SwePub
Sök i SwePub databas

  Extended search

Träfflista för sökning "WFRF:(Goumans Marie Josè) "

Search: WFRF:(Goumans Marie Josè)

  • Result 1-10 of 17
Sort/group result
   
EnumerationReferenceCoverFind
1.
  • Chiara, Federica, et al. (author)
  • A gain of function mutation in the activation loop of platelet-derived growth factor beta-receptor deregulates its kinase activity
  • 2004
  • In: Journal of Biological Chemistry. - 0021-9258 .- 1083-351X. ; 279:41, s. 42516-42527
  • Journal article (peer-reviewed)abstract
    • The platelet-derived growth factor receptors (PDGFRs) are receptor tyrosine kinases implicated in multiple aspects of cell growth, differentiation, and survival. Recently, a gain of function mutation in the activation loop of the human PDGFRalpha has been found in patients with gastrointestinal stromal tumors. Here we show that a mutation in the corresponding codon in the activation loop of the murine PDGFRbeta, namely an exchange of asparagine for aspartic acid at amino acid position 849 (D849N), confers transforming characteristics to embryonic fibroblasts from mutant mice, generated by a knock-in strategy. By comparing the enzymatic properties of the wild-type versus the mutant receptor protein, we demonstrate that the D849N mutation lowers the threshold for kinase activation, causes a dramatic alteration in the pattern of tyrosine phosphorylation kinetics following ligand stimulation, and induces a ligand-independent phosphorylation of several tyrosine residues. These changes result in deregulated recruitment of specific signal transducers. The GTPase-activating protein for Ras (RasGAP), a negative regulator of the Ras mitogenic pathway, displayed a delayed binding to the mutant receptor. Moreover, we have observed enhanced ligand-independent ERK1/2 activation and an increased proliferation of mutant cells. The p85 regulatory subunit of the phosphatidylinositol 3 '-kinase was constitutively associated with the mutant receptor, and this ligand-independent activation of the phosphatidylinositol 3'-kinase pathway may explain the observed strong protection against apoptosis and increased motility in cellular wounding assays. Our findings support a model whereby an activating point mutation results in a deregulated PDGFRbeta with oncogenic predisposition.
  •  
2.
  • Cunha, Sara I, et al. (author)
  • Genetic and pharmacological targeting of activin receptor-like kinase 1 impairs tumor growth and angiogenesis
  • 2010
  • In: Journal of Experimental Medicine. - : The Rockefeller University Press. - 0022-1007 .- 1540-9538. ; 207:1, s. 85-100
  • Journal article (peer-reviewed)abstract
    • Members of the transforming growth factor beta (TGF-beta) family have been genetically linked to vascular formation during embryogenesis. However, contradictory studies about the role of TGF-beta and other family members with reported vascular functions, such as bone morphogenetic protein (BMP) 9, in physiological and pathological angiogenesis make the need for mechanistic studies apparent. We demonstrate, by genetic and pharmacological means, that the TGF-beta and BMP9 receptor activin receptor-like kinase (ALK) 1 represents a new therapeutic target for tumor angiogenesis. Diminution of ALK1 gene dosage or systemic treatment with the ALK1-F(c) fusion protein RAP-041 retarded tumor growth and progression by inhibition of angiogenesis in a transgenic mouse model of multistep tumorigenesis. Furthermore, RAP-041 significantly impaired the in vitro and in vivo angiogenic response toward vascular endothelial growth factor A and basic fibroblast growth factor. In seeking the mechanism for the observed effects, we uncovered an unexpected signaling synergy between TGF-beta and BMP9, through which the combined action of the two factors augmented the endothelial cell response to angiogenic stimuli. We delineate a decisive role for signaling by TGF-beta family members in tumor angiogenesis and offer mechanistic insight for the forthcoming clinical development of drugs blocking ALK1 in oncology.
  •  
3.
