Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Susanna Valanne is active.

Publication


Featured researches published by Susanna Valanne.


Journal of Immunology | 2011

The Drosophila Toll signaling pathway.

Susanna Valanne; Jing-Huan Wang; Mika Rämet

The identification of the Drosophila melanogaster Toll pathway cascade and the subsequent characterization of TLRs have reshaped our understanding of the immune system. Ever since, Drosophila NF-κB signaling has been actively studied. In flies, the Toll receptors are essential for embryonic development and immunity. In total, nine Toll receptors are encoded in the Drosophila genome, including the Toll pathway receptor Toll. The induction of the Toll pathway by Gram-positive bacteria or fungi leads to the activation of cellular immunity as well as the systemic production of certain antimicrobial peptides. The Toll receptor is activated when the proteolytically cleaved ligand Spatzle binds to the receptor, eventually leading to the activation of the NF-κB factors Dorsal-related immunity factor or Dorsal. In this study, we review the current literature on the Toll pathway and compare the Drosophila and mammalian NF-κB pathways.


The EMBO Journal | 2005

Inhibitor of apoptosis 2 and TAK1-binding protein are components of the Drosophila Imd pathway

Anni Kleino; Susanna Valanne; Johanna Ulvila; Jenni Kallio; Henna Myllymäki; Heidi Enwald; Svenja Stöven; Mickael Poidevin; Ryu Ueda; Dan Hultmark; Bruno Lemaitre; Mika Rämet

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.


Journal of Immunology | 2008

Pirk Is a Negative Regulator of the Drosophila Imd Pathway

Anni Kleino; Henna Myllymäki; Jenni Kallio; Leena-Maija Vanha-aho; Kaisa E. Oksanen; Johanna Ulvila; Dan Hultmark; Susanna Valanne; Mika Rämet

NF-κB 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.


Journal of Immunology | 2014

The Drosophila Imd Signaling Pathway

Henna Myllymäki; Susanna Valanne; Mika Rämet

The fruit fly, Drosophila melanogaster, has helped us to understand how innate immunity is activated. In addition to the Toll receptor and the Toll signaling pathway, the Drosophila immune response is regulated by another evolutionarily conserved signaling cascade, the immune deficiency (Imd) pathway, which activates NF-κB. In fact, the Imd pathway controls the expression of most of the antimicrobial peptides in Drosophila; thus, it is indispensable for normal immunity in flies. In this article, we review the current literature on the Drosophila Imd pathway, with special emphasis on its role in the (patho)physiology of different organs. We discuss the systemic response, as well as local responses, in the epithelial and mucosal surfaces and the nervous system.


Journal of Immunology | 2010

Genome-Wide RNA Interference in Drosophila Cells Identifies G Protein-Coupled Receptor Kinase 2 as a Conserved Regulator of NF-κB Signaling

Susanna Valanne; Henna Myllymäki; Jenni Kallio; Martin R. Schmid; Anni Kleino; Astrid Murumägi; Laura Airaksinen; Tapio Kotipelto; Meri Kaustio; Johanna Ulvila; Shiva Seyedoleslami Esfahani; Ylva Engström; Olli Silvennoinen; Dan Hultmark; Mataleena Parikka; Mika Rämet

Because NF-κB 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-κB pathways. We analyzed 16,025 dsRNA-treatments and identified 10 novel NF-κB 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 IκB homolog Cactus, but is not required in Cactus degradation, indicating a novel mechanism for NF-κB 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-κB signaling. Moreover, small interfering RNA silencing of the human ortholog GRK5 in HeLa cells impaired NF-κB reporter activity. Gprk2 RNAi flies are susceptible to infection with Enterococcus faecalis and Gprk2 RNAi rescues Toll10b-induced blood cell activation in Drosophila larvae in vivo. We conclude that Gprk2/GRK5 has an evolutionarily conserved role in regulating NF-κB signaling.


The FASEB Journal | 2010

Eye transformer is a negative regulator of Drosophila JAK/STAT signaling

Jenni Kallio; Henna Myllymäki; Juha Grönholm; Morag Armstrong; Leena-Maija Vanha-aho; Leena Mäkinen; Olli Silvennoinen; Susanna Valanne; Mika Rämet

JAK/STAT signaling pathway is evolutionarily conserved and tightly regulated. We carried out a reporter‐based genome‐wide RNAi in vitro screen to identify genes that regulate Drosophila JAK/STAT pathway and found 5 novel regulators. Of these, CG14225 is a negative regulator structurally related to the Drosophila JAK/STAT pathway receptor Domeless, especially in the extracellular domain, and to the mammalian IL‐6 receptor and the signal transducer gp130. CG14225 coimmunoprecipitates with Domeless and its associated kinase hopscotch in S2 cells. CG14225 RNAi caused hyperphosphorylation of the transcription factor Stat92E in S2 cells on stimulation with the Drosophila JAK/STAT pathway ligand unpaired. CG14225 RNAi in vivo hyperactivated JAK/STAT target genes on septic injury and enhanced unpaired‐induced eye overgrowth, and was thus named the eye transformer (ET). In the gastrointestinal infection model, where JAK/STAT signaling is important for stem cell renewal, CG14225/ET RNAi was protective in vivo. In conclusion, we have identified ET as a novel negative regulator of the Drosophila JAK/STAT pathway both in vitro and in vivo, and it functions in regulating Stat92E phosphorylation.—Kallio, J., Myllymäki, H., Grönholm, J., Armstrong, M., Vanha‐aho, L.‐M., Mäkinen, L., Silvennoinen, O., Valanne, S., Rämet, M. Eye transformer is a negative regulator of Drosophila JAK/STAT signaling. FASEBJ. 24, 4467–4479 (2010). www.fasebj.org


The FASEB Journal | 2012

Not4 enhances JAK/STAT pathway-dependent gene expression in Drosophila and in human cells

Juha Grönholm; Meri Kaustio; Henna Myllymäki; Jenni Kallio; Juha Saarikettu; Jesper Kronhamn; Susanna Valanne; Olli Silvennoinen; Mika Rämet

The JAK/STAT pathway is essential for organogenesis, innate immunity, and stress responses in Drosophila melanogaster. The JAK/STAT pathway and its associated regulators have been highly conserved in evolution from flies to humans. We have used a genome‐wide RNAi screen in Drosophila S2 cells to identify regulators of the JAK/STAT pathway, and here we report the characterization of Not4 as a positive regulator of the JAK/STAT pathway. Overexpression of Not4 enhanced Stat92E‐mediated gene responses in vitro and in vivo in Drosophila. Specifically, Not4 increased Stat92E‐mediated reporter gene activation in S2 cells; and in flies, Not4 overexpression resulted in an 8‐fold increase in Turandot M (TotM) and in a 4‐fold increase in Turandot A (TotA) stress gene activation when compared to wild‐type flies. Drosophila Not4 is structurally related to human CNOT4, which was found to regulate interferon‐γ‐ and interleukin‐4‐induced STAT‐mediated gene responses in human HeLa cells. Not4 was found to coimmunoprecipitate with Stat92E but not to affect tyrosine phosphorylation of Stat92E in Drosophila cells. However, Not4 is required for binding of Stat92E to its DNA recognition sequence in the TotM gene promoter. In summary, Not4/CNOT4 is a novel positive regulator of the JAK/STAT pathway in Drosophila and in humans.—Grönholm, J., Kaustio, M., Myllymäki, H., Kallio, J., Saarikettu, J., Kronhamn, J., Valanne, S., Silvennoinen, O., Rämet, M. Not4 enhances JAK/STAT pathway‐dependent gene expression in Drosophila and in human cells. FASEB J. 26, 1239‐1250 (2012). www.fasebj.org


World Journal of Biological Chemistry | 2010

Drosophila as a model for antiviral immunity

Jing-Huan Wang; Susanna Valanne; Mika Rämet

The fruit fly Drosophila melanogaster has been successfully used to study numerous biological processes including immune response. Flies are naturally infected with more than twenty RNA viruses making it a valid model organism to study host-pathogen interactions during viral infections. The Drosophila antiviral immunity includes RNA interference, activation of the JAK/STAT and other signaling cascades and other mechanisms such as autophagy and interactions with other microorganisms. Here we review Drosophila as an immunological research model as well as recent advances in the field of Drosophila antiviral immunity.


Developmental and Comparative Immunology | 2012

Large-scale RNAi screens add both clarity and complexity to Drosophila NF-κB signaling.

Susanna Valanne; Jenni Kallio; Anni Kleino; Mika Rämet

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.


PLOS Pathogens | 2015

Edin Expression in the Fat Body Is Required in the Defense Against Parasitic Wasps in Drosophila melanogaster

Leena-Maija Vanha-aho; Ines Anderl; Laura Vesala; Dan Hultmark; Susanna Valanne; Mika Rämet

The cellular immune response against parasitoid wasps in Drosophila involves the activation, mobilization, proliferation and differentiation of different blood cell types. Here, we have assessed the role of Edin (elevated during infection) in the immune response against the parasitoid wasp Leptopilina boulardi in Drosophila melanogaster larvae. The expression of edin was induced within hours after a wasp infection in larval fat bodies. Using tissue-specific RNAi, we show that Edin is an important determinant of the encapsulation response. Although edin expression in the fat body was required for the larvae to mount a normal encapsulation response, it was dispensable in hemocytes. Edin expression in the fat body was not required for lamellocyte differentiation, but it was needed for the increase in plasmatocyte numbers and for the release of sessile hemocytes into the hemolymph. We conclude that edin expression in the fat body affects the outcome of a wasp infection by regulating the increase of plasmatocyte numbers and the mobilization of sessile hemocytes in Drosophila larvae.

Collaboration


Dive into the Susanna Valanne's collaboration.

Top Co-Authors

Avatar

Mika Rämet

Oulu University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge