101
|
An D, Hao F, Zhang F, Kong W, Chun J, Xu X, Cui MZ. CD14 is a key mediator of both lysophosphatidic acid and lipopolysaccharide induction of foam cell formation. J Biol Chem 2017; 292:14391-14400. [PMID: 28705936 DOI: 10.1074/jbc.m117.781807] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 07/08/2017] [Indexed: 01/19/2023] Open
Abstract
Macrophage uptake of oxidized low-density lipoprotein (oxLDL) plays an important role in foam cell formation and the pathogenesis of atherosclerosis. We report here that lysophosphatidic acid (LPA) enhances lipopolysaccharide (LPS)-induced oxLDL uptake in macrophages. Our data revealed that both LPA and LPS highly induce the CD14 expression at messenger RNA and protein levels in macrophages. The role of CD14, one component of the LPS receptor cluster, in LPA-induced biological functions has been unknown. We took several steps to examine the role of CD14 in LPA signaling pathways. Knockdown of CD14 expression nearly completely blocked LPA/LPS-induced oxLDL uptake in macrophages, demonstrating for the first time that CD14 is a key mediator responsible for both LPA- and LPS-induced oxLDL uptake/foam cell formation. To determine the molecular mechanism mediating CD14 function, we demonstrated that both LPA and LPS significantly induce the expression of scavenger receptor class A type I (SR-AI), which has been implicated in lipid uptake process, and depletion of CD14 levels blocked LPA/LPS-induced SR-AI expression. We further showed that the SR-AI-specific antibody, which quenches SR-AI function, blocked LPA- and LPS-induced foam cell formation. Thus, SR-AI is the downstream mediator of CD14 in regulating LPA-, LPS-, and LPA/LPS-induced foam cell formation. Taken together, our results provide the first experimental evidence that CD14 is a novel connecting molecule linking both LPA and LPS pathways and is a key mediator responsible for LPA/LPS-induced foam cell formation. The LPA/LPS-CD14-SR-AI nexus might be the new convergent pathway, contributing to the worsening of atherosclerosis.
Collapse
Affiliation(s)
- Dong An
- From the Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee 37996.,College of Life Sciences and
| | - Feng Hao
- From the Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee 37996
| | - Fuqiang Zhang
- From the Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee 37996.,Science and Research Center, China-Japan Union Hospital, Jilin University, Changchun 130021, China, and
| | | | - Jerold Chun
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037
| | - Xuemin Xu
- From the Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee 37996
| | - Mei-Zhen Cui
- From the Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee 37996,
| |
Collapse
|
102
|
Brazee PL, Soni PN, Tokhtaeva E, Magnani N, Yemelyanov A, Perlman HR, Ridge KM, Sznajder JI, Vagin O, Dada LA. FXYD5 Is an Essential Mediator of the Inflammatory Response during Lung Injury. Front Immunol 2017; 8:623. [PMID: 28620381 PMCID: PMC5451504 DOI: 10.3389/fimmu.2017.00623] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 05/10/2017] [Indexed: 12/28/2022] Open
Abstract
The alveolar epithelium secretes cytokines and chemokines that recruit immune cells to the lungs, which is essential for fighting infections but in excess can promote lung injury. Overexpression of FXYD5, a tissue-specific regulator of the Na,K-ATPase, in mice, impairs the alveolo-epithelial barrier, and FXYD5 overexpression in renal cells increases C-C chemokine ligand-2 (CCL2) secretion in response to lipopolysaccharide (LPS). The aim of this study was to determine whether FXYD5 contributes to the lung inflammation and injury. Exposure of alveolar epithelial cells (AEC) to LPS increased FXYD5 levels at the plasma membrane, and FXYD5 silencing prevented both the activation of NF-κB and the secretion of cytokines in response to LPS. Intratracheal instillation of LPS into mice increased FXYD5 levels in the lung. FXYD5 overexpression increased the recruitment of interstitial macrophages and classical monocytes to the lung in response to LPS. FXYD5 silencing decreased CCL2 levels, number of cells, and protein concentration in bronchoalveolar lavage fluid (BALF) after LPS treatment, indicating that FXYD5 is required for the NF-κB-stimulated epithelial production of CCL2, the influx of immune cells, and the increase in alveolo-epithelial permeability in response to LPS. Silencing of FXYD5 also prevented the activation of NF-κB and cytokine secretion in response to interferon α and TNF-α, suggesting that pro-inflammatory effects of FXYD5 are not limited to the LPS-induced pathway. Furthermore, in the absence of other stimuli, FXYD5 overexpression in AEC activated NF-κB and increased cytokine production, while FXYD5 overexpression in mice increased cytokine levels in BALF, indicating that FXYD5 is sufficient to induce the NF-κB-stimulated cytokine secretion by the alveolar epithelium. The FXYD5 overexpression also increased cell counts in BALF, which was prevented by silencing the CCL2 receptor (CCR2), or by treating mice with a CCR2-blocking antibody, confirming that FXYD5-induced CCL2 production leads to the recruitment of monocytes to the lung. Taken together, the data demonstrate that FXYD5 is a key contributor to inflammatory lung injury.
Collapse
Affiliation(s)
- Patricia L Brazee
- Pulmonary and Critical Care Division, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Pritin N Soni
- Pulmonary and Critical Care Division, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Elmira Tokhtaeva
- Department of Physiology, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States.,Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Natalia Magnani
- Pulmonary and Critical Care Division, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Alex Yemelyanov
- Pulmonary and Critical Care Division, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Harris R Perlman
- Division of Rheumatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Karen M Ridge
- Pulmonary and Critical Care Division, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jacob I Sznajder
- Pulmonary and Critical Care Division, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Olga Vagin
- Department of Physiology, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States.,Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Laura A Dada
- Pulmonary and Critical Care Division, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
103
|
Battin C, Hennig A, Mayrhofer P, Kunert R, Zlabinger GJ, Steinberger P, Paster W. A human monocytic NF-κB fluorescent reporter cell line for detection of microbial contaminants in biological samples. PLoS One 2017; 12:e0178220. [PMID: 28542462 PMCID: PMC5443541 DOI: 10.1371/journal.pone.0178220] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 05/09/2017] [Indexed: 12/16/2022] Open
Abstract
Sensing of pathogens by innate immune cells is essential for the initiation of appropriate immune responses. Toll-like receptors (TLRs), which are highly sensitive for various structurally and evolutionary conserved molecules derived from microbes have a prominent role in this process. TLR engagement results in the activation of the transcription factor NF-κB, which induces the expression of cytokines and other inflammatory mediators. The exquisite sensitivity of TLR signalling can be exploited for the detection of bacteria and microbial contaminants in tissue cultures and in protein preparations. Here we describe a cellular reporter system for the detection of TLR ligands in biological samples. The well-characterized human monocytic THP-1 cell line was chosen as host for an NF-ᴋB-inducible enhanced green fluorescent protein reporter gene. We studied the sensitivity of the resultant reporter cells for a variety of microbial components and observed a strong reactivity towards TLR1/2 and TLR2/6 ligands. Mycoplasma lipoproteins are potent TLR2/6 agonists and we demonstrate that our reporter cells can be used as reliable and robust detection system for mycoplasma contaminations in cell cultures. In addition, a TLR4-sensitive subline of our reporters was engineered, and probed with recombinant proteins expressed in different host systems. Bacterially expressed but not mammalian expressed proteins induced strong reporter activity. We also tested proteins expressed in an E. coli strain engineered to lack TLR4 agonists. Such preparations also induced reporter activation in THP-1 cells highlighting the importance of testing recombinant protein preparations for microbial contaminations beyond endotoxins. Our results demonstrate the usefulness of monocytic reporter cells for high-throughput screening for microbial contaminations in diverse biological samples, including tissue culture supernatants and recombinant protein preparations. Fluorescent reporter assays can be measured on standard flow cytometers and in contrast to established detection methods, like luciferase-based systems or Limulus Amebocyte Lysate tests, they do not require costly reagents.
Collapse
Affiliation(s)
- Claire Battin
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Annika Hennig
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Patrick Mayrhofer
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Renate Kunert
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Gerhard J. Zlabinger
- Division of Clinical and Experimental Immunology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Peter Steinberger
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- * E-mail: (PS); (WP), (WP)
| | - Wolfgang Paster
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- * E-mail: (PS); (WP), (WP)
| |
Collapse
|
104
|
Kieser KJ, Kagan JC. Multi-receptor detection of individual bacterial products by the innate immune system. Nat Rev Immunol 2017; 17:376-390. [PMID: 28461704 DOI: 10.1038/nri.2017.25] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The receptors of the innate immune system detect specific microbial ligands to promote effective inflammatory and adaptive immune responses. Although this idea is well appreciated, studies in recent years have highlighted the complexity of innate immune detection, with multiple host receptors recognizing the same microbial ligand. Understanding the collective actions of diverse receptors that recognize common microbial signatures represents a new frontier in the study of innate immunity, and is the focus of this Review. Here, we discuss examples of individual bacterial cell wall components that are recognized by at least two and as many as four different receptors of the innate immune system. These receptors survey the extracellular or cytosolic spaces for their cognate ligands and operate in a complementary manner to induce distinct cellular responses. We further highlight that, despite this genetic diversity in receptors and pathways, common features exist to explain the operation of these receptors. These common features may help to provide unifying organizing principles associated with host defence.
Collapse
Affiliation(s)
- Karen J Kieser
- Department of Pediatrics, Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, Massachusetts 02115, USA
| | - Jonathan C Kagan
- Department of Pediatrics, Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, Massachusetts 02115, USA
| |
Collapse
|
105
|
Structural basis for lipopolysaccharide extraction by ABC transporter LptB2FG. Nat Struct Mol Biol 2017; 24:469-474. [DOI: 10.1038/nsmb.3399] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 03/14/2017] [Indexed: 12/17/2022]
|
106
|
Bowman JD, Surani S, Horseman MA. Endotoxin, Toll-like Receptor-4, and Atherosclerotic Heart Disease. Curr Cardiol Rev 2017; 13:86-93. [PMID: 27586023 PMCID: PMC5452150 DOI: 10.2174/1573403x12666160901145313] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 07/28/2016] [Accepted: 08/25/2016] [Indexed: 02/07/2023] Open
Abstract
Background: Endotoxin is a lipopolysaccharide (LPS) constituent of the outer membrane of most gram negative bacteria. Ubiquitous in the environment, it has been implicated as a cause or con-tributing factor in several disparate disorders from sepsis to heatstroke and Type II diabetes mellitus. Starting at birth, the innate immune system develops cellular defense mechanisms against environmen-tal microbes that are in part modulated through a series of receptors known as toll-like receptors. Endo-toxin, often referred to as LPS, binds to toll-like receptor 4 (TLR4)/ myeloid differentiation protein 2 (MD2) complexes on various tissues including cells of the innate immune system, smooth muscle and endothelial cells of blood vessels including coronary arteries, and adipose tissue. Entry of LPS into the systemic circulation ultimately leads to intracellular transcription of several inflammatory mediators. The subsequent inflammation has been implicated in the development and progression atherosclerosis and subsequent coronary artery disease and heart failure. Objective: The potential roles of endotoxin and TLR4 are reviewed regarding their role in the pathogen-esis of atherosclerotic heart disease. Conclusion: Atherosclerosis is initiated by inflammation in arterial endothelial and subendothelial cells, and inflammatory processes are implicated in its progression to clinical heart disease. Endotoxin and TLR4 play a central role in the inflammatory process, and represent potential targets for therapeutic intervention. Therapy with HMG-CoA inhibitors may reduce the expression of TLR4 on monocytes. Other therapeutic interventions targeting TLR4 expression or function may prove beneficial in athero-sclerotic disease prevention and treatment.
Collapse
Affiliation(s)
- John D Bowman
- Department of Pharmacy Practice, Rangel College of Pharmacy, Texas A&M Health Science Center, Kingsville, TX, United States
| | - Salim Surani
- Department of Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Michael A Horseman
- Department of Pharmacy Practice, Rangel College of Pharmacy, Texas A&M Health Science Center, Kingsville, TX, United States
| |
Collapse
|
107
|
Anti-neuroinflammatory effects of grossamide from hemp seed via suppression of TLR-4-mediated NF-κB signaling pathways in lipopolysaccharide-stimulated BV2 microglia cells. Mol Cell Biochem 2017; 428:129-137. [DOI: 10.1007/s11010-016-2923-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 12/21/2016] [Indexed: 02/04/2023]
|
108
|
Mokkala K, Röytiö H, Ekblad U, Laitinen K. Opportunities for probiotics and polyunsaturated fatty acids to improve metabolic health of overweight pregnant women. Benef Microbes 2017; 8:3-15. [DOI: 10.3920/bm2016.0068] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Overweight during pregnancy predisposes both the mother and foetus to health complications. Maternal complications include gestational diabetes, obstetric problems and type 2 diabetes later in life. Complications for the offspring are not only restricted to the foetal period or birth, such as prematurity and foetal macrosomia, but may also have long-term metabolic health implications through the mechanism of early nutrition programming. One of the key metabolic components characterising overweight in the non-pregnant state is low-grade inflammation manifested by elevated levels of circulatory pro-inflammatory cytokines. In pregnancy, in addition to adipose tissue and placenta, inflammatory response may originate from the gut. The extent to which overweight induces metabolic maladaptation during pregnancy and further compromises maternal and child health is currently poorly understood. In this review, we evaluate recent scientific literature and describe the suggested links between overweight, gut and low-grade inflammation associated metabolic disorders. We focus on overweight pregnant women and gestational diabetes, and discuss how specific dietary factors, probiotics and long-chain polyunsaturated fatty acids (fish oil), might confer health benefits in combatting against metabolic risk factors.
Collapse
Affiliation(s)
- K. Mokkala
- Institute of Biomedicine, Department of Medical Microbiology and Immunology, and Functional Foods Forum, University of Turku, 20014 University of Turku, Finland
| | - H. Röytiö
- Institute of Biomedicine and Functional Foods Forum, University of Turku, 20014 University of Turku, Finland
| | - U. Ekblad
- Department of Obstetrics and Gynaecology, University of Turku, 20014 University of Turku, Finland
- Turku University Central Hospital, Kiinamyllynkatu 4-8, 20520 Turku, Finland
| | - K. Laitinen
- Institute of Biomedicine and Functional Foods Forum, University of Turku, 20014 University of Turku, Finland
| |
Collapse
|
109
|
Rosadini CV, Kagan JC. Early innate immune responses to bacterial LPS. Curr Opin Immunol 2017; 44:14-19. [PMID: 27842237 PMCID: PMC5426986 DOI: 10.1016/j.coi.2016.10.005] [Citation(s) in RCA: 233] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 10/28/2016] [Indexed: 12/14/2022]
Abstract
A mammalian receptor for bacterial lipopolysaccharide (LPS), Toll-like receptor 4 (TLR4), plays a beneficial role in controlling bacterial infections, but is also a main driver of aberrant inflammation in lethal sepsis. As a result, investigation of TLR4 signaling has been a major area of research. Despite this focus, our understanding of the mechanisms that regulate TLR4 activities remains primitive. Nowhere is our knowledge of TLR4 biology more lacking than at the receptor-proximal level, where many factors act in concert to regulate LPS signaling. Several recent studies have begun filling these gaps in our knowledge. In this review, we discuss the importance of these receptor proximal activities in the spatiotemporal regulation of TLR4 signaling, and suggest interesting areas for future research.
Collapse
Affiliation(s)
- Charles V Rosadini
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Jonathan C Kagan
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
110
|
Brooks MB, Turk JR, Guerrero A, Narayanan PK, Nolan JP, Besteman EG, Wilson DW, Thomas RA, Fishman CE, Thompson KL, Ellinger-Ziegelbauer H, Pierson JB, Paulman A, Chiang AY, Schultze AE. Non-Lethal Endotoxin Injection: A Rat Model of Hypercoagulability. PLoS One 2017; 12:e0169976. [PMID: 28081568 PMCID: PMC5233421 DOI: 10.1371/journal.pone.0169976] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 12/24/2016] [Indexed: 02/04/2023] Open
Abstract
Systemic inflammation co-activates coagulation, which unchecked culminates in a lethal syndrome of multi-organ microvascular thrombosis known as disseminated intravascular coagulation (DIC). We studied an endotoxin-induced inflammatory state in rats to identify biomarkers of hemostatic imbalance favoring hypercoagulability. Intraperitoneal injection of LPS at 15 mg/kg body weight resulted in peripheral leukopenia and widespread neutrophilic sequestration characteristic of an acute systemic inflammatory response. Early indicators of hemostatic pathway activation developed within 4 hours, including increased circulating concentrations of procoagulant extracellular vesicles (EVs), EVs expressing endothelial cell and platelet membrane markers, and high concentration of soluble intercellular adhesion molecule-1 (sICAM-1), plasminogen activator inhibitor-1 (PAI-1), and D-dimers. Inflammation persisted throughout the 48-hour observation period; however, increases were found in a subset of serum microRNA (miRNA) that coincided with gradual resolution of hemostatic protein abnormalities and reduction in EV counts. Dose-adjusted LPS treatment in rats provides a time-course model to develop biomarker profiles reflecting procoagulant imbalance and rebalance under inflammatory conditions.
Collapse
Affiliation(s)
- Marjory B. Brooks
- Comparative Coagulation Section, Department of Population Medicine and Diagnostic Sciences, Cornell University, Ithaca, NY, United States of America
| | - James R. Turk
- Comparative Biology and Safety Sciences, Amgen Inc., Thousand Oaks, CA, United States of America
| | - Abraham Guerrero
- Comparative Biology and Safety Sciences, Amgen Inc., Thousand Oaks, CA, United States of America
| | - Padma K. Narayanan
- Comparative Biology and Safety Sciences, Amgen Inc., Thousand Oaks, CA, United States of America
| | - John P. Nolan
- Scintillon Institute, San Diego, CA, United States of America
| | - Elizabeth G. Besteman
- Department of Pathology, Safety Assessment and Laboratory Animal Resources, Merck Research Laboratories, West Point, PA, United States of America
| | - Dennis W. Wilson
- Department of Pathology Microbiology and Immunology, School of Veterinary Medicine, University of California-Davis, Davis, CA, United States of America
| | - Roberta A. Thomas
- GlaxoSmithKline, Research and Development, King of Prussia, Pennsylvania, United States of America
| | - Cindy E. Fishman
- GlaxoSmithKline, Research and Development, King of Prussia, Pennsylvania, United States of America
| | - Karol L. Thompson
- Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, United States of America
| | | | - Jennifer B. Pierson
- Health and Environmental Sciences Institute, Suite, Washington, DC, United States of America
| | - April Paulman
- Department of Pathology, Covance Laboratories, Greenfield, IN, United States of America
| | - Alan Y. Chiang
- Global Statistical Sciences, Lilly Research Laboratories, Indianapolis, IN, United States of America
| | - Albert E. Schultze
- Pathology Department, Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN, United States of America
- * E-mail:
| |
Collapse
|
111
|
Tan Y, Kagan JC. Microbe-inducible trafficking pathways that control Toll-like receptor signaling. Traffic 2017; 18:6-17. [PMID: 27731905 PMCID: PMC5182131 DOI: 10.1111/tra.12454] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 10/07/2016] [Accepted: 10/07/2016] [Indexed: 12/12/2022]
Abstract
The receptors of the mammalian innate immune system are designed for rapid microbial detection, and are located in organelles that are conducive to serve these needs. However, emerging evidence indicates that the sites of microbial detection are not the sites of innate immune signal transduction. Rather, microbial detection triggers the movement of receptors to regions of the cell where factors called sorting adaptors detect active receptors and promote downstream inflammatory responses. These findings highlight the critical role that membrane trafficking pathways play in the initiation of innate immunity to infection. In this review, we describe pathways that promote the microbe-inducible endocytosis of Toll-like receptors (TLRs), and the microbe-inducible movement of TLRs between intracellular compartments. We highlight a new class of proteins called Transporters Associated with the eXecution of Inflammation (TAXI), which have the unique ability to transport TLRs and their microbial ligands to signaling-competent regions of the cell, and we discuss the means by which the subcellular sites of signal transduction are defined.
Collapse
Affiliation(s)
- Yunhao Tan
- Harvard Medical School and Division of Gastroenterology, Boston Children’s Hospital, Boston, MA, 02115, USA
| | - Jonathan C. Kagan
- Harvard Medical School and Division of Gastroenterology, Boston Children’s Hospital, Boston, MA, 02115, USA
| |
Collapse
|
112
|
Norsworthy AN, Pearson MM. From Catheter to Kidney Stone: The Uropathogenic Lifestyle of Proteus mirabilis. Trends Microbiol 2016; 25:304-315. [PMID: 28017513 DOI: 10.1016/j.tim.2016.11.015] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 11/17/2016] [Accepted: 11/28/2016] [Indexed: 12/15/2022]
Abstract
Proteus mirabilis is a model organism for urease-producing uropathogens. These diverse bacteria cause infection stones in the urinary tract and form crystalline biofilms on indwelling urinary catheters, frequently leading to polymicrobial infection. Recent work has elucidated how P. mirabilis causes all of these disease states. Particularly exciting is the discovery that this bacterium forms large clusters in the bladder lumen that are sites for stone formation. These clusters, and other steps of infection, require two virulence factors in particular: urease and MR/P fimbriae. Highlighting the importance of MR/P fimbriae is the cotranscribed regulator, MrpJ, which globally controls virulence. Overall, P. mirabilis exhibits an extraordinary lifestyle, and further probing will answer exciting basic microbiological and clinically relevant questions.
Collapse
Affiliation(s)
- Allison N Norsworthy
- Department of Microbiology, New York University Medical Center, New York, NY, USA
| | - Melanie M Pearson
- Department of Microbiology, New York University Medical Center, New York, NY, USA; Department of Urology, New York University Medical Center, New York, NY, USA; Current address: University of Michigan Medical School, Department of Microbiology and Immunology, 5641 Medical Science Building II, 1150 West Medical Center Dr., Ann Arbor, MI 48109-0620, USA.
| |
Collapse
|
113
|
Ryu JK, Kim SJ, Rah SH, Kang JI, Jung HE, Lee D, Lee HK, Lee JO, Park BS, Yoon TY, Kim HM. Reconstruction of LPS Transfer Cascade Reveals Structural Determinants within LBP, CD14, and TLR4-MD2 for Efficient LPS Recognition and Transfer. Immunity 2016; 46:38-50. [PMID: 27986454 DOI: 10.1016/j.immuni.2016.11.007] [Citation(s) in RCA: 257] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 10/08/2016] [Accepted: 10/21/2016] [Indexed: 01/29/2023]
Abstract
Lipopolysaccharide (LPS), the major component of the outer membrane of Gram-negative bacteria, binds Toll-like receptor 4 (TLR4)-MD2 complex and activates innate immune responses. LPS transfer to TLR4-MD2 is catalyzed by both LPS binding protein (LBP) and CD14. To define the sequential molecular interactions underlying this transfer, we reconstituted in vitro the entire LPS transfer process from LPS micelles to TLR4-MD2. Using electron microscopy and single-molecule approaches, we characterized the dynamic intermediate complexes for LPS transfer: LBP-LPS micelles, CD14-LBP-LPS micelle, and CD14-LPS-TLR4-MD2 complex. A single LBP molecule bound longitudinally to LPS micelles catalyzed multi-rounds of LPS transfer to CD14s that rapidly dissociated from LPB-LPS complex upon LPS transfer via electrostatic interactions. Subsequently, the single LPS molecule bound to CD14 was transferred to TLR4-MD2 in a TLR4-dependent manner. The definition of the structural determinants of the LPS transfer cascade to TLR4 may enable the development of targeted therapeutics for intervention in LPS-induced sepsis.
Collapse
Affiliation(s)
- Je-Kyung Ryu
- National Creative Research Initiative Center for Single-Molecule Systems Biology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea; Department of Physics, KAIST, Daejeon 34141, Korea
| | - Soo Jin Kim
- Biomedical Science and Engineering Interdisciplinary Program, KAIST, Daejeon 34141, Korea
| | - Sang-Hyun Rah
- National Creative Research Initiative Center for Single-Molecule Systems Biology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea; Department of Physics, KAIST, Daejeon 34141, Korea; Center for Nanomedicine, Institute for Basic Science (IBS), Yonsei University, Seoul 03722, Korea; Yonsei-IBS Institute, Yonsei University, Seoul 03722, Korea
| | - Ji In Kang
- Biomedical Science and Engineering Interdisciplinary Program, KAIST, Daejeon 34141, Korea
| | - Hi Eun Jung
- Biomedical Science and Engineering Interdisciplinary Program, KAIST, Daejeon 34141, Korea
| | - Dongsun Lee
- Graduate School of Medical Science & Engineering, KAIST, Daejeon 34141, Korea
| | - Heung Kyu Lee
- Biomedical Science and Engineering Interdisciplinary Program, KAIST, Daejeon 34141, Korea; Graduate School of Medical Science & Engineering, KAIST, Daejeon 34141, Korea
| | - Jie-Oh Lee
- Department of Chemistry, KAIST, Daejeon 34141, Korea
| | - Beom Seok Park
- Department of Biomedical Laboratory Science, College of Health Science, Eulji University, Seongnam 461-713, Korea
| | - Tae-Young Yoon
- National Creative Research Initiative Center for Single-Molecule Systems Biology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea; Department of Physics, KAIST, Daejeon 34141, Korea; Center for Nanomedicine, Institute for Basic Science (IBS), Yonsei University, Seoul 03722, Korea; Yonsei-IBS Institute, Yonsei University, Seoul 03722, Korea.
| | - Ho Min Kim
- Biomedical Science and Engineering Interdisciplinary Program, KAIST, Daejeon 34141, Korea; Graduate School of Medical Science & Engineering, KAIST, Daejeon 34141, Korea.
| |
Collapse
|
114
|
Karch CP, Burkhard P. Vaccine technologies: From whole organisms to rationally designed protein assemblies. Biochem Pharmacol 2016; 120:1-14. [PMID: 27157411 PMCID: PMC5079805 DOI: 10.1016/j.bcp.2016.05.001] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 05/04/2016] [Indexed: 11/16/2022]
Abstract
Vaccines have been the single most significant advancement in public health, preventing morbidity and mortality in millions of people annually. Vaccine development has traditionally focused on whole organism vaccines, either live attenuated or inactivated vaccines. While successful for many different infectious diseases whole organisms are expensive to produce, require culture of the infectious agent, and have the potential to cause vaccine associated disease in hosts. With advancing technology and a desire to develop safe, cost effective vaccine candidates, the field began to focus on the development of recombinantly expressed antigens known as subunit vaccines. While more tolerable, subunit vaccines tend to be less immunogenic. Attempts have been made to increase immunogenicity with the addition of adjuvants, either immunostimulatory molecules or an antigen delivery system that increases immune responses to vaccines. An area of extreme interest has been the application of nanotechnology to vaccine development, which allows for antigens to be expressed on a particulate delivery system. One of the most exciting examples of nanovaccines are rationally designed protein nanoparticles. These nanoparticles use some of the basic tenants of structural biology, biophysical chemistry, and vaccinology to develop protective, safe, and easily manufactured vaccines. Rationally developed nanoparticle vaccines are one of the most promising candidates for the future of vaccine development.
Collapse
MESH Headings
- Adjuvants, Immunologic/adverse effects
- Adjuvants, Immunologic/chemistry
- Adjuvants, Immunologic/therapeutic use
- Allergy and Immunology/history
- Allergy and Immunology/trends
- Animals
- Antigens/adverse effects
- Antigens/chemistry
- Antigens/immunology
- Antigens/therapeutic use
- Biopharmaceutics/history
- Biopharmaceutics/methods
- Biopharmaceutics/trends
- Chemistry, Pharmaceutical/history
- Chemistry, Pharmaceutical/trends
- Communicable Disease Control/history
- Communicable Disease Control/trends
- Communicable Diseases/immunology
- Communicable Diseases/veterinary
- Drug Delivery Systems/adverse effects
- Drug Delivery Systems/trends
- Drug Delivery Systems/veterinary
- Drug Design
- History, 19th Century
- History, 20th Century
- History, 21st Century
- Humans
- Nanoparticles/adverse effects
- Nanoparticles/chemistry
- Nanoparticles/therapeutic use
- Protein Engineering/trends
- Protein Engineering/veterinary
- Protein Folding
- Recombinant Proteins/adverse effects
- Recombinant Proteins/chemistry
- Recombinant Proteins/immunology
- Recombinant Proteins/therapeutic use
- Vaccines/adverse effects
- Vaccines/chemistry
- Vaccines/immunology
- Vaccines/therapeutic use
- Vaccines, Subunit/adverse effects
- Vaccines, Subunit/chemistry
- Vaccines, Subunit/immunology
- Vaccines, Subunit/therapeutic use
- Vaccines, Synthetic/adverse effects
- Vaccines, Synthetic/chemistry
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/therapeutic use
- Veterinary Drugs/adverse effects
- Veterinary Drugs/chemistry
- Veterinary Drugs/immunology
- Veterinary Drugs/therapeutic use
Collapse
Affiliation(s)
- Christopher P Karch
- The Institute of Materials Science, 97 North Eagleville Road, Storrs, CT 06269, United States
| | - Peter Burkhard
- The Institute of Materials Science, 97 North Eagleville Road, Storrs, CT 06269, United States; Department of Molecular and Cell Biology, 93 North Eagleville Road, Storrs, CT 06269, United States.
| |
Collapse
|
115
|
De Santis R, Liepelt A, Mossanen JC, Dueck A, Simons N, Mohs A, Trautwein C, Meister G, Marx G, Ostareck-Lederer A, Ostareck DH. miR-155 targets Caspase-3 mRNA in activated macrophages. RNA Biol 2016; 13:43-58. [PMID: 26574931 DOI: 10.1080/15476286.2015.1109768] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
To secure the functionality of activated macrophages in the innate immune response, efficient life span control is required. Recognition of bacterial lipopolysaccharides (LPS) by toll-like receptor 4 (TLR4) induces downstream signaling pathways, which merge to induce the expression of cytokine genes and anti-apoptotic genes. MicroRNAs (miRNAs) have emerged as important inflammatory response modulators, but information about their functional impact on apoptosis is scarce. To identify miRNAs differentially expressed in response to LPS, cDNA libraries from untreated and LPS-activated murine macrophages were analyzed by deep sequencing and regulated miRNA expression was verified by Northern blotting and qPCR. Employing TargetScan(TM) we identified CASPASE-3 (CASP-3) mRNA that encodes a key player in apoptosis as potential target of LPS-induced miR-155. LPS-dependent primary macrophage activation revealed TLR4-mediated enhancement of miR-155 expression and CASP-3 mRNA reduction. Endogenous CASP-3 and cleaved CASP-3 protein declined in LPS-activated macrophages. Accumulation of miR-155 and CASP-3 mRNA in miRNA-induced silencing complexes (miRISC) was demonstrated by ARGONAUTE 2 (AGO2) immunoprecipitation. Importantly, specific antagomir transfection effectively reduced mature miR-155 and resulted in significantly elevated CASP-3 mRNA levels in activated macrophages. In vitro translation assays demonstrated that the target site in the CASP-3 mRNA 3'UTR mediates miR-155-dependent Luciferase reporter mRNA destabilization. Strikingly, Annexin V staining of macrophages transfected with antagomir-155 and stimulated with LPS prior to staurosporine (SSP) treatment implied that LPS-induced miR-155 prevents apoptosis through CASP-3 mRNA down-regulation. In conclusion, we report that miR-155-mediated CASP-3 mRNA destabilization in LPS-activated RAW 264.7 macrophages suppresses apoptosis, as a prerequisite to maintain their crucial function in inflammation.
Collapse
Affiliation(s)
- Rebecca De Santis
- a Department of Intensive Care and Intermediate Care , University Hospital, RWTH Aachen University , Pauwelsstrasse 30, 52074 , Aachen , Germany
| | - Anke Liepelt
- a Department of Intensive Care and Intermediate Care , University Hospital, RWTH Aachen University , Pauwelsstrasse 30, 52074 , Aachen , Germany.,b Department of Internal Medicine III , University Hospital, RWTH Aachen University , Pauwelsstrasse 30, 52074 , Aachen , Germany
| | - Jana C Mossanen
- a Department of Intensive Care and Intermediate Care , University Hospital, RWTH Aachen University , Pauwelsstrasse 30, 52074 , Aachen , Germany
| | - Anne Dueck
- c Biochemistry Center Regensburg (BZR) , Laboratory for RNA Biology, University of Regensburg , Universitätsstrasse 31, 93053 , Regensburg , Germany
| | - Nadine Simons
- a Department of Intensive Care and Intermediate Care , University Hospital, RWTH Aachen University , Pauwelsstrasse 30, 52074 , Aachen , Germany
| | - Antje Mohs
- b Department of Internal Medicine III , University Hospital, RWTH Aachen University , Pauwelsstrasse 30, 52074 , Aachen , Germany
| | - Christian Trautwein
- b Department of Internal Medicine III , University Hospital, RWTH Aachen University , Pauwelsstrasse 30, 52074 , Aachen , Germany
| | - Gunter Meister
- c Biochemistry Center Regensburg (BZR) , Laboratory for RNA Biology, University of Regensburg , Universitätsstrasse 31, 93053 , Regensburg , Germany
| | - Gernot Marx
- a Department of Intensive Care and Intermediate Care , University Hospital, RWTH Aachen University , Pauwelsstrasse 30, 52074 , Aachen , Germany
| | - Antje Ostareck-Lederer
- a Department of Intensive Care and Intermediate Care , University Hospital, RWTH Aachen University , Pauwelsstrasse 30, 52074 , Aachen , Germany
| | - Dirk H Ostareck
- a Department of Intensive Care and Intermediate Care , University Hospital, RWTH Aachen University , Pauwelsstrasse 30, 52074 , Aachen , Germany
| |
Collapse
|
116
|
LPS-Induced Macrophage Activation and Plasma Membrane Fluidity Changes are Inhibited Under Oxidative Stress. J Membr Biol 2016; 249:789-800. [PMID: 27619206 DOI: 10.1007/s00232-016-9927-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 08/30/2016] [Indexed: 10/21/2022]
Abstract
Macrophage activation is essential for a correct and efficient response of innate immunity. During oxidative stress membrane receptors and/or membrane lipid dynamics can be altered, leading to dysfunctional cell responses. Our aim is to analyze membrane fluidity modifications and cell function under oxidative stress in LPS-activated macrophages. Membrane fluidity of individual living THP-1 macrophages was evaluated by the technique two-photon microscopy. LPS-activated macrophage function was determined by TNFα secretion. It was shown that LPS activation causes fluidification of macrophage plasma membrane and production of TNFα. However, oxidative stress induces rigidification of macrophage plasma membrane and inhibition of cell activation, which is evidenced by a decrease of TNFα secretion. Thus, under oxidative conditions macrophage proinflammatory response might develop in an inefficient manner.
Collapse
|
117
|
Li L, Yu Y, Zhou Z, Zhou JM. Plant pattern-recognition receptors controlling innate immunity. SCIENCE CHINA-LIFE SCIENCES 2016; 59:878-88. [DOI: 10.1007/s11427-016-0115-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 06/02/2016] [Indexed: 11/24/2022]
|
118
|
Rösch C, Taverne N, Venema K, Gruppen H, Wells JM, Schols HA. Effects ofin vitrofermentation of barley β-glucan and sugar beet pectin using human fecal inocula on cytokine expression by dendritic cells. Mol Nutr Food Res 2016; 61. [DOI: 10.1002/mnfr.201600243] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 06/14/2016] [Accepted: 06/19/2016] [Indexed: 12/20/2022]
Affiliation(s)
- Christiane Rösch
- Laboratory of Food Chemistry; Wageningen University; Wageningen the Netherlands
- Top Institute Food and Nutrition; Wageningen the Netherlands
| | - Nico Taverne
- Host Microbe Interactomics; Wageningen University; Wageningen the Netherlands
| | - Koen Venema
- Top Institute Food and Nutrition; Wageningen the Netherlands
- Beneficial Microbes Consultancy; Wageningen the Netherlands
| | - Harry Gruppen
- Laboratory of Food Chemistry; Wageningen University; Wageningen the Netherlands
| | - Jerry M. Wells
- Host Microbe Interactomics; Wageningen University; Wageningen the Netherlands
| | - Henk A. Schols
- Laboratory of Food Chemistry; Wageningen University; Wageningen the Netherlands
- Top Institute Food and Nutrition; Wageningen the Netherlands
| |
Collapse
|
119
|
Affiliation(s)
- Stefanie Ranf
- Phytopathology, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising-Weihenstephan, Germany
- * E-mail:
| |
Collapse
|
120
|
He Z, Riva M, Björk P, Swärd K, Mörgelin M, Leanderson T, Ivars F. CD14 Is a Co-Receptor for TLR4 in the S100A9-Induced Pro-Inflammatory Response in Monocytes. PLoS One 2016; 11:e0156377. [PMID: 27228163 PMCID: PMC4881898 DOI: 10.1371/journal.pone.0156377] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 05/13/2016] [Indexed: 01/23/2023] Open
Abstract
The cytosolic Ca2+-binding S100A9 and S100A8 proteins form heterodimers that are primarily expressed in human neutrophils and monocytes. We have recently shown that S100A9 binds to TLR4 in vitro and induces TLR4-dependent NF-κB activation and a pro-inflammatory cytokine response in monocytes. In the present report we have further investigated the S100A9-mediated stimulation of TLR4 in monocytes. Using transmission immunoelectron microscopy, we detected focal binding of S100A9 to monocyte membrane subdomains containing the caveolin-1 protein and TLR4. Furthermore, the S100A9 protein was detected in early endosomes of the stimulated cells, indicating that the protein could be internalized by endocytosis. Although stimulation of monocytes with S100A9 was strictly TLR4-dependent, binding of S100A9 to the plasma membrane and endocytosis of S100A9 was still detectable and coincided with CD14 expression in TLR4-deficient cells. We therefore investigated whether CD14 would be involved in the TLR4-dependent stimulation and could show that the S100A9-induced cytokine response was inhibited both in CD14-deficient cells and in cells exposed to CD14 blocking antibodies. Further, S100A9 was not internalized into CD14-deficient cells suggesting a direct role of CD14 in endocytosis of S100A9. Finally, we could detect satiable binding of S100A9 to CD14 in surface plasmon resonance experiments. Taken together, these results indicate that CD14 is a co-receptor of TLR4 in the S100A9-induced cytokine response.
Collapse
Affiliation(s)
- Zhifei He
- Immunology group, Section for Immunology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Matteo Riva
- Immunology group, Section for Immunology, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Active Biotech AB, Lund, Sweden
| | | | - Karl Swärd
- Section for Cell and Tissue Biology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Matthias Mörgelin
- Section for Infection Biology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Tomas Leanderson
- Immunology group, Section for Immunology, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Active Biotech AB, Lund, Sweden
| | - Fredrik Ivars
- Immunology group, Section for Immunology, Department of Experimental Medical Science, Lund University, Lund, Sweden
- * E-mail:
| |
Collapse
|
121
|
Liu J, Wang R, Ma D, Ouyang D, Xi Z. Efficient construction of stable gene nanoparticles through polymerase chain reaction with flexible branched primers for gene delivery. Chem Commun (Camb) 2016; 51:9208-11. [PMID: 25952052 DOI: 10.1039/c5cc01788b] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Flexible branched primers were designed to construct stable gene nanoparticles with multiple target gene copies through polymerase chain reaction, which can be used as an efficient transcription template in eukaryotic cells for gene delivery.
Collapse
Affiliation(s)
- Jianbing Liu
- Department of Chemical Biology, State Key Laboratory of Elemento-Organic Chemistry, National Engineering Research Center of Pesticide (Tianjin), Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, China.
| | | | | | | | | |
Collapse
|
122
|
Sun J, Li N, Oh KS, Dutta B, Vayttaden SJ, Lin B, Ebert TS, De Nardo D, Davis J, Bagirzadeh R, Lounsbury NW, Pasare C, Latz E, Hornung V, Fraser IDC. Comprehensive RNAi-based screening of human and mouse TLR pathways identifies species-specific preferences in signaling protein use. Sci Signal 2016; 9:ra3. [PMID: 26732763 PMCID: PMC5381726 DOI: 10.1126/scisignal.aab2191] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Toll-like receptors (TLRs) are a major class of pattern recognition receptors, which mediate the responses of innate immune cells to microbial stimuli. To systematically determine the roles of proteins in canonical TLR signaling pathways, we conducted an RNA interference (RNAi)-based screen in human and mouse macrophages. We observed a pattern of conserved signaling module dependencies across species, but found notable species-specific requirements at the level of individual proteins. Among these, we identified unexpected differences in the involvement of members of the interleukin-1 receptor-associated kinase (IRAK) family between the human and mouse TLR pathways. Whereas TLR signaling in mouse macrophages depended primarily on IRAK4 and IRAK2, with little or no role for IRAK1, TLR signaling and proinflammatory cytokine production in human macrophages depended on IRAK1, with knockdown of IRAK4 or IRAK2 having less of an effect. Consistent with species-specific roles for these kinases, IRAK4 orthologs failed to rescue signaling in IRAK4-deficient macrophages from the other species, and only mouse macrophages required the kinase activity of IRAK4 to mediate TLR responses. The identification of a critical role for IRAK1 in TLR signaling in humans could potentially explain the association of IRAK1 with several autoimmune diseases. Furthermore, this study demonstrated how systematic screening can be used to identify important characteristics of innate immune responses across species, which could optimize therapeutic targeting to manipulate human TLR-dependent outputs.
Collapse
Affiliation(s)
- Jing Sun
- Signaling Systems Unit, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ning Li
- Signaling Systems Unit, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kyu-Seon Oh
- Signaling Systems Unit, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bhaskar Dutta
- Bioinformatics Team, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sharat J Vayttaden
- Signaling Systems Unit, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bin Lin
- Signaling Systems Unit, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Thomas S Ebert
- Institute of Molecular Medicine, University Hospital, University of Bonn, 53127 Bonn, Germany
| | - Dominic De Nardo
- Institute of Innate Immunity, University Hospital, Biomedical Centre, University of Bonn, 53127 Bonn, Germany. Inflammation Division, Walter and Eliza Hall Institute, Parkville,Victoria 3052, Australia. Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Joie Davis
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rustam Bagirzadeh
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Nicolas W Lounsbury
- Signaling Systems Unit, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chandrashekhar Pasare
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital, Biomedical Centre, University of Bonn, 53127 Bonn, Germany. German Center for Neurodegenerative Diseases (DZNE), 53175 Bonn, Germany. Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Veit Hornung
- Institute of Molecular Medicine, University Hospital, University of Bonn, 53127 Bonn, Germany
| | - Iain D C Fraser
- Signaling Systems Unit, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
123
|
Liu J, Li Y, Ma D, Ouyang D, Xi Z. Flexible DNA junction assisted efficient construction of stable gene nanoparticles for gene delivery. Chem Commun (Camb) 2016; 52:1953-6. [DOI: 10.1039/c5cc07949g] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A flexible DNA junction was designed to construct stable gene nanoparticles, which can be used as efficient gene cargo for eukaryotic cells.
Collapse
Affiliation(s)
- Jianbing Liu
- Department of Chemical Biology
- State Key Laboratory of Elemento-Organic Chemistry
- National Engineering Research Center of Pesticide (Tianjin)
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin)
- Nankai University
| | - Yanyan Li
- Department of Chemical Biology
- State Key Laboratory of Elemento-Organic Chemistry
- National Engineering Research Center of Pesticide (Tianjin)
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin)
- Nankai University
| | - Dejun Ma
- Department of Chemical Biology
- State Key Laboratory of Elemento-Organic Chemistry
- National Engineering Research Center of Pesticide (Tianjin)
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin)
- Nankai University
| | - Di Ouyang
- Department of Chemical Biology
- State Key Laboratory of Elemento-Organic Chemistry
- National Engineering Research Center of Pesticide (Tianjin)
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin)
- Nankai University
| | - Zhen Xi
- Department of Chemical Biology
- State Key Laboratory of Elemento-Organic Chemistry
- National Engineering Research Center of Pesticide (Tianjin)
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin)
- Nankai University
| |
Collapse
|
124
|
Rowe HM, Huntley JF. From the Outside-In: The Francisella tularensis Envelope and Virulence. Front Cell Infect Microbiol 2015; 5:94. [PMID: 26779445 PMCID: PMC4688374 DOI: 10.3389/fcimb.2015.00094] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 12/07/2015] [Indexed: 12/20/2022] Open
Abstract
Francisella tularensis is a highly-infectious bacterium that causes the rapid, and often lethal disease, tularemia. Many studies have been performed to identify and characterize the virulence factors that F. tularensis uses to infect a wide variety of hosts and host cell types, evade immune defenses, and induce severe disease and death. This review focuses on the virulence factors that are present in the F. tularensis envelope, including capsule, LPS, outer membrane, periplasm, inner membrane, secretion systems, and various molecules in each of aforementioned sub-compartments. Whereas, no single bacterial molecule or molecular complex single-handedly controls F. tularensis virulence, we review here how diverse bacterial systems work in conjunction to subvert the immune system, attach to and invade host cells, alter phagosome/lysosome maturation pathways, replicate in host cells without being detected, inhibit apoptosis, and induce host cell death for bacterial release and infection of adjacent cells. Given that the F. tularensis envelope is the outermost layer of the bacterium, we highlight herein how many of these molecules directly interact with the host to promote infection and disease. These and future envelope studies are important to advance our collective understanding of F. tularensis virulence mechanisms and offer targets for future vaccine development efforts.
Collapse
Affiliation(s)
- Hannah M Rowe
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences Toledo, OH, USA
| | - Jason F Huntley
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences Toledo, OH, USA
| |
Collapse
|
125
|
Wolfson ML, Schander JA, Bariani MV, Correa F, Franchi AM. Progesterone modulates the LPS-induced nitric oxide production by a progesterone-receptor independent mechanism. Eur J Pharmacol 2015; 769:110-6. [DOI: 10.1016/j.ejphar.2015.11.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 10/30/2015] [Accepted: 11/02/2015] [Indexed: 12/15/2022]
|
126
|
Kudo K, Uchida T, Sawada M, Nakamura Y, Yoneda A, Fukami K. Phospholipase C δ1 in macrophages negatively regulates TLR4-induced proinflammatory cytokine production and Fcγ receptor-mediated phagocytosis. Adv Biol Regul 2015; 61:68-79. [PMID: 26643908 DOI: 10.1016/j.jbior.2015.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 11/14/2015] [Accepted: 11/14/2015] [Indexed: 12/26/2022]
Abstract
Macrophages are key players in the innate immune response. Turnover of phosphoinositides (PI), particularly phosphatidylinositol 4,5 bisphosphate (PI(4,5)P2), has been implicated in macrophage functions such as toll-like receptor (TLR)-mediated cytokine production and phagocytosis. However, PI metabolizing enzymes responsible for macrophage functions are not well defined. The phospholipase C (PLC) family of enzymes is critical in PI(4,5)P2 turnover. In this study, we investigated the role of PLCδ1, a prototype PLC, in macrophages on the expression of inflammation-associated genes and phagocytosis. Lipopolysaccharides (LPS) signal through TLR4 to produce proinflammatory cytokines such as interleukin (IL)-1β. LPS stimulation of both RAW264.7 murine macrophages and murine bone marrow-derived macrophages resulted in lower PLCδ1 mRNA and protein expression levels, compared to that in the control. Using chemical inhibitor compounds, we demonstrated that the up-regulation of p38 MAPK activity led to down-regulation of PLCδ1 mRNA expression in macrophages. PLCδ1 reduction by RNAi or gene deletion resulted in greater LPS-induced IL-1β expression than that observed in the control siRNA-treated cells, without increasing TLR4 cell surface expression. PLCδ1 also negatively regulated LPS-induced cell spreading. Analysis of Fcγ receptor-mediated phagocytosis demonstrated an increased phagocytosis index after PLCδ1 knockdown in RAW264.7 cells. Conversely, overexpression of PLCδ1 reduced phagocytosis whereas catalytic inactive PLCδ1 had no effect. Altered levels of PLCδ1 affected the binding of opsonized latex beads with cells, rather than the phagocytic activity. Taken together, the data suggest that PLCδ1 negatively regulates LPS-induced production of IL-1β and Fcγ receptor-mediated phagocytosis in macrophages.
Collapse
Affiliation(s)
- Kohya Kudo
- Laboratory of Genome and Biosignals, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Takafumi Uchida
- Laboratory of Genome and Biosignals, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Mayu Sawada
- Laboratory of Genome and Biosignals, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yoshikazu Nakamura
- Laboratory of Genome and Biosignals, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan; AMED-CREST, AMED, Tokyo, Japan
| | - Atsuko Yoneda
- Laboratory of Genome and Biosignals, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan; AMED-CREST, AMED, Tokyo, Japan
| | - Kiyoko Fukami
- Laboratory of Genome and Biosignals, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan; AMED-CREST, AMED, Tokyo, Japan.
| |
Collapse
|
127
|
Tan Y, Zanoni I, Cullen TW, Goodman AL, Kagan JC. Mechanisms of Toll-like Receptor 4 Endocytosis Reveal a Common Immune-Evasion Strategy Used by Pathogenic and Commensal Bacteria. Immunity 2015; 43:909-22. [PMID: 26546281 PMCID: PMC4685471 DOI: 10.1016/j.immuni.2015.10.008] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 09/28/2015] [Accepted: 10/05/2015] [Indexed: 01/20/2023]
Abstract
Microbe-induced receptor trafficking has emerged as an essential means to promote innate immune signal transduction. Upon detection of bacterial lipopolysaccharides (LPS), CD14 induces an inflammatory endocytosis pathway that delivers Toll-like receptor 4 (TLR4) to endosomes. Although several regulators of CD14-dependent TLR4 endocytosis have been identified, the cargo-selection mechanism during this process remains unknown. We reveal that, in contrast to classic cytosolic interactions that promoted the endocytosis of transmembrane receptors, TLR4 was selected as cargo for inflammatory endocytosis entirely through extracellular interactions. Mechanistically, the extracellular protein MD-2 bound to and dimerized TLR4 in order to promote this endocytic event. Our analysis of LPS variants from human pathogens and gut commensals revealed a common mechanism by which bacteria prevent inflammatory endocytosis. We suggest that evasion of CD14-dependent endocytosis is an attribute that transcends the concept of pathogenesis and might be a fundamental feature of bacteria that inhabit eukaryotic hosts.
Collapse
Affiliation(s)
- Yunhao Tan
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Ivan Zanoni
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan 20126, Italy; Unit of Cell Signalling and Innate Immunity, Humanitas Clinical and Research Center, Rozzano 20089, Italy
| | - Thomas W Cullen
- Department of Microbial Pathogenesis and Microbial Sciences Institute, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Andrew L Goodman
- Department of Microbial Pathogenesis and Microbial Sciences Institute, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jonathan C Kagan
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
128
|
Aedo JE, Reyes AE, Avendaño-Herrera R, Molina A, Valdés JA. Bacterial lipopolysaccharide induces rainbow trout myotube atrophy via Akt/FoxO1/Atrogin-1 signaling pathway. Acta Biochim Biophys Sin (Shanghai) 2015; 47:932-7. [PMID: 26341977 DOI: 10.1093/abbs/gmv087] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 06/28/2015] [Indexed: 02/07/2023] Open
Abstract
Lipopolysaccharide (LPS) is considered as a powerful inducer of muscle atrophy in higher vertebrates due to skeletal muscle cell recognition of the endotoxin and a consequent activation of catabolic signaling pathways. In contrast, there is no evidence of LPS directly inducing skeletal muscle atrophy in lower vertebrates, such as fish. For years it has been assumed that fish are resistant to LPS, mainly due to differences in the key features of toll-like receptor (TLR) signaling pathways when compared with mammals. In this study, we report that the stimulation of cultured rainbow trout (Oncorhynchus mykiss) myotubes with LPS (100 ng/ml) resulted in a transient decrease in the pAkt/Akt ratio, a subsequent reduction in the pFoxO1/FoxO1 ratio, and a significant increase in atrogin-1 transcript expression. Preincubation with polymyxin B, an LPS-neutralizing agent, and 740 Y-P, an agonist of p85-PI3K, blocked the effects of LPS. Additionally, LPS treatment induced an increase in protein ubiquitination and a reduction in myotube diameter, both of which are associated with muscular atrophy that is not observed under polymyxin B and 740 Y-P pretreatments. Finally, rainbow trout myotubes expressed the genes tlr1, tlr3, tlr5m, tlr8a1, tlr8a2, tlr9, and tlr22, with significantly increased expressions of tlr5m and tlr9 under LPS stimulation. These results indicate that LPS is an inducer of fish skeletal muscle atrophy and suggest that TLR5M and TLR9 may play important roles in detecting LPS, which supports for the first time the hypothesis that LPS is a direct inducer of skeletal muscle atrophy in teleost species.
Collapse
Affiliation(s)
- J E Aedo
- Facultad de Ciencias Biológicas, Universidad Andrés Bello, Santiago 8370146, Chile
| | - A E Reyes
- Facultad de Ciencias Biológicas, Universidad Andrés Bello, Santiago 8370146, Chile Interdisciplinary Center for Aquaculture Research (INCAR), Víctor Lamas 1290, PO Box 160-C, Concepción, Chile
| | - R Avendaño-Herrera
- Facultad de Ciencias Biológicas, Universidad Andrés Bello, Santiago 8370146, Chile Interdisciplinary Center for Aquaculture Research (INCAR), Víctor Lamas 1290, PO Box 160-C, Concepción, Chile
| | - A Molina
- Facultad de Ciencias Biológicas, Universidad Andrés Bello, Santiago 8370146, Chile Interdisciplinary Center for Aquaculture Research (INCAR), Víctor Lamas 1290, PO Box 160-C, Concepción, Chile
| | - J A Valdés
- Facultad de Ciencias Biológicas, Universidad Andrés Bello, Santiago 8370146, Chile Interdisciplinary Center for Aquaculture Research (INCAR), Víctor Lamas 1290, PO Box 160-C, Concepción, Chile
| |
Collapse
|
129
|
Encyclopedia of bacterial gene circuits whose presence or absence correlate with pathogenicity--a large-scale system analysis of decoded bacterial genomes. BMC Genomics 2015; 16:773. [PMID: 26459834 PMCID: PMC4603813 DOI: 10.1186/s12864-015-1957-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 09/28/2015] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Bacterial infections comprise a global health challenge as the incidences of antibiotic resistance increase. Pathogenic potential of bacteria has been shown to be context dependent, varying in response to environment and even within the strains of the same genus. RESULTS We used the KEGG repository and extensive literature searches to identify among the 2527 bacterial genomes in the literature those implicated as pathogenic to the host, including those which show pathogenicity in a context dependent manner. Using data on the gene contents of these genomes, we identified sets of genes highly abundant in pathogenic but relatively absent in commensal strains and vice versa. In addition, we carried out genome comparison within a genus for the seventeen largest genera in our genome collection. We projected the resultant lists of ortholog genes onto KEGG bacterial pathways to identify clusters and circuits, which can be linked to either pathogenicity or synergy. Gene circuits relatively abundant in nonpathogenic bacteria often mediated biosynthesis of antibiotics. Other synergy-linked circuits reduced drug-induced toxicity. Pathogen-abundant gene circuits included modules in one-carbon folate, two-component system, type-3 secretion system, and peptidoglycan biosynthesis. Antibiotics-resistant bacterial strains possessed genes modulating phagocytosis, vesicle trafficking, cytoskeletal reorganization, and regulation of the inflammatory response. Our study also identified bacterial genera containing a circuit, elements of which were previously linked to Alzheimer's disease. CONCLUSIONS Present study produces for the first time, a signature, in the form of a robust list of gene circuitry whose presence or absence could potentially define the pathogenicity of a microbiome. Extensive literature search substantiated a bulk majority of the commensal and pathogenic circuitry in our predicted list. Scanning microbiome libraries for these circuitry motifs will provide further insights into the complex and context dependent pathogenicity of bacteria.
Collapse
|
130
|
Choudhury S, Ghosh S, Gupta P, Mukherjee S, Chattopadhyay S. Inflammation-induced ROS generation causes pancreatic cell death through modulation of Nrf2/NF-κB and SAPK/JNK pathway. Free Radic Res 2015; 49:1371-83. [PMID: 26189548 DOI: 10.3109/10715762.2015.1075016] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chronic pancreatitis is characterized by progressive loss of exocrine and endocrine functions of the pancreas and is considered to be the single most important cause for development of pancreatic cancer. Recent evidence suggests that inflammation and oxidative stress play pivotal roles in the development of clinical conditions like pancreatitis, type 2 diabetes mellitus, and metabolic syndrome. Nonetheless, molecular signaling pathways linking inflammation, oxidative stress, and pancreatic cell death are not yet well defined. In this study, bacterial lipopolysaccharide (LPS) was used (injected twice a week for three weeks) to emulate a chronic systemic inflammatory state in experimental Swiss albino mice. Using this model, we traced the genesis of inflammation-induced pancreatic dysfunction and mapped the signaling events which contribute to the induction of this state. Histopathological studies revealed the appearance of cell injuries and increased collagen content in LPS-exposed group, indicative of fibrosis. Assays for intraperitoneal glucose tolerance, insulin levels, and insulin receptor mRNA expression signified inflammation-induced insulin insensitivity. For the first time we present evidence that cellular inflammation and subsequent oxidative stress modulate the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)/NF-E2-related factor 2 or Nuclear factor (erythroid-derived 2)-like 2 pathway and initiates pancreatic cell death by activation of stress-responsive Rho/stress-activated protein kinase or SAPK/Jun-N-terminal kinase (JNK) pathway. Scavenging of intracellular reactive oxygen species (ROS) by a standard antioxidant N-acetyl cysteine led to pancreatic cell survival. The data obtained strongly indicates that the LPS/toll-like receptor-4 or TLR-4/ROS/NF-κB pathway is critically involved in the initiation of inflammation, oxidative stress, and pancreatic cell death and might prove to be an excellent choice as a target for novel therapeutic strategies in the management of metabolic disorders.
Collapse
Affiliation(s)
- S Choudhury
- a Department of Physiology , University of Calcutta , Kolkata , India
| | - S Ghosh
- a Department of Physiology , University of Calcutta , Kolkata , India
| | - P Gupta
- a Department of Physiology , University of Calcutta , Kolkata , India
| | - S Mukherjee
- a Department of Physiology , University of Calcutta , Kolkata , India
| | - S Chattopadhyay
- a Department of Physiology , University of Calcutta , Kolkata , India.,b Centre for Research in Nanoscience and Nanotechnology, University of Calcutta , Kolkata , India
| |
Collapse
|
131
|
Chebrolu C, Artner D, Sigmund AM, Buer J, Zamyatina A, Kirschning CJ. Species and mediator specific TLR4 antagonism in primary human and murine immune cells by βGlcN(1↔1)αGlc based lipid A mimetics. Mol Immunol 2015; 67:636-41. [PMID: 26319313 DOI: 10.1016/j.molimm.2015.07.037] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 06/18/2015] [Accepted: 07/29/2015] [Indexed: 12/25/2022]
Abstract
Immune stimulatory pathogen associated molecular patterns (PAMPs) are major drivers of infection pathology. Infections with Gram-negative bacteria or negatively polar and single stranded RNA influenza virus are prominent causes of morbidity and mortality. Toll-like receptor (TLR) 4 is a major host sensor for both of the two infections. In order to inhibit TLR4 driven immune activation we recently developed synthetic tetra-acylated lipid A mimetics based on a conformationally restricted βGlcN(1↔1)αGlcN disaccharide scaffold (DA-compounds) that antagonized ectopically overexpressed human and murine TLR4/MD-2 complexes. Here we comparatively analyzed human peripheral blood mononuclear cell (hPBMC) and murine bone marrow derived macrophage (mBM) activation upon 30 min of preincubation in vitro with six variably acylated DA-compounds. 16 h subsequent to consequent LPS challenge, we sampled culture supernatants for cytokine and NO concentration analysis. Four compounds significantly inhibited release of both TNF and IL-6 by hPBMCs upon LPS challenge. In contrast, three compounds effectively inhibited mBM production of MIP-2 and KC, and even five of them inhibited IL-6 and NO production. LPS driven like other TLR ligand driven mBM TNF release was largely unimpaired. The inhibitory effect was specific in that Clo75 driven cytokine release by both hPBMCs and mBMs was unimpaired by the compounds analyzed. Our results indicate biological species specificity of LPS antagonism by variably tetraacylated lipid A mimetics and validate three out of six DA-antagonists as promising candidates for development of therapeutically applicable anti-inflammatory compounds.
Collapse
Affiliation(s)
- Chiranjeevi Chebrolu
- Institute of Medical Microbiology, University of Duisburg-Essen, 45147 Essen, Germany
| | - Daniel Artner
- Department of Chemistry, University of Natural Resources and Life Sciences, 1190 Vienna, Austria
| | - Anna M Sigmund
- Institute of Medical Microbiology, University of Duisburg-Essen, 45147 Essen, Germany
| | - Jan Buer
- Institute of Medical Microbiology, University of Duisburg-Essen, 45147 Essen, Germany
| | - Alla Zamyatina
- Department of Chemistry, University of Natural Resources and Life Sciences, 1190 Vienna, Austria
| | - Carsten J Kirschning
- Institute of Medical Microbiology, University of Duisburg-Essen, 45147 Essen, Germany.
| |
Collapse
|
132
|
Wang Y, Shan X, Chen G, Jiang L, Wang Z, Fang Q, Liu X, Wang J, Zhang Y, Wu W, Liang G. MD-2 as the target of a novel small molecule, L6H21, in the attenuation of LPS-induced inflammatory response and sepsis. Br J Pharmacol 2015; 172:4391-405. [PMID: 26076332 DOI: 10.1111/bph.13221] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 05/27/2015] [Accepted: 06/06/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Myeloid differentiation 2 (MD-2) recognizes LPS, which is required for TLR4 activation, and represents an attractive therapeutic target for severe inflammatory disorders. We previously found that a chalcone derivative, L6H21, could inhibit LPS-induced overexpression of TNF-α and IL-6 in macrophages. Here, we performed a series of biochemical experiments to investigate whether L6H21 specifically targets MD-2 and inhibits the interaction and signalling transduction of LPS-TLR4/MD-2. EXPERIMENTAL APPROACH The binding affinity of L6H21 to MD-2 protein was analysed using computer docking, surface plasmon resonance analysis, elisa, fluorescence measurements and flow cytometric analysis. The effects of L6H21 on MAPK and NF-κB signalling were determined using EMSA, fluorescence staining, Western blotting and immunoprecipitation. The anti-inflammatory effects of L6H21 were confirmed using elisa and RT-qPCR in vitro. The anti-inflammatory effects of L6H21 were also evaluated in septic C57BL/6 mice. KEY RESULTS Compound L6H21 inserted into the hydrophobic region of the MD-2 pocket, forming hydrogen bonds with Arg(90) and Tyr(102) in the MD-2 pocket. In vitro, L6H21 subsequently suppressed MAPK phosphorylation, NF-κB activation and cytokine expression in macrophages stimulated by LPS. In vivo, L6H21 pretreatment improved survival, prevented lung injury, decreased serum and hepatic cytokine levels in mice subjected to LPS. In addition, mice with MD-2 gene knockout were universally protected from the effects of LPS-induced septic shock. CONCLUSIONS AND IMPLICATIONS Overall, this work demonstrated that the new chalcone derivative, L6H21, is a potential candidate for the treatment of sepsis. More importantly, the data confirmed that MD-2 is an important therapeutic target for inflammatory disorders.
Collapse
Affiliation(s)
- Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoou Shan
- Department of Paediatrics, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Gaozhi Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lili Jiang
- Department of Paediatrics, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhe Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qilu Fang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xing Liu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jingying Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yali Zhang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wencan Wu
- Department of Orbital and Oculoplastic Surgery, The Eye Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
133
|
Kell D, Potgieter M, Pretorius E. Individuality, phenotypic differentiation, dormancy and 'persistence' in culturable bacterial systems: commonalities shared by environmental, laboratory, and clinical microbiology. F1000Res 2015; 4:179. [PMID: 26629334 PMCID: PMC4642849 DOI: 10.12688/f1000research.6709.2] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/04/2015] [Indexed: 01/28/2023] Open
Abstract
For bacteria, replication mainly involves growth by binary fission. However, in a very great many natural environments there are examples of phenotypically dormant, non-growing cells that do not replicate immediately and that are phenotypically 'nonculturable' on media that normally admit their growth. They thereby evade detection by conventional culture-based methods. Such dormant cells may also be observed in laboratory cultures and in clinical microbiology. They are usually more tolerant to stresses such as antibiotics, and in clinical microbiology they are typically referred to as 'persisters'. Bacterial cultures necessarily share a great deal of relatedness, and inclusive fitness theory implies that there are conceptual evolutionary advantages in trading a variation in growth rate against its mean, equivalent to hedging one's bets. There is much evidence that bacteria exploit this strategy widely. We here bring together data that show the commonality of these phenomena across environmental, laboratory and clinical microbiology. Considerable evidence, using methods similar to those common in environmental microbiology, now suggests that many supposedly non-communicable, chronic and inflammatory diseases are exacerbated (if not indeed largely caused) by the presence of dormant or persistent bacteria (the ability of whose components to cause inflammation is well known). This dormancy (and resuscitation therefrom) often reflects the extent of the availability of free iron. Together, these phenomena can provide a ready explanation for the continuing inflammation common to such chronic diseases and its correlation with iron dysregulation. This implies that measures designed to assess and to inhibit or remove such organisms (or their access to iron) might be of much therapeutic benefit.
