Network


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

Hotspot


Dive into the research topics where Stine Granum is active.

Publication


Featured researches published by Stine Granum.


Genes and Immunity | 2011

Exploring the CLEC16A gene reveals a MS-associated variant with correlation to the relative expression of CLEC16A isoforms in thymus.

Inger-Lise Mero; Maria Ban; Åslaug R. Lorentzen; Cathrine Smestad; Elisabeth G. Celius; Sæther H; Saeedi H; Marte K. Viken; Beate Skinningsrud; Dag E. Undlien; Jan Harald Aarseth; Kjell-Morten Myhr; Stine Granum; Anne Spurkland; Stephen Sawcer; A. Compston; Benedicte A. Lie; Hanne F. Harbo

Genomewide association studies have implicated the CLEC16A gene in several autoimmune diseases, including multiple sclerosis (MS) and type 1 diabetes. However, the most associated single-nucleotide polymorphism (SNP) varies, and causal variants are still to be defined. In MS, two SNPs in partial linkage disequilibrium with each other, rs6498169 and rs12708716, have been validated at genomewide significance level. To explore the CLEC16A association in MS in more detail, we genotyped 57 SNPs in 807 Norwegian MS patients and 1027 Norwegian controls. Six highly associated SNPs emerged and were then replicated in two large independent sample sets (Norwegian and British), together including 1153 MS trios, 2308 MS patients and 4044 healthy controls. In combined analyses, SNP rs12708716 gave the strongest association signal in MS (P=5.3 × 10−8, odds ratio 1.18, 95% confidence interval=1.11–1.25), and was found to be superior to the other SNP associations in conditional logistic regression analyses. Expression analysis revealed that rs12708716 genotype was significantly associated with the relative expression levels of two different CLEC16A transcripts in thymus (P=0.004), but not in blood, possibly implying a thymus- or cell-specific splice regulation.


European Journal of Immunology | 2005

The C terminus of T cell-specific adapter protein (TSAd) is necessary for TSAd-mediated inhibition of Lck activity.

Vibeke Sundvold-Gjerstad; Stine Granum; Tomas Mustelin; Thorny Cecilie Bie Andersen; Tone Berge; Michael J. Shapiro; Virginia Smith Shapiro; Anne Spurkland; Tor Lea

T cell‐specific adapter protein (TSAd), encoded by the SH2D2A gene, is expressed in activated T cells. The function of TSAd is as yet unknown. We previously showed that TSAd may modulate T cell receptor‐triggered signaling events. TSAd contains a Src homology (SH)2 domain, ten tyrosines and a C‐terminal proline‐rich region. Here, we show that human TSAd interacts with Lck through the Lck SH2 and SH3 domains and is a substrate for Lck. The TSAd C terminus, including the proline‐rich region and five tyrosines, is both necessary and sufficient for TSAd interaction with and phosphorylation by Lck. Expression of TSAd in Jurkat TAg cells results in hyperphosphorylation of endogenous Lck on Y394 and to an even larger extent on Y505, resulting in a reduced Y394/Y505 phosphorylation ratio in these cells. Furthermore, full‐length TSAd, but not TSAd lacking the C terminus, inhibits the hyperactive Lck Y505F mutant when both are expressed in Jurkat T cells. In contrast, expression of the TSAd C terminus alone is sufficient to inhibit Lck Y505F in phosphorylating its substrates in Jurkat T cells. Our results indicate that the TSAd C terminus is essential for inhibition of Lck activity by TSAd, and suggest a mechanism for how TSAd may inhibit early T cell activation events.


PLOS ONE | 2010

T Cell Specific Adapter Protein (TSAd) Interacts with Tec Kinase ITK to Promote CXCL12 Induced Migration of Human and Murine T Cells

Tone Berge; Vibeke Sundvold-Gjerstad; Stine Granum; Thorny Cecilie Bie Andersen; Gunn B. Holthe; Lena Claesson-Welsh; Amy H. Andreotti; Marit Inngjerdingen; Anne Spurkland

Background The chemokine CXCL12/SDF-1α interacts with its G-protein coupled receptor CXCR4 to induce migration of lymphoid and endothelial cells. T cell specific adapter protein (TSAd) has been found to promote migration of Jurkat T cells through interaction with the G protein β subunit. However, the molecular mechanisms for how TSAd influences cellular migration have not been characterized in detail. Principal Findings We show that TSAd is required for tyrosine phosphorylation of the Lck substrate IL2-inducible T cell kinase (Itk). Presence of Itk Y511 was necessary to boost TSAds effect on CXCL12 induced migration of Jurkat T cells. In addition, TSAds ability to promote CXCL12-induced actin polymerization and migration of Jurkat T lymphocytes was dependent on the Itk-interaction site in the proline-rich region of TSAd. Furthermore, TSAd-deficient murine thymocytes failed to respond to CXCL12 with increased Itk phosphorylation, and displayed reduced actin polymerization and cell migration responses. Conclusion We propose that TSAd, through its interaction with both Itk and Lck, primes Itk for Lck mediated phosphorylation and thereby regulates CXCL12 induced T cell migration and actin cytoskeleton rearrangements.


