Network


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

Hotspot


Dive into the research topics where Anders Kallin is active.

Publication


Featured researches published by Anders Kallin.


Journal of Biological Chemistry | 2005

JAK1 and Tyk2 Activation by the Homologous Polycythemia Vera JAK2 V617F Mutation CROSS-TALK WITH IGF1 RECEPTOR

Judith Staerk; Anders Kallin; Jean-Baptiste Demoulin; William Vainchenker; Stefan N. Constantinescu

The majority of polycythemia vera (PV) patients harbor a unique somatic mutation (V617F) in the pseudokinase domain of JAK2, which leads to constitutive signaling. Here we show that the homologous mutations in JAK1 (V658F) and in Tyk2 (V678F) lead to constitutive activation of these kinases. Their expression induces autonomous growth of cytokine-dependent cells and constitutive activation of STAT5, STAT3, mitogen-activated protein kinase, and Akt signaling in Ba/F3 cells. The mutant JAKs exhibit constitutive signaling also when expressed in fibrosarcoma cells deficient in JAK proteins. Expression of the JAK2 V617F mutant renders Ba/F3 cells hypersensitive to insulin-like growth factor 1 (IGF1), which is a hallmark of PV erythroid progenitors. Upon selection of Ba/F3 cells for autonomous growth induced by the JAK2 V617F mutant, cells respond to IGF1 by activating STAT5, STAT3, Erk1/2, and Akt on top of the constitutive activation characteristic of autonomous cells. The synergic effect on proliferation and STAT activation appears specific to the JAK2 V617F mutant. Our results show that the homologous V617F mutation induces activation of JAK1 and Tyk2, suggesting a common mechanism of activation for the JAK1, JAK2, and Tyk2 mutants. JAK3 is not activated by the homologous mutation M592F, despite the presence of the conserved GVC preceding sequence. We suggest that mutations in the JAK1 and Tyk2 genes may be identified as initial molecular defects in human cancers and autoimmune diseases.


Nucleic Acids Research | 2010

Transcription factor regulation can be accurately predicted from the presence of target gene signatures in microarray gene expression data

Ahmed Essaghir; Federica Toffalini; Laurent Knoops; Anders Kallin; Jacques van Helden; Jean-Baptiste Demoulin

Deciphering transcription factor networks from microarray data remains difficult. This study presents a simple method to infer the regulation of transcription factors from microarray data based on well-characterized target genes. We generated a catalog containing transcription factors associated with 2720 target genes and 6401 experimentally validated regulations. When it was available, a distinction between transcriptional activation and inhibition was included for each regulation. Next, we built a tool (www.tfacts.org) that compares submitted gene lists with target genes in the catalog to detect regulated transcription factors. TFactS was validated with published lists of regulated genes in various models and compared to tools based on in silico promoter analysis. We next analyzed the NCI60 cancer microarray data set and showed the regulation of SOX10, MITF and JUN in melanomas. We then performed microarray experiments comparing gene expression response of human fibroblasts stimulated by different growth factors. TFactS predicted the specific activation of Signal transducer and activator of transcription factors by PDGF-BB, which was confirmed experimentally. Our results show that the expression levels of transcription factor target genes constitute a robust signature for transcription factor regulation, and can be efficiently used for microarray data mining.


Journal of Biological Chemistry | 2004

Platelet-derived growth factor stimulates membrane lipid synthesis through activation of phosphatidylinositol 3-kinase and sterol regulatory element-binding proteins

Jean-Baptiste Demoulin; Johan Ericsson; Anders Kallin; Charlotte Rorsman; Lars Rönnstrand; Carl-Henrik Heldin

We analyzed the transcriptional program elicited by stimulation of normal human fibroblasts with platelet-derived growth factor (PDGF) using cDNA microarrays. 103 significantly regulated transcripts that had not been previously linked to PDGF signaling were identified. Among them, a cluster of genes involved in fatty acid and cholesterol biosynthesis, including stearoyl-CoA desaturase (SCD), fatty acid synthase, and hydroxymethylglutaryl-CoA synthase (HMGCS), was up-regulated by PDGF after 24 h of treatment, and their expression correlated with increased membrane lipid production. These genes are known to be controlled by sterol regulatory element-binding proteins (SREBP). PDGF increased the amount of mature SREBP-1 and regulated the promoters of SCD and HMGCS in an SREBP-dependent manner. In line with these results, blocking SREBP processing by addition of 25-hydroxycholesterol blunted the effects of PDGF on lipogenic enzymes. SREBP activation was dependent on the phosphatidylinositol 3-kinase (PI3K) pathway, as judged from the effects of the inhibitor LY294002 and mutation of the PDGFβ receptor tyrosines that bind the PI3K adaptor subunit p85. Fibroblast growth factors (FGF-2 and FGF-4) and other growth factors mimicked the effects of PDGF on NIH3T3 and human fibroblasts. In conclusion, our results suggest that growth factors induce membrane lipid synthesis via the activation SREBP and PI3K.


Oncogene | 1999

SHP-2 binds to Tyr763 and Tyr1009 in the PDGF beta-receptor and mediates PDGF-induced activation of the Ras/MAP kinase pathway and chemotaxis.

Lars Rönnstrand; Ann-Kristin Arvidsson; Anders Kallin; Charlotte Rorsman; Ulf Hellman; Ulla Engström; Christer Wernstedt; Carl-Henrik Heldin

Activation of the β-receptor for platelet-derived growth factor (PDGF) by its ligand leads to autophosphorylation on a number of tyrosine residues. Here we show that Tyr763 in the kinase insert region is a novel autophosphorylation site, which after phosphorylation binds the protein tyrosine phosphatase SHP-2. SHP-2 has also previously been shown to bind to phosphorylated Tyr1009 in the PDGF β-receptor. Porcine aortic endothelial (PAE) cells transfected with a PDGF β-receptor in which Tyr763 and Tyr1009 were mutated to phenylalanine residues failed to associate with SHP-2 after ligand stimulation. Morover, PDGF-BB-induced Ras GTP-loading and Erk2 activation were severely compromised in the receptor mutant. Whereas the mitogenic response to PDGF-BB remained at the same level as in cells expressing wild-type PDGF β-receptor, chemotaxis induced by PDGF-BB was significantly decreased in the case of the Y763F/Y1009F mutant cells, suggesting an important role for SHP-2 in chemotactic signaling.


Journal of Lipid Research | 2007

SREBP-1 regulates the expression of heme oxygenase 1 and the phosphatidylinositol-3 kinase regulatory subunit p55 gamma.

Anders Kallin; Lene E Johannessen; Patrice D. Cani; Catherine Y. Marbehant; Ahmed Essaghir; Fabienne Foufelle; Pascal Ferré; Carl-Henrik Heldin; Nathalie M. Delzenne; Jean-Baptiste Demoulin

Sterol-regulatory element binding proteins (SREBPs) control the expression of genes involved in fatty acid and cholesterol biosynthesis. Using microarrays, we observed that mature SREBP-1 also induced the expression of genes unrelated to lipid metabolism, such as heme oxygenase 1 (HMOX1), plasma glutathione peroxidase, the phosphatidylinositol-3 kinase regulatory subunit p55γ, synaptic vesicle glycoprotein 2A, and COTE1. The expression of these genes was repressed upon addition of sterols, which block endogenous SREBP cleavage, and was induced by the statin drug mevinolin. Stimulation of fibroblasts with platelet-derived growth factor, which activates SREBP-1, had a similar effect. Fasted mice that were refed with a high-carbohydrate diet presented an increased expression of HMOX1 and p55γ in the liver. Overall, the transcriptional signature of SREBP-1 in fibroblasts stimulated by growth factors was very similar to that described in liver cells. We analyzed the HMOX1 promoter and found one SREBP binding site of the E-box type, which was required for regulation by SREBP-1a and SREBP-1c but was insensitive to SREBP-2. In conclusion, our data suggest that SREBP-1 regulates the expression of stress response and signaling genes, which could contribute to the metabolic response to insulin and growth factors in various tissues.


Oncogene | 2002

SHP-2 is involved in heterodimer specific loss of phosphorylation of Tyr771 in the PDGF-beta receptor

Simon Ekman; Anders Kallin; Ulla Engström; Carl-Henrik Heldin; Lars Rönnstrand

We have previously shown that the binding site for GTPase activating protein of Ras (RasGAP) in the PDGF β-receptor, Tyr771, is phosphorylated to a much lower extent in the heterodimeric configuration of PDGF α- and β-receptors, compared to the PDGF β-receptor homodimer. The decreased recruitment of the RasGAP to the receptor leads to prolonged activation of the Ras/MAP kinase pathway, which could explain the increase in mitogenicity seen upon induction of heterodimers. The molecular mechanism underlying these differences was investigated. We could show that the loss of phosphorylation of Tyr771 was dependent on presence of intact binding sites for the protein tyrosine phosphatase SHP-2 on the PDGF β-receptor. Thus, in PDGF receptor mutants in which binding of SHP-2 was lost, a higher degree of phosphorylation of Tyr771 was seen, while other phosphorylation sites in the receptor remained virtually unaffected. Thus, SHP-2 appears to play an important role in modulating phosphorylation of Y771, thereby controlling RasGAP recruitment and Ras/MAP kinase signaling in the heterodimeric configuration of the PDGF receptors.


