51
|
Cevallos VM, Díaz V, Sirois CM. Particulate matter air pollution from the city of Quito, Ecuador, activates inflammatory signaling pathways in vitro. Innate Immun 2017; 23:392-400. [PMID: 28409539 DOI: 10.1177/1753425917699864] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Automobile traffic, industrial processes and natural phenomena cause notable air pollution, including gaseous and particulate contaminants, in urban centers. Exposure to particulate matter (PM) air pollution affects human health, and has been linked to respiratory, cardiovascular and neurological diseases. The mechanisms underlying inflammation in these diverse diseases, and to what extent health effects are different for PM obtained from different sources or locations, are still unclear. This study investigated the in vitro toxicity of ambient course (PM10) and fine (PM2.5) particulate matter collected at seven sites in the urban and periurban zones of Quito, Ecuador. Material from all sites was capable of activating TLR2 and TLR4 signaling pathways, with differences in the activation related to particle size. Additionally, airborne particulate matter from Quito is an effective activator of the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Victoria M Cevallos
- 1 Center for Translational Research, Universidad de Las Américas, Quito, Ecuador
| | - Valeria Díaz
- 2 Secretariat of the Environment, Municipality of the Quito Metropolitan District, Quito, Ecuador.,3 School of Medicine, College of Health Sciences, Universidad de Las Américas, Quito, Ecuador
| | - Cherilyn M Sirois
- 1 Center for Translational Research, Universidad de Las Américas, Quito, Ecuador.,3 School of Medicine, College of Health Sciences, Universidad de Las Américas, Quito, Ecuador.,4 Center for Biomedical Research, Eugenio Espejo College of Health Sciences, Universidad Tecnológica Equinoccial, Quito, Ecuador
| |
Collapse
|
52
|
Abstract
Urinary tract infections (UTIs) cause a huge burden of morbidity worldwide with recurrent UTIs becoming increasingly frequent owing to the emergence of antibiotic-resistant bacterial strains. Interactions between the innate and adaptive immune responses to pathogens colonizing the urinary tract have been the focus of much research. Inflammasomes are part of the innate immune defence and can respond rapidly to infectious insult. Assembly of the multiprotein inflammasome complex activates caspase-1, processes proinflammatory cytokines IL-1β and IL-18, and induces pyroptosis. These effector pathways, in turn, act at different levels to either prevent or resolve infection, or eliminate the infectious agent itself. In certain instances, inflammasome activation promotes tissue pathology; however, the precise functions of inflammasomes in UTIs remain unexplored. An improved understanding of inflammasomes could provide novel approaches for the design of diagnostics and therapeutics for complicated UTIs, enabling us to overcome the challenge of drug resistance.
Collapse
|
53
|
Borghesi A, Stronati M, Fellay J. Neonatal Group B Streptococcal Disease in Otherwise Healthy Infants: Failure of Specific Neonatal Immune Responses. Front Immunol 2017; 8:215. [PMID: 28326082 PMCID: PMC5339282 DOI: 10.3389/fimmu.2017.00215] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 02/15/2017] [Indexed: 12/26/2022] Open
Abstract
Only a small proportion of newborn infants exposed to a pathogenic microorganism develop overt infection. Susceptibility to infection in preterm infants and infants with known comorbidities has a likely multifactorial origin and can be often attributed to the concurrence of iatrogenic factors, environmental determinants, underlying pathogenic processes, and probably genetic predisposition. Conversely, infection occurring in otherwise healthy full-term newborn infants is unexplained in most cases. Microbial virulence factors and the unique characteristics of the neonatal immune system only partially account for the interindividual variability in the neonatal immune responses to pathogens. We here suggest that neonatal infection occurring in otherwise healthy infants is caused by a failure of the specific protective immunity to the microorganism. To explain infection in term and preterm infants, we propose an extension of the previously proposed model of the genetic architecture of infectious diseases in humans. We then focus on group B streptococcus (GBS) disease, the best characterized neonatal infection, and outline the potential molecular mechanisms underlying the selective failure of the immune responses against GBS. In light of the recent discoveries of pathogen-specific primary immunodeficiencies and of the role of anticytokine autoantibodies in increasing susceptibility to specific infections, we hypothesize that GBS disease occurring in otherwise healthy infants could reflect an immunodeficiency caused either by rare genetic defects in the infant or by transmitted maternal neutralizing antibodies. These hypotheses are consistent with available epidemiological data, with clinical and epidemiological observations, and with the state of the art of neonatal physiology and disease. Studies should now be designed to comprehensively search for genetic or immunological factors involved in susceptibility to severe neonatal infections.
Collapse
Affiliation(s)
- Alessandro Borghesi
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Neonatal Intensive Care Unit, San Matteo Hospital, Pavia, Italy
| | - Mauro Stronati
- Neonatal Intensive Care Unit, San Matteo Hospital, Pavia, Italy
| | - Jacques Fellay
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| |
Collapse
|
54
|
Inflammasomes in the lung. Mol Immunol 2017; 86:44-55. [PMID: 28129896 DOI: 10.1016/j.molimm.2017.01.014] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/12/2017] [Accepted: 01/13/2017] [Indexed: 12/11/2022]
Abstract
Innate immune responses act as first line defences upon exposure to potentially noxious stimuli. The innate immune system has evolved numerous intracellular and extracellular receptors that undertake surveillance for potentially damaging particulates. Inflammasomes are intracellular innate immune multiprotein complexes that form and are activated following interaction with these stimuli. Inflammasome activation leads to the cleavage of pro-IL-1β and release of the pro-inflammatory cytokine, IL-1β, which initiates acute phase pro-inflammatory responses, and other responses are also involved (IL-18, pyroptosis). However, excessive activation of inflammasomes can result in chronic inflammation, which has been implicated in a range of chronic inflammatory diseases. The airways are constantly exposed to a wide variety of stimuli. Inflammasome activation and downstream responses clears these stimuli. However, excessive activation may drive the pathogenesis of chronic respiratory diseases such as severe asthma and chronic obstructive pulmonary disease. Thus, there is currently intense interest in the role of inflammasomes in chronic inflammatory lung diseases and in their potential for therapeutic targeting. Here we review the known associations between inflammasome-mediated responses and the development and exacerbation of chronic lung diseases.
Collapse
|
55
|
Boldenow E, Gendrin C, Ngo L, Bierle C, Vornhagen J, Coleman M, Merillat S, Armistead B, Whidbey C, Alishetti V, Santana-Ufret V, Ogle J, Gough M, Srinouanprachanh S, MacDonald JW, Bammler TK, Bansal A, Liggitt HD, Rajagopal L, Adams Waldorf KM. Group B Streptococcus circumvents neutrophils and neutrophil extracellular traps during amniotic cavity invasion and preterm labor. Sci Immunol 2016; 1:1/4/eaah4576. [PMID: 27819066 DOI: 10.1126/sciimmunol.aah4576] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Preterm birth is a leading cause of neonatal morbidity and mortality. Although microbial invasion of the amniotic cavity (MIAC) is associated with the majority of early preterm births, the temporal events that occur during MIAC and preterm labor are not known. Group B Streptococci (GBS) are β-hemolytic, gram-positive bacteria, which commonly colonize the vagina but have been recovered from the amniotic fluid in preterm birth cases. To understand temporal events that occur during MIAC, we utilized a unique chronically catheterized nonhuman primate model that closely emulates human pregnancy. This model allows monitoring of uterine contractions, timing of MIAC and immune responses during pregnancy-associated infections. Here, we show that adverse outcomes such as preterm labor, MIAC, and fetal sepsis were observed more frequently during infection with hemolytic GBS when compared to nonhemolytic GBS. Although MIAC was associated with systematic progression in chorioamnionitis beginning with chorionic vasculitis and progressing to neutrophilic infiltration, the ability of the GBS hemolytic pigment toxin to induce neutrophil cell death and subvert killing by neutrophil extracellular traps (NETs) in placental membranes in vivo facilitated MIAC and fetal injury. Furthermore, compared to maternal neutrophils, fetal neutrophils exhibit decreased neutrophil elastase activity and impaired phagocytic functions to GBS. Collectively, our studies demonstrate how a unique bacterial hemolytic lipid toxin enables GBS to circumvent neutrophils and NETs in placental membranes to induce fetal injury and preterm labor.
Collapse
Affiliation(s)
- Erica Boldenow
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Claire Gendrin
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Lisa Ngo
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Craig Bierle
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Jay Vornhagen
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America; Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Michelle Coleman
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Sean Merillat
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Blair Armistead
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America; Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Christopher Whidbey
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America; Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Varchita Alishetti
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Veronica Santana-Ufret
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Jason Ogle
- Washington National Primate Center, University of Washington, Seattle, Washington, United States of America
| | - Michael Gough
- Washington National Primate Center, University of Washington, Seattle, Washington, United States of America
| | - Sengkeo Srinouanprachanh
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - James W MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Theo K Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Aasthaa Bansal
- Department of Pharmacy, University of Washington, Seattle, Washington, United States of America
| | - H Denny Liggitt
- Department of Comparative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Lakshmi Rajagopal
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America; Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Kristina M Adams Waldorf
- Department of Obstetrics & Gynecology, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
56
|
Yu JW, Lee MS. Mitochondria and the NLRP3 inflammasome: physiological and pathological relevance. Arch Pharm Res 2016; 39:1503-1518. [PMID: 27600432 DOI: 10.1007/s12272-016-0827-4] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 08/29/2016] [Indexed: 01/12/2023]
Abstract
The NLRP3 inflammasome is assembled and activated in certain types of myeloid cells upon sensing microbe-derived toxins or host-derived danger signals. Activation of the NLRP3 inflammasome by endogenous ligands has been discovered in various disorders, including metabolic syndrome, type 2 diabetes, atherosclerosis, gout, reperfusion injury of the heart, neurodegeneration, such as Alzheimer's disease, chronic kidney diseases, and macular degeneration of the eyes. Despite the potential significance of the NLRP3 inflammasome in the pathogenesis of several diseases, details on the activation mechanism of the NLRP3 inflammasome by a variety of stimulators have yet to be reported. Emerging evidence suggests that mitochondrial events are associated with NLRP3 activation in disease conditions. Mitochondrial dysfunction acts upstream of NLRP3 activation by providing reactive oxygen species (ROS) to trigger NLRP3 oligomerization or by inducing α-tubulin acetylation to relocate mitochondria to the proximity of NLRP3. In addition, mitochondria work as a platform for inflammasome assembly. Mitochondrial events may also lie downstream of NLRP3 activation. While the molecular mechanisms of mitochondrial dysfunction associated with NLRP3 activation are still unclear, they may involve the perturbation of mitochondria by K+ efflux and subsequent intracellular disequilibrium. Thus, mitochondria and NLRP3 machinery appear to be closely interwoven at multiple levels.
Collapse
Affiliation(s)
- Je-Wook Yu
- Department of Microbiology and Immunology, BK 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Myung-Shik Lee
- Severance Biomedical Science Institute and Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea.
| |
Collapse
|
57
|
Mohammadi N, Midiri A, Mancuso G, Patanè F, Venza M, Venza I, Passantino A, Galbo R, Teti G, Beninati C, Biondo C. Neutrophils Directly Recognize Group B Streptococci and Contribute to Interleukin-1β Production during Infection. PLoS One 2016; 11:e0160249. [PMID: 27509078 PMCID: PMC4980021 DOI: 10.1371/journal.pone.0160249] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 07/15/2016] [Indexed: 11/29/2022] Open
Abstract
Previous studies have shown that the pro-inflammatory cytokine IL-1β has a crucial role in host defenses against group B streptococcus (GBS), a frequent human pathogen, by recruiting neutrophils to infection sites. We examined here the cell types and mechanisms involved in IL-1β production during infection. Using a GBS-induced peritonitis model in mice, we first found that a large proportion of exudate cells contain intracellular IL-1β by immunofluorescence. Of the IL-1β positive cells, 82 and 7% were neutrophils and macrophages, respectively, suggesting that the former cell type might significantly contribute to IL-1β production. Accordingly, depletion of neutrophils with anti-Ly6G antibodies resulted in a significant reduction in the levels of IL-1β, but not of TNF-α or IL-6. We next found that neutrophils are capable of releasing mature IL-1β and TNF-α directly in response to in vitro stimulation with GBS. The production of pro-IL-1β and TNF-α in these cells required the Toll-like receptor (TLR) adaptor MyD88 and the chaperone protein UNC93B1, which is involved in mobilization of a subfamily of TLRs to the endosomes. Moreover, pro-IL-1β processing and IL-1β release was triggered by GBS hemolysin and required components of the canonical inflammasome, including caspase-1, ASC and NLRP3. Collectively our findings indicate that neutrophils make a significant contribution to IL-1β production during GBS infection, thereby amplifying their own recruitment. These cells directly recognize GBS by means of endosomal TLRs and cytosolic sensors, leading to activation of the caspase-1 inflammasome.
Collapse
Affiliation(s)
- Nastaran Mohammadi
- Department of Clinical and Experimental Medicine, University of Messina, 98125, Messina, Italy
| | - Angelina Midiri
- Department of Human Pathology, University of Messina, 98125, Messina, Italy
| | - Giuseppe Mancuso
- Department of Human Pathology, University of Messina, 98125, Messina, Italy
| | - Francesco Patanè
- Department of Human Pathology, University of Messina, 98125, Messina, Italy
| | - Mario Venza
- Department of Clinical and Experimental Medicine, University of Messina, 98125, Messina, Italy
| | - Isabella Venza
- Department of Clinical and Experimental Medicine, University of Messina, 98125, Messina, Italy
| | | | - Roberta Galbo
- Department of Biological, Chemical and Environmental Sciences, University of Messina, 98125, Messina, Italy
| | - Giuseppe Teti
- Department of Clinical and Experimental Medicine, University of Messina, 98125, Messina, Italy
- Charybdis Vaccines Srl, 98125, Messina, Italy
- * E-mail:
| | - Concetta Beninati
- Department of Human Pathology, University of Messina, 98125, Messina, Italy
- Scylla Biotech Srl, 98125, Messina, Italy
| | - Carmelo Biondo
- Department of Human Pathology, University of Messina, 98125, Messina, Italy
| |
Collapse
|
58
|
Protracted Myalgia Syndrome as the Presenting Sign of Familial Mediterranean Fever: Is Group A β-Hemolytic Streptococcus Infection a Causative Factor? Indian J Pediatr 2016; 83:890-1. [PMID: 27056245 DOI: 10.1007/s12098-016-2101-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 03/21/2016] [Indexed: 10/22/2022]
|
59
|
Leclercq SY, Sullivan MJ, Ipe DS, Smith JP, Cripps AW, Ulett GC. Pathogenesis of Streptococcus urinary tract infection depends on bacterial strain and β-hemolysin/cytolysin that mediates cytotoxicity, cytokine synthesis, inflammation and virulence. Sci Rep 2016; 6:29000. [PMID: 27383371 PMCID: PMC4935997 DOI: 10.1038/srep29000] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 06/07/2016] [Indexed: 01/22/2023] Open
Abstract
Streptococcus agalactiae can cause urinary tract infection (UTI) including cystitis and asymptomatic bacteriuria (ABU). The early host-pathogen interactions that occur during S. agalactiae UTI and subsequent mechanisms of disease pathogenesis are poorly defined. Here, we define the early interactions between human bladder urothelial cells, monocyte-derived macrophages, and mouse bladder using uropathogenic S. agalactiae (UPSA) 807 and ABU-causing S. agalactiae (ABSA) 834 strains. UPSA 807 adhered, invaded and killed bladder urothelial cells more efficiently compared to ABSA 834 via mechanisms including low-level caspase-3 activation, and cytolysis, according to lactate dehydrogenase release measures and cell viability. Severe UPSA 807-induced cytotoxicity was mediated entirely by the bacterial β-hemolysin/cytolysin (β-H/C) because an β-H/C-deficient UPSA 807 isogenic mutant, UPSA 807ΔcylE, was not cytotoxic in vitro; the mutant was also significantly attenuated for colonization in the bladder in vivo. Analysis of infection-induced cytokines, including IL-8, IL-1β, IL-6 and TNF-α in vitro and in vivo revealed that cytokine and chemokine responses were dependent on expression of β-H/C that also elicited severe bladder neutrophilia. Thus, virulence of UPSA 807 encompasses adhesion to, invasion of and killing of bladder cells, pro-inflammatory cytokine/chemokine responses that elicit neutrophil infiltration, and β-H/C-mediated subversion of innate immune-mediated bacterial clearance from the bladder.
Collapse
Affiliation(s)
- Sophie Y Leclercq
- School of Medical Science, and Menzies Health Institute Queensland, Griffith University, Parklands 4222, Australia.,Research and Development Center, Ezequiel Dias Foundation (Funed), Belo Horizonte, MG, Brazil
| | - Matthew J Sullivan
- School of Medical Science, and Menzies Health Institute Queensland, Griffith University, Parklands 4222, Australia
| | - Deepak S Ipe
- School of Medical Science, and Menzies Health Institute Queensland, Griffith University, Parklands 4222, Australia
| | - Joshua P Smith
- School of Medical Science, and Menzies Health Institute Queensland, Griffith University, Parklands 4222, Australia
| | - Allan W Cripps
- School of Medical Science, and Menzies Health Institute Queensland, Griffith University, Parklands 4222, Australia
| | - Glen C Ulett
- School of Medical Science, and Menzies Health Institute Queensland, Griffith University, Parklands 4222, Australia
| |
Collapse
|
60
|
Tzeng TC, Schattgen S, Monks B, Wang D, Cerny A, Latz E, Fitzgerald K, Golenbock DT. A Fluorescent Reporter Mouse for Inflammasome Assembly Demonstrates an Important Role for Cell-Bound and Free ASC Specks during In Vivo Infection. Cell Rep 2016; 16:571-582. [PMID: 27346360 DOI: 10.1016/j.celrep.2016.06.011] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 04/05/2015] [Accepted: 05/26/2016] [Indexed: 11/24/2022] Open
Abstract
Inflammasome activation is associated with numerous diseases. However, in vivo detection of the activated inflammasome complex has been limited by a dearth of tools. We have developed transgenic mice that ectopically express the fluorescent adaptor protein, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) and characterized the formation of assembled inflammasome complexes ("specks") in primary cells and tissues. In addition to hematopoietic cells, we have found that a stromal population in the lung tissues formed specks during the early phase of influenza infection, whereas myeloid cells showed speck formation after 2 days. In a peritonitis and group B streptococcus infection model, a higher percentage of neutrophils formed specks at early phases of infection, while dendritic cells formed specks at later time points. Furthermore, speck-forming cells underwent pyroptosis and extensive release of specks to the extracellular milieu in vivo. These data underscore the importance of free specks during inflammatory processes in vivo.
Collapse
Affiliation(s)
- Te-Chen Tzeng
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Stefan Schattgen
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Brian Monks
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Institute of Innate Immunity, University of Bonn, Sigmund-Freud-Strasse 25, 53127 Bonn, Germany
| | - Donghai Wang
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Anna Cerny
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Eicke Latz
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Institute of Innate Immunity, University of Bonn, Sigmund-Freud-Strasse 25, 53127 Bonn, Germany
| | - Katherine Fitzgerald
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Douglas T Golenbock
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
61
|
Maltez VI, Miao EA. Reassessing the Evolutionary Importance of Inflammasomes. THE JOURNAL OF IMMUNOLOGY 2016; 196:956-62. [PMID: 26802061 DOI: 10.4049/jimmunol.1502060] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Inflammasomes monitor the cytosol for microbial contamination or perturbation and, thus, are predicted to provide potent defense against infection. However, the compendium of data from murine infection models suggests that inflammasomes merely delay the course of disease, allowing the host time to mount an adaptive response. Interpretations of such results are confounded by inflammasome-evasion strategies of vertebrate-adapted pathogens. Conversely, environmental opportunistic pathogens have not evolved in the context of inflammasomes and, therefore, are less likely to evade them. Indeed, opportunistic pathogens do not normally cause disease in wild-type animals. Accordantly, the extreme virulence of two opportunistic bacterial pathogens, Burkholderia thailandensis and Chromobacterium violaceum, is fully counteracted by inflammasomes in murine models. This leads us to propose a new hypothesis: perhaps animals maintain inflammasomes over evolutionary time not to defend against vertebrate-adapted pathogens but instead to counteract infection by a plethora of undiscovered opportunistic pathogens residing in the environment.
