Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Helicobacter ; 23(1)2018 Feb.
Article in English | MEDLINE | ID: mdl-29110387

ABSTRACT

BACKGROUND: Non-Helicobacter pylori helicobacters (NHPHs) besides H. pylori infect human stomachs and cause chronic gastritis and mucosa-associated lymphoid tissue lymphoma. Cholesteryl-α-glucosides have been identified as unique glycolipids present in H. pylori and some Helicobacter species. Cholesterol-α-glucosyltransferase (αCgT), a key enzyme for the biosynthesis of cholesteryl-α-glucosides, plays crucial roles in the pathogenicity of H. pylori. Therefore, it is important to examine αCgTs of NHPHs. MATERIALS AND METHODS: Six gastric NHPHs were isolated from Japanese patients and maintained in mouse stomachs. The αCgT genes were amplified by PCR and inverse PCR. We retrieved the αCgT genes of other Helicobacter species by BLAST searches in GenBank. RESULTS: αCgT genes were present in most Helicobacter species and in all Japanese isolates examined. However, we could find no candidate gene for αCgT in the whole genome of Helicobacter cinaedi and several enterohepatic species. Phylogenic analysis demonstrated that the αCgT genes of all Japanese isolates show high similarities to that of a zoonotic group of gastric NHPHs including Helicobacter suis, Helicobacter heilmannii, and Helicobacter ailurogastricus. Of 6 Japanese isolates, the αCgT genes of 4 isolates were identical to that of H. suis, and that of another 2 isolates were similar to that of H. heilmannii and H. ailurogastricus. CONCLUSIONS: All gastric NHPHs examined showed presence of αCgT genes, indicating that αCgT may be beneficial for these helicobacters to infect human and possibly animal stomachs. Our study indicated that NHPHs could be classified into 2 groups, NHPHs with αCgT genes and NHPHs without αCgT genes.


Subject(s)
Glucosyltransferases/genetics , Helicobacter Infections/microbiology , Helicobacter/enzymology , Helicobacter/genetics , Lymphoma, B-Cell, Marginal Zone/microbiology , Animals , Female , Gastritis/microbiology , Gastritis/pathology , Genome, Bacterial/genetics , Helicobacter/classification , Helicobacter Infections/pathology , Humans , Japan , Lymphoma, B-Cell, Marginal Zone/pathology , Mice , Molecular Sequence Data , Phylogeny , Polymerase Chain Reaction , Sequence Alignment , Sequence Homology, Amino Acid , Virulence/genetics
2.
Histochem Cell Biol ; 148(4): 463-471, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28534071

ABSTRACT

Helicobacter suis (H. suis), formerly called Helicobacter heilmannii type 1 (H. heilmannii), is a gram-negative bacterium of the Helicobacter species. This pathogen infects the stomach of humans and animals such as dogs, cats, pigs, and rodents, the latter giving rise to zoonotic infection. Here, we generated a H. suis-specific antibody useful for immunohistochemistry with formalin-fixed, paraffin-embedded tissue sections. To do so, we began by cloning the gene encoding H. suis cholesterol α-glucosyltransferase (αCgT). αCgT is the key enzyme responsible for biosynthesis of cholesteryl α-D-glucopyranoside (CGL), a major cell wall component of Helicobacter species including H. suis. The deduced amino acid sequence of H. suis αCgT had 56% identity with the corresponding Helicobacter pylori (H. pylori). We then developed a polyclonal antibody (anti-Hh-I205R) by immunizing rabbits with a 205 amino acid H. suis αCgT fragment. Immunohistochemistry with the anti-Hh-I205R antibody could differentiate H. suis from H. pylori in gastric mucosa sections derived from mice infected with either pathogen. We then probed formalin-fixed, paraffin-embedded sections of human gastric mucosa positive for H. suis infection with the anti-Hh-I205R antibody and detected positive staining. These results indicate that anti-Hh-I205R antibody is specific for H. suis αCgT and useful to detect H. suis in gastric specimens routinely analyzed in pathological examinations.


Subject(s)
Antibodies/metabolism , Cholesterol/analysis , Gastric Mucosa/chemistry , Glucosyltransferases/analysis , Helicobacter heilmannii/enzymology , Animals , Cell Differentiation , Cell Wall/chemistry , Cell Wall/metabolism , Cholesterol/genetics , Cholesterol/metabolism , Cloning, Molecular , Formaldehyde , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Humans , Immunohistochemistry , Mice , Paraffin Embedding
3.
Arterioscler Thromb Vasc Biol ; 35(1): 127-36, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25378412

ABSTRACT

OBJECTIVE: Abdominal aortic aneurysm (AAA) is considered a chronic inflammatory disease; however, the molecular basis underlying the sterile inflammatory response involved in the process of AAA remains unclear. We previously showed that the inflammasome, which regulates the caspase-1-dependent interleukin-1ß production, mediates the sterile cardiovascular inflammatory responses. Therefore, we hypothesized that the inflammasome is a key mediator of initial inflammation in AAA formation. APPROACH AND RESULTS: Apoptosis-associated speck-like protein containing a caspase recruitment domain is highly expressed in adventitial macrophages in human and murine AAA tissues. Using an established mouse model of AAA induced by continuous infusion of angiotensin II in Apoe(-/-) mice, NLR family pyrin domain containing 3 (NLRP3), apoptosis-associated speck-like protein containing a caspase recruitment domain, and caspase-1 deficiency in Apoe(-/-) mice were shown to decrease the incidence, maximal diameter, and severity of AAA along with adventitial fibrosis and inflammatory responses significantly, such as inflammatory cell infiltration and cytokine expression in the vessel wall. NLRP3, apoptosis-associated speck-like protein containing a caspase recruitment domain, and caspase-1 deficiency in Apoe(-/-) mice also reduced elastic lamina degradation and metalloproteinase activation in the early phase of AAA formation. Furthermore, angiotensin II stimulated generation of mitochondria-derived reactive oxygen species in the adventitial macrophages, and this mitochondria-derived reactive oxygen species generation was inhibited by NLRP3, apoptosis-associated speck-like protein containing a caspase recruitment domain, and caspase-1 deficiency. In vitro experiments revealed that angiotensin II stimulated the NLRP3 inflammasome activation and subsequent interleukin-1ß release in macrophages, and this activation was mediated through an angiotensin type I receptor/mitochondria-derived reactive oxygen species-dependent pathway. CONCLUSIONS: Our results demonstrate the importance of the NLRP3 inflammasome in the initial inflammatory responses in AAA formation, indicating its potential as a novel therapeutic target for preventing AAA progression.


Subject(s)
Angiotensin II , Aorta, Abdominal/metabolism , Aortic Aneurysm, Abdominal/metabolism , Inflammasomes/metabolism , Macrophage Activation , Macrophages/metabolism , Mitochondria/metabolism , Oxidative Stress , Aged , Animals , Aorta, Abdominal/immunology , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/immunology , Aortic Aneurysm, Abdominal/pathology , Aortic Aneurysm, Abdominal/prevention & control , Apolipoproteins E , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , CARD Signaling Adaptor Proteins , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspase 1/deficiency , Caspase 1/genetics , Cells, Cultured , Disease Models, Animal , Female , Fibrosis , Humans , Inflammasomes/genetics , Inflammasomes/immunology , Inflammation Mediators/metabolism , Interleukin-1beta/metabolism , Macrophages/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/immunology , NLR Family, Pyrin Domain-Containing 3 Protein , Reactive Oxygen Species/metabolism , Signal Transduction , Time Factors
4.
J Immunol ; 192(9): 4342-51, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24696236

ABSTRACT

Inflammation plays a key role in the pathophysiology of hepatic ischemia-reperfusion (I/R) injury. However, the mechanism by which hepatic I/R induces inflammatory responses remains unclear. Recent evidence indicates that a sterile inflammatory response triggered by I/R is mediated through a multiple-protein complex called the inflammasome. Therefore, we investigated the role of the inflammasome in hepatic I/R injury and found that hepatic I/R stimuli upregulated the inflammasome-component molecule, nucleotide-binding oligomerization domain-like receptor (NLR) family pyrin domain-containing 3 (NLRP3), but not apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC). NLRP3(-/-) mice, but not ASC(-/-) and caspase-1(-/-) mice, had significantly less liver injury after hepatic I/R. NLRP3(-/-) mice showed reduced inflammatory responses, reactive oxygen species production, and apoptosis in I/R liver. Notably, infiltration of neutrophils, but not macrophages, was markedly inhibited in the I/R liver of NLRP3(-/-) mice. Bone marrow transplantation experiments showed that NLRP3 not only in bone marrow-derived cells, but also in non-bone marrow-derived cells contributed to liver injury after I/R. In vitro experiments revealed that keratinocyte-derived chemokine-induced activation of heterotrimeric G proteins was markedly diminished. Furthermore, NLRP3(-/-) neutrophils decreased keratinocyte-derived chemokine-induced concentrations of intracellular calcium elevation, Rac activation, and actin assembly formation, thereby resulting in impaired migration activity. Taken together, NLRP3 regulates chemokine-mediated functions and recruitment of neutrophils, and thereby contributes to hepatic I/R injury independently of inflammasomes. These findings identify a novel role of NLRP3 in the pathophysiology of hepatic I/R injury.


Subject(s)
Carrier Proteins/immunology , Liver/immunology , Neutrophils/immunology , Reperfusion Injury/immunology , Animals , Apoptosis/immunology , Blotting, Western , Carrier Proteins/metabolism , Chemotaxis, Leukocyte , Flow Cytometry , Immunohistochemistry , Inflammasomes/immunology , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Neutrophils/metabolism , Real-Time Polymerase Chain Reaction , Reperfusion Injury/pathology , Reverse Transcriptase Polymerase Chain Reaction
5.
Am J Pathol ; 184(5): 1287-98, 2014 May.
Article in English | MEDLINE | ID: mdl-24606883

ABSTRACT

Inflammation plays a crucial role in the pathophysiological characteristics of chronic kidney disease; however, the inflammatory mechanisms underlying the chronic kidney disease process remain unclear. Recent evidence indicates that sterile inflammation triggered by tissue injury is mediated through a multiprotein complex called the inflammasome. Therefore, we investigated the role of the inflammasome in the development of chronic kidney disease using a murine unilateral ureteral obstruction (UUO) model. Inflammasome-related molecules were up-regulated in the kidney after UUO. Apoptosis-associated speck-like protein containing a caspase recruitment domain deficiency significantly reduced inflammatory responses, such as inflammatory cell infiltration and cytokine expression, and improved subsequent renal injury and fibrosis. Furthermore, apoptosis-associated speck-like protein containing a caspase recruitment domain was specifically up-regulated in collecting duct (CD) epithelial cells of the UUO-treated kidney. In vitro experiments showed that extracellular adenosine triphosphate (ATP) induced inflammasome activation in CD epithelial cells through P2X7-potassium efflux and reactive oxygen species-dependent pathways. These results demonstrate the molecular basis for the inflammatory response in the process of chronic kidney disease and suggest the CD inflammasome as a potential therapeutic target for preventing chronic kidney disease progression.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Inflammation/complications , Kidney Tubules, Collecting/pathology , Ureteral Obstruction/complications , Animals , Apoptosis , Apoptosis Regulatory Proteins/deficiency , CARD Signaling Adaptor Proteins , Cytokines/metabolism , Fibrosis , Inflammation/metabolism , Inflammation/pathology , Male , Mice, Inbred C57BL , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology
6.
J Immunol ; 191(8): 3995-9, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24043892

ABSTRACT

Activation of the NLRP3 inflammasome by diverse stimuli requires a priming signal from TLRs and an activation signal from purinergic receptors or pore-forming toxins. In this study, we demonstrate, through detailed analysis of NLRP3 activation in macrophages deficient in key downstream TLR signaling molecules, that MyD88 is required for an immediate early phase, whereas Toll/IL-1R domain-containing adapter inducing IFN-ß is required for a subsequent intermediate phase of posttranslational NLRP3 activation. Both IL-1R-associated kinase (IRAK) 1 and IRAK4 are critical for rapid activation of NLRP3 through the MyD88 pathway, but only IRAK1 is partially required in the Toll/IL-1R domain-containing adapter inducing IFN-ß pathway. IRAK1 and IRAK4 are also required for rapid activation of NLRP3 by Listeria monocytogenes, as deletion of IRAK1 or IRAK4 led to defective inflammasome activation. These findings define the pathways that lead to rapid NLRP3 activation and identify IRAK1 as a critical mediator of a transcription-independent,inflammasome-dependent early warning response to pathogenic infection.


Subject(s)
Carrier Proteins/metabolism , Inflammasomes , Interleukin-1 Receptor-Associated Kinases/immunology , Interleukin-1 Receptor-Associated Kinases/metabolism , Myeloid Differentiation Factor 88/metabolism , Toll-Like Receptors/immunology , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Enzyme Activation , Interferon-beta/metabolism , Listeria monocytogenes/immunology , Listeria monocytogenes/metabolism , Macrophages/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Receptors, Interleukin-1/metabolism , Signal Transduction , Toll-Like Receptors/metabolism
7.
Circulation ; 123(6): 594-604, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21282498

ABSTRACT

Background- Inflammation plays a key role in the pathophysiology of myocardial ischemia/reperfusion (I/R) injury; however, the mechanism by which myocardial I/R induces inflammation remains unclear. Recent evidence indicates that a sterile inflammatory response triggered by tissue damage is mediated through a multiple-protein complex called the inflammasome. Therefore, we hypothesized that the inflammasome is an initial sensor for danger signal(s) in myocardial I/R injury. Methods and Results- We demonstrate that inflammasome activation in cardiac fibroblasts, but not in cardiomyocytes, is crucially involved in the initial inflammatory response after myocardial I/R injury. We found that inflammasomes are formed by I/R and that its subsequent activation of inflammasomes leads to interleukin-1ß production, resulting in inflammatory responses such as inflammatory cell infiltration and cytokine expression in the heart. In mice deficient for apoptosis-associated speck-like adaptor protein and caspase-1, these inflammatory responses and subsequent injuries, including infarct development and myocardial fibrosis and dysfunction, were markedly diminished. Bone marrow transplantation experiments with apoptosis-associated speck-like adaptor protein-deficient mice revealed that inflammasome activation in bone marrow cells and myocardial resident cells such as cardiomyocytes or cardiac fibroblasts plays an important role in myocardial I/R injury. In vitro experiments revealed that hypoxia/reoxygenation stimulated inflammasome activation in cardiac fibroblasts, but not in cardiomyocytes, and that hypoxia/reoxygenation-induced activation was mediated through reactive oxygen species production and potassium efflux. Conclusions- Our results demonstrate the molecular basis for the initial inflammatory response after I/R and suggest that the inflammasome is a potential novel therapeutic target for preventing myocardial I/R injury.


Subject(s)
Fibroblasts/metabolism , Inflammasomes/metabolism , Myocardial Reperfusion Injury/metabolism , Animals , Caspase 1/metabolism , Cytokines/biosynthesis , Humans , Inflammation/metabolism , Interleukin-1beta/biosynthesis , Male , Mice , Mice, Inbred C57BL , Middle Aged , Myocardial Infarction/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Potassium/metabolism , Reactive Oxygen Species/metabolism
8.
Biochem Biophys Res Commun ; 425(2): 162-8, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22819845

ABSTRACT

OBJECTIVE: Recent investigations have suggested that the inflammasome plays a role in the development of vascular inflammation and atherosclerosis; however, its precise role remains controversial. We produced double-deficient mice for apolipoprotien E (Apoe) and caspase-1 (Casp1), a key component molecule of the inflammasome, and investigated the effect of caspase-1 deficiency on vascular inflammation and atherosclerosis. METHODS AND RESULTS: Atherosclerotic plaque areas in whole aortas and aortic root of Western diet (WD)-fed Apoe(-/-)Casp1(-/-) mice were significantly reduced compared to those in Apoe(-/-) mice. The amount of macrophages and vascular smooth muscle cells in the plaques was also reduced in Apoe(-/-)Casp1(-/-) mice. No significant differences in plasma lipid profiles and body weight change were observed between these mice. Expression of interleukin (IL)-1ß in the plaques as well as plasma levels of IL-1ß, IL-1α, IL-6, CCL2, and TNF-α, in Apoe(-/-)Casp1(-/-) mice were lower than those in Apoe(-/-) mice. In vitro experiments showed that calcium phosphate crystals induced caspase-1 activation and secretion of IL-1ß and IL-1α in macrophages. CONCLUSION: Our findings suggest that caspase-1 plays a critical role in vascular inflammation and atherosclerosis, and that modulation of caspase-1 could be a potential target for prevention and treatment of atherosclerosis.


Subject(s)
Atherosclerosis/enzymology , Caspase 1/physiology , Diet/adverse effects , Vasculitis/enzymology , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/etiology , Atherosclerosis/genetics , Caspase 1/genetics , Inflammasomes/metabolism , Macrophages/enzymology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Vasculitis/etiology , Vasculitis/genetics
9.
J Immunol ; 184(10): 5874-84, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20393137

ABSTRACT

Many members of the nucleotide-binding and oligomerization domain (NOD)- and leucine-rich-repeat-containing protein (NLR) family play important roles in pathogen recognition and inflammation. However, we previously reported that human PYNOD/NLRP10, an NLR-like protein consisting of a pyrin domain and a NOD, inhibits inflammatory signal mediated by caspase-1 and apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) in reconstitution experiments using HEK293 cells. In this study, we investigated the molecular mechanism of PYNOD's anti-inflammatory activity in vitro and its expression and function in mice. Human PYNOD inhibited the autoprocessing of caspase-1 and caspase-1-mediated IL-1beta processing and suppressed the aggregation of ASC, a hallmark of ASC activation. Interestingly, the NOD of human PYNOD was sufficient to inhibit caspase-1-mediated IL-1beta secretion, whereas its pyrin domain was sufficient to inhibit ASC-mediated NF-kappaB activation and apoptosis and to reduce ASC's ability to promote caspase-1-mediated IL-1beta production. Mouse PYNOD protein was detected in the skin, tongue, heart, colon, peritoneal macrophages, and several cell lines of hematopoietic and myocytic lineages. Mouse PYNOD colocalized with ASC aggregates in LPS + R837-stimulated macrophages; however, unlike human PYNOD, mouse PYNOD failed to inhibit ASC aggregation. Macrophages and neutrophils from PYNOD-transgenic mice exhibited reduced IL-1beta processing and secretion upon microbial infection, although mouse PYNOD failed to inhibit caspase-1 processing, which was inhibited by caspase-4 inhibitor z-LEED-fluoromethylketone. These results suggest that mouse PYNOD colocalizes with ASC and inhibits caspase-1-mediated IL-1beta processing without inhibiting caspase-4 (mouse caspase-11)-mediated caspase-1 processing. Furthermore, PYNOD-transgenic mice were resistant to lethal endotoxic shock. Thus, PYNOD is the first example of an NLR that possesses an anti-inflammatory function in vivo.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Carrier Proteins/physiology , Inflammation Mediators/physiology , Adaptor Proteins, Signal Transducing , Animals , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/genetics , Apoptosis/immunology , Apoptosis Regulatory Proteins , CARD Signaling Adaptor Proteins , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspase Inhibitors , Caspases/physiology , Caspases, Initiator , Cell Line , Cell Line, Tumor , Cells, Cultured , Cytokines/biosynthesis , Cytoskeletal Proteins/blood , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/physiology , Enzyme Precursors/antagonists & inhibitors , Humans , Inflammation Mediators/metabolism , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/blood , Lipopolysaccharides/toxicity , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Shock, Septic/immunology , Shock, Septic/prevention & control , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/blood
10.
J Immunol ; 184(9): 5287-97, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20348425