Haematologica | 2009

The fusion proteins TEL-PDGFRβ and FIP1L1-PDGFRα escape ubiquitination and degradation

Federica Toffalini; Anders Kallin; Peter Vandenberghe; Pascal Pierre; Lucienne Michaux; Jan Cools; Jean-Baptiste Demoulin

Upon growth factor-induced activation, receptor tyrosine kinases such as the PDGF and FGF receptors are targeted for lysosomal degradation via a mechanism that involves ubiquitination of receptor lysines. In this study, it is shown that constitutively active oncogenic fusion proteins that contain PDGF or FGF receptor moieties, caused by specific chromosomal translocations in chronic myeloid neoplasms, escape this negative regulatory mechanism. Background Chimeric oncogenes encoding constitutively active protein tyrosine kinases are associated with chronic myeloid neoplasms. TEL-PDGFRβ (TPβ, also called ETV6-PDGFRB) is a hybrid protein produced by the t(5;12) translocation, FIP1L1-PDGFRα (FPα) results from a deletion on chromosome 4q12 and ZNF198-FGFR1 is created by the t(8;13) translocation. These fusion proteins are found in patients with myeloid neoplasms associated with eosinophilia. Wild-type receptor tyrosine kinases are efficiently targeted for degradation upon activation, in a process that requires Cbl-mediated monoubiquitination of receptor lysines. Since protein degradation pathways have been identified as useful targets for cancer therapy, the aim of this study was to compare the degradation of hybrid and wild-type receptor tyrosine kinases. Design and Methods We used Ba/F3 as a model cell line, as well as leukocytes from two patients, to analyze hybrid protein degradation. Results In contrast to the corresponding wild-type receptors, which are quickly degraded upon activation, we observed that TPβ, FPα and the ZNF198-FGFR1 hybrids escaped down-regulation in Ba/F3 cells. The high stability of TPβ and FPα hybrid proteins was confirmed in leukocytes from leukemia patients. Ubiquitination of TPβ and FPα was much reduced compared to that of wild-type receptors, despite marked Cbl phosphorylation in cells expressing hybrid receptors. The fusion of a destabilizing domain to TPβ induced protein degradation. Instability was reverted by adding the destabilizing domain ligand, Shield1. The destabilization of this modified TPβ reduced cell transformation and STAT5 activation. Conclusions We have shown that chimeric receptor tyrosine kinases escape ubiquitination and down-regulation and that their stabilization is critical to efficient stimulation of cell proliferation.


Haematologica | 2009

The fusion proteins TEL-PDGFRbeta and FIP1L1-PDGFRalpha escape ubiquitination and degradation

Federica Toffalini; Anders Kallin; Peter Vandenberghe; Pascal Pierre; Lucienne Michaux; Jan Cools; Jean-Baptiste Demoulin

Upon growth factor-induced activation, receptor tyrosine kinases such as the PDGF and FGF receptors are targeted for lysosomal degradation via a mechanism that involves ubiquitination of receptor lysines. In this study, it is shown that constitutively active oncogenic fusion proteins that contain PDGF or FGF receptor moieties, caused by specific chromosomal translocations in chronic myeloid neoplasms, escape this negative regulatory mechanism. Background Chimeric oncogenes encoding constitutively active protein tyrosine kinases are associated with chronic myeloid neoplasms. TEL-PDGFRβ (TPβ, also called ETV6-PDGFRB) is a hybrid protein produced by the t(5;12) translocation, FIP1L1-PDGFRα (FPα) results from a deletion on chromosome 4q12 and ZNF198-FGFR1 is created by the t(8;13) translocation. These fusion proteins are found in patients with myeloid neoplasms associated with eosinophilia. Wild-type receptor tyrosine kinases are efficiently targeted for degradation upon activation, in a process that requires Cbl-mediated monoubiquitination of receptor lysines. Since protein degradation pathways have been identified as useful targets for cancer therapy, the aim of this study was to compare the degradation of hybrid and wild-type receptor tyrosine kinases. Design and Methods We used Ba/F3 as a model cell line, as well as leukocytes from two patients, to analyze hybrid protein degradation. Results In contrast to the corresponding wild-type receptors, which are quickly degraded upon activation, we observed that TPβ, FPα and the ZNF198-FGFR1 hybrids escaped down-regulation in Ba/F3 cells. The high stability of TPβ and FPα hybrid proteins was confirmed in leukocytes from leukemia patients. Ubiquitination of TPβ and FPα was much reduced compared to that of wild-type receptors, despite marked Cbl phosphorylation in cells expressing hybrid receptors. The fusion of a destabilizing domain to TPβ induced protein degradation. Instability was reverted by adding the destabilizing domain ligand, Shield1. The destabilization of this modified TPβ reduced cell transformation and STAT5 activation. Conclusions We have shown that chimeric receptor tyrosine kinases escape ubiquitination and down-regulation and that their stabilization is critical to efficient stimulation of cell proliferation.


Haematologica | 2009

The fusion proteins TEL-PDGFR and FIP1L1-PDGFR escape ubiquitination and degradation

Federica Toffalini; Anders Kallin; Peter Vandenberghe; Pascal Pierre; Lucienne Michaux; Jan Cools; Jean-Baptiste Demoulin

Upon growth factor-induced activation, receptor tyrosine kinases such as the PDGF and FGF receptors are targeted for lysosomal degradation via a mechanism that involves ubiquitination of receptor lysines. In this study, it is shown that constitutively active oncogenic fusion proteins that contain PDGF or FGF receptor moieties, caused by specific chromosomal translocations in chronic myeloid neoplasms, escape this negative regulatory mechanism. Background Chimeric oncogenes encoding constitutively active protein tyrosine kinases are associated with chronic myeloid neoplasms. TEL-PDGFRβ (TPβ, also called ETV6-PDGFRB) is a hybrid protein produced by the t(5;12) translocation, FIP1L1-PDGFRα (FPα) results from a deletion on chromosome 4q12 and ZNF198-FGFR1 is created by the t(8;13) translocation. These fusion proteins are found in patients with myeloid neoplasms associated with eosinophilia. Wild-type receptor tyrosine kinases are efficiently targeted for degradation upon activation, in a process that requires Cbl-mediated monoubiquitination of receptor lysines. Since protein degradation pathways have been identified as useful targets for cancer therapy, the aim of this study was to compare the degradation of hybrid and wild-type receptor tyrosine kinases. Design and Methods We used Ba/F3 as a model cell line, as well as leukocytes from two patients, to analyze hybrid protein degradation. Results In contrast to the corresponding wild-type receptors, which are quickly degraded upon activation, we observed that TPβ, FPα and the ZNF198-FGFR1 hybrids escaped down-regulation in Ba/F3 cells. The high stability of TPβ and FPα hybrid proteins was confirmed in leukocytes from leukemia patients. Ubiquitination of TPβ and FPα was much reduced compared to that of wild-type receptors, despite marked Cbl phosphorylation in cells expressing hybrid receptors. The fusion of a destabilizing domain to TPβ induced protein degradation. Instability was reverted by adding the destabilizing domain ligand, Shield1. The destabilization of this modified TPβ reduced cell transformation and STAT5 activation. Conclusions We have shown that chimeric receptor tyrosine kinases escape ubiquitination and down-regulation and that their stabilization is critical to efficient stimulation of cell proliferation.


Journal of Biological Chemistry | 2003

The Tyrosine Kinase Pyk2 Regulates Arf1 Activity by Phosphorylation and Inhibition of the Arf-GTPase-activating Protein ASAP1

Anamarija Kruljac-Letunic; Jörg Moelleken; Anders Kallin; Felix T. Wieland; Andree Blaukat

Collaboration


Dive into the Anders Kallin's collaboration.

Top Co-Authors

Avatar

Jean-Baptiste Demoulin

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Federica Toffalini

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carl-Henrik Heldin

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Peter Vandenberghe

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Ahmed Essaghir

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Jan Cools

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Lucienne Michaux

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Charlotte Rorsman

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Pascal Pierre

Université catholique de Louvain

View shared research outputs
Researchain Logo
Decentralizing Knowledge