Journal of Biological Chemistry | 2008

Modulation of Lck Function through Multisite Docking to T Cell-specific Adapter Protein

Stine Granum; Thorny Cesilie Bie Andersen; Morten Sørlie; Marit Jørgensen; Lise Koll; Tone Berge; Tor Lea; Burkhard Fleckenstein; Anne Spurkland; Vibeke Sundvold-Gjerstad

T cell-specific adapter protein (TSAd), encoded by the SH2D2A gene, interacts with Lck through its C terminus and thus modulates Lck activity. Here we mapped Lck phosphorylation and interaction sites on TSAd and evaluated their functional importance. The three C-terminal TSAd tyrosines Tyr280, Tyr290, and Tyr305 were phosphorylated by Lck and functioned as docking sites for the Lck Src homology 2 (SH2) domain. Binding affinities of the TSAd Tyr(P)280 and Tyr(P)290 phosphopeptides to the isolated Lck SH2 domain were similar to that observed for the Lck Tyr(P)505 phosphopeptide, whereas the TSAd Tyr(P)305 peptide displayed a 10-fold higher affinity. The proline-rich Lck SH3-binding site on TSAd as well as the Lck SH2 domain were required for efficient tyrosine phosphorylation of TSAd by Lck. Interaction sites on TSAd for both Lck SH2 and Lck SH3 were necessary for TSAd-mediated modulation of proximal TCR signaling events. We found that 20–30% of TSAd molecules are phosphorylated in activated T cells and that the proportion of TSAd to Lck molecules in such cells is ∼1:1. Therefore, in activated T cells, a considerable number of Lck molecules may potentially be engaged by TSAd. In conclusion, Lck binds to TSAd prolines and phosphorylates and interacts with the three C-terminal TSAd tyrosines. We propose that through multivalent interactions with Lck, TSAd diverts Lck from phosphorylating other substrates, thus modulating its functional activity through substrate competition.


BMC Immunology | 2006

Structure function analysis of SH2D2A isoforms expressed in T cells reveals a crucial role for the proline rich region encoded by SH2D2A exon 7

Stine Granum; Vibeke Sundvold-Gjerstad; Ke-Zheng Dai; Kristin M. Kolltveit; Kjersti Hildebrand; Henrik S. Huitfeldt; Tor Lea; Anne Spurkland

BackgroundThe activation induced T cell specific adapter protein (TSAd), encoded by SH2D2A, interacts with and modulates Lck activity. Several transcript variants of TSAd mRNA exist, but their biological significance remains unknown. Here we examined expression of SH2D2A transcripts in activated CD4+ T cells and used the SH2D2A variants as tools to identify functionally important regions of TSAd.ResultsTSAd was found to interact with Lck in human CD4+ T cells ex vivo. Three interaction modes of TSAd with Lck were identified. TSAd aa239–256 conferred binding to the Lck-SH3 domain, whereas one or more of the four tyrosines within aa239–334 encoded by SH2D2A exon 7 was found to confer interaction with the Lck-SH2-domain. Finally the TSAd-SH2 domain was found to interact with Lck. The SH2D2A exon 7 encoding TSAd aa 239–334 was found to harbour information essential not only for TSAd interaction with Lck, but also for TSAd modulation of Lck activity and translocation of TSAd to the nucleus. All five SH2D2A transcripts were found to be expressed in CD3 stimulated CD4+ T cells.ConclusionThese data show that TSAd and Lck may interact through several different domains and that Lck TSAd interaction occurs in CD4+ T cells ex vivo. Alternative splicing of exon 7 encoding aa239–334 results in loss of the majority of protein interaction motives of TSAd and yields truncated TSAd molecules with altered ability to modulate Lck activity. Whether TSAd is regulated through differential alternative splicing of the SH2D2A transcript remains to be determined.


Science Signaling | 2014

The kinase Itk and the adaptor TSAd change the specificity of the kinase Lck in T cells by promoting the phosphorylation of Tyr192

Stine Granum; Vibeke Sundvold-Gjerstad; Ramakrishna Prabhu Gopalakrishnan; Tone Berge; Lise Koll; Greger Abrahamsen; Morten Sørlie; Anne Spurkland

Phosphorylation of a tyrosine residue in the SH2 domain of the kinase Lck alters its substrate specificity in T cells. Switching Lck’s Attention The Src family kinase (SFK) member Lck has distinct early and late functions in T cell signaling. Early after activation of the T cell receptor (TCR), Lck mediates signals close in space and time to the TCR; later, Lck regulates the actin cytoskeleton, reorganization of which is required for the stable interaction between T cells and antigen-presenting cells (APCs). Lck activity is determined by the balance in the phosphorylation status of two tyrosine residues, one activating and the other inhibitory. Granum et al. identified a third tyrosine residue, Tyr192, which was phosphorylated in response to TCR stimulation. T cells expressing a mutant Lck lacking this tyrosine failed to form stable interactions with APCs. Lck phosphorylated at Tyr192 preferentially associated with regulators of the actin cytoskeleton. Thus, phosphorylation of Tyr192 in Lck enables Lck to switch its attention from the TCR to the actin cytoskeleton, thereby fulfilling different functions at different stages of T cell activation. The substrate specificity of Src family kinases (SFKs) is partly determined by their Src homology 2 (SH2) domains. Thus, transient alterations in the SH2 domain of SFKs might change their binding partners and affect intracellular signaling pathways. Lck is an SFK that is central to the initiation of T cell activation in response to ligand binding to the T cell receptor (TCR) and is also critical for later signaling processes. The kinase activity of Lck requires both the phosphorylation of an activating tyrosine residue and the dephosphorylation of an inhibitory tyrosine residue. We found that a third conserved tyrosine phosphorylation site (Tyr192) within the SH2 domain of Lck was required for proper T cell activation and formation of cell-cell conjugates between T cells and antigen-presenting cells. Through phosphopeptide arrays and biochemical assays, we identified several regulators of the actin cytoskeleton that preferentially bound to Lck phosphorylated at Tyr192 compared to Lck that was not phosphorylated at this site. Two of these phosphorylation-dependent binding partners, the kinase Itk (interleukin-2–inducible Tec kinase) and the adaptor protein TSAd (T cell–specific adaptor), promoted the TCR-dependent phosphorylation of Lck at Tyr192. Our data suggest that phosphorylation transiently alters SH2 domain specificity and provide a potential mechanism whereby SFKs may be rewired from one signaling program to another to enable appropriate cell activation.


Cell Communication and Signaling | 2015

T cell specific adaptor protein (TSAd) promotes interaction of Nck with Lck and SLP-76 in T cells

Cecilie Dahl Hem; Vibeke Sundvold-Gjerstad; Stine Granum; Lise Koll; Greger Abrahamsen; László Buday; Anne Spurkland

BackgroundThe Lck and Src binding adaptor protein TSAd (T cell specific adaptor) regulates actin polymerization in T cells and endothelial cells. The molecular details as to how TSAd regulates this process remain to be elucidated.ResultsTo identify novel interaction partners for TSAd, we used a scoring matrix-assisted ligand algorithm (SMALI), and found that the Src homology 2 (SH2) domain of the actin regulator Non-catalytic region of tyrosine kinase adaptor protein (Nck) potentially binds to TSAd phosphorylated on Tyr280 (pTyr280) and pTyr305. These predictions were confirmed by peptide array analysis, showing direct binding of recombinant Nck SH2 to both pTyr280 and pTyr305 on TSAd. In addition, the SH3 domains of Nck interacted with the proline rich region (PRR) of TSAd. Pull-down and immunoprecipitation experiments further confirmed the Nck-TSAd interactions through Nck SH2 and SH3 domains. In line with this Nck and TSAd co-localized in Jurkat cells as assessed by confocal microscopy and imaging flow cytometry. Co-immunoprecipitation experiments in Jurkat TAg cells lacking TSAd revealed that TSAd promotes interaction of Nck with Lck and SLP-76, but not Vav1. TSAd expressing Jurkat cells contained more polymerized actin, an effect dependent on TSAd exon 7, which includes interactions sites for both Nck and Lck.ConclusionsTSAd binds to and co-localizes with Nck. Expression of TSAd increases both Nck-Lck and Nck-SLP-76 interaction in T cells. Recruitment of Lck and SLP-76 to Nck by TSAd could be one mechanism by which TSAd promotes actin polymerization in activated T cells.


