Logo: to the web site of Uppsala University

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

Direct link
Moustakas, AristidisORCID iD iconorcid.org/0000-0001-9131-3827
Publications (10 of 137) 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
Moustakas, A. (2024). Crosstalk between TGF-β and EGF receptors via direct phosphorylation. Journal of Cell Biology, 223(4), Article ID e202403075.
Open this publication in new window or tab >>Crosstalk between TGF-β and EGF receptors via direct phosphorylation
2024 (English)In: Journal of Cell Biology, ISSN 0021-9525, E-ISSN 1540-8140, Vol. 223, no 4, article id e202403075Article in journal, Editorial material (Other academic) Published
Abstract [en]

Aristidis Moustakas discusses work from Ye-Guang Chen and colleagues (https://doi.org/10.1083/jcb.202307138) on a new mechanism by which TGF-β modulates HER2 signaling in mammary epithelia.

Place, publisher, year, edition, pages
Rockefeller University Press, 2024
National Category
Cell and Molecular Biology
Identifiers
urn:nbn:se:uu:diva-533852 (URN)10.1083/jcb.202403075 (DOI)001188538000001 ()38506732 (PubMedID)
Funder
Swedish Childhood Cancer Foundation, 2018-0091Swedish Cancer Society, CAN 2018/469Swedish Research Council, 2023-02865
Available from: 2024-07-01 Created: 2024-07-01 Last updated: 2024-07-01Bibliographically 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
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
Mendes Rodrigues Junior, D. & Moustakas, A. (2024). Unboxing the network among long non-coding RNAs and TGF-β signaling in cancer. Upsala Journal of Medical Sciences, 129(S1), Article ID e10614.
Open this publication in new window or tab >>Unboxing the network among long non-coding RNAs and TGF-β signaling in cancer
2024 (English)In: Upsala Journal of Medical Sciences, ISSN 0300-9734, E-ISSN 2000-1967, Vol. 129, no S1, article id e10614Article, review/survey (Refereed) Published
Abstract [en]

Deeper analysis of molecular mechanisms arising in tumor cells is an unmet need to provide new diagnostic and therapeutic strategies to prevent and treat tumors. The transforming growth factor β (TGF-β) signaling has been steadily featured in tumor biology and linked to poor prognosis of cancer patients. One pro-tumorigenic mechanism induced by TGF-β is the epithelial-to-mesenchymal transition (EMT), which can initiate cancer dissemination, enrich the tumor stem cell population, and increase chemoresistance. TGF-β signals via SMAD proteins, ubiquitin ligases, and protein kinases and modulates the expression of protein-coding and non-coding RNA genes, including those encoding larger than 500 nt transcripts, defined as long non-coding RNAs (lncRNAs). Several reports have shown lncRNAs regulating malignant phenotypes by directly affecting epigenetic processes, transcription, and post-transcriptional regulation. Thus, this review aims to update and summarize the impact of TGF-β signaling on the expression of lncRNAs and the function of such lncRNAs as regulators of TGF-β signaling, and how these networks might impact specific hallmarks of cancer.

Place, publisher, year, edition, pages
Upsala Medical Society, 2024
Keywords
Cancer, epigenetics, non-coding RNA, signal transduction, transforming growth factor-0
National Category
Cancer and Oncology Cell and Molecular Biology Cell Biology
Identifiers
urn:nbn:se:uu:diva-536798 (URN)10.48101/ujms.v129.10614 (DOI)001273186000001 ()38571882 (PubMedID)
Funder
Swedish Childhood Cancer Foundation, PR2018-0091Swedish Childhood Cancer Foundation, PR2023-0115Swedish Cancer Society, CAN2021/1506Pj01HSwedish Research Council, 2023-02865Lars Hierta Memorial Foundation, FO2023-0501O.E. och Edla Johanssons vetenskapliga stiftelse
Available from: 2024-08-27 Created: 2024-08-27 Last updated: 2024-08-27Bibliographically 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
Peleli, M., Antoniadou, I., Mendes Rodrigues Junior, D., Savvoulidou, O., Caja, L., Katsouda, A., . . . Papapetropoulos, A. (2023). Cystathionine gamma-lyase (CTH) inhibition attenuates glioblastoma formation. Redox Biology, 64, Article ID 102773.
Open this publication in new window or tab >>Cystathionine gamma-lyase (CTH) inhibition attenuates glioblastoma formation
Show others...
2023 (English)In: Redox Biology, E-ISSN 2213-2317, Vol. 64, article id 102773Article in journal (Refereed) Published
Abstract [en]

Purpose: Glioblastoma (GBM) is the most common type of adult brain tumor with extremely poor survival. Cystathionine-gamma lyase (CTH) is one of the main Hydrogen Sulfide (H2S) producing enzymes and its expression contributes to tumorigenesis and angiogenesis but its role in glioblastoma development remains poorly understood.

Methods: and Principal Results: An established allogenic immunocompetent in vivo GBM model was used in C57BL/6J WT and CTH KO mice where the tumor volume and tumor microvessel density were blindly measured by stereological analysis. Tumor macrophage and stemness markers were measured by blinded immunohisto-chemistry. Mouse and human GBM cell lines were used for cell-based analyses. In human gliomas, the CTH expression was analyzed by bioinformatic analysis on different databases.

In vivo, the genetic ablation of CTH in the host led to a significant reduction of the tumor volume and the protumorigenic and stemness transcription factor sex determining region Y-box 2 (SOX2). The tumor microvessel density (indicative of angiogenesis) and the expression levels of peritumoral macrophages showed no significant changes between the two genotypes. Bioinformatic analysis in human glioma tumors revealed that higher CTH expression is positively correlated to SOX2 expression and associated with worse overall survival in all grades of gliomas. Patients not responding to temozolomide have also higher CTH expression. In mouse or human GBM cells, pharmacological inhibition (PAG) or CTH knockdown (siRNA) attenuates GBM cell proliferation, migration and stem cell formation frequency.

Major Conclusions: Inhibition of CTH could be a new promising target against glioblastoma formation.

Place, publisher, year, edition, pages
Elsevier, 2023
Keywords
Cystathionine gamma-lyase (CTH), Glioblastoma stem cells (GSC), Brain blood vessels, Sex determining region Y -Box 2 (SOX2)
National Category
Cancer and Oncology Cell and Molecular Biology
Identifiers
urn:nbn:se:uu:diva-508161 (URN)10.1016/j.redox.2023.102773 (DOI)001021217700001 ()37300955 (PubMedID)
Funder
Swedish Research Council, 2019-00534Swedish Cancer Society, CAN2018/469Swedish Cancer Society, CAN2021/1506Pj01HNovo Nordisk, 0064142
Note

De två sista författarna delar sistaförfattarskapet.

Available from: 2023-08-09 Created: 2023-08-09 Last updated: 2024-01-04Bibliographically 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
Gélabert, C., Papoutsoglou, P., Cancela Golan, I., Ahlström, E., Ameur, A., Heldin, C.-H., . . . Moustakas, A. (2023). The long non-coding RNA LINC00707 interacts with Smad proteins to regulate TGFβ signaling and cancer cell invasion. Cell Communication and Signaling
Open this publication in new window or tab >>The long non-coding RNA LINC00707 interacts with Smad proteins to regulate TGFβ signaling and cancer cell invasion
Show others...
2023 (English)In: Cell Communication and Signaling, E-ISSN 1478-811XArticle in journal (Refereed) Published
Abstract [en]

Background: Long non-coding RNAs (lncRNAs) regulate cellular processes by interacting with RNAs or proteins. Transforming growth factor β (TGFβ) signaling via Smad proteins regulates gene networks that control diverse biological processes, including cancer cell migration. LncRNAs have emerged as TGFβ targets, yet, their mechanism of action and biological role in cancer remains poorly understood.

Methods: Whole-genome transcriptomics identified lncRNA genes regulated by TGFβ. Protein kinase inhibitors and RNA-silencing, in combination with cDNA cloning, provided loss- and gain-of-function analyses. Cancer cell-based assays coupled to RNA-immunoprecipitation and protein screening sought mechanistic evidence. Functional validation of TGFβ-regulated lncRNAs was based on new transcriptomics and by combining RNAscope with immunohistochemical analysis in tumor tissue.

Results: Transcriptomics of TGFβ signaling responses revealed down-regulation of the predominantly cytoplasmic long intergenic non-protein coding RNA 707 (LINC00707). Expression of LINC00707 required Smad and mitogen-activated protein kinase inputs. By limiting the binding of Krüppel-like factor 6 to the LINC00707 promoter, TGFβ led to LINC00707 repression. Functionally, LINC00707 suppressed cancer cell invasion, as well as key fibrogenic and pro-mesenchymal responses to TGFβ, as also attested by RNA-sequencing analysis. LINC00707 also suppressed Smad-dependent signaling. Mechanistically, LINC00707 interacted with and retained Smad proteins in the cytoplasm. Upon TGFβ stimulation, LINC00707 elimination allowed Smad accumulation in the nucleus. In vivo, LINC00707 expression was negatively correlated with Smad2 activation in tumor tissues.

Conclusions: TGFβ signaling decreases LINC00707 expression, which facilitates Smad-dependent signaling, favoring cancer cell invasion.

Place, publisher, year, edition, pages
BioMed Central (BMC), 2023
Keywords
cell invasion, lncRNA, signal transduction, RNA-protein interaction, transforming growth factor (TGFβ)
National Category
Cell and Molecular Biology
Research subject
Biomedical Laboratory Science; Biology with specialization in Molecular Cell Biology
Identifiers
urn:nbn:se:uu:diva-500360 (URN)10.1186/s12964-023-01273-3 (DOI)001079282800002 ()37784093 (PubMedID)
Funder
Ludwig Institute for Cancer ResearchSwedish Cancer Society, CAN 2015/438Swedish Cancer Society, CAN2018/469Swedish Cancer Society, CAN2021/1506Pj01HSwedish Research Council, 2017-01588Swedish Research Council, 2018-02757Swedish Research Council, 2015-02757Swedish Research Council, 2020-01291EU, European Research Council, 787472O.E. och Edla Johanssons vetenskapliga stiftelseHedlund foundation, M2019-1065Swedish Society of Medicine, SLS-887701Magnus Bergvall Foundation, 2019-03444Magnus Bergvall Foundation, 2020-03781Uppsala University
Available from: 2023-04-14 Created: 2023-04-14 Last updated: 2024-01-12Bibliographically approved
Mendes Rodrigues Junior, D., Raminelli, C., Hassanie, H., Trossini, G. H., Perecim, G. P., Caja, L., . . . Vettore, A. L. (2022). Aporphine and isoquinoline derivatives block glioblastoma cell stemness and enhance temozolomide cytotoxicity. Scientific Reports, 12(1), 21113, Article ID 21113.
Open this publication in new window or tab >>Aporphine and isoquinoline derivatives block glioblastoma cell stemness and enhance temozolomide cytotoxicity
Show others...
2022 (English)In: Scientific Reports, E-ISSN 2045-2322, Vol. 12, no 1, p. 21113-, article id 21113Article in journal (Refereed) Published
Abstract [en]

Glioblastoma (GBM) is the most aggressive and common primary malignant brain tumor with limited available therapeutic approaches. Despite improvements in therapeutic options for GBM patients, efforts to develop new successful strategies remain as major unmet medical needs. Based on the cytotoxic properties of aporphine compounds, we evaluated the biological effect of 12 compounds obtained through total synthesis of ( ±)-apomorphine hydrochloride (APO) against GBM cells. The compounds 2,2,2-trifluoro-1-(1-methylene-3,4-dihydroisoquinolin-2(1H)-yl)ethenone (A5) and ( ±)-1-(10,11-dimethoxy-6a,7-dihydro-4H-dibenzo[de,g]quinolin-6(5H)-yl)ethenone (C1) reduced the viability of GBM cells, with 50% inhibitory concentration ranging from 18 to 48 μM in patient-derived GBM cultures. Our data show that APO, A5 or C1 modulate the expression of DNA damage and apoptotic markers, impair 3D-gliomasphere growth and reduce the expression of stemness markers. Potential activity and protein targets of A5, C1 or APO were predicted in silico based on PASS and SEA software. Dopamine receptors (DRD1 and 5), CYP2B6, CYP2C9 and ABCB1, whose transcripts were differentially expressed in the GBM cells, were among the potential A5 or C1 target proteins. Docking analyses (HQSAR and 3D-QSAR) were performed to characterize possible interactions of ABCB1 and CYP2C9 with the compounds. Notably, A5 or C1 treatment, but not temozolomide (TMZ), reduced significantly the levels of extracellular ATP, suggesting ABCB1 negative regulation, which was correlated with stronger cytotoxicity induced by the combination of TMZ with A5 or C1 on GBM cells. Hence, our data reveal a potential therapeutic application of A5 and C1 as cytotoxic agents against GBM cells and predicted molecular networks that can be further exploited to characterize the pharmacological effects of these isoquinoline-containing substances.

Place, publisher, year, edition, pages
Springer Nature, 2022
National Category
Cell and Molecular Biology Cancer and Oncology
Identifiers
urn:nbn:se:uu:diva-490571 (URN)10.1038/s41598-022-25534-2 (DOI)000905463900045 ()36477472 (PubMedID)
Funder
Swedish Cancer Society, CAN2018/469Swedish Cancer Society, CAN2021/1506Pj01HSwedish Research Council, 2018-02757
Available from: 2022-12-16 Created: 2022-12-16 Last updated: 2023-04-03Bibliographically approved
Projects
TGF-beta signaling as a determinant of stroma remodeling and immune response in lung cancer [2021-02693_VR]; Uppsala UniversityRole of TGFβ-induced EMT in tumor stemness and treatment-resistance [2023-02865_VR]; Uppsala University
Organisations
Identifiers
ORCID iD: ORCID iD iconorcid.org/0000-0001-9131-3827

Search in DiVA

Show all publications