneously. Reporter gene studies indicate that each element of the cluster contributes to lipopolysaccharide-induced transcriptional activation in Mono Mac 6 cells. These findings suggest that NF-kappaB acts in a complex manner to activate TNF transcription in human monocytes.
Collapse
Affiliation(s)
- I A Udalova
- Oxford University Department of Paediatrics, Institute of Molecular Medicine, John Radcliffe Hospital, Oxford OX3 9DS, United Kingdom.
| | | | | | | | | |
Collapse
|
49
|
Affiliation(s)
- J A Cook
- Medical University of South Carolina, Department of Physiology, Charleston 29425, USA
| |
Collapse
|
50
|
Sellati TJ, Bouis DA, Kitchens RL, Darveau RP, Pugin J, Ulevitch RJ, Gangloff SC, Goyert SM, Norgard MV, Radolf JD. Treponema pallidum and Borrelia burgdorferi Lipoproteins and Synthetic Lipopeptides Activate Monocytic Cells via a CD14-Dependent Pathway Distinct from That Used by Lipopolysaccharide. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.160.11.5455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
Lipoproteins of Treponema pallidum and Borrelia burgdorferi possess potent proinflammatory properties and, thus, have been implicated as major proinflammatory agonists in syphilis and Lyme disease. Here we used purified B. burgdorferi outer surface protein A (OspA) and synthetic lipopeptides corresponding to the N-termini of OspA and the 47-kDa major lipoprotein immunogen of T. pallidum to clarify the contribution of CD14 to monocytic cell activation by spirochetal lipoproteins and lipopeptides. As with LPS, mouse anti-human CD14 Abs blocked the activation of 1,25-dihydroxyvitamin D3-matured human myelomonocytic THP-1 cells by OspA and the two lipopeptides. The existence of a CD14-dependent pathway was corroborated by using undifferentiated THP-1 cells transfected with CD14 and peritoneal macrophages from CD14-deficient BALB/c mice. Unlike LPS, cell activation by lipoproteins and lipopeptides was serum independent and was not augmented by exogenous LPS-binding protein. Two observations constituted evidence that LPS and lipoprotein/lipopeptide signaling proceed via distinct transducing elements downstream of CD14: 1) CHO cells transfected with CD14 were exquisitely sensitive to LPS but were lipoprotein/lipopeptide nonresponsive; and 2) substoichiometric amounts of deacylated LPS that block LPS signaling at a site distal to CD14 failed to antagonize activation by lipoproteins and lipopeptides. The combined results demonstrate that spirochetal lipoproteins and lipopeptides use a CD14-dependent pathway that differs in at least two fundamental respects from the well-characterized LPS recognition pathway.
Collapse
Affiliation(s)
| | | | | | - Richard P. Darveau
- ‡Bristol-Myers Squibb Pharmaceutical Research Institute, Seattle, WA 98121
| | - Jerome Pugin
- §Medical Intensive Care Unit, University of Geneva, Geneva, Switzerland
| | - Richard J. Ulevitch
- ¶Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037; and
| | - Sophie C. Gangloff
- ∥Department of Molecular Medicine, North Shore University Hospital/Cornell University Medical College, Manhasset, NY 11030
| | - Sanna M. Goyert
- ∥Department of Molecular Medicine, North Shore University Hospital/Cornell University Medical College, Manhasset, NY 11030
| | - Michael V. Norgard
- †Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75235
| | - Justin D. Radolf
- *Internal Medicine and
- †Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75235
| |
Collapse
|