Logo: to the web site of Uppsala University

uu.sePublications from Uppsala University
Change search
Link to record
Permanent link

Direct link
Alternative names
Publications (10 of 250) Show all publications
Mendes Rodrigues Junior, D., Tsirigoti, C., Psatha, K., Kletsas, D., Aivaliotis, M., Heldin, C.-H. & Moustakas, A. (2025). TGF-β induces cholesterol accumulation to regulate the secretion of tumor-derived extracellular vesicles. Journal of Experimental & Clinical Cancer Research, 44(1), Article ID 42.
Open this publication in new window or tab >>TGF-β induces cholesterol accumulation to regulate the secretion of tumor-derived extracellular vesicles
Show others...
2025 (English)In: Journal of Experimental & Clinical Cancer Research, E-ISSN 1756-9966, Vol. 44, no 1, article id 42Article in journal (Refereed) Published
Abstract [en]

Background

Cancer cells are avid extracellular vesicle (EV) producers. EVs transport transforming growth factor-beta (TGF-beta), which is commonly activated under late stages of cancer progression. Nevertheless, whether TGF-beta signaling coordinates EV biogenesis is a relevant topic that remains minimally explored.

Method

We sought after specific TGF-beta pathway mediators that could regulate EV release. To this end, we used a large number of cancer cell models, coupled to EV cell biological assays, unbiased proteomic and transcriptomic screens, followed by signaling and cancer biology analyses, including drug resistance assays.

Results

We report that TGF-beta, by activating its type I receptor and MEK-ERK1/2 signaling, increased the numbers of EVs released by human cancer cells. Upon examining cholesterol as a mediator of EV biogenesis, we delineated a pathway whereby ERK1/2 acted by phosphorylating sterol regulatory element-binding protein-2 that transcriptionally induced 7-dehydrocholesterol reductase expression, thus raising cholesterol abundance at both cellular and EV levels. Notably, inhibition of MEK or cholesterol synthesis, which impaired TGF-beta-induced EV secretion, sensitized cancer cells to chemotherapeutic drugs. Furthermore, proteomic profiling of two distinct EV populations revealed that EVs secreted by TGF-beta-stimulated cells were either depleted or enriched for different sets of cargo proteins. Among these, latent-TGF-beta 1 present in the EVs was not affected by TGF-beta signaling, while TGF-beta pathway-related molecules (e.g., matrix metalloproteinases, including MMP9) were either uniquely enriched on EVs or strongly enhanced after TGF-beta stimulation. EV-associated latent-TGF-beta 1 activated SMAD signaling, even when EV uptake was blocked by heparin, indicating competent signaling capacity from target cell surface receptors. MMP inhibitor or proteinase treatment blocked EV-mediated SMAD signaling, suggesting that EVs require MMP activity to release the active TGF-beta from its latent complex, a function also linked to the EV-mediated transfer of pro-migratory potential and ability of cancer cells to survive in the presence of cytotoxic drugs.

Conclusion

Hence, we delineated a novel signaling cascade that leads to high rates of EV generation by cancer cells in response to TGF-beta, with cholesterol being a key intermediate step in this mechanism.Graphical Abstract center dot TGF-beta increases EV release by activating a MEK-ERK1/2-SREBP2-DHCR7 signaling and transcriptional pathway.center dot TGF-beta-induced DHCR7 expression raises cholesterol abundance that promotes EV release.center dot EVs carry surface latent TGF-beta and MMP9 that can activate TGF-beta receptor signaling on the surface of recipient cells.

Place, publisher, year, edition, pages
BioMed Central (BMC), 2025
Keywords
Cancer, Cholesterol, Extracellular, Vesicles, Matrix metalloproteinase, Transforming growth factor beta
National Category
Cell and Molecular Biology Cancer and Oncology Molecular Biology
Identifiers
urn:nbn:se:uu:diva-551460 (URN)10.1186/s13046-025-03291-0 (DOI)001415312500001 ()39910665 (PubMedID)2-s2.0-85218180431 (Scopus ID)
Available from: 2025-02-28 Created: 2025-02-28 Last updated: 2025-02-28Bibliographically approved
Rubin Sander, M., Tsiatsiou, A. K., Wang, K., Papadopoulos, N., Rorsman, C., Olsson, F., . . . Lennartsson, J. (2024). PDGF-induced internalisation promotes proteolytic cleavage of PDGFRβ in mesenchymal cells. Growth Factors, 42(4), 147-160
Open this publication in new window or tab >>PDGF-induced internalisation promotes proteolytic cleavage of PDGFRβ in mesenchymal cells
Show others...
2024 (English)In: Growth Factors, ISSN 0897-7194, E-ISSN 1029-2292, Vol. 42, no 4, p. 147-160Article in journal (Refereed) Published
Abstract [en]