Collapse
Affiliation(s)
- Douglas Kell
- School of Chemistry and The Manchester Institute of Biotechnology, The University of Manchester, Manchester, Lancashire, M1 7DN, UK
| | - Marnie Potgieter
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia, 0007, South Africa
| | - Etheresia Pretorius
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia, 0007, South Africa
| |
Collapse
|
134
|
Kell D, Potgieter M, Pretorius E. Individuality, phenotypic differentiation, dormancy and 'persistence' in culturable bacterial systems: commonalities shared by environmental, laboratory, and clinical microbiology. F1000Res 2015; 4:179. [PMID: 26629334 DOI: 10.12688/f1000research.6709.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/29/2015] [Indexed: 01/28/2023] Open
Abstract
For bacteria, replication mainly involves growth by binary fission. However, in a very great many natural environments there are examples of phenotypically dormant, non-growing cells that do not replicate immediately and that are phenotypically 'nonculturable' on media that normally admit their growth. They thereby evade detection by conventional culture-based methods. Such dormant cells may also be observed in laboratory cultures and in clinical microbiology. They are usually more tolerant to stresses such as antibiotics, and in clinical microbiology they are typically referred to as 'persisters'. Bacterial cultures necessarily share a great deal of relatedness, and inclusive fitness theory implies that there are conceptual evolutionary advantages in trading a variation in growth rate against its mean, equivalent to hedging one's bets. There is much evidence that bacteria exploit this strategy widely. We here bring together data that show the commonality of these phenomena across environmental, laboratory and clinical microbiology. Considerable evidence, using methods similar to those common in environmental microbiology, now suggests that many supposedly non-communicable, chronic and inflammatory diseases are exacerbated (if not indeed largely caused) by the presence of dormant or persistent bacteria (the ability of whose components to cause inflammation is well known). This dormancy (and resuscitation therefrom) often reflects the extent of the availability of free iron. Together, these phenomena can provide a ready explanation for the continuing inflammation common to such chronic diseases and its correlation with iron dysregulation. This implies that measures designed to assess and to inhibit or remove such organisms (or their access to iron) might be of much therapeutic benefit.
Collapse
Affiliation(s)
- Douglas Kell
- School of Chemistry and The Manchester Institute of Biotechnology, The University of Manchester, Manchester, Lancashire, M1 7DN, UK
| | - Marnie Potgieter
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia, 0007, South Africa
| | - Etheresia Pretorius
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia, 0007, South Africa
| |
Collapse
|
135
|
|
136
|
IL-12 Inhibits Lipopolysaccharide Stimulated Osteoclastogenesis in Mice. J Immunol Res 2015; 2015:214878. [PMID: 26064997 PMCID: PMC4433692 DOI: 10.1155/2015/214878] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 12/04/2014] [Indexed: 11/18/2022] Open
Abstract
Lipopolysaccharide (LPS) is related to osteoclastogenesis in osteolytic diseases. Interleukin- (IL-) 12 is an inflammatory cytokine that plays a critical role in host defense. In this study, we investigated the effects of IL-12 on LPS-induced osteoclastogenesis. LPS was administered with or without IL-12 into the supracalvariae of mice, and alterations in the calvarial suture were evaluated histochemically. The number of osteoclasts in the calvarial suture and the mRNA level of tartrate-resistant acid phosphatase (TRAP), an osteoclast marker, were lower in mice administered LPS with IL-12 than in mice administered LPS alone. The serum level of tartrate-resistant acid phosphatase 5b (TRACP 5b), a bone resorption marker, was also lower in mice administered LPS with IL-12 than in mice administered LPS alone. These results revealed that IL-12 might inhibit LPS-induced osteoclastogenesis and bone resorption. In TdT-mediated dUTP-biotin nick end-labeling (TUNEL) assays, apoptotic changes in cells were recognized in the calvarial suture in mice administered LPS with IL-12. Furthermore, the mRNA levels of both Fas and FasL were increased in mice administered LPS with IL-12. Taken together, the findings demonstrate that LPS-induced osteoclastogenesis is inhibited by IL-12 and that this might arise through apoptotic changes in osteoclastogenesis-related cells induced by Fas/FasL interactions.
Collapse
|
137
|
Transcriptional analysis of the MrpJ network: modulation of diverse virulence-associated genes and direct regulation of mrp fimbrial and flhDC flagellar operons in Proteus mirabilis. Infect Immun 2015; 83:2542-56. [PMID: 25847961 DOI: 10.1128/iai.02978-14] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 03/29/2015] [Indexed: 01/14/2023] Open
Abstract
The enteric bacterium Proteus mirabilis is associated with a significant number of catheter-associated urinary tract infections (UTIs). Strict regulation of the antagonistic processes of adhesion and motility, mediated by fimbriae and flagella, respectively, is essential for disease progression. Previously, the transcriptional regulator MrpJ, which is encoded by the mrp fimbrial operon, has been shown to repress both swimming and swarming motility. Here we show that MrpJ affects an array of cellular processes beyond adherence and motility. Microarray analysis found that expression of mrpJ mimicking levels observed during UTIs leads to differential expression of 217 genes related to, among other functions, bacterial virulence, type VI secretion, and metabolism. We probed the molecular mechanism of transcriptional regulation by MrpJ using transcriptional reporters and chromatin immunoprecipitation (ChIP). Binding of MrpJ to two virulence-associated target gene promoters, the promoters of the flagellar master regulator flhDC and mrp itself, appears to be affected by the condensation state of the native chromosome, although both targets share a direct MrpJ binding site proximal to the transcriptional start. Furthermore, an mrpJ deletion mutant colonized the bladders of mice at significantly lower levels in a transurethral model of infection. Additionally, we observed that mrpJ is widely conserved in a collection of recent clinical isolates. Altogether, these findings support a role of MrpJ as a global regulator of P. mirabilis virulence.
Collapse
|
138
|
Ranf S, Gisch N, Schäffer M, Illig T, Westphal L, Knirel YA, Sánchez-Carballo PM, Zähringer U, Hückelhoven R, Lee J, Scheel D. A lectin S-domain receptor kinase mediates lipopolysaccharide sensing in Arabidopsis thaliana. Nat Immunol 2015; 16:426-33. [PMID: 25729922 DOI: 10.1038/ni.3124] [Citation(s) in RCA: 202] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 02/11/2014] [Indexed: 12/25/2022]
Abstract
The sensing of microbe-associated molecular patterns (MAMPs) triggers innate immunity in animals and plants. Lipopolysaccharide (LPS) from Gram-negative bacteria is a potent MAMP for mammals, with the lipid A moiety activating proinflammatory responses via Toll-like receptor 4 (TLR4). Here we found that the plant Arabidopsis thaliana specifically sensed LPS of Pseudomonas and Xanthomonas. We isolated LPS-insensitive mutants defective in the bulb-type lectin S-domain-1 receptor-like kinase LORE (SD1-29), which were hypersusceptible to infection with Pseudomonas syringae. Targeted chemical degradation of LPS from Pseudomonas species suggested that LORE detected mainly the lipid A moiety of LPS. LORE conferred sensitivity to LPS onto tobacco after transient expression, which demonstrated a key function in LPS sensing and indicated the possibility of engineering resistance to bacteria in crop species.
Collapse
Affiliation(s)
- Stefanie Ranf
- 1] Stress and Developmental Biology, Leibniz Institute of Plant Biochemistry, Halle, Germany. [2] Phytopathology, TUM School of Life Sciences Weihenstephan, Technische Universität München, Freising-Weihenstephan, Germany
| | - Nicolas Gisch
- Division of Immunochemistry/Bioanalytical Chemistry, Priority Area Infections, Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Borstel, Germany
| | - Milena Schäffer
- Phytopathology, TUM School of Life Sciences Weihenstephan, Technische Universität München, Freising-Weihenstephan, Germany
| | - Tina Illig
- Phytopathology, TUM School of Life Sciences Weihenstephan, Technische Universität München, Freising-Weihenstephan, Germany
| | - Lore Westphal
- Stress and Developmental Biology, Leibniz Institute of Plant Biochemistry, Halle, Germany
| | - Yuriy A Knirel
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Patricia M Sánchez-Carballo
- Division of Immunochemistry/Bioanalytical Chemistry, Priority Area Infections, Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Borstel, Germany
| | - Ulrich Zähringer
- Division of Immunochemistry/Bioanalytical Chemistry, Priority Area Infections, Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Borstel, Germany
| | - Ralph Hückelhoven
- Phytopathology, TUM School of Life Sciences Weihenstephan, Technische Universität München, Freising-Weihenstephan, Germany
| | - Justin Lee
- Stress and Developmental Biology, Leibniz Institute of Plant Biochemistry, Halle, Germany
| | - Dierk Scheel
- Stress and Developmental Biology, Leibniz Institute of Plant Biochemistry, Halle, Germany
| |
Collapse
|
139
|
Ardila CM, Perez-Valencia AY, Rendon-Osorio WL. Tannerella forsythia is associated with increased levels of atherogenic low density lipoprotein and total cholesterol in chronic periodontitis. J Clin Exp Dent 2015; 7:e254-60. [PMID: 26155342 PMCID: PMC4483333 DOI: 10.4317/jced.52128] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 02/05/2015] [Indexed: 12/29/2022] Open
Abstract
Background Accumulating evidence suggests that acute and chronic infections with periodontopathogens are associated with an increased risk of cardiovascular disease. The objective of this study was to assess whether Tanerella forsythia and Porphyromonas gingivalis are associated with increased levels of atherogenic low-density lipoprotein (LDL), high-density lipoprotein, total cholesterol (TC), triglycerides and body mass index (BMI) in chronic periodontitis patients. Material and Methods Medical history and clinical and radiographic examination were conducted in 80 chronic periodontitis patients and 28 healthy individuals. Fasting blood samples were drawn for the measurement of the parameters of dyslipidemia. Anthropometric measurements such as height in meters and weight in kilograms were recorded. Both periodontitis and control subjects were asked to answer a questionnaire with regard to their socio-demographic and smoking status. The presence of T. forsythia, and P. gingivalis was detected using primers designed to target the respective 16S rRNA gene sequences. Results The occurrence of T. forsythia and P. gingivalis was higher in the group of subjects with periodontitis. Superior levels of triglycerides were observed in chronic periodontitis patients compared to healthy individuals. High levels of TC in periodontitis persons were significantly associated with increased bleeding on probing. Greater mean levels of TC and LDL were shown in the presence of T. forsythia (P<0.05). Likewise, higher proportions of patients with BMI ≥25 kg/m2 related with T. forsythia (P<0.05). T. forsythia was a significant discriminating factor in the multivariate linear regression model emerging as significant explanatory of increased levels of TC (β=17,879, 95% CI = 4,357-31,401; p=0.01) and LDL (β=17,162, 95% CI= 4,009-30,316; p=0.01). Conclusions Higher levels of serum total cholesterol and LDL were observed in the occurrence of T. forsythia and the presence of this periodontopathogen may increase the atherogenic potency of low-density lipoprotein that may augment the risk for atherosclerosis in periodontal disease patients. Key words:Periodontitis, dyslipidemia, Tannerella forsythia, cardiovascular disease.
Collapse
Affiliation(s)
- Carlos M Ardila
- Periodontist. Ph.D in Epidemiology, Biomedical Stomatology Group, Universidad de Antioquia U de A, Medellín, Colombia, Department of Periodontology, School of Dentistry, Universidad de Antioquia
| | | | | |
Collapse
|
140
|
Genomic analysis of LPS-stimulated myeloid cells identifies a common pro-inflammatory response but divergent IL-10 anti-inflammatory responses. Sci Rep 2015; 5:9100. [PMID: 25765318 PMCID: PMC4650320 DOI: 10.1038/srep09100] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 02/12/2015] [Indexed: 01/02/2023] Open
Abstract
Inflammation is an essential physiological response to infection and injury that must be kept within strict bounds. The IL-10/STAT3 anti-inflammatory response (AIR) is indispensable for controlling the extent of inflammation, although the complete mechanisms downstream of STAT3 have not yet been elucidated. The AIR is widely known to extend to other myeloid cells, but it has best been characterized in macrophages. Here we set out to characterize the LPS-mediated pro-inflammatory response and the AIR across a range of myeloid cells. We found that whereas the LPS-induced pro-inflammatory response is broadly similar among macrophages, dendritic cells, neutrophils, mast cells and eosinophils, the AIR is drastically different across all myeloid cell types that respond to IL-10 (all bar eosinophils). We propose a model whereby the IL-10/STAT3 AIR works by selectively inhibiting specific pathways in distinct cell types: in macrophages the AIR most likely works through the inhibition of NF-κB target genes; in DCs and mast cells through indirect IRF disruption; and in neutrophils through IRF disruption and possibly also indirect NF-κB inhibition. In summary, no conserved IL-10/STAT3 AIR effectors were identified; instead a cell type-specific model of the AIR is proposed.
Collapse
|
141
|
Wiedemann A, Virlogeux-Payant I, Chaussé AM, Schikora A, Velge P. Interactions of Salmonella with animals and plants. Front Microbiol 2015; 5:791. [PMID: 25653644 PMCID: PMC4301013 DOI: 10.3389/fmicb.2014.00791] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 12/22/2014] [Indexed: 12/16/2022] Open
Abstract
Salmonella enterica species are Gram-negative bacteria, which are responsible for a wide range of food- and water-borne diseases in both humans and animals, thereby posing a major threat to public health. Recently, there has been an increasing number of reports, linking Salmonella contaminated raw vegetables and fruits with food poisoning. Many studies have shown that an essential feature of the pathogenicity of Salmonella is its capacity to cross a number of barriers requiring invasion of a large variety of cells and that the extent of internalization may be influenced by numerous factors. However, it is poorly understood how Salmonella successfully infects hosts as diversified as animals or plants. The aim of this review is to describe the different stages required for Salmonella interaction with its hosts: (i) attachment to host surfaces; (ii) entry processes; (iii) multiplication; (iv) suppression of host defense mechanisms; and to point out similarities and differences between animal and plant infections.
Collapse
Affiliation(s)
- Agnès Wiedemann
- Institut National de la Recherche Agronomique, UMR1282 Infectiologie et Santé Publique Nouzilly, France ; UMR1282 Infectiologie et Santé Publique, Université François Rabelais Tours, France
| | - Isabelle Virlogeux-Payant
- Institut National de la Recherche Agronomique, UMR1282 Infectiologie et Santé Publique Nouzilly, France ; UMR1282 Infectiologie et Santé Publique, Université François Rabelais Tours, France
| | - Anne-Marie Chaussé
- Institut National de la Recherche Agronomique, UMR1282 Infectiologie et Santé Publique Nouzilly, France ; UMR1282 Infectiologie et Santé Publique, Université François Rabelais Tours, France
| | - Adam Schikora
- Institute for Phytopathology, Research Center for BioSystems, Land Use and Nutrition (IFZ), Justus Liebig University Giessen Giessen, Germany
| | - Philippe Velge
- Institut National de la Recherche Agronomique, UMR1282 Infectiologie et Santé Publique Nouzilly, France ; UMR1282 Infectiologie et Santé Publique, Université François Rabelais Tours, France
| |
Collapse
|
142
|
Manček-Keber M, Jerala R. Postulates for validating TLR4 agonists. Eur J Immunol 2015; 45:356-70. [DOI: 10.1002/eji.201444462] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Revised: 10/20/2014] [Accepted: 12/01/2014] [Indexed: 02/03/2023]
Affiliation(s)
- Mateja Manček-Keber
- Department of Biotechnology; National Institute of Chemistry; Ljubljana Slovenia
- EN-FIST Centre of Excellence; Ljubljana Slovenia
| | - Roman Jerala
- Department of Biotechnology; National Institute of Chemistry; Ljubljana Slovenia
- EN-FIST Centre of Excellence; Ljubljana Slovenia
| |
Collapse
|
143
|
Bianchi MG, Allegri M, Costa AL, Blosi M, Gardini D, Del Pivo C, Prina-Mello A, Di Cristo L, Bussolati O, Bergamaschi E. Titanium dioxide nanoparticles enhance macrophage activation by LPS through a TLR4-dependent intracellular pathway. Toxicol Res (Camb) 2015. [DOI: 10.1039/c4tx00193a] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
TiO2nanoparticles enhance LPS-dependent NO production and cytokine secretion through a mechanism that involves TLR4-mediated p38-signalling and requires phagocytosis.
Collapse
Affiliation(s)
- Massimiliano G. Bianchi
- Unit of Occupational Medicine
- Department of Clinical and Experimental Medicine
- University of Parma
- 43026 Parma
- Italy
| | - Manfredi Allegri
- Unit of General Pathology
- Department of Biomedical
- Biotechnological and Translational Sciences
- University of Parma
- 43025 Parma
| | - Anna L. Costa
- Institute of Science and Technology for Ceramics (CNR-ISTEC)
- National Research Council of Italy
- 48018 Faenza (RA)
- Italy
| | - Magda Blosi
- Institute of Science and Technology for Ceramics (CNR-ISTEC)
- National Research Council of Italy
- 48018 Faenza (RA)
- Italy
| | - Davide Gardini
- Institute of Science and Technology for Ceramics (CNR-ISTEC)
- National Research Council of Italy
- 48018 Faenza (RA)
- Italy
| | - Camilla Del Pivo
- Institute of Science and Technology for Ceramics (CNR-ISTEC)
- National Research Council of Italy
- 48018 Faenza (RA)
- Italy
| | - Adriele Prina-Mello
- Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN) and School of Medicine
- Trinity College Dublin
- Dublin
- Ireland
| | - Luisana Di Cristo
- Unit of Occupational Medicine
- Department of Clinical and Experimental Medicine
- University of Parma
- 43026 Parma
- Italy
| | - Ovidio Bussolati
- Unit of General Pathology
- Department of Biomedical
- Biotechnological and Translational Sciences
- University of Parma
- 43025 Parma
| | - Enrico Bergamaschi
- Unit of Occupational Medicine
- Department of Clinical and Experimental Medicine
- University of Parma
- 43026 Parma
- Italy
| |
Collapse
|
144
|
Wang Z, Chen G, Chen L, Liu X, Fu W, Zhang Y, Li C, Liang G, Cai Y. Insights into the binding mode of curcumin to MD-2: studies from molecular docking, molecular dynamics simulations and experimental assessments. MOLECULAR BIOSYSTEMS 2015; 11:1933-8. [DOI: 10.1039/c5mb00085h] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The residues R90 and Y102 of MD-2 are hot spot residues that contribute significantly to the affinity of curcumin binding.
Collapse
Affiliation(s)
- Zhe Wang
- Chemical Biology Research Center
- School of Pharmaceutical Sciences
- Wenzhou Medical University
- Wenzhou
- China
| | - Gaozhi Chen
- Chemical Biology Research Center
- School of Pharmaceutical Sciences
- Wenzhou Medical University
- Wenzhou
- China
| | - Linfeng Chen
- Chemical Biology Research Center
- School of Pharmaceutical Sciences
- Wenzhou Medical University
- Wenzhou
- China
| | - Xing Liu
- Chemical Biology Research Center
- School of Pharmaceutical Sciences
- Wenzhou Medical University
- Wenzhou
- China
| | - Weitao Fu
- Chemical Biology Research Center
- School of Pharmaceutical Sciences
- Wenzhou Medical University
- Wenzhou
- China
| | - Yali Zhang
- Chemical Biology Research Center
- School of Pharmaceutical Sciences
- Wenzhou Medical University
- Wenzhou
- China
| | - Chenglong Li
- Chemical Biology Research Center
- School of Pharmaceutical Sciences
- Wenzhou Medical University
- Wenzhou
- China
| | - Guang Liang
- Chemical Biology Research Center
- School of Pharmaceutical Sciences
- Wenzhou Medical University
- Wenzhou
- China
| | - Yuepiao Cai
- Chemical Biology Research Center
- School of Pharmaceutical Sciences
- Wenzhou Medical University
- Wenzhou
- China
| |
Collapse
|
145
|
Dissociation between sickness behavior and emotionality during lipopolysaccharide challenge in lymphocyte deficient Rag2(-/-) mice. Behav Brain Res 2014; 278:74-82. [PMID: 25257108 DOI: 10.1016/j.bbr.2014.09.030] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 09/05/2014] [Accepted: 09/15/2014] [Indexed: 12/21/2022]
Abstract
Inflammatory diseases are highly associated with affective disorders including depression and anxiety. While the role of the innate immune system on emotionality has been extensively studied, the role of adaptive immunity is less understood. Considering that chronic inflammatory conditions are mediated largely by maladaptive lymphocyte function, the role of these cells on brain function and behavior during inflammation warrants investigation. In the present study we employed mice deficient in lymphocyte function and studied behavioral and inflammatory responses during challenge with bacterial lipopolysaccharides (LPS). Rag2(-/-) mice lacking mature lymphocytes were susceptible to death under sub-septic (5 mg/kg) doses of LPS and survived only to moderate (1 mg/kg) doses of LPS. Under these conditions, they displayed attenuated TNF-alpha responses and behavioral symptoms of sickness when compared with immunocompetent mice. Nevertheless, Rag2(-/-) mice had protracted motivational impairments after recovery from sickness suggesting a specific function for lymphocytes on the re-establishment of motivational states after activation of the innate immune system. The behavioral impairments in Rag2(-/-) mice were paralleled by an elevation in plasma corticosterone after behavioral tests. These results provide evidence that the absence of adaptive immunity may be associated with emotional deficits during inflammation and suggest that depressive states associated with medical illness may be mediated in part by impaired lymphocyte responses.
Collapse
|