ABSTRACT

Vibrio vulnificus and Vibrio cholerae are Gram-negative pathogens that cause serious infectious disease in humans. The beta form of pro-IL-1 is thought to be involved in inflammatory responses and disease development during infection with these pathogens, but the mechanism of beta form of pro-IL-1 production remains poorly defined. In this study, we demonstrate that infection of mouse macrophages with two pathogenic Vibrio triggers the activation of caspase-1 via the NLRP3 inflammasome. Activation of the NLRP3 inflammasome was mediated by hemolysins and multifunctional repeat-in-toxins produced by the pathogenic bacteria. NLRP3 activation in response to V. vulnificus infection required NF-kappaB activation, which was mediated via TLR signaling. V. cholerae-induced NLRP3 activation also required NF-kappaB activation but was independent of TLR stimulation. Studies with purified V. cholerae hemolysin revealed that toxin-stimulated NLRP3 activation was induced by TLR and nucleotide-binding oligomerization domain 1/2 ligand-mediated NF-kappaB activation. Our results identify the NLRP3 inflammasome as a sensor of Vibrio infections through the action of bacterial cytotoxins and differential activation of innate signaling pathways acting upstream of NF-kappaB.


Subject(s)
Bacterial Toxins/pharmacology , Carrier Proteins/metabolism , NF-kappa B/physiology , Nod1 Signaling Adaptor Protein/physiology , Nod2 Signaling Adaptor Protein/physiology , Signal Transduction/immunology , Toll-Like Receptors/physiology , Vibrio cholerae/pathogenicity , Vibrio vulnificus/pathogenicity , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/microbiology , Bone Marrow Cells/pathology , Carrier Proteins/genetics , Carrier Proteins/physiology , Caspase 1/metabolism , Immunity, Innate/genetics , Inflammation/enzymology , Inflammation/immunology , Inflammation/microbiology , Interleukin-1beta/metabolism , Ligands , Macrophages/immunology , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Signal Transduction/genetics , Vibrio cholerae/immunology , Vibrio vulnificus/immunology
11.
Nat Cell Biol ; 6(2): 121-8, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14730312

ABSTRACT

The apoptosis-associated speck-like protein (ASC) is an unusual adaptor protein that contains the Pyrin/PAAD death domain in addition to the CARD protein-protein interaction domain. Here, we present evidence that ASC can function as an adaptor molecule for Bax and regulate a p53-Bax mitochondrial pathway of apoptosis. When ectopically expressed, ASC interacted directly with Bax, colocalized with Bax to the mitochondria, induced cytochrome c release with a significant reduction of mitochondrial membrane potential and resulted in the activation of caspase-9, -2 and -3. The rapid induction of apoptosis by ASC was not observed in Bax-deficient cells. We also show that induction of ASC after exposure to genotoxic stress is dependent on p53. Blocking of endogenous ASC expression by small-interfering RNA (siRNA) reduced the apoptotic response and inhibited translocation of Bax to mitochondria in response to p53 or genotoxic insult, suggesting that ASC is required to translocate Bax to the mitochondria. Our findings demonstrate that ASC has an essential role in the intrinsic mitochondrial pathway of apoptosis through a p53-Bax network.


Subject(s)
Apoptosis/physiology , Cytoskeletal Proteins/metabolism , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Base Sequence , CARD Signaling Adaptor Proteins , Caspases/metabolism , Cell Line, Tumor , Cytochromes c/metabolism , Cytoskeletal Proteins/genetics , Enzyme Activation , Humans , Membrane Potentials/physiology , Molecular Sequence Data , Protein Structure, Tertiary , Protein Transport , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , bcl-2-Associated X Protein
12.
J Exp Med ; 196(12): 1605-15, 2002 Dec 16.
Article in English | MEDLINE | ID: mdl-12486103

