Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Viruses ; 11(5)2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31052481

RESUMO

The host immune response and virus-encoded immune evasion proteins pose constant, mutual selective pressure on each other. Virally encoded immune evasion proteins also indicate which host pathways must be inhibited to allow for viral replication. Here, we show that IIV-6 is capable of inhibiting the two Drosophila NF-κB signaling pathways, Imd and Toll. Antimicrobial peptide (AMP) gene induction downstream of either pathway is suppressed when cells infected with IIV-6 are also stimulated with Toll or Imd ligands. We find that cleavage of both Imd and Relish, as well as Relish nuclear translocation, three key points in Imd signal transduction, occur in IIV-6 infected cells, indicating that the mechanism of viral inhibition is farther downstream, at the level of Relish promoter binding or transcriptional activation. Additionally, flies co-infected with both IIV-6 and the Gram-negative bacterium, Erwinia carotovora carotovora, succumb to infection more rapidly than flies singly infected with either the virus or the bacterium. These findings demonstrate how pre-existing infections can have a dramatic and negative effect on secondary infections, and establish a Drosophila model to study confection susceptibility.


Assuntos
Proteínas de Drosophila/imunologia , Drosophila melanogaster/imunologia , Drosophila melanogaster/virologia , Iridovirus/fisiologia , Receptores Toll-Like/imunologia , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Interações Hospedeiro-Patógeno , Imunidade Inata , Iridovirus/genética , Receptores Toll-Like/genética , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia , Replicação Viral
2.
Insect Biochem Mol Biol ; 108: 16-23, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30857831

RESUMO

Fruit flies elicit effective defense responses against numerous microbes. The responses against Gram-negative bacteria are mediated by the Imd pathway, an evolutionarily conserved NF-κB pathway recognizing meso-diaminopimelic acid (DAP)-type peptidoglycan from bacterial cell walls. Several reviews already provide a detailed view of ligand recognition and signal transduction during Imd signaling, but the formation and regulation of the signaling complex immediately downstream of the peptidoglycan-sensing receptors is still elusive. In this review, we focus on the formation of the Imd amyloidal signaling center and post-translational modifications in the assembly and disassembly of the Imd signaling complex.


Assuntos
Amiloide/metabolismo , Drosophila/metabolismo , NF-kappa B/metabolismo , Amiloide/imunologia , Animais , Drosophila/imunologia , Proteínas de Drosophila/metabolismo , Imunidade Inata , Processamento de Proteína Pós-Traducional , Receptores de Superfície Celular/metabolismo , Transdução de Sinais
3.
Immunity ; 47(4): 635-647.e6, 2017 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-29045898

RESUMO

In the Drosophila immune response, bacterial derived diaminopimelic acid-type peptidoglycan binds the receptors PGRP-LC and PGRP-LE, which through interaction with the adaptor protein Imd leads to activation of the NF-κB homolog Relish and robust antimicrobial peptide gene expression. PGRP-LC, PGRP-LE, and Imd each contain a motif with some resemblance to the RIP Homotypic Interaction Motif (RHIM), a domain found in mammalian RIPK proteins forming functional amyloids during necroptosis. Here we found that despite sequence divergence, these Drosophila cryptic RHIMs formed amyloid fibrils in vitro and in cells. Amyloid formation was required for signaling downstream of Imd, and in contrast to the mammalian RHIMs, was not associated with cell death. Furthermore, amyloid formation constituted a regulatable step and could be inhibited by Pirk, an endogenous feedback regulator of this pathway. Thus, diverse sequence motifs are capable of forming amyloidal signaling platforms, and the formation of these platforms may present a regulatory point in multiple biological processes.


Assuntos
Amiloide/imunologia , Proteínas de Transporte/imunologia , Proteínas de Drosophila/imunologia , NF-kappa B/imunologia , Receptores de Superfície Celular/imunologia , Transdução de Sinais/imunologia , Motivos de Aminoácidos/genética , Motivos de Aminoácidos/imunologia , Sequência de Aminoácidos , Amiloide/metabolismo , Animais , Sítios de Ligação/genética , Sítios de Ligação/imunologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Linhagem Celular , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Drosophila melanogaster/imunologia , Feminino , Expressão Gênica/imunologia , Masculino , Microscopia Confocal , Modelos Imunológicos , Mutação , NF-kappa B/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/imunologia , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos
4.
Immunity ; 45(5): 951-953, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27851920

RESUMO

Contradictory to previous reports, Iatsenko et al. (2016) reveal that PGRP-SD regulates the Imd signaling pathway rather than the Toll pathway in Drosophila and shed light on a decade-old mystery of conflicting structural and phenotypic data.