  •  
4.
  • Hawinkels, Lukas J. A. C., et al. (author)
  • Activin Receptor-like Kinase 1 Ligand Trap Reduces Microvascular Density and Improves Chemotherapy Efficiency to Various Solid Tumors
  • 2016
  • In: Clinical Cancer Research. - 1078-0432 .- 1557-3265. ; 22:1, s. 96-106
  • Journal article (peer-reviewed)abstract
    • Purpose: Antiangiogenic therapy, mostly targeting VEGF, has been applied in cancer patients for the last decade. However, resistance to anti-VEGF therapy and/or no significant benefit as monotherapeutic agent is often observed. Therefore, new antiangiogenic strategies are needed. In the current study, we investigated the therapeutic effect of interfering with the bone morphogenetic protein (BMP)9/activin receptor-like kinase (ALK)1 signaling pathway by using an ALK1-Fc ligand trap. Experimental Design: We analyzed the potential antiangiogenic and antitumor effects of ALK1-Fc protein as monotherapy and in combination with chemotherapy in vivo in mouse models of melanoma, head and neck cancer, and invasive lobular breast carcinomas. ALK1-Fc sequesters BMP9 and 10 and prevents binding of these ligands to endothelial ALK1, which regulates angiogenesis. Results: Treatment of mice with ALK1-Fc strongly decreased the tumors' microvascular density in the three different mouse cancer models. However, this effect was not accompanied by a reduction in tumor volume. Animmunohistochemical analysis of the tumor samples revealed that ALK1-Fc treatment increased the pericyte coverage of the remaining tumor vessels and decreased the hypoxia within the tumor. Next, we observed that combining ALK1-Fc with cisplatin inhibited tumor growth in the breast and head and neck cancer models more efficiently than chemotherapy alone. Conclusions: The addition of ALK1-Fc to the cisplatin treatment was able to enhance the cytotoxic effect of the chemotherapy. Our results provide strong rationale to explore combined targeting of ALK1 with chemotherapy in a clinical setting, especially in the ongoing phase II clinical trials with ALK1-Fc.
  •  
5.
  • Itoh, Fumiko, et al. (author)
  • Poor vessel formation in embryos from knock-in mice expressing ALK5 with L45 loop mutation defective in Smad activation
  • 2009
  • In: Laboratory Investigation. - : Elsevier BV. - 1530-0307 .- 0023-6837. ; 89:7, s. 800-810
  • Journal article (peer-reviewed)abstract
    • Transforming growth factor (TGF)-beta regulates vascular development through two type I receptors: activin receptor-like kinase (ALK) 1 and ALK5, each of which activates a different downstream Smad pathway. The endothelial cell (EC)-specific ALK1 increases EC proliferation and migration, whereas the ubiquitously expressed ALK5 inhibits both of these processes. As ALK1 requires the kinase activity of ALK5 for optimal activation, the lack of ALK5 in ECs results in defective phosphorylation of both Smad pathways on TGF-beta stimulation. To understand why TGF-beta signaling through ALK1 and ALK5 has opposing effects on ECs and whether this takes place in vivo, we carefully compared the phenotype of ALK5 knock-in (ALK5(KI/KI)) mice, in which the aspartic acid residue 266 in the L45 loop of ALK5 was replaced by an alanine residue, with the phenotypes of ALK5 knock-out (ALK5(-/-)) and wild-type mice. The ALK5(KI/KI) mice showed angiogenic defects with embryonic lethality at E10.5-11.5. Although the phenotype of the ALK5(KI/KI) mice was quite similar to that of the ALK5(-/-) mice, the hierarchical structure of blood vessels formed in the ALK5(KI/KI) embryos was more developed than that in the ALK5(-/-) mutants. Thus, the L45 loop mutation in ALK5 partially rescued the earliest vascular defects in the ALK5(-/-) embryos. This study supports our earlier observation that vascular maturation in vivo requires both TGF-beta/ALK1/BMP-Smad and TGF-beta/ALK5/activin-Smad pathways for normal vascular development. Laboratory Investigation (2009) 89, 800-810; doi:10.1038/labinvest.2009.37; published online 27 April 2009
  •  
6.
  •  
7.
  • Karlsson, Göran, et al. (author)
  • Gene expression profiling demonstrates that TGF-{beta}1 signals exclusively through receptor complexes involving Alk5 and identifies targets of TGF-{beta} signaling.
  • 2005
  • In: Physiological Genomics. - : American Physiological Society. - 1094-8341 .- 1531-2267. ; 21:3, s. 396-403
  • Journal article (peer-reviewed)abstract
    • Transforming growth factor-β 1 (TGF-β) regulates cellular functions like proliferation, differentiation, and apoptosis. On the cell surface, TGF-β binds to receptor complexes consisting of TGF-β receptor type II (Tβ RII) and activin-like kinase receptor-5 (Alk5), and the downstream signaling is transduced by Smad and MAPK proteins. Recent data have shown that alternative receptor combinations aside from the classical pairing of Tβ RII/Alk5 can be relevant for TGF-β signaling. We have screened for alternative receptors for TGF-β and also for gene targets of TGF-β signaling, by performing functional assays and microarray analysis in murine embryonic fibroblast (MEF) cell lines lacking Alk5. Data from TGF-β-stimulated Alk5(-/-) cells show them to be completely unaffected by TGF-β. Additionally, 465 downstream targets of Alk5 signaling were identified when comparing Alk5(-/-) or TGF-β-stimulated Alk5(+/+) MEFs with unstimulated Alk5(+/+) cells. Our results demonstrate that, in MEFs, TGF-β signals exclusively through complexes involving Alk5, and give insight to its downstream effector genes.
  •  
8.
  • Larsson, Jonas, et al. (author)
  • Abnormal angiogenesis but intact hematopoietic potential in TGF-beta type I receptor-deficient mice
  • 2001
  • In: EMBO Journal. - : Wiley. - 1460-2075. ; 20:7, s. 1663-1673
  • Journal article (peer-reviewed)abstract
    • Deletion of the transforming growth factor beta1 (TGF-beta1) gene in mice has previously suggested that it regulates both hematopoiesis and angiogenesis. To define the function of TGF-beta more precisely, we inactivated the TGF-beta type I receptor (T beta RI) gene by gene targeting. Mice lacking T beta RI die at midgestation, exhibiting severe defects in vascular development of the yolk sac and placenta, and an absence of circulating red blood cells. However, despite obvious anemia in the T beta RI-/- yolk sacs, clonogenic assays on yolk sac-derived hematopoietic precursors in vitro revealed that T beta RI-/- mice exhibit normal hematopoietic potential compared with wild-type and heterozygous siblings, Endothelial cells derived from T beta RI-deficient embryos show enhanced cell proliferation, improper migratory behavior and impaired fibronectin production in vitro, defects that are associated with the vascular defects seen in vivo. We thus demonstrate here that, while T beta RI is crucial for the function of TGF-beta during vascular development and can not be compensated for by the activin receptor-like kinase-1 (ALK-1), functional hematopoiesis and development of hematopoietic progenitors is not dependent on TGF-beta signaling via T beta RI.
  •  
9.
  • Larsson, Jonas, et al. (author)
  • TGF-{beta} signaling-deficient hematopoietic stem cells have normal self-renewal and regenerative ability in vivo despite increased proliferative capacity in vitro.
  • 2003
  • In: Blood. - : American Society of Hematology. - 1528-0020 .- 0006-4971. ; 102:9, s. 3129-3135
  • Journal article (peer-reviewed)abstract
    • Studies in vitro implicate transforming growth factor β (TGF-β) as a key regulator of hematopoiesis with potent inhibitory effects on progenitor and stem cell proliferation. In vivo studies have been hampered by early lethality of knock-out mice for TGF-β isoforms and the receptors. To directly assess the role of TGF-β signaling for hematopoiesis and hematopoietic stem cell (HSC) function in vivo, we generated a conditional knock-out model in which a disruption of the TGF-β type I receptor (TβRI) gene was induced in adult mice. HSCs from induced mice showed increased proliferation recruitment when cultured as single cells under low stimulatory conditions in vitro, consistent with an inhibitory role of TGF-β in HSC proliferation. However, induced TβRI null mice show normal in vivo hematopoiesis with normal numbers and differentiation ability of hematopoietic progenitor cells. Furthermore HSCs from TβRI null mice exhibit a normal cell cycle distribution and do not differ in their ability long term to repopulate primary and secondary recipient mice following bone marrow transplantation. These findings challenge the classical view that TGF-β is an essential negative regulator of hematopoietic stem cells under physiologic conditions in vivo.
  •  
10.
  • Levéen, Per, et al. (author)
  • TGF-{beta} type II receptor deficient thymocytes develop normally but demonstrate increased CD8+ proliferation in vivo.
  • 2005
  • In: Blood. - : American Society of Hematology. - 1528-0020 .- 0006-4971. ; 106:13, s. 4234-4240
  • Journal article (peer-reviewed)abstract
    • We have taken advantage of the Cre/lox system to generate a mouse model with inducible deficiency of transforming growth factor β receptor II (TβRII). Using this approach, transforming growth factor β (TGF-β) signaling deficiency can be restricted to the hematopoietic system by bone marrow transplantation. Mice that received transplants with TβRII-/- bone marrow develop a lethal inflammatory disorder closely resembling that of TGF-β1-null mice. Previous in vitro studies have suggested multiple roles for TGF-β in T-cell development, including proliferation, apoptosis, and differentiation. We used our transplantation model to ask whether T-cell development is normal in the absence of TGF-β signaling. The findings show for the first time in vivo and in fetal thymus organ culture (FTOC) that TGF-β is not required for thymocytes to differentiate along the entire pathway of thymic T-cell development, as defined by the expression patterns of CD4, CD8, CD25, and CD44. In contrast to previous investigations, no increase of thymocyte apoptosis was observed. However, TβRII-deficient CD8+ thymocytes displayed a 2-fold increase in proliferation rate, as determined by bromodeoxyuridine (BrdU) incorporation in vivo. These results reinforce the importance of TGF-β as an immune regulator critical for T-cell function.
  •  
Skapa referenser, mejla, bekava och länka
  • Result 1-10 of 17
Type of publication
journal article (15)
other publication (1)
research review (1)
Type of content
peer-reviewed (16)
other academic/artistic (1)
Author/Editor
Goumans, Marie-José (17)
ten Dijke, Peter (11)
Itoh, Susumu (6)
Larsson, Jonas (5)
Karlsson, Stefan (5)
Heldin, Carl-Henrik (3)
show more...
Levéen, Per (3)
Lebrin, Franck (2)
Mummery, Christine L ... (2)
Heldin, Carl-Henrik, ... (2)
Pardali, Evangelia (2)
Thorikay, Midory (2)
Aspenström, Pontus (1)
Heuchel, Rainer (1)
Karlsson, Göran (1)
Williams, Eleanor (1)
Ward, Dorien (1)
Borén, Jan, 1963 (1)
Cilio, Corrado (1)
Ringnér, Markus (1)
Larsson, Erik, 1975 (1)
Akyürek, Levent, 196 ... (1)
Zhou, Xianghua, 1973 (1)
Ehinger, Mats (1)
Wernstedt, Christer (1)
Sartipy, Peter (1)
Pietras, Kristian (1)
Anderberg, Charlotte (1)
Sanchez-Duffhues, Go ... (1)
Xu, Xiufeng (1)
Thorgeirsson, Snorri ... (1)
Synnergren, Jane (1)
Hellberg, Carina (1)
Helgadottir, Hildur (1)
Wang, Qing-Dong (1)
Björnsson, Jon Mar (1)
Blank Savukinas, Ulr ... (1)
Åhgren, Aive (1)
Brolén, Gabriella (1)
Fan, Xiaolong (1)
Forsberg, Henrik (1)
Carlsén, Maria (1)
Chiara, Federica (1)
Rasola, Andrea (1)
Cunha, Sara I. (1)
Hawinkels, Lukas (1)
Seehra, Jasbir (1)
De Vinuesa, Amaya Ga ... (1)
Mezzanotte, Laura (1)
Gittenberger-de Groo ... (1)
show less...
University
Uppsala University (10)
Lund University (6)
University of Gothenburg (1)
University of Skövde (1)
Karolinska Institutet (1)
Language
English (13)
Undefined language (4)
Research subject (UKÄ/SCB)
Medical and Health Sciences (8)
Natural sciences (1)

Year

Kungliga biblioteket hanterar dina personuppgifter i enlighet med EU:s dataskyddsförordning (2018), GDPR. Läs mer om hur det funkar här.
Så här hanterar KB dina uppgifter vid användning av denna tjänst.

 
pil uppåt Close

Copy and save the link in order to return to this view