ABSTRACT

Apoptosis-associated speck-like protein containing a Caspase recruitment domain (ASC) belongs to a large family of proteins that contain a Pyrin, AIM, ASC, and death domain-like (PAAD) domain (also known as PYRIN, DAPIN, Pyk). Recent data have suggested that ASC functions as an adaptor protein linking various PAAD-family proteins to pathways involved in nuclear factor (NF)-kappaB and pro-Caspase-1 activation. We present evidence here that the role of ASC in modulating NF-kappaB activation pathways is much broader than previously suspected, as it can either inhibit or activate NF-kappaB, depending on cellular context. While coexpression of ASC with certain PAAD-family proteins such as Pyrin and Cryopyrin increases NF-kappaB activity, ASC has an inhibitory influence on NF-kappaB activation by various proinflammatory stimuli, including tumor necrosis factor (TNF)alpha, interleukin 1beta, and lipopolysaccharide (LPS). Elevations in ASC protein levels or of the PAAD domain of ASC suppressed activation of IkappaB kinases in cells exposed to pro-inflammatory stimuli. Conversely, reducing endogenous levels of ASC using siRNA enhanced TNF- and LPS-induced degradation of the IKK substrate, IkappaBalpha. Our findings suggest that ASC modulates diverse NF-kappaB induction pathways by acting upon the IKK complex, implying a broad role for this and similar proteins containing PAAD domains in regulation of inflammatory responses.


Subject(s)
Cytoskeletal Proteins/metabolism , NF-kappa B/metabolism , Binding Sites , CARD Signaling Adaptor Proteins , Cell Line , Cytoskeletal Proteins/genetics , Genes, Reporter , Humans , I-kappa B Kinase , Macromolecular Substances , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Proteins/genetics , Proteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , TNF Receptor-Associated Factor 1 , Tumor Necrosis Factor-alpha/metabolism
13.
Circulation ; 117(24): 3079-87, 2008 Jun 17.
Article in English | MEDLINE | ID: mdl-18541743

ABSTRACT

BACKGROUND: Inflammatory cytokines such as interleukin (IL)-1 beta and IL-18 play an important role in the development of atherosclerosis and restenosis. Apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) is an adaptor protein that regulates caspase-1-dependent IL-1 beta and IL-18 generation; however, the role of ASC in vascular injury remains undefined. Here, we investigated the contribution of ASC to neointimal formation after vascular injury in ASC-deficient (ASC(-/-)) mice. METHODS AND RESULTS: Wire-mediated vascular injury was produced in the femoral artery of ASC(-/-) and wild-type mice. Immunohistochemical analysis revealed that ASC was markedly expressed at the site of vascular injury. Neointimal formation was significantly attenuated in ASC(-/-) mice after injury. IL-1 beta and IL-18 were expressed in the neointimal lesion in wild-type mice but showed decreased expression in the lesion of ASC(-/-) mice. To investigate the contribution of bone marrow-derived cells, we developed bone marrow-transplanted mice and found that neointimal formation was significantly decreased in wild-type mice in which bone marrow was replaced with ASC(-/-) bone marrow cells. Furthermore, in vitro experiments showed that the proliferation activity of ASC(-/-) vascular smooth muscle cells was not impaired. CONCLUSIONS: These findings suggest that bone marrow-derived ASC is critical for neointimal formation after vascular injury and identify ASC as a novel therapeutic target for atherosclerosis and restenosis.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Cytoskeletal Proteins/deficiency , Tunica Intima/physiopathology , Vascular Diseases/pathology , Vascular Diseases/physiopathology , Animals , Apoptosis , Apoptosis Regulatory Proteins , Bone Marrow Transplantation , CARD Signaling Adaptor Proteins , Caspases/deficiency , Caspases/genetics , Caspases/metabolism , Cell Culture Techniques , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/physiology , Extracellular Signal-Regulated MAP Kinases/genetics , Femoral Artery/injuries , Femoral Artery/pathology , Femoral Artery/physiopathology , Immunohistochemistry , Inflammation/pathology , Inflammation/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Tunica Intima/pathology
14.
Mediators Inflamm ; 2009: 287387, 2009.
Article in English | MEDLINE | ID: mdl-19759850

ABSTRACT

The apoptosis-associated speck-like protein containing a caspase recruit domain (ASC) is involved in apoptosis and innate immunity and is a major adaptor molecule responsible for procaspase-1 activation. ASC mRNA is encoded by three exons: exons 1 and 3 encode a pyrin domain (PYD) and caspase recruit domain (CARD), respectively, and exon 2 encodes a proline and glycine-rich (PGR) domain. Here, we identified a variant ASC protein (vASC) lacking the PGR domain that was smaller than full length ASC (fASC) derived from fully transcribed mRNA and searched for differences in biochemical and biological nature. Both fASC and vASC were found to activate procaspase-1 to a similar degree, but the efficiency of IL-1beta excretion was significantly higher for vASC. There was also a marked structural difference observed in the fibrous aggregates formed by fASC and vASC. These results suggest that although the PGR domain is dispensable for procaspase-1 activation, it plays an important role in the regulation of the molecular structure and activity of ASC.


