98 results on '"Sangfelt O"'
Search Results
2. FBXW7 regulates glucocorticoid response in T-cell acute lymphoblastic leukaemia by targeting the glucocorticoid receptor for degradation
- Author
-
Malyukova, A, Brown, S, Papa, R, O'Brien, R, Giles, J, Trahair, T N, Dalla Pozza, L, Sutton, R, Liu, T, Haber, M, Norris, M D, Lock, R B, Sangfelt, O, and Marshall, G M
- Published
- 2013
- Full Text
- View/download PDF
3. FBXO44 promotes DNA replication-coupled repetitive element silencing in cancer cells
- Author
-
Shen, J.Z., Qiu, Z., Wu, Q., Finlay, D., Garcia, G., Sun, D., Rantala, J., Barshop, W., Hope, J.L., Gimple, R.C., Sangfelt, O., Bradley, L.M., Wohlschlegel, J., Rich, J.N., and Spruck, C.
- Subjects
DNA Replication ,Adult ,Male ,Cell Survival ,Drug Resistance ,Methylation ,SUV39H1 ,Medical and Health Sciences ,Repetitive Sequences ,Cell Line ,H3K9me3 ,Histones ,Double-Stranded ,Genetic ,Neoplasms ,Genetics ,Humans ,2.1 Biological and endogenous factors ,Aetiology ,Immune Checkpoint Inhibitors ,Cell Proliferation ,Cancer ,Neoplastic ,Tumor ,Nucleic Acid ,Lysine ,F-Box Proteins ,DNA Breaks ,Immunity ,FBXO44 ,Middle Aged ,Biological Sciences ,Nucleosomes ,Neoplasm Proteins ,repetitive elements ,Treatment Outcome ,Infectious Diseases ,Gene Expression Regulation ,Neoplasm ,Female ,Interferons ,immunotherapy ,Transcription ,Signal Transduction ,Biotechnology ,Developmental Biology - Abstract
Repetitive elements (REs) compose ∼50% of the human genome and are normally transcriptionally silenced, although the mechanism has remained elusive. Through an RNAi screen, we identified FBXO44 as an essential repressor of REs in cancer cells. FBXO44 bound H3K9me3-modified nucleosomes at the replication fork and recruited SUV39H1, CRL4, and Mi-2/NuRD to transcriptionally silence REs post-DNA replication. FBXO44/SUV39H1 inhibition reactivated REs, leading to DNA replication stress and stimulation of MAVS/STING antiviral pathways and interferon (IFN) signaling in cancer cells to promote decreased tumorigenicity, increased immunogenicity, and enhanced immunotherapy response. FBXO44 expression inversely correlated with replication stress, antiviral pathways, IFN signaling, and cytotoxic Tcell infiltration in human cancers, while a FBXO44-immune gene signature correlated with improved immunotherapy response in cancer patients. FBXO44/SUV39H1 were dispensable in normal cells. Collectively, FBXO44/SUV39H1 are crucial repressors of RE transcription, and their inhibition selectively induces DNA replication stress and viral mimicry in cancer cells.
- Published
- 2021
4. A novel combination therapy targeting ubiquitin-specific protease 5 in MYCN-driven neuroblastoma
- Author
-
Cheung, BB, Kleynhans, A, Mittra, R, Kim, PY, Holien, JK, Nagy, Z, Ciampa, OC, Seneviratne, JA, Mayoh, C, Raipuria, M, Gadde, S, Massudi, H, Wong, IPL, Tan, O, Gong, A, Suryano, A, Diakiw, SM, Liu, B, Arndt, GM, Liu, T, Kumar, N, Sangfelt, O, Zhu, S, Norris, MD, Haber, M, Carter, DR, Parker, MW, Marshall, GM, Cheung, BB, Kleynhans, A, Mittra, R, Kim, PY, Holien, JK, Nagy, Z, Ciampa, OC, Seneviratne, JA, Mayoh, C, Raipuria, M, Gadde, S, Massudi, H, Wong, IPL, Tan, O, Gong, A, Suryano, A, Diakiw, SM, Liu, B, Arndt, GM, Liu, T, Kumar, N, Sangfelt, O, Zhu, S, Norris, MD, Haber, M, Carter, DR, Parker, MW, and Marshall, GM
- Abstract
Histone deacetylase (HDAC) inhibitors are effective in MYCN-driven cancers, because of a unique need for HDAC recruitment by the MYCN oncogenic signal. However, HDAC inhibitors are much more effective in combination with other anti-cancer agents. To identify novel compounds which act synergistically with HDAC inhibitor, such as suberanoyl hydroxamic acid (SAHA), we performed a cell-based, high-throughput drug screen of 10,560 small molecule compounds from a drug-like diversity library and identified a small molecule compound (SE486-11) which synergistically enhanced the cytotoxic effects of SAHA. Effects of drug combinations on cell viability, proliferation, apoptosis and colony forming were assessed in a panel of neuroblastoma cell lines. Treatment with SAHA and SE486-11 increased MYCN ubiquitination and degradation, and markedly inhibited tumorigenesis in neuroblastoma xenografts, and, MYCN transgenic zebrafish and mice. The combination reduced ubiquitin-specific protease 5 (USP5) levels and increased unanchored polyubiquitin chains. Overexpression of USP5 rescued neuroblastoma cells from the cytopathic effects of the combination and reduced unanchored polyubiquitin, suggesting USP5 is a therapeutic target of the combination. SAHA and SE486-11 directly bound to USP5 and the drug combination exhibited a 100-fold higher binding to USP5 than individual drugs alone in microscale thermophoresis assays. MYCN bound to the USP5 promoter and induced USP5 gene expression suggesting that USP5 and MYCN expression created a forward positive feedback loop in neuroblastoma cells. Thus, USP5 acts as an oncogenic cofactor with MYCN in neuroblastoma and the novel combination of HDAC inhibitor with SE486-11 represents a novel therapeutic approach for the treatment of MYCN-driven neuroblastoma.
- Published
- 2021
5. The Transcription Map of the 13q14 Region Frequently Deleted in B-cell Chronic Lymphocytic Leukemia
- Author
-
Tyazhelova, T. V., Ivanov, D. V., Makeeva, N. V., Kapanadze, B. I., Nikitin, E. A., Semov, A. B., Sangfelt, O., Grander, D., Vorobiev, A. I., Einhorn, S., Yankovsky, N. K., and Baranova, A. V.
- Published
- 2001
- Full Text
- View/download PDF
6. Apoptosis and cell growth inhibition as antitumor effector functions of interferons
- Author
-
Sangfelt, O. and Strander, H.
- Published
- 2001
- Full Text
- View/download PDF
7. Oncoprotein stabilization in brain tumors
- Author
-
Hede, Sanna-Maria, Savov, Vasil, Weishaupt, Holger, Sangfelt, O., Swartling, Fredrik Johansson, Hede, Sanna-Maria, Savov, Vasil, Weishaupt, Holger, Sangfelt, O., and Swartling, Fredrik Johansson
- Abstract
Proteins involved in promoting cell proliferation and viability need to be timely expressed and carefully controlled for the proper development of the brain but also efficiently degraded in order to prevent cells from becoming brain cancer cells. A major pathway for targeted protein degradation in cells is the ubiquitin-proteasome system (UPS). Oncoproteins that drive tumor development and tumor maintenance are often deregulated and stabilized in malignant cells. This can occur when oncoproteins escape degradation by the UPS because of mutations in either the oncoprotein itself or in the UPS components responsible for recognition and ubiquitylation of the oncoprotein. As the pathogenic accumulation of an oncoprotein can lead to effectively sustained cell growth, viability and tumor progression, it is an indisputable target for cancer treatment. The most common types of malignant brain tumors in children and adults are medulloblastoma and glioma, respectively. Here, we review different ways of how deregulated proteolysis of oncoproteins involved in major signaling cancer pathways contributes to medulloblastoma and glioma development. We also describe means of targeting relevant oncoproteins in brain tumors with treatments affecting their stability or therapeutic strategies directed against the UPS itself.