Collapse
Affiliation(s)
- Vivien I Maltez
- Department of Microbiology and Immunology, Center for Gastrointestinal Biology and Disease, and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Edward A Miao
- Department of Microbiology and Immunology, Center for Gastrointestinal Biology and Disease, and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
62
|
Group B Streptococcus Induces a Robust IFN-γ Response by CD4(+) T Cells in an In Vitro and In Vivo Model. J Immunol Res 2016; 2016:5290604. [PMID: 26989699 PMCID: PMC4771917 DOI: 10.1155/2016/5290604] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 01/06/2016] [Indexed: 11/17/2022] Open
Abstract
Group B Streptococcus (GBS) serotype III causes life-threatening infections. Cytokines have emerged as important players for the control of disease, particularly IFN-γ. Although potential sources of this cytokine have been proposed, no specific cell line has ever been described as a leading contributor. In this study, CD4+ T cell activation profiles in response to GBS were evaluated through in vivo, ex vivo, and in vitro approaches. Total splenocytes readily produce a type 1 proinflammatory response by releasing IFN-γ, TNF-α, and IL-6 and actively recruit T cells via chemokines like CXCL9, CXCL10, and CCL3. Responding CD4+ T cells differentiate into Th1 cells producing large amounts of IFN-γ, TNF-α, and IL-2. In vitro studies using dendritic cell and CD4+ T cell cocultures infected with wild-type GBS or a nonencapsulated mutant suggested that GBS capsular polysaccharide, one of the major bacterial virulence factors, differentially modulates surface expression of CD69 and IFN-γ production. Overall, CD4+ T cells are important producers of IFN-γ and might thus influence the course of GBS infection through the expression balance of this cytokine.
Collapse
|
63
|
Sahoo M, Lantier L, Re F. Role of Canonical and Non-canonical Inflammasomes During Burkholderia Infection. Curr Top Microbiol Immunol 2016; 397:199-214. [PMID: 27460811 DOI: 10.1007/978-3-319-41171-2_10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Burkholderia pseudomallei is a Gram-negative flagellate bacterium that causes melioidosis, a disease endemic to Southeast Asia and other tropical regions. Following infection of macrophages and other non-phagocytic cell types, B. pseudomallei or B. thailandensis (a related species that causes disease in mice but not humans) are able to escape the phagosome and replicate in the host cell cytoplasm. Resistance to infection with Burkholderia is dependent on the Nlrp3 and Nlrc4 inflammasomes and the non-canonical caspase-11 inflammasome. Nlrc4 mediates protection through induction of pyroptosis in the early phase of infection. As the infection progresses and as IL-18-dependent IFNγ production increases, caspase-11-dependent pyroptosis acquires a preponderant protective role. Production of IL-1β and IL-18 during infection is primarily mediated by Nlrp3. IL-18 is essential for survival because of its ability to induce IFNγ production, which in turn activates macrophage microbicidal functions and primes for caspase-11 expression. In contrast, during melioidosis, IL-1β has deleterious effects due to excessive recruitment of neutrophils to the lung and consequent tissue damage.
Collapse
Affiliation(s)
- Manoranjan Sahoo
- Department of Microbiology & Immunology, Rosalind Franklin University of Medicine and Science, 3333, Green Bay Road, North Chicago, IL, 60064, USA
| | - Louis Lantier
- Department of Microbiology & Immunology, Rosalind Franklin University of Medicine and Science, 3333, Green Bay Road, North Chicago, IL, 60064, USA
| | - Fabio Re
- Department of Microbiology & Immunology, Rosalind Franklin University of Medicine and Science, 3333, Green Bay Road, North Chicago, IL, 60064, USA.
| |
Collapse
|
64
|
Whidbey C, Vornhagen J, Gendrin C, Boldenow E, Samson JM, Doering K, Ngo L, Ezekwe EAD, Gundlach JH, Elovitz MA, Liggitt D, Duncan JA, Adams Waldorf KM, Rajagopal L. A streptococcal lipid toxin induces membrane permeabilization and pyroptosis leading to fetal injury. EMBO Mol Med 2015; 7:488-505. [PMID: 25750210 PMCID: PMC4403049 DOI: 10.15252/emmm.201404883] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Group B streptococci (GBS) are Gram-positive bacteria that cause infections in utero and in newborns. We recently showed that the GBS pigment is hemolytic and increased pigment production promotes bacterial penetration of human placenta. However, mechanisms utilized by the hemolytic pigment to induce host cell lysis and the consequence on fetal injury are not known. Here, we show that the GBS pigment induces membrane permeability in artificial lipid bilayers and host cells. Membrane defects induced by the GBS pigment trigger K+ efflux leading to osmotic lysis of red blood cells or pyroptosis in human macrophages. Macrophages lacking the NLRP3 inflammasome recovered from pigment-induced cell damage. In a murine model of in utero infection, hyperpigmented GBS strains induced fetal injury in both an NLRP3 inflammasome-dependent and NLRP3 inflammasome-independent manner. These results demonstrate that the dual mechanism of action of the bacterial pigment/lipid toxin leading to hemolysis or pyroptosis exacerbates fetal injury and suggest that preventing both activities of the hemolytic lipid is likely critical to reduce GBS fetal injury and preterm birth.
Collapse
Affiliation(s)
- Christopher Whidbey
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, WA, USA Department of Global Health, University of Washington, Seattle, WA, USA
| | - Jay Vornhagen
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, WA, USA Department of Global Health, University of Washington, Seattle, WA, USA
| | - Claire Gendrin
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, WA, USA
| | - Erica Boldenow
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, WA, USA
| | - Jenny Mae Samson
- Department of Physics, University of Washington, Seattle, WA, USA
| | - Kenji Doering
- Department of Physics, University of Washington, Seattle, WA, USA
| | - Lisa Ngo
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, WA, USA
| | - Ejiofor A D Ezekwe
- Department of Medicine, Division of Infectious Diseases and Pharmacology, School of Medicine and Lineberger Comprehensive Cancer Center University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jens H Gundlach
- Department of Physics, University of Washington, Seattle, WA, USA
| | - Michal A Elovitz
- Maternal and Child Health Research Program, Department of Obstetrics and Gynecology, Center for Research on Reproduction and Women's Health Perelman School of Medicine University of Pennsylvania, Philadelphia, PA, USA
| | - Denny Liggitt
- Department of Comparative Medicine, School of Medicine University of Washington, Seattle, WA, USA
| | - Joseph A Duncan
- Department of Medicine, Division of Infectious Diseases and Pharmacology, School of Medicine and Lineberger Comprehensive Cancer Center University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kristina M Adams Waldorf
- Department of Obstetrics and Gynecology, School of Medicine University of Washington, Seattle, WA, USA
| | - Lakshmi Rajagopal
- Department of Pediatric Infectious Diseases, University of Washington and Seattle Children's Research Institute, Seattle, WA, USA Department of Global Health, University of Washington, Seattle, WA, USA
| |
Collapse
|
65
|
Greaney AJ, Leppla SH, Moayeri M. Bacterial Exotoxins and the Inflammasome. Front Immunol 2015; 6:570. [PMID: 26617605 PMCID: PMC4639612 DOI: 10.3389/fimmu.2015.00570] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 10/26/2015] [Indexed: 11/24/2022] Open
Abstract
The inflammasomes are intracellular protein complexes that play an important role in innate immune sensing. Activation of inflammasomes leads to activation of caspase-1 and maturation and secretion of the pro-inflammatory cytokines interleukin (IL)-1β and IL-18. In certain myeloid cells, this activation can also lead to an inflammatory cell death (pyroptosis). Inflammasome sensor proteins have evolved to detect a range of microbial ligands and bacterial exotoxins either through direct interaction or by detection of host cell changes elicited by these effectors. Bacterial exotoxins activate the inflammasomes through diverse processes, including direct sensor cleavage, modulation of ion fluxes through plasma membrane pore formation, and perturbation of various host cell functions. In this review, we summarize the findings on some of the bacterial exotoxins that activate the inflammasomes.
Collapse
Affiliation(s)
- Allison J Greaney
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, MD , USA
| | - Stephen H Leppla
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, MD , USA
| | - Mahtab Moayeri
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, MD , USA
| |
Collapse
|
66
|
Jo EK, Kim JK, Shin DM, Sasakawa C. Molecular mechanisms regulating NLRP3 inflammasome activation. Cell Mol Immunol 2015; 13:148-59. [PMID: 26549800 DOI: 10.1038/cmi.2015.95] [Citation(s) in RCA: 970] [Impact Index Per Article: 107.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 10/14/2015] [Accepted: 10/14/2015] [Indexed: 02/06/2023] Open
Abstract
Inflammasomes are multi-protein signaling complexes that trigger the activation of inflammatory caspases and the maturation of interleukin-1β. Among various inflammasome complexes, the NLRP3 inflammasome is best characterized and has been linked with various human autoinflammatory and autoimmune diseases. Thus, the NLRP3 inflammasome may be a promising target for anti-inflammatory therapies. In this review, we summarize the current understanding of the mechanisms by which the NLRP3 inflammasome is activated in the cytosol. We also describe the binding partners of NLRP3 inflammasome complexes activating or inhibiting the inflammasome assembly. Our knowledge of the mechanisms regulating NLRP3 inflammasome signaling and how these influence inflammatory responses offers further insight into potential therapeutic strategies to treat inflammatory diseases associated with dysregulation of the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon 301-747, South Korea.,Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon 301-747, South Korea
| | - Jin Kyung Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon 301-747, South Korea.,Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon 301-747, South Korea
| | - Dong-Min Shin
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon 301-747, South Korea.,Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon 301-747, South Korea
| | - Chihiro Sasakawa
- Medical Mycology Research Center, Chiba University, Chiba 260-8673, Japan.,Nippon Institute for Biological Science, Tokyo 198-0024, Japan
| |
Collapse
|
67
|
Naganuma Y, Takakubo Y, Hirayama T, Tamaki Y, Pajarinen J, Sasaki K, Goodman SB, Takagi M. Lipoteichoic acid modulates inflammatory response in macrophages after phagocytosis of titanium particles through Toll-like receptor 2 cascade and inflammasomes. J Biomed Mater Res A 2015; 104:435-44. [PMID: 26440284 DOI: 10.1002/jbm.a.35581] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Revised: 09/12/2015] [Accepted: 10/01/2015] [Indexed: 12/25/2022]
Abstract
Toll-like receptor 2 (TLR2) and nucleotide-binding and oligomerization domain-like receptors with a pyrin domain 3 (NLRP3) inflammasomes have been presumed to participate in the pathogenesis of aseptic implant loosening. The aim of this study is to analyze the cellular localization of TLR2 and NLRP3 inflammasomes in the periprosthetic tissue from aseptically loose hip implants as well as the expression of these molecules in macrophages stimulated in vitro with titanium particles (Ti) coated with lipoteichoic acid (LTA). Using immunohistochemistry, immunoreactivity of TLR2 and NLRP3 inflammasomes was found in macrophages within the foreign body granulomatosis. Using RAW264.7 cells, stimulation with Ti increased the messenger RNA (mRNA) levels of TLR2 and TNF-α. Stimulation with LTA-coated Ti enhanced mRNA levels of NLRP3 and IL-1β, whereas reinforced secretion of IL-1β was not detected in spite of marked release of TNF-α. Finally, the same cells with silenced Irak2, an adaptor protein in the TLR2 cascade, suppressed this NLRP3 upregulation. This study suggests that TLR2 and NLRP3 inflammasomes are factors involved in cross-talk mediating the foreign body type response to wear particles. In addition, discrepant behavior in the release between TNF-α and IL-1β release may explain the variable pathomechanisms of aseptic implant loosening without acute inflammatory reactions.
Collapse
Affiliation(s)
- Yasushi Naganuma
- Department of Orthopaedic Surgery, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Yuya Takakubo
- Department of Orthopaedic Surgery, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Tomoyuki Hirayama
- Department of Orthopaedic Surgery, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Yasunobu Tamaki
- Department of Clinical Medicine, Yamagata Saisei Hospital, Yamagata, Japan
| | - Jukka Pajarinen
- Department of Orthopaedic Surgery, Stanford University School of Medicine, California, 94063.,Department of Medicine, Biomedicum Helsinki, University of Helsinki, Helsinki, FIN-00014 HY, Finland
| | - Kan Sasaki
- Department of Orthopaedic Surgery, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, California, 94063
| | - Michiaki Takagi
- Department of Orthopaedic Surgery, Yamagata University Faculty of Medicine, Yamagata, Japan
| |
Collapse
|
68
|
LaRock CN, Nizet V. Inflammasome/IL-1β Responses to Streptococcal Pathogens. Front Immunol 2015; 6:518. [PMID: 26500655 PMCID: PMC4597127 DOI: 10.3389/fimmu.2015.00518] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 09/24/2015] [Indexed: 02/06/2023] Open
Abstract
Inflammation mediated by the inflammasome and the cytokine IL-1β are some of the earliest and most important alarms to infection. These pathways are responsive to the virulence factors that pathogens use to subvert immune processes, and thus are typically activated only by microbes with potential to cause severe disease. Among the most serious human infections are those caused by the pathogenic streptococci, in part because these species numerous strategies for immune evasion. Since the virulence factor armament of each pathogen is unique, the role of IL-1β and the pathways leading to its activation varies for each infection. This review summarizes the role of IL-1β during infections caused by streptococcal pathogens, with emphasis on emergent mechanisms and concepts countering paradigms determined for other organisms.
Collapse
Affiliation(s)
- Christopher N LaRock
- Department of Pediatrics, University of California San Diego , La Jolla, CA , USA
| | - Victor Nizet
- Department of Pediatrics, University of California San Diego , La Jolla, CA , USA ; Skaggs School of Medicine and Pharmaceutical Sciences, University of California San Diego , La Jolla, CA , USA
| |
Collapse
|
69
|
Gendrin C, Vornhagen J, Ngo L, Whidbey C, Boldenow E, Santana-Ufret V, Clauson M, Burnside K, Galloway DP, Adams Waldorf KM, Piliponsky AM, Rajagopal L. Mast cell degranulation by a hemolytic lipid toxin decreases GBS colonization and infection. SCIENCE ADVANCES 2015; 1:e1400225. [PMID: 26425734 PMCID: PMC4584422 DOI: 10.1126/sciadv.1400225] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 06/04/2015] [Indexed: 05/08/2023]
Abstract
Ascending infection of microbes from the lower genital tract into the amniotic cavity increases the risk of preterm birth, stillbirth, and newborn infections. Host defenses that are critical for preventing ascending microbial infection are not completely understood. Group B Streptococcus (GBS) are Gram-positive bacteria that frequently colonize the lower genital tract of healthy women but cause severe infections during pregnancy, leading to preterm birth, stillbirth, or early-onset newborn infections. We recently described that the GBS pigment is hemolytic, and increased pigment expression promotes GBS penetration of human placenta. Here, we show that the GBS hemolytic pigment/lipid toxin and hyperpigmented GBS strains induce mast cell degranulation, leading to the release of preformed and proinflammatory mediators. Mast cell-deficient mice exhibit enhanced bacterial burden, decreased neutrophil mobilization, and decreased immune responses during systemic GBS infection. In a vaginal colonization model, hyperpigmented GBS strains showed increased persistence in mast cell-deficient mice compared to mast cell-proficient mice. Consistent with these observations, fewer rectovaginal GBS isolates from women in their third trimester of pregnancy were hyperpigmented/hyperhemolytic. Our work represents the first example of a bacterial hemolytic lipid that induces mast cell degranulation and emphasizes the role of mast cells in limiting genital colonization by hyperpigmented GBS.
Collapse
Affiliation(s)
- Claire Gendrin
- Department of Pediatric Infectious Diseases, University of Washington, Seattle, WA 98101, USA
- Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Jay Vornhagen
- Seattle Children’s Research Institute, Seattle, WA 98101, USA
- Department of Global Health, University of Washington, Seattle, WA 98195, USA
| | - Lisa Ngo
- Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Christopher Whidbey
- Seattle Children’s Research Institute, Seattle, WA 98101, USA
- Department of Global Health, University of Washington, Seattle, WA 98195, USA
| | - Erica Boldenow
- Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | | | - Morgan Clauson
- Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Kellie Burnside
- Department of Pediatric Infectious Diseases, University of Washington, Seattle, WA 98101, USA
| | - Dionne P. Galloway
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA
| | | | - Adrian M. Piliponsky
- Department of Pediatric Infectious Diseases, University of Washington, Seattle, WA 98101, USA
- Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Lakshmi Rajagopal
- Department of Pediatric Infectious Diseases, University of Washington, Seattle, WA 98101, USA
- Seattle Children’s Research Institute, Seattle, WA 98101, USA
- Department of Global Health, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
70
|
Eigenbrod T, Dalpke AH. Bacterial RNA: An Underestimated Stimulus for Innate Immune Responses. THE JOURNAL OF IMMUNOLOGY 2015; 195:411-8. [DOI: 10.4049/jimmunol.1500530] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
71
|
Zhang L, Zhou J, Sokol L. Hereditary and acquired hemophagocytic lymphohistiocytosis. Cancer Control 2015; 21:301-12. [PMID: 25310211 DOI: 10.1177/107327481402100406] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Hemophagocytic lymphohistiocytosis (HLH) is a rare but life-threatening hyperinflammatory/hypercytokinemia syndrome clinicopathologically manifested by fever, hepatosplenomegaly, cytopenias, liver dysfunction, and hemophagocytosis. METHODS We searched the medical literature for English-written articles and analyzed data regarding the diagnosis, pathoetiology, prognosis, and management of HLH. RESULTS HLH can be subcategorized into primary/genetic (PHLH) or secondary/acquired (SHLH) according to etiology. PHLH, including familial HLH and inherited immune deficiency syndromes, typically occurs in children harboring underlying genetic defects, whereas SHLH frequently manifests in adults and is associated with infection, autoimmunity, immune suppression, or malignancy. The pathogenesis of HLH is still elusive. Its known mechanisms include somatic mutations in gene coding for proteins implicated in the cytotoxic pathways of cytotoxic T or natural killer cells. The impaired ability of these cells to kill target cells leads to an uncontrolled hypercytokinemia and hyperinflammatory process, triggering hemophagocytosis and multiorgan failure. Corticosteroids, chemotherapy, and immunotherapy are the mainstay therapeutic strategies. The consolidation with allogeneic hematopoietic stem cell transplantation is a potentially curative option for PHLH and refractory or relapsed SHLH. CONCLUSIONS Understanding of the pathophysiology of HLH has improved in the last decade. The establishment of diagnostic and treatment guidelines for PHLH and SHLH has resulted in earlier diagnoses and the rapid initiation of therapy, both of which are associated with favorable outcomes.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Hematopathology and Laboratory Medicine, Moffitt Cancer Center, Tampa, FL 33612, USA.
| | | | | |
Collapse
|
72
|
Yoshikawa FSY, Ferreira LG, de Almeida SR. IL-1 signaling inhibits Trichophyton rubrum conidia development and modulates the IL-17 response in vivo. Virulence 2015; 6:449-57. [PMID: 25950847 DOI: 10.1080/21505594.2015.1020274] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Dermatophytosis are one of the most common fungal infections in the world. They compromise keratinized tissues and the main etiological agent is Trichophyton rubrum. Macrophages are key cells in innate immunity and prominent sources of IL-1β, a potent inflammatory cytokine whose main production pathway is by the activation of inflammasomes and caspase-1. However, the role of inflammasomes and IL-1 signaling against T.rubrum has not been reported. In this work, we observed that bone marrow-derived macrophages produce IL-1β in response to T.rubrum conidia in a NLRP3-, ASC- and caspase-1-dependent fashion. Curiously, lack of IL-1 signaling promoted hyphae development, uncovering a protective role for IL-1β in macrophages. In addition, mice lacking IL-1R showed reduced IL-17 production, a key cytokine in the antifungal defense, in response to T.rubrum. Our findings point to a prominent role of IL-1 signaling in the immune response to T.rubrum, opening the venue for the study of this pathway in other fungal infections.
Collapse
Affiliation(s)
- Fábio Seiti Yamada Yoshikawa
- a Faculty of Pharmaceutical Sciences; Department of Clinical e Toxicological Analysis; University of São Paulo ; São Paulo , Brazil
| | | | | |
Collapse
|
73
|
Non-canonical manifestations of familial Mediterranean fever: a changing paradigm. Clin Rheumatol 2015; 34:1503-11. [PMID: 25761640 DOI: 10.1007/s10067-015-2916-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Accepted: 03/01/2015] [Indexed: 01/04/2023]
Abstract
Paroxysmal crises of fever and systemic inflammation herald familial Mediterranean fever (FMF), considered as the archetype of all inherited systemic autoinflammatory diseases. Inflammatory bouts are characterized by short-term and self-limited abdominal, thoracic, and/or articular symptoms which subside spontaneously. Erysipelas-like findings, orchitis, and different patterns of myalgia may appear in a minority of patients. In recent years, many non-classical manifestations have been reported in the clinical context of FMF, such as vasculitides and thrombotic manifestations, neurologic and sensory organ abnormalities, gastrointestinal diseases, and even macrophage activation syndrome. As FMF left unrecognized and untreated is ominously complicated by the occurrence of AA-amyloidosis, it is highly desirable that diagnosis of this autoinflammatory disorder with its multiple clinical faces can be contemplated at whatever age and brought forward.
Collapse
|
74
|
Venza M, Visalli M, Biondo C, Oteri R, Agliano F, Morabito S, Caruso G, Caffo M, Teti D, Venza I. Epigenetic effects of cadmium in cancer: focus on melanoma. Curr Genomics 2015; 15:420-35. [PMID: 25646071 PMCID: PMC4311387 DOI: 10.2174/138920291506150106145932] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 11/07/2014] [Accepted: 11/17/2014] [Indexed: 01/01/2023] Open
Abstract
Cadmium is a highly toxic heavy metal, which has a destroying impact on organs. Exposure to cadmium causes severe health problems to human beings due to its ubiquitous environmental presence and features of the pathologies associated with pro-longed exposure. Cadmium is a well-established carcinogen, although the underlying mechanisms have not been fully under-stood yet. Recently, there has been considerable interest in the impact of this environmental pollutant on the epigenome. Be-cause of the role of epigenetic alterations in regulating gene expression, there is a potential for the integration of cadmium-induced epigenetic alterations as critical elements in the cancer risk assessment process. Here, after a brief review of the ma-jor diseases related to cadmium exposure, we focus our interest on the carcinogenic potential of this heavy metal. Among the several proposed pathogenetic mechanisms, particular attention is given to epigenetic alterations, including changes in DNA methylation, histone modifications and non-coding RNA expression. We review evidence for a link between cadmium-induced epigenetic changes and cell transformation, with special emphasis on melanoma. DNA methylation, with reduced expression of key genes that regulate cell proliferation and apoptosis, has emerged as a possible cadmium-induced epigenetic mechanism in melanoma. A wider comprehension of mechanisms related to this common environmental contaminant would allow a better cancer risk evaluation.
Collapse
Affiliation(s)
- Mario Venza
- Department of Experimental Specialistic Medical, Surgical and Odontostomatology Sciences, University of Messina, Messina, Italy
| | - Maria Visalli
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Carmelo Biondo
- Department of Pediatric, Gynecological, Microbiological and Biomedical Sciences, University of Messina, Messina, Italy
| | - Rosaria Oteri
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Federica Agliano
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Silvia Morabito
- Department of Experimental Specialistic Medical, Surgical and Odontostomatology Sciences, University of Messina, Messina, Italy
| | - Gerardo Caruso
- Department of Neurosciences, University of Messina, Messina, Italy
| | - Maria Caffo
- Department of Neurosciences, University of Messina, Messina, Italy
| | - Diana Teti
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Isabella Venza
- Department of Experimental Specialistic Medical, Surgical and Odontostomatology Sciences, University of Messina, Messina, Italy
| |
Collapse
|
75
|
Lemire P, Roy D, Fittipaldi N, Okura M, Takamatsu D, Bergman E, Segura M. Implication of TLR- but not of NOD2-signaling pathways in dendritic cell activation by group B Streptococcus serotypes III and V. PLoS One 2014; 9:e113940. [PMID: 25436906 PMCID: PMC4250082 DOI: 10.1371/journal.pone.0113940] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 10/31/2014] [Indexed: 12/31/2022] Open
Abstract
Group B Streptococcus (GBS) is an important agent of life-threatening invasive infection. It has been previously shown that encapsulated type III GBS is easily internalized by dendritic cells (DCs), and that this internalization had an impact on cytokine production. The receptors underlying these processes are poorly characterized. Knowledge on the mechanisms used by type V GBS to activate DCs is minimal. In this work, we investigated the role of Toll-like receptor (TLR)/MyD88 signaling pathway, the particular involvement of TLR2, and that of the intracellular sensing receptor NOD2 in the activation of DCs by types III and V GBS. The role of capsular polysaccharide (CPS, one of the most important GBS virulence factors) in bacterial-DC interactions was evaluated using non-encapsulated mutants. Despite differences in the role of CPS between types III and V GBS in bacterial internalization and intracellular survival, no major differences were observed in their capacity to modulate release of cytokines by DC. For both serotypes, CPS had a minor role in this response. Production of cytokines by DCs was shown to strongly rely on MyD88-dependent signaling pathways, suggesting that DCs recognize GBS and become activated mostly through TLR signaling. Yet, GBS-infected TLR2-/- DCs only showed a partial reduction in the production of IL-6 and CXCL1 compared to control DCs. Surprisingly, CXCL10 release by type III or type V GBS-infected DCs was MyD88-independent. No differences in DC activation were observed between NOD2-/- and control DCs. These results demonstrate the involvement of various receptors and the complexity of the cytokine production pathways activated by GBS upon DC infection.
Collapse
Affiliation(s)
- Paul Lemire
- Laboratory of Immunology, Faculty of Veterinary Medicine, University of Montreal, St-Hyacinthe, Quebec, Canada
| | - David Roy
- Laboratory of Immunology, Faculty of Veterinary Medicine, University of Montreal, St-Hyacinthe, Quebec, Canada
| | - Nahuel Fittipaldi
- Public Health Ontario, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Masatoshi Okura
- Bacterial and Parasitic Diseases Research Division, National Institute of Animal Health, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
| | - Daisuke Takamatsu
- Bacterial and Parasitic Diseases Research Division, National Institute of Animal Health, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
- The United Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan
| | - Eugenia Bergman
- Laboratory of Immunology, Faculty of Veterinary Medicine, University of Montreal, St-Hyacinthe, Quebec, Canada
| | - Mariela Segura
- Laboratory of Immunology, Faculty of Veterinary Medicine, University of Montreal, St-Hyacinthe, Quebec, Canada
- * E-mail:
| |
Collapse
|
76
|
Vick EJ, Park HS, Huff KA, Brooks KM, Farone AL, Farone MB. Gardnerella vaginalis triggers NLRP3 inflammasome recruitment in THP-1 monocytes. J Reprod Immunol 2014; 106:67-75. [DOI: 10.1016/j.jri.2014.08.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 08/05/2014] [Accepted: 08/19/2014] [Indexed: 01/09/2023]
|
77
|
Landwehr-Kenzel S, Henneke P. Interaction of Streptococcus agalactiae and Cellular Innate Immunity in Colonization and Disease. Front Immunol 2014; 5:519. [PMID: 25400631 PMCID: PMC4212683 DOI: 10.3389/fimmu.2014.00519] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 10/05/2014] [Indexed: 12/18/2022] Open
Abstract
Streptococcus agalactiae (Group B streptococcus, GBS) is highly adapted to humans, where it is a normal constituent of the intestinal and vaginal flora. Yet, GBS has highly invasive potential and causes excessive inflammation, sepsis, and death at the beginning of life, in the elderly and in diabetic patients. Thus, GBS is a model pathobiont that thrives in the healthy host, but has not lost its potential virulence during coevolution with mankind. It remains incompletely understood how the innate immune system contains GBS in the natural niches, the intestinal and genital tracts, and which molecular events underlie breakdown of mucocutaneous resistance. Newborn infants between days 7 and 90 of life are at risk of a particularly striking sepsis manifestation (late-onset disease), where the transition from colonization to invasion and dissemination, and thus from health to severe sepsis is typically fulminant and not predictable. The great majority of late-onset sepsis cases are caused by one clone, GBS ST17, which expresses HvgA as a signature virulence factor and adhesin. In mice, HvgA promotes the crossing of both the mucosal and the blood–brain barrier. Expression levels of HvgA and other GBS virulence factors, such as pili and toxins, are regulated by the upstream two-component control system CovR/S. This in turn is modulated by acidic epithelial pH, high glucose levels, and during the passage through the mouse intestine. After invasion, GBS has the ability to subvert innate immunity by mechanisms like glycerinaldehyde-3-phosphate-dehydrogenase-dependent induction of IL-10 and β-protein binding to the inhibitory phagocyte receptors sialic acid binding immunoglobulin-like lectin 5 and 14. On the host side, sensing of GBS nucleic acids and lipopeptides by both Toll-like receptors and the inflammasome appears to be critical for host resistance against GBS. Yet, comprehensive models on the interplay between GBS and human immune cells at the colonizing site are just emerging.
Collapse
Affiliation(s)
- Sybille Landwehr-Kenzel
- Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine Berlin , Berlin , Germany ; Berlin-Brandenburg School for Regenerative Therapies, Charité University Medicine Berlin , Berlin , Germany ; Department of Pediatric Pulmonology and Immunology, Charité University Medicine Berlin , Berlin , Germany
| | - Philipp Henneke
- Center for Pediatrics and Adolescent Medicine, University Medical Center Freiburg , Freiburg , Germany ; Center for Chronic Immunodeficiency, University Medical Center Freiburg , Freiburg , Germany
| |
Collapse
|
78
|
Boldenow E, Hogan KA, Chames MC, Aronoff DM, Xi C, Loch-Caruso R. Role of cytokine signaling in group B Streptococcus-stimulated expression of human beta defensin-2 in human extraplacental membranes. Am J Reprod Immunol 2014; 73:263-72. [PMID: 25263616 DOI: 10.1111/aji.12325] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 08/27/2014] [Indexed: 01/16/2023] Open
Abstract
PROBLEM Group B Streptococcus (GBS) is a leading cause of neonatal morbidity and mortality. We tested the hypothesis that the choriodecidua plays a role in GBS-stimulated human beta defensin(HBD)-2 increases in amnion cells through a secreted factor of choriodecidual origin. METHOD OF STUDY Human amnion epithelial cells were treated with choriodecidual GBS-conditioned medium, live GBS, lipoteichoic acid (LTA), or lipopolysaccharide (LPS), with and without IL-1 inhibitors. RESULTS Choriodecidual tissue punches released IL-1α and IL-1β in response to GBS and this medium significantly stimulated release of HBD-2 by amnion cell cultures. Inhibitors of IL-1 significantly impaired the release of HBD-2 from amnion cells treated with GBS choriodecidual conditioned medium. Direct stimulation of amnion cells with GBS, LTA, or LPS did not increase HBD-2 release. CONCLUSION Paracrine signaling involving IL-1 of choriodecidual origin is likely a critical driver for amnion HBD-2 increases in response to GBS infection of extraplacental membranes.
Collapse
Affiliation(s)
- Erica Boldenow
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | | | | | | | | | | |
Collapse
|
79
|
Role of Toll-like receptor 13 in innate immune recognition of group B streptococci. Infect Immun 2014; 82:5013-22. [PMID: 25225249 DOI: 10.1128/iai.02282-14] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Murine Toll-like receptor 13 (TLR13), an endosomal receptor that is not present in humans, is activated by an unmethylated motif present in the large ribosomal subunit of bacterial RNA (23S rRNA). Little is known, however, of the impact of TLR13 on antibacterial host defenses. Here we examined the role of this receptor in the context of infection induced by the model pathogen group B streptococcus (GBS). To this end, we used bacterial strains masked from TLR13 recognition by virtue of constitutive expression of the ErmC methyltransferase, which results in dimethylation of the 23S rRNA motif at a critical adenine residue. We found that TLR13-mediated rRNA recognition was required for optimal induction of tumor necrosis factor alpha and nitrous oxide in dendritic cell and macrophage cultures stimulated with heat-killed bacteria or purified bacterial RNA. However, TLR13-dependent recognition was redundant when live bacteria were used as a stimulus. Moreover, masking bacterial rRNA from TLR13 recognition did not increase the ability of GBS to avoid host defenses and replicate in vivo. In contrast, increased susceptibility to infection was observed under conditions in which signaling by all endosomal TLRs was abolished, i.e., in mice with a loss-of-function mutation in the chaperone protein UNC93B1. Our data lend support to the conclusion that TLR13 participates in GBS recognition, although blockade of the function of this receptor can be compensated for by other endosomal TLRs. Lack of selective pressure by bacterial infections might explain the evolutionary loss of TLR13 in humans. However, further studies using different bacterial species are needed to prove this hypothesis.
Collapse
|
80
|
Abstract
Signal transduction via MyD88, an adaptor protein engaged by the Toll-like receptor (TLR) and interleukin-1 receptor (IL-1R) family receptors, has a crucial role in host defenses against group B streptococcus (GBS). To examine the contribution of IL-1R signaling to MyD88-dependent host defenses, we analyzed GBS infection in type I IL-1R (IL-1RI)-deficient mice. Most of these animals displayed clinical signs of sepsis and neurological disease and died after a challenge with a bacterial dose that did not cause illness or death in any of the wild-type animals. Moreover, bacterial numbers in the blood and brains of the immunodefective mice were considerably increased. The ability of blood leukocytes or bone marrow-derived macrophages to kill GBS in vitro was not affected by a lack of IL-1RI. However, it was found in a newly developed model of GBS-induced peritoneal inflammation that IL-1 signaling selectively promoted the production of the chemokines KC and MIP-1α and neutrophil recruitment. Moreover, the secretion of KC and MIP-1α, but not tumor necrosis factor alpha, by peritoneal macrophages stimulated with GBS was significantly decreased in the absence of IL-1RI. Accordingly, the number of neutrophils in the blood and the concentration of myeloperoxidase, a neutrophil marker, in infected organs were severely reduced in the immunodefective mice during GBS disease, concomitantly with a reduction in tissue KC and MIP-1α levels. In conclusion, IL-1RI plays a crucial role in host defenses against GBS by inducing the high-level production of chemokines and the subsequent recruitment of neutrophilic polymorphonuclear leukocytes to infection sites. Group B streptococcus (GBS) is a serious and frequent human pathogen. Experimental infection with this bacterium has been widely used to understand the mechanism whereby the body’s first line of defense, represented by cells and molecules of the innate immune system, fights infections. In both humans and mice, defective function of the adaptor molecule MyD88 has been associated with extreme susceptibility to infection by GBS and other extracellular bacteria. We show here that lack of signaling by interleukin-1 (IL-1) cytokines can largely, although not completely, explain the increased susceptibility to infection observed in the absence of MyD88 function. We show, in particular, that IL-1 signaling through the IL-1 receptor promotes the production of the leukocyte attractant chemokines KC and MIP-1α and recruitment of neutrophils to GBS infection sites, thereby enabling these leukocytes to clear the infection. Our findings indicate that stimulation of IL-1 signaling may be useful as an alternative therapeutic strategy to treat GBS infections.
Collapse
|
81
|
Rosa-Fraile M, Dramsi S, Spellerberg B. Group B streptococcal haemolysin and pigment, a tale of twins. FEMS Microbiol Rev 2014; 38:932-46. [PMID: 24617549 PMCID: PMC4315905 DOI: 10.1111/1574-6976.12071] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 02/18/2014] [Accepted: 03/03/2014] [Indexed: 12/11/2022] Open
Abstract
Group B streptococcus [(GBS or Streptococcus agalactiae)] is a leading cause of neonatal meningitis and septicaemia. Most clinical isolates express simultaneously a β-haemolysin/cytolysin and a red polyenic pigment, two phenotypic traits important for GBS identification in medical microbiology. The genetic determinants encoding the GBS haemolysin and pigment have been elucidated and the molecular structure of the pigment has been determined. The cyl operon involved in haemolysin and pigment production is regulated by the major two-component system CovS/R, which coordinates the expression of multiple virulence factors of GBS. Genetic analyses indicated strongly that the haemolysin activity was due to a cytolytic toxin encoded by cylE. However, the biochemical nature of the GBS haemolysin has remained elusive for almost a century because of its instability during purification procedures. Recently, it has been suggested that the haemolytic and cytolytic activity of GBS is due to the ornithine rhamnopolyenic pigment and not to the CylE protein. Here we review and summarize our current knowledge of the genetics, regulation and biochemistry of these twin GBS phenotypic traits, including their functions as GBS virulence factors.
Collapse
Affiliation(s)
| | - Shaynoor Dramsi
- Unité de Biologie des Bactéries Pathogènes à Gram positif, Institut PasteurParis, France
- CNRS ERL 3526Paris, France
| | - Barbara Spellerberg
- Institute of Medical Microbiology and Hygiene, University Hospital UlmUlm, Germany
| |
Collapse
|
82
|
The interleukin-1β/CXCL1/2/neutrophil axis mediates host protection against group B streptococcal infection. Infect Immun 2014; 82:4508-17. [PMID: 25114117 DOI: 10.1128/iai.02104-14] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Previous studies have indicated that group B streptococcus (GBS), a frequent human pathogen, potently induces the release of interleukin-1β (IL-1β), an important mediator of inflammatory responses. Since little is known about the role of this cytokine in GBS disease, we analyzed the outcome of infection in IL-1β-deficient mice. These animals were markedly sensitive to GBS infection, with most of them dying under challenge conditions that caused no deaths in wild-type control mice. Lethality was due to the inability of the IL-1β-deficient mice to control local GBS replication and dissemination to target organs, such as the brain and the kidneys. Moreover, in a model of inflammation induced by the intraperitoneal injection of killed GBS, a lack of IL-1β was associated with selective impairment in the production of the neutrophil chemokines CXCL1 and CXCL2 and in neutrophil recruitment to the peritoneal cavity. Decreased blood neutrophil counts and impaired neutrophil recruitment to the brain and kidneys were also observed during GBS infection in IL-1β-deficient mice concomitantly with a reduction in CXCL1 and CXCL2 tissue levels. Notably, the hypersusceptibility to GBS infection observed in the immune-deficient animals was recapitulated by neutrophil depletion with anti-Gr1 antibodies. Collectively, our data identify a cytokine circuit that involves IL-1β-induced production of CXCL1 and CXCL2 and leads the recruitment of neutrophils to GBS infection sites. Moreover, our data point to an essential role of these cells in controlling the progression and outcome of GBS disease.
Collapse
|
83
|
Okumura CYM, Nizet V. Subterfuge and sabotage: evasion of host innate defenses by invasive gram-positive bacterial pathogens. Annu Rev Microbiol 2014; 68:439-58. [PMID: 25002085 DOI: 10.1146/annurev-micro-092412-155711] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The development of a severe invasive bacterial infection in an otherwise healthy individual is one of the most striking and fascinating aspects of human medicine. A small cadre of gram-positive pathogens of the genera Streptococcus and Staphylococcus stand out for their unique invasive disease potential and sophisticated ability to counteract the multifaceted components of human innate defense. This review illustrates how these leading human disease agents evade host complement deposition and activation, impede phagocyte recruitment and activation, resist the microbicidal activities of host antimicrobial peptides and reactive oxygen species, escape neutrophil extracellular traps, and promote and accelerate phagocyte cell death through the action of pore-forming cytolysins. Understanding the molecular basis of bacterial innate immune resistance can open new avenues for therapeutic intervention geared to disabling specific virulence factors and resensitizing the pathogen to host innate immune clearance.
Collapse
Affiliation(s)
- Cheryl Y M Okumura
- Department of Biology, Occidental College, Los Angeles, California 90041;
| | | |
Collapse
|
84
|
Two novel functions of hyaluronidase from Streptococcus agalactiae are enhanced intracellular survival and inhibition of proinflammatory cytokine expression. Infect Immun 2014; 82:2615-25. [PMID: 24711564 DOI: 10.1128/iai.00022-14] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Streptococcus agalactiae is the causative agent of septicemia and meningitis in fish. Previous studies have shown that hyaluronidase (Hyl) is an important virulence factor in many Gram-positive bacteria. To investigate the role of S. agalactiae Hyl during interaction with macrophages, we inactivated the gene encoding extracellular hyaluronidase, hylB, in a clinical Hyl(+) isolate. The isogenic hylb mutant (Δhylb) displayed reduced survival in macrophages compared to the wild type and stimulated a significantly higher release of proinflammatory cytokines, such as interleukin-1β (IL-1β), IL-6, and tumor necrosis factor alpha (TNF-α), than the wild type in macrophages as well as in mice. Furthermore, only Hyl(+) strains could grow utilizing hyaluronic acid (HA) as the sole carbon source, suggesting that Hyl permits the organism to utilize host HA as an energy source. Fifty percent lethal dose (LD50) determinations in zebrafish demonstrated that the hylb mutant was highly attenuated relative to the wild-type strain. Experimental infection of BALB/c mice revealed that bacterial loads in the blood, spleen, and brain at 16 h postinfection were significantly reduced in the ΔhylB mutant compared to those in wild-type-infected mice. In conclusion, hyaluronidase has a strong influence on the intracellular survival of S. agalactiae and proinflammatory cytokine expression, suggesting that it plays a key role in S. agalactiae pathogenicity.
Collapse
|
85
|
Gupta R, Ghosh S, Monks B, DeOliveira RB, Tzeng TC, Kalantari P, Nandy A, Bhattacharjee B, Chan J, Ferreira F, Rathinam V, Sharma S, Lien E, Silverman N, Fitzgerald K, Firon A, Trieu-Cuot P, Henneke P, Golenbock DT. RNA and β-hemolysin of group B Streptococcus induce interleukin-1β (IL-1β) by activating NLRP3 inflammasomes in mouse macrophages. J Biol Chem 2014; 289:13701-5. [PMID: 24692555 DOI: 10.1074/jbc.c114.548982] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The inflammatory cytokine IL-1β is critical for host responses against many human pathogens. Here, we define Group B Streptococcus (GBS)-mediated activation of the Nod-like receptor-P3 (NLRP3) inflammasome in macrophages. NLRP3 activation requires GBS expression of the cytolytic toxin, β-hemolysin, lysosomal acidification, and leakage. These processes allow the interaction of GBS RNA with cytosolic NLRP3. The present study supports a model in which GBS RNA, along with lysosomal components including cathepsins, leaks out of lysosomes and interacts with NLRP3 to induce IL-1β production.
Collapse
Affiliation(s)
- Rahul Gupta
- From the Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Shubhendu Ghosh
- From the Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Brian Monks
- From the Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Rosane B DeOliveira
- From the Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Te-Chen Tzeng
- From the Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Parisa Kalantari
- From the Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Anubhab Nandy
- From the Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Bornali Bhattacharjee
- From the Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Jennie Chan
- From the Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Fabianno Ferreira
- the Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro (UFRJ), 21941-902 Rio de Janeiro, Brazil, and
| | - Vijay Rathinam
- From the Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Shruti Sharma
- From the Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Egil Lien
- From the Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Neal Silverman
- From the Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Katherine Fitzgerald
- From the Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Arnaud Firon
- the Institut Pasteur, Unité de Biologie des Bactéries Pathogènes à Gram-Positif, 75724 Paris Cedex 15, France
| | - Patrick Trieu-Cuot
- the Institut Pasteur, Unité de Biologie des Bactéries Pathogènes à Gram-Positif, 75724 Paris Cedex 15, France
| | - Philipp Henneke
- the Center of Chronic Immunodeficiency and Center of Pediatrics and Adolescent Medicine, University Medical Center Freiburg, 79106 Freiburg im Breisgau, Germany
| | - Douglas T Golenbock
- From the Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts 01605,
| |
Collapse
|
86
|
Activation and regulation of cellular inflammasomes: gaps in our knowledge for central nervous system injury. J Cereb Blood Flow Metab 2014; 34:369-75. [PMID: 24398940 PMCID: PMC3948131 DOI: 10.1038/jcbfm.2013.227] [Citation(s) in RCA: 246] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 11/19/2013] [Accepted: 12/01/2013] [Indexed: 02/07/2023]
Abstract
The inflammasome is an intracellular multiprotein complex involved in the activation of caspase-1 and the processing of the proinflammatory cytokines interleukin-1β (IL-1β) and IL-18. The inflammasome in the central nervous system (CNS) is involved in the generation of an innate immune inflammatory response through IL-1 cytokine release and in cell death through the process of pyroptosis. In this review, we consider the different types of inflammasomes (NLRP1, NLRP2, NLRP3, and AIM2) that have been described in CNS cells, namely neurons, astrocytes, and microglia. Importantly, we focus on the role of the inflammasome after brain and spinal cord injury and cover the potential activators of the inflammasome after CNS injury such as adenosine triphosphate and DNA, and the therapeutic potential of targeting the inflammasome to improve outcomes after CNS trauma.
Collapse
|
87
|
Beardsley PM, Hauser KF. Glial modulators as potential treatments of psychostimulant abuse. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2014; 69:1-69. [PMID: 24484974 DOI: 10.1016/b978-0-12-420118-7.00001-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Glia (including astrocytes, microglia, and oligodendrocytes), which constitute the majority of cells in the brain, have many of the same receptors as neurons, secrete neurotransmitters and neurotrophic and neuroinflammatory factors, control clearance of neurotransmitters from synaptic clefts, and are intimately involved in synaptic plasticity. Despite their prevalence and spectrum of functions, appreciation of their potential general importance has been elusive since their identification in the mid-1800s, and only relatively recently have they been gaining their due respect. This development of appreciation has been nurtured by the growing awareness that drugs of abuse, including the psychostimulants, affect glial activity, and glial activity, in turn, has been found to modulate the effects of the psychostimulants. This developing awareness has begun to illuminate novel pharmacotherapeutic targets for treating psychostimulant abuse, for which targeting more conventional neuronal targets has not yet resulted in a single, approved medication. In this chapter, we discuss the molecular pharmacology, physiology, and functional relationships that the glia have especially in the light in which they present themselves as targets for pharmacotherapeutics intended to treat psychostimulant abuse disorders. We then review a cross section of preclinical studies that have manipulated glial processes whose behavioral effects have been supportive of considering the glia as drug targets for psychostimulant-abuse medications. We then close with comments regarding the current clinical evaluation of relevant compounds for treating psychostimulant abuse, as well as the likelihood of future prospects.
Collapse
Affiliation(s)
| | - Kurt F Hauser
- Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
88
|
Zhou Y, Wang Y, Wang J, Anne Stetler R, Yang QW. Inflammation in intracerebral hemorrhage: from mechanisms to clinical translation. Prog Neurobiol 2013; 115:25-44. [PMID: 24291544 DOI: 10.1016/j.pneurobio.2013.11.003] [Citation(s) in RCA: 455] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 11/15/2013] [Accepted: 11/18/2013] [Indexed: 02/07/2023]
Abstract
Intracerebral hemorrhage (ICH) accounts for 10-15% of all strokes and is associated with high mortality and morbidity. Currently, no effective medical treatment is available to improve functional outcomes in patients with ICH. Potential therapies targeting secondary brain injury are arousing a great deal of interest in translational studies. Increasing evidence has shown that inflammation is the key contributor of ICH-induced secondary brain injury. Inflammation progresses in response to various stimuli produced after ICH. Hematoma components initiate inflammatory signaling via activation of microglia, subsequently releasing proinflammatory cytokines and chemokines to attract peripheral inflammatory infiltration. Hemoglobin (Hb), heme, and iron released after red blood cell lysis aggravate ICH-induced inflammatory injury. Danger associated molecular patterns such as high mobility group box 1 protein, released from damaged or dead cells, trigger inflammation in the late stage of ICH. Preclinical studies have identified inflammatory signaling pathways that are involved in microglial activation, leukocyte infiltration, toll-like receptor (TLR) activation, and danger associated molecular pattern regulation in ICH. Recent advances in understanding the pathogenesis of ICH-induced inflammatory injury have facilitated the identification of several novel therapeutic targets for the treatment of ICH. This review summarizes recent progress concerning the mechanisms underlying ICH-induced inflammation. We focus on the inflammatory signaling pathways involved in microglial activation and TLR signaling, and explore potential therapeutic interventions by targeting the removal of hematoma components and inhibition of TLR signaling.
Collapse
Affiliation(s)
- Yu Zhou
- Department of Neurology, Xinqiao Hospital & The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Yanchun Wang
- Department of Neurology, Xinqiao Hospital & The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Jian Wang
- Department of Anesthesiology/Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - R Anne Stetler
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Qing-Wu Yang
- Department of Neurology, Xinqiao Hospital & The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China.
| |
Collapse
|
89
|
Abstract
Human beings are constantly exposed to pathogens. The innate immune system is the first line of defense against microbes. It has evolved to recognize conserved microbial motifs (PAMP or pathogen-associated molecular patterns) thanks to a limited array of receptors termed pattern recognition receptors (PRR). Upon activation, most PRR trigger a transcriptional response leading to neosynthesis of hundreds of genes. In contrast, engagement of various PRR in the recently identified inflammasome complexes lead to activation of a cysteine protease, caspase-1. This inflammatory caspase has a dual activity: it triggers the release of very potent proinflammatory cytokines IL-1β and IL-18 and, an hyperinflammatory cell death termed pyroptosis. In this review, we describe the inflammasome receptors and their ligands, the molecular mechanisms leading to the assembly of this innate immune platform and the role of the inflammasome during viral and bacterial infections.
Collapse
Affiliation(s)
- Yvan Jamilloux
- Service de médecine interne, centre hospitalo-universitaire de la Croix-Rousse, 103, Grande rue de la Croix Rousse, 69004 Lyon, France - Centre international de recherche en infectiologie (CIRI), université de Lyon, Lyon, France - Inserm U1111, 21, avenue Tony Garnier, 69007 Lyon, France - CNRS, UMR 5308, Lyon, France - École normale supérieure, Lyon, France
| | | |
Collapse
|
90
|
Lappas M. Caspase-1 activation is increased with human labour in foetal membranes and myometrium and mediates infection-induced interleukin-1β secretion. Am J Reprod Immunol 2013; 71:189-201. [PMID: 24238269 DOI: 10.1111/aji.12174] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 10/11/2013] [Indexed: 12/21/2022] Open
Abstract
PROBLEM Interleukin-1β (IL-1β) is a pro-inflammatory cytokine that is involved in human parturition, especially in the context of infection-induced preterm birth. Caspase-1 is a key component of inflammasomes, which are activated upon infection to trigger the maturation of IL-1β. METHOD OF STUDY To determine the effect of human labour on caspase-1 activation in human foetal membranes and myometrium. In addition, the mechanisms by which inflammasome activation regulates IL-1β production were also be assessed. RESULTS Higher caspase-1 gene and protein expression were detected in foetal membranes myometrium obtained from term labouring women when compared with samples taken from non labouring women. Lipopolysaccharide induced the transcription and secretion of IL-1β from foetal membranes and myometrium; both events were dependent on nuclear factor kappa B (NF-κB). However, levels of extracellular IL-1β were greatly increased by subsequent treatment with the potassium-proton ionophore Adenosine triphosphate (ATP) or nigericin; an effect that was dependent on active caspase-1. Additionally, ATP induced IL-1β secretion via the purinergic P2X7 receptor, whereas the pannexin-1 channel was required for nigericin induced IL-1β secretion. CONCLUSION Taken together, these results demonstrate that caspase-1 activation is increased with human labour in foetal membranes and myometrium, and is required for infection-induced IL-1β secretion.
Collapse
Affiliation(s)
- Martha Lappas
- Department of Obstetrics and Gynaecology, University of Melbourne, Heidelberg, Vic., Australia; Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, Vic., Australia
| |
Collapse
|
91
|
Lemire P, Calzas C, Segura M. The NOD2 receptor does not play a major role in the pathogenesis of Group B Streptococcus in mice. Microb Pathog 2013; 65:41-7. [PMID: 24107312 DOI: 10.1016/j.micpath.2013.09.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 09/25/2013] [Accepted: 09/26/2013] [Indexed: 01/08/2023]
Abstract
Group B Streptococcus (GBS) capsular type III is an important agent of life-threatening invasive infections. It has been previously shown that encapsulated GBS is easily internalized by dendritic cells (DCs) and this internalization has an impact on cytokine production. The intracellular receptors or pathways underlying this response are not well understood. In this work, we investigated the role of NOD2 in the pathogenesis of GBS using a mouse model of infection. NOD2(-/-) mice showed similar levels of survival and bacteremia than control mice. Interestingly, ex vivo analysis of total spleen cells from infected animals showed that the absence of NOD2 results in reduced production of inflammatory cytokines. However this abridged inflammatory response does not seem to improve mouse survival. In conclusion, we demonstrated that NOD2 is not a crucial receptor to fight GBS infection and only partially contributes to the inflammatory response.
Collapse
Affiliation(s)
- Paul Lemire
- Laboratory of Immunology, Faculté de médecine vétérinaire, Université de Montréal, St-Hyacinthe, Québec, Canada
| | | | | |
Collapse
|
92
|
Kim JJ, Jo EK. NLRP3 inflammasome and host protection against bacterial infection. J Korean Med Sci 2013; 28:1415-23. [PMID: 24133343 PMCID: PMC3792593 DOI: 10.3346/jkms.2013.28.10.1415] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 08/20/2013] [Indexed: 01/30/2023] Open
Abstract
The inflammasome is a multi-protein complex that induces maturation of inflammatory cytokines interleukin (IL)-1β and IL-18 through activation of caspase-1. Several nucleotide binding oligomerization domain-like receptor family members, including NLRP3, recognize unique microbial and danger components and play a central role in inflammasome activation. The NLRP3 inflammasome is critical for maintenance of homeostasis against pathogenic infections. However, inflammasome activation acts as a double-edged sword for various bacterial infections. When the IL-1 family of cytokines is secreted excessively, they cause tissue damage and extensive inflammatory responses that are potentially hazardous for the host. Emerging evidence has shown that diverse bacterial pathogens or their components negatively regulate inflammasome activation to escape the immune response. In this review, we discuss the current knowledge of the roles and regulation of the NLRP3 inflammasome during bacterial infections. Activation and regulation of the NLRP3 inflammasome should be tightly controlled to prevent virulence and pathology during infections. Understanding the roles and regulatory mechanisms of the NLRP3 inflammasome is essential for developing potential treatment approaches against pathogenic infections.
Collapse
Affiliation(s)
- Jwa-Jin Kim
- Department of Microbiology and Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon, Korea
| | - Eun-Kyeong Jo
- Department of Microbiology and Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon, Korea
| |
Collapse
|
93
|
Andrade EB, Alves J, Madureira P, Oliveira L, Ribeiro A, Cordeiro-da-Silva A, Correia-Neves M, Trieu-Cuot P, Ferreira P. TLR2-Induced IL-10 Production Impairs Neutrophil Recruitment to Infected Tissues during Neonatal Bacterial Sepsis. THE JOURNAL OF IMMUNOLOGY 2013; 191:4759-68. [DOI: 10.4049/jimmunol.1301752] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
94
|
Barichello T, Fagundes GD, Generoso JS, Elias SG, Simões LR, Teixeira AL. Pathophysiology of neonatal acute bacterial meningitis. J Med Microbiol 2013; 62:1781-1789. [PMID: 23946474 DOI: 10.1099/jmm.0.059840-0] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Neonatal meningitis is a severe acute infectious disease of the central nervous system and an important cause of morbidity and mortality worldwide. The inflammatory reaction involves the meninges, the subarachnoid space and the brain parenchymal vessels and contributes to neuronal injury. Neonatal meningitis leads to deafness, blindness, cerebral palsy, seizures, hydrocephalus or cognitive impairment in approximately 25-50 % of survivors. Bacterial pathogens can reach the blood-brain barrier and be recognized by antigen-presenting cells through the binding of Toll-like receptors. They induce the activation of NFκB or mitogen-activated protein kinase pathways and subsequently upregulate leukocyte populations and express numerous proteins involved in inflammation and the immune response. Many brain cells can produce cytokines, chemokines and other pro-inflammatory molecules in response to bacterial stimuli, and polymorphonuclear leukocytes are attracted, activated and released in large amounts of superoxide anion and nitric oxide, leading to peroxynitrite formation and generating oxidative stress. This cascade leads to lipid peroxidation, mitochondrial damage and breakdown of the blood-brain barrier, thus contributing to cell injury during neonatal meningitis. This review summarizes information on the pathophysiology and adjuvant treatment of acute bacterial meningitis in neonates.
Collapse
Affiliation(s)
- Tatiana Barichello
- Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil.,Laboratório de Microbiologia Experimental e Instituto Nacional de Ciência e Tecnologia Translacional em Medicina, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil.,Núcleo de Excelência em Neurociências Aplicadas de Santa Catarina (NENASC), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil.,Center for Experimental Models in Psychiatry, Department of Psychiatry and Behavioral Sciences, Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Glauco D Fagundes
- Núcleo de Excelência em Neurociências Aplicadas de Santa Catarina (NENASC), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil.,Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil.,Laboratório de Microbiologia Experimental e Instituto Nacional de Ciência e Tecnologia Translacional em Medicina, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Jaqueline S Generoso
- Núcleo de Excelência em Neurociências Aplicadas de Santa Catarina (NENASC), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil.,Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil.,Laboratório de Microbiologia Experimental e Instituto Nacional de Ciência e Tecnologia Translacional em Medicina, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Samuel Galvão Elias
- Núcleo de Excelência em Neurociências Aplicadas de Santa Catarina (NENASC), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil.,Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil.,Laboratório de Microbiologia Experimental e Instituto Nacional de Ciência e Tecnologia Translacional em Medicina, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Lutiana R Simões
- Núcleo de Excelência em Neurociências Aplicadas de Santa Catarina (NENASC), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil.,Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil.,Laboratório de Microbiologia Experimental e Instituto Nacional de Ciência e Tecnologia Translacional em Medicina, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Antonio Lucio Teixeira
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
95
|
Ling E, Landau D, Krymko H. Protracted febrile myalgia in an afebrile child with familial Mediterranean fever. Int J Rheum Dis 2013; 17:212-3. [PMID: 24576277 DOI: 10.1111/1756-185x.12118] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Eduard Ling
- Pediatric Rheumatology Unit, Saban Pediatric Medical Center, Beer Sheva, Israel; Pediatrics Department "B", Saban Pediatric Medical Center, Beer Sheva, Israel
| | | | | |
Collapse
|
96
|
How does the brain limit the severity of inflammation and tissue injury during bacterial meningitis? J Neuropathol Exp Neurol 2013; 72:370-85. [PMID: 23584204 DOI: 10.1097/nen.0b013e3182909f2f] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The most devastating CNS bacterial infection, bacterial meningitis, has both acute and long-term neurologic consequences. The CNS defends itself against bacterial invasion through a combination of physical barriers (i.e. blood-brain barrier, meninges, and ependyma), which contain macrophages that express a range of pattern-recognition receptors that detect pathogens before they gain access to the CNS and cerebrospinal fluid. This activates an antipathogen response consisting of inflammatory cytokines, complement, and chemoattractants. Regulation of the antipathogen inflammatory response is essential for preventing irreversible brain injury and protecting stem cell populations in the ventricle wall. The severity of brain inflammation is regulated by the clearance of apoptotic inflammatory cells and neurons. Death signaling pathways are expressed by glia to stimulate apoptosis of neutrophils, lymphocytes, and damaged neurons and to regulate in flammation and remove necrotic cells. The emerging group of neuroimmunoregulatory molecules adjusts the balance of the anti-inflammatory and proinflammatory response to provide optimal conditions for effective clearance of pathogens and apoptotic cells but reduce the severity of the inflammatory response to prevent injury to brain cells, including stem cell populations. The neuroimmunoregulatory molecules and other CNS anti-inflammatory pathways represent potential therapeutic targets capable of reducing brain injury caused by bacterial infection.
Collapse
|
97
|
Chen KW, Schroder K. Antimicrobial functions of inflammasomes. Curr Opin Microbiol 2013; 16:311-8. [DOI: 10.1016/j.mib.2013.02.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 02/04/2013] [Accepted: 02/07/2013] [Indexed: 12/14/2022]
|
98
|
Higa N, Toma C, Nohara T, Nakasone N, Takaesu G, Suzuki T. Lose the battle to win the war: bacterial strategies for evading host inflammasome activation. Trends Microbiol 2013; 21:342-9. [PMID: 23712018 DOI: 10.1016/j.tim.2013.04.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 03/31/2013] [Accepted: 04/22/2013] [Indexed: 01/20/2023]
Abstract
The inflammasome is composed of nucleotide-binding, oligomerization domain (NOD)-like receptor (NLR) proteins, and leads to caspase-1 activation and subsequent secretion of the proinflammatory cytokines interleukin 1β (IL-1β) and interleukin-18 (IL-18). After certain pathogenic bacteria infect host cells, such as macrophages, NLR-mediated inflammasome activation is triggered to form part of the host defenses against the invading pathogens. However, recent evidence has shown that bacteria have strategies for evading inflammasome activation in host cells. In this review, we focus on NLR-mediated inflammasome activation and bacterial evasion of the inflammasome as part of the battle between the host defenses and pathogens.
Collapse
Affiliation(s)
- Naomi Higa
- Department of Molecular Bacteriology and Immunology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Okinawa 903-0125, Japan
| | | | | | | | | | | |
Collapse
|
99
|
Neonatal immune adaptation of the gut and its role during infections. Clin Dev Immunol 2013; 2013:270301. [PMID: 23737810 PMCID: PMC3659470 DOI: 10.1155/2013/270301] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 04/03/2013] [Indexed: 12/22/2022]
Abstract
The intestinal tract is engaged in a relationship with a dense and complex microbial ecosystem, the microbiota. The establishment of this symbiosis is essential for host physiology, metabolism, and immune homeostasis. Because newborns are essentially sterile, the first exposure to microorganisms and environmental endotoxins during the neonatal period is followed by a crucial sequence of active events leading to immune tolerance and homeostasis. Contact with potent immunostimulatory molecules starts immediately at birth, and the discrimination between commensal bacteria and invading pathogens is essential to avoid an inappropriate immune stimulation and/or host infection. The dysregulation of these tight interactions between host and microbiota can be responsible for important health disorders, including inflammation and sepsis. This review summarizes the molecular events leading to the establishment of postnatal immune tolerance and how pathogens can avoid host immunity and induce neonatal infections and sepsis.
Collapse
|
100
|
Role of pore-forming toxins in neonatal sepsis. Clin Dev Immunol 2013; 2013:608456. [PMID: 23710203 PMCID: PMC3655490 DOI: 10.1155/2013/608456] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 03/27/2013] [Indexed: 11/17/2022]
Abstract
Protein toxins are important virulence factors contributing to neonatal sepsis. The major pathogens of neonatal sepsis, group B Streptococci, Escherichia coli, Listeria monocytogenes, and Staphylococcus aureus, secrete toxins of different molecular nature, which are key for defining the disease. Amongst these toxins are pore-forming exotoxins that are expressed as soluble monomers prior to engagement of the target cell membrane with subsequent formation of an aqueous membrane pore. Membrane pore formation is not only a means for immediate lysis of the targeted cell but also a general mechanism that contributes to penetration of epithelial barriers and evasion of the immune system, thus creating survival niches for the pathogens. Pore-forming toxins, however, can also contribute to the induction of inflammation and hence to the manifestation of sepsis. Clearly, pore-forming toxins are not the sole factors that drive sepsis progression, but they often act in concert with other bacterial effectors, especially in the initial stages of neonatal sepsis manifestation.
Collapse
|