Subject(s)
Alternative Splicing , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Interleukin-1beta/metabolism , Blotting, Western , CARD Signaling Adaptor Proteins , Caspase 1/metabolism , HL-60 Cells , Humans , Mass Spectrometry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
15.
Oral Oncol ; 41(1): 38-47, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15598584

ABSTRACT

Dihydropyrimidine dehydrogenase (DPD) is the rate-limiting enzyme of 5-fluorouracil (5-Fu) catabolism. The aim of this study was to evaluate the prognostic value of DPD expression and the correlation between DPD expression and efficacy of 5-Fu. Retrospective analysis of DPD expression was performed immunohistochemically in 103 patients with oral squamous cell carcinoma (OSCC), in which staining intensity of DPD expression and degree of heterogeneity of DPD expression were categorized. Expression of DPD correlated with lymph node metastasis, mode of invasion and differentiation. Expression of DPD was an independent significant factor for survival outcome and was more predictive than conventional clinical factors. Furthermore, heterogeneous expression of DPD was more effective than homogeneous expression of DPD in neoplastic cells when evaluated in patients treated with chemotherapy including tegafur/uracil (UFT). Expression of DPD is an independent predictor for clinical outcome. Furthermore, heterogeneity of DPD expression may be a clue for predicting sensitivity to 5-Fu in patients with OSCC.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/drug therapy , Dihydrouracil Dehydrogenase (NADP)/metabolism , Fluorouracil/therapeutic use , Mouth Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/pathology , Female , Humans , Immunoenzyme Techniques , Lymphatic Metastasis , Male , Middle Aged , Mouth Neoplasms/enzymology , Mouth Neoplasms/pathology , Neoplasm Invasiveness , Neoplasm Proteins/metabolism , Prognosis , Proliferating Cell Nuclear Antigen/metabolism , Retrospective Studies , Survival Analysis , Treatment Outcome , Tumor Suppressor Protein p53/metabolism
16.
Clin Cancer Res ; 10(2): 572-80, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14760079

ABSTRACT

PURPOSE: Moesin is a linking protein of the submembraneous cytoskeleton and plays a key role in the control of cell morphology, adhesion, and motility. The aim of the present study was to elucidate the clinical significance of expression patterns of moesin in patients with oral squamous cell carcinoma (OSCC). EXPERIMENTAL DESIGN: Immunohistochemistry for moesin monoclonal antibody was performed on 103 paraffin-embedded specimens from patients with primary OSCC, including 30 patients with locoregional lymph node metastasis, and in the sections from nude mice transplanted with two cell lines derived from a single human tongue cancer (SQUU-A and SQUU-B). RESULTS: Expression patterns of moesin in OSCCs were divided into three groups: membranous pattern; mixed pattern; and cytoplasmic pattern. These expression patterns correlated with tumor size, lymph node metastasis, mode of invasion, differentiation, and lymphocytic infiltration. In about two-thirds of the patients with metastatic lymph node, homogeneous cytoplasmic expression was detected in the metastatic lymph nodes. In addition, SQUU-B with high metastatic potential showed more reduced levels of membrane-bound moesin than SQUU-A with low metastatic potential. A multivariate analysis demonstrated that expression patterns of moesin can be an independent prognostic factor. CONCLUSIONS: Our results suggest that moesin expression contributed to discriminating between patients with the potentiality for locoregional lymph node metastasis and those with a better prognosis and might improve the definition of suitable therapy for each.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Microfilament Proteins/biosynthesis , Mouth Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Animals , Antibodies, Monoclonal , Carcinoma, Squamous Cell/pathology , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cytoplasm/metabolism , Epithelium/metabolism , Female , Humans , Immunohistochemistry , Lymphatic Metastasis , Male , Mice , Mice, Inbred BALB C , Microfilament Proteins/metabolism , Middle Aged , Mouth Neoplasms/pathology , Multivariate Analysis , Neoplasm Metastasis , Prognosis , Time Factors
17.
FEBS Open Bio ; 5: 348-56, 2015.
Article in English | MEDLINE | ID: mdl-25973362

ABSTRACT

Increasing evidence indicates that caspase recruitment domain (CARD)-mediated caspase-1 (CASP1) assembly is an essential process for its activation and subsequent interleukin (IL)-1ß release, leading to the initiation of inflammation. Both CARD16 and CARD17 were previously reported as inhibitory homologs of CASP1; however, their molecular function remains unclear. Here, we identified that oligomerization activity allows CARD16 to function as a CASP1 activator. We investigated the molecular characteristics of CARD16 and CARD17 in transiently transfected HeLa cells. Although both CARD16 and CARD17 interacted with CASP1CARD, only CARD16 formed a homo-oligomer. Oligomerized CARD16 formed a filament-like structure with CASP1CARD and a speck with apoptosis-associated speck-like protein containing a CARD. A filament-like structure formed by CARD16 promoted CASP1 filament assembly and IL-1ß release. In contrast, CARD17 did not form a homo-oligomer or filaments and inhibited CASP1-dependent IL-1ß release. Mutated CARD16D27G, mimicking the CARD17 amino acid sequence, formed a homo-oligomer but failed to form a filament-like structure. Consequently, CARD16D27G weakly promoted CASP1 filament assembly and subsequent IL-1ß release. These results suggest that oligomerized CARD16 promotes CARD-mediated molecular assembly and CASP1 activation.

