Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
FASEB J ; 29(5): 1869-78, 2015 May.
Article in English | MEDLINE | ID: mdl-25667218

ABSTRACT

A20 protects against pathologic vascular remodeling by inhibiting the inflammatory transcription factor NF-κB. A20's function has been attributed to ubiquitin editing of receptor-interacting protein 1 (RIP1) to influence activity/stability. The validity of this mechanism was tested using a murine model of transplant vasculopathy and human cells. Mouse C57BL/6 aortae transduced with adenoviruses containing A20 (or ß-galactosidase as a control) were allografted into major histocompatibility complex-mismatched BALB/c mice. Primary endothelial cells, smooth muscle cells, or transformed epithelial cells (all human) were transfected with wild-type A20 or with catalytically inactive mutants as a control. NF-κB activity and intracellular localization of RIP1 was monitored by reporter gene assay, immunofluorescent staining, and Western blotting. Native and catalytically inactive versions of A20 had similar inhibitory effects on NF-κB activity (-70% vs. -76%; P > 0.05). A20 promoted localization of RIP1 to insoluble aggresomes in murine vascular allografts and in human cells (53% vs. 0%) without altering RIP1 expression, and this process was increased by the assembly of polyubiquitin chains (87% vs. 28%; P < 0.05). A20 captures polyubiquitinated signaling intermediaries in insoluble aggresomes, thus reducing their bioavailability for downstream NF-κB signaling. This novel mechanism contributes to protection from vasculopathy in transplanted organs treated with exogenous A20.


Subject(s)
Aorta/transplantation , Carotid Arteries/surgery , Cysteine Endopeptidases/metabolism , GTPase-Activating Proteins/metabolism , Inflammation/immunology , Intracellular Signaling Peptides and Proteins/metabolism , NF-kappa B/metabolism , Protein Aggregates/physiology , Adenoviridae/genetics , Allografts , Animals , Aorta/metabolism , Aorta/pathology , Blotting, Western , Carotid Arteries/metabolism , Carotid Arteries/pathology , Cell Proliferation , Cells, Cultured , Cysteine Endopeptidases/genetics , GTPase-Activating Proteins/genetics , Graft Rejection/etiology , Graft Rejection/metabolism , Graft Rejection/pathology , Histocompatibility , Humans , Immunity, Innate/immunology , Immunoenzyme Techniques , Inflammation/metabolism , Inflammation/pathology , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , NF-kappa B/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tumor Necrosis Factor alpha-Induced Protein 3 , Ubiquitin/metabolism , Ubiquitination
2.
Adv Exp Med Biol ; 809: 33-48, 2014.
Article in English | MEDLINE | ID: mdl-25302364

ABSTRACT

A20 is a ubiquitin-editing molecule. It belongs to a novel family of deubiquitinating cysteine proteases, called the ovarian tumor (OTU) family, which can cleave monoubiquitin from modified proteins. In addition, A20 contains seven Cys2-Cys2 zinc fingers, one of which is believed to regulate E3 ubiquitin ligase activity. Here we review the biology of human genes that encode OTU domains or contain A20-type zinc fingers. The human genome contains 15 members of the OTU family including the deubiquitinating enzymes Cezanne, VCIP135 and Otubain 1. Genomic analysis also identified 10 genes that contain A20-type zinc fingers including Rabex5, Znf216 and AWP1. In Rabex5 the A20-zinc finger regulates E3 ligase activity whereas A20-type zinc fingers of Znf216 and AWP1 function as ubiquitin-binding motifs. A20 and its relatives regulate highly divergent physiological activities including NF-kappaB activity (A20, Cezanne, Znf216, Rabex5), endocytosis (Rabex5, AWP1), skeletal muscle atrophy (Znf216), Golgi membrane fusion (VCIP135) and T-cell anergy (Otubain 1). Further studies are required to characterize the biology of other A20-related molecules whose function remains largely undefined.


Subject(s)
DNA-Binding Proteins/physiology , Intracellular Signaling Peptides and Proteins/physiology , Nuclear Proteins/physiology , Amino Acid Sequence , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Molecular Sequence Data , NF-kappa B/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Sequence Homology, Amino Acid , Tumor Necrosis Factor alpha-Induced Protein 3 , Ubiquitin/physiology , Ubiquitin-Specific Proteases/metabolism , Zinc Fingers
3.
J Biol Chem ; 288(34): 24569-80, 2013 Aug 23.
Article in English | MEDLINE | ID: mdl-23846693

ABSTRACT

Double-stranded (ds) RNA of viral origin, a ligand for Melanoma Differentiation-associated gene 5 (MDA5) and Toll-Like Receptor 3 (TLR3), induces the TANK-Binding Kinase 1 (TBK1)-dependent phosphorylation and activation of Interferon Regulatory Factor 3 (IRF3) and the E3 ubiquitin ligase Pellino1, which are required for interferon ß (IFNß) gene transcription. Here, we report that Pellino1 interacts with the transcription factor Deformed Epidermal Autoregulatory Factor 1 (DEAF1). The interaction is independent of the E3 ligase activity of Pellino1, but weakened by the phosphorylation of Pellino1. We show that DEAF1 binds to the IFNß promoter and to IRF3 and IRF7, that it is required for the transcription of the IFNß gene and IFNß secretion in MEFs infected with Sendai virus or transfected with poly(I:C). DEAF1 is also needed for TLR3-dependent IFNß production. Taken together, our results identify DEAF1 as a novel component of the signal transduction network by which dsRNA of viral origin stimulates IFNß production.


Subject(s)
Interferon-beta/biosynthesis , Nuclear Proteins/metabolism , RNA, Double-Stranded/pharmacology , Respirovirus Infections/metabolism , Sendai virus/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , DNA-Binding Proteins , HEK293 Cells , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon-Induced Helicase, IFIH1 , Interferon-beta/genetics , Mice , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Respirovirus Infections/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism , Transcription Factors/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Ubiquitin-Protein Ligases/genetics
4.
J Biol Chem ; 287(41): 34825-35, 2012 Oct 05.
Article in English | MEDLINE | ID: mdl-22902624

ABSTRACT

Viral double-stranded RNA, a ligand for Toll-like Receptor 3 (TLR3) and the cytoplasmic RNA receptors RIG1 and MDA5, activate a signaling network in which the IKK-related protein kinase TBK1 phosphorylates the transcription factor Interferon Regulatory Factor 3 (IRF3) and the E3 ubiquitin ligase Pellino1. IRF3 then translocates to the nucleus where it stimulates transcription of the interferonß (IFNß) gene, but the function of Pellino1 in this pathway is unknown. Here, we report that myeloid cells and embryonic fibroblasts from knock-in mice expressing an E3 ligase-deficient mutant of Pellino1 produce reduced levels of IFNß mRNA and secrete much less IFNß in response to viral double-stranded RNA because the interaction of IRF3 with the IFNß promoter is impaired. These results identify Pellino1 as a novel component of the signal transduction network by which viral double-stranded RNA stimulates IFNß gene transcription.


Subject(s)
Cell Nucleus/metabolism , Fibroblasts/metabolism , Interferon-beta/biosynthesis , Nuclear Proteins/metabolism , RNA, Double-Stranded/metabolism , RNA, Messenger/biosynthesis , RNA, Viral/metabolism , Active Transport, Cell Nucleus , Animals , Cell Nucleus/genetics , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fibroblasts/cytology , Gene Knock-In Techniques , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon-Induced Helicase, IFIH1 , Interferon-beta/genetics , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Promoter Regions, Genetic/physiology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Double-Stranded/genetics , RNA, Messenger/genetics , RNA, Viral/genetics , Signal Transduction/physiology , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism , Transcription, Genetic/physiology , Ubiquitin-Protein Ligases
5.
J Mol Med (Berl) ; 86(12): 1329-39, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18813897

ABSTRACT

Ischaemia followed by reperfusion (I/R) can induce inflammation and injury and is a risk factor for delayed graft function and rejection of transplanted kidneys. Inflammation is regulated by NF-kappaB transcription factors which induce pro-inflammatory molecules in endothelial cells (EC). We examined whether A20, a negative regulator of NF-kappaB, can protect kidneys from I/R injury. To mimic the fluctuations in endothelial oxygenation that occur during I/R we exposed cultured human umbilical vein EC (HUVEC) to hypoxia (1% O(2) for 4 h) followed by re-oxygenation (21% O(2) for 1 h-24 h). We observed transient expression of pro-inflammatory molecules (E-selectin, VCAM-1 and IL-8) and sustained expression of A20 in HUVEC exposed to hypoxia/re-oxygenation. The effect of A20 on endothelial responses to hypoxia/re-oxygenation was assessed. We observed that pre-treatment of HUVEC with an adenovirus containing A20 (Ad-A20) suppressed activation of NF-kappaB and induction of pro-inflammatory molecules by hypoxia/re-oxygenation, whereas a control adenovirus had little or no effect. Thus the induction of A20 may form a negative feedback loop in pro-inflammatory signalling in cells exposed to hypoxia/re-oxygenation. To validate our cell culture experiments we examined the role of A20 in renal responses to I/R. We observed that A20 was induced in rat kidneys exposed to I/R. Moreover, pre-treatment of animals with Ad-A20 significantly reduced acute tubular necrosis, renal expression of VCAM-1 and NF-kappaB activation in response to I/R, whereas pre-treatment with control adenovirus did not. Our observations suggest that A20 maintains physiological homeostasis in kidneys exposed to I/R by protecting them from inflammation and injury.