Assuntos
Proteínas de Transporte/química , Imunidade Inata/imunologia , Animais , Drosophila/imunologia , Proteínas de Drosophila/química , Transdução de Sinais
5.
Dev Comp Immunol ; 42(1): 25-35, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23721820

RESUMO

The IMD pathway signaling plays a pivotal role in the Drosophila defense against bacteria. During the last two decades, significant progress has been made in identifying the components and deciphering the molecular mechanisms underlying this pathway, including the means of bacterial sensing and signal transduction. While these findings have contributed to the understanding of the immune signaling in insects, they have also provided new insights in studying the mammalian NF-κB signaling pathways. Here, we summarize the current view of the IMD pathway focusing on how it regulates the humoral immune response of Drosophila.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/imunologia , Infecções/imunologia , Complexo de Endopeptidases do Proteassoma/imunologia , Receptores de Reconhecimento de Padrão/imunologia , Animais , Proteínas de Drosophila/imunologia , Humanos , Imunidade Humoral , Transdução de Sinais/imunologia , Ubiquitinação
6.
EMBO J ; 32(11): 1626-38, 2013 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-23652443

RESUMO

Throughout the animal kingdom, steroid hormones have been implicated in the defense against microbial infection, but how these systemic signals control immunity is unclear. Here, we show that the steroid hormone ecdysone controls the expression of the pattern recognition receptor PGRP-LC in Drosophila, thereby tightly regulating innate immune recognition and defense against bacterial infection. We identify a group of steroid-regulated transcription factors as well as two GATA transcription factors that act as repressors and activators of the immune response and are required for the proper hormonal control of PGRP-LC expression. Together, our results demonstrate that Drosophila use complex mechanisms to modulate innate immune responses, and identify a transcriptional hierarchy that integrates steroid signalling and immunity in animals.


Assuntos
Proteínas de Transporte/metabolismo , Drosophila/imunologia , Ecdisona/metabolismo , Regulação da Expressão Gênica , Transdução de Sinais , Animais , Proteínas de Transporte/genética , Linhagem Celular , Drosophila/genética , Drosophila/microbiologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Enterobacter cloacae/fisiologia , Fatores de Transcrição GATA/genética , Fatores de Transcrição GATA/metabolismo , Imunidade Inata , Estimativa de Kaplan-Meier , Modelos Moleculares , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , Pectobacterium carotovorum/fisiologia , Interferência de RNA , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
7.
PLoS One ; 7(5): e37153, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22606343

RESUMO

Drosophila is a well-established model organism for studying innate immunity because of its high resistance against microbial infections and lack of adaptive immunity. In addition, the immune signaling cascades found in Drosophila are evolutionarily conserved. Upon infection, activation of the immune signaling pathways, Toll and Imd, leads to the expression of multiple immune response genes, such as the antimicrobial peptides (AMPs). Previously, we identified an uncharacterized gene edin among the genes, which were strongly induced upon stimulation with Escherichia coli in Drosophila S2 cells. Edin has been associated with resistance against Listeria monocytogenes, but its role in Drosophila immunity remains elusive. In this study, we examined the role of Edin in the immune response of Drosophila both in vitro and in vivo. We report that edin expression is dependent on the Imd-pathway NF-κB transcription factor Relish and that it is expressed upon infection both in vitro and in vivo. Edin encodes a pro-protein, which is further processed in S2 cells. In our experiments, Edin did not bind microbes, nor did it possess antimicrobial activity to tested microbial strains in vitro or in vivo. Furthermore, edin RNAi did not significantly affect the expression of AMPs in vitro or in vivo. However, edin RNAi flies showed modestly impaired resistance to E. faecalis infection. We conclude that Edin has no potent antimicrobial properties but it appears to be important for E. faecalis infection via an uncharacterized mechanism. Further studies are still required to elucidate the exact role of Edin in the Drosophila immune response.


Assuntos
Proteínas de Drosophila/genética , Proteínas de Drosophila/imunologia , Drosophila melanogaster/genética , Drosophila melanogaster/imunologia , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Peptídeos Catiônicos Antimicrobianos/genética , Peptídeos Catiônicos Antimicrobianos/imunologia , Sequência de Bases , Linhagem Celular , Primers do DNA/genética , Proteínas de Drosophila/antagonistas & inibidores , Drosophila melanogaster/microbiologia , Genes de Insetos , Infecções por Bactérias Gram-Negativas/genética , Infecções por Bactérias Gram-Negativas/imunologia , Imunidade Inata , Dados de Sequência Molecular , Interferência de RNA , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Especificidade da Espécie , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia
8.
Cell Host Microbe ; 11(4): 320-2, 2012 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-22520459

RESUMO

The mechanisms by which epithelial cells distinguish pathogens from commensal microbes have long puzzled us. Now, McEwan et al. (2012) and Dunbar et al. (2012), in this issue of Cell Host & Microbe, demonstrate that in C. elegans, microbial toxin-induced inhibition of host cellular functions, especially blockade of protein translation, activates the effector-triggered immune response dependent on the transcription factor ZIP-2.

9.
Dev Comp Immunol ; 37(1): 9-18, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-21930155

RESUMO

NF-κB signaling is an immune response mechanism remarkably conserved through phylogeny. The genetically tractable model animal Drosophila melanogaster is an important model organism for studying NF-κB signaling in the immune response. Fruit flies have two NF-κB signaling pathways: the Toll and the Imd pathway. Traditional genetic screens have revealed many important aspects about the regulation of Drosophila NF-κB signaling and have helped us to also understand the immune response in humans. For example, the discovery that Toll like receptors are the main immune signaling molecules in mammals was based on work in flies. During the past decade high throughput RNA interference (RNAi)-based screening in cultured Drosophila cells has become a common method for identifying novel genes required for numerous cellular processes including NF-κB signaling. These screens have identified many novel positive and negative regulators of Drosophila NF-κB signaling thus enhancing our understanding of these signaling cascades.


Assuntos
Drosophila/genética , NF-kappa B/metabolismo , Interferência de RNA , Transdução de Sinais/genética , Animais , Drosophila/imunologia , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Receptores Toll-Like/metabolismo
10.
J Leukoc Biol ; 89(5): 649-59, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21208897

RESUMO

Phagocytosis is an ancient cellular process that plays an important role in host defense. In Drosophila melanogaster phagocytic, macrophage-like hemocytes recognize and ingest microbes. We performed an RNAi-based in vitro screen in the Drosophila hemocyte cell line S2 and identified Abi, cpa, cofilin regulator 14-3-3ζ, tlk, CG2765, and CG15609 as mediators of bacterial phagocytosis. Of these identified genes, 14-3-3ζ had an evolutionarily conserved role in phagocytosis: bacterial phagocytosis was compromised when 14-3-3ζ was targeted with RNAi in primary Drosophila hemocytes and when the orthologous genes Ywhab and Ywhaz were silenced in zebrafish and mouse RAW 264.7 cells, respectively. In Drosophila and zebrafish infection models, 14-3-3ζ was required for resistance against Staphylococcus aureus. We conclude that 14-3-3ζ is essential for phagocytosis and microbial resistance in insects and vertebrates.


Assuntos
Proteínas 14-3-3/genética , Fatores de Despolimerização de Actina/genética , Evolução Biológica , Drosophila melanogaster/genética , Drosophila melanogaster/microbiologia , Escherichia coli/patogenicidade , Fagocitose/fisiologia , Proteínas 14-3-3/antagonistas & inibidores , Animais , Células Cultivadas , Drosophila melanogaster/imunologia , Infecções por Escherichia coli/genética , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Feminino , Inativação Gênica , Genes de Insetos , Hemócitos/metabolismo , Masculino , Camundongos , RNA Interferente Pequeno/genética , Peixe-Zebra/genética , Peixe-Zebra/imunologia , Peixe-Zebra/microbiologia
11.
J Immunol ; 184(11): 6188-98, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20421637

RESUMO

Because NF-kappaB signaling pathways are highly conserved in evolution, the fruit fly Drosophila melanogaster provides a good model to study these cascades. We carried out an RNA interference (RNAi)-based genome-wide in vitro reporter assay screen in Drosophila for components of NF-kappaB pathways. We analyzed 16,025 dsRNA-treatments and identified 10 novel NF-kappaB regulators. Of these, nine dsRNA-treatments affect primarily the Toll pathway. G protein-coupled receptor kinase (Gprk)2, CG15737/Toll pathway activation mediating protein, and u-shaped were required for normal Drosomycin response in vivo. Interaction studies revealed that Gprk2 interacts with the Drosophila IkappaB homolog Cactus, but is not required in Cactus degradation, indicating a novel mechanism for NF-kappaB regulation. Morpholino silencing of the zebrafish ortholog of Gprk2 in fish embryos caused impaired cytokine expression after Escherichia coli infection, indicating a conserved role in NF-kappaB signaling. Moreover, small interfering RNA silencing of the human ortholog GRK5 in HeLa cells impaired NF-kappaB reporter activity. Gprk2 RNAi flies are susceptible to infection with Enterococcus faecalis and Gprk2 RNAi rescues Toll(10b)-induced blood cell activation in Drosophila larvae in vivo. We conclude that Gprk2/GRK5 has an evolutionarily conserved role in regulating NF-kappaB signaling.


Assuntos
Proteínas de Drosophila/imunologia , Quinase 2 de Receptor Acoplado a Proteína G/imunologia , Quinase 5 de Receptor Acoplado a Proteína G/metabolismo , Imunidade Inata , NF-kappa B/imunologia , Transdução de Sinais/fisiologia , Animais , Western Blotting , Drosophila , Proteínas de Drosophila/metabolismo , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Quinase 5 de Receptor Acoplado a Proteína G/imunologia , Bactérias Gram-Negativas Quimiolitotróficas/imunologia , Bactérias Gram-Negativas Quimiolitotróficas/metabolismo , Humanos , Imuno-Histoquímica , Imunoprecipitação , NF-kappa B/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Peixe-Zebra
12.
J Immunol ; 180(8): 5413-22, 2008 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-18390723

RESUMO

NF-kappaB transcription factors are involved in evolutionarily conserved signaling pathways controlling multiple cellular processes including apoptosis and immune and inflammatory responses. Immune response of the fruit fly Drosophila melanogaster to Gram-negative bacteria is primarily mediated via the Imd (immune deficiency) pathway, which closely resembles the mammalian TNFR signaling pathway. Instead of cytokines, the main outcome of Imd signaling is the production of antimicrobial peptides. The pathway activity is delicately regulated. Although many of the Imd pathway components are known, the mechanisms of negative regulation are more elusive. In this study we report that a previously uncharacterized gene, pirk, is highly induced upon Gram-negative bacterial infection in Drosophila in vitro and in vivo. pirk encodes a cytoplasmic protein that coimmunoprecipitates with Imd and the cytoplasmic tail of peptidoglycan recognition protein LC (PGRP-LC). RNA interference-mediated down-regulation of Pirk caused Imd pathway hyperactivation upon infection with Gram-negative bacteria, while overexpression of pirk reduced the Imd pathway response both in vitro and in vivo. Furthermore, pirk-overexpressing flies were more susceptible to Gram-negative bacterial infection than wild-type flies. We conclude that Pirk is a negative regulator of the Imd pathway.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/imunologia , Drosophila melanogaster/microbiologia , Enterobacter cloacae/imunologia , Sequência de Aminoácidos , Animais , Proteínas de Transporte/química , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Proteínas de Drosophila/imunologia , Drosophila melanogaster/metabolismo , Enterobacter cloacae/fisiologia , Genes de Insetos , Dados de Sequência Molecular , NF-kappa B/imunologia , NF-kappa B/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Interferência de RNA , Alinhamento de Sequência , Transdução de Sinais
13.
Dev Comp Immunol ; 31(10): 991-1001, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17343912

RESUMO

Fruit flies have effective immune response against Gram-negative bacteria. Upon infection, early JNK-signaling pathway mediated response is followed by the action of the Immune deficiency (Imd) signaling cascade, a Drosophila equivalent of mammalian TNF-receptor pathway, leading to the release of antimicrobial peptides. Recently, Tak1-binding protein 2 (Tab2) and Inhibitor of apoptosis 2 (Iap2) were identified as components of the Imd pathway. In this study, we carried out a genome-wide kinetic analysis of the role of Tab2 and Iap2 for immune response in Drosophila S2 cells using oligonucleotide microarrays. Tab2 RNAi abolished the induction of all immune response genes in S2 cells indicating its requirement for signaling both via the Imd and the JNK pathway. The role of Iap2 was more specific. Kinetic analysis indicated that Iap2 is required to sustain antimicrobial peptide gene expression in S2 cells. Furthermore, inactivation of Iap2 by RNAi resulted in impaired microbial resistance in Drosophila in vivo.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila melanogaster/imunologia , Proteínas Inibidoras de Apoptose/fisiologia , Transdução de Sinais/imunologia , Animais , Peptídeos Catiônicos Antimicrobianos/biossíntese , Peptídeos Catiônicos Antimicrobianos/genética , Linhagem Celular , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Imunidade Inata/genética , Proteínas Inibidoras de Apoptose/genética , Testes de Sensibilidade Microbiana , Transdução de Sinais/genética
14.
J Biol Chem ; 281(20): 14370-5, 2006 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-16531407

RESUMO

Double-stranded RNA (dsRNA) fragments are readily internalized and processed by Drosophila S2 cells, making these cells a widely used tool for the analysis of gene function by gene silencing through RNA interference (RNAi). The underlying mechanisms are insufficiently understood. To identify components of the RNAi pathway in S2 cells, we developed a screen based on rescue from RNAi-induced lethality. We identified Argonaute 2, a core component of the RNAi machinery, and three gene products previously unknown to be involved in RNAi in Drosophila: DEAD-box RNA helicase Belle, 26 S proteasome regulatory subunit 8 (Pros45), and clathrin heavy chain, a component of the endocytic machinery. Blocking endocytosis in S2 cells impaired RNAi, suggesting that dsRNA fragments are internalized by receptor-mediated endocytosis. Indeed, using a candidate gene approach, we identified two Drosophila scavenger receptors, SR-CI and Eater, which together accounted for more than 90% of the dsRNA uptake into S2 cells. When expressed in mammalian cells, SR-CI was sufficient to mediate internalization of dsRNA fragments. Our data provide insight into the mechanism of dsRNA internalization by Drosophila cells. These results have implications for dsRNA delivery into mammalian cells.


Assuntos
Endocitose , RNA de Cadeia Dupla/química , Receptores Depuradores/metabolismo , Animais , Linhagem Celular , Drosophila melanogaster , Biblioteca Gênica , Inativação Gênica , Técnicas Genéticas , Luciferases/metabolismo , Interferência de RNA
15.
EMBO J ; 24(19): 3423-34, 2005 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-16163390

RESUMO

The Imd signaling cascade, similar to the mammalian TNF-receptor pathway, controls antimicrobial peptide expression in Drosophila. We performed a large-scale RNAi screen to identify novel components of the Imd pathway in Drosophila S2 cells. In all, 6713 dsRNAs from an S2 cell-derived cDNA library were analyzed for their effect on Attacin promoter activity in response to Escherichia coli. We identified seven gene products required for the Attacin response in vitro, including two novel Imd pathway components: inhibitor of apoptosis 2 (Iap2) and transforming growth factor-activated kinase 1 (TAK1)-binding protein (TAB). Iap2 is required for antimicrobial peptide response also by the fat body in vivo. Both these factors function downstream of Imd. Neither TAB nor Iap2 is required for Relish cleavage, but may be involved in Relish nuclear localization in vitro, suggesting a novel mode of regulation of the Imd pathway. Our results show that an RNAi-based approach is suitable to identify genes in conserved signaling cascades.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Proteínas Inibidoras de Apoptose/genética , Transdução de Sinais/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Western Blotting , Células Cultivadas , DNA/genética , Primers do DNA , Proteínas de Drosophila/imunologia , Drosophila melanogaster/imunologia , Drosophila melanogaster/microbiologia , Eletroforese em Gel de Poliacrilamida , Escherichia coli/imunologia , Biblioteca Gênica , Imuno-Histoquímica , Proteínas Inibidoras de Apoptose/metabolismo , Proteínas de Insetos/metabolismo , Luciferases , Modelos Biológicos , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Transdução de Sinais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...