18.
Nanotoxicology ; 9(5): 554-67, 2015.
Article in English | MEDLINE | ID: mdl-25211550

ABSTRACT

Despite the increasing commercial use of nanoparticles, little is known about their effects on placental inflammation and pregnancy complications. In this study, nanosilica (NS) particles upregulated the inflammasome component nucleotide-binding oligomerization domain-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) and induced placental inflammation and reactive oxygen species (ROS) generation, resulting in pregnancy complications. Furthermore, NS-induced pregnancy complications were markedly improved in Nlrp3(-/-) mice but not in component apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC)-deficient (Asc(-/-)) mice, indicating the independence of NLRP3 inflammasomes. Pregnancy complications in Nlrp3(-/-) and Asc(-/-) mice phenotypes were dependent on the balance between interleukin (IL)-1α and IL-10. NS-induced pregnancy complications were completely prevented by either inhibition of ROS generation or forced expression of IL-10. Our findings provide important information about NS-induced placental inflammation and pregnancy complications and the novel pathophysiological roles of NLRP3 and ASC in pregnancy.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Carrier Proteins/metabolism , Inflammasomes/immunology , Nanoparticles/toxicity , Placenta/drug effects , Pregnancy Complications/chemically induced , Silicon Dioxide/toxicity , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , CARD Signaling Adaptor Proteins , Carrier Proteins/genetics , Female , Inflammasomes/metabolism , Interleukin-10/immunology , Interleukin-1alpha/immunology , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Nanoparticles/chemistry , Placenta/immunology , Placenta/pathology , Pregnancy , Pregnancy Complications/immunology , Pregnancy Complications/pathology , Reactive Oxygen Species/metabolism , Silicon Dioxide/chemistry
19.
Nat Commun ; 6: 7515, 2015 Jun 24.
Article in English | MEDLINE | ID: mdl-26104484

ABSTRACT

TLR2 promotes NLRP3 inflammasome activation via an early MyD88-IRAK1-dependent pathway that provides a priming signal (signal 1) necessary for activation of the inflammasome by a second potassium-depleting signal (signal 2). Here we show that TLR3 binding to dsRNA promotes post-translational inflammasome activation through intermediate and late TRIF/RIPK1/FADD-dependent pathways. Both pathways require the scaffolding but not the catalytic function of caspase-8 or RIPK1. Only the late pathway requires kinase competent RIPK3 and MLKL function. Mechanistically, FADD/caspase-8 scaffolding function provides a post-translational signal 1 in the intermediate pathway, whereas in the late pathway it helps the oligomerization of RIPK3, which together with MLKL provides both signal 1 and 2 for inflammasome assembly. Cytoplasmic dsRNA activates NLRP3 independent of TRIF, RIPK1, RIPK3 or mitochondrial DRP1, but requires FADD/caspase-8 in wildtype macrophages to remove RIPK3 inhibition. Our study provides a comprehensive analysis of pathways that lead to NLRP3 inflammasome activation in response to dsRNA.


Subject(s)
Carrier Proteins/metabolism , Caspase 8/metabolism , Macrophages/metabolism , Protein Kinases/metabolism , RNA, Double-Stranded/metabolism , Toll-Like Receptor 3/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Carrier Proteins/genetics , Caspase 8/genetics , Dynamins/genetics , Dynamins/metabolism , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Mice , Mice, Knockout , Mitochondria/metabolism , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Protein Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
20.
Cancer Lett ; 202(1): 101-8, 2003 Dec 08.
Article in English | MEDLINE | ID: mdl-14643031

ABSTRACT

ASC/TMS1, a proapoptotic activator of procaspase-1, was reported to be aberrantly methylated in human breast cancer. We found that ASC was methylated in three of five human colon cancer cell lines lacking ASC protein expression. Demethylation treatment of these cell lines lacking ASC with 5-aza-2'-deoxycytidine partially restored ASC expression. Methylated ASC was also detected in six of ten colorectal cancer tissues. Although clear down-regulation of ASC in the whole region of a tumor tissue was hardly observed by immunostaining with anti-ASC mAb, complete suppression of ASC was identified in a minor population of the colorectal tumor cells. The biological significance of ASC methylation inducible ASC suppression in colorectal cancer will be discussed.


Subject(s)
Caspases/metabolism , Colorectal Neoplasms/metabolism , Cytoskeletal Proteins/metabolism , DNA Methylation , Enzyme Precursors/metabolism , Proteins/metabolism , Adult , Aged , Aged, 80 and over , Apoptosis , Azacitidine/pharmacology , CARD Signaling Adaptor Proteins , Case-Control Studies , Caspase 1 , Colorectal Neoplasms/pathology , Cytoskeletal Proteins/genetics , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Primers/chemistry , DNA, Neoplasm/genetics , Down-Regulation , Enzyme Activation , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Male , Middle Aged , Polymerase Chain Reaction , Proteins/genetics , Sulfites/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL