Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 205
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 19(4): 327-341, 2018 04.
Article in English | MEDLINE | ID: mdl-29507356

ABSTRACT

Trauma can affect any individual at any location and at any time over a lifespan. The disruption of macrobarriers and microbarriers induces instant activation of innate immunity. The subsequent complex response, designed to limit further damage and induce healing, also represents a major driver of complications and fatal outcome after injury. This Review aims to provide basic concepts about the posttraumatic response and is focused on the interactive events of innate immunity at frequent sites of injury: the endothelium at large, and sites within the lungs, inside and outside the brain and at the gut barrier.


Subject(s)
Immunity, Innate/immunology , Wounds and Injuries/immunology , Animals , Humans
2.
Immunity ; 47(1): 3-5, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28723551

ABSTRACT

Following a severe primary infection or trauma, the risk of developing pneumonia increases due to acquired immune defects collectively known as sepsis-induced immunosuppression. In this issue of Immunity, Roquilly et al. (2017) show that dendritic cells and macrophages developing in the lung after the resolution of a severe infection acquire tolerogenic properties that contribute to persistent immunosuppression and susceptibility to secondary infections.


Subject(s)
Immune Tolerance , Sepsis/immunology , Dendritic Cells/immunology , Humans , Immunosuppression Therapy , Macrophages/immunology
3.
Proc Natl Acad Sci U S A ; 120(40): e2215421120, 2023 10 03.
Article in English | MEDLINE | ID: mdl-37756334

ABSTRACT

Externalized histones erupt from the nucleus as extracellular traps, are associated with several acute and chronic lung disorders, but their implications in the molecular pathogenesis of interstitial lung disease are incompletely defined. To investigate the role and molecular mechanisms of externalized histones within the immunologic networks of pulmonary fibrosis, we studied externalized histones in human and animal bronchoalveolar lavage (BAL) samples of lung fibrosis. Neutralizing anti-histone antibodies were administered in bleomycin-induced fibrosis of C57BL/6 J mice, and subsequent studies used conditional/constitutive knockout mouse strains for TGFß and IL-27 signaling along with isolated platelets and cultured macrophages. We found that externalized histones (citH3) were significantly (P < 0.01) increased in cell-free BAL fluids of patients with idiopathic pulmonary fibrosis (IPF; n = 29) as compared to healthy controls (n = 10). The pulmonary sources of externalized histones were Ly6G+CD11b+ neutrophils and nonhematopoietic cells after bleomycin in mice. Neutralizing monoclonal anti-histone H2A/H4 antibodies reduced the pulmonary collagen accumulation and hydroxyproline concentration. Histones activated platelets to release TGFß1, which signaled through the TGFbRI/TGFbRII receptor complex on LysM+ cells to antagonize macrophage-derived IL-27 production. TGFß1 evoked multiple downstream mechanisms in macrophages, including p38 MAPK, tristetraprolin, IL-10, and binding of SMAD3 to the IL-27 promotor regions. IL-27RA-deficient mice displayed more severe collagen depositions suggesting that intact IL-27 signaling limits fibrosis. In conclusion, externalized histones inactivate a safety switch of antifibrotic, macrophage-derived IL-27 by boosting platelet-derived TGFß1. Externalized histones are accessible to neutralizing antibodies for improving the severity of experimental pulmonary fibrosis.


Subject(s)
Idiopathic Pulmonary Fibrosis , Interleukin-27 , Humans , Mice , Animals , Mice, Inbred C57BL , Histones , Blood Platelets , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/genetics
4.
Immunity ; 39(5): 801-2, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24238336

ABSTRACT

Special proresolving mediators (SPMs) limit the intensity of inflammation; however, their regulation is poorly understood. In this issue of Immunity, Li et al. (2013) describe bipolar roles for miR-466l in the promotion of inflammation and the induction of SPMs.


Subject(s)
Inflammation Mediators/metabolism , Macrophages/metabolism , MicroRNAs/physiology , Neutrophils/metabolism , Peritonitis/metabolism , Sepsis/blood , Animals , Humans , Male
5.
J Immunol ; 205(1): 251-260, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32444389

ABSTRACT

Over the first days of polymicrobial sepsis, there is robust activation of the innate immune system, causing the appearance of proinflammatory cytokines and chemokines, along with the appearance of extracellular histones, which are highly proinflammatory and prothrombotic. In the current study, we studied different innate immune responses in mice with knockout (KO) of complement protein 6 (C6). Polymorphonuclear neutrophils (PMNs) from these KO mice had defective innate immune responses, including defective expression of surface adhesion molecules, generation of superoxide anion, and appearance of reactive oxygen species and histone release after activation of PMNs, along with defective phagocytosis. In addition, in C6-/- mice, the NLRP3 inflammasome was defective both in PMNs and in macrophages. When these KO mice were subjected to polymicrobial sepsis, their survival was improved, associated with reduced levels in the plasma of proinflammatory cytokines and chemokines and lower levels of histones in plasma. In addition, sepsis-induced cardiac dysfunction was attenuated in these KO mice. In a model of acute lung injury induced by LPS, C6-/- mice showed reduced PMN buildup and less lung epithelial/endothelial cell dysfunction (edema and hemorrhage). These data indicate that C6-/- mice have reduced innate immune responses that result in less organ injury and improved survival after polymicrobial sepsis.


Subject(s)
Acute Lung Injury/immunology , Cardiomyopathies/immunology , Coinfection/immunology , Complement C6/metabolism , Immunity, Innate , Sepsis/immunology , Acute Lung Injury/diagnosis , Acute Lung Injury/pathology , Animals , Cardiomyopathies/diagnosis , Cardiomyopathies/pathology , Coinfection/complications , Coinfection/diagnosis , Coinfection/pathology , Complement C6/genetics , Disease Models, Animal , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Male , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Sepsis/complications , Sepsis/diagnosis , Sepsis/genetics , Severity of Illness Index
6.
J Immunol ; 202(3): 931-942, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30578307

ABSTRACT

Sepsis is the leading cause of death in the intensive care unit with an overall mortality rate of 20%. Individuals who are obese and have type 2 diabetes have increased recurrent, chronic, nosocomial infections that worsen the long-term morbidity and mortality from sepsis. Additionally, animal models of sepsis have shown that obese, diabetic mice have lower survival rates compared with nondiabetic mice. Neutrophils are essential for eradication of bacteria, prevention of infectious complications, and sepsis survival. In diabetic states, there is a reduction in neutrophil chemotaxis, phagocytosis, and reactive oxygen species (ROS) generation; however, few studies have investigated the extent to which these deficits compromise infection eradication and mortality. Using a cecal ligation and puncture model of sepsis in lean and in diet-induced obese mice, we demonstrate that obese diabetic mice have decreased "emergency hematopoiesis" after an acute infection. Additionally, both neutrophils and monocytes in obese, diabetic mice have functional defects, with decreased phagocytic ability and a decreased capacity to generate ROS. Neutrophils isolated from obese diabetic mice have decreased transcripts of Axl and Mertk, which partially explains the phagocytic dysfunction. Furthermore, we found that exogenous GM-CSF administration improves sepsis survival through enhanced neutrophil and monocytes phagocytosis and ROS generation abilities in obese, diabetic mice with sepsis.


Subject(s)
Diabetes Mellitus, Experimental/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Immunity, Innate/drug effects , Obesity/immunology , Sepsis/immunology , Animals , Bacteria , Cytokines/genetics , Cytokines/immunology , Diabetes Mellitus, Experimental/microbiology , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Monocytes/immunology , Monocytes/pathology , Neutrophils/immunology , Neutrophils/pathology , Obesity/microbiology , Phagocytosis , Sepsis/drug therapy , Sepsis/microbiology
7.
Immunol Rev ; 274(1): 330-353, 2016 11.
Article in English | MEDLINE | ID: mdl-27782333

ABSTRACT

Sepsis occurs when an infection exceeds local tissue containment and induces a series of dysregulated physiologic responses that result in organ dysfunction. A subset of patients with sepsis progress to septic shock, defined by profound circulatory, cellular, and metabolic abnormalities, and associated with a greater mortality. Historically, sepsis-induced organ dysfunction and lethality were attributed to the complex interplay between the initial inflammatory and later anti-inflammatory responses. With advances in intensive care medicine and goal-directed interventions, early 30-day sepsis mortality has diminished, only to steadily escalate long after "recovery" from acute events. As so many sepsis survivors succumb later to persistent, recurrent, nosocomial, and secondary infections, many investigators have turned their attention to the long-term sepsis-induced alterations in cellular immune function. Sepsis clearly alters the innate and adaptive immune responses for sustained periods of time after clinical recovery, with immune suppression, chronic inflammation, and persistence of bacterial representing such alterations. Understanding that sepsis-associated immune cell defects correlate with long-term mortality, more investigations have centered on the potential for immune modulatory therapy to improve long-term patient outcomes. These efforts are focused on more clearly defining and effectively reversing the persistent immune cell dysfunction associated with long-term sepsis mortality.


Subject(s)
Immune System , Immunity , Immunomodulation , Inflammation/immunology , Sepsis/immunology , Animals , Disease Progression , Humans , Immunosuppression Therapy , Inflammation/therapy , Secondary Prevention , Sepsis/therapy , Time Factors , Treatment Outcome
8.
FASEB J ; 31(9): 4129-4139, 2017 09.
Article in English | MEDLINE | ID: mdl-28572445

ABSTRACT

Polymicrobial sepsis in mice causes myocardial dysfunction after generation of the complement anaphylatoxin, complement component 5a (C5a). C5a interacts with its receptors on cardiomyocytes (CMs), resulting in redox imbalance and cardiac dysfunction that can be functionally measured and quantitated using Doppler echocardiography. In this report we have evaluated activation of MAPKs and Akt in CMs exposed to C5a in vitro and after cecal ligation and puncture (CLP) in vivo In both cases, C5a in vitro caused activation (phosphorylation) of MAPKs and Akt in CMs, which required availability of both C5a receptors. Using immunofluorescence technology, activation of MAPKs and Akt occurred in left ventricular (LV) CMs, requiring both C5a receptors, C5aR1 and -2. Use of a water-soluble p38 inhibitor curtailed activation in vivo of MAPKs and Akt in LV CMs as well as the appearance of cytokines and histones in plasma from CLP mice. When mouse macrophages were exposed in vitro to LPS, activation of MAPKs and Akt also occurred. The copresence of the p38 inhibitor blocked these activation responses. Finally, the presence of the p38 inhibitor in CLP mice reduced the development of cardiac dysfunction. These data suggest that polymicrobial sepsis causes cardiac dysfunction that appears to be linked to activation of MAPKs and Akt in heart.-Fattahi, F., Kalbitz, M., Malan, E. A., Abe, E., Jajou, L., Huber-Lang, M. S., Bosmann, M., Russell, M. W., Zetoune, F. S., Ward, P. A. Complement-induced activation of MAPKs and Akt during sepsis: role in cardiac dysfunction.


Subject(s)
Complement C5a/metabolism , Gene Expression Regulation/physiology , Heart Diseases/etiology , Mitogen-Activated Protein Kinase Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sepsis/metabolism , Animals , Complement C5a/genetics , Heart Diseases/metabolism , Interleukins , Male , Mitogen-Activated Protein Kinase Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Rats , Rats, Sprague-Dawley , Receptor, Anaphylatoxin C5a/genetics , Receptor, Anaphylatoxin C5a/metabolism
9.
J Immunol ; 197(6): 2353-61, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27521340

ABSTRACT

There is accumulating evidence during sepsis that cardiomyocyte (CM) homeostasis is compromised, resulting in cardiac dysfunction. An important role for complement in these outcomes is now demonstrated. Addition of C5a to electrically paced CMs caused prolonged elevations of intracellular Ca(2+) concentrations during diastole, together with the appearance of spontaneous Ca(2+) transients. In polymicrobial sepsis in mice, we found that three key homeostasis-regulating proteins in CMs were reduced: Na(+)/K(+)-ATPase, which is vital for effective action potentials in CMs, and two intracellular Ca(2+) concentration regulatory proteins, that is, sarcoplasmic/endoplasmic reticulum calcium ATPase 2 and the Na(+)/Ca(2+) exchanger. Sepsis caused reduced mRNA levels and reductions in protein concentrations in CMs for all three proteins. The absence of either C5a receptor mitigated sepsis-induced reductions in the three regulatory proteins. Absence of either C5a receptor (C5aR1 or C5aR2) diminished development of defective systolic and diastolic echocardiographic/Doppler parameters developing in the heart (cardiac output, left ventricular stroke volume, isovolumic relaxation, E' septal annulus, E/E' septal annulus, left ventricular diastolic volume). We also found in CMs from septic mice the presence of defective current densities for Ik1, l-type calcium channel, and Na(+)/Ca(2+) exchanger. These defects were accentuated in the copresence of C5a. These data suggest complement-related mechanisms responsible for development of cardiac dysfunction during sepsis.


Subject(s)
Coinfection/immunology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/pathology , Sepsis/immunology , Sepsis/physiopathology , Animals , Calcium/metabolism , Calcium Channels, L-Type/immunology , Coinfection/microbiology , Coinfection/physiopathology , Complement C5a/immunology , Cytoplasm/chemistry , Cytoplasm/metabolism , Heart/physiopathology , Mice , Myocytes, Cardiac/microbiology , Receptor, Anaphylatoxin C5a/deficiency , Receptor, Anaphylatoxin C5a/immunology , Receptor, Anaphylatoxin C5a/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/immunology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sepsis/complications
10.
PLoS Pathog ; 11(9): e1005140, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26402732

ABSTRACT

The in utero environment profoundly impacts childhood neurodevelopment and behaviour. A substantial proportion of pregnancies in Africa are at risk of malaria in pregnancy (MIP) however the impact of in utero exposure to MIP on fetal neurodevelopment is unknown. Complement activation, in particular C5a, may contribute to neuropathology and adverse outcomes during MIP. We used an experimental model of MIP and standardized neurocognitive testing, MRI, micro-CT and HPLC analysis of neurotransmitter levels, to test the hypothesis that in utero exposure to malaria alters neurodevelopment through a C5a-C5aR dependent pathway. We show that malaria-exposed offspring have persistent neurocognitive deficits in memory and affective-like behaviour compared to unexposed controls. These deficits were associated with reduced regional brain levels of major biogenic amines and BDNF that were rescued by disruption of C5a-C5aR signaling using genetic and functional approaches. Our results demonstrate that experimental MIP induces neurocognitive deficits in offspring and suggest novel targets for intervention.


Subject(s)
Complement C5a/metabolism , Host-Parasite Interactions , Malaria/physiopathology , Neurocognitive Disorders/etiology , Neurogenesis , Pregnancy Complications, Parasitic/physiopathology , Receptor, Anaphylatoxin C5a/metabolism , Animals , Biogenic Amines/metabolism , Brain/blood supply , Brain/immunology , Brain/metabolism , Brain/pathology , Brain-Derived Neurotrophic Factor/metabolism , Cerebrovascular Circulation , Down-Regulation , Female , Fetal Development , Malaria/immunology , Malaria/metabolism , Malaria/parasitology , Male , Mice, Inbred BALB C , Mice, Knockout , Neurocognitive Disorders/immunology , Neurocognitive Disorders/metabolism , Neurocognitive Disorders/pathology , Neurons/immunology , Neurons/metabolism , Neurons/pathology , Plasmodium berghei/immunology , Plasmodium berghei/physiology , Pregnancy , Pregnancy Complications, Parasitic/immunology , Pregnancy Complications, Parasitic/metabolism , Pregnancy Complications, Parasitic/parasitology , Receptor, Anaphylatoxin C5a/genetics , Signal Transduction
11.
FASEB J ; 30(12): 3997-4006, 2016 12.
Article in English | MEDLINE | ID: mdl-27543123

ABSTRACT

Cardiac dysfunction develops during sepsis in humans and rodents. In the model of polymicrobial sepsis induced by cecal ligation and puncture (CLP), we investigated the role of the NLRP3 inflammasome in the heart. Mouse heart homogenates from sham-procedure mice contained high mRNA levels of NLRP3 and IL-1ß. Using the inflammasome protocol, exposure of cardiomyocytes (CMs) to LPS followed by ATP or nigericin caused release of mature IL-1ß. Immunostaining of left ventricular frozen sections before and 8 h after CLP revealed the presence of NLRP3 and IL-1ß proteins in CMs. CLP caused substantial increases in mRNAs for IL-1ß and NLRP3 in CMs which are reduced in the absence of either C5aR1 or C5aR2. After CLP, NLRP3-/- mice showed reduced plasma levels of IL-1ß and IL-6. In vitro exposure of wild-type CMs to recombinant C5a (rC5a) caused elevations in both cytosolic and nuclear/mitochondrial reactive oxygen species (ROS), which were C5a-receptor dependent. Use of a selective NOX2 inhibitor prevented increased cytosolic and nuclear/mitochondrial ROS levels and release of IL-1ß. Finally, NLRP3-/- mice had reduced defects in echo/Doppler parameters in heart after CLP. These studies establish that the NLRP3 inflammasome contributes to the cardiomyopathy of polymicrobial sepsis.-Kalbitz, M., Fattahi, F., Grailer, J. J., Jajou, L., Malan, E. A., Zetoune, F. S., Huber-Lang, M., Russell, M. W., Ward, P. A. Complement-induced activation of the cardiac NLRP3 inflammasome in sepsis.


Subject(s)
Complement C5a/metabolism , Inflammasomes/metabolism , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Sepsis/metabolism , Animals , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Mice , Rats , Reactive Oxygen Species/metabolism
12.
J Immunol ; 194(3): 868-72, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25539817

ABSTRACT

In the early stages of sepsis, lymphocytes undergo apoptosis, resulting in lymphopenia and immunosuppression. The trigger for septic lymphopenia is unknown. Using the polymicrobial model of murine sepsis, we investigated the role of C5a receptors in septic lymphopenia. In wild-type mice, cecal ligation and puncture resulted in splenocyte apoptosis and significant lymphopenia after 3 d, which was not observed in C5aR1(-/-) or C5aR2(-/-) mice. Our data show that mouse neutrophils exposed to recombinant mouse C5a cause release of histones in a dose-dependent and time-dependent manner. Histone levels in spleen were significantly elevated following cecal ligation and puncture but were reduced by the absence of C5aR1. Histones induced significant lymphocyte apoptosis in vitro. Ab-mediated neutralization of histones prevented the development of lymphopenia in sepsis. Together, these results describe a new pathway of septic lymphopenia involving complement and extracellular histones. Targeting of this pathway may have therapeutic benefit for patients with sepsis or other serious illness.


Subject(s)
Lymphopenia/etiology , Lymphopenia/metabolism , Receptor, Anaphylatoxin C5a/metabolism , Sepsis/complications , Animals , Apoptosis , Complement C5a/immunology , Complement System Proteins/immunology , Complement System Proteins/metabolism , Disease Models, Animal , Extracellular Space , Histones/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Male , Mice , Mice, Knockout , Receptor, Anaphylatoxin C5a/genetics
13.
Trends Immunol ; 34(3): 129-36, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23036432

ABSTRACT

The pathophysiology of sepsis and its accompanying systemic inflammatory response syndrome (SIRS) and the events that lead to multiorgan failure and death are poorly understood. It is known that, in septic humans and rodents, the development of SIRS is associated with a loss of the redox balance, but SIRS can also develop in noninfectious states. In addition, a hyperinflammatory state develops, together with impaired innate immune functions of phagocytes, immunosuppression, and complement activation, collectively leading to septic shock and lethality. Here, we discuss recent insights into the signaling pathways in immune and phagocytic cells that underlie sepsis and SIRS and consider how these might be targeted for therapeutic interventions to reverse or attenuate pathways that lead to lethality during sepsis.


Subject(s)
Inflammation , Sepsis/immunology , Sepsis/physiopathology , Systemic Inflammatory Response Syndrome , Animals , Cytokines/blood , Humans , Inflammation/immunology , Inflammation/physiopathology , Phagocytosis , Sepsis/complications , Signal Transduction , Systemic Inflammatory Response Syndrome/complications , Systemic Inflammatory Response Syndrome/immunology , Systemic Inflammatory Response Syndrome/physiopathology
14.
FASEB J ; 29(9): 3762-72, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25999468

ABSTRACT

Excessive activation of the complement system is detrimental in acute inflammatory disorders. In this study, we analyzed the role of complement-derived anaphylatoxins in the pathogenesis of experimental acute lung injury/acute respiratory distress syndrome (ALI/ARDS) in C57BL/6J mice. Intratracheal administration of recombinant mouse complement component (C5a) caused alveolar inflammation with abundant recruitment of Ly6-G(+)CD11b(+) leukocytes to the alveolar spaces and severe alveolar-capillary barrier dysfunction (C5a-ALI; EC(50[C5a]) = 20 ng/g body weight). Equimolar concentrations of C3a or desarginated C5a (C5a(desArg)) did not induce alveolar inflammation. The severity of C5a-ALI was aggravated in C5-deficient mice. Depletion of Ly6-G(+) cells and use of C5aR1(-/-) bone marrow chimeras suggested an essential role of C5aR1(+) hematopoietic cells in C5a-ALI. Blockade of PI3K/Akt and MEK1/2 kinase pathways completely abrogated lung injury. The mechanistic description is that C5a altered the alveolar cytokine milieu and caused significant release of CC-chemokines. Mice with genetic deficiency of CC-chemokine receptor (CCR) type 5, the common receptor of chemokine (C-C motif) ligand (CCL) 3, CCL4, and CCL5, displayed reduced lung damage. Moreover, treatment with a CCR5 antagonist, maraviroc, was protective against C5a-ALI. In summary, our results suggest that the detrimental effects of C5a in this model are partly mediated through CCR5 activation downstream of C5aR1, which may be evaluated for potential therapeutic exploitation in ALI/ARDS.


Subject(s)
Acute Lung Injury/metabolism , Complement Activation , Complement C3a/metabolism , Complement C5a, des-Arginine/metabolism , Pulmonary Alveoli/metabolism , Receptors, CCR5/metabolism , Acute Lung Injury/drug therapy , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Animals , CCR5 Receptor Antagonists/pharmacology , Complement C3a/genetics , Complement C5a, des-Arginine/genetics , Cyclohexanes/pharmacology , Leukocytes/metabolism , Leukocytes/pathology , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/genetics , MAP Kinase Kinase 2/metabolism , Maraviroc , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Alveoli/pathology , Receptor, Anaphylatoxin C5a/genetics , Receptor, Anaphylatoxin C5a/metabolism , Receptors, CCR5/genetics , Triazoles/pharmacology
15.
FASEB J ; 29(5): 2185-93, 2015 May.
Article in English | MEDLINE | ID: mdl-25681459

ABSTRACT

The purpose of this study was to define the relationship in polymicrobial sepsis (in adult male C57BL/6 mice) between heart dysfunction and the appearance in plasma of extracellular histones. Procedures included induction of sepsis by cecal ligation and puncture and measurement of heart function using echocardiogram/Doppler parameters. We assessed the ability of histones to cause disequilibrium in the redox status and intracellular [Ca(2+)]i levels in cardiomyocytes (CMs) (from mice and rats). We also studied the ability of histones to disturb both functional and electrical responses of hearts perfused with histones. Main findings revealed that extracellular histones appearing in septic plasma required C5a receptors, polymorphonuclear leukocytes (PMNs), and the Nacht-, LRR-, and PYD-domains-containing protein 3 (NLRP3) inflammasome. In vitro exposure of CMs to histones caused loss of homeostasis of the redox system and in [Ca(2+)]i, as well as defects in mitochondrial function. Perfusion of hearts with histones caused electrical and functional dysfunction. Finally, in vivo neutralization of histones in septic mice markedly reduced the parameters of heart dysfunction. Histones caused dysfunction in hearts during polymicrobial sepsis. These events could be attenuated by histone neutralization, suggesting that histones may be targets in the setting of sepsis to reduce cardiac dysfunction.


Subject(s)
Cardiomyopathies/etiology , Disease Models, Animal , Histones/adverse effects , Mitochondria/pathology , Sepsis/complications , Animals , Calcium/metabolism , Cardiomyopathies/blood , Cardiomyopathies/diagnosis , Carrier Proteins/physiology , Caspase 1/physiology , Cells, Cultured , Histones/blood , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Receptor, Anaphylatoxin C5a/metabolism , Sepsis/blood , Sepsis/pathology , Toll-Like Receptor 2/physiology , Toll-Like Receptor 4/physiology
16.
J Pineal Res ; 60(4): 405-14, 2016 May.
Article in English | MEDLINE | ID: mdl-26888116

ABSTRACT

Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are clinically severe respiratory disorders, and there are currently no Food and Drug Administration-approved drug therapies. Melatonin is a well-known anti-inflammatory molecule, which has proven to be effective in ALI induced by many conditions. Emerging studies suggest that the NLRP3 inflammasome plays a critical role during ALI. How melatonin directly blocks activation of the NLRP3 inflammasome in ALI remains unclear. In this study, using an LPS-induced ALI mouse model, we found intratracheal (i.t.) administration of melatonin markedly reduced the pulmonary injury and decreased the infiltration of macrophages and neutrophils into lung. During ALI, the NLRP3 inflammasome is significantly activated with a large amount of IL-1ß and the activated caspase-1 occurring in the lung. Melatonin inhibits the activation of the NLRP3 inflammasome by both suppressing the release of extracellular histones and directly blocking histone-induced NLRP3 inflammasome activation. Notably, i.t. route of melatonin administration opens a more efficient therapeutic approach for treating ALI.


Subject(s)
Acute Lung Injury/pathology , Anti-Inflammatory Agents/pharmacology , Inflammasomes/drug effects , Melatonin/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Acute Lung Injury/metabolism , Animals , Blotting, Western , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Inflammasomes/metabolism , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , X-Ray Microtomography
17.
J Immunol ; 192(12): 5974-83, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24795455

ABSTRACT

The inflammasome is a key factor in innate immunity and senses soluble pathogen and danger-associated molecular patterns as well as biological crystals (urate, cholesterol, etc.), resulting in expression of IL-1ß and IL-18. Using a standard model of acute lung injury (ALI) in mice featuring airway instillation of LPS, ALI was dependent on availability of NLRP3 as well as caspase-1, which are known features of the NLRP3 inflammasome. The appearance of IL-1ß, a product of NLRP3 inflammasome activation, was detected in bronchoalveolar lavage fluids (BALF) in a macrophage- and neutrophil-dependent manner. Neutrophil-derived extracellular histones appeared in the BALF during ALI and directly activated the NLRP3 inflammasome. Ab-mediated neutralization of histones significantly reduced IL-1ß levels in BALF during ALI. Inflammasome activation by extracellular histones in LPS-primed macrophages required NLRP3 and caspase-1 as well as extrusion of K(+), increased intracellular Ca(2+) concentration, and generation of reactive oxygen species. NLRP3 and caspase-1 were also required for full extracellular histone presence during ALI, suggesting a positive feedback mechanism. Extracellular histone and IL-1ß levels in BALF were also elevated in C5a-induced and IgG immune complex ALI models, suggesting a common inflammatory mechanism. These data indicate an interaction between extracellular histones and the NLRP3 inflammasome, resulting in ALI. Such findings suggest novel targets for treatment of ALI, for which there is currently no known efficacious drug.


Subject(s)
Acute Lung Injury/immunology , Carrier Proteins/immunology , Inflammasomes/immunology , Macrophages, Alveolar/immunology , Neutrophils/immunology , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Animals , Bronchoalveolar Lavage Fluid/immunology , Carrier Proteins/genetics , Caspase 1/genetics , Caspase 1/immunology , Disease Models, Animal , Histones/genetics , Histones/immunology , Inflammasomes/genetics , Interleukin-18/genetics , Interleukin-18/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Lipopolysaccharides/pharmacology , Macrophages, Alveolar/pathology , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Neutrophils/pathology
18.
J Immunol ; 193(11): 5668-77, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25348624

ABSTRACT

Severe sepsis and septic shock are leading causes of morbidity and mortality worldwide. Infection-associated inflammation promotes the development and progression of adverse outcomes in sepsis. The effects of heterodimeric IL-27 (p28/EBI3) have been implicated in the natural course of sepsis, whereas the molecular mechanisms underlying the regulation of gene expression and release of IL-27 in sepsis are poorly understood. We studied the events regulating the p28 subunit of IL-27 in endotoxic shock and polymicrobial sepsis following cecal ligation and puncture. Neutralizing Abs to IL-27(p28) improved survival rates, restricted cytokine release, and reduced bacterial burden in C57BL/6 mice during sepsis. Genetic disruption of IL-27 signaling enhanced the respiratory burst of macrophages. Experiments using splenectomized mice or treatment with clodronate liposomes suggested that macrophages in the spleen may be a significant source of IL-27(p28) during sepsis. In cultures of TLR4-activated macrophages, the frequency of F4/80(+)CD11b(+)IL-27(p28)(+) cells was reduced by the addition of IL-10. IL-10 antagonized both MyD88-dependent and TRIF-dependent release of IL-27(p28). Genetic deletion of STAT3 in Tie2-Cre/STAT3flox macrophages completely interrupted the inhibition of IL-27(p28) by IL-10 after TLR4 activation. In contrast, IL-10 remained fully active to suppress IL-27(p28) with deletion of SOCS3 in Tie2-Cre/SOCS3flox macrophages. Blockade of IL-10R by Ab or genetic deficiency of IL-10 resulted in 3-5-fold higher concentrations of IL-27(p28) in endotoxic shock and polymicrobial sepsis. Our studies identify IL-10 as a critical suppressing factor for IL-27(p28) production during infection-associated inflammation. These findings may be helpful for a beneficial manipulation of adverse IL-27(p28) release during sepsis.


Subject(s)
Interleukin-10/metabolism , Interleukins/metabolism , Macrophages/physiology , STAT3 Transcription Factor/metabolism , Sepsis/immunology , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Antibodies, Blocking/administration & dosage , Bacterial Load , Cecum/surgery , Cells, Cultured , Disease Models, Animal , Humans , Interleukin-10/genetics , Interleukins/immunology , Macrophages/drug effects , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Oxidative Stress/drug effects , Oxidative Stress/genetics , Receptors, Cytokine/genetics , Receptors, Interleukin , STAT3 Transcription Factor/genetics , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Toll-Like Receptor 4/immunology
19.
Mediators Inflamm ; 2016: 1340156, 2016.
Article in English | MEDLINE | ID: mdl-27382187

ABSTRACT

C5a is an inflammatory mediator generated by complement activation that positively regulates various arms of immune defense, including Toll-like receptor 4 (TLR4) signaling. The NOD-like receptor pyrin domain-containing protein 3 (NLRP3) inflammasome is activated by pathogen products and cellular/tissue damage products and is a major contributor of IL-1ß. In this study, we investigate whether C5a modulates lipopolysaccharide- (LPS-) induced NLRP3 inflammasome activation in myeloid cells. Appearance of plasma IL-1ß during endotoxemia was reduced in C5aR1(-/-) mice when compared to wild-type mice. In vitro, C5a significantly enhanced LPS-induced production of IL-1ß in bone marrow Ly6C-high inflammatory monocytes, accompanied by augmented intracellular pro-IL-1ß expression. This effect was abolished during p38 blockade by SB 203580 and in the absence of C5aR1. Conversely, C5a suppressed LPS-induced macrophage production of IL-1ß, which was accompanied by attenuated levels of pro-IL-1ß, NLRP3, and caspase-1 expression. C5a's suppressive effects were negated during phosphoinositide 3-kinase (PI3K) inhibition by wortmannin but were largely preserved in the absence of C5aR1. Thus, C5a bidirectionally amplifies TLR4-mediated NLRP3 inflammasome activation in monocytes while suppressing this pathway in macrophages. However, as C5aR1 deficiency attenuates the IL-1ß response to LPS challenge in vivo, our results suggest overall that C5a augments physiologic inflammasome responses.


Subject(s)
Inflammasomes/metabolism , Macrophages/metabolism , Monocytes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Receptor, Anaphylatoxin C5a/metabolism , Animals , Blotting, Western , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Complement C5a/metabolism , Complement C5a/pharmacology , Disease Models, Animal , Endotoxemia/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Inflammasomes/drug effects , Interleukin-1beta/metabolism , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Receptor, Anaphylatoxin C5a/deficiency , Receptor, Anaphylatoxin C5a/genetics , Reverse Transcriptase Polymerase Chain Reaction
20.
J Immunol ; 190(8): 4215-25, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23479227

ABSTRACT

During experimental sepsis, excessive generation of the anaphylatoxin C5a results in reduction of the C5a receptor (C5aR) on neutrophils. These events have been shown to result in impaired innate immunity. However, the regulation and fate of C5aR on neutrophils during sepsis are largely unknown. In contrast to 30 healthy volunteers, 60 patients in septic shock presented evidence of complement activation with significantly increased serum levels of C3a, C5a, and C5b-9. In the septic shock group, the corresponding decrease in complement hemolytic activity distinguished survivors from nonsurvivors. Neutrophils from patients in septic shock exhibited decreased C5aR expression, which inversely correlated with serum concentrations of C-reactive protein (CRP) and clinical outcome. In vitro exposure of normal neutrophils to native pentameric CRP led to a dose- and time-dependent loss of C5aR expression on neutrophils, whereas the monomeric form of CRP, as well as various other inflammatory mediators, failed to significantly alter C5aR levels on neutrophils. A circulating form of C5aR (cC5aR) was detected in serum by immunoblotting and a flow-based capture assay, suggestive of an intact C5aR molecule. Levels of cC5aR were significantly enhanced during septic shock, with serum levels directly correlating with lethality. The data suggest that septic shock in humans is associated with extensive complement activation, CRP-dependent loss of C5aR on neutrophils, and appearance of cC5aR in serum, which correlated with a poor outcome. Therefore, cC5aR may represent a new sepsis marker to be considered in tailoring individualized immune-modulating therapy.


Subject(s)
Neutrophils/immunology , Neutrophils/metabolism , Receptors, Complement/blood , Shock, Septic/blood , Shock, Septic/immunology , Adult , Aged , Animals , Female , Humans , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Receptor, Anaphylatoxin C5a , Receptors, Complement/antagonists & inhibitors , Shock, Septic/mortality , Survival
SELECTION OF CITATIONS
SEARCH DETAIL