Subject(s)
DNA-Binding Proteins/immunology , Endothelial Cells/immunology , Intracellular Signaling Peptides and Proteins/immunology , Kidney/immunology , Nuclear Proteins/immunology , Reperfusion Injury/immunology , Reperfusion Injury/prevention & control , Animals , Cells, Cultured , Endothelial Cells/drug effects , Humans , Intracellular Signaling Peptides and Proteins/genetics , Kidney/drug effects , Male , NF-kappa B/genetics , NF-kappa B/immunology , Nuclear Proteins/genetics , Rats , Rats, Inbred F344 , Transfection , Tumor Necrosis Factor alpha-Induced Protein 3 , Tumor Necrosis Factor-alpha/immunology
6.
Circ Res ; 103(7): 726-32, 2008 Sep 26.
Article in English | MEDLINE | ID: mdl-18723442

ABSTRACT

Atherosclerosis is a chronic inflammatory disease of arteries. It is triggered by proinflammatory mediators which induce adhesion molecules (eg, vascular cell adhesion molecule [VCAM]-1) in endothelial cells (ECs) by activating p38 and c-Jun N-terminal kinase (JNK) mitogen-activated protein (MAP) kinases by phosphorylation. Blood flow influences atherosclerosis by exerting shear stress (mechanical drag) on the inner surface of arteries, a force that alters endothelial physiology. Regions of the arterial tree exposed to high shear are protected from endothelial activation, inflammation, and atherosclerosis, whereas regions exposed to low or oscillatory shear are susceptible. We examined whether MAP kinase phosphatase (MKP)-1, a negative regulator of p38 and JNK, mediates the antiinflammatory effects of shear stress. We observed that expression of MKP-1 in cultured ECs was elevated by shear stress, whereas the expression of VCAM-1 was reduced. MKP-1 induction was shown to be necessary for the antiinflammatory effects of shear stress because gene silencing of MKP-1 restored VCAM-1 expression in sheared ECs. Immunostaining revealed that MKP-1 is preferentially expressed by ECs in a high-shear, protected region of the mouse aorta and is necessary for suppression of EC activation at this site, because p38 activation and VCAM-1 expression was enhanced by genetic deletion of MKP-1. We conclude that MKP-1 induction is required for the antiinflammatory effects of shear stress. Thus, our findings reveal a novel molecular mechanism contributing to the spatial distribution of vascular inflammation and atherosclerosis.


Subject(s)
Atherosclerosis/enzymology , Dual Specificity Phosphatase 1/biosynthesis , Endothelial Cells/enzymology , Gene Expression Regulation, Enzymologic , Animals , Aorta/enzymology , Atherosclerosis/genetics , Cells, Cultured , Chronic Disease , Dual Specificity Phosphatase 1/genetics , Enzyme Activation/genetics , Humans , Inflammation/enzymology , Inflammation/genetics , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Knockout , Phosphorylation , Shear Strength , Stress, Mechanical , Vascular Cell Adhesion Molecule-1/biosynthesis , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
7.
J Biol Chem ; 283(27): 18582-90, 2008 Jul 04.
Article in English | MEDLINE | ID: mdl-18474597

ABSTRACT

NF-kappaB transcription factors induce pro-inflammatory molecules (e.g. IL-8) in response to cytokines (e.g. TNFalpha, IL-1beta) or other stimuli. In the basal state, they are sequestered in the cytoplasm by inhibitory IkappaB proteins. Pro-inflammatory signaling triggers polyubiquitination of intermediaries (e.g. RIP1), which activate IkappaB kinases that trigger Ser phosphorylation and degradation of IkappaBalpha, thereby promoting nuclear translocation of NF-kappaB. A negative feedback loop exists whereby NF-kappaB drives resynthesis of IkappaBalpha, which promotes export of NF-kappaB from the nucleus to the cytoplasm. This process relies on Cezanne, a deubiquitinating cysteine protease that stabilizes resynthesized IkappaBalpha by removing polyubiquitin from modified intermediaries. H(2)O(2) is generated during inflammation. Here we examined the effects of H(2)O(2) on NF-kappaB dynamics and pro-inflammatory activation in cultured cells co-stimulated with TNFalpha or IL-1beta. Quantitative reverse transcription-PCR and enzyme-linked immunosorbent assay revealed that H(2)O(2) enhanced the induction of IL-8 by TNFalpha or IL-1beta. We demonstrated by using assays of NF-kappaB nuclear localization and by imaging of live cells expressing a fluorescent form of NF-kappaB that H(2)O(2) prolonged NF-kappaB nuclear localization in cells co-stimulated with TNFalpha or IL-1beta by suppressing its export from the nucleus. We provide evidence that H(2)O(2) suppresses NF-kappaB export by prolonging polyubiquitination of signaling intermediaries, which promotes Ser phosphorylation and destabilization of newly synthesized IkappaBalpha proteins. Finally, we observed that the catalytic activity of Cezanne and its ability to suppress RIP1 polyubiquitination and NF-kappaB transcriptional activity were inhibited by H(2)O(2). We conclude that H(2)O(2) prolongs NF-kappaB activation in co-stimulated cells by suppressing the negative regulatory functions of Cezanne and IkappaBalpha.


Subject(s)
Cell Nucleus/metabolism , Hydrogen Peroxide/pharmacology , NF-kappa B/metabolism , Oxidants/pharmacology , Signal Transduction/drug effects , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/physiology , Cytokines/metabolism , Cytokines/pharmacology , Endopeptidases/metabolism , HeLa Cells , Humans , I-kappa B Kinase/metabolism , I-kappa B Proteins/metabolism , Inflammation Mediators/metabolism , Inflammation Mediators/pharmacology , Nuclear Pore Complex Proteins/metabolism , Phosphorylation/drug effects , RNA-Binding Proteins/metabolism , Signal Transduction/physiology , Ubiquitin/metabolism , Ubiquitination/drug effects , Ubiquitination/physiology
8.
Cardiovasc Res ; 79(3): 472-80, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18375498

ABSTRACT

AIMS: The molecular mechanisms that regulate cardiomyocyte apoptosis and their role in human heart failure (HF) are uncertain. Expression of the apoptosis regulator p53 is governed by minute double minute 2 (MDM2), an E3 enzyme that targets p53 for ubiquitination and proteasomal processing, and by the deubiquitinating enzyme, herpesvirus-associated ubiquitin-specific protease (HAUSP), which rescues p53 by removing ubiquitin chains from it. Here, we examined whether elevated expression of p53 was associated with dysregulation of ubiquitin-proteasome system (UPS) components and activation of downstream effectors of apoptosis in human dilated cardiomyopathy (DCM). METHODS AND RESULTS: Left ventricular myocardial samples were obtained from patients with DCM (n = 12) or from non-failing (donor) hearts (n = 17). Western blotting and immunohistochemistry revealed that DCM tissues contained elevated levels of p53 and its regulators MDM2 and HAUSP (all P < 0.01) compared with non-failing hearts. DCM tissues also contained elevated levels of polyubiquitinated proteins and possessed enhanced 20S-proteasome chymotrypsin-like activities (P < 0.04) as measured in vitro using a fluorogenic substrate. DCM tissues contained activated caspases-9 and -3 (P < 0.001) and reduced expression of the caspase substrate PARP-1 (P < 0.05). Western blotting and immunohistochemistry revealed that DCM tissues contained elevated expression levels of caspase-3-activated DNAse (CAD; P < 0.001), which is a key effector of DNA fragmentation in apoptosis and also contained elevated expression of a potent inhibitor of CAD (ICAD-S; P < 0.01). CONCLUSION: Expression of p53 in human DCM is associated with dysregulation of UPS components, which are known to regulate p53 stability. Elevated p53 expression and caspase activation in DCM was not associated with activation of both CAD and its inhibitor, ICAD-S. Our findings are consistent with the concept that apoptosis may be interrupted and therefore potentially reversible in human HF.


Subject(s)
Apoptosis , Cardiomyopathy, Dilated/enzymology , Myocardium/enzymology , Proteasome Endopeptidase Complex/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitin/metabolism , Adult , Apoptosis Regulatory Proteins/metabolism , Cardiomyopathy, Dilated/pathology , Caspase 3/metabolism , Caspase 9/metabolism , DNA Fragmentation , Deoxyribonucleases/metabolism , Female , Humans , Male , Middle Aged , Myocardium/pathology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Specific Peptidase 7 , Up-Regulation , Young Adult
9.
J Biol Chem ; 283(11): 7036-45, 2008 Mar 14.
Article in English | MEDLINE | ID: mdl-18178551

ABSTRACT

Transcription factors belonging to the NF-kappaB family regulate inflammation by inducing pro-inflammatory molecules (e.g. interleukin (IL)-8) in response to cytokines (e.g. tumor necrosis factor (TNF) alpha, IL-1) or other stimuli. Several negative regulators of NF-kappaB, including the ubiquitin-editing enzyme A20, participate in the resolution of inflammatory responses. We report that Cezanne, a member of the A20 family of the deubiquitinating cysteine proteases, can be induced by TNFalpha in cultured cells. Silencing of endogenous Cezanne using small interfering RNA led to elevated NF-kappaB luciferase reporter gene activity and enhanced expression of IL-8 transcripts in TNFalpha-treated cells. Thus we conclude that endogenous Cezanne can attenuate NF-kappaB activation and the induction of pro-inflammatory transcripts in response to TNF receptor (TNFR) signaling. Overexpression studies revealed that Cezanne suppressed NF-kappaB nuclear translocation and transcriptional activity by targeting the TNFR signaling pathway at the level of the IkappaB kinase complex or upstream from it. These effects were not observed in a form of Cezanne that was mutated at the catalytic cysteine residue (Cys209), indicating that the deubiquitinating activity of Cezanne is essential for NF-kappaB regulation. Finally, we demonstrate that Cezanne can be recruited to activated TNFRs where it suppresses the build-up of polyubiquitinated RIP1 signal adapter proteins. Thus we conclude that Cezanne forms a novel negative feedback loop in pro-inflammatory signaling and that it suppresses NF-kappaB activation by targeting RIP1 signaling intermediaries for deubiquitination.


Subject(s)
Endopeptidases/metabolism , Gene Expression Regulation, Enzymologic , Inflammation , NF-kappa B/metabolism , Nuclear Pore Complex Proteins/metabolism , RNA-Binding Proteins/metabolism , Ubiquitin/metabolism , DNA-Binding Proteins , Endothelial Cells/cytology , Humans , Interleukin-8/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lysine/chemistry , Models, Biological , Mutation , Nuclear Proteins/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , Tumor Necrosis Factor alpha-Induced Protein 3
10.
FASEB J ; 21(13): 3553-61, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17557931

ABSTRACT

Regions of the arterial tree exposed to laminar flow, which exerts high shear stress, are protected from inflammation, endothelial cell (EC) death and atherosclerosis. TNFalpha activates NF-kappaB transcription factors, which potentially exert dual functions by inducing both proinflammatory and cytoprotective transcripts. We assessed whether laminar shear stress protects EC by modulating NF-kappaB function. Human umbilical vein EC (HUVEC) were cultured under shear stress (12 dynes/cm2 for 16 h) using a parallel-plate flow chamber or were maintained in static conditions. Comparative real-time PCR revealed that preshearing significantly alters transcriptional responses to TNFalpha by enhancing the expression of cytoprotective molecules (Bcl-2, MnSOD, GADD45beta, A1) and suppressing proinflammatory transcripts (E-selectin, VCAM-1, IL-8). We demonstrated using assays of nuclear localization, NF-kappaB subunit phosphorylation, DNA-binding, and transcriptional activity that NF-kappaB is activated by TNFalpha in presheared HUVEC. Furthermore, a specific inhibitor revealed that NF-kappaB is essential for the induction of cytoprotective transcripts in presheared EC. Finally, we observed that NF-kappaB can be activated in vascular endothelium exposed to laminar shear stress in NF-kappaB-luciferase reporter mice, thus validating our cell culture experiments. We conclude that shear stress primes EC for enhanced NF-kappaB-dependent cytoprotective responsiveness while attenuating proinflammatory activation. Thus modulation of NF-kappaB function may underlie the atheroprotective effects of laminar shear stress.


Subject(s)
Endothelium, Vascular/metabolism , NF-kappa B/metabolism , Stress, Mechanical , Base Sequence , Cells, Cultured , DNA Primers , Endothelium, Vascular/cytology , Humans , NF-kappa B/genetics , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...