Platelet-derived growth factor (PDGF)-induced signalling via PDGF receptor β (PDGFRβ) leads to activation of downstream signalling pathways which regulate multiple cellular responses. It is unclear how PDGFRβ is degraded; both lysosomal and proteasomal degradation have been suggested. In this study, we have characterised the proteolytic cleavage of ligand-activated PDGFRβ, which results in two fragments: a larger fragment containing the extracellular domain, the transmembrane segment, and a part of the intracellular juxtamembrane region with a molecular mass of ∼130 kDa, and an intracellular ∼70 kDa fragment released into the cytoplasm. The proteolytic processing did not take place without internalisation of PDGFRβ. In addition, chelation of intracellular Ca2+ inhibited proteolytic processing. Inhibition of the proteasome affected signal transduction by increasing the phosphorylation of PDGFRβ, PLCγ, and STAT3 while reducing it on Erk1/2 and not affecting Akt. The proteolytic cleavage was observed in fibroblasts or cells that had undergone epithelial-mesenchymal transition.

Place, publisher, year, edition, pages
Taylor & Francis, 2024
Keywords
PDGFR, RTK, bortezomib, cleavage, proteasome, proteolysis
National Category
Cell and Molecular Biology
Identifiers
urn:nbn:se:uu:diva-548858 (URN)10.1080/08977194.2024.2413623 (DOI)001329833100001 ()39387439 (PubMedID)2-s2.0-85206197452 (Scopus ID)
Funder
Swedish Cancer Society, 222363PjH02HSwedish Cancer Society, 211427Pj01HSwedish Cancer Society, 222306Pj
Available from: 2025-01-29 Created: 2025-01-29 Last updated: 2025-01-30Bibliographically approved
Voytyuk, O., Ohata, Y., Moustakas, A., ten Dijke, P. & Heldin, C.-H. (2024). Smad7 palmitoylation by the S-acyltransferase zDHHC17 enhances its inhibitory effect on TGF-β/Smad signaling. Journal of Biological Chemistry, 300(7), Article ID 107462.
Open this publication in new window or tab >>Smad7 palmitoylation by the S-acyltransferase zDHHC17 enhances its inhibitory effect on TGF-β/Smad signaling
Show others...
2024 (English)In: Journal of Biological Chemistry, ISSN 0021-9258, E-ISSN 1083-351X, Vol. 300, no 7, article id 107462Article in journal (Refereed) Published
Abstract [en]

Intracellular signaling by the pleiotropic cytokine transforming growth factor- (3 (TGF-(3) is inhibited by Smad7 in a feedback control mechanism. The activity of Smad7 is tightly regulated by multiple post-translational modifications. Using resin-assisted capture and metabolic labeling methods, we show here that Smad7 is S-palmitoylated in mammary epithelial cell models that are widely studied because of their strong responses to TGF-(3 and their biological relevance to mammary development and tumor progression. S-palmitoylation of Smad7 is mediated by zDHHC17, a member of a family of 23 S-acyltransferase enzymes. Moreover, we identified four cysteine residues (Cys202, Cys225, Cys415, and Cys417) in Smad7 as palmitoylation acceptor sites. S-palmitoylation of Smad7 on Cys415 and Cys417 promoted the translocation of Smad7 from the nucleus to the cytoplasm, enhanced the stability of the Smad7 protein, and enforced its inhibitory effect on TGF-(3-induced Smad transcriptional response. Thus, our fi ndings reveal a new post-translational modification of Smad7, and highlight an important role of S-palmitoylation to enhance inhibition of TGF-(3/Smad signaling by Smad7.

Place, publisher, year, edition, pages
Elsevier, 2024
National Category
Cell and Molecular Biology
Identifiers
urn:nbn:se:uu:diva-544793 (URN)10.1016/j.jbc.2024.107462 (DOI)001364997000001 ()38876303 (PubMedID)
Funder
EU, European Research Council, 787,472Swedish Research Council, 2020-01291
Available from: 2024-12-10 Created: 2024-12-10 Last updated: 2024-12-10Bibliographically approved
Westermark, B. & Heldin, C.-H. (2024). Special issue: frontiers in recent advances on cancer diagnosis and treatment. Upsala Journal of Medical Sciences, 129, Article ID e11919.
Open this publication in new window or tab >>Special issue: frontiers in recent advances on cancer diagnosis and treatment
2024 (English)In: Upsala Journal of Medical Sciences, ISSN 0300-9734, E-ISSN 2000-1967, Vol. 129, article id e11919Article in journal, Editorial material (Other academic) Published
Place, publisher, year, edition, pages
Upsala Medical Society, 2024
National Category
Cancer and Oncology
Identifiers
urn:nbn:se:uu:diva-550028 (URN)10.48101/ujms.v129.11919 (DOI)001399023300001 ()39780954 (PubMedID)2-s2.0-85214353252 (Scopus ID)
Available from: 2025-02-11 Created: 2025-02-11 Last updated: 2025-02-11Bibliographically approved
ten Dijke, P., Miyazono, K., Heldin, C.-H. & Moustakas, A. (2024). Special issue: TGF-β and epithelial-mesenchymal transition in cancer. Seminars in Cancer Biology, 102, 1-3
Open this publication in new window or tab >>Special issue: TGF-β and epithelial-mesenchymal transition in cancer
2024 (English)In: Seminars in Cancer Biology, ISSN 1044-579X, E-ISSN 1096-3650, Vol. 102, p. 1-3Article in journal, Editorial material (Other academic) Published
Place, publisher, year, edition, pages
Elsevier, 2024
Keywords
Cancer, Epithelial-mesenchymal transition, Metastasis, Signal transduction, Transcription, Transforming growthfactor-beta
National Category
Cancer and Oncology
Identifiers
urn:nbn:se:uu:diva-536084 (URN)10.1016/j.semcancer.2024.06.002 (DOI)001266725100001 ()38944133 (PubMedID)
Funder
Swedish Research Council, 2020-01291EU, European Research Council, 787472Swedish Cancer Society, CAN2021/1506Pj01HSwedish Childhood Cancer Foundation, PR2023-0115Swedish Research Council, 2023-02865
Available from: 2024-08-13 Created: 2024-08-13 Last updated: 2024-08-13Bibliographically approved
Vasilaki, E., Bai, Y., Ali, M. M., Sundqvist, A., Moustakas, A. & Heldin, C.-H. (2024). ΔNp63 bookmarks and creates an accessible epigenetic environment for TGFβ-induced cancer cell stemness and invasiveness. Cell Communication and Signaling, 22(1), Article ID 411.
Open this publication in new window or tab >>ΔNp63 bookmarks and creates an accessible epigenetic environment for TGFβ-induced cancer cell stemness and invasiveness
Show others...
2024 (English)In: Cell Communication and Signaling, E-ISSN 1478-811X, Vol. 22, no 1, article id 411Article in journal (Refereed) Published
Abstract [en]

Background: p63 is a transcription factor with intrinsic pioneer factor activity and pleiotropic functions. Transforming growth factor beta (TGF beta) signaling via activation and cooperative action of canonical, SMAD, and non-canonical, MAP-kinase (MAPK) pathways, elicits both anti- and pro-tumorigenic properties, including cell stemness and invasiveness. TGF beta activates the Delta Np63 transcriptional program in cancer cells; however, the link between TGF beta and p63 in unmasking the epigenetic landscape during tumor progression allowing chromatin accessibility and gene transcription, is not yet reported.

Methods: Small molecule inhibitors, including protein kinase inhibitors and RNA-silencing, provided loss of function analyses. Sphere formation assays in cancer cells, chromatin immunoprecipitation and mRNA expression assays were utilized in order to gain mechanistic evidence. Mass spectrometry analysis coupled to co-immunoprecipitation assays revealed novel p63 interactors and their involvement in p63-dependent transcription.

Results: The sphere-forming capacity of breast cancer cells was enhanced upon TGF beta stimulation and significantly decreased upon Delta Np63 depletion. Activation of TGF beta signaling via p38 MAPK signaling induced Delta Np63 phosphorylation at Ser 66/68 resulting in stabilized Delta Np63 protein with enhanced DNA binding properties. TGF beta stimulation altered the ratio of H3K27ac and H3K27me3 histone modification marks, pointing towards higher H3K27ac and increased p300 acetyltransferase recruitment to chromatin. By silencing the expression of Delta Np63, the TGF beta effect on chromatin remodeling was abrogated. Inhibition of H3K27me3, revealed the important role of TGF beta as the upstream signal for guiding Delta Np63 to the TGF beta/SMAD gene loci, as well as the indispensable role of Delta Np63 in recruiting histone modifying enzymes, such as p300, to these genomic regions, regulating chromatin accessibility and gene transcription. Mechanistically, TGF beta through SMAD activation induced dissociation of Delta Np63 from NURD or NCOR/SMRT histone deacetylation complexes, while promoted the assembly of Delta Np63-p300 complexes, affecting the levels of histone acetylation and the outcome of Delta Np63-dependent transcription.

Conclusions: Delta Np63, phosphorylated and recruited by TGF beta to the TGF beta/SMAD/Delta Np63 gene loci, promotes chromatin accessibility and transcription of target genes related to stemness and cell invasion.

Place, publisher, year, edition, pages
Springer Nature, 2024
Keywords
p63, Transforming growth factor beta (TGF beta), Signal transduction, Transcription, Chromatin accessibility, Protein-protein interaction
National Category
Cancer and Oncology
Identifiers
urn:nbn:se:uu:diva-537760 (URN)10.1186/s12964-024-01794-5 (DOI)001296725200001 ()39180088 (PubMedID)
Available from: 2024-09-17 Created: 2024-09-17 Last updated: 2024-10-16Bibliographically approved
Lin, C.-Y., Basu, K., Ruusala, A., Kozlova, I., Li, Y.-S., Skandalis, S. S., . . . Heldin, P. (2023). Hyaluronan-Induced CD44-iASPP Interaction Affects Fibroblast Migration and Survival. Cancers, 15(4), Article ID 1082.
Open this publication in new window or tab >>Hyaluronan-Induced CD44-iASPP Interaction Affects Fibroblast Migration and Survival
Show others...
2023 (English)In: Cancers, ISSN 2072-6694, Vol. 15, no 4, article id 1082Article in journal (Refereed) Published
Abstract [en]

In the present study, we show that the inhibitor of the apoptosis-stimulating protein of p53 (iASPP) physically interacts with the hyaluronan receptor CD44 in normal and transformed cells. We noticed that the CD44 standard isoform (CD44s), but not the variant isoform (CD44v), bound to iASPP via the ankyrin-binding domain in CD44s. The formation of iASPP-CD44s complexes was promoted by hyaluronan stimulation in fibroblasts but not in epithelial cells. The cellular level of p53 affected the amount of the iASPP-CD44 complex. iASPP was required for hyaluronan-induced CD44-dependent migration and adhesion of fibroblasts. Of note, CD44 altered the sub-cellular localization of the iASPP-p53 complex; thus, ablation of CD44 promoted translocation of iASPP from the nucleus to the cytoplasm, resulting in increased formation of a cytoplasmic iASPP-p53 complex in fibroblasts. Overexpression of iASPP decreased, but CD44 increased the level of intracellular reactive oxygen species (ROS). Knock-down of CD44s, in the presence of p53, led to increased cell growth and cell density of fibroblasts by suppression of p27 and p53. Our observations suggest that the balance of iASPP-CD44 and iASPP-p53 complexes affect the survival and migration of fibroblasts.

Place, publisher, year, edition, pages
MDPI, 2023
Keywords
CD44, p53, iASPP, hyaluronan, apoptosis, cancer, tumor microenvironment
National Category
Cell and Molecular Biology
Identifiers
urn:nbn:se:uu:diva-498563 (URN)10.3390/cancers15041082 (DOI)000938950500001 ()36831425 (PubMedID)
Funder
Swedish Cancer Society, 180657Swedish Cancer Society, 190066Pj01HAgnes and Mac Rudberg Foundation
Note

De två första författarna delar förstaförfattarskapet.

Available from: 2023-03-17 Created: 2023-03-17 Last updated: 2023-12-12Bibliographically approved
Wang, K., Papadopoulos, N., Hamidi, A., Lennartsson, J. & Heldin, C.-H. (2023). SUMOylation of PDGF receptor α affects signaling via PLCγ and STAT3, and cell proliferation. BMC Molecular and Cell Biology, 24(1), Article ID 19.
Open this publication in new window or tab >>SUMOylation of PDGF receptor α affects signaling via PLCγ and STAT3, and cell proliferation
Show others...
2023 (English)In: BMC Molecular and Cell Biology, E-ISSN 2661-8850, Vol. 24, no 1, article id 19Article in journal (Refereed) Published
Abstract [en]

Background

The platelet-derived growth factor (PDGF) family of ligands exerts their cellular effects by binding to α- and β-tyrosine kinase receptors (PDGFRα and PDGFRβ, respectively). SUMOylation is an important posttranslational modification (PTM) which regulates protein stability, localization, activation and protein interactions. A mass spectrometry screen has demonstrated SUMOylation of PDGFRα. However, the functional role of SUMOylation of PDGFRα has remained unknown.

Results

In the present study, we validated that PDGFRα is SUMOylated on lysine residue 917 as was previously reported using a mass spectrometry approach. Mutation of lysine residue 917 to arginine (K917R) in PDGFRα substantially decreased SUMOylation, indicating that this amino acid residue is a major SUMOylation site. Whereas no difference in the stability of wild-type and mutant receptor was observed, the K917R mutant PDGFRα was less ubiquitinated than wild-type PDGFRα. The internalization and trafficking of the receptor to early and late endosomes were not affected by the mutation, neither was the localization of the PDGFRα to Golgi. However, the K917R mutant PDGFRα showed delayed activation of PLC-γ and enhanced activation of STAT3. Functional assays showed that the mutation of K917 of PDGFRα decreased cell proliferation in response to PDGF-BB stimulation.

Conclusions

SUMOylation of PDGFRα decreases ubiquitination of the receptor and affects ligand-induced signaling and cell proliferation.

Place, publisher, year, edition, pages
BioMed Central (BMC), 2023
National Category
Cell and Molecular Biology
Identifiers
urn:nbn:se:uu:diva-496383 (URN)10.1186/s12860-023-00481-6 (DOI)000991979900001 ()37193980 (PubMedID)
Funder
Swedish Cancer Society, 190066 Pj 01 HUppsala University
Note

Title in Web of Science: SUMOylation of PDGF receptor alpha affects signaling via PLC gamma and STAT3, and cell proliferation

Title in the list of papers of Kehuan Wang's thesis: SUMOylation of PDGF receptor α affects signaling via PLCγ and STAT3 and promotes cell proliferation

Available from: 2023-02-11 Created: 2023-02-11 Last updated: 2023-10-05Bibliographically approved
Sarri, N., Papadopoulos, N., Lennartsson, J. & Heldin, C.-H. (2023). The E3 Ubiquitin Ligase TRIM21 Regulates Basal Levels of PDGFRβ. International Journal of Molecular Sciences, 24(9), Article ID 7782.
Open this publication in new window or tab >>The E3 Ubiquitin Ligase TRIM21 Regulates Basal Levels of PDGFRβ
2023 (English)In: International Journal of Molecular Sciences, ISSN 1661-6596, E-ISSN 1422-0067, Vol. 24, no 9, article id 7782Article in journal (Refereed) Published
Abstract [en]

Activation of platelet-derived growth factor (PDGF) receptors α and β (PDGFRα and PDGFRβ) at the cell surface by binding of PDGF isoforms leads to internalization of receptors, which affects the amplitude and kinetics of signaling. Ubiquitination of PDGF receptors in response to ligand stimulation is mediated by the Casitas b-lineage lymphoma (Cbl) family of ubiquitin ligases, promoting internalization and serving as a sorting signal for vesicular trafficking of receptors. We report here that another E3 ligase, i.e., tripartite motif-containing protein 21 (TRIM21), contributes to the ubiquitination of PDGFRβ in human primary fibroblasts AG1523 and the osteosarcoma cell line U2OS and regulates basal levels of PDGFRβ. We found that siRNA-mediated depletion of TRIM21 led to decreased ubiquitination of PDGFRβ in response to PDGF-BB stimulation, while internalization from the cell surface and the rate of ligand-induced degradation of the receptor were not affected. Moreover, induction of TRIM21 decreased the levels of PDGFRβ in serum-starved cells, and even more in growing cells, in the absence of PDGF stimulation. Consistently, siRNA knockdown of TRIM21 caused accumulation of the total amount of PDGFRβ, both in the cytoplasm and on the cell surface, without affecting mRNA levels of the receptor. We conclude that TRIM21 acts post-translationally and maintains basal levels of PDGFRβ, thus suggesting that ubiquitination of PDGFRβ by TRIM21 may direct a portion of receptor for degradation in growing cells in a ligand-independent manner.

Place, publisher, year, edition, pages
MDPI, 2023
Keywords
PDGFRβ, TRIM21, autophagy, Cbl, ubiquitination, receptor tyrosine kinase, degradation
National Category
Cell and Molecular Biology
Research subject
Medical Cell Biology; Medical Biochemistry
Identifiers
urn:nbn:se:uu:diva-489071 (URN)10.3390/ijms24097782 (DOI)000986934200001 ()37175489 (PubMedID)
Funder
Swedish Cancer Society, 222363PjH02H
Note

Title in the list of papers of Niki Sarri's thesis: The E3 ubiquitin ligase TRIM21 modulates the basal levels of PDGFRβ

Title in Web of Science: The E3 Ubiquitin Ligase TRIM21 Regulates Basal Levels of PDGFR beta

Available from: 2022-11-26 Created: 2022-11-26 Last updated: 2023-06-19Bibliographically approved
Tzavlaki, K., Ohata, Y., Morén, A., Watanabe, Y., Eriksson, J., Tsuchiya, M., . . . Moustakas, A. (2023). The liver kinase B1 supports mammary epithelial morphogenesis by inhibiting critical factors that mediate epithelial-mesenchymal transition. Journal of Cellular Physiology, 238(4), 790-812
Open this publication in new window or tab >>The liver kinase B1 supports mammary epithelial morphogenesis by inhibiting critical factors that mediate epithelial-mesenchymal transition
Show others...
2023 (English)In: Journal of Cellular Physiology, ISSN 0021-9541, E-ISSN 1097-4652, Vol. 238, no 4, p. 790-812Article in journal (Refereed) Published
Abstract [en]

The liver kinase B1 (LKB1) controls cellular metabolism and cell polarity across species. We previously established a mechanism for negative regulation of transforming growth factor β (TGFβ) signaling by LKB1. The impact of this mechanism in the context of epithelial polarity and morphogenesis remains unknown. After demonstrating that human mammary tissue expresses robust LKB1 protein levels, whereas invasive breast cancer exhibits significantly reduced LKB1 levels, we focused on mammary morphogenesis studies in three dimensional (3D) acinar organoids. CRISPR/Cas9-introduced loss-of-function mutations of STK11 (LKB1) led to profound defects in the formation of 3D organoids, resulting in amorphous outgrowth and loss of rotation of young organoids embedded in matrigel. This defect was associated with an enhanced signaling by TGFβ, including TGFβ auto-induction and induction of transcription factors that mediate epithelial-mesenchymal transition (EMT). Protein marker analysis confirmed a more efficient EMT response to TGFβ signaling in LKB1 knockout cells. Accordingly, chemical inhibition of the TGFβ type I receptor kinase largely restored the morphogenetic defect of LKB1 knockout cells. Similarly, chemical inhibition of the bone morphogenetic protein pathway or the TANK-binding kinase 1, or genetic silencing of the EMT factor SNAI1, partially restored the LKB1 knockout defect. Thus, LKB1 sustains mammary epithelial morphogenesis by limiting pathways that promote EMT. The observed downregulation of LKB1 expression in breast cancer is therefore predicted to associate with enhanced EMT induced by SNAI1 and TGFβ family members.

Place, publisher, year, edition, pages
John Wiley & Sons, 2023
Keywords
bone morphogenetic protein (BMP), epithelial-mesenchymal transition (EMT), liver kinase B1 (LKB1), morphogenesis, transforming growth factor beta (TGF beta)
National Category
Endocrinology and Diabetes Cancer and Oncology
Identifiers
urn:nbn:se:uu:diva-501860 (URN)10.1002/jcp.30975 (DOI)000933624600001 ()36791282 (PubMedID)
Funder
Magnus Bergvall FoundationSwedish Research CouncilSwedish Cancer SocietyEU, Horizon 2020Swedish Childhood Cancer Foundation, PR2020-0088Swedish Childhood Cancer Foundation, TJ2021-0078Hedlund foundationLars Hierta Memorial FoundationO.E. och Edla Johanssons vetenskapliga stiftelseSwedish Society of MedicineLudwig Institute for Cancer Research
Available from: 2023-05-22 Created: 2023-05-22 Last updated: 2023-05-22Bibliographically approved
Projects
Molecular mechanisms of the protumorigenic effects of TGFbeta [2015-02757_VR]; Uppsala UniversityMolecular mechanisms of TGFbeta signaling [2020-01291_VR]; Uppsala University; Publications
ten Dijke, P., Miyazono, K., Heldin, C.-H. & Moustakas, A. (2024). Special issue: TGF-β and epithelial-mesenchymal transition in cancer. Seminars in Cancer Biology, 102, 1-3
Molecular mechanisms of TGFbeta signaling [2024-03002_VR]; Uppsala University
Organisations
Identifiers
ORCID iD: ORCID iD iconorcid.org/0000-0002-9508-896x

Search in DiVA

Show all publications