Scandinavian Journal of Immunology | 2017

CD6 and Linker of Activated T cells are potential interaction partners for T cell Specific Adaptor protein

Cecilie Dahl Hem; Marianne Ekornhol; Stine Granum; Vibeke Sundvold-Gjerstad; Anne Spurkland

The T cell‐specific adaptor protein (TSAd) contains several protein interaction domains, and is merging as a modulator of T cell activation. Several interaction partners for the TSAd proline‐rich region and phosphotyrosines have been identified, including the Src and Tec family kinases lymphocyte‐specific protein tyrosine kinase and interleukin 2‐inducible T cell kinase. Via its Src homology 2 (SH2) domain, TSAd may thus function as a link between these enzymes and other signalling molecules. However, few binding partners to the TSAd SH2 domain in T cells are hitherto known. Through the use of in silico ligand prediction, peptide spot arrays, pull‐down and immunoprecipitation experiments, we here report novel interactions between the TSAd SH2 domain and CD6 phosphotyrosine (pTyr)629 and linker of activated T cells (LAT) pTyr171, pTyr191 and pTyr226.


European Journal of Oral Sciences | 2010

Expression of the T‐cell‐specific adapter protein in oral epithelium

Kristin M. Kolltveit; Olav Schreurs; Janet Østrem; Tine M. Søland; Cuong Khuu; Tone Berge; Edward B. Messelt; Katsuhiko Hayashi; Stine Granum; Anne Spurkland; Karl Schenck

The multifunctional T-cell-specific adapter protein (TSAd) was originally described in T cells but is also expressed in epithelial cells from the respiratory tract and in endothelium. In this study, we found expression of TSAd messenger RNA (mRNA) and protein in both human and murine oral mucosal epithelium as well as in human primary oral keratinocyte cell cultures. In TSAd(-/-) mice, the mucosa and skin appeared macroscopically normal, but severe disturbances were observed in the fine structures of the basal membrane and intercellular epithelial spaces upon analysis using transmission electron microscopy. Oral epithelial cells from TSAd(-/-) mice displayed decreased migration compared with cells from wild-type mice, whereas overexpression of TSAd in a human epithelial cell line resulted in impaired proliferation. This study is the first to show that TSAd is expressed in normal oral mucosa, that it is important for the normal ultrastructural morphology of the epithelium and the basal membrane, and that it is involved in the migration and proliferation of oral keratinocytes.


PLOS ONE | 2012

SH2D2A Modulates T Cell Mediated Protection to a B Cell Derived Tumor in Transgenic Mice

Tone Berge; Ingrid Helene Bø Grønningsæter; Kristina Berg Lorvik; Greger Abrahamsen; Stine Granum; Vibeke Sundvold-Gjerstad; Alexandre Corthay; Bjarne Bogen; Anne Spurkland

Background T cell specific adapter protein (TSAd), encoded by the SH2D2A gene, modulates signaling downstream of the T cell receptor (TCR). Young, unchallenged SH2D2A-deficient C57BL/6 mice exhibit a relatively normal immune phenotype. To address whether SH2D2A regulates physiologic immune responses, SH2D2A-deficient TCR-transgenic BALB/c mice were generated. The transgenic TCR recognizes a myeloma-derived idiotypic (Id) peptide in the context of the major histocompatibility complex (MHC) class II molecule I-Ed, and confers T cell mediated resistance to transplanted multiple myeloma development in vivo. Principal Findings The immune phenotype of SH2D2A-deficient C57BL/6 and BALB/c mice did not reveal major differences compared to the corresponding wild type mice. When challenged with myeloma cells, Id-specific TCR-transgenic BALB/c mice lacking SH2D2A displayed increased resistance towards tumor development. Tumor free TCR-transgenic SH2D2A-deficient mice had higher numbers of Id-specific single positive CD4+ thymocytes compared to TCR-transgenic wild-type mice. Conclusion Our results suggest a modulatory role for SH2D2A in T cell mediated immune surveillance of cancer. However, it remains to be established whether its effect is T-cell intrinsic. Further studies are required to determine whether targeting SH2D2A function in T cells may be a potential adjuvant in cancer immunotherapy.

Collaboration


Dive into the Stine Granum's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tone Berge

Oslo University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tor Lea

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Morten Sørlie

Norwegian University of Life Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge