51
|
Novakovic B, Habibi E, Wang SY, Arts RJW, Davar R, Megchelenbrink W, Kim B, Kuznetsova T, Kox M, Zwaag J, Matarese F, van Heeringen SJ, Janssen-Megens EM, Sharifi N, Wang C, Keramati F, Schoonenberg V, Flicek P, Clarke L, Pickkers P, Heath S, Gut I, Netea MG, Martens JHA, Logie C, Stunnenberg HG. β-Glucan Reverses the Epigenetic State of LPS-Induced Immunological Tolerance. Cell 2017; 167:1354-1368.e14. [PMID: 27863248 PMCID: PMC5927328 DOI: 10.1016/j.cell.2016.09.034] [Citation(s) in RCA: 461] [Impact Index Per Article: 57.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 08/05/2016] [Accepted: 09/20/2016] [Indexed: 12/21/2022]
Abstract
Innate immune memory is the phenomenon whereby innate immune cells such as monocytes or macrophages undergo functional reprogramming after exposure to microbial components such as lipopolysaccharide (LPS). We apply an integrated epigenomic approach to characterize the molecular events involved in LPS-induced tolerance in a time-dependent manner. Mechanistically, LPS-treated monocytes fail to accumulate active histone marks at promoter and enhancers of genes in the lipid metabolism and phagocytic pathways. Transcriptional inactivity in response to a second LPS exposure in tolerized macrophages is accompanied by failure to deposit active histone marks at promoters of tolerized genes. In contrast, β-glucan partially reverses the LPS-induced tolerance in vitro. Importantly, ex vivo β-glucan treatment of monocytes from volunteers with experimental endotoxemia re-instates their capacity for cytokine production. Tolerance is reversed at the level of distal element histone modification and transcriptional reactivation of otherwise unresponsive genes.
Collapse
Affiliation(s)
- Boris Novakovic
- Department of Molecular Biology, Faculty of Science, Radboud University, 6525 GA Nijmegen, the Netherlands
| | - Ehsan Habibi
- Department of Molecular Biology, Faculty of Science, Radboud University, 6525 GA Nijmegen, the Netherlands
| | - Shuang-Yin Wang
- Department of Molecular Biology, Faculty of Science, Radboud University, 6525 GA Nijmegen, the Netherlands
| | - Rob J W Arts
- Department of Internal Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), 6525 GA Nijmegen, the Netherlands
| | - Robab Davar
- Department of Molecular Biology, Faculty of Science, Radboud University, 6525 GA Nijmegen, the Netherlands
| | - Wout Megchelenbrink
- Department of Molecular Biology, Faculty of Science, Radboud University, 6525 GA Nijmegen, the Netherlands
| | - Bowon Kim
- Department of Molecular Biology, Faculty of Science, Radboud University, 6525 GA Nijmegen, the Netherlands
| | - Tatyana Kuznetsova
- Department of Molecular Biology, Faculty of Science, Radboud University, 6525 GA Nijmegen, the Netherlands
| | - Matthijs Kox
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), 6500 HB Nijmegen, the Netherlands
| | - Jelle Zwaag
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), 6500 HB Nijmegen, the Netherlands
| | - Filomena Matarese
- Department of Molecular Biology, Faculty of Science, Radboud University, 6525 GA Nijmegen, the Netherlands
| | - Simon J van Heeringen
- Department of Molecular Developmental Biology, Faculty of Science, Radboud University, 6525 GA Nijmegen, the Netherlands
| | - Eva M Janssen-Megens
- Department of Molecular Biology, Faculty of Science, Radboud University, 6525 GA Nijmegen, the Netherlands
| | - Nilofar Sharifi
- Department of Molecular Biology, Faculty of Science, Radboud University, 6525 GA Nijmegen, the Netherlands
| | - Cheng Wang
- Department of Molecular Biology, Faculty of Science, Radboud University, 6525 GA Nijmegen, the Netherlands
| | - Farid Keramati
- Department of Molecular Biology, Faculty of Science, Radboud University, 6525 GA Nijmegen, the Netherlands
| | - Vivien Schoonenberg
- Department of Molecular Biology, Faculty of Science, Radboud University, 6525 GA Nijmegen, the Netherlands
| | - Paul Flicek
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Laura Clarke
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), 6500 HB Nijmegen, the Netherlands
| | - Simon Heath
- Centro Nacional de Análisis Genómico (CNAG), Parc Científic de Barcelona, 08028 Barcelona, Spain
| | - Ivo Gut
- Centro Nacional de Análisis Genómico (CNAG), Parc Científic de Barcelona, 08028 Barcelona, Spain
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Center, Radboud Center for Infectious Diseases (RCI), 6525 GA Nijmegen, the Netherlands
| | - Joost H A Martens
- Department of Molecular Biology, Faculty of Science, Radboud University, 6525 GA Nijmegen, the Netherlands
| | - Colin Logie
- Department of Molecular Biology, Faculty of Science, Radboud University, 6525 GA Nijmegen, the Netherlands
| | - Hendrik G Stunnenberg
- Department of Molecular Biology, Faculty of Science, Radboud University, 6525 GA Nijmegen, the Netherlands.
| |
Collapse
|
52
|
Kiers D, Koch RM, Hamers L, Gerretsen J, Thijs EJM, van Ede L, Riksen NP, Kox M, Pickkers P. Characterization of a model of systemic inflammation in humans in vivo elicited by continuous infusion of endotoxin. Sci Rep 2017; 7:40149. [PMID: 28054645 PMCID: PMC5215288 DOI: 10.1038/srep40149] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 12/01/2016] [Indexed: 01/06/2023] Open
Abstract
Investigating the systemic inflammatory response in patients with critical illness such as sepsis, trauma and burns is complicated due to uncertainties about the onset, duration and severity of the insult. Therefore, in vivo models of inflammation are essential to study the pathophysiology and to evaluate immunomodulatory therapies. Intravenous bolus administration of endotoxin to healthy volunteers is a well-established model of a short-lived systemic inflammatory response, characterized by increased plasma cytokine levels, flu-like symptoms and fever. In contrast, patients suffering from systemic inflammation are often exposed to inflammatory stimuli for an extended period of time. Therefore, continuous infusion of endotoxin may better reflect the kinetics of the inflammatory response encountered in these patients. Herein, we characterize a novel model of systemic inflammation elicited by a bolus infusion of 1 ng/kg, followed by a 3hr continuous infusion of 1 ng/kg/h of endotoxin in healthy volunteers, and compared it with models of bolus administrations of 1 and 2 ng/kg of endotoxin. The novel model was well-tolerated and resulted in a more pronounced increase in plasma cytokine levels with different kinetics and more prolonged symptoms and fever compared with the bolus-only models. Therefore, the continuous endotoxin infusion model provides novel insights into kinetics of the inflammatory response during continuous inflammatory stimuli and accommodates a larger time window to evaluate immunomodulating therapies.
Collapse
Affiliation(s)
- D Kiers
- Radboud university medical center, 6500 HB, Radboud Institute for Molecular Life Sciences, Department of Intensive Care Medicine, 6500 HB, Nijmegen, The Netherlands.,Radboud university medical center, 6500 HB, Radboud Institute for Molecular Life Sciences, Department of Pediatrics, 6500 HB, Nijmegen, The Netherlands
| | - R M Koch
- Radboud university medical center, 6500 HB, Radboud Institute for Molecular Life Sciences, Department of Intensive Care Medicine, 6500 HB, Nijmegen, The Netherlands.,Radboud university medical center, 6500 HB, Radboud Institute for Molecular Life Sciences, Department of Pediatrics, 6500 HB, Nijmegen, The Netherlands
| | - L Hamers
- Radboud university medical center, 6500 HB, Radboud Institute for Molecular Life Sciences, Department of Intensive Care Medicine, 6500 HB, Nijmegen, The Netherlands.,Radboud university medical center, 6500 HB, Radboud Institute for Molecular Life Sciences, Department of Pediatrics, 6500 HB, Nijmegen, The Netherlands
| | - J Gerretsen
- Radboud university medical center, 6500 HB, Radboud Institute for Molecular Life Sciences, Department of Intensive Care Medicine, 6500 HB, Nijmegen, The Netherlands.,Radboud university medical center, 6500 HB, Radboud Institute for Molecular Life Sciences, Department of Pediatrics, 6500 HB, Nijmegen, The Netherlands
| | - E J M Thijs
- Radboud university medical center, 6500 HB, Radboud Institute for Molecular Life Sciences, Department of Intensive Care Medicine, 6500 HB, Nijmegen, The Netherlands
| | - L van Ede
- Radboud university medical center, 6500 HB, Radboud Institute for Molecular Life Sciences, Department of Intensive Care Medicine, 6500 HB, Nijmegen, The Netherlands
| | - N P Riksen
- Radboud university medical center, 6500 HB, Radboud Institute for Molecular Life Sciences, Department of Internal Medicine, 6500 HB, Nijmegen, The Netherlands
| | - M Kox
- Radboud university medical center, 6500 HB, Radboud Institute for Molecular Life Sciences, Department of Intensive Care Medicine, 6500 HB, Nijmegen, The Netherlands.,Radboud university medical center, 6500 HB, Radboud Institute for Molecular Life Sciences, Department of Pediatrics, 6500 HB, Nijmegen, The Netherlands
| | - P Pickkers
- Radboud university medical center, 6500 HB, Radboud Institute for Molecular Life Sciences, Department of Intensive Care Medicine, 6500 HB, Nijmegen, The Netherlands.,Radboud university medical center, 6500 HB, Radboud Institute for Molecular Life Sciences, Department of Pediatrics, 6500 HB, Nijmegen, The Netherlands
| |
Collapse
|
53
|
Immune dysfunction in patients with obstructive jaundice before and after endoscopic retrograde cholangiopancreatography. Clin Sci (Lond) 2016; 130:1535-44. [PMID: 27252406 PMCID: PMC4947925 DOI: 10.1042/cs20160326] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 06/01/2016] [Indexed: 02/07/2023]
Abstract
This prospective observational study investigated monocyte cytokine responses to lipopolysaccharide (LPS) in patients with obstructive jaundice (OJ) before and after endoscopic biliary drainage. Dendritic cell (DC) subsets and their expression of co-stimulatory molecules were also studied. Forty patients with OJ and ten non-jaundiced patients with normal gastroscopy findings were recruited. Ten healthy volunteers provided control blood samples for immunological assays. Patients with OJ had blood and duodenal mucosa sampled at the time of endoscopic retrograde cholangiopancreatography (ERCP) and further blood sampled during the recovery phase. Monocyte cytokine responses to LPS, DC subsets and co-stimulatory molecule expression were compared with controls. Duodenal morphology and occludin expression were also assessed. Monocytes obtained before ERCP from jaundiced patients demonstrated reduced cytokine responses to endotoxin compared with controls (IL-1β: 2678 compared with 4631 pg/ml, P=0.04 and IL-6: 3442 compared with 6157 pg/ml, P=0.002). Monocytes from patients with malignancy had poorer responses to endotoxin than from those with benign OJ (IL-1β: 2025 compared with 3332 pg/ml, P=0.001). After ERCP, the secretion of inflammatory cytokines by monocytes obtained from jaundiced patients increased (IL-1β: 2150 compared with 2520 pg/ml, P=0.03 and IL-6: 2488 compared with 3250 pg/ml, P=0.01). Occludin expression (85 compared with 95%, P=0.004) and mean duodenal villus height (334 compared with 404 μm, P=0.03) were lower in jaundiced patients. Before biliary drainage, patients with OJ had a higher percentage of myeloid dendritic cells (mDCs) and greater mDC expression of CD40 (P=0.04) and CD86 (P=0.04). Monocytes from patients with OJ had lower proinflammatory cytokine secretion in response to LPS, an effect reversed following biliary drainage.
Collapse
|
54
|
Fullerton JN, Segre E, De Maeyer RPH, Maini AAN, Gilroy DW. Intravenous Endotoxin Challenge in Healthy Humans: An Experimental Platform to Investigate and Modulate Systemic Inflammation. J Vis Exp 2016. [PMID: 27213711 PMCID: PMC4942172 DOI: 10.3791/53913] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Activation of inflammatory pathways represents a central mechanism in multiple disease states both acute and chronic. Triggered via either pathogen or tissue damage-associated molecular motifs, common biochemical pathways lead to conserved yet variable physiological and immunological alterations. Dissection and delineation of the determinants and mechanisms underlying phenotypic variance in response is expected to yield novel therapeutic advances. Intravenous (IV) administration of endotoxin (gram-negative bacterial lipopolysaccharide), a specific Toll-like receptor 4 agonist, represents an in vivo model of systemic inflammation in man. National Institutes for Health Clinical Center Reference Endotoxin (CCRE, Escherichia coli O:113:H10:K negative) is employed to reliably and reproducibly generate vascular, hematological, endocrine, immunological and organ-specific functional effects that parallel, to varying degrees, those seen in the early stages of pathological states. Alteration of dose (0.06 - 4 ng/kg) and time-scale of exposure (bolus vs. infusion) allows replication of either acute or chronic inflammation and a range of severity to be elicited, with higher doses (2 - 4 ng/kg) frequently being used to create a 'sepsis-like' state. Established and novel medicinal compounds may additionally be administered prior to or post endotoxin exposure to appreciate their effect on the inflammatory cascade. Despite limitations in scope and generalizability, human IV endotoxin challenge offers a unique platform to gain mechanistic insights into inducible physiological responses and inflammatory pathways. Rationally employed it may aid translation of this knowledge into therapeutic innovations.
Collapse
Affiliation(s)
- James N Fullerton
- Centre for Clinical Pharmacology, Division of Medicine, University College London;
| | - Elisabetta Segre
- Centre for Clinical Pharmacology, Division of Medicine, University College London
| | - Roel P H De Maeyer
- Centre for Clinical Pharmacology, Division of Medicine, University College London
| | - Alexander A N Maini
- Centre for Clinical Pharmacology, Division of Medicine, University College London
| | - Derek W Gilroy
- Centre for Clinical Pharmacology, Division of Medicine, University College London
| |
Collapse
|
55
|
Boutagy NE, McMillan RP, Frisard MI, Hulver MW. Metabolic endotoxemia with obesity: Is it real and is it relevant? Biochimie 2016; 124:11-20. [PMID: 26133659 PMCID: PMC4695328 DOI: 10.1016/j.biochi.2015.06.020] [Citation(s) in RCA: 279] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 06/23/2015] [Indexed: 02/06/2023]
Abstract
Obesity is associated with metabolic derangements in multiple tissues, which contribute to the progression of insulin resistance and the metabolic syndrome. The underlying stimulus for these metabolic derangements in obesity are not fully elucidated, however recent evidence in rodents and humans suggests that systemic, low level elevations of gut derived endotoxin (lipopolysaccharide, LPS) may play an important role in obesity related, whole-body and tissue specific metabolic perturbations. LPS initiates a well-characterized signaling cascade that elicits many pro- and anti-inflammatory pathways when bound to its receptor, Toll-Like Receptor 4 (TLR4). Low-grade elevation in plasma LPS has been termed "metabolic endotoxemia" and this state is associated with a heightened pro-inflammatory and oxidant environment often observed in obesity. Given the role of inflammatory and oxidative stress in the etiology of obesity related cardio-metabolic disease risk, it has been suggested that metabolic endotoxemia may serve a key mediator of metabolic derangements observed in obesity. This review provides supporting evidence of mechanistic associations with cell and animal models, and provides complimentary evidence of the clinical relevance of metabolic endotoxemia in obesity as it relates to inflammation and metabolic derangements in humans. Discrepancies with endotoxin detection are considered, and an alternate method of reporting metabolic endotoxemia is recommended until a standardized measurement protocol is set forth.
Collapse
Affiliation(s)
- Nabil E Boutagy
- The Department of Human Nutrition, Foods, and Exercise, 295 West Campus Drive, Virginia Tech, Blacksburg, VA 24061, USA; The Fralin Translational Obesity Research Center, 1981 Kraft Drive, Virginia Tech, Blacksburg, VA 24061, USA; The Metabolic Phenotyping Core, 1981 Kraft Drive, Virginia Tech, Blacksburg, VA 24061, USA.
| | - Ryan P McMillan
- The Department of Human Nutrition, Foods, and Exercise, 295 West Campus Drive, Virginia Tech, Blacksburg, VA 24061, USA; The Fralin Translational Obesity Research Center, 1981 Kraft Drive, Virginia Tech, Blacksburg, VA 24061, USA; The Metabolic Phenotyping Core, 1981 Kraft Drive, Virginia Tech, Blacksburg, VA 24061, USA.
| | - Madlyn I Frisard
- The Department of Human Nutrition, Foods, and Exercise, 295 West Campus Drive, Virginia Tech, Blacksburg, VA 24061, USA; The Fralin Translational Obesity Research Center, 1981 Kraft Drive, Virginia Tech, Blacksburg, VA 24061, USA; The Metabolic Phenotyping Core, 1981 Kraft Drive, Virginia Tech, Blacksburg, VA 24061, USA.
| | - Matthew W Hulver
- The Department of Human Nutrition, Foods, and Exercise, 295 West Campus Drive, Virginia Tech, Blacksburg, VA 24061, USA; The Fralin Translational Obesity Research Center, 1981 Kraft Drive, Virginia Tech, Blacksburg, VA 24061, USA; The Metabolic Phenotyping Core, 1981 Kraft Drive, Virginia Tech, Blacksburg, VA 24061, USA.
| |
Collapse
|
56
|
Wu Y, Lan C, Ren D, Chen GY. Induction of Siglec-1 by Endotoxin Tolerance Suppresses the Innate Immune Response by Promoting TGF-β1 Production. J Biol Chem 2016; 291:12370-82. [PMID: 27129263 DOI: 10.1074/jbc.m116.721258] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Indexed: 12/22/2022] Open
Abstract
Sepsis is one of the leading causes of death worldwide. Although the prevailing theory for the sepsis syndrome is a condition of uncontrolled inflammation in response to infection, sepsis is increasingly being recognized as an immunosuppressive state known as endotoxin tolerance. We found sialylation of cell surface was significantly increased on LPS-induced tolerant cells; knockdown of Neu1 in macrophage cell line RAW 264.7 cells resulted in enhanced LPS-induced tolerance, whereas overexpression of Neu1 or treatment with sialidase abrogated LPS-induced tolerance, as defined by measuring TNF-α levels in the culture supernatants. We also found that the expression of Siglec-1 (a member of sialic acid-binding Ig (I)-like lectin family members, the predominant sialic acid-binding proteins on cell surface) was specifically up-regulated in endotoxin tolerant cells and the induction of Siglec-1 suppresses the innate immune response by promoting TGF-β1 production. The enhanced TGF-β1 production by Siglec-1 was significantly attenuated by spleen tyrosine kinase (Syk) inhibitor. Knockdown of siglec-1 in RAW 264.7 cells resulted in inhibiting the production of TGF-β1 by ubiquitin-dependent degradation of Syk. Mechanistically, Siglec-1 associates with adaptor protein DNAX-activation protein of 12 kDa (DAP12) and transduces a signal to Syk to control the production of TGF-β1 in endotoxin tolerance. Thus, Siglec-1 plays an important role in the development of endotoxin tolerance and targeted manipulation of this process could lead to a new therapeutic opportunity for patients with sepsis.
Collapse
Affiliation(s)
- Yin Wu
- From the Children's Foundation Research Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee 38103
| | - Chao Lan
- From the Children's Foundation Research Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee 38103
| | - Dongren Ren
- From the Children's Foundation Research Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee 38103
| | - Guo-Yun Chen
- From the Children's Foundation Research Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee 38103
| |
Collapse
|
57
|
Neagos J, Standiford TJ, Newstead MW, Zeng X, Huang SK, Ballinger MN. Epigenetic Regulation of Tolerance to Toll-Like Receptor Ligands in Alveolar Epithelial Cells. Am J Respir Cell Mol Biol 2016; 53:872-81. [PMID: 25965198 DOI: 10.1165/rcmb.2015-0057oc] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
To protect the host against exuberant inflammation and injury responses, cells have the ability to become hyporesponsive or "tolerized" to repeated stimulation by microbial and nonmicrobial insults. The lung airspace is constantly exposed to a variety of exogenous and endogenous Toll-like receptor (TLR) ligands, yet the ability of alveolar epithelial cells (AECs) to be tolerized has yet to be examined. We hypothesize that type II AECs will develop a tolerance phenotype upon repeated TLR agonist exposure. To test this hypothesis, primary AECs isolated from the lungs of mice and a murine AEC cell line (MLE-12) were stimulated with either a vehicle control or a TLR ligand for 18 hours, washed, then restimulated with either vehicle or TLR ligand for an additional 6 hours. Tolerance was assessed by measurement of TLR ligand-stimulated chemokine production (monocyte chemoattractant protein [MCP]-1/CCL2, keratinocyte chemoattractant [KC]/CXCL1, and macrophage inflammatory protein [MIP]-2/CXCL2). Sequential treatment of primary AECs or MLE-12 cells with TLR agonists resulted in induction of either tolerance or cross-tolerance. The induction of tolerance was not due to expression of specific negative regulators of TLR signaling (interleukin-1 receptor associated kinase [IRAK]-M, Toll-interacting protein [Tollip], single Ig IL-1-related receptor [SIGIRR], or suppressor of cytokine signaling [SOCS]), inhibitory microRNAs (miRs; specifically, miR-155 and miR146a), or secretion of inhibitory or regulatory soluble mediators (prostaglandin E2, IL-10, transforming growth factor-β, or IFN-α/β). Moreover, inhibition of histone demethylation or DNA methylation did not prevent the development of tolerance. However, treatment of AECs with the histone deacetylase inhibitors trichostatin A or suberoylanilide hyrozamine resulted in reversal of the tolerance phenotype. These findings indicate a novel mechanism by which epigenetic modification regulates the induction of tolerance in AECs.
Collapse
Affiliation(s)
- Jacqueline Neagos
- 1 Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan; and
| | - Theodore J Standiford
- 1 Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan; and
| | - Michael W Newstead
- 1 Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan; and
| | - Xianying Zeng
- 1 Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan; and
| | - Steven K Huang
- 1 Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan; and
| | - Megan N Ballinger
- 2 Department of Internal Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Ohio State University, Columbus, Ohio
| |
Collapse
|
58
|
Scully EP, Lockhart A, Garcia-Beltran W, Palmer CD, Musante C, Rosenberg E, Allen TM, Chang JJ, Bosch RJ, Altfeld M. Innate immune reconstitution with suppression of HIV-1. JCI Insight 2016; 1:e85433. [PMID: 27158667 DOI: 10.1172/jci.insight.85433] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Progressive HIV-1 infection leads to both profound immune suppression and pathologic inflammation in the majority of infected individuals. While adaptive immune dysfunction, as evidenced by CD4+ T cell depletion and exhaustion, has been extensively studied, less is known about the functional capacity of innate immune cell populations in the context of HIV-1 infection. Given the broad susceptibility to opportunistic infections and the dysregulated inflammation observed in progressive disease, we hypothesized that there would be significant changes in the innate cellular responses. Using a cohort of patients with multiple samplings before and after antiretroviral therapy (ART) initiation, we demonstrated increased responses to innate immune stimuli following viral suppression, as measured by the production of inflammatory cytokines. Plasma viral load itself had the strongest association with this change in innate functional capacity. We further identified epigenetic modifications in the TNFA promoter locus in monocytes that are associated with viremia, suggesting a molecular mechanism for the observed changes in innate immune function following initiation of ART. These data indicate that suppression of HIV-1 viremia is associated with changes in innate cellular function that may in part determine the restoration of protective immune responses.
Collapse
Affiliation(s)
- Eileen P Scully
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, USA; Division of Infectious Diseases, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Ainsley Lockhart
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, USA
| | - Wilfredo Garcia-Beltran
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, USA
| | - Christine D Palmer
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, USA
| | - Chelsey Musante
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, USA
| | - Eric Rosenberg
- Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Todd M Allen
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, USA
| | - J Judy Chang
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, USA
| | - Ronald J Bosch
- Harvard School of Public Health, Boston, Massachusetts, USA
| | - Marcus Altfeld
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, USA; Heinrich-Pette-Institut, Hamburg, Germany
| |
Collapse
|
59
|
Galbraith N, Walker S, Galandiuk S, Gardner S, Polk HC. The Significance and Challenges of Monocyte Impairment: For the Ill Patient and the Surgeon. Surg Infect (Larchmt) 2016; 17:303-12. [PMID: 26958709 DOI: 10.1089/sur.2015.245] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Trauma, major elective surgery, and overt sepsis can lead to a cascade of immunological change. A subset of these patients will have a degree of immune suppression that leads to hyporesponsive innate defenses, increasing the risk of infective co-morbidity and death. This article is an overview of monocyte impairment in the high-risk surgical patient. Specifically, our primary focus is on observations made pertaining to monocyte function and pathophysiological mechanisms underpinning this impairment. Clinical factors influencing monocyte function are also discussed. METHODS A Pubmed search was conducted to review aspects of monocyte impairment in the surgical patient. Search terms included "monocyte impairment," "immunoparalysis," and "endotoxin tolerance" cross-referenced against terms including "trauma," "major surgery," and "sepsis." RESULTS Findings revealed a broad variety of monocyte defects reported in surgical patients. They ranged from altered cytokine responses, particularly ex vivo TNF-α production, to impaired antigen presentation such as depressed HLA-DR expression. The latter is the most commonly described marker of secondary infection and death. Studies of underlying mechanisms have commonly utilized a model of endotoxin tolerance with in vitro monocytes, revealing a complex array of dysregulated pathways. For our purposes, endotoxin tolerance and monocyte impairment are sufficiently similar entities to permit further study as a single subject. In the high risk patient, microRNAs (also referred to as miRNA or miR) are emerging as potential biomarkers that may modify such pathways. Creation of a reliable impaired human monocyte model could be important to all such considerations. CONCLUSION Impairment of monocyte function continues to be predictive of nosocomial infection, multi-organ failure, and death in some surgical patients. However, the optimal marker that could identify a patient as high risk early enough, and whether it might guide potential therapy, still is yet to be proven.
Collapse
Affiliation(s)
- Norman Galbraith
- Department of Surgery, University of Louisville School of Medicine , Louisville, Kentucky
| | - Samuel Walker
- Department of Surgery, University of Louisville School of Medicine , Louisville, Kentucky
| | - Susan Galandiuk
- Department of Surgery, University of Louisville School of Medicine , Louisville, Kentucky
| | - Sarah Gardner
- Department of Surgery, University of Louisville School of Medicine , Louisville, Kentucky
| | - Hiram C Polk
- Department of Surgery, University of Louisville School of Medicine , Louisville, Kentucky
| |
Collapse
|
60
|
Timmermans K, Kox M, Gerretsen J, Peters E, Scheffer GJ, van der Hoeven JG, Pickkers P, Hoedemaekers CW. The Involvement of Danger-Associated Molecular Patterns in the Development of Immunoparalysis in Cardiac Arrest Patients. Crit Care Med 2016. [PMID: 26196352 DOI: 10.1097/ccm.0000000000001204] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
OBJECTIVES After cardiac arrest, patients are highly vulnerable toward infections, possibly due to a suppressed state of the immune system called "immunoparalysis." We investigated if immunoparalysis develops following cardiac arrest and whether the release of danger-associated molecular patterns could be involved. DESIGN Observational study. SETTING ICU of a university medical center. PATIENTS Fourteen post-cardiac arrest patients treated with mild therapeutic hypothermia for 24 hours and 11 control subjects. MEASUREMENTS AND MAIN RESULTS Plasma cytokines showed highest levels within 24 hours after cardiac arrest and decreased during the next 2 days. By contrast, ex vivo production of cytokines interleukin-6, tumor necrosis factor-α, and interleukin-10 by lipopolysaccharide-stimulated leukocytes was severely impaired compared with control subjects, with most profound effects observed at day 0, and only partially recovering afterward. Compared with incubation at 37°C, incubation at 32°C resulted in higher interleukin-6 and lower interleukin-10 production by lipopolysaccharide-stimulated leukocytes of control subjects, but not of patients. Plasma nuclear DNA, used as a marker for general danger-associated molecular pattern release, and the specific danger-associated molecular patterns (EN-RAGE and heat shock protein 70) were substantially higher in patients at days 0 and 1 compared with control subjects. Furthermore, plasma heat shock protein 70 levels were negatively correlated with ex vivo production of inflammatory mediators interleukin-6, tumor necrosis factor-α, and interleukin-10. Extracellular newly identified receptor for advanced glycation end products-binding protein levels only showed a significant negative correlation with ex vivo production of interleukin-6 and tumor necrosis factor-α and a borderline significant inverse correlation with interleukin-10. No significant correlations were observed between plasma nuclear DNA levels and ex vivo cytokine production. INTERVENTIONS None. CONCLUSIONS Release of danger-associated molecular patterns during the first days after cardiac arrest is associated with the development of immunoparalysis. This could explain the increased susceptibility toward infections in cardiac arrest patients.
Collapse
Affiliation(s)
- Kim Timmermans
- 1Department of Intensive Care Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands. 2Department of Anesthesiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands. 3Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
61
|
Scriven JE, Graham LM, Schutz C, Scriba TJ, Wilkinson KA, Wilkinson RJ, Boulware DR, Urban BC, Lalloo DG, Meintjes G. A Glucuronoxylomannan-Associated Immune Signature, Characterized by Monocyte Deactivation and an Increased Interleukin 10 Level, Is a Predictor of Death in Cryptococcal Meningitis. J Infect Dis 2016; 213:1725-34. [PMID: 26768248 PMCID: PMC4857465 DOI: 10.1093/infdis/jiw007] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 12/23/2015] [Indexed: 11/14/2022] Open
Abstract
Background. Cryptococcal meningitis remains a significant cause of death among human immunodeficiency virus type 1 (HIV)–infected persons in Africa. We aimed to better understand the pathogenesis and identify immune correlates of mortality, particularly the role of monocyte activation. Methods. A prospective cohort study was conducted in Cape Town, South Africa. Patients with a first episode of cryptococcal meningitis were enrolled, and their immune responses were assessed in unstimulated and stimulated blood specimens, using flow cytometry and cytokine analysis. Results. Sixty participants were enrolled (median CD4+ T-cell count, 34 cells/µL). Mortality was 23% (14 of 60 participants) at 14 days and 39% (22 of 57) at 12 weeks. Nonsurvivors were more likely to have an altered consciousness and higher cerebrospinal fluid fungal burden at presentation. Principal component analysis identified an immune signature associated with early mortality, characterized by monocyte deactivation (reduced HLA-DR expression and tumor necrosis factor α response to lipopolysaccharide); increased serum interleukin 6, CXCL10, and interleukin 10 levels; increased neutrophil counts; and decreased T-helper cell type 1 responses. This immune signature remained an independent predictor of early mortality after adjustment for consciousness level and fungal burden and was associated with higher serum titers of cryptococcal glucuronoxylomannan. Conclusions. Cryptococcal-related mortality is associated with monocyte deactivation and an antiinflammatory blood immune signature, possibly due to Cryptococcus modulation of the host immune response. Validation in other cohorts is required.
Collapse
Affiliation(s)
- James E Scriven
- Liverpool School of Tropical Medicine Wellcome Trust Liverpool Glasgow Centre for Global Health Research, Liverpool Clinical Infectious Diseases Research Initiative Department of Medicine, University of Cape Town and Groote Schuur Hospital, South Africa
| | | | - Charlotte Schutz
- Clinical Infectious Diseases Research Initiative Department of Medicine, University of Cape Town and Groote Schuur Hospital, South Africa
| | - Thomas J Scriba
- South African TB Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine Department of Paediatrics and Child Health, University of Cape Town
| | | | - Robert J Wilkinson
- Department of Medicine, Imperial College London Mill Hill Laboratory, Francis Crick Institute, London, United Kingdom Clinical Infectious Diseases Research Initiative
| | | | | | - David G Lalloo
- Liverpool School of Tropical Medicine Wellcome Trust Liverpool Glasgow Centre for Global Health Research, Liverpool
| | - Graeme Meintjes
- Department of Medicine, Imperial College London Clinical Infectious Diseases Research Initiative Department of Medicine, University of Cape Town and Groote Schuur Hospital, South Africa
| |
Collapse
|
62
|
Chiariotti L, Coretti L, Pero R, Lembo F. Epigenetic Alterations Induced by Bacterial Lipopolysaccharides. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 879:91-105. [PMID: 26659265 DOI: 10.1007/978-3-319-24738-0_5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Lipopolysaccharide (LPS) is one of the principal bacterial products known to elicit inflammation. Cells of myeloid lineage such as monocytes and macrophages, but also epithelial cells give rise to an inflammatory response upon LPS stimulation. This phenomenon implies reprogramming of cell specific gene expression that can occur through different mechanisms including epigenetic modifications. Given their intrinsic nature, epigenetic modifications may be involved both in the acute response to LPS and in the establishment of a preconditioned genomic state (epigenomic memory) that may potentially influence the host response to further contacts with microorganisms. Information has accumulated during the last years aimed at elucidating the epigenetic mechanisms which underlie the cellular LPS response. These findings, summarized in this chapter, will hopefully be a good basis for a definition of the complete cascade of LPS-induced epigenetic events and their biological significance in different cell types.
Collapse
Affiliation(s)
- Lorenzo Chiariotti
- Department of Medicina Molecolare e Biotecnologie Mediche, University of Naples "Federico II", Naples, Italy. .,Istituto di Endocrinologia ed Oncologia Sperimentale IEOS, C.N.R., EPIGEN Laboratories, Naples, Italy.
| | - Lorena Coretti
- Department of Medicina Molecolare e Biotecnologie Mediche, University of Naples "Federico II", Naples, Italy.,Istituto di Endocrinologia ed Oncologia Sperimentale IEOS, C.N.R., EPIGEN Laboratories, Naples, Italy
| | - Raffaela Pero
- Department of Medicina Molecolare e Biotecnologie Mediche, University of Naples "Federico II", Naples, Italy
| | - Francesca Lembo
- Istituto di Endocrinologia ed Oncologia Sperimentale IEOS, C.N.R., EPIGEN Laboratories, Naples, Italy. .,Department of Pharmacy, University of Naples "Federico II", Naples, Italy.
| |
Collapse
|
63
|
Endothelial cell tolerance to lipopolysaccharide challenge is induced by monophosphoryl lipid A. Clin Sci (Lond) 2015; 130:451-61. [PMID: 26669797 DOI: 10.1042/cs20150592] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 12/15/2015] [Indexed: 01/18/2023]
Abstract
Prior exposure to lipopolysaccharide (LPS) produces a reduced or "tolerant" inflammatory response to subsequent challenges with LPS, however the potent pro-inflammatory effects of LPS limit its clinical benefit. The adjuvant monophosphoryl lipid A (MPLA) is a weak toll-like receptor 4 (TLR4) agonist that induces negligible inflammation but retains potent immunomodulatory properties. We postulated that pre-treatment with MPLA would inhibit the inflammatory response of endothelial cells to secondary LPS challenge. Human umbilical vein endothelial cells (HUVECs), were exposed to MPLA (10 μg/ml), LPS (100 ng/ml) or vehicle control. HUVECs were then washed and maintained in culture for 24 h before being challenged with LPS (100 ng/ml). Supernatants were collected and examined for cytokine production in the presence or absence of siRNA inhibitors of critical TLR4 signalling proteins. Pre-treatment with MPLA attenuated interleukin (IL)-6 production to secondary LPS challenge to a similar degree as LPS. The application of myeloid differentiation primary response gene 88 (MyD88) siRNA dramatically reduced MPLA-induced tolerance while TIR-domain-containing adapter-inducing interferon-β (TRIF) siRNA had no effect. The tolerant phenotype in endothelial cells was associated with reduced IκB kinase (IKK), p38 and c-Jun N-terminal kinase (JNK) phosphorylation and enhanced IL-1 receptor associated kinase-M (IRAK-M) expression for LPS-primed HUVECs, but less so in MPLA primed cells. Instead, MPLA-primed HUVECs demonstrated enhanced p-extracellular-signal-regulated kinase (ERK) phosphorylation. In contrast with leucocytes in which tolerance is largely TRIF-dependent, MyD88 signalling mediated endotoxin tolerance in endothelial cells. Most importantly, MPLA, a vaccine adjuvant with a wide therapeutic window, induced tolerance to LPS in endothelial cells.
Collapse
|
64
|
Palmer CD, Romero-Tejeda M, Sirignano M, Sharma S, Allen TM, Altfeld M, Jost S. Naturally Occurring Subclinical Endotoxemia in Humans Alters Adaptive and Innate Immune Functions through Reduced MAPK and Increased STAT1 Phosphorylation. THE JOURNAL OF IMMUNOLOGY 2015; 196:668-77. [PMID: 26643479 DOI: 10.4049/jimmunol.1501888] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 11/02/2015] [Indexed: 11/19/2022]
Abstract
Multiple studies have shown correlates of immune activation with microbial translocation and plasma LPS during HIV infection. It is unclear whether this activation is due to LPS, residual viral replication, or both. Few studies have addressed the effects of persistent in vivo levels of LPS on specific immune functions in humans in the absence of chronic viral infection or pathological settings such as sepsis. We previously reported on a cohort of HIV-negative men with subclinical endotoxemia linked to alterations in CD4/CD8 T cell ratio and plasma cytokine levels. This HIV-negative cohort allowed us to assess cellular immune functions in the context of different subclinical plasma LPS levels ex vivo without confounding viral effects. By comparing two samples of differing plasma LPS levels from each individual, we now show that subclinical levels of plasma LPS in vivo significantly alter T cell proliferative capacity, monocyte cytokine release, and HLA-DR expression, and induce TLR cross-tolerance by decreased phosphorylation of MAPK pathway components. Using this human in vivo model of subclinical endotoxemia, we furthermore show that plasma LPS leads to constitutive activation of STAT1 through autocrine cytokine signaling, suggesting that subclinical endotoxemia in healthy individuals might lead to significant changes in immune function that have thus far not been appreciated.
Collapse
Affiliation(s)
| | | | | | | | - Todd M Allen
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139; and
| | - Marcus Altfeld
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139; and Department of Viral Immunology, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, 20251 Hamburg, Germany
| | - Stephanie Jost
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139; and
| |
Collapse
|
65
|
Walshe CM, Laffey JG, Kevin L, O’Toole D. Sepsis protects the myocardium and other organs from subsequent ischaemic/reperfusion injury via a MAPK-dependent mechanism. Intensive Care Med Exp 2015; 3:35. [PMID: 26215802 PMCID: PMC4513033 DOI: 10.1186/s40635-014-0035-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 12/09/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Sepsis has been shown to precondition the intact heart against ischaemia/reperfusion (IR) injury, and prior endotoxin exposure of cells in in vitro models has shown evidence of protection against subsequent simulated ischaemia. Our aim in this study is to validate these findings and further investigate the signaling pathways involved. METHODS Adult male Sprague Dawley rats were randomised to control (n = 7) or caecal ligation and perforation (CLP)-induced sepsis (n = 7). Hearts were harvested at 48 h, suspended in Langendorff mode and subjected to 30-min global ischaemia followed by 90-min reperfusion. In subsequent experiments, designed to determine the mechanisms by which sepsis protected against ischaemic injury, endotoxin-stimulated isolated cardiomyocytes, pulmonary A549 cells and renal HK2 cells were subjected to normoxic and hypoxic conditions. The roles of key pathways, including mitogen-activated protein (MAP) kinases extracellular-regulated protein kinase (ERK) 1/2, p38 MAPK (p38), c-Jun NH2-terminal protein kinase (JNK)), and nuclear factor-kappaB (NF-κB) were examined. RESULTS Systemic sepsis protected isolated hearts from subsequent ischaemic/reperfusion-induced injury, enhancing functional recovery on reperfusion [developed left ventricular pressure ((d)LVP) mean(SE) 66.63(±10.7) mmHg vs. 54.13(±9.9) mmHg; LVPmax at 60 min 67.29(±11.9) vs. 72.48(±9.3), sepsis vs. control] despite significantly reduced baseline LV function in CLP animals (p < 0.001). Septic preconditioning significantly reduced infarct size after IR injury (p < 0.05). Endotoxin exposure protected isolated cardiomyocytes against hypoxia-induced cell death (p < 0.001). This effect appeared mediated in part via the p38, JNK and NF-κB pathways, but was independent of the ERK pathway, and did not appear to be mediated via HMGB1. The preconditioning effect of endotoxin was also demonstrated in isolated kidney and lung cells, suggesting that this preconditioning effect of sepsis is not confined to the myocardium. CONCLUSIONS Sepsis preconditions the isolated rat heart against myocardial IR injury. These effects appeared to be mediated in part via the p38, JNK and NF-κB and pathways, but were independent of the ERK and HMGB pathways.
Collapse
Affiliation(s)
- Criona M Walshe
- Department of Anaesthesia, Galway University Hospitals and National University of Ireland, University Road, Galway, Ireland
| | - John G Laffey
- Department of Anaesthesia, Galway University Hospitals and National University of Ireland, University Road, Galway, Ireland
| | - Leo Kevin
- Department of Anaesthesia, Galway University Hospitals and National University of Ireland, University Road, Galway, Ireland
| | - Daniel O’Toole
- Department of Anaesthesia, Galway University Hospitals and National University of Ireland, University Road, Galway, Ireland
| |
Collapse
|
66
|
Gnauck A, Lentle RG, Kruger MC. The Characteristics and Function of Bacterial Lipopolysaccharides and Their Endotoxic Potential in Humans. Int Rev Immunol 2015; 35:189-218. [PMID: 26606737 DOI: 10.3109/08830185.2015.1087518] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Cross-talk between enteral microbiota and human host is essential for the development and maintenance of the human gastrointestinal and systemic immune systems. The presence of lipopolysaccharides (LPS) lysed from the cell membrane of Gram-negative bacteria in the gut lumen is thought to promote the development of a balanced gut immune response whilst the entry of the same LPS into systemic circulation may lead to a deleterious pro-inflammatory systemic immune response. Recent data suggest that chronically low levels of circulating LPS may be associated with the development of metabolic diseases such as insulin resistance, type 2 diabetes, atherosclerosis and cardiovascular disease. This review focuses on the cross-talk between enteral commensal bacteria and the human immune system via LPS. We explain the structural characterisation of the LPS molecule and its function in the bacteria. We then examine how LPS is recognised by various elements of the human immune system and the signalling pathways that are activated by the structure of the LPS molecule and the effect of various concentrations. Further, we discuss the sequelae of this signalling in the gut-associated and systemic immune systems i.e. the neutralisation of LPS and the development of tolerance to LPS.
Collapse
Affiliation(s)
- Anne Gnauck
- a School of Food and Nutrition, College of Health , Massey University , Palmerston North , New Zealand
| | - Roger G Lentle
- a School of Food and Nutrition, College of Health , Massey University , Palmerston North , New Zealand
| | - Marlena C Kruger
- a School of Food and Nutrition, College of Health , Massey University , Palmerston North , New Zealand
| |
Collapse
|
67
|
Julian MW, Strange HR, Ballinger MN, Hotchkiss RS, Papenfuss TL, Crouser ED. Tolerance and Cross-Tolerance following Toll-Like Receptor (TLR)-4 and -9 Activation Are Mediated by IRAK-M and Modulated by IL-7 in Murine Splenocytes. PLoS One 2015. [PMID: 26218271 PMCID: PMC4517781 DOI: 10.1371/journal.pone.0132921] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Objective Immune suppression during critical illness predisposes to serious infections. We sought to determine the mechanisms regulating tolerance and cross-tolerance to common pro-inflammatory danger signals in a model that recapitulates the intact in vivo immune response. Materials and Methods Flt3-expanded splenocytes obtained from wild-type or matching IRAK-M knockout (IRAK-M-/-), C57BL/6, male mice (8–10 weeks old) were treated repeatedly or alternately with either LPS or CpGA DNA, agonists of Toll-like receptor (TLR)-4 and -9, respectively, over successive 24-hour periods. Supernatants were collected following each 24-hour period with cytokine release (ELISA) and splenocyte IRAK-M expression (Western blot) determined. Tolerance and cross-tolerance were assessed in the absence or presence of programmed death receptor (PD)-1 blocking antibody or IL-7 pre-treatment. Main Results Splenocytes notably exhibited both tolerance and cross-tolerance to subsequent treatments with either LPS or CpGA DNA. The character of tolerance and cross-tolerance in this model was distinct following initial LPS or CpGA treatment in that TNFα and IFNγ release (not IL-10) were suppressed following LPS; whereas, initial CpGA treatment suppressed TNFα, IFNγ and IL-10 release in response to subsequent stimulation (LPS or CpGA). Tolerance and cross-tolerance were unrelated to IL-10 release or PD-1 but were attenuated in IRAK-M-/- splenocytes. IL-7 significantly suppressed IRAK-M expression and restored TNFα and IFNγ production without influencing IL-10 release. Conclusions In summary, acute immune tolerance and cross-tolerance in response to LPS or CpGA were distinct in that LPS selectively suppressed pro-inflammatory cytokine responses; whereas, CpGA suppressed both pro- and anti-inflammatory responses. The induction of tolerance and cross-tolerance in response to common danger signals was mechanistically unrelated to IL-10 or PD-1 but was directly influenced by IRAK-M expression. IL-7 reduced IRAK-M expression and attenuated immune tolerance induced by either LPS or CpGA, and thus may be useful for reversal of immune tolerance in the setting of critical illness.
Collapse
Affiliation(s)
- Mark W. Julian
- Dorothy M. Davis Heart and Lung Research Institute, Division of Pulmonary Allergy, Critical Care, and Sleep Medicine, Wexner Medical Center, Columbus, OH, United States of America
| | - Heather R. Strange
- College of Veterinary Medicine, Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States of America
| | - Megan N. Ballinger
- Dorothy M. Davis Heart and Lung Research Institute, Division of Pulmonary Allergy, Critical Care, and Sleep Medicine, Wexner Medical Center, Columbus, OH, United States of America
| | - Richard S. Hotchkiss
- Departments of Anesthesiology, Medicine and Surgery, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Tracey L. Papenfuss
- College of Veterinary Medicine, Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States of America
| | - Elliott D. Crouser
- Dorothy M. Davis Heart and Lung Research Institute, Division of Pulmonary Allergy, Critical Care, and Sleep Medicine, Wexner Medical Center, Columbus, OH, United States of America
- * E-mail:
| |
Collapse
|
68
|
Shi L, Song L, Maurer K, Sharp J, Zhang Z, Sullivan KE. Endotoxin tolerance in monocytes can be mitigated by α2-interferon. J Leukoc Biol 2015. [PMID: 26206900 DOI: 10.1189/jlb.4a0914-450rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Endotoxin tolerance is characterized by diminished expression of inflammatory cytokines after sequential exposure to Toll-like receptor stimuli. Many mechanisms contribute to tolerance; however, chromatin remodeling appears to be the most significant regulator. The type II interferon, IFN-γ, has been recognized as being able to reverse or abrogate the establishment of tolerance. Type I interferons have not been investigated previously, and they bind a distinct receptor. We found that α2-interferon was able to abrogate or diminish tolerance by endotoxin, as defined by measuring mRNA levels at recognized tolerance targets. We also found that α2-interferon treatment during tolerization was associated with increased H3K4me3 and H3K4me2 levels at promoters of tolerance targets in THP1 cells. These marks were normalized after exposure of the cells to α2-interferon. Interferon regulatory factor 1 is a transcription factor activated and induced by types I and II interferons. We found recruitment of this transcription factor paralleled tolerance and inhibition of tolerance at target genes. Therefore, there are at least 2 distinct pathways by which endotoxin tolerance may be mitigated. A type I interferon, in spite of binding to a different receptor, was just as able to inhibit tolerance as the type II interferon and also appeared to act by modifying chromatin at tolerance target genes.
Collapse
Affiliation(s)
- Lihua Shi
- *Division of Allergy Immunology and The Center for Biomedical Informatics, The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Li Song
- *Division of Allergy Immunology and The Center for Biomedical Informatics, The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Kelly Maurer
- *Division of Allergy Immunology and The Center for Biomedical Informatics, The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - James Sharp
- *Division of Allergy Immunology and The Center for Biomedical Informatics, The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Zhe Zhang
- *Division of Allergy Immunology and The Center for Biomedical Informatics, The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Kathleen E Sullivan
- *Division of Allergy Immunology and The Center for Biomedical Informatics, The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
69
|
Hormone and Cytokine Responses to Repeated Endotoxin Exposures—No Evidence of Endotoxin Tolerance After 5 Weeks in Humans. Shock 2015; 44:32-5. [DOI: 10.1097/shk.0000000000000384] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
70
|
Perez-Cruz M, Costa C, Manez R. Divergence of the response induced by xenogenic immunization in the sepsis survival of rats. PLoS One 2015; 10:e0125472. [PMID: 25984763 PMCID: PMC4436005 DOI: 10.1371/journal.pone.0125472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 03/24/2015] [Indexed: 11/18/2022] Open
Abstract
We have previously described that boosted natural xenoantibodies in rats cross-react to bacteria by targeting carbohydrate antigens. This type of immunization is associated with reduced survival after cecal ligation and puncture (CLP). In the present study, we investigated further this phenomenon by immunizing Lewis rats with three intraperitoneal injections, every other day, of hamster blood compared to saline-injected control animals. One day after the last injection, CLP was performed to produce a low-grade sepsis. Induction of xenoantibodies was associated with a reduction in animal survival after CLP relative to controls (45% vs. 90%, p<0.01). No bacterial blood load was observed after CLP in this model either with or without xenoantibody enhancement, indicating that the augmented mortality was not mediated by a direct effect of boosted xenoantibodies over blood bacteria. Nevertheless, the xenoimmunization produced a systemic inflammatory response in all rats. Additionally, a lack of weight gain at the time of CLP was present in animals that died after the procedure, which was not observed in surviving rats and controls. The cytokine profile at the time of CLP in animals that died after the procedure was characterized by an increase in the serum level of several cytokines, particularly adipokines. In contrast, the cytokine profile at CLP of xenoimmunized rats that survived the procedure was characterized by a reduction in the level of cytokines. In conclusion, this study failed to show a direct effect of boosted xenoantibodies over blood bacterial isolates as cause for the decreased survival after CLP. However, it evidenced that non-infectious systemic inflammation may lead to a pattern of augmented cytokines, particularly adipokines, which impairs survival after subsequent CLP. Therefore, the profile of cytokines existing before the infectious insult appears more crucial than that resulting from the condition for the outcome of sepsis.
Collapse
Affiliation(s)
- Magdiel Perez-Cruz
- Infectious Diseases and Transplantation Division, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain
| | - Cristina Costa
- Infectious Diseases and Transplantation Division, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain
| | - Rafael Manez
- Infectious Diseases and Transplantation Division, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain
- Intensive Care Department, Bellvitge University Hospital, L’Hospitalet de Llobregat, Barcelona, Spain
- * E-mail:
| |
Collapse
|
71
|
Patel PN, Shah RY, Ferguson JF, Reilly MP. Human experimental endotoxemia in modeling the pathophysiology, genomics, and therapeutics of innate immunity in complex cardiometabolic diseases. Arterioscler Thromb Vasc Biol 2015; 35:525-34. [PMID: 25550206 PMCID: PMC4344396 DOI: 10.1161/atvbaha.114.304455] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 12/18/2014] [Indexed: 01/16/2023]
Abstract
Inflammation is a fundamental feature of several complex cardiometabolic diseases. Indeed, obesity, insulin resistance, metabolic dyslipidemia, and atherosclerosis are all closely linked inflammatory states. Increasing evidence suggests that the infectious, biome-related, or endogenous activation of the innate immune system may contribute to the development of metabolic syndrome and cardiovascular disease. Here, we describe the human experimental endotoxemia model for the specific study of innate immunity in understanding further the pathogenesis of cardiometabolic disease. In a controlled, experimental setting, administration of an intravenous bolus of purified Escherichia coli endotoxin activates innate immunity in healthy human volunteers. During endotoxemia, changes emerge in glucose metabolism, lipoprotein composition, and lipoprotein functions that closely resemble those observed chronically in inflammatory cardiovascular disease risk states. In this review, we describe the transient systemic inflammation and specific metabolic consequences that develop during human endotoxemia. Such a model provides a controlled induction of systemic inflammation, eliminates confounding, undermines reverse causation, and possesses unique potential as a starting point for genomic screening and testing of novel therapeutics for treatment of the inflammatory underpinning of cardiometabolic disease.
Collapse
Affiliation(s)
- Parth N Patel
- From the Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (P.N.P., R.Y.S., M.P.R.); and Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN (J.F.F.)
| | - Rhia Y Shah
- From the Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (P.N.P., R.Y.S., M.P.R.); and Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN (J.F.F.)
| | - Jane F Ferguson
- From the Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (P.N.P., R.Y.S., M.P.R.); and Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN (J.F.F.)
| | - Muredach P Reilly
- From the Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (P.N.P., R.Y.S., M.P.R.); and Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN (J.F.F.).
| |
Collapse
|
72
|
BCG-induced protection: Effects on innate immune memory. Semin Immunol 2014; 26:512-7. [DOI: 10.1016/j.smim.2014.09.006] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 09/17/2014] [Accepted: 09/21/2014] [Indexed: 01/23/2023]
|
73
|
Hussen J, Frank C, Düvel A, Koy M, Schuberth HJ. The chemokine CCL5 induces selective migration of bovine classical monocytes and drives their differentiation into LPS-hyporesponsive macrophages in vitro. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2014; 47:169-177. [PMID: 25064684 DOI: 10.1016/j.dci.2014.07.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 07/17/2014] [Accepted: 07/18/2014] [Indexed: 06/03/2023]
Abstract
Human and mouse studies indicate distinct roles of selected chemokines for monocyte subset attraction. We therefore analyzed the still unknown sensitivity and response of bovine monocyte subsets toward two monocyte-attracting chemokines (CCL2, CCL5). Only CCL5 induced a significant Ca(2+)influx and migration response in bovine monocytes, with classical and intermediate monocytes being significantly stimulated and attracted compared to nonclassical monocytes. The presence of CCL5 during in vitro macrophage differentiation did not alter their capacity to phagocytize or to generate reactive oxygen species upon stimulation with E. coli. However, macrophages differentiated in the presence of CCL5 displayed an altered phenotype with significantly less expressed CD14 and MHC class II molecules, whereas CD16 was upregulated. Moreover, CCL5-differentiated macrophages displayed a reduced upregulation of CXCL8, ARG1, IL6 and IL10 mRNA. Taken together, CCL5 but not CCL2 mainly attract bovine classical monocytes and promote their differentiation into LPS-hypo-responsive macrophages.
Collapse
Affiliation(s)
- Jamal Hussen
- Immunology Unit, University of Veterinary Medicine Foundation, Bischofsholer Damm 15, D-30173 Hannover, Germany
| | - Constanze Frank
- Immunology Unit, University of Veterinary Medicine Foundation, Bischofsholer Damm 15, D-30173 Hannover, Germany
| | - Anna Düvel
- Immunology Unit, University of Veterinary Medicine Foundation, Bischofsholer Damm 15, D-30173 Hannover, Germany; HIPRA Deutschland GmbH, Feldstraße 21, D-40479 Düsseldorf, Germany
| | - Mirja Koy
- Immunology Unit, University of Veterinary Medicine Foundation, Bischofsholer Damm 15, D-30173 Hannover, Germany
| | - Hans-Joachim Schuberth
- Immunology Unit, University of Veterinary Medicine Foundation, Bischofsholer Damm 15, D-30173 Hannover, Germany.
| |
Collapse
|
74
|
Dillingh MR, van Poelgeest EP, Malone KE, Kemper EM, Stroes ESG, Moerland M, Burggraaf J. Characterization of inflammation and immune cell modulation induced by low-dose LPS administration to healthy volunteers. JOURNAL OF INFLAMMATION-LONDON 2014. [DOI: 10.1186/s12950-014-0028-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
75
|
Eftekhari G, Hajiasgharzadeh K, Ahmadi-Soleimani SM, Dehpour AR, Semnanian S, Mani AR. Activation of central muscarinic receptor type 1 prevents development of endotoxin tolerance in rat liver. Eur J Pharmacol 2014; 740:436-41. [PMID: 25008070 DOI: 10.1016/j.ejphar.2014.06.050] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 06/24/2014] [Accepted: 06/26/2014] [Indexed: 12/13/2022]
Abstract
Endotoxin tolerance is a mechanism in which cells receiving low doses of endotoxin, enter a transient phase with less inflammatory response to the next endotoxin challenges. Central nervous system is known to modulate systemic inflammation through activation of the cholinergic system; however, the role of central anti-inflammatory pathway in pathophysiology of hepatic endotoxin tolerance is unknown. Our study was designed to assess the effect central muscarinic type 1 receptor (M1) activation on development of endotoxin tolerance in rat liver. Endotoxin tolerance was induced by daily intraperitoneal injection of endotoxin (1 mg/kg) for 5 days. Animals were randomly divided into two groups which received intracerebroventricular injection of either MCNA-343 (an M1 agonist, 5 ng/kg) or saline 1h after intraperitoneal injection of saline or endotoxin. The responsiveness to endotoxin was assessed by measuring hepatic MCP-1 (monocyte chemotactic protein-1), iNOS (inducible nitric oxide synthase) and TNF-α (tumor necrosis-α) mRNA expression 3h after intraperitoneal administration of endotoxin using quantitative RT-PCR. A significant reduction in hepatic expression of MCP-1, iNOS and TNF-α was observed in rats with 5 days endotoxin challenge in comparison with rats given a single dose of endotoxin. There was no significant difference in hepatic expression of MCP-1, iNOS or TNF-α between acute and chronic LPS-treated groups in rats given MCNA-343. Central MCNA-343 stimulation could prevent the induction of hepatic endotoxin tolerance in animals receiving repeated doses of endotoxin. This indicates that M1 cholinergic receptor activation in the central nervous system can modulate endotoxin tolerance in rat liver.
Collapse
Affiliation(s)
- Golnar Eftekhari
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Khalil Hajiasgharzadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Ahmad R Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Semnanian
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ali R Mani
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
76
|
Aggarwal NR, Tsushima K, Eto Y, Tripathi A, Mandke P, Mock JR, Garibaldi BT, Singer BD, Sidhaye VK, Horton MR, King LS, D'Alessio FR. Immunological priming requires regulatory T cells and IL-10-producing macrophages to accelerate resolution from severe lung inflammation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 192:4453-4464. [PMID: 24688024 PMCID: PMC4001810 DOI: 10.4049/jimmunol.1400146] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Overwhelming lung inflammation frequently occurs following exposure to both direct infectious and noninfectious agents and is a leading cause of mortality worldwide. In that context, immunomodulatory strategies may be used to limit severity of impending organ damage. We sought to determine whether priming the lung by activating the immune system, or immunological priming, could accelerate resolution of severe lung inflammation. We assessed the importance of alveolar macrophages, regulatory T cells, and their potential interaction during immunological priming. We demonstrate that oropharyngeal delivery of low-dose LPS can immunologically prime the lung to augment alveolar macrophage production of IL-10 and enhance resolution of lung inflammation induced by a lethal dose of LPS or by Pseudomonas bacterial pneumonia. IL-10-deficient mice did not achieve priming and were unable to accelerate lung injury resolution. Depletion of lung macrophages or regulatory T cells during the priming response completely abrogated the positive effect of immunological priming on resolution of lung inflammation and significantly reduced alveolar macrophage IL-10 production. Finally, we demonstrated that oropharyngeal delivery of synthetic CpG-oligonucleotides elicited minimal lung inflammation compared with low-dose LPS but nonetheless primed the lung to accelerate resolution of lung injury following subsequent lethal LPS exposure. Immunological priming is a viable immunomodulatory strategy used to enhance resolution in an experimental acute lung injury model with the potential for therapeutic benefit against a wide array of injurious exposures.
Collapse
Affiliation(s)
- Neil R Aggarwal
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Kenji Tsushima
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Yoshiki Eto
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Ashutosh Tripathi
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Pooja Mandke
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Jason R Mock
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Brian T Garibaldi
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Benjamin D Singer
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Venkataramana K Sidhaye
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Maureen R Horton
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Landon S King
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Franco R D'Alessio
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21224
| |
Collapse
|
77
|
Lv Y, Hu S, Lu J, Dong N, Liu Q, Du M, Zhang H. Upregulating nonneuronal cholinergic activity decreases TNF release from lipopolysaccharide-stimulated RAW264.7 cells. Mediators Inflamm 2014; 2014:873728. [PMID: 24733966 PMCID: PMC3964895 DOI: 10.1155/2014/873728] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 01/03/2014] [Accepted: 01/16/2014] [Indexed: 11/18/2022] Open
Abstract
Nonneuronal cholinergic system plays a primary role in maintaining homeostasis. It has been proved that endogenous neuronal acetylcholine (ACh) could play an anti-inflammatory role, and exogenous cholinergic agonists could weaken macrophages inflammatory response to lipopolysaccharide (LPS) stimulation through activation of α7 subunit-containing nicotinic acetylcholine receptor (α7nAChR). We assumed that nonneuronal cholinergic system existing in macrophages could modulate inflammation through autocrine ACh and expressed α7nAChR on the cells. Therefore, we explored whether LPS continuous stimulation could upregulate the nonneuronal cholinergic activity in macrophages and whether increasing autocrine ACh could decrease TNF release from the macrophages. The results showed that, in RAW264.7 cells incubated with LPS for 20 hours, the secretion of ACh was significantly decreased at 4 h and then gradually increased, accompanied with the enhancement of α7nAChR expression level. The release of TNF was greatly increased from RAW264.7 cells at 4 h and 8 h exposure to LPS; however, it was suppressed at 20 h. Upregulating choline acetyltransferase (ChAT) expression through ChAT gene transfection could enhance ACh secretion and reduce TNF release from the infected RAW264. 7cells. The results indicated that LPS stimulation could modulate the activity of nonneuronal cholinergic system of RAW264.7 cells. Enhancing autocrine ACh production could attenuate TNF release from RAW264.7 cells.
Collapse
Affiliation(s)
- Yi Lv
- Laboratory of Shock and Multiple Organ Dysfunction, Burns Institute, First Hospital Affiliated to the People's Liberation Army General Hospital, 51 Fu Cheng Road, Beijing 100048, China
| | - Sen Hu
- Laboratory of Shock and Multiple Organ Dysfunction, Burns Institute, First Hospital Affiliated to the People's Liberation Army General Hospital, 51 Fu Cheng Road, Beijing 100048, China
| | - Jiangyang Lu
- Department of Pathology, First Hospital Affiliated to the People's Liberation Army General Hospital, 51 Fu Cheng Road, Beijing 100048, China
| | - Ning Dong
- Laboratory of Shock and Multiple Organ Dysfunction, Burns Institute, First Hospital Affiliated to the People's Liberation Army General Hospital, 51 Fu Cheng Road, Beijing 100048, China
| | - Qian Liu
- Department of Pathology, First Hospital Affiliated to the People's Liberation Army General Hospital, 51 Fu Cheng Road, Beijing 100048, China
| | - Minghua Du
- Laboratory of Shock and Multiple Organ Dysfunction, Burns Institute, First Hospital Affiliated to the People's Liberation Army General Hospital, 51 Fu Cheng Road, Beijing 100048, China
| | - Huiping Zhang
- Laboratory of Shock and Multiple Organ Dysfunction, Burns Institute, First Hospital Affiliated to the People's Liberation Army General Hospital, 51 Fu Cheng Road, Beijing 100048, China
| |
Collapse
|
78
|
Tsigou E, Aloizos S, Stavros A, Myrianthefs P, Pavlos M, Gourgiotis S, Stavros G, Tsakris A, Athanassios T, Baltopoulos G, George B. The immune response after stimulation with wall components of gram-positive bacteria and fungi. Immunol Lett 2014; 159:23-9. [PMID: 24440200 DOI: 10.1016/j.imlet.2013.12.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Revised: 12/05/2013] [Accepted: 12/11/2013] [Indexed: 10/25/2022]
Abstract
Although several components of the microbial wall of gram-positive bacteria and fungi possess immunostimulatory properties, their pathogenetic role remains incompletely evaluated. The purpose of this study was to assess the basic immune status of patients susceptible to infections and their capability for cytokine production after stimulation with wall components of gram-positive bacteria and fungi. We measured serum cytokine levels as well as cytokine production after ex vivo lipoteichoic acid (LTA) and mannan stimulation of whole blood. The blood was taken from 10 healthy volunteers, 10 patients with end-stage renal disease (ESRD), 10 patients with diabetes mellitus (DM), and 10 patients on their 2nd day of stay in the Intensive Care Unit (ICU), who suffered from non septic systemic inflammatory response syndrome (SIRS) and had an APACHE II score ≥25. We used 1 μg/ml LTA and 100 μg/ml mannan for an incubation period of 8 h to stimulate 100 μl aliquots of whole blood. All patient groups had higher baseline values of TNF-α, IL-6, IL-1β, and IL-10 compared to the control group, but only for ICU patients the difference was statistically significant. The ratio IL-10/IL-6 was found 0.33, 0.22, and 0.96 in healthy persons, ESRD, and DM patients respectively, and 1.32 in ICU patients. In all examined groups, the levels of cytokines significantly increased after stimulation by LTA and mannan, although in severely ill patients this change was considerably smaller, possibly reflecting a state of monocytes' depression and relative hyporesponsiveness. No significant differences between the LTA and the mannan stimulation were observed.
Collapse
Affiliation(s)
- Evdoxia Tsigou
- Department of Intensive Care Medicine, A. Anargiroi Hospital, Athens, Greece.
| | | | - Aloizos Stavros
- Department of Intensive Care Medicine, A. Anargiroi Hospital, Athens, Greece
| | | | - Myrianthefs Pavlos
- Department of Intensive Care Medicine, A. Anargiroi Hospital, Athens, Greece
| | | | - Gourgiotis Stavros
- Department of Intensive Care Medicine, A. Anargiroi Hospital, Athens, Greece
| | | | - Tsakris Athanassios
- Department of Intensive Care Medicine, A. Anargiroi Hospital, Athens, Greece
| | | | - Baltopoulos George
- Department of Intensive Care Medicine, A. Anargiroi Hospital, Athens, Greece
| |
Collapse
|
79
|
López-Collazo E, del Fresno C. Pathophysiology of endotoxin tolerance: mechanisms and clinical consequences. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2013; 17:242. [PMID: 24229432 PMCID: PMC4059412 DOI: 10.1186/cc13110] [Citation(s) in RCA: 167] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Endotoxin tolerance was first described in a study that exposed animals to a sublethal dose of bacterial endotoxin. The animals subsequently survived a lethal injection of endotoxin. This refractory state is associated with the innate immune system and, in particular, with monocytes and macrophages, which act as the main participants. Several mechanisms are involved in the control of endotoxin tolerance; however, a full understanding of this phenomenon remains elusive. A number of recent reports indicate that clinical examples of endotoxin tolerance include not only sepsis but also diseases such as cystic fibrosis and acute coronary syndrome. In these pathologies, the risk of new infections correlates with a refractory state. This review integrates the molecular basis and clinical implications of endotoxin tolerance in various pathologies.
Collapse
|
80
|
MicroRNA-146a and RBM4 form a negative feed-forward loop that disrupts cytokine mRNA translation following TLR4 responses in human THP-1 monocytes. Immunol Cell Biol 2013; 91:532-40. [PMID: 23897118 PMCID: PMC3770753 DOI: 10.1038/icb.2013.37] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 06/13/2013] [Accepted: 07/07/2013] [Indexed: 01/13/2023]
Abstract
Within hours after its initiation, the severe systemic inflammatory response of sepsis shifts to an adaptive anti-inflammatory state with coincident immunosuppression. This anti-inflammatory phenotype is characterized by diminished proinflammatory cytokine gene expression in response to toll-like receptor (TLR) stimulation with bacterial endotoxin/lipopolysaccharide (LPS), also known as endotoxin tolerance/adaptation. Our and other studies have established that gene-specific reprogramming following TLR4 responses independently represses transcription and translation of proinflammatory genes such as tumor necrosis factor alpha (TNFα). We also previously demonstrated that TNFα and interleukin (IL)-6 mRNA translation is repressed in endotoxin-adapted THP-1 human monocytes by an miRNA-based mechanism involving the argonaute family protein argonaute 2 (Ago2). Here, we further define the molecular nature of reprogramming translation by showing that TLR4-induced microRNA-146 promotes a feed-forward loop that modifies the subcellular localization of the RNA-binding protein RBM4 (RNA-binding motif protein 4) and promotes its interaction with Ago2. This interaction results in the assembly of a translation-repressor complex that disrupts TNFα and IL-6 cytokine synthesis in endotoxin-adapted THP-1 monocytes. This novel molecular path prevents the phosphorylation of RBM4 on serine-309 by p38 MAPK (mitogen-activated protein kinase), which leads to RBM4 accumulation in the cytosol and interaction with Ago2. We further find that microRNA-146a knockdown by antagomirs or protein phosphatase inhibition by okadaic acid increases p38 MAPK phosphorylation and results in RBM4 serine-309 phosphorylation and nuclear relocalization, which disrupts RBM4 and Ago2 interactions and restores TLR4-dependent synthesis of TNFα and IL-6. We conclude that miR-146a has a diverse and critical role in limiting an excessive acute inflammatory reaction.
Collapse
|
81
|
Kubera M, Curzytek K, Duda W, Leskiewicz M, Basta-Kaim A, Budziszewska B, Roman A, Zajicova A, Holan V, Szczesny E, Lason W, Maes M. A new animal model of (chronic) depression induced by repeated and intermittent lipopolysaccharide administration for 4 months. Brain Behav Immun 2013; 31:96-104. [PMID: 23313516 DOI: 10.1016/j.bbi.2013.01.001] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 01/02/2013] [Accepted: 01/03/2013] [Indexed: 02/07/2023] Open
Abstract
Chronic activation of immune-inflammatory and oxidative and nitrosative stress (O&NS) pathways plays an important role in the pathophysiology of clinical depression. Increased IgA responses directed against LPS of gram-negative bacteria, indicating increased bacterial translocation, may be one of the drivers underpinning these pathways. There is a strong association between signs of bacterial translocation and chronicity of depression and O&NS, but not pro-inflammatory cytokines. The aims of the present study were to: (1) develop a new neurobehavioral model of (chronic) depression (anhedonic behavior) that may reflect chronic LPS stimulation and is associated with increased oxidative stress, and (2) to delineate the effects of fluoxetine on this new depression model. We established that in female mice repeated LPS injections once daily for 5 days (from 750 μg/kg to a maximal dose 1250 μg/kg; increasing doses for the first three days which were then gradually decreased on day 4 and 5) at a one-month interval and this repeated for 4 consecutive months induced chronic anhedonia (estimated by the preference to drink a 1% sucrose) lasting for at least 7 weeks. Chronic LPS administration significantly decreased thymus weight, proliferative activity of splenocytes, production of interferon (IFN)γ and interleukin-(IL)10, and increased superoxide and corticosterone production. Treatment with fluoxetine for 3 weeks abolished the neurobehavioral effects of LPS. The antidepressant effect of fluoxetine was accompanied by increased production of IL-10 and reduced superoxide and corticosterone production. Our results suggest that repeated intermittent LPS injections to female mice may be a useful model of chronic depression and in particular for the depressogenic effects of long standing activation of the toll-like receptor IV complex.
Collapse
Affiliation(s)
- Marta Kubera
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Science, 31-343 Krakow, Poland.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
van Swelm RP, Laarakkers CM, Pertijs JC, Verweij V, Masereeuw R, Russel FG. Urinary proteomic profiling reveals diclofenac-induced renal injury and hepatic regeneration in mice. Toxicol Appl Pharmacol 2013; 269:141-9. [DOI: 10.1016/j.taap.2013.03.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 03/03/2013] [Accepted: 03/06/2013] [Indexed: 12/25/2022]
|
83
|
Benn CS, Netea MG, Selin LK, Aaby P. A small jab - a big effect: nonspecific immunomodulation by vaccines. Trends Immunol 2013; 34:431-9. [PMID: 23680130 DOI: 10.1016/j.it.2013.04.004] [Citation(s) in RCA: 388] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 03/18/2013] [Accepted: 04/07/2013] [Indexed: 02/07/2023]
Abstract
Recent epidemiological studies have shown that, in addition to disease-specific effects, vaccines against infectious diseases have nonspecific effects on the ability of the immune system to handle other pathogens. For instance, in randomized trials tuberculosis and measles vaccines are associated with a substantial reduction in overall child mortality, which cannot be explained by prevention of the target disease. New research suggests that the nonspecific effects of vaccines are related to cross-reactivity of the adaptive immune system with unrelated pathogens, and to training of the innate immune system through epigenetic reprogramming. Hence, epidemiological findings are backed by immunological data. This generates a new understanding of the immune system and about how it can be modulated by vaccines to impact the general resistance to disease.
Collapse
Affiliation(s)
- Christine S Benn
- Research Center for Vitamins and Vaccines (CVIVA), Statens Serum Institut, Institute of Clinical Research, University of Southern Denmark, and Odense University Hospital, Denmark.
| | | | | | | |
Collapse
|
84
|
Maes M, Kubera M, Leunis JC, Berk M, Geffard M, Bosmans E. In depression, bacterial translocation may drive inflammatory responses, oxidative and nitrosative stress (O&NS), and autoimmune responses directed against O&NS-damaged neoepitopes. Acta Psychiatr Scand 2013; 127:344-54. [PMID: 22900942 DOI: 10.1111/j.1600-0447.2012.01908.x] [Citation(s) in RCA: 155] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Depression is accompanied by activation of immuno-inflammatory and oxidative and nitrosative stress (IO&NS) pathways, and increased IgM/IgA responses to lipopolysaccharide (LPS) of gram-negative commensal bacteria. The latter suggests that bacterial translocation has caused IgM/IgA responses directed against LPS. Bacterial translocation may drive IO&NS responses. METHOD To examine the associations between IgM/IgA responses to LPS and IO&NS measurements, including plasma/serum interleukin-1 (IL-1), tumor necrosis factor (TNF)α, neopterin, lysozyme, oxidized LDL (oxLDL) antibodies, peroxides, and IgM (auto)immune responses against malondialdehyde (MDA), azelaic acid, phophatidyl inositol (Pi), NO-tryptophan and NO-tyrosine in depressed patients and controls. RESULTS We found significant positive associations between IgM/IgA responses to LPS and oxLDL antibodies, IgM responses against MDA, azelaic acid, Pi, NO-tryptophan, and NO-tyrosine. The IgA responses to LPS were correlated with lysozyme. There were no significant positive correlations between the IgM/IgA responses to LPS and IL-1 and neopterin. CONCLUSION The findings show that in depression there is an association between increased bacterial translocation and lysozyme production, an antibacterial compound, O&NS processes, and autoimmune responses directed against O&NS generated neoantigenic determinants. It is suggested that bacterial translocation may drive IO&NS pathways in depression and thus play a role in its pathophysiology.
Collapse
Affiliation(s)
- M Maes
- Maes Clinics @ Tria, Bangkok, Thailand.
| | | | | | | | | | | |
Collapse
|
85
|
Haddadian Z, Eftekhari G, Mazloom R, Jazaeri F, Dehpour AR, Mani AR. Effect of endotoxin on heart rate dynamics in rats with cirrhosis. Auton Neurosci 2013; 177:104-13. [PMID: 23511062 DOI: 10.1016/j.autneu.2013.02.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 02/11/2013] [Accepted: 02/25/2013] [Indexed: 12/11/2022]
Abstract
Reduced heart rate variability (HRV) is a hallmark of systemic inflammation which carries negative prognostic information in sepsis. Decreased HRV is associated with partial uncoupling of cardiac pacemaker from cholinergic neural control during systemic inflammation. Sepsis is a common complication in liver cirrhosis with high mortality. The present study was aimed to explore the hypothesis that endotoxin uncouples cardiac pacemaker from autonomic neural control and reduces HRV in an experimental model of cirrhosis. Cirrhosis was induced by surgical ligation of the bile duct in rats. Cirrhotic rats were given intraperitoneal injection of either saline or lipopolysaccharide (endotoxin, 1mg/kg). Changes in HRV indices were studied in conscious rats using implanted telemetric probes. The atria were isolated and chronotropic responsiveness to cholinergic stimulation was assessed in vitro. Endotoxin injection induced a significant tachycardia and decreased short-term and long-term HRV indices in control rats. However, endotoxin was unable to increase heart rate in cirrhotic animals. In contrast with control rats, endotoxin induced biphasic changes in short-term HRV in cirrhotic rats. Acute endotoxin challenge reduced long-term HRV with 60-min delay in comparison with control animals. Endotoxin injection was associated with a significant hypo-responsiveness to cholinergic stimulation in control rats in vitro. Endotoxin did not change atrial chronotropic responsiveness to cholinergic stimulation in cirrhotic rats. Our data shows that cirrhosis is associated with development of tolerance to cardiac chronotropic effect of endotoxin in rats.
Collapse
Affiliation(s)
- Zahra Haddadian
- Department of Physiology, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | | | | | | | | |
Collapse
|
86
|
Castegren M, Skorup P, Lipcsey M, Larsson A, Sjölin J. Endotoxin tolerance variation over 24 h during porcine endotoxemia: association with changes in circulation and organ dysfunction. PLoS One 2013; 8:e53221. [PMID: 23326400 PMCID: PMC3542331 DOI: 10.1371/journal.pone.0053221] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 11/27/2012] [Indexed: 01/05/2023] Open
Abstract
Endotoxin tolerance (ET), defined as reduced inflammatory responsiveness to endotoxin challenge following a first encounter with endotoxin, is an extensively studied phenomenon. Although reduced mortality and morbidity in the presence of ET has been demonstrated in animal studies, little is known about the temporal development of ET. Further, in acute respiratory distress syndrome ET correlates to the severity of the disease, suggesting a complicated relation between ET and organ dysfunction. Eighteen pigs were subjected to intensive care and a continuous endotoxin infusion for 24 h with the aim to study the time course of early ET and to relate ET to outcome in organ dysfunction. Three animals served as non-endotoxemic controls. Blood samples for cytokine analyses were taken and physiological variables registered every third hour. Production of TNF-α, IL-6, and IL-10 before and after endotoxin stimulation ex vivo was measured. The difference between cytokine values after and before ex vivo LPS stimulation (Δ-values) was calculated for all time points. ΔTNF-α was employed as the principal marker of ET and lower ΔTNF-α values were interpreted as higher levels of ET. During endotoxin infusion, there was suppression of ex vivo productions of TNF-α and IL-6 but not of IL-10 in comparison with that at 0 h. The ex vivo TNF-α values followed another time concentration curve than those in vivo. ΔTNF-α was at the lowest already at 6 h, followed by an increase during the ensuing hours. ΔTNF-α at 6 h correlated positively to blood pressure and systemic vascular resistance and negatively to cardiac index at 24 h. In this study a temporal variation of ET was demonstrated that did not follow changes in plasma TNF-α concentrations. Maximal ET occurred early in the course and the higher the ET, the more hyperdynamic the circulation 18 h later.
Collapse
Affiliation(s)
- Markus Castegren
- Centre for Clinical Research Sörmland, Uppsala University, Uppsala, Sweden.
| | | | | | | | | |
Collapse
|
87
|
Rinsky JL, Hoppin JA, Blair A, He K, Beane Freeman LE, Chen H. Agricultural exposures and stroke mortality in the Agricultural Health Study. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2013; 76:798-814. [PMID: 24028665 PMCID: PMC3773612 DOI: 10.1080/15287394.2013.819308] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Exposures associated with common agricultural activities may increase risk of stroke. The authors evaluated associations between self-reported agricultural activities including pesticide use and handling of crops and stroke mortality among 51,603 male pesticide applicators enrolled in the Agricultural Health Study (AHS). Vital status was obtained through 2008. Stroke mortality was defined by underlying or contributing cause of death (ICD-9 430-438, ICD-10 I60-I69). Information regarding lifetime pesticide use, working with crops or animals, engagement in other agricultural activities, and potential confounders was self-reported at enrollment. Cox proportional hazards models, with age as the time scale, were used to estimate hazard ratios (HR) and 95% confidence intervals (CI) adjusted for state of residence, smoking status, and alcohol consumption. Median follow-up time was 13 yr, during which 308 stroke deaths occurred. No measure of overall or specific pesticide use was positively associated with mortality due to stroke. Stroke mortality was inversely associated with handling hay, grain, or silage at least once each year as reported at enrollment (HR: 0.75; 95% CI: 0.58, 0.98). There was no evidence of an association between pesticide use and stroke mortality. The inverse association between handling of hays and grains and stroke mortality may be due to (1) those engaging in such activities being healthier than those who did not or (2) exposure to some biological agent present in hays and grains. Further investigation of incident stroke, rather than stroke mortality, as well as stroke subtypes, is needed to determine the full role of agricultural exposures and stroke.
Collapse
Affiliation(s)
- Jessica L. Rinsky
- Department of Epidemiology, University of North Carolina Gillings School of Global Public Health. Chapel Hill, NC
| | - Jane A. Hoppin
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services. Research Triangle Park, NC
| | - Aaron Blair
- Occupational and Environmental Epidemiology Branch, National Cancer Institute, National Institutes of Health, Department of Health and Human Services. Bethesda, MD
| | - Ka He
- Departments of Nutrition and Epidemiology, University of North Carolina Gillings School of Global Public Health, Chapel Hill, NC and Department of Epidemiology and Biostatistics, School of Public Health, Indiana University Bloomington. Bloomington, IN
| | - Laura E. Beane Freeman
- Occupational and Environmental Epidemiology Branch, National Cancer Institute, National Institutes of Health, Department of Health and Human Services. Bethesda, MD
| | - Honglei Chen
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services. Research Triangle Park, NC
| |
Collapse
|
88
|
Leentjens J, Kox M, Koch RM, Preijers F, Joosten LAB, van der Hoeven JG, Netea MG, Pickkers P. Reversal of Immunoparalysis in HumansIn Vivo. Am J Respir Crit Care Med 2012; 186:838-45. [DOI: 10.1164/rccm.201204-0645oc] [Citation(s) in RCA: 166] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
89
|
Souza-Fonseca-Guimaraes F, Parlato M, Philippart F, Misset B, Cavaillon JM, Adib-Conquy M. Toll-like receptors expression and interferon-γ production by NK cells in human sepsis. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2012; 16:R206. [PMID: 23098236 PMCID: PMC3682310 DOI: 10.1186/cc11838] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 10/23/2012] [Indexed: 12/19/2022]
Abstract
Introduction During the course of infection, natural killer (NK) cells contribute to innate immunity by producing cytokines, particularly interferon-gamma (IFN-γ). In addition to their beneficial effects against infection, NK cells may play a detrimental role during systemic inflammation, causing lethality during sepsis. Little is known on the immune status of NK cells in patients with systemic inflammatory response syndrome (SIRS) or sepsis in terms of cell surface markers expression and IFN-γ production. Methods We investigated 27 sepsis patients and 11 patients with non-infectious SIRS. CD56bright and CD56dim NK cell subsets were identified by flow cytometry and Toll-like receptor (TLR)2, TLR4, TLR9, CX3CR1, CD16 and CD69 expression were analyzed, as well as ex vivo IFN-γ production by NK cells in whole blood samples. Results We first showed that in NK cells from healthy controls, TLR2 and TLR4 expression is mainly intracellular, similarly to TLR9. Intracellular levels of TLR2 and TLR4, in both CD56bright and CD56dim NK cell subsets from sepsis patients, were increased compared to healthy subjects. In addition, the percentage of CD69+ cells was higher among NK cells of sepsis patients. No difference was observed for TLR9, CX3CR1, and CD16 expression. The ex vivo stimulation by TLR4 or TLR9 agonists, or whole bacteria in synergy with accessory cytokines (IL-15+IL-18), resulted in significant production of IFN-γ by NK cells of healthy controls. In contrast, for SIRS and sepsis patients this response was dramatically reduced. Conclusions This study reports for the first time an intracellular expression of TLR2 and TLR4 in human NK cells. Surface TLR4 expression allows discriminating sepsis and SIRS. Furthermore, during these pathologies, NK cells undergo an alteration of their immune status characterized by a profound reduction of their capacity to release IFN-γ.
Collapse
|
90
|
Gresnigt MS, Joosten LAB, Verschueren I, van der Meer JWM, Netea MG, Dinarello CA, van de Veerdonk FL. Neutrophil-mediated inhibition of proinflammatory cytokine responses. THE JOURNAL OF IMMUNOLOGY 2012; 189:4806-15. [PMID: 23053514 DOI: 10.4049/jimmunol.1103551] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Neutrophils (polymorphonuclear neutrophils [PMNs]) play an elaborate role in the innate immune response against invading pathogens. Recent research provided evidence that PMNs can play a modulatory role in inflammation next to their primary role of phagocytosis. In the current study, we investigated whether neutrophils can modulate the innate immune response against Candida albicans. Production of the proinflammatory cytokines IL-1β and TNF-α by human PBMCs in response to C. albicans or LPS was decreased by coculture of PMNs; however, the anti-inflammatory cytokine IL-10 remained unaffected. Using Transwells and cells of patients with chronic granulomatous disease, we show that this downregulation of proinflammatory cytokine production was independent of phagocytosis and reactive oxygen species but was dependent on a soluble factor. We suggest that neutrophil-derived proteases are responsible for the downregulation of IL-1β and TNF-α, as cytokine production could be recovered by addition of α1-antitrypsin, an endogenous inhibitor of serine proteases. PMN lysates and neutrophil elastase could degrade recombinant human IL-1β and TNF-α but not IL-10, and this could be inhibited by addition of α1-antitrypsin. Moreover, we also provide evidence that the dampening effect of PMNs is present in vivo in a murine zymosan-induced arthritis model and a murine experimental endotoxemia model. Altogether, our data show that PMNs can dampen the proinflammatory response to C. albicans by protease-mediated degradation of cytokines. This observation suggest that PMNs might play a important regulatory role in the host defense against C. albicans and can be important for understanding the regulation of inflammation in general.
Collapse
Affiliation(s)
- Mark S Gresnigt
- Department of Medicine, Nijmegen Institute for Infection, Inflammation, and Immunity, Radboud, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
91
|
Lee KH, Biswas A, Liu YJ, Kobayashi KS. Proteasomal degradation of Nod2 protein mediates tolerance to bacterial cell wall components. J Biol Chem 2012; 287:39800-11. [PMID: 23019338 DOI: 10.1074/jbc.m112.410027] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The innate immune system serves as the first line of defense by detecting microbes and initiating inflammatory responses. Although both Toll-like receptor (TLR) and nucleotide binding domain and leucine-rich repeat (NLR) proteins are important for this process, their excessive activation is hazardous to hosts; thus, tight regulation is required. Endotoxin tolerance is refractory to repeated lipopolysaccharide (LPS) stimulation and serves as a host defense mechanism against septic shock caused by an excessive TLR4 response during gram-negative bacterial infection. Gram-positive bacteria as well as their cell wall components also induce shock. However, the mechanism underlying tolerance is not understood. Here, we show that activation of Nod2 by its ligand, muramyl dipeptide (MDP) in the bacterial cell wall, induces rapid degradation of Nod2, which confers MDP tolerance in vitro and in vivo. Nod2 is constitutively associated with a chaperone protein, Hsp90, which is required for Nod2 stability and protects Nod2 from degradation. Upon MDP stimulation, Hsp90 rapidly dissociates from Nod2, which subsequently undergoes ubiquitination and proteasomal degradation. The SOCS-3 protein induced by Nod2 activation further facilitates this degradation process. Therefore, Nod2 protein stability is a key factor in determining responsiveness to MDP stimulation. This indicates that TLRs and NLRs induce a tolerant state through distinct molecular mechanisms that protect the host from septic shock.
Collapse
Affiliation(s)
- Kyoung-Hee Lee
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | | | | | | |
Collapse
|
92
|
Differences in organ dysfunction in endotoxin-tolerant pigs under intensive care exposed to a second hit of endotoxin. Shock 2012; 37:501-10. [PMID: 22266970 DOI: 10.1097/shk.0b013e318249bb0d] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Endotoxin tolerance is a well-studied phenomenon associated with a reduced inflammatory response. In the switch from an inflammatory to an anti-inflammatory response in clinical sepsis, the concept of endotoxin tolerance is of obvious interest. However, only limited data exist regarding the effect of endotoxin tolerance on organ dysfunction, and therefore, this was investigated in a porcine intensive care sepsis model. Twenty-seven healthy pigs, including nine control animals, were included in the study. Twelve pigs pre-exposed to 24 h of intravenous endotoxin infusion and intensive care and six unexposed pigs were given either a high- or low-dose endotoxin challenge for 6 h. Inflammatory, circulatory, hypoperfusion, and organ dysfunction parameters were followed. The inflammatory responses as well as parameters representing circulation, hypoperfusion, and cardiac and renal function were all markedly attenuated in animals pre-exposed to endotoxin and intensive care as compared with animals not pre-exposed. In animals pre-exposed to endotoxin and given the high-dose of endotoxin challenge, deterioration in pulmonary function was equal to or even worse than in animals not pre-exposed. In contrast to the overall protective effect of endotoxin tolerance observed in other organ systems, the lungs of endotoxin-tolerant animals demonstrated an increased responsiveness to high-dose endotoxin challenge.
Collapse
|
93
|
Lipopolysaccharide- and Superantigen-Modulated Superoxide Production and Monocyte Hyporesponsiveness to Activating Stimuli in Sepsis. Shock 2012; 38:43-8. [DOI: 10.1097/shk.0b013e318257ed62] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
94
|
Souza-Fonseca-Guimaraes F, Parlato M, Fitting C, Cavaillon JM, Adib-Conquy M. NK cell tolerance to TLR agonists mediated by regulatory T cells after polymicrobial sepsis. THE JOURNAL OF IMMUNOLOGY 2012; 188:5850-8. [PMID: 22566566 DOI: 10.4049/jimmunol.1103616] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
As sensors of infection, innate immune cells are able to recognize pathogen-associated molecular patterns by receptors such as TLRs. NK cells present in many tissues contribute to inflammatory processes, particularly through the production of IFN-γ. They may display a protective role during infection but also a detrimental role during sterile or infectious systemic inflammatory response syndrome. Nevertheless, the exact status of NK cells during bacterial sepsis and their capacity directly to respond to TLR agonists remain unclear. The expression of TLRs in NK cells has been widely studied by analyzing the mRNA of these receptors. The aim of this study was to gain insight into TLR2/TLR4/TLR9 expression on/in murine NK cells at the protein level and determine if their agonists were able to induce cytokine production. We show, by flow cytometry, a strong intracellular expression of TLR2 and a low of TLR4 in freshly isolated murine spleen NK cells, similar to that of TLR9. In vitro, purified NK cells respond to TLR2, TLR4, and TLR9 agonists, in synergy with activating cytokines (IL-2, IL-15, and/or IL-18), and produce proinflammatory cytokines (IFN-γ and GM-CSF). Finally, we explored the possible tolerance of NK cells to TLR agonists after a polymicrobial sepsis (experimental peritonitis). For the first time, to our knowledge, NK cells are shown to become tolerant in terms of proinflammatory cytokines production after sepsis. We show that this tolerance is associated with a reduction of the CD27(+)CD11b(-) subset in the spleen related to the presence of regulatory T cells and mainly mediated by TGF-β.
Collapse
|
95
|
Inducible cardiac ischaemia is related to a decrease in the whole-blood Toll-like receptor 2 and 4 response. Clin Sci (Lond) 2012; 122:527-33. [PMID: 22188581 DOI: 10.1042/cs20110323] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
TLR (Toll-like receptor) activation-induced inflammatory responses are important in the progression of atherosclerosis. We previously showed that TLR-dependent leucocyte responsiveness is acutely attenuated following percutaneous coronary intervention or vascular surgery. Furthermore, cytokine release following whole-blood TLR-2 and TLR-4 stimulation is negatively correlated with fractional flow reserve, suggesting that chronic ischaemia can elicit an enhanced inflammatory response. In the present study, we assessed the association between leucocyte TLR-2 and TLR-4 responsiveness and pre-existent and inducible ischaemia in patients undergoing SPECT (single-photon emission computed tomography)-MPI (myocardial perfusion imaging). TLR-2, TLR-4 and CD11b expression on monocytes were measured in blood samples that were obtained from 100 patients with suspected coronary artery disease before and after myocardial stress testing for SPECT-MPI. IL-8 (interleukin-8) levels were determined after whole-blood stimulation with Pam3Cys (TLR-2) and LPS (lipopolysaccharide; TLR-4). On the basis of SPECT-MPI, patients were categorized into three groups: reversible defect, irreversible defect and no defect. Myocardial stress induced a reduction in TLR-4 expression (2.46±0.21 compared with 2.17±0.16 arbitrary units, P=0.001) and CD11b expression (83.2±1.73 compared with 76.0±1.89 arbitrary units, P<0.001). TLR-induced IL-8 production before myocardial stress induction was not associated with the results of SPECT-MPI. However, a significant decrease in IL-8 production following TLR stimulation was observed after stress, which was more pronounced in patients with a reversible defect. In conclusion, inducible ischaemia is associated with a decrease in whole-blood TLR-2 and TLR-4 response. These results point to a regulating role of TLRs in order to prevent excessive inflammatory events known to occur during acute ischaemia.
Collapse
|
96
|
Cole TS, Zhang M, Standiford TJ, Newstead M, Luther J, Zhang J, Chen CC, Kao JY. IRAK-M modulates expression of IL-10 and cell surface markers CD80 and MHC II after bacterial re-stimulation of tolerized dendritic cells. Immunol Lett 2012; 144:49-59. [PMID: 22472665 DOI: 10.1016/j.imlet.2012.03.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Revised: 03/04/2012] [Accepted: 03/12/2012] [Indexed: 01/22/2023]
Abstract
BACKGROUND As essential components of the innate immune system, dendritic cells (DCs) can interact directly with pathogens as well as participate in the adaptive immune response. In cells closely related to DCs such as macrophages and monocytes, prior exposure to minute amounts of endotoxin can lead to a refractory period where subsequent exposure to higher doses fails to induce an inflammatory response; little research has investigated this effect on DCs. This study tested if murine bone marrow-derived dendritic cells (BM-DCs) respond to endotoxin- and bacterial sonicate-induced tolerance by decreased inflammatory and increased anti-inflammatory response, and the role of IRAK-M, an intracellular negative regulator of TLR signaling, in this tolerance. RESULTS Tolerized BM-DCs exhibited a significant drop in TNF-α and IL-12p70 production and increased IL-10 expression compared to untolerized cells. BM-DCs also showed the ability to develop heterotolerance, in which the LPS exposure alone was able to induce tolerance to Helicobacter pylori sonicate and TLR2 agonist Pam3Cys. Furthermore, the expression of IRAK-M was increased after restimulation of tolerized BM-DCs as determined qPCR and Western blot. IRAK-M exhibited a suppressive effect on surface expression of major histocompatibilty complex class II (MHC II) and CD80 in LPS-tolerized BM-DCs. IL-10 expression in bacterial sonicate-tolerized IRAK-M-/- BM-DCs was altered as compared to wild type BM-DCs, with tolerance-induced expression of IL-10 mitigated in tolerized IRAK-M-/- BM-DCs. CONCLUSION Along with endotoxin, bacterial sonicate is able to induce refractory tolerance in BM-DCs, and IRAK-M plays a role in modulating cell surface expression of MHC class II and CD80 and release of IL-10 during this tolerance.
Collapse
Affiliation(s)
- Tyler S Cole
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI 48109, United States
| | | | | | | | | | | | | | | |
Collapse
|
97
|
Shalova IN, Kajiji T, Lim JY, Gómez-Piña V, Fernández-Ruíz I, Arnalich F, Iau PTC, López-Collazo E, Wong SC, Biswas SK. CD16 regulates TRIF-dependent TLR4 response in human monocytes and their subsets. THE JOURNAL OF IMMUNOLOGY 2012; 188:3584-93. [PMID: 22427642 DOI: 10.4049/jimmunol.1100244] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Blood monocytes recognize Gram-negative bacteria through the TLR4, which signal via MyD88- and TRIF-dependent pathway to trigger an immune-inflammatory response. However, a dysregulated inflammatory response by these cells often leads to severe pathologies such as sepsis. We investigated the role of CD16 in the regulation of human monocyte response to Gram-negative endotoxin and sepsis. Blood monocytes from sepsis patients demonstrated an upregulation of several TRIF-dependent genes as well as a selective expansion of CD16-expressing (CD16(+)) monocytes. Gene expression and biochemical studies revealed CD16 to regulate the TRIF-dependent TLR4 pathway in monocytes by activating Syk, IFN regulatory factor 3, and STAT1, which resulted in enhanced expression of IFNB, CCL5, and CXCL10. CD16 also upregulated the expression of IL-1R-associated kinase M and IL-1 receptor antagonist, which are negative regulators of the MyD88-dependent pathway. CD16 overexpression or small interfering RNA knockdown in monocytes confirmed the above findings. Interestingly, these results were mirrored in the CD16(+) monocyte subset isolated from sepsis patients, providing an in vivo confirmation to our findings. Collectively, the results from the current study demonstrate CD16 as a key regulator of the TRIF-dependent TLR4 pathway in human monocytes and their CD16-expressing subset, with implications in sepsis.
Collapse
Affiliation(s)
- Irina N Shalova
- Singapore Immunology Network, Agency for Science, Technology, and Research (A*STAR), Singapore
| | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Lee CT, Zhong L, Mace TA, Repasky EA. Elevation in body temperature to fever range enhances and prolongs subsequent responsiveness of macrophages to endotoxin challenge. PLoS One 2012; 7:e30077. [PMID: 22253887 PMCID: PMC3254634 DOI: 10.1371/journal.pone.0030077] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 12/13/2011] [Indexed: 12/31/2022] Open
Abstract
Macrophages are often considered the sentries in innate immunity, sounding early immunological alarms, a function which speeds the response to infection. Compared to the large volume of studies on regulation of macrophage function by pathogens or cytokines, relatively little attention has been devoted to the role of physical parameters such as temperature. Given that temperature is elevated during fever, a long-recognized cardinal feature of inflammation, it is possible that macrophage function is responsive to thermal signals. To explore this idea, we used LPS to model an aseptic endotoxin-induced inflammatory response in BALB/c mice and found that raising mouse body temperature by mild external heat treatment significantly enhances subsequent LPS-induced release of TNF-α into the peritoneal fluid. It also reprograms macrophages, resulting in sustained subsequent responsiveness to LPS, i.e., this treatment reduces “endotoxin tolerance” in vitro and in vivo. At the molecular level, elevating body temperature of mice results in a increase in LPS-induced downstream signaling including enhanced phosphorylation of IKK and IκB, NF-κB nuclear translocation and binding to the TNF-α promoter in macrophages upon secondary stimulation. Mild heat treatment also induces expression of HSP70 and use of HSP70 inhibitors (KNK437 or Pifithrin-µ) largely abrogates the ability of the thermal treatment to enhance TNF-α, suggesting that the induction of HSP70 is important for mediation of thermal effects on macrophage function. Collectively, these results support the idea that there has been integration between the evolution of body temperature regulation and macrophage function that could help to explain the known survival benefits of fever in organisms following infection.
Collapse
Affiliation(s)
- Chen-Ting Lee
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, New York, United States of America
| | - Lingwen Zhong
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, New York, United States of America
| | - Thomas A. Mace
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, New York, United States of America
| | - Elizabeth A. Repasky
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, New York, United States of America
- * E-mail:
| |
Collapse
|
99
|
Homji NF, Mao X, Langsdorf EF, Chang SL. Endotoxin-induced cytokine and chemokine expression in the HIV-1 transgenic rat. J Neuroinflammation 2012; 9:3. [PMID: 22216977 PMCID: PMC3322344 DOI: 10.1186/1742-2094-9-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2011] [Accepted: 01/04/2012] [Indexed: 01/07/2023] Open
Abstract
Background Repeated exposure to a low dose of a bacterial endotoxin such as lipopolysaccharide (LPS) causes immune cells to become refractory to a subsequent endotoxin challenge, a phenomenon known as endotoxin tolerance (ET). During ET, there is an imbalance in pro- and anti-inflammatory cytokine and chemokine production, leading to a dysregulated immune response. HIV-1 viral proteins are known to have an adverse effect on the immune system. However, the effects of HIV-1 viral proteins during ET have not been investigated. Methods In this study, HIV-1 transgenic (HIV-1Tg) rats and control F344 rats (n = 12 ea) were randomly treated with 2 non-pyrogenic doses of LPS (LL) to induce ET, or saline (SS), followed by a high challenge dose of LPS (LL+L, SS+L) or saline (LL+S, SS+S). The gene expression of 84 cytokines, chemokines, and their receptors in the brain and spleen was examined by relative quantitative PCR using a PCR array, and protein levels in the brain, spleen, and serum of 7 of these 84 genes was determined using an electrochemiluminescent assay. Results In the spleen, there was an increase in key pro-inflammatory (IL1α, IL-1β, IFN-γ) and anti-inflammatory (IL-10) cytokines, and inflammatory chemokines (Ccl2, Ccl7, and Ccl9,) in response to LPS in the SS+L and LL+L (ET) groups of both the HIV-1Tg and F344 rats, but was greater in the HIV-1Tg rats than in the F344. In the ET HIV-1Tg and F344 (LL+L) rats in the spleen, the LPS-induced increase in pro-inflammatory cytokines was diminished and that of the anti-inflammatory cytokine was enhanced compared to the SS+L group rats. In the brain, IL-1β, as well as the Ccl2, Ccl3, and Ccl7 chemokines were increased to a greater extent in the HIV-1Tg rats compared to the F344; whereas Cxcl1, Cxcl10, and Cxcl11 were increased to a greater extent in the F344 rats compared to the HIV-1Tg rats in the LL+L and SS+L groups. Conclusion Our data indicate that the continuous presence of HIV-1 viral proteins can have tissue-dependent effects on endotoxin-induced cytokine and chemokine expression in the ET state.
Collapse
Affiliation(s)
- Natasha F Homji
- Institute of NeuroImmune Pharmacology, Seton Hall University, South Orange, NJ 07079, USA
| | | | | | | |
Collapse
|
100
|
|