Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
Add more filters

Publication year range
1.
Hepatology ; 78(1): 225-242, 2023 07 01.
Article in English | MEDLINE | ID: mdl-36862512

ABSTRACT

BACKGROUND AIMS: Prolonged systemic inflammation contributes to poor clinical outcomes in severe alcohol-associated hepatitis (AH) even after the cessation of alcohol use. However, mechanisms leading to this persistent inflammation remain to be understood. APPROACH RESULTS: We show that while chronic alcohol induces nucleotide-binding oligomerization domain-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome activation in the liver, alcohol binge results not only in NLRP3 inflammasome activation but also in increased circulating extracellular apoptosis-associated speck-like protein containing a caspase recruitment domain (ex-ASC) specks and hepatic ASC aggregates both in patients with AH and in mouse models of AH. These ex-ASC specks persist in circulation even after the cessation of alcohol use. Administration of alcohol-induced-ex-ASC specks in vivo in alcohol-naive mice results in sustained inflammation in the liver and circulation and causes liver damage. Consistent with the key role of ex-ASC specks in mediating liver injury and inflammation, alcohol binge failed to induce liver damage or IL-1ß release in ASC-deficient mice. Our data show that alcohol induces ex-ASC specks in liver macrophages and hepatocytes, and these ex-ASC specks can trigger IL-1ß release in alcohol-naive monocytes, a process that can be prevented by the NLRP3 inhibitor, MCC950. In vivo administration of MCC950 reduced hepatic and ex-ASC specks, caspase-1 activation, IL-1ß production, and steatohepatitis in a murine model of AH. CONCLUSIONS: Our study demonstrates the central role of NLRP3 and ASC in alcohol-induced liver inflammation and unravels the critical role of ex-ASC specks in the propagation of systemic and liver inflammation in AH. Our data also identify NLRP3 as a potential therapeutic target in AH.


Subject(s)
Hepatitis, Alcoholic , Hepatitis , Animals , Mice , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Hepatitis/etiology , Inflammation , Hepatitis, Alcoholic/etiology , Ethanol/adverse effects , Caspase 1/metabolism , Interleukin-1beta/metabolism , CARD Signaling Adaptor Proteins/metabolism
2.
Immunity ; 38(6): 1088-90, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23809158

ABSTRACT

Activation of inflammasome signaling can produce harmful inflammation. In this issue of Immunity, Yan et al. (2013) suggest that omega-3 fatty acids commonly found in marine oils can suppress activation of NLRP3 and NLRP1b inflammasomes.


Subject(s)
Carrier Proteins/metabolism , Diabetes Mellitus, Type 2/drug therapy , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Inflammasomes/metabolism , Inflammation/prevention & control , Macrophages/drug effects , Animals , NLR Family, Pyrin Domain-Containing 3 Protein
3.
Immunity ; 37(1): 96-107, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22840842

ABSTRACT

Yersinia pestis, the causative agent of plague, is able to suppress production of inflammatory cytokines IL-18 and IL-1ß, which are generated through caspase-1-activating nucleotide-binding domain and leucine-rich repeat (NLR)-containing inflammasomes. Here, we sought to elucidate the role of NLRs and IL-18 during plague. Lack of IL-18 signaling led to increased susceptibility to Y. pestis, producing tetra-acylated lipid A, and an attenuated strain producing a Y. pseudotuberculosis-like hexa-acylated lipid A. We found that the NLRP12 inflammasome was an important regulator controlling IL-18 and IL-1ß production after Y. pestis infection, and NLRP12-deficient mice were more susceptible to bacterial challenge. NLRP12 also directed interferon-γ production via induction of IL-18, but had minimal effect on signaling to the transcription factor NF-κB. These studies reveal a role for NLRP12 in host resistance against pathogens. Minimizing NLRP12 inflammasome activation may have been a central factor in evolution of the high virulence of Y. pestis.


Subject(s)
Inflammasomes/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Plague/immunology , Plague/metabolism , Yersinia pestis/immunology , Animals , Inflammasomes/immunology , Interferon-gamma/biosynthesis , Interleukin-18/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Plague/mortality , Signal Transduction
4.
Biochem Biophys Res Commun ; 523(4): 867-873, 2020 03 19.
Article in English | MEDLINE | ID: mdl-31955888

ABSTRACT

Inflammation is centrally involved in the development of cardiac hypertrophy and the processes of remodelling. The complement system and Toll-like receptor (TLR) family, two upstream arms of the innate immune system, have previously been reported to be involved in cardiac remodelling. However, the role of complement component 3 (C3), TLR co-receptor CD14 and the synergy between them have not been addressed during pressure overload-induced cardiac remodelling. Here, we examined angiotensin II-induced cardiac hypertrophy and remodelling for 7 days in male C57Bl/6 J mice deficient in C3, CD14, or both (C3CD14), and WT controls. Angiotensin II infusion induced a mild concentric hypertrophic phenotype in WT mice with increased left ventricle weight, wall thicknesses and reduced ventricular internal diameter, associated with increased cardiac fibrosis. However, there were no differences between WT mice and mice deficient for C3, CD14 or C3CD14, as systolic blood pressure, cardiac function and structure and levels of fibrosis were comparable between WT mice and the three other genotypes. C5a did not change in angiotensin II treated mice, whereas Mac2 levels were increased in angiotensin II treated mice, but did not differ between genotypes. The inflammatory IL-6 response was comparable between WT and C3 deficient mice, however, it was decreased in CD14 and C3CD14 deficient mice. We conclude that deficiency in C3, CD14 or C3CD14 had no effect on cardiac remodelling following angiotensin II-induced pressure overload. This suggests that C3 and CD14 are not involved in angiotensin II-induced adverse cardiac remodelling.


Subject(s)
Angiotensin II/pharmacology , Complement C3/metabolism , Lipopolysaccharide Receptors/metabolism , Toll-Like Receptors/metabolism , Ventricular Remodeling/drug effects , Animals , Biomarkers/blood , Blood Pressure/drug effects , Cardiomegaly/blood , Cardiomegaly/genetics , Fibrosis , Hypertrophy , Interleukin-6/genetics , Interleukin-6/metabolism , Mice , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Organ Size/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Systole/drug effects
5.
Alcohol Clin Exp Res ; 44(6): 1300-1311, 2020 06.
Article in English | MEDLINE | ID: mdl-32282939

ABSTRACT

BACKGROUND: Activation of NLRP3 in liver macrophages contributes to alcohol-associated liver disease (ALD). Molecular chaperone heat shock protein (HSP) 90 facilitates NLRP3 inflammasome activity during infections and inflammatory diseases. We previously reported that HSP90 is induced in ALD and regulates proinflammatory cytokines, tumor necrosis factor alpha, and IL-6. Whether HSP90 affects IL-1ß and IL-18 regulated by NLRP3 inflammasome in ALD is unknown. Here, we hypothesize that HSP90 modulated NLRP3 inflammasome activity and affects IL-1ß and IL-18 secretion in ALD. METHODS: The expression of HSP90AA1 and NLRP3 inflammasome genes was evaluated in human alcoholic livers and in mouse model of ALD. The importance of HSP90 on NLRP3 inflammasome activation in ALD was evaluated by administering HSP90 inhibitor, 17-dimethylaminoethylamino-17-demethoxygeldanamycin (17-DMAG) to mice subjected to ALD, and in vitro to bone marrow-derived macrophages (BMDM) stimulated with LPS and ATP. The effect of activation of HSF1/HSPA1A axis during HSP90 inhibition or direct activation during heat shock of BMDMs on NLRP3 activity and secretion of downstream cytokines was evaluated. RESULTS: We found positive correlation between induction of HSP90 and NLRP3 inflammasome genes in human alcoholic cirrhotic livers. Administration of 17-DMAG in mouse model of ALD significantly down-regulated NLRP3 inflammasome-mediated caspase-1 (CASP-1) activity and cytokine secretion, with reduction in ALD. 17-DMAG-mediated decrease in NLRP3 was restricted to liver macrophages. Using BMDMs, we show that inhibition of HSP90 prevented CASP-1 activity, and Gasdermin D (GSDMD) cleavage, important in release of active IL-1ß and IL-18. Interestingly, activation of the heat shock factor 1 (HSF1)/HSPA1A axis, either during HSP90 inhibition or by heat shock, decreased NLRP3 inflammasome activity and reduced secretion of cytokines. CONCLUSION: Our studies indicate that inhibition of HSP90 and activation of HSF1/HSPA1A reduce IL-1ß and IL-18 via decrease in NLRP3/CASP-1 and GSDMD activity in ALD.


Subject(s)
Liver Diseases, Alcoholic/genetics , Adult , Aged , Animals , Benzoquinones/pharmacology , Caspase 1/drug effects , Caspase 1/metabolism , Cytokines/metabolism , Disease Models, Animal , Female , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/genetics , Heat Shock Transcription Factors/metabolism , Humans , In Vitro Techniques , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Lactams, Macrocyclic/pharmacology , Liver Cirrhosis, Alcoholic/genetics , Liver Cirrhosis, Alcoholic/metabolism , Liver Diseases, Alcoholic/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Middle Aged , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neoplasm Proteins , RNA, Messenger/metabolism , Young Adult
6.
Immunity ; 33(4): 583-96, 2010 Oct 29.
Article in English | MEDLINE | ID: mdl-20933442

ABSTRACT

Toll-like receptor 4 (TLR4) is indispensable for recognition of Gram-negative bacteria. We described a trafficking pathway for TLR4 from the endocytic recycling compartment (ERC) to E. coli phagosomes. We found a prominent colocalization between TLR4 and the small GTPase Rab11a in the ERC, and Rab11a was involved in the recruitment of TLR4 to phagosomes in a process requiring TLR4 signaling. Also, Toll-receptor-associated molecule (TRAM) and interferon regulatory factor-3 (IRF3) localized to E. coli phagosomes and internalization of E. coli was required for a robust interferon-ß induction. Suppression of Rab11a reduced TLR4 in the ERC and on phagosomes leading to inhibition of the IRF3 signaling pathway induced by E. coli, whereas activation of the transcription factor NF-κB was unaffected. Moreover, Rab11a silencing reduced the amount of TRAM on phagosomes. Thus, Rab11a is an important regulator of TLR4 and TRAM transport to E. coli phagosomes thereby controlling IRF3 activation from this compartment.


Subject(s)
Phagosomes/metabolism , Toll-Like Receptor 4/physiology , rab GTP-Binding Proteins/physiology , Endocytosis , Escherichia coli/immunology , Humans , Interferon Regulatory Factor-3/metabolism , Interferon-beta/biosynthesis , Phagocytosis , Signal Transduction , Staphylococcus aureus/immunology
7.
Am J Physiol Endocrinol Metab ; 315(5): E912-E923, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30016155

ABSTRACT

Inflammasomes are multiprotein inflammatory platforms that induce caspase-1 activation and subsequently interleukin (IL)-1ß and IL-18 processing. The NLRP3 inflammasome is activated by different forms of oxidative stress, and, based on the central role of IL-1ß in the destruction of pancreatic islets, it could be related to the development of diabetes. We therefore investigated responses in wild-type C57Bl/6 (WT) mice, NLRP3-/- mice, and mice deficient in apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC) after exposing islets to short-term hypoxia or alloxan-induced islet damage. NLRP3-deficient islets compared with WT islets had preserved function ex vivo and were protected against hypoxia-induced cell death. Furthermore, NLRP3 and ASC-deficient mice were protected against oxidative stress-induced diabetes caused by repetitive low-dose alloxan administration, and this was associated with reduced ß-cell death and reduced macrophage infiltration. This suggests that the beneficial effect of NLRP3 inflammasome deficiency on oxidative stress-mediated ß-cell damage could involve reduced macrophage infiltration and activation. To support the role of macrophage activation in alloxan-induced diabetes, we injected WT mice with liposomal clodronate, which causes macrophage depletion before induction of a diabetic phenotype by alloxan treatment, resulting in improved glucose homeostasis in WT mice. We show here that the NLRP3 inflammasome acts as a mediator of hypoxia and oxidative stress in insulin-producing cells, suggesting that inhibition of the NLRP3 inflammasome could have beneficial effects on ß-cell preservation.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Inflammasomes/metabolism , Islets of Langerhans/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidative Stress/physiology , Animals , Apoptosis/physiology , Insulin-Secreting Cells/metabolism , Interleukin-1beta/metabolism , Macrophage Activation/physiology , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics
8.
PLoS Pathog ; 12(12): e1006035, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27911947

ABSTRACT

Type III secretion systems (T3SS) are central virulence factors for many pathogenic Gram-negative bacteria, and secreted T3SS effectors can block key aspects of host cell signaling. To counter this, innate immune responses can also sense some T3SS components to initiate anti-bacterial mechanisms. The Yersinia pestis T3SS is particularly effective and sophisticated in manipulating the production of pro-inflammatory cytokines IL-1ß and IL-18, which are typically processed into their mature forms by active caspase-1 following inflammasome formation. Some effectors, like Y. pestis YopM, may block inflammasome activation. Here we show that YopM prevents Y. pestis induced activation of the Pyrin inflammasome induced by the RhoA-inhibiting effector YopE, which is a GTPase activating protein. YopM blocks YopE-induced Pyrin-mediated caspase-1 dependent IL-1ß/IL-18 production and cell death. We also detected YopM in a complex with Pyrin and kinases RSK1 and PKN1, putative negative regulators of Pyrin. In contrast to wild-type mice, Pyrin deficient mice were also highly susceptible to an attenuated Y. pestis strain lacking YopM, emphasizing the importance of inhibition of Pyrin in vivo. A complex interplay between the Y. pestis T3SS and IL-1ß/IL-18 production is evident, involving at least four inflammasome pathways. The secreted effector YopJ triggers caspase-8- dependent IL-1ß activation, even when YopM is present. Additionally, the presence of the T3SS needle/translocon activates NLRP3 and NLRC4-dependent IL-1ß generation, which is blocked by YopK, but not by YopM. Taken together, the data suggest YopM specificity for obstructing the Pyrin pathway, as the effector does not appear to block Y. pestis-induced NLRP3, NLRC4 or caspase-8 dependent caspase-1 processing. Thus, we identify Y. pestis YopM as a microbial inhibitor of the Pyrin inflammasome. The fact that so many of the Y. pestis T3SS components are participating in regulation of IL-1ß/IL-18 release suggests that these effects are essential for maximal control of innate immunity during plague.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Inflammasomes/immunology , Plague/immunology , Pyrin/immunology , Animals , Disease Models, Animal , Mice , Mice, Knockout , Yersinia pestis/immunology
9.
J Biol Chem ; 291(19): 9894-905, 2016 May 06.
Article in English | MEDLINE | ID: mdl-26884330

ABSTRACT

Innate immunity plays a central role in resolving infections by pathogens. Host survival during plague, caused by the Gram-negative bacterium Yersinia pestis, is favored by a robust early innate immune response initiated by IL-1ß and IL-18. These cytokines are produced by a two-step mechanism involving NF-κB-mediated pro-cytokine production and inflammasome-driven maturation into bioactive inflammatory mediators. Because of the anti-microbial effects induced by IL-1ß/IL-18, it may be desirable for pathogens to manipulate their production. Y. pestis type III secretion system effectors YopJ and YopM can interfere with different parts of this process. Both effectors have been reported to influence inflammasome caspase-1 activity; YopJ promotes caspase-8-dependent cell death and caspase-1 cleavage, whereas YopM inhibits caspase-1 activity via an incompletely understood mechanism. However, neither effector appears essential for full virulence in vivo Here we report that the sum of influences by YopJ and YopM on IL-1ß/IL-18 release is suppressive. In the absence of YopM, YopJ minimally affects caspase-1 cleavage but suppresses IL-1ß, IL-18, and other cytokines and chemokines. Importantly, we find that Y. pestis containing combined deletions of YopJ and YopM induces elevated levels of IL-1ß/IL-18 in vitro and in vivo and is significantly attenuated in a mouse model of bubonic plague. The reduced virulence of the YopJ-YopM mutant is dependent on the presence of IL-1ß, IL-18, and caspase-1. Thus, we conclude that Y. pestis YopJ and YopM can both exert a tight control of host IL-1ß/IL-18 production to benefit the bacteria, resulting in a redundant impact on virulence.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/metabolism , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Virulence/immunology , Yersinia Infections/immunology , Yersinia pestis/pathogenicity , Animals , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/immunology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Cells, Cultured , Immunity, Innate/immunology , Inflammasomes/genetics , Inflammasomes/immunology , Inflammasomes/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice , Real-Time Polymerase Chain Reaction , Yersinia Infections/microbiology
10.
J Biol Chem ; 291(3): 1123-36, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26555265

ABSTRACT

Many immunostimulants act as vaccine adjuvants via activation of the innate immune system, although in many cases it is unclear which specific molecules contribute to the stimulatory activity. QS-21 is a defined, highly purified, and soluble saponin adjuvant currently used in licensed and exploratory vaccines, including vaccines against malaria, cancer, and HIV-1. However, little is known about the mechanisms of cellular activation induced by QS-21. We observed QS-21 to elicit caspase-1-dependent IL-1ß and IL-18 release in antigen-presenting cells such as macrophages and dendritic cells when co-stimulated with the TLR4-agonist adjuvant monophosphoryl lipid A. Furthermore, our data suggest that the ASC-NLRP3 inflammasome is responsible for QS-21-induced IL-1ß/IL-18 release. At higher concentrations, QS-21 induced macrophage and dendritic cell death in a caspase-1-, ASC-, and NLRP3-independent manner, whereas the presence of cholesterol rescued cell viability. A nanoparticulate adjuvant that contains QS-21 as part of a heterogeneous mixture of saponins also induced IL-1ß in an NLRP3-dependent manner. Interestingly, despite the role NLRP3 plays for cellular activation in vitro, NLRP3-deficient mice immunized with HIV-1 gp120 and QS-21 showed significantly higher levels of Th1 and Th2 antigen-specific T cell responses and increased IgG1 and IgG2c compared with wild type controls. Thus, we have identified QS-21 as a nonparticulate single molecular saponin that activates the NLRP3 inflammasome, but this signaling pathway may contribute to decreased antigen-specific responses in vivo.


Subject(s)
Adjuvants, Immunologic/pharmacology , Carrier Proteins/metabolism , Dendritic Cells/drug effects , Immunity, Innate/drug effects , Inflammasomes/drug effects , Macrophages/drug effects , Saponins/pharmacology , AIDS Vaccines/agonists , AIDS Vaccines/immunology , Adjuvants, Immunologic/analysis , Adjuvants, Immunologic/chemistry , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Carrier Proteins/genetics , Cell Survival/drug effects , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/immunology , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , HIV Envelope Protein gp120/agonists , HIV Envelope Protein gp120/immunology , Immunoglobulin G/analysis , Immunoglobulin G/biosynthesis , Inflammasomes/immunology , Inflammasomes/metabolism , Lipid A/agonists , Lipid A/analogs & derivatives , Lipid A/pharmacology , Macrophages/cytology , Macrophages/immunology , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Saponins/analysis , Saponins/chemistry , Solubility , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/metabolism
11.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(2): 234-245, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27845246

ABSTRACT

Palmitate triggers inflammatory responses in several cell types, but its effects on cardiac fibroblasts are at present unknown. The aims of the study were to (1) assess the potential of palmitate to promote inflammatory signaling in cardiac fibroblasts through TLR4 and the NLRP3 inflammasome and (2) characterize the cellular phenotype of cardiac fibroblasts exposed to palmitate. We examined whether palmitate induces inflammatory responses in cardiac fibroblasts from WT, NLRP3-/- and ASC-/-mice (C57BL/6 background). Exposure to palmitate caused production of TNF, IL-6 and CXCL2 via TLR4 activation. NLRP3 inflammasomes are activated in a two-step manner. Whereas palmitate did not prime the NLRP3 inflammasome, it induced activation in LPS-primed cardiac fibroblasts as indicated by IL-1ß, IL-18 production and NLRP3-ASC co-localization. Palmitate-induced NLRP3 inflammasome activation in LPS-primed cardiac fibroblasts was associated with reduced AMPK activity, mitochondrial reactive oxygen species production and mitochondrial dysfunction. The cardiac fibroblast phenotype caused by palmitate, in an LPS and NLRP3 independent manner, was characterized by decreased cellular proliferation, contractility, collagen and MMP-2 expression, as well as increased senescence-associated ß-galactosidase activity, and consistent with a state of cellular senescence. This study establishes that in vitro palmitate exposure of cardiac fibroblasts provides inflammatory responses via TLR4 and NLRP3 inflammasome activation. Palmitate also modulates cardiac fibroblast functionality, in a NLRP3 independent manner, resulting in a phenotype related to cellular senescence. These effects of palmitate could be of importance for myocardial dysfunction in obese and diabetic patients.


Subject(s)
Cellular Senescence/drug effects , Fibroblasts/drug effects , Heart/drug effects , Inflammation/chemically induced , Palmitates/pharmacology , Animals , Apoptosis Regulatory Proteins/metabolism , Chemokine CXCL2/metabolism , Fibroblasts/metabolism , Inflammasomes/metabolism , Inflammation/metabolism , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , beta-Galactosidase/metabolism
12.
Proc Natl Acad Sci U S A ; 111(20): 7391-6, 2014 May 20.
Article in English | MEDLINE | ID: mdl-24799678

ABSTRACT

A number of pathogens cause host cell death upon infection, and Yersinia pestis, infamous for its role in large pandemics such as the "Black Death" in medieval Europe, induces considerable cytotoxicity. The rapid killing of macrophages induced by Y. pestis, dependent upon type III secretion system effector Yersinia outer protein J (YopJ), is minimally affected by the absence of caspase-1, caspase-11, Fas ligand, and TNF. Caspase-8 is known to mediate apoptotic death in response to infection with several viruses and to regulate programmed necrosis (necroptosis), but its role in bacterially induced cell death is poorly understood. Here we provide genetic evidence for a receptor-interacting protein (RIP) kinase-caspase-8-dependent macrophage apoptotic death pathway after infection with Y. pestis, influenced by Toll-like receptor 4-TIR-domain-containing adapter-inducing interferon-ß (TLR4-TRIF). Interestingly, macrophages lacking either RIP1, or caspase-8 and RIP3, also had reduced infection-induced production of IL-1ß, IL-18, TNF, and IL-6; impaired activation of the transcription factor NF-κB; and greatly compromised caspase-1 processing. Cleavage of the proform of caspase-1 is associated with triggering inflammasome activity, which leads to the maturation of IL-1ß and IL-18, cytokines important to host responses against Y. pestis and many other infectious agents. Our results identify a RIP1-caspase-8/RIP3-dependent caspase-1 activation pathway after Y. pestis challenge. Mice defective in caspase-8 and RIP3 were also highly susceptible to infection and displayed reduced proinflammatory cytokines and myeloid cell death. We propose that caspase-8 and the RIP kinases are key regulators of macrophage cell death, NF-κB and inflammasome activation, and host resistance after Y. pestis infection.


Subject(s)
Caspase 8/metabolism , Cell Death , Immunity, Innate , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis , Bacterial Proteins/genetics , Bone Marrow Cells/cytology , Cytokines/metabolism , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Yersinia Infections/microbiology , Yersinia pestis/genetics
13.
J Biol Chem ; 290(6): 3209-22, 2015 Feb 06.
Article in English | MEDLINE | ID: mdl-25505250

ABSTRACT

Toll-like receptors (TLRs) are involved in sensing invading microbes by host innate immunity. TLR2 recognizes bacterial lipoproteins/lipopeptides, and lipopolysaccharide activates TLR4. TLR2 and TLR4 signal via the Toll/interleukin-1 receptor adaptors MyD88 and MAL, leading to NF-κB activation. TLR4 also utilizes the adaptors TRAM and TRIF, resulting in activation of interferon regulatory factor (IRF) 3. Here, we report a new role for TRAM and TRIF in TLR2 regulation and signaling. Interestingly, we observed that TLR2-mediated induction of the chemokine Ccl5 was impaired in TRAM or TRIF deficient macrophages. Inhibition of endocytosis reduced Ccl5 release, and the data also suggested that TRAM and TLR2 co-localize in early endosomes, supporting the hypothesis that signaling may occur from an intracellular compartment. Ccl5 release following lipoprotein challenge additionally involved the kinase Tbk-1 and Irf3, as well as MyD88 and Irf1. Induction of Interferon-ß and Ccl4 by lipoproteins was also partially impaired in cells lacking TRIF cells. Our results show a novel function of TRAM and TRIF in TLR2-mediated signal transduction, and the findings broaden our understanding of how Toll/interleukin-1 receptor adaptor proteins may participate in signaling downstream from TLR2.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Receptors, Interleukin/metabolism , Signal Transduction , Toll-Like Receptor 2/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Animals , Cells, Cultured , Chemokine CCL4/genetics , Chemokine CCL4/metabolism , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Endocytosis , Endosomes/metabolism , HEK293 Cells , Humans , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon-beta/genetics , Interferon-beta/metabolism , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/metabolism , Mice , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptors, Interleukin/genetics , Toll-Like Receptor 2/agonists
14.
Nature ; 464(7293): 1357-61, 2010 Apr 29.
Article in English | MEDLINE | ID: mdl-20428172

ABSTRACT

The inflammatory nature of atherosclerosis is well established but the agent(s) that incite inflammation in the artery wall remain largely unknown. Germ-free animals are susceptible to atherosclerosis, suggesting that endogenous substances initiate the inflammation. Mature atherosclerotic lesions contain macroscopic deposits of cholesterol crystals in the necrotic core, but their appearance late in atherogenesis had been thought to disqualify them as primary inflammatory stimuli. However, using a new microscopic technique, we revealed that minute cholesterol crystals are present in early diet-induced atherosclerotic lesions and that their appearance in mice coincides with the first appearance of inflammatory cells. Other crystalline substances can induce inflammation by stimulating the caspase-1-activating NLRP3 (NALP3 or cryopyrin) inflammasome, which results in cleavage and secretion of interleukin (IL)-1 family cytokines. Here we show that cholesterol crystals activate the NLRP3 inflammasome in phagocytes in vitro in a process that involves phagolysosomal damage. Similarly, when injected intraperitoneally, cholesterol crystals induce acute inflammation, which is impaired in mice deficient in components of the NLRP3 inflammasome, cathepsin B, cathepsin L or IL-1 molecules. Moreover, when mice deficient in low-density lipoprotein receptor (LDLR) were bone-marrow transplanted with NLRP3-deficient, ASC (also known as PYCARD)-deficient or IL-1alpha/beta-deficient bone marrow and fed on a high-cholesterol diet, they had markedly decreased early atherosclerosis and inflammasome-dependent IL-18 levels. Minimally modified LDL can lead to cholesterol crystallization concomitant with NLRP3 inflammasome priming and activation in macrophages. Although there is the possibility that oxidized LDL activates the NLRP3 inflammasome in vivo, our results demonstrate that crystalline cholesterol acts as an endogenous danger signal and its deposition in arteries or elsewhere is an early cause rather than a late consequence of inflammation. These findings provide new insights into the pathogenesis of atherosclerosis and indicate new potential molecular targets for the therapy of this disease.


Subject(s)
Atherosclerosis/metabolism , Atherosclerosis/pathology , Carrier Proteins/metabolism , Cholesterol/chemistry , Cholesterol/metabolism , Animals , Apoptosis Regulatory Proteins , Atherosclerosis/chemically induced , Bone Marrow Transplantation , CARD Signaling Adaptor Proteins , Carrier Proteins/genetics , Cathepsin B/metabolism , Cathepsin L/metabolism , Cholesterol/pharmacology , Crystallization , Cytoskeletal Proteins/deficiency , Diet, Atherogenic , Female , Humans , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Interleukin-1/deficiency , Interleukin-18/metabolism , Lysosomes/drug effects , Lysosomes/pathology , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Peritoneal Cavity/pathology , Phagocytes/drug effects , Phagocytes/pathology , Phagocytes/physiology , Receptors, LDL/deficiency , Time Factors
15.
J Biol Chem ; 289(20): 13701-5, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24692555

ABSTRACT

The inflammatory cytokine IL-1ß 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, ß-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-1ß production.


Subject(s)
Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Hemolysin Proteins/metabolism , Inflammasomes/metabolism , Interleukin-1beta/biosynthesis , Macrophages/metabolism , RNA, Bacterial/metabolism , Streptococcus agalactiae/physiology , Animals , Humans , Interleukin-1beta/metabolism , Lysosomes/metabolism , Lysosomes/microbiology , Macrophages/cytology , Macrophages/microbiology , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Phagosomes/metabolism , Phagosomes/microbiology , Streptococcus agalactiae/metabolism
16.
Am J Physiol Lung Cell Mol Physiol ; 309(4): L378-87, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26071556

ABSTRACT

Pulmonary hypertension is a serious condition that can lead to premature death. The mechanisms involved are incompletely understood although a role for the immune system has been suggested. Inflammasomes are part of the innate immune system and consist of the effector caspase-1 and a receptor, where nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 3 (NLRP3) is the best characterized and interacts with the adaptor protein apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC). To investigate whether ASC and NLRP3 inflammasome components are involved in hypoxia-induced pulmonary hypertension, we utilized mice deficient in ASC and NLRP3. Active caspase-1, IL-18, and IL-1ß, which are regulated by inflammasomes, were measured in lung homogenates in wild-type (WT), ASC(-/-), and NLRP3(-/-) mice, and phenotypical changes related to pulmonary hypertension and right ventricular remodeling were characterized after hypoxic exposure. Right ventricular systolic pressure (RVSP) of ASC(-/-) mice was significantly lower than in WT exposed to hypoxia (40.8 ± 1.5 mmHg vs. 55.8 ± 2.4 mmHg, P < 0.001), indicating a substantially reduced pulmonary hypertension in mice lacking ASC. Magnetic resonance imaging further supported these findings by demonstrating reduced right ventricular remodeling. RVSP of NLRP3(-/-) mice exposed to hypoxia was not significantly altered compared with WT hypoxia. Whereas hypoxia increased protein levels of caspase-1, IL-18, and IL-1ß in WT and NLRP3(-/-) mice, this response was absent in ASC(-/-) mice. Moreover, ASC(-/-) mice displayed reduced muscularization and collagen deposition around arteries. In conclusion, hypoxia-induced elevated right ventricular pressure and remodeling were attenuated in mice lacking the inflammasome adaptor protein ASC, suggesting that inflammasomes play an important role in the pathogenesis of pulmonary hypertension.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Hypertension, Pulmonary/metabolism , Inflammasomes/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Arteries/pathology , CARD Signaling Adaptor Proteins , Cell Hypoxia , Collagen/metabolism , Gene Expression , Hypertrophy, Right Ventricular/metabolism , Interleukin-18/blood , Leukocytes/immunology , Lung/immunology , Lung/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Ventricular Remodeling
17.
mBio ; 14(2): e0011023, 2023 04 25.
Article in English | MEDLINE | ID: mdl-36852999

ABSTRACT

Influenza A virus (IAV) triggers multiple programmed cell death pathways, including MLKL-dependent necroptosis, caspase-8-dependent apoptosis, and caspase-1-dependent pyroptosis in myeloid cells. All three pathways share common upstream regulators, namely, ZBP1 and RIPK3. Yet, the molecular mechanism underlying IAV-induced inflammasome activation remains unclear. Here, we demonstrate that MLKL promotes inflammasome activation and IL-1ß processing in IAV-infected macrophages. MLKL drives NLRP3 inflammasome activation through potassium efflux. In the absence of the MLKL-inflammasome axis, caspase-8 coordinates the maturation and secretion of IL-1ß. MLKL alone is dispensable for host inflammatory responses to IAV in vivo. Taken together, MLKL and caspase-8 serve as redundant mechanisms by which to drive an inflammatory form of cell death in response to an IAV infection. IMPORTANCE Influenza A virus (IAV) induces multiple types of cell death, which play important roles in the host antiviral responses but can also cause unwanted inflammation and tissue damage. In this study, we dissect the interplay of cell death pathways and demonstrate that macrophages utilize redundant mechanisms to drive an inflammatory form of cell death upon IAV infection. MLKL, the executor of necroptosis, promotes inflammasome activation and pyroptotic cell death. When the MLKL-inflammasome axis is inhibited, cells divert to caspase-8-dependent inflammatory cell death. Our findings advance the current understanding of the innate immune response to IAV infection as well as broader contexts involving multifaceted cell death.


Subject(s)
Influenza A virus , Influenza, Human , Humans , Inflammasomes/metabolism , Caspase 8/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Cell Death , Apoptosis , Caspase 1/metabolism , Protein Kinases/metabolism
18.
Cell Rep Med ; 4(7): 101098, 2023 07 18.
Article in English | MEDLINE | ID: mdl-37390829

ABSTRACT

During differentiation, neutrophils undergo a spontaneous pro-inflammatory program that is hypothesized here to be under caspase-8 control. In mice, intraperitoneal administration of the caspase-8 inhibitor z-IETD-fmk is sufficient to unleash the production of pro-inflammatory cytokines and neutrophil influx in the absence of cell death. These effects are due to selective inhibition of caspase-8 and require tonic interferon-ß (IFN-ß) production and RIPK3 but not MLKL, the essential downstream executioner of necroptotic cell death. In vitro, stimulation with z-IETD-fmk is sufficient to induce significant cytokine production in murine neutrophils but not in macrophages. Therapeutic administration of z-IETD-fmk improves clinical outcome in models of lethal bacterial peritonitis and pneumonia by augmenting cytokine release, neutrophil influx, and bacterial clearance. Moreover, the inhibitor protects mice against high-dose endotoxin shock. Collectively, our data unveil a RIPK3- and IFN-ß-dependent pathway that is constitutively activated in neutrophils and can be harnessed therapeutically using caspase-8 inhibition.


Subject(s)
Apoptosis , Bacterial Infections , Animals , Mice , Bacterial Infections/drug therapy , Caspase 8/metabolism , Caspase 8/pharmacology , Cytokines/metabolism , Neutrophil Activation
19.
Life Sci Alliance ; 6(12)2023 12.
Article in English | MEDLINE | ID: mdl-37788908

ABSTRACT

Inflammation plays a crucial role in the development and progression of many diseases, and is often caused by dysregulation of signalling from pattern recognition receptors, such as TLRs. Inhibition of key protein-protein interactions is an attractive target for treating inflammation. Recently, we demonstrated that the signalling lymphocyte activation molecule family 1 (SLAMF1) positively regulates signalling downstream of TLR4 and identified the interaction interface between SLAMF1 and the TLR4 adaptor protein TRIF-related adapter molecule (TRAM). Based on these findings, we developed a SLAMF1-derived peptide, P7, which is linked to a cell-penetrating peptide for intracellular delivery. We found that P7 peptide inhibits the expression and secretion of IFNß and pro-inflammatory cytokines (TNF, IL-1ß, IL-6) induced by TLR4, and prevents death in mice subjected to LPS shock. The mechanism of action of P7 peptide is based on interference with several intracellular protein-protein interactions, including TRAM-SLAMF1, TRAM-Rab11FIP2, and TIRAP-MyD88 interactions. Overall, P7 peptide has a unique mode of action and demonstrates high efficacy in inhibiting TLR4-mediated signalling in vitro and in vivo.


Subject(s)
Signal Transduction , Toll-Like Receptor 4 , Animals , Mice , Signaling Lymphocytic Activation Molecule Family/metabolism , Peptides/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Inflammation
SELECTION OF CITATIONS
SEARCH DETAIL