1
|
Fu YL, Harrison RE. Microbial Phagocytic Receptors and Their Potential Involvement in Cytokine Induction in Macrophages. Front Immunol 2021; 12:662063. [PMID: 33995386 PMCID: PMC8117099 DOI: 10.3389/fimmu.2021.662063] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/30/2021] [Indexed: 11/13/2022] Open
Abstract
Phagocytosis is an essential process for the uptake of large (>0.5 µm) particulate matter including microbes and dying cells. Specialized cells in the body perform phagocytosis which is enabled by cell surface receptors that recognize and bind target cells. Professional phagocytes play a prominent role in innate immunity and include macrophages, neutrophils and dendritic cells. These cells display a repertoire of phagocytic receptors that engage the target cells directly, or indirectly via opsonins, to mediate binding and internalization of the target into a phagosome. Phagosome maturation then proceeds to cause destruction and recycling of the phagosome contents. Key subsequent events include antigen presentation and cytokine production to alert and recruit cells involved in the adaptive immune response. Bridging the innate and adaptive immunity, macrophages secrete a broad selection of inflammatory mediators to orchestrate the type and magnitude of an inflammatory response. This review will focus on cytokines produced by NF-κB signaling which is activated by extracellular ligands and serves a master regulator of the inflammatory response to microbes. Macrophages secrete pro-inflammatory cytokines including TNFα, IL1β, IL6, IL8 and IL12 which together increases vascular permeability and promotes recruitment of other immune cells. The major anti-inflammatory cytokines produced by macrophages include IL10 and TGFβ which act to suppress inflammatory gene expression in macrophages and other immune cells. Typically, macrophage cytokines are synthesized, trafficked intracellularly and released in response to activation of pattern recognition receptors (PRRs) or inflammasomes. Direct evidence linking the event of phagocytosis to cytokine production in macrophages is lacking. This review will focus on cytokine output after engagement of macrophage phagocytic receptors by particulate microbial targets. Microbial receptors include the PRRs: Toll-like receptors (TLRs), scavenger receptors (SRs), C-type lectin and the opsonic receptors. Our current understanding of how macrophage receptor stimulation impacts cytokine production is largely based on work utilizing soluble ligands that are destined for endocytosis. We will instead focus this review on research examining receptor ligation during uptake of particulate microbes and how this complex internalization process may influence inflammatory cytokine production in macrophages.
Collapse
Affiliation(s)
- Yan Lin Fu
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, Canada
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, ON, Canada
| | - Rene E. Harrison
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, Canada
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, ON, Canada
| |
Collapse
|
2
|
Checa J, Aran JM. Airway Redox Homeostasis and Inflammation Gone Awry: From Molecular Pathogenesis to Emerging Therapeutics in Respiratory Pathology. Int J Mol Sci 2020; 21:E9317. [PMID: 33297418 PMCID: PMC7731288 DOI: 10.3390/ijms21239317] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 12/05/2020] [Indexed: 02/06/2023] Open
Abstract
As aerobic organisms, we are continuously and throughout our lifetime subjected to an oxidizing atmosphere and, most often, to environmental threats. The lung is the internal organ most highly exposed to this milieu. Therefore, it has evolved to confront both oxidative stress induced by reactive oxygen species (ROS) and a variety of pollutants, pathogens, and allergens that promote inflammation and can harm the airways to different degrees. Indeed, an excess of ROS, generated intrinsically or from external sources, can imprint direct damage to key structural cell components (nucleic acids, sugars, lipids, and proteins) and indirectly perturb ROS-mediated signaling in lung epithelia, impairing its homeostasis. These early events complemented with efficient recognition of pathogen- or damage-associated recognition patterns by the airway resident cells alert the immune system, which mounts an inflammatory response to remove the hazards, including collateral dead cells and cellular debris, in an attempt to return to homeostatic conditions. Thus, any major or chronic dysregulation of the redox balance, the air-liquid interface, or defects in epithelial proteins impairing mucociliary clearance or other defense systems may lead to airway damage. Here, we review our understanding of the key role of oxidative stress and inflammation in respiratory pathology, and extensively report current and future trends in antioxidant and anti-inflammatory treatments focusing on the following major acute and chronic lung diseases: acute lung injury/respiratory distress syndrome, asthma, chronic obstructive pulmonary disease, pulmonary fibrosis, and cystic fibrosis.
Collapse
Affiliation(s)
| | - Josep M. Aran
- Immune-Inflammatory Processes and Gene Therapeutics Group, IDIBELL, L’Hospitalet de Llobregat, 08908 Barcelona, Spain;
| |
Collapse
|
3
|
Shaw TD, McAuley DF, O’Kane CM. Emerging drugs for treating the acute respiratory distress syndrome. Expert Opin Emerg Drugs 2019; 24:29-41. [DOI: 10.1080/14728214.2019.1591369] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Timothy D. Shaw
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, UK
| | - Daniel F. McAuley
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, UK
- Regional Intensive Care Unit, Royal Victoria Hospital, Belfast, UK
| | - Cecilia M. O’Kane
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, UK
| |
Collapse
|
4
|
Nakamura M, Takeuchi T, Shirakawa K, Furusako S. Anti-human CD14 monoclonal antibody improves survival following sepsis induced by endotoxin, but not following polymicrobial infection. Eur J Pharmacol 2017; 806:18-24. [PMID: 28322834 DOI: 10.1016/j.ejphar.2017.03.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 02/28/2017] [Accepted: 03/15/2017] [Indexed: 12/15/2022]
Abstract
Cluster of differentiation 14 (CD14), a pattern recognition receptor expressed on myeloid cells and a critical component of the innate immune system, mediates local and systemic host responses to gram-negative bacterial products, including lipopolysaccharide (LPS). Therefore, CD14 is an attractive target for development of sepsis therapies, and several monoclonal anti-CD14 antibodies have been reported. In this study, we prepared an anti-human CD14 monoclonal antibody, F1024-1-3, which suppressed LPS-induced upregulation of pro-inflammatory cytokines and an adhesion molecule in human peripheral mononuclear cells and human vascular endothelial cells. Half-maximal inhibitory concentrations in these assays ranged from 0.1 to 1μg/ml. In rabbits, intravenous administration (3mg/kg) as well as in vitro exposure of F1024-1-3 suppressed LPS-induced cytokine production in whole blood. In endotoxemia models generated by three sequential injections of LPS, intravenous administration of F1024-1-3 at 0.3-3mg/kg sharply reduced pro-inflammatory responses in a dose-dependent manner and moderately attenuated pro-coagulant responses; at 1mg/kg, the protein protected rabbits from lethality even when administered 2h after the initial LPS injection. However, F1024-1-3 (10mg/kg) given 2h post-surgery did not prevent death of rabbits in a cecal ligation and puncture model. Thus, suppression of CD14-mediated activation of leukocytes and endothelial cells alone may not be clinically efficacious for the treatment of severe sepsis and septic shock.
Collapse
Affiliation(s)
- Masaki Nakamura
- Discovery Research, Mochida Pharmaceutical Co., Ltd., Shizuoka, Japan.
| | - Takashi Takeuchi
- Discovery Research, Mochida Pharmaceutical Co., Ltd., Shizuoka, Japan
| | - Kamon Shirakawa
- Discovery Research, Mochida Pharmaceutical Co., Ltd., Shizuoka, Japan
| | - Shoji Furusako
- Discovery Research, Mochida Pharmaceutical Co., Ltd., Shizuoka, Japan
| |
Collapse
|
5
|
Nakamura M, Takeuchi T, Kawahara T, Hirose J, Nakayama K, Hosaka Y, Furusako S. Simultaneous targeting of CD14 and factor XIa by a fusion protein consisting of an anti-CD14 antibody and the modified second domain of bikunin improves survival in rabbit sepsis models. Eur J Pharmacol 2017; 802:60-68. [PMID: 28249709 DOI: 10.1016/j.ejphar.2017.02.045] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 02/05/2017] [Accepted: 02/24/2017] [Indexed: 02/03/2023]
Abstract
Severe sepsis is a complex, multifactorial, and rapidly progressing disease characterized by excessive inflammation and coagulation following bacterial infection. To simultaneously suppress pro-inflammatory and pro-coagulant responses, we genetically engineered a novel fusion protein (MR1007) consisting of an anti-CD14 antibody and the modified second domain of bikunin, and evaluated the potential of MR1007 as an anti-sepsis agent. Suppressive effects of MR1007 on lipopolysaccharide (LPS)-induced inflammatory responses were assessed using peripheral blood mononuclear cells or endothelial cells. Its inhibitory activity against the coagulation factor XIa was assessed using a purified enzyme and a chromogenic substrate. Anticoagulant activity was assessed using human or rabbit plasma. Anti-inflammatory and anti-coagulant effects and/or survival benefits were evaluated in an endotoxemia model and a cecal ligation and puncture model. MR1007 inhibited LPS-induced cytokine production in peripheral blood mononuclear cells and endothelial cells, inhibited factor XIa, and exhibited anticoagulant activity. In an endotoxemia model, 0.3-3mg/kg MR1007 suppressed pro-inflammatory and pro-coagulant responses in a dose-dependent manner; at a dose of 3mg/kg, the protein improved survival even when administered 8h after the LPS injection. In addition, 10mg/kg MR1007 administered 2h post cecal ligation and puncture improved survival. However, MR1007 administered at doses up to 30mg/kg did not increase ear bleeding time or bacterial counts in the cecal ligation and puncture model. Thus, simultaneous targeting of CD14 and factor XIa improves survival in the rabbit endotoxemia and sepsis models and represents a promising approach for the treatment of severe sepsis.
Collapse
Affiliation(s)
- Masaki Nakamura
- Discovery Research, Mochida Pharmaceutical Co., Ltd., 722 Jimba-aza-Uenohara, Gotemba, Shizuoka 412-8524, Japan.
| | - Takashi Takeuchi
- Discovery Research, Mochida Pharmaceutical Co., Ltd., 722 Jimba-aza-Uenohara, Gotemba, Shizuoka 412-8524, Japan
| | - Tetsushi Kawahara
- Discovery Research, Mochida Pharmaceutical Co., Ltd., 722 Jimba-aza-Uenohara, Gotemba, Shizuoka 412-8524, Japan
| | - Jiro Hirose
- Discovery Research, Mochida Pharmaceutical Co., Ltd., 722 Jimba-aza-Uenohara, Gotemba, Shizuoka 412-8524, Japan
| | - Kazuyuki Nakayama
- Discovery Research, Mochida Pharmaceutical Co., Ltd., 722 Jimba-aza-Uenohara, Gotemba, Shizuoka 412-8524, Japan
| | - Yoshitaka Hosaka
- Discovery Research, Mochida Pharmaceutical Co., Ltd., 722 Jimba-aza-Uenohara, Gotemba, Shizuoka 412-8524, Japan
| | - Shoji Furusako
- Discovery Research, Mochida Pharmaceutical Co., Ltd., 722 Jimba-aza-Uenohara, Gotemba, Shizuoka 412-8524, Japan
| |
Collapse
|
6
|
Axtelle T, Pribble J. An overview of clinical studies in healthy subjects and patients with severe sepsis with IC14, a CD14-specific chimeric monoclonal antibody. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519030090061301] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
An overview and discussion of clinical studies with IC14, a chimeric monoclonal antibody directed against human CD14 is presented. These studies include phase 1 trials in: (i) healthy subjects; (ii) healthy subjects challenged with lipopolysaccharide (LPS); and (iii) patients with severe sepsis. The results from clinical studies of IC14 demonstrate this chimeric monoclonal antibody diminishes the systemic inflammatory response directed against LPS and may have potential as a treatment to prevent organ dysfunction in patients with severe sepsis. Furthermore, IC14 treatment was generally well-tolerated in both healthy subjects and severe sepsis patients and did not increase the incidence of secondary infections. Analysis of pharmacology data indicate that higher doses of IC14 are required to saturate membrane-bound CD14 receptors effectively in patients with severe sepsis than in healthy subjects. Finally, the results of non-clinical studies indicate that IC14 should only be administered with concurrent antimicrobial therapy in patients with infection.
Collapse
|
7
|
Hiki N, Mimura Y, Ogawa T, Kojima J, Hatao F, Kaminishi M. Pathophysiological relevance of the CD14 receptor in surgical patients: biological activity of endotoxin is regulated by the CD14 receptor. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519010070060101] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Endotoxins (lipopolysaccharides, LPSs) are potent bacterial poisons, and they are always present in the intestine in considerable numbers. Stress, such that as a resulting from multiple injuries, burns, hypovolemia, hypoxia, intestinal ischemia, and surgery can lead to a breakdown of the gut barrier, allowing endotoxins to enter the systemic circulation via translocation. However, estimating the biological activity of translocated circulating endotoxins and identification of the mechanisms regulating their biological activities remain complex problems. CD14 has been found to exist as a soluble protein in the serum and as a glycosylphosphatidylinositol (GPI)-anchored protein of myeloid lineage cells. It plays key roles in both LPS-induced activation and in LPS internalization by cells. In this article, we outline: (i) the biological activity of circulating endotoxin; and (ii) the role of membrane and/or soluble CD14 regulating the bioactivity of circulating endotoxin in a human model of postoperative endotoxemia.
Collapse
Affiliation(s)
- Naoki Hiki
- Department of Surgery, University of Tokyo, Tokyo, Japan,
| | | | | | - Junichi Kojima
- Department of Surgery, University of Tokyo, Tokyo, Japan
| | - Fumihiko Hatao
- Department of Surgery, University of Tokyo, Tokyo, Japan
| | | |
Collapse
|
8
|
Axtelle T, Pribble J. IC14, a CD14 specific monoclonal antibody, is a potential treatment for patients with severe sepsis. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519010070040201] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
CD14 is a pattern recognition receptor for the bacterial cell wall components from Gram-positive and Gram-negative bacteria as well as mycobacteria. Binding of lipopolysaccharide (LPS) or other cell wall constituents to CD14 initiates signal transduction through the Toll-like receptors resulting in the release of pro-inflammatory cytokines and the initiation of the systemic inflammatory response. In rabbits and non-human primates, CD14 specific antibodies were shown to attenuate responses to LPS or Escherichia coli challenge including pro-inflammatory cytokine release, acute lung injury, hypotension and changes in lung, liver, spleen and adrenal gland morphology. In healthy human subjects, single doses of a chimeric CD14 antibody (IC14) have been shown to be well tolerated and result in a dose-related degree of saturation of CD14 receptors on monocytes and granulocytes. Pretreatment of healthy subjects with IC14 2 h prior to LPS resulted in an attenuation of the LPS-induced fever, clinical symptoms, and leukocyte activation and degranulation. IC14 inhibited the release of TNF-α, IL-6, and IL-10 and delayed the release of sTNFRI and IL-1ra. Further studies are in progress to characterize the safety and clinical pharmacology of IC14 in patients with severe sepsis.
Collapse
|
9
|
Carey AJ, Sullivan MJ, Duell BL, Crossman DK, Chattopadhyay D, Brooks AJ, Tan CK, Crowley M, Sweet MJ, Schembri MA, Ulett GC. Uropathogenic Escherichia coli Engages CD14-Dependent Signaling to Enable Bladder-Macrophage-Dependent Control of Acute Urinary Tract Infection. J Infect Dis 2015; 213:659-68. [PMID: 26324782 DOI: 10.1093/infdis/jiv424] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 08/11/2015] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND CD14, a coreceptor for several pattern recognition receptors and a widely used monocyte/macrophage marker, plays a key role in host responses to gram-negative bacteria. Despite the central role of CD14 in the inflammatory response to lipopolysaccharide and other microbial products and in the dissemination of bacteria in some infections, the signaling networks controlled by CD14 during urinary tract infection (UTI) are unknown. METHODS We used uropathogenic Escherichia coli (UPEC) infection of wild-type (WT) C57BL/6 and Cd14(-/-) mice and RNA sequencing to define the CD14-dependent transcriptional signature and the role of CD14 in host defense against UTI in the bladder. RESULTS UPEC induced the upregulation of Cd14 and the monocyte/macrophage-related genes Emr1/F4/80 and Csf1r/c-fms, which was associated with lower UPEC burdens in WT mice, compared with Cd14(-/-) mice. Exacerbation of infection in Cd14(-/-) mice was associated with the absence of a 491-gene transcriptional signature in the bladder that encompassed multiple host networks not previously associated with this receptor. CD14-dependent pathways included immune cell trafficking, differential cytokine production in macrophages, and interleukin 17 signaling. Depletion of monocytes/macrophages in the bladder by administration of liposomal clodronate led to higher UPEC burdens. CONCLUSIONS This study identifies new host protective and signaling roles for CD14 in the bladder during UPEC UTI.
Collapse
Affiliation(s)
- Alison J Carey
- Menzies Health Institute Queensland, Griffith University, Gold Coast School of Medical Science, Griffith University, Gold Coast
| | - Matthew J Sullivan
- Menzies Health Institute Queensland, Griffith University, Gold Coast School of Medical Science, Griffith University, Gold Coast
| | - Benjamin L Duell
- Menzies Health Institute Queensland, Griffith University, Gold Coast School of Medical Science, Griffith University, Gold Coast
| | - David K Crossman
- Heflin Center for Human Genetics, School of Medicine, University of Alabama at Birmingham
| | - Debasish Chattopadhyay
- Division of Infectious Diseases, School of Medicine, University of Alabama at Birmingham
| | - Andrew J Brooks
- University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute
| | - Chee K Tan
- Menzies Health Institute Queensland, Griffith University, Gold Coast School of Medical Science, Griffith University, Gold Coast
| | - Michael Crowley
- Heflin Center for Human Genetics, School of Medicine, University of Alabama at Birmingham
| | - Matthew J Sweet
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Australia Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Mark A Schembri
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Australia School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Australia
| | - Glen C Ulett
- Menzies Health Institute Queensland, Griffith University, Gold Coast School of Medical Science, Griffith University, Gold Coast Division of Infectious Diseases, School of Medicine, University of Alabama at Birmingham
| |
Collapse
|
10
|
Chang MY, Tanino Y, Vidova V, Kinsella MG, Chan CK, Johnson PY, Wight TN, Frevert CW. Reprint of: A rapid increase in macrophage-derived versican and hyaluronan in infectious lung disease. Matrix Biol 2014; 35:162-73. [PMID: 24727035 PMCID: PMC4096977 DOI: 10.1016/j.matbio.2014.04.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 01/21/2014] [Accepted: 01/21/2014] [Indexed: 01/08/2023]
Abstract
The goals of this study were to characterize the changes in chondroitin sulfate proteoglycans and hyaluronan in lungs in acute response to gram-negative bacterial infection and to identify cellular components responsible for these changes. Mice were treated with intratracheal (IT) live Escherichia coli, E. coli lipopolysaccharide (LPS), or PBS. Both E. coli and LPS caused rapid selective increases in mRNA expression of versican and hyaluronan synthase (Has) isoforms 1 and 2 associated with increased immunohistochemical and histochemical staining for versican and hyaluronan in the lungs. Versican was associated with a subset of alveolar macrophages. To examine whether macrophages contribute to versican and hyaluronan accumulation, in vitro studies with primary cultures of bone marrow-derived and alveolar macrophages were performed. Unstimulated macrophages expressed very low levels of versican and hyaluronan synthase mRNA, with no detectible versican protein or hyaluronan product. Stimulation with LPS caused rapid increases in versican mRNA and protein, a rapid increase in Has1 mRNA, and concomitant inhibition of hyaluronidases 1 and 2, the major hyaluronan degrading enzymes. Hyaluronan could be detected following chloroquine pre-treatment, indicating rapid turnover and degradation of hyaluronan by macrophages. In addition, the effects of LPS, the M1 macrophage classical activation agonist, were compared to those of IL-4/IL-13 or IL-10, the M2a and M2c alternative activation agonists, respectively. Versican and Has1 increased only in response to M1 activation. Finally, the up-regulation of versican and Has1 in the whole lungs of wild-type mice following IT LPS was completely abrogated in TLR-4(-/-) mice. These findings suggest that versican and hyaluronan synthesis may play an important role in the innate immune response to gram-negative lung infection.
Collapse
Affiliation(s)
- Mary Y Chang
- Comparative Pathology Program, Department of Comparative Medicine, University of Washington School of Medicine, Seattle, WA, United States.
| | - Yoshinori Tanino
- Fukushima Medical University School of Medicine, Department of Pulmonary Medicine, Fukushima, Japan
| | - Veronika Vidova
- Comparative Pathology Program, Department of Comparative Medicine, University of Washington School of Medicine, Seattle, WA, United States
| | - Michael G Kinsella
- Hope Heart Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - Christina K Chan
- Hope Heart Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - Pamela Y Johnson
- Hope Heart Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - Thomas N Wight
- Hope Heart Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - Charles W Frevert
- Comparative Pathology Program, Department of Comparative Medicine, University of Washington School of Medicine, Seattle, WA, United States; Division of Pulmonary/Critical Care Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
11
|
Chang MY, Tanino Y, Vidova V, Kinsella MG, Chan CK, Johnson PY, Wight TN, Frevert CW. A rapid increase in macrophage-derived versican and hyaluronan in infectious lung disease. Matrix Biol 2014; 34:1-12. [PMID: 24472738 DOI: 10.1016/j.matbio.2014.01.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 01/21/2014] [Accepted: 01/21/2014] [Indexed: 02/08/2023]
Abstract
The goals of this study were to characterize the changes in chondroitin sulfate proteoglycans and hyaluronan in lungs in acute response to gram-negative bacterial infection and to identify cellular components responsible for these changes. Mice were treated with intratracheal (IT) live Escherichia coli, E. coli lipopolysaccharide (LPS), or PBS. Both E. coli and LPS caused rapid selective increases in mRNA expression of versican and hyaluronan synthase (Has) isoforms 1 and 2 associated with increased immunohistochemical and histochemical staining for versican and hyaluronan in the lungs. Versican was associated with a subset of alveolar macrophages. To examine whether macrophages contribute to versican and hyaluronan accumulation, in vitro studies with primary cultures of bone marrow-derived and alveolar macrophages were performed. Unstimulated macrophages expressed very low levels of versican and hyaluronan synthase mRNA, with no detectible versican protein or hyaluronan product. Stimulation with LPS caused rapid increases in versican mRNA and protein, a rapid increase in Has1 mRNA, and concomitant inhibition of hyaluronidases 1 and 2, the major hyaluronan degrading enzymes. Hyaluronan could be detected following chloroquine pre-treatment, indicating rapid turnover and degradation of hyaluronan by macrophages. In addition, the effects of LPS, the M1 macrophage classical activation agonist, were compared to those of IL-4/IL-13 or IL-10, the M2a and M2c alternative activation agonists, respectively. Versican and Has1 increased only in response to M1 activation. Finally, the up-regulation of versican and Has1 in the whole lungs of wild-type mice following IT LPS was completely abrogated in TLR-4(-/-) mice. These findings suggest that versican and hyaluronan synthesis may play an important role in the innate immune response to gram-negative lung infection.
Collapse
Affiliation(s)
- Mary Y Chang
- Comparative Pathology Program, Department of Comparative Medicine, University of Washington School of Medicine, Seattle, WA, United States.
| | - Yoshinori Tanino
- Fukushima Medical University School of Medicine, Department of Pulmonary Medicine, Fukushima, Japan
| | - Veronika Vidova
- Comparative Pathology Program, Department of Comparative Medicine, University of Washington School of Medicine, Seattle, WA, United States
| | - Michael G Kinsella
- Hope Heart Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - Christina K Chan
- Hope Heart Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - Pamela Y Johnson
- Hope Heart Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - Thomas N Wight
- Hope Heart Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - Charles W Frevert
- Comparative Pathology Program, Department of Comparative Medicine, University of Washington School of Medicine, Seattle, WA, United States; Division of Pulmonary/Critical Care Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
12
|
Zanoni I, Granucci F. Role of CD14 in host protection against infections and in metabolism regulation. Front Cell Infect Microbiol 2013; 3:32. [PMID: 23898465 PMCID: PMC3721004 DOI: 10.3389/fcimb.2013.00032] [Citation(s) in RCA: 187] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 07/05/2013] [Indexed: 01/14/2023] Open
Abstract
CD14 is a glycosylphosphatidylinositol (GPI)-anchored receptor known to serve as a co-receptor for several Toll-like Receptors (TLRs) both at the cell surface and in the endosomal compartment. CD14 can be expressed by cells of both hematopoietic and non-hematopoietic origin as a cell membrane or secreted protein. Although CD14 was discovered more than 20 years ago, its activities remain largely to be defined. Most of the information available concerns CD14's role as a co-receptor working with TLR4 and facilitating cellular responses to low doses of lipopolysaccharide (LPS). Recent studies have highlighted and molecularly defined many other functions of this pattern recognition receptor (PRR). These functions include the mechanisms through which CD14 allows the activation of the TLR4-TRAM-TRIF pathway upon LPS stimulation; the capacity of CD14 to transduce a TLR4-independent signaling pathway leading to the activation of NFAT transcription factor family members with important consequences in myeloid cells; the CD14 influence on cell metabolism in conditions predisposing to obesity. In this review, we summarize recent progresses toward the molecular definition of the multiple roles exerted by CD14 in innate immune cells in response to LPS and the consequences of CD14 activation in physiologic and pathologic conditions.
Collapse
Affiliation(s)
- Ivan Zanoni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Milan, Italy.
| | | |
Collapse
|
13
|
Preuß S, Omam FD, Scheiermann J, Stadelmann S, Winoto-Morbach S, von Bismarck P, Adam-Klages S, Knerlich-Lukoschus F, Lex D, Wesch D, Held-Feindt J, Uhlig S, Schütze S, Krause MF. Topical application of phosphatidyl-inositol-3,5-bisphosphate for acute lung injury in neonatal swine. J Cell Mol Med 2012; 16:2813-26. [PMID: 22882773 PMCID: PMC4118249 DOI: 10.1111/j.1582-4934.2012.01618.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 08/03/2012] [Indexed: 11/28/2022] Open
Abstract
Hypoxemic respiratory failure of the neonatal organism involves increased acid sphingomyelinase (aSMase) activity and production of ceramide, a second messenger of a pro-inflammatory pathway that promotes increased vascular permeability, surfactant alterations and alveolar epithelial apoptosis. We comparatively assessed the benefits of topical aSMase inhibition by either imipramine (Imi) or phosphatidylinositol-3,5-bisphosphate (PIP2) when administered into the airways together with surfactant (S) for fortification. In this translational study, a triple-hit acute lung injury model was used that entails repeated airway lavage, injurious ventilation and tracheal lipopolysaccharide instillation in newborn piglets subject to mechanical ventilation for 72 hrs. After randomization, we administered an air bolus (control), S, S+Imi, or S+PIP2. Only in the latter two groups we observed significantly improved oxygenation and ventilation, dynamic compliance and pulmonary oedema. S+Imi caused systemic aSMase suppression and ceramide reduction, whereas the S+PIP2 effect remained compartmentalized in the airways because of the molecule's bulky structure. The surfactant surface tensions improved by S+Imi and S+PIP2 interventions, but only to a minor extent by S alone. S+PIP2 inhibited the migration of monocyte-derived macrophages and granulocytes into airways by the reduction of CD14/CD18 expression on cell membranes and the expression of epidermal growth factors (amphiregulin and TGF-β1) and interleukin-6 as pro-fibrotic factors. Finally we observed reduced alveolar epithelial apoptosis, which was most apparent in S+PIP2 lungs. Exogenous surfactant "fortified" by PIP2, a naturally occurring surfactant component, improves lung function by topical suppression of aSMase, providing a potential treatment concept for neonates with hypoxemic respiratory failure.
Collapse
Affiliation(s)
- Stefanie Preuß
- Universitätsklinikum Schleswig-Holstein, Campus Kiel, Department of PediatricsKiel, Germany
| | - Friede D Omam
- Universitätsklinikum Schleswig-Holstein, Campus Kiel, Department of PediatricsKiel, Germany
| | - Julia Scheiermann
- Universitätsklinikum Schleswig-Holstein, Campus Kiel, Department of PediatricsKiel, Germany
| | - Sabrina Stadelmann
- Universitätsklinikum Schleswig-Holstein, Campus Kiel, Department of PediatricsKiel, Germany
| | - Supandi Winoto-Morbach
- Universitätsklinikum Schleswig-Holstein, Campus Kiel, Institute of ImmunologyKiel, Germany
| | - Philipp von Bismarck
- Universitätsklinikum Schleswig-Holstein, Campus Kiel, Department of PediatricsKiel, Germany
| | - Sabine Adam-Klages
- Universitätsklinikum Schleswig-Holstein, Campus Kiel, Institute of ImmunologyKiel, Germany
| | | | - Dennis Lex
- Universitätsklinikum, RWTH Aachen, Institute of Pharmacology and ToxicologyAachen, Germany
| | - Daniela Wesch
- Universitätsklinikum Schleswig-Holstein, Campus Kiel, Institute of ImmunologyKiel, Germany
| | - Janka Held-Feindt
- Universitätsklinikum Schleswig-Holstein, Campus Kiel, Department of NeurosurgeryKiel, Germany
| | - Stefan Uhlig
- Universitätsklinikum, RWTH Aachen, Institute of Pharmacology and ToxicologyAachen, Germany
| | - Stefan Schütze
- Universitätsklinikum Schleswig-Holstein, Campus Kiel, Institute of ImmunologyKiel, Germany
| | - Martin F Krause
- Universitätsklinikum Schleswig-Holstein, Campus Kiel, Department of PediatricsKiel, Germany
| |
Collapse
|
14
|
Panda SK, Kumar S, Tupperwar NC, Vaidya T, George A, Rath S, Bal V, Ravindran B. Chitohexaose activates macrophages by alternate pathway through TLR4 and blocks endotoxemia. PLoS Pathog 2012; 8:e1002717. [PMID: 22654663 PMCID: PMC3359989 DOI: 10.1371/journal.ppat.1002717] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 04/09/2012] [Indexed: 01/07/2023] Open
Abstract
Sepsis is a consequence of systemic bacterial infections leading to hyper activation of immune cells by bacterial products resulting in enhanced release of mediators of inflammation. Endotoxin (LPS) is a major component of the outer membrane of Gram negative bacteria and a critical factor in pathogenesis of sepsis. Development of antagonists that inhibit the storm of inflammatory molecules by blocking Toll like receptors (TLR) has been the main stay of research efforts. We report here that a filarial glycoprotein binds to murine macrophages and human monocytes through TLR4 and activates them through alternate pathway and in the process inhibits LPS mediated classical activation which leads to inflammation associated with endotoxemia. The active component of the nematode glycoprotein mediating alternate activation of macrophages was found to be a carbohydrate residue, Chitohexaose. Murine macrophages and human monocytes up regulated Arginase-1 and released high levels of IL-10 when incubated with chitohexaose. Macrophages of C3H/HeJ mice (non-responsive to LPS) failed to get activated by chitohexaose suggesting that a functional TLR4 is critical for alternate activation of macrophages also. Chitohexaose inhibited LPS induced production of inflammatory molecules TNF-α, IL-1β and IL-6 by macropahges in vitro and in vivo in mice. Intraperitoneal injection of chitohexaose completely protected mice against endotoxemia when challenged with a lethal dose of LPS. Furthermore, Chitohexaose was found to reverse LPS induced endotoxemia in mice even 6/24/48 hrs after its onset. Monocytes of subjects with active filarial infection displayed characteristic alternate activation markers and were refractory to LPS mediated inflammatory activation suggesting an interesting possibility of subjects with filarial infections being less prone to develop of endotoxemia. These observations that innate activation of alternate pathway of macrophages by chtx through TLR4 has offered novel opportunities to cell biologists to study two mutually exclusive activation pathways of macrophages being mediated through a single receptor. Sepsis is one of the leading causes of death contributing to mortality as high as 54 percent in intensive care units across the world. Hyper inflammation induced by bacteria or bacterial products through Toll like receptors leads to sepsis and hence current approaches are directed towards blockade such receptors. While many such candidate antagonists have shown promise they also result in induction of inappropriate innate immune responses thus increasing risk of development of shock leading to death. In this study we describe a novel approach to treat endotoxemia associated with sepsis, fundamentally different from other reports. Chitohexaose a small molecular weight polysaccharide by virtue of its ability to bind to active sites of TLR4 inhibited LPS induced production of inflammatory mediators by murine macrophages and human monocytes. Administration of chitohexaose with LPS blocked endotoxemia leading to mortality of mice. More significantly, Chitohexaose reversed inflammation and protected mice even 24/48 hrs after onset of endotoxemia. Apart from competitively inhibiting LPS induced inflammation chitohexaose also activated alternate pathway of macrophages. Such macrophages are known to display increased phagocytic activity, are resistant to LPS induced activation and associated with resolution of inflammation and tissue repair.
Collapse
Affiliation(s)
| | - Sunil Kumar
- Institute of Life Sciences, Bhubaneswar, India
| | | | - Tushar Vaidya
- Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Anna George
- National Institute of Immunology, New Delhi, India
| | | | - Vineeta Bal
- National Institute of Immunology, New Delhi, India
| | | |
Collapse
|
15
|
Muthu K, He LK, Szilagyi A, Strotmon P, Gamelli RL, Shankar R. ß-adrenergic stimulation increases macrophage CD14 expression and E. coli phagocytosis through PKA signaling mechanisms. J Leukoc Biol 2010; 88:715-24. [PMID: 20643814 DOI: 10.1189/jlb.0410186] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
CD14 is a glycoprotein that binds bacterial LPS in MØ. It is an essential component of the phagocytic system and is increased in septic shock. Critical injury and sepsis result in elevated endogenous CA levels. CAs have a significant impact on MØ inflammatory functions. We tested the hypothesis that β-adrenergic stimulation regulates CD14 expression and bacterial phagocytosis in BMØ. Murine BMØ stimulated with isoproterenol (>8 h) induced a dose-dependent increase in cell surface CD14 expression. Specific PKA inhibitor (H-89) and gene-silencing (siRNA) studies demonstrated the role of cAMP-dependent PKA in mediating this response. In addition, we observed a correlation between an isoproterenol-mediated increase in CD14 expression and live Escherichia coli uptake in BMØ. Further, the essential role of CD14 in an isoproterenol-mediated increase in E. coli uptake was highlighted from experiments using CD14(-/-) mice. Moreover, the dose response of isoproterenol stimulation to CD14 expression and E. coli phagocytosis overlapped with similar EC50. Additionally, isoproterenol-mediated E. coli phagocytosis was prevented by H-89, suggesting that β-adrenergic stimulus in BMØ increases CD14 expression and live E. coli phagocytosis through a common signaling pathway. Our studies indicate the potential impact of β-adrenergic agents on important innate immune functions.
Collapse
Affiliation(s)
- Kuzhali Muthu
- Department of Surgery, Loyola University Medical Center, Maywood, IL 60153, USA.
| | | | | | | | | | | |
Collapse
|
16
|
Anas A, van der Poll T, de Vos AF. Role of CD14 in lung inflammation and infection. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2010; 14:209. [PMID: 20236452 PMCID: PMC2887102 DOI: 10.1186/cc8850] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
This article is one of ten reviews selected from the Yearbook of Intensive Care and Emergency Medicine 2010 (Springer Verlag) and co-published as a series in Critical Care. Other articles in the series can be found online at http://ccforum.com/series/yearbook. Further information about the Yearbook of Intensive Care and Emergency Medicine is available from http://www.springer.com/series/2855.
Collapse
Affiliation(s)
- Adam Anas
- Center for Experimental and Molecular Medicine, Center of Infection and Immunity, Academic Medical Center, Meibergdreef 9, Amsterdam, Netherlands
| | | | | |
Collapse
|
17
|
Anas A, Van Der Poll T, De Vas AF. Role of CD14 in Lung Inflammation and Infection. Intensive Care Med 2010. [PMCID: PMC7120299 DOI: 10.1007/978-1-4419-5562-3_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Toll-like receptors (TLR) on the surface of cells of the respiratory tract play an essential role in sensing the presence of microorganisms in the airways and lungs. These receptors trigger inflammatory responses, activate innate immune responses, and prime adaptive immune responses to eradicate invading microbes [1]. TLR are members of a family of pattern-recognition receptors, which recognize molecular structures of bacteria, viruses, fungi and protozoa (pathogen-associated molecular patterns or PAMPs), as well as endogenous structures and proteins released during inflammation (damage/danger-associated molecular patterns or DAMPs). To date, ten different TLR have been identified in humans and twelve in mice. TLR are expressed on all cells of the immune system, but also on parenchymal cells of many organs and tissues. The binding of a PAMP to a TLR results in cellular activation and initiates a variety of effector functions, including cytokine secretion, proliferation’ co-stimulation or phagocyte maturation. To facilitate microbial recognition and to amplify cellular responses, certain TLR require additional proteins, such as lipopolysaccharide (LPS) binding protein (LBP), CD14, CD36 and high mobility group box-l protein (HMGB-l). In this chapter, the role of CD14 as an accessory receptor for TLR in lung inflammation and infection is discussed. The central role of CD14 in the recognition of various PAMPs and amplification of immune and inflammatory responses in the lung is depicted in Fig. 1. Central role of CD14 in pathogen- and pathogen-associated molecular pattern (PAMP)-induced responses in the lung. CD14, which lacks an intracellular domain for signal transduction, is expressed on the surface of alveolar macrophages, infiltrating monocytes and neutrophils, and at lower levels also on epithelial and endothelial cells in the lung. CD14 recognizes and binds various structures from invading microbes, such as lipopolysaccharide (LPS) from Gram-negative bacteria, lipoteichoic acid (LTA) from Gram-positive bacteria, lipoarabinomannan (LAM) from mycobacteria, viral double stranded (ds) RNA and F glycoprotein (F-gp) from respiratory syncytial virus (RSV). CD14 subsequently transfers these bound components to Toll-like receptors (TLR) which than trigger cell activation. Binding of LPS to CD14 is regulated by additional accessory receptors in the lung, including LPS-binding protein (LBP) and a number of surfactant proteins (SP). Furthermore, soluble CD14 (sCD14) enhances LPS-induced activation of cells with low CD14 expression. Depending on the microbe and the PAMPs it expresses, CD14-amplified responses can either be beneficial to the host by induction of an adequate inflammatory and immune response to eradicate the invading microbe, or detrimental to the host by excessive inflammation and/or dissemination of the pathogen. ![]()
Collapse
|
18
|
Role of CD14 in Lung Inflammation and Infection. YEARBOOK OF INTENSIVE CARE AND EMERGENCY MEDICINE 2010. [PMCID: PMC7124092 DOI: 10.1007/978-3-642-10286-8_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Toll-like receptors (TLR) on the surface of cells of the respiratory tract play an essential role in sensing the presence of microorganisms in the airways and lungs. These receptors trigger inflammatory responses, activate innate immune responses, and prime adaptive immune responses to eradicate invading microbes [1]. TLR are members of a family of pattern-recognition receptors, which recognize molecular structures of bacteria, viruses, fungi and protozoa (pathogen-associated molecular patterns or PAMPs), as well as endogenous structures and proteins released during inflammation (damage/danger-associated molecular patterns or DAMPs). To date, ten different TLR have been identified in humans and twelve in mice. TLR are expressed on all cells of the immune system, but also on parenchymal cells of many organs and tissues. The binding of a PAMP to a TLR results in cellular activation and initiates a variety of effector functions, including cytokine secretion, proliferation, co-stimulation or phagocyte maturation. To facilitate microbial recognition and to amplify cellular responses, certain TLR require additional proteins, such as lipopolysaccharide (LPS) binding protein (LBP), CD14, CD36 and high mobility group box-1 protein (HMGB-1). In this chapter, the role of CD14 as an accessory receptor for TLR in lung inflammation and infection is discussed. The central role of CD14 in the recognition of various PAMPs and amplification of immune and inflammatory responses in the lung is depicted in Figure 1.
Central role of CD14 in pathogen- and pathogen-associated molecular pattern (PAMP)-induced responses in the lung. CD14, which lacks an intracellular domain for signal transduction, is expressed on the surface of alveolar macrophages, infiltrating monocytes and neutrophils, and at lower levels also on epithelial and endothelial cells in the lung. CD14 recognizes and binds various structures from invading microbes, such as lipopolysaccharide (LPS) from Gram-negative bacteria, lipoteichoic acid (LTA) from Grampositive bacteria, lipoarabinomannan (LAM) from mycobacteria, viral double stranded (ds) RNA and F glycoprotein (F-gp) from respiratory syncytial virus (RSV). CD14 subsequently transfers these bound components to Toll-like receptors (TLR) which than trigger cell activation. Binding of LPS to CD14 is regulated by additional accessory receptors in the lung, including LPS-binding protein (LBP) and a number of surfactant proteins (SP). Furthermore, soluble CD14 (sCD14) enhances LPS-induced activation of cells with low CD14 expression. Depending on the microbe and the PAMPs it expresses, CD14-amplified responses can either be beneficial to the host by induction of an adequate inflammatory and immune response to eradicate the invading microbe, or detrimental to the host by excessive inflammation and/or dissemination of the pathogen. ![]()
Collapse
|
19
|
Thorgersen EB, Hellerud BC, Nielsen EW, Barratt-Due A, Fure H, Lindstad JK, Pharo A, Fosse E, Tønnessen TI, Johansen HT, Castellheim A, Mollnes TE. CD14 inhibition efficiently attenuates early inflammatory and hemostatic responses in Escherichia coli sepsis in pigs. FASEB J 2009; 24:712-22. [PMID: 19841036 DOI: 10.1096/fj.09-140798] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Sepsis is a severe infection-induced systemic inflammatory syndrome. Inhibition of downstream inflammatory mediators of sepsis, e.g., TNF-alpha, has failed in clinical trials. The aim of this study was to investigate the effects of inhibiting CD14, a key upstream innate immunity molecule, on the early inflammatory and hemostatic responses in a pig model of gram-negative sepsis. The study comprised two arms, whole live Escherichia coli bacteria and E. coli lipopolysaccharide (LPS) (n=25 and n=9 animals, respectively). The animals were allocated into treatment (anti-CD14) and control (IgG isotype or saline) groups. Inflammatory, hemostatic, physiological, and microbiological parameters were measured. The proinflammatory cytokines TNF-alpha, IL-1beta, IL-6, and IL-8, but not the anti-inflammatory cytokine IL-10, were efficiently inhibited by anti-CD14. Furthermore, anti-CD14 preserved the leukocyte count and significantly reduced granulocyte enzyme matrix metalloproteinase-9 release and expression of the granulocyte membrane activation molecule wCD11R3 (pig CD11b). The hemostatic markers thrombin-antithrombin III complexes and plasminogen activator inhibitor-1 were significantly attenuated. Anti-CD14 did not affect LPS or E. coli DNA levels. This study documents that CD14 inhibition efficiently attenuates the proinflammatory cytokine response and granulocyte activation and reverses the procoagulant state but does not interfere with LPS levels or bacterial counts in E. coli-induced sepsis.-Thorgersen, E. B., Hellerud, B. C., Nielsen, E. W., Barratt-Due, A., Fure, H., Lindstad, J. K., Pharo, A., Fosse, E., Tønnessen, T. I., Johansen, H. T., Castellheim, A., Mollnes, T. E. CD14 inhibition efficiently attenuates early inflammatory and hemostatic responses in Escherichia coli sepsis in pigs.
Collapse
|
20
|
Su X, Looney M, Robriquet L, Fang X, Matthay MA. DIRECT VISUAL INSTILLATION AS A METHOD FOR EFFICIENT DELIVERY OF FLUID INTO THE DISTAL AIRSPACES OF ANESTHETIZED MICE. Exp Lung Res 2009; 30:479-93. [PMID: 15524406 DOI: 10.1080/01902140490476382] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Although several methods have been used to deliver fluid into the distal airspaces of the lung, the efficiency of these methods has been variable. Therefore, the authors have modified prior techniques to design a better method for direct visual instillation (DVI) of fluid into the trachea and compared its efficiency with two commonly used methods: nasal inhalation and invasive intratracheal instillation (delivery of the instillate by needle puncture of the trachea). The results showed that this method (DVI) can deliver fluid efficiently into either both lungs or into a single lung. Using an 131I-albumin labeling technique, DVI resulted in 92 +/- 1% retention of the labeled albumin in the lungs 1 hour after instillation, significantly greater than nasal inhalation (48 +/- 3%, P < .01) and invasive intratracheal instillation (77 +/- 3%, P < .05). Also, when bacteria (Escherichia coli) were instilled with the DVI method, the severity of gram-negative pneumonia was greater (6.5 +/- 0.5 g water/g dry weight) compared to delivery by nasal inhalation (5.5 +/- 0.4 g water/g dry weight, P < .05) or by invasive intratracheal instillation (5.9 +/- 0.4g water/g dry weight, P < .05). The authors conclude that DVI is more efficient than nasal inhalation and invasive intratracheal instillation for delivering experimental fluids into the distal airspaces of anesthetized mice. This method should be valuable for experimental lung studies in mice.
Collapse
Affiliation(s)
- Xiao Su
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143-0130, USA
| | | | | | | | | |
Collapse
|
21
|
Protection from lethal gram-negative bacterial sepsis by targeting Toll-like receptor 4. Proc Natl Acad Sci U S A 2009; 106:2348-52. [PMID: 19181857 DOI: 10.1073/pnas.0808146106] [Citation(s) in RCA: 226] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Toll-like receptor 4 (TLR4), the signal-transducing molecule of the LPS receptor complex, plays a fundamental role in the sensing of LPS from gram-negative bacteria. Activation of TLR4 signaling pathways by LPS is a critical upstream event in the pathogenesis of gram-negative sepsis, making TLR4 an attractive target for novel antisepsis therapy. To validate the concept of TLR4-targeted treatment strategies in gram-negative sepsis, we first showed that TLR4(-/-) and myeloid differentiation primary response gene 88 (MyD88)(-/-) mice were fully resistant to Escherichia coli-induced septic shock, whereas TLR2(-/-) and wild-type mice rapidly died of fulminant sepsis. Neutralizing anti-TLR4 antibodies were then generated using a soluble chimeric fusion protein composed of the N-terminal domain of mouse TLR4 (amino acids 1-334) and the Fc portion of human IgG1. Anti-TLR4 antibodies inhibited intracellular signaling, markedly reduced cytokine production, and protected mice from lethal endotoxic shock and E. coli sepsis when administered in a prophylactic and therapeutic manner up to 13 h after the onset of bacterial sepsis. These experimental data provide strong support for the concept of TLR4-targeted therapy for gram-negative sepsis.
Collapse
|
22
|
Wiersinga WJ, de Vos AF, Wieland CW, Leendertse M, Roelofs JJTH, van der Poll T. CD14 impairs host defense against gram-negative sepsis caused by Burkholderia pseudomallei in mice. J Infect Dis 2008; 198:1388-97. [PMID: 18855560 DOI: 10.1086/592220] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND CD14 is a pattern-recognition receptor that can facilitate the presentation of bacterial components to either Toll-like receptor 2 (TLR2) or TLR4. We have recently shown that during melioidosis, a severe infection caused by the gram-negative bacterium Burkholderia pseudomallei, TLR2 but not TLR4 impacts the immune response of the intact host in vivo. METHODS The function of CD14 in melioidosis was analyzed by means of in vitro and in vivo approaches, using wild-type (WT) and CD14 knockout (KO) mice. RESULTS CD14-deficient macrophages and whole blood leukocytes released less tumor necrosis factor (TNF)-alpha on stimulation with B. pseudomallei or B. pseudomallei lipopolysaccharide in vitro, compared with WT cells. Strikingly, CD14 KO mice intranasally inoculated with B. pseudomallei demonstrated reduced lethality and significantly decreased bacterial outgrowth, compared with WT mice. Administration of recombinant soluble CD14 to CD14 KO mice partially reversed their phenotype to that of WT mice. Lastly, CD14 deficiency did not alter the capacity of macrophages or neutrophils to phagocytose or kill B. pseudomallei. CONCLUSION CD14 is crucially involved in the recognition of B. pseudomallei by innate immune cells but plays a remarkable detrimental role in the host response against B. pseudomallei. Inhibition of CD14 may be a novel treatment strategy in melioidosis.
Collapse
Affiliation(s)
- W Joost Wiersinga
- Centers for Infection and Immunity Amsterdam, Academic Medical Center, Amsterdam, the Netherlands.
| | | | | | | | | | | |
Collapse
|
23
|
Smith LS, Kajikawa O, Elson G, Wick M, Mongovin S, Kosco-Vilbois M, Martin TR, Frevert CW. Effect of Toll-like receptor 4 blockade on pulmonary inflammation caused by mechanical ventilation and bacterial endotoxin. Exp Lung Res 2008; 34:225-43. [PMID: 18465402 DOI: 10.1080/01902140802022492] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Mechanical ventilation (MV) and lipopolysaccharide (LPS) synergistically increase inflammation and lung injury. The goal of this study was to determine whether blockade of CD14 or Toll-like receptor 4 (TLR4) would reduce inflammation caused by LPS and MV. Rabbits were pretreated with anti-TLR4 or anti-CD14 monoclonal antibodies, followed by endobronchial LPS and MV. Blockade of TLR4 reduced the number of neutrophils and the amount of CXCL8 in bronchoalveolar lavage fluid. In contrast, blockade of CD14 did not significantly decrease the number of neutrophils or the amount of CXCL8. These data show that TLR4 blockade reduces pulmonary inflammation caused by the combination of LPS and Mechanical ventilation.
Collapse
Affiliation(s)
- Lincoln S Smith
- Division of Pediatric Critical Care, Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Su X, Johansen M, Looney MR, Brown EJ, Matthay MA. CD47 deficiency protects mice from lipopolysaccharide-induced acute lung injury and Escherichia coli pneumonia. THE JOURNAL OF IMMUNOLOGY 2008; 180:6947-53. [PMID: 18453616 DOI: 10.4049/jimmunol.180.10.6947] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
CD47 modulates neutrophil transmigration toward the sites of infection or injury. Mice lacking CD47 are susceptible to Escherichia coli (E. coli) peritonitis. However, less is known concerning the role of CD47 in the development of acute lung inflammation and injury. In this study, we show that mice lacking CD47 are protected from LPS-induced acute lung injury and E. coli pneumonia with a significant reduction in pulmonary edema, lung vascular permeability, and bacteremia. Reconstitution of CD47(+/-) mice with CD47(-/-) neutrophils significantly reduced lung edema and neutrophil infiltration, thus demonstrating that CD47(+) neutrophils are required for the development of lung injury from E. coli pneumonia. Importantly, CD47-deficient mice with E. coli pneumonia had an improved survival rate. Taken together, deficiency of CD47 protects mice from LPS-induced acute lung injury and E. coli pneumonia. Targeting CD47 may be a novel pathway for treatment of acute lung injury.
Collapse
Affiliation(s)
- Xiao Su
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143, USA.
| | | | | | | | | |
Collapse
|
25
|
Affiliation(s)
- Joseph P Mizgerd
- Molecular and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston, MA 02115, USA.
| |
Collapse
|
26
|
Daubeuf B, Mathison J, Spiller S, Hugues S, Herren S, Ferlin W, Kosco-Vilbois M, Wagner H, Kirschning CJ, Ulevitch R, Elson G. TLR4/MD-2 Monoclonal Antibody Therapy Affords Protection in Experimental Models of Septic Shock. THE JOURNAL OF IMMUNOLOGY 2007; 179:6107-14. [DOI: 10.4049/jimmunol.179.9.6107] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
27
|
Dessing MC, van der Sluijs KF, Florquin S, van der Poll T. CD14 plays a limited role during influenza A virus infection in vivo. Immunol Lett 2007; 113:47-51. [PMID: 17825924 DOI: 10.1016/j.imlet.2007.07.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2007] [Revised: 07/02/2007] [Accepted: 07/22/2007] [Indexed: 01/12/2023]
Abstract
Influenza A is a single stranded (ss)RNA virus that can cause upper respiratory tract infections that in rare cases may progress to pneumonia. Toll-like receptors (TLRs) and CD14 are receptors which recognize viral proteins and nucleic acid of several viruses. CD14 is required for influenza-induced cytokine production during infection of mouse macrophages. In addition, CD14 was shown to bind ssRNA, suggesting an important role for CD14 during infection with influenza. To investigate the role of CD14 during influenza pneumonia we inoculated WT and CD14 KO mice with a non-lethal dose of a mouse adapted strain of influenza A. CD14 KO mice displayed a reduced viral load in the lungs, 2 and 14 days after infection with influenza. Pulmonary cytokine production in CD14 KO mice was reduced at day 2 and elevated at day 8 compared to WT mice. CD14 deficiency did not influence lymphocyte recruitment or lymphocyte activation in lungs and draining lymph nodes 8 days after infection. These data show that CD14 plays a limited role in host defense against infection with influenza.
Collapse
Affiliation(s)
- Mark C Dessing
- Center of Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands.
| | | | | | | |
Collapse
|
28
|
Metkar S, Awasthi S, Denamur E, Kim KS, Gangloff SC, Teichberg S, Haziot A, Silver J, Goyert SM. Role of CD14 in responses to clinical isolates of Escherichia coli: effects of K1 capsule expression. Infect Immun 2007; 75:5415-24. [PMID: 17709409 PMCID: PMC2168279 DOI: 10.1128/iai.00601-07] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Severe bacterial infections leading to sepsis or septic shock can be induced by bacteria that utilize different factors to drive pathogenicity and/or virulence, leading to disease in the host. One major factor expressed by all clinical isolates of gram-negative bacteria is lipopolysaccharide (LPS); a second factor expressed by some Escherichia coli strains is a K1 polysaccharide capsule. To determine the role of the CD14 LPS receptor in the pathogenic effects of naturally occurring E. coli, the responses of CD14-/- and CD14+/+ mice to three different isolates of E. coli obtained from sepsis patients were compared; two isolates express both smooth LPS and the K1 antigen, while the third isolate expresses only LPS and is negative for K1. An additional K1-positive isolate obtained from a newborn with meningitis and a K1-negative isogenic mutant of this strain were also used for these studies. CD14-/- mice were resistant to the lethal effects of the K1-negative isolates. This resistance was accompanied by significantly lower levels of systemic tumor necrosis factor alpha (TNF-alpha) and interleukin-6 (IL-6) in these mice than in CD14+/+ mice, enhanced clearance of the bacteria, and significantly fewer additional gross symptoms. In contrast, CD14-/- mice were as sensitive as CD14+/+ mice to the lethal effects of the K1-positive isolates, even though they had significantly lower levels of TNF-alpha and IL-6 than CD14+/+ mice. These studies show that different bacterial isolates can use distinctly different mechanisms to cause disease and suggest that new, nonantibiotic therapeutics need to be directed against multiple targets.
Collapse
Affiliation(s)
- Shalaka Metkar
- Department of Microbiology and Immunology, CUNY Medical School, and Sophie Davis School for Biomedical Sciences, City College of New York, New York, New York, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Lingnau M, Höflich C, Volk HD, Sabat R, Döcke WD. Interleukin-10 enhances the CD14-dependent phagocytosis of bacteria and apoptotic cells by human monocytes. Hum Immunol 2007; 68:730-8. [PMID: 17869646 DOI: 10.1016/j.humimm.2007.06.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2007] [Revised: 06/19/2007] [Accepted: 06/19/2007] [Indexed: 10/23/2022]
Abstract
Monocytes are centrally involved in both specific and nonspecific immunity by secretion of regulatory immune mediators, phagocytosis, and presentation of antigens. Recent work has shown that monocytes can phagocytose bacteria independently from Fc gamma, complement, and scavenger receptors via a CD14-mediated process. Furthermore, incorporation of cells undergoing apoptosis is also mediated by CD14. In this study we investigated the regulation of monocytic CD14-dependent phagocytosis by the immunoregulatory cytokines interleukin-10 (IL-10), interferon-gamma (IFN-gamma) and transforming growth factor-beta1 (TGF-beta1). In this study an in vitro human whole-blood assay was used to test regulation of CD14-dependent phagocytosis of fluorescence-labeled E. coli by IL-10, IFN-gamma, and TGF-beta1 in monocytes from healthy donors. Phagocytosis by monocytes from a patient with paroxysmal nocturnal hemoglobinuria (PNH) and its regulation by IL-10 was also investigated. Finally, regulation of monocytic incorporation of apoptotic Jurkat cells by IL-10 was analyzed. For the CD14 blockade, murine anti-CD14 IgG2a antibody RMO52 was used. We observed that IL-10, suggested to be a monocyte-deactivating cytokine, strongly increased the monocytic CD14-dependent phagocytosis of E. coli. In contrast, IFN-gamma and TGF-beta1 depressed monocytic CD14 incorporation of E. coli. Compatible with this, IL-10 upregulated CD14 expression on monocytes, whereas IFN-gamma and TGF-beta1 downregulated its expression. IL-10 also increased the monocytic CD14-dependent and -independent phagocytosis of apoptotic cells. As expected, IL-10 strongly increased the CD14-independent phagocytosis but had no influence on the CD14-dependent phagocytosis of monocytes from a PNH patient. In conclusion, our data support a general role of IL-10 for activating monocytic scavenger functions, which are at least partly mediated by CD14. This is in line with the fact that IL-10 promotes the development of monocytes to macrophages. The contrasting effects of IL-10 and IFN-gamma on monocytic CD14-dependent phagocytosis may reflect a further mechanism counterbalancing antigen-presentation and nonimmunogenic scavenging of bacterial and cellular debris. TGF-beta, however, may be an inhibitor of both systems.
Collapse
Affiliation(s)
- Marcel Lingnau
- Institute of Medical Immunology, University Hospital Charité, Berlin, Germany
| | | | | | | | | |
Collapse
|
30
|
Brass DM, Hollingsworth JW, McElvania-Tekippe E, Garantziotis S, Hossain I, Schwartz DA. CD14 is an essential mediator of LPS-induced airway disease. Am J Physiol Lung Cell Mol Physiol 2007; 293:L77-83. [PMID: 17384086 DOI: 10.1152/ajplung.00282.2006] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Chronic lipopolysaccharide (LPS) inhalation in rodents recapitulates many classic features of chronic obstructive pulmonary disease seen in humans, including airways hyperresponsiveness, neutrophilic inflammation, cytokine production in the lung, and small airways remodeling. CD14-deficient mice (C57BL/6(CD14-/-)) have an altered response to systemic LPS, and yet the role of CD14 in the response to inhaled LPS has not been defined. We observed that C57BL/6(CD14-/-) mice demonstrate no discernable physiological or inflammatory response to a single LPS inhalation challenge. However, the physiological (airways hyperresponsiveness) and inflammatory (presence of neutrophils and TNF-alpha in whole lung lavage fluid) responsiveness to inhaled LPS in C57BL/6(CD14-/-) mice was restored by instilling soluble CD14 intratracheally. Intratracheal instillation of wild-type macrophages into C57BL/6(CD14-/-) mice restored neutrophilic inflammation only and failed to restore airways hyperresponsiveness or TNF-alpha protein in whole lung lavage. These findings demonstrate that CD14 is critical to LPS-induced airway disease and that macrophage CD14 is sufficient to initiate neutrophil recruitment into the airways but that CD14 may need to interact with other cell types as well for the development of airways hyperresponsiveness and for cytokine production.
Collapse
Affiliation(s)
- David M Brass
- National Institute of Environmental Health Sciences, Rall Bldg., Rm. C224, PO Box 12233 MD C2-15, 111 Alexander Dr., Research Triangle Park, NC 27709, USA.
| | | | | | | | | | | |
Collapse
|
31
|
Nita IM, Serapinas D, Janciauskiene SM. α1-Antitrypsin regulates CD14 expression and soluble CD14 levels in human monocytes in vitro. Int J Biochem Cell Biol 2007; 39:1165-76. [PMID: 17448722 DOI: 10.1016/j.biocel.2007.02.017] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2006] [Revised: 02/23/2007] [Accepted: 02/26/2007] [Indexed: 11/24/2022]
Abstract
The recognition of bacterial lipopolysaccharide (LPS) is principally mediated by either membrane-bound or soluble form of the glycoprotein CD14 and CD14-associated signal transducer, toll-like receptor 4 (TLR4). Recent findings indicate that the serine protease inhibitor, alpha1-antitrypsin (AAT), may not only afford protection against proteolytic injury, but may also neutralize microbial activities and affect regulation of innate immunity. We postulated that AAT affects monocyte responses to LPS by regulating CD14 expression and soluble CD14 release. Here we show that a short-term (up to 2h) monocyte exposure to AAT alone or in combination with LPS leads to a remarkable induction of CD14 levels. In parallel, a short-term (2h) cell exposure to AAT/LPS significantly enhances LPS-induced NF kappaB (p50 and p65) activation in conjunction with increased TNFalpha, IL-1 beta and IL-8 release. In contrast, longer term incubation (18 h) of monocytes with combined AAT/LPS results in a significant reduction in expression of both CD14 and TLR4, inhibition of LPS-induced TNFalpha, IL-1 beta and IL-8 mRNA and protein expression. These findings provide evidence that AAT is an important regulator of CD14 expression and release in monocytes and suggest that AAT may be involved in LPS neutralization and prevention of over-activation of monocytes in vivo.
Collapse
Affiliation(s)
- Izabela M Nita
- Department of Clinical Sciences, Wallenberg Laboratory, University Hospital Malmö, Lund University, SE-20502 Malmö, Sweden
| | | | | |
Collapse
|
32
|
Knapp S, Schultz MJ, van der Poll T. Pneumonia models and innate immunity to respiratory bacterial pathogens. Shock 2006; 24 Suppl 1:12-8. [PMID: 16374367 DOI: 10.1097/01.shk.0000191385.41689.f3] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Preclinical sepsis models have been used for decades to study the pathophysiologic processes during sepsis and shock. Although these studies revealed promising immunomodulating agents for the treatment of sepsis, clinical trials evaluating the efficacy of these new agents in patients with sepsis were disappointing. The main reason for this unsatisfactory experience might be that unlike the clinical situation, most of these preclinical models are devoid of a localized infectious source from which the infection disseminates. Studies on the effects of several immunomodulating strategies have demonstrated strikingly opposite results when sepsis models with a more natural route of infection, such as pneumonia, were used. In this review, we will give insights into pneumonia models and discuss results and differences in the innate immune responses during distinct pulmonary infection models.
Collapse
Affiliation(s)
- Sylvia Knapp
- Department of Internal Medicine, Medical University, Department of Intensive Care, University of Amsterdam, 1105AZ Amsterdam, The Netherlands.
| | | | | |
Collapse
|
33
|
Dessing MC, Knapp S, Florquin S, de Vos AF, van der Poll T. CD14 facilitates invasive respiratory tract infection by Streptococcus pneumoniae. Am J Respir Crit Care Med 2006; 175:604-11. [PMID: 17185649 DOI: 10.1164/rccm.200606-824oc] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE CD14 is a pattern recognition receptor that can interact with a variety of bacterial ligands. During gram-negative infection, CD14 plays an important role in the induction of a protective immune response by virtue of its capacity to recognize lipopolysaccharide in the bacterial cell wall. Knowledge of the contribution of CD14 to host defense against gram-positive infections is limited. OBJECTIVES To study the role of CD14 in gram-positive bacterial pneumonia. METHODS CD14 knockout (KO) and normal wild-type (WT) mice were intranasally infected with Streptococcus pneumoniae. MEASUREMENTS AND MAIN RESULTS CD14 KO mice demonstrated a strongly reduced lethality, which was accompanied by a more than 10-fold lower bacterial load in lung homogenates but not in bronchoalveolar lavage fluid at 48 hours after infection. Strikingly, CD14 KO mice failed to develop positive blood cultures, whereas WT mice had positive blood cultures from 24 hours onward and eventually invariably had evidence of systemic infection. Lung inflammation was attenuated in CD14 KO mice at 48 hours after infection, as evaluated by histopathology and cytokine and chemokine levels. Intrapulmonary delivery of recombinant soluble CD14 to CD14 KO mice rendered them equally susceptible to S. pneumoniae as WT mice, resulting in enhanced bacterial growth in lung homogenates and bacteremia, indicating that the presence of soluble CD14 in the bronchoalveolar compartment is sufficient to cause invasive pneumococcal disease. CONCLUSION These data suggest that S. pneumoniae uses (soluble) CD14 present in the bronchoalveolar space to cause invasive respiratory tract infection.
Collapse
Affiliation(s)
- Mark C Dessing
- Center for Infection and Immunity Amsterdam, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
34
|
Stearns-Kurosawa DJ, Lupu F, Taylor FB, Kinasewitz G, Kurosawa S. Sepsis and pathophysiology of anthrax in a nonhuman primate model. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 169:433-44. [PMID: 16877346 PMCID: PMC1698797 DOI: 10.2353/ajpath.2006.051330] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/04/2006] [Indexed: 12/22/2022]
Abstract
Studies that define natural responses to bacterial sepsis assumed new relevance after the lethal bioterrorist attacks with Bacillus anthracis (anthrax), a spore-forming, toxigenic gram-positive bacillus. Considerable effort has focused on identifying adjunctive therapeutics and vaccines to prevent future deaths, but translation of promising compounds into the clinical setting necessitates an animal model that recapitulates responses observed in humans. Here we describe a nonhuman primate (Papio c. cynocephalus) model of B. anthracis infection using infusion of toxigenic B. anthracis Sterne 34F2 bacteria (5 x 10(5) to 6.5 x 10(9) CFU/kg). Similar to that seen in human patients, we observed changes in vascular permeability, disseminated intravascular coagulation, and systemic inflammation. The lung was a primary target organ with serosanguinous pleural effusions, intra-alveolar edema, and hemorrhagic lesions. This animal model reveals that a fatal outcome is dominated by the host septic response, thereby providing important insights into approaches for treatment and prevention of anthrax in humans.
Collapse
Affiliation(s)
- Deborah J Stearns-Kurosawa
- Department of Free Radical Biology and Aging Research, Oklahoma Medical Research Foundation, 825 NE 13th St., Oklahoma City, OK 73104, USA
| | | | | | | | | |
Collapse
|
35
|
Genth-Zotz S, von Haehling S, Bolger AP, Kalra PR, Wensel R, Coats AJS, Volk HD, Anker SD. The anti-CD14 antibody IC14 suppresses ex vivo endotoxin stimulated tumor necrosis factor-alpha in patients with chronic heart failure. Eur J Heart Fail 2006; 8:366-72. [PMID: 16337191 DOI: 10.1016/j.ejheart.2005.10.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2004] [Revised: 08/02/2005] [Accepted: 10/13/2005] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Activation of the endotoxin (LPS) receptor, CD14, leads to tumor necrosis factor-alpha (TNF) production. Plasma LPS activity is elevated in patients with severe chronic heart failure (CHF). An anti-CD14 antibody, IC14, blocks TNF production in healthy volunteers. It is not known whether IC14 prevents TNF production in CHF patients. METHODS AND RESULTS Blood from 20 CHF patients (age 64+/-2.1 years, NYHA class 2.2+/-0.1, LVEF 27+/-3%, mean+/-SEM) was pre-incubated with 0.5, 1.0, 5.0, 10 and 50 microg/mL IC14 for 1 h followed by incubation with 1 or 10 ng/mL LPS for 6 h. Fourteen subjects served as controls (58+/-2.4 years). LPS-stimulated TNF release was 76% and 60% greater at 1 and 10 ng/mL LPS, respectively, in CHF patients versus controls (p=0.07 and p=0.008). IC14 at concentrations of 5.0, 10 and 50 microg/mL substantially reduced TNF production in response to stimulation with LPS (all p<0.05). CD14 receptor density was similar in patients and controls. In controls, but not in CHF patients, there was a positive correlation between CD14 receptor density and TNF production (r=0.61, p=0.03). CONCLUSION IC14 suppresses LPS-stimulated whole blood TNF production in patients with CHF and in normal subjects and therefore may represent a novel therapeutic strategy for CHF patients with systemic immune activation.
Collapse
Affiliation(s)
- Sabine Genth-Zotz
- Clinical Cardiology, National Heart and Lung Institute, Imperial College School of Medicine, London, UK.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Innate immunity is a primordial system that has a primary role in lung antimicrobial defenses. Recent advances in understanding the recognition systems by which cells of the innate immune system recognize and respond to microbial products have revolutionized the understanding of host defenses in the lungs and other tissues. The innate immune system includes lung leukocytes and also epithelial cells lining the alveolar surface and the conducting airways. The innate immune system drives adaptive immunity in the lungs and has important interactions with other systems, including apoptosis pathways and signaling pathways induced by mechanical stretch. Human diversity in innate immune responses could explain some of the variability seen in the responses of patients to bacterial, fungal, and viral infections in the lungs. New strategies to modify innate immune responses could be useful in limiting the adverse consequences of some inflammatory reactions in the lungs.
Collapse
Affiliation(s)
- Thomas R Martin
- Pulmonary Research Laboratories, VA Puget Sound Health Care System, 151L, 1660 South Columbian Way, Seattle, WA 98108, USA.
| | | |
Collapse
|
37
|
Jeyaseelan S, Manzer R, Young SK, Yamamoto M, Akira S, Mason RJ, Worthen GS. Toll-IL-1 Receptor Domain-Containing Adaptor Protein Is Critical for Early Lung Immune Responses against Escherichia coli Lipopolysaccharide and Viable Escherichia coli. THE JOURNAL OF IMMUNOLOGY 2005; 175:7484-95. [PMID: 16301656 DOI: 10.4049/jimmunol.175.11.7484] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pulmonary bacterial diseases are a leading cause of mortality in the U.S. Innate immune response is vital for bacterial clearance from the lung, and TLRs play a critical role in this process. Toll-IL-1R domain-containing adaptor protein (TIRAP) is a key molecule in the TLR4 and 2 signaling. Despite its potential importance, the role of TIRAP-mediated signaling in lung responses has not been examined. Our goals were to determine the role of TIRAP-dependent signaling in the induction of lung innate immune responses against Escherichia coli LPS and viable E. coli, and in lung defense against E. coli in mice. LPS-induced neutrophil sequestration; NF-kappaB translocation; keratinocyte cell-derived chemokine, MIP-2, TNF-alpha, and IL-6 expression; histopathology; and VCAM-1 and ICAM-1 expression were abolished in the lungs of TIRAP-/- mice. A cell-permeable TIRAP blocking peptide attenuated LPS-induced lung responses. Furthermore, immune responses in the lungs of TIRAP-/- mice were attenuated against E. coli compared with TIRAP+/+ mice. TIRAP-/- mice also had early mortality, higher bacterial burden in the lungs, and more bacterial dissemination following E. coli inoculation. Moreover, we used human alveolar macrophages to examine the role of TIRAP signaling in the human system. The TIRAP blocking peptide abolished LPS-induced TNF-alpha, IL-6, and IL-8 expression in alveolar macrophages, whereas it attenuated E. coli-induced expression of these cytokines and chemokines. Taken together, this is the first study illustrating the crucial role of TIRAP in the generation of an effective early immune response against E. coli LPS and viable E. coli, and in lung defense against a bacterial pathogen.
Collapse
Affiliation(s)
- Samithamby Jeyaseelan
- Division of Respiratory Infections, Department of Medicine, National Jewish Medical and Research Center, Denver, CO 80206, USA.
| | | | | | | | | | | | | |
Collapse
|
38
|
Van Gucht S, Van Reeth K, Nauwynck H, Pensaert M. Porcine reproductive and respiratory syndrome virus infection increases CD14 expression and lipopolysaccharide-binding protein in the lungs of pigs. Viral Immunol 2005; 18:116-26. [PMID: 15802956 DOI: 10.1089/vim.2005.18.116] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is a respiratory virus of swine that plays an important role in multifactorial respiratory disease. European strains of PRRSV cause mild or no respiratory signs on their own, but can sensitize the lungs for the production of proinflammatory cytokines and respiratory signs upon exposure to bacterial lipopolysaccharides (LPS). The inflammatory effect of LPS depends on the binding to the LPS receptor complex. Therefore, we quantified the levels of CD14 expression and LPS-binding protein (LBP) in the lungs of pigs throughout a PRRSV infection. Twenty-four gnotobiotic pigs were inoculated intranasally with PRRSV (10(6) 50% tissue culture infectious doses per pig, Lelystad strain) or phosphate-buffered saline (PBS), and euthanized 1-52 days later. Lungs were examined for CD14 expression (immunofluorescence and image analysis), LBP (ELISA), and virus replication. PRRSV infection caused a clear increase of CD14 expression from 3 to 40 days post-inoculation (DPI) and LBP from 7 to 14 DPI. Both parameters peaked at 9-10 DPI (40 and 14 times higher than PBS control pigs, respectively) and were correlated tightly with virus replication in the lungs. Double immunofluorescence labelings demonstrated that resident macrophages expressed little CD14 and that the increase of CD14 expression in the PRRSV-infected lungs was probably due to infiltration of highly CD14-positive monocytes in the interstitium. As both CD14 and LBP potentiate the inflammatory effects of LPS, their increase in the lungs could explain why PRRSV sensitizes the lungs for the production of proinflammatory cytokines and respiratory signs upon exposure to LPS.
Collapse
Affiliation(s)
- Steven Van Gucht
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, B-9820 Merelbeke, Belgium
| | | | | | | |
Collapse
|
39
|
Martin AC, Laing IA, Zhang G, Brennan S, Winfield K, Sly PD, Stick SM, Goldblatt J, LeSouef PN. CD14 C-159T and early infection with Pseudomonas aeruginosa in children with cystic fibrosis. Respir Res 2005; 6:63. [PMID: 15975149 PMCID: PMC1168907 DOI: 10.1186/1465-9921-6-63] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2004] [Accepted: 06/23/2005] [Indexed: 11/10/2022] Open
Abstract
Early acquisition of Pseudomonas aeruginosa is associated with a poorer prognosis in patients with cystic fibrosis. We investigated whether polymorphisms in CD14, the lipopolysaccharide receptor, increase the risk of early infection. Forty-five children with cystic fibrosis were investigated with annual bronchoalveolar lavage (BAL) and plasma sCD14 levels. Plasma sCD14 levels were significantly lower in children from whom P.aeruginosa was subsequently isolated (492.75 microg/ml vs. 1339.43 microg/ml, p = 0.018). Those with the CD14 -159CC genotype had a significantly increased risk of early infection with P.aeruginosa suggesting that CD14 C-159T plays a role in determining the risk of early infection with P.aeruginosa.
Collapse
Affiliation(s)
- AC Martin
- School of Paediatrics and Child Health, University of Western Australia, Perth, Western Australia 6001
- Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia 6008
| | - IA Laing
- School of Paediatrics and Child Health, University of Western Australia, Perth, Western Australia 6001
| | - G Zhang
- School of Paediatrics and Child Health, University of Western Australia, Perth, Western Australia 6001
| | - S Brennan
- Division of Clinical Science, Telethon Institute for Child Health Research, Perth, Western Australia 6008
| | - K Winfield
- Division of Clinical Science, Telethon Institute for Child Health Research, Perth, Western Australia 6008
| | - PD Sly
- Division of Clinical Science, Telethon Institute for Child Health Research, Perth, Western Australia 6008
- Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia 6008
| | - SM Stick
- Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia 6008
| | - J Goldblatt
- School of Paediatrics and Child Health, University of Western Australia, Perth, Western Australia 6001
| | - PN LeSouef
- School of Paediatrics and Child Health, University of Western Australia, Perth, Western Australia 6001
- Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia 6008
| |
Collapse
|
40
|
Echchannaoui H, Frei K, Letiembre M, Strieter RM, Adachi Y, Landmann R. CD14 deficiency leads to increased MIP-2 production, CXCR2 expression, neutrophil transmigration, and early death in pneumococcal infection. J Leukoc Biol 2005; 78:705-15. [PMID: 15941778 DOI: 10.1189/jlb.0205063] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
CD14 is a myeloid receptor for bacterial cell membrane/wall components, for which we previously showed a strong induction in cerebrospinal fluid (CSF) during meningitis. Here, we studied CD14 function in murine Streptococcus pneumoniae meningitis by using wild-type (WT), CD14(-/-) mice, and WT mice pretreated with neutralizing anti-CD14 antibodies. Early polymorphonuclear leukocytes (PMN) immigration was more pronounced in CSF of CD14(-/-) than of WT mice. This was not a result of altered adherence molecule expression in blood and CSF PMN or brain endothelial cells. Macrophage inflammatory protein-2 (MIP-2) and keratinocyte-derived chemokine levels were similar in CSF in both strains, but MIP-2 was higher in infected brain and in brain-derived endothelial cells infected in vitro in CD14(-/-) than in WT mice. CD14(-/-) PMN demonstrated increased expression of CXC chemokine receptor 2 (CXCR2) after infection and stronger in vitro chemotaxis than WT PMN toward CSF from WT or CD14(-/-) mice and toward MIP-2. Excess PMN migration in CD14(-/-) mice did not result in improved bacterial clearing but in increased tumor necrosis factor in CSF, higher disease severity, and earlier death. Pretreatment with anti-CXCR2 reduced PMN infiltration into CSF and brain MIP-2 production and abolished earlier mortality in CD14(-/-) mice. In conclusion, CD14 plays a protective role in pneumococcal meningitis by slowing PMN migration via MIP-2 and CXCR2 modulation.
Collapse
Affiliation(s)
- Hakim Echchannaoui
- Department of Research, University Hospital, Hebelstrasse 20, CH-4031 Basel, Switzerland
| | | | | | | | | | | |
Collapse
|
41
|
Benhnia MREI, Wroblewski D, Akhtar MN, Patel RA, Lavezzi W, Gangloff SC, Goyert SM, Caimano MJ, Radolf JD, Sellati TJ. Signaling through CD14 attenuates the inflammatory response to Borrelia burgdorferi, the agent of Lyme disease. THE JOURNAL OF IMMUNOLOGY 2005; 174:1539-48. [PMID: 15661914 DOI: 10.4049/jimmunol.174.3.1539] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Lyme disease is a chronic inflammatory disorder caused by the spirochetal bacterium, Borrelia burgdorferi. In vitro evidence suggests that binding of spirochetal lipoproteins to CD14, a pattern recognition receptor expressed on monocytes/macrophages and polymorphonuclear cells, is a critical requirement for cellular activation and the subsequent release of proinflammatory cytokines that most likely contribute to symptomatology and clinical manifestations. To test the validity of this notion, we assessed the impact of CD14 deficiency on Lyme disease in C3H/HeN mice. Contrary to an anticipated diminution in pathology, CD14(-/-) mice exhibited more severe and persistent inflammation than did CD14(+/+) mice. This disparity reflects altered gene regulation within immune cells that may engender the higher bacterial burden and serum cytokine levels observed in CD14(-/-) mice. Comparing their in vitro stimulatory activity, live spirochetes, but not lysed organisms, were a potent CD14-independent stimulus of cytokine production, triggering an exaggerated response by CD14(-/-) macrophages. Collectively, our in vivo and in vitro findings support the provocative notion that: 1) pattern recognition by CD14 is entirely dispensable for elaboration of an inflammatory response to B. burgdorferi, and 2) CD14-independent signaling pathways are inherently more destructive than CD14-dependent pathways. Continued study of CD14-independent signaling pathways may provide mechanistic insight into the inflammatory processes that underlie development of chronic inflammation.
Collapse
|
42
|
Sutherland AM, Walley KR, Russell JA. Polymorphisms in CD14, mannose-binding lectin, and Toll-like receptor-2 are associated with increased prevalence of infection in critically ill adults*. Crit Care Med 2005; 33:638-44. [PMID: 15753758 DOI: 10.1097/01.ccm.0000156242.44356.c5] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVE To test for the association of single nucleotide polymorphisms of the innate immunity receptors cluster of differentiation (CD)-14, mannose-binding lectin, and Toll-like receptor-2 with clinical phenotype in critically ill patients with systemic inflammatory response syndrome. DESIGN Genetic association study. SETTING Tertiary care mixed medical-surgery intensive care unit at St. Paul's Hospital, Vancouver, BC, a teaching hospital associated with the University of British Columbia. PATIENTS A cohort of 252 critically ill Caucasians with systemic inflammatory response syndrome. INTERVENTIONS DNA was extracted from discarded blood. Clinical data were gathered by retrospective chart review. MEASUREMENTS AND MAIN RESULTS C-159T CD14, the X/Y and B, C, and D polymorphisms of mannose-binding lectin, and T-16933A Toll-like receptor-2 were genotyped using polymerase chain reaction-restriction fragment length polymorphism. We tested for association of genotype with prevalence of positive bacterial cultures, type of organism (Gram-positive, Gram-negative, other), sepsis and septic shock at admission to the intensive care unit, and 28-day survival. CD14 -159TT was associated with increased prevalence of positive bacterial cultures and with Gram-negative bacteria. Mannose-binding lectin haplotype pairs XO/O and O/O were also associated with increased prevalence of positive bacterial cultures but not with a specific organism class. Toll-like receptor-2 -16933AA was associated with increased prevalence of sepsis and with Gram-positive bacteria. In contrast, the polymorphisms were not associated with increased prevalence of septic shock or altered 28-day survival. CONCLUSIONS Single nucleotide polymorphisms in CD14, mannose-binding lectin, and Toll-like receptor-2 are associated with increased prevalence of positive bacterial cultures and sepsis but not with altered prevalence of septic shock or decreased 28-day survival. Furthermore, CD14 single nucleotide polymorphisms were associated with Gram-negative bacteria and Toll-like receptor-2 with Gram-positive bacteria, whereas mannose-binding lectin was not associated with a particular organism class. Thus, single nucleotide polymorphisms in innate immunity receptors may alter recognition and clearance of bacteria without changing outcomes of critically ill adults with systemic inflammatory response syndrome.
Collapse
Affiliation(s)
- Ainsley M Sutherland
- University of British Columbia Critical Care Research Laboratories, James Hogg iCAPTURE Centre/St. Paul's Hospital, Vancouver, BC, Canada
| | | | | |
Collapse
|
43
|
Sinclair SE, Altemeier WA, Matute-Bello G, Chi EY. Augmented lung injury due to interaction between hyperoxia and mechanical ventilation*. Crit Care Med 2004; 32:2496-501. [PMID: 15599157 DOI: 10.1097/01.ccm.0000148231.04642.8d] [Citation(s) in RCA: 153] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Mechanical overdistension and hyperoxia can independently cause lung injury, yet little is known about their combined effects. We hypothesized that hyperoxia exacerbates lung injury caused by large tidal volume ventilation. DESIGN Experimental study. SETTING University laboratory. SUBJECTS Anesthetized, paralyzed rabbits. INTERVENTIONS In experiment 1, 12 rabbits were ventilated with 25 mL/kg tidal volumes at positive end-expiratory pressure of 0 cm H2O for 4 hrs with either hyperoxia (HO; FiO2 = 0.5) or normoxia (NO; FiO2 = 0.21). In experiment 2, a separate group of animals were randomized to one of four groups to assess the interaction of tidal volume and inspired oxygen concentration on potential mediators of injury after 2 hrs of ventilation, before significant injury occurs: a) NO+normal tidal volume (NV; VT = 10 mL/kg); b) HO+NV; c) NO+high tidal volume (HV; VT = 25 mL/kg); d) HO+HV (n = 3 per group). MEASUREMENTS AND MAIN RESULTS : In the first study, HO compared with the NO group had significantly reduced PaO2/FiO2 ratio (320 +/- 110 vs. 498 +/- 98, p = .014) and increased lung injury scores at 4 hrs. Hyperoxia also significantly increased polymorphonuclear leukocytes, growth-related oncogene-alpha (2073 +/- 535 vs. 463 +/- 236 pg/mL, p = .02), and monocyte chemotactic protein-1 (7517 +/- 1612 vs. 2983 +/- 1289 pg/mL, p = .05) concentrations in bronchoalveolar lavage fluid. The second study showed increased alveolar-capillary permeability to a 70-kD fluorescent-labeled dextran only in response to the combination of both HO and HV. Chemokines and bronchoalveolar lavage fluid neutrophils were elevated in both HV groups; however, hyperoxia did not further increase chemokine or neutrophil counts over normoxia. No difference in lipid peroxidation was seen between groups. CONCLUSIONS Moderate hyperoxia exacerbates lung injury in a large tidal volume model of ventilator-induced lung injury. The mechanism by which this occurs is not mediated by increased production of CXC chemokines or lipid peroxidation.
Collapse
Affiliation(s)
- Scott E Sinclair
- Departments of Medicine and Physiology, University of Tennessee, Memphis, TN 38163, USA.
| | | | | | | |
Collapse
|
44
|
Kajikawa O, Frevert CW, Lin SM, Goodman RB, Mongovin SM, Wong V, Ballman K, Daubeuf B, Elson G, Martin TR. Gene expression of Toll-like receptor-2, Toll-like receptor-4, and MD2 is differentially regulated in rabbits with Escherichia coli pneumonia. Gene 2004; 344:193-202. [PMID: 15656985 DOI: 10.1016/j.gene.2004.09.032] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2004] [Revised: 09/01/2004] [Accepted: 09/23/2004] [Indexed: 02/04/2023]
Abstract
Sepsis, a common sequela to Gram-negative pneumonia, results in considerable morbidity and mortality in hospitalized patients. The goal of this study was to determine whether Gram-negative pneumonia alters the expression TLR2, TLR4, and MD2 in lungs or in organs distant to the site of the primary infection. The cDNA sequence coding open reading frames for rabbit TLR2, TLR4, and MD2 were cloned and expressed in Escherichia coli, and specific polyclonal antibodies and polymerase chain reaction (PCR) probes were produced to identify changes in these receptors in rabbits with Gram-negative pneumonia. Using tissues from lungs and distant organs, we show that TLR2, TLR4, and MD2 gene expression is differentially regulated in rabbits with E. coli pneumonia. The increased expression of TLR2 and TLR4 could play an important role in the innate immune response to bacterial infection in the lungs, and improve pathogen recognition and bacterial clearance. In contrast, the increased gene expression of TLR2, TLR4, and MD2 in organs distant to the primary site of infection may contribute to the deleterious systemic inflammatory response observed in patients with sepsis.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, Surface/genetics
- Antigens, Surface/metabolism
- Base Sequence
- Cloning, Molecular
- DNA, Complementary/chemistry
- DNA, Complementary/genetics
- Escherichia coli Infections/genetics
- Gene Expression Profiling
- Immunohistochemistry
- Lung/metabolism
- Lung/microbiology
- Lung/pathology
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Molecular Sequence Data
- Pneumonia, Bacterial/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rabbits
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sequence Analysis, DNA
- Toll-Like Receptor 2
- Toll-Like Receptor 4
- Toll-Like Receptors
Collapse
Affiliation(s)
- Osamu Kajikawa
- Pulmonary Research Laboratories at the Seattle Department of Veterans Affairs Medical Center, University of Washington School of Medicine, 151L 1660 South Columbian Way, Seattle, WA 98108, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
|
46
|
Reinhart K, Glück T, Ligtenberg J, Tschaikowsky K, Bruining A, Bakker J, Opal S, Moldawer LL, Axtelle T, Turner T, Souza S, Pribble J. CD14 receptor occupancy in severe sepsis: Results of a phase I clinical trial with a recombinant chimeric CD14 monoclonal antibody (IC14)*. Crit Care Med 2004; 32:1100-8. [PMID: 15190957 DOI: 10.1097/01.ccm.0000124870.42312.c4] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Binding of bacterial cell wall components to CD14 and co-receptors on myeloid cells results in cellular activation and production of proinflammatory mediators. A recombinant anti-CD14 monoclonal antibody (IC14) has been shown to decrease lipopolysaccharide-induced responses in animal and human models of endotoxemia. This study was performed to evaluate the safety, pharmacokinetics, pharmacodynamics, and clinical pharmacology of IC14 in patients with severe sepsis. DESIGN Randomized, double-blind, placebo-controlled, dose-ranging, multiple-center trial. SETTING Six medical and surgical intensive care units located in Germany and The Netherlands. PATIENTS Forty patients with severe sepsis. INTERVENTIONS IC14 was administered intravenously to eight patients/cohort as single (1 mg/kg or 4 mg/kg) or multiple doses (4 mg/kg daily for 4 days, or 4 mg/kg on day 1 followed by 2 mg/kg daily for 3 days). A placebo group (two patients/cohort) was also included. MEASUREMENTS AND MAIN RESULTS The overall incidence and types of adverse events were similar among treatment groups. One patient in the group receiving multiple-dose IC14 4 mg/kg daily for 4 days experienced an anaphylactic reaction after receiving the first dose of study drug. IC14 did not induce antibody formation or increase the incidence of secondary bacterial infection. A mean IC14 serum concentration of approximately 1 microg/mL was required to achieve 50% of maximum membrane-bound CD14 receptor occupancy on peripheral blood monocytes. The pattern of proinflammatory and anti-inflammatory cytokines, chemokine, soluble receptor, soluble E-selectin, and acute phase proteins in response to treatment was highly variable by patient and IC14 treatment group. CONCLUSIONS Single and multiple doses of IC14 were generally well tolerated and did not induce antibody formation or increase the incidence of secondary bacterial infection. The results suggest that CD14 blockade with IC14 warrants further clinical investigation to determine its ability to attenuate the proinflammatory response due to infection.
Collapse
Affiliation(s)
- Konrad Reinhart
- Department of Anesthesia and Critical Care, University Medical Center, University of Jena, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lin SM, Frevert CW, Kajikawa O, Wurfel MM, Ballman K, Mongovin S, Wong VA, Selk A, Martin TR. Differential regulation of membrane CD14 expression and endotoxin-tolerance in alveolar macrophages. Am J Respir Cell Mol Biol 2004; 31:162-70. [PMID: 15059784 PMCID: PMC4096031 DOI: 10.1165/rcmb.2003-0307oc] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
CD14 is important in the clearance of bacterial pathogens from lungs. However, the mechanisms that regulate the expression of membrane CD14 (mCD14) on alveolar macrophages (AM) have not been studied in detail. This study examines the regulation of mCD14 on AM exposed to Escherichia coli in vivo and in vitro, and explores the consequences of changes in mCD14 expression. The expression of mCD14 was decreased on AM exposed to E. coli in vivo and AM incubated with lipopolysaccharide (LPS) or E. coli in vitro. Polymyxin B abolished LPS effects, but only partially blocked the effects of E. coli. Blockade of extracellular signal-regulated kinase pathways attenuated LPS and E. coli-induced decrease in mCD14 expression. Inhibition of proteases abrogated the LPS-induced decrease in mCD14 expression on AM and the release of sCD14 into the supernatants, but did not affect the response to E. coli. The production of tumor necrosis factor-alpha in response to a second challenge with Staphylococcus aureus or zymosan was decreased in AM after incubation with E. coli but not LPS. These studies show that distinct mechanisms regulate the expression of mCD14 and the induction of endotoxin tolerance in AM, and suggest that AM function is impaired at sites of bacterial infection.
Collapse
Affiliation(s)
- Shu-Min Lin
- Pulmonary Research Laboratories at the VA Puget Sound Medical Center, and the Division of Pulmonary/Critical Care Medicine, Department of Medicine; University of Washington School of Medicine, Seattle, WA, U.S.A
- Department of Thoracic Medicine II, Chang Gung Memorial Hospital, Taipei, Taiwan Supported in part by NIH grants GM37696, HL30542
| | - Charles W. Frevert
- Pulmonary Research Laboratories at the VA Puget Sound Medical Center, and the Division of Pulmonary/Critical Care Medicine, Department of Medicine; University of Washington School of Medicine, Seattle, WA, U.S.A
| | - Osamu Kajikawa
- Pulmonary Research Laboratories at the VA Puget Sound Medical Center, and the Division of Pulmonary/Critical Care Medicine, Department of Medicine; University of Washington School of Medicine, Seattle, WA, U.S.A
| | - Mark M. Wurfel
- Pulmonary Research Laboratories at the VA Puget Sound Medical Center, and the Division of Pulmonary/Critical Care Medicine, Department of Medicine; University of Washington School of Medicine, Seattle, WA, U.S.A
| | - Kimberly Ballman
- Pulmonary Research Laboratories at the VA Puget Sound Medical Center, and the Division of Pulmonary/Critical Care Medicine, Department of Medicine; University of Washington School of Medicine, Seattle, WA, U.S.A
| | - Stephen Mongovin
- Pulmonary Research Laboratories at the VA Puget Sound Medical Center, and the Division of Pulmonary/Critical Care Medicine, Department of Medicine; University of Washington School of Medicine, Seattle, WA, U.S.A
| | - Venus A. Wong
- Pulmonary Research Laboratories at the VA Puget Sound Medical Center, and the Division of Pulmonary/Critical Care Medicine, Department of Medicine; University of Washington School of Medicine, Seattle, WA, U.S.A
| | - Amy Selk
- Pulmonary Research Laboratories at the VA Puget Sound Medical Center, and the Division of Pulmonary/Critical Care Medicine, Department of Medicine; University of Washington School of Medicine, Seattle, WA, U.S.A
| | - Thomas R. Martin
- Pulmonary Research Laboratories at the VA Puget Sound Medical Center, and the Division of Pulmonary/Critical Care Medicine, Department of Medicine; University of Washington School of Medicine, Seattle, WA, U.S.A
| |
Collapse
|
48
|
Rensing H. [Endotoxins. Pathogenetic meaning of sepsis]. Anaesthesist 2004; 52 Suppl 1:S7-S13. [PMID: 14727044 DOI: 10.1007/s00101-003-0587-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- H Rensing
- Klinik für Anästhesiologie und Intensivmedizin, Universitätskliniken des Saarlandes, Homburg/Saar.
| |
Collapse
|
49
|
Abstract
During the past 3 years new insights have been gained into the fundamental elements that underlie the pathogenesis of sepsis, and after years of frustrating failures, progress in the basic understanding of sepsis has translated into successful new therapies. These new treatment strategies have significantly improved the outcome of patients experiencing the puzzling syndrome of severe sepsis. More effective supportive therapies with early, goal-oriented therapy including volume resuscitation, catecholamine therapy and transfusion improve the chances for survival in septic shock. Novel endocrine management with hydrocortisone replacement therapy for relative adrenal insufficiency in septic shock patients and strict blood glucose control provide a survival advantage in critically ill patients. Administering appropriate antimicrobial therapy, nutritional support and ventilation protocols with low tidal volumes have now been shown to benefit septic patients. Finally, human recombinant activated protein C (drotrecogin alfa), which ameliorates sepsis-induced disseminated intravascular coagulation and exerts several other favourable effects on endothelial cells, has been shown to reduce mortality in patients with severe sepsis. On the basis of newly discovered pathophysiological mechanisms of sepsis, several other adjuvant therapies for sepsis are in various stages of preclinical and clinical development. Individualised and optimal supportive care with efforts to reverse the precipitating cause of sepsis remains the mainstay of therapy for severe sepsis. How these new and often expensive regimens will fit into the standard treatment approach to sepsis remains to be defined by future clinical investigations.
Collapse
Affiliation(s)
- Thomas Glück
- Klinik und Poliklinik für Innere Medizin I, Universitätsklinikum Regensburg, Regensburg, Germany
| | | |
Collapse
|
50
|
Abstract
Experimental models of pulmonary infection are being discussed, focused on various aspects of good experimental design, such as choice of animal species and infecting strain, and route of infection/inoculation techniques (intranasal inoculation, aerosol inoculation, and direct instillation into the lower respiratory tract). In addition, parameters to monitor pulmonary infection are being reviewed such as general clinical signs, pulmonary-associated signs, complication of the pulmonary infection, mortality rate, and parameters after dissection of animals. Examples of pulmonary infection models caused by bacteria, fungi, viruses or parasites in experimental animals with intact or impaired host defense mechanisms are shortly summarized including key-references.
Collapse
Affiliation(s)
- Irma A J M Bakker-Woudenberg
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, P.O. Box 1738, 3000 DR Rotterdam, The Netherlands.
| |
Collapse
|