- Published
- 2014
- Full Text
- View/download PDF
8. CDK-mediated activation of the SCFFBXO28 ubiquitin ligase promotes MYC-driven transcription and tumourigenesis and predicts poor survival in breast cancer
- Author
-
Cepeda, D., Ng, H. -F, Sharifi, H. R., Mahmoudi, S., Cerrato, V. S., Fredlund, E., Magnusson, K., Nilsson, H., Malyukova, A., Rantala, J., Klevebring, D., Viñals, F., Bhaskaran, N., Zakaria, S. M., Rahmanto, A. S., Grotegut, S., Nielsen, M. L., Al-Khalili Szigyarto, Cristina, Sun, D., Lerner, M., Navani, S., Widschwendter, M., Uhlén, Mathias, Jirström, K., Pontén, F., Wohlschlegel, J., Grandér, D., Spruck, C., Larsson, L. -G, Sangfelt, O., Cepeda, D., Ng, H. -F, Sharifi, H. R., Mahmoudi, S., Cerrato, V. S., Fredlund, E., Magnusson, K., Nilsson, H., Malyukova, A., Rantala, J., Klevebring, D., Viñals, F., Bhaskaran, N., Zakaria, S. M., Rahmanto, A. S., Grotegut, S., Nielsen, M. L., Al-Khalili Szigyarto, Cristina, Sun, D., Lerner, M., Navani, S., Widschwendter, M., Uhlén, Mathias, Jirström, K., Pontén, F., Wohlschlegel, J., Grandér, D., Spruck, C., Larsson, L. -G, and Sangfelt, O.
- Abstract
SCF (Skp1/Cul1/F-box) ubiquitin ligases act as master regulators of cellular homeostasis by targeting key proteins for ubiquitylation. Here, we identified a hitherto uncharacterized F-box protein, FBXO28 that controls MYC-dependent transcription by non-proteolytic ubiquitylation. SCFFBXO28 activity and stability are regulated during the cell cycle by CDK1/2-mediated phosphorylation of FBXO28, which is required for its efficient ubiquitylation of MYC and downsteam enhancement of the MYC pathway. Depletion of FBXO28 or overexpression of an F-box mutant unable to support MYC ubiquitylation results in an impairment of MYC-driven transcription, transformation and tumourigenesis. Finally, in human breast cancer, high FBXO28 expression and phosphorylation are strong and independent predictors of poor outcome. In conclusion, our data suggest that SCFFBXO28 plays an important role in transmitting CDK activity to MYC function during the cell cycle, emphasizing the CDK-FBXO28-MYC axis as a potential molecular drug target in MYC-driven cancers, including breast cancer. FBXO28 is identified as part of a SCF complex acting as a regulator of tumor cell proliferation and an important modifier of MYC function. FBXO28 may be a new prognostic factor in breast cancer and a new potential drug target in MYC- driven tumors., QC 20131120
- Published
- 2013
- Full Text
- View/download PDF
9. Proteomic screen reveals Fbw7 as a modulator of the NF-kappa B pathway
- Author
-
Arabi, A., Ullah, K., Branca, R. M. M., Johansson, J., Bandarra, D., Haneklaus, M., Fu, J., Ariës, I., Nilsson, Peter, Den Boer, M. L., Pokrovskaja, K., Grandér, D., Xiao, G., Rocha, S., Lehtiö, J., Sangfelt, O., Arabi, A., Ullah, K., Branca, R. M. M., Johansson, J., Bandarra, D., Haneklaus, M., Fu, J., Ariës, I., Nilsson, Peter, Den Boer, M. L., Pokrovskaja, K., Grandér, D., Xiao, G., Rocha, S., Lehtiö, J., and Sangfelt, O.
- Abstract
Fbw7 is a ubiquitin-ligase that targets several oncoproteins for proteolysis, but the full range of Fbw7 substrates is not known. Here we show that by performing quantitative proteomics combined with degron motif searches, we effectively screened for a more complete set of Fbw7 targets. We identify 89 putative Fbw7 substrates, including several disease-associated proteins. The transcription factor NF-κB2 (p100/p52) is one of the candidate Fbw7 substrates. We show that Fbw7 interacts with p100 via a conserved degron and that it promotes degradation of p100 in a GSK3 2 phosphorylation-dependent manner. Fbw7 inactivation increases p100 levels, which in the presence of NF-κB pathway stimuli, leads to increased p52 levels and activity. Accordingly, the apoptotic threshold can be increased by loss of Fbw7 in a p100-dependent manner. In conclusion, Fbw7-mediated destruction of p100 is a regulatory component restricting the response to NF-κB2 pathway stimulation., QC 20120903
- Published
- 2012
- Full Text
- View/download PDF
10. Oncoprotein stabilization in brain tumors
- Author
-
Hede, S-M, primary, Savov, V, additional, Weishaupt, H, additional, Sangfelt, O, additional, and Swartling, F J, additional
- Published
- 2013
- Full Text
- View/download PDF
11. FBXW7 regulates glucocorticoid response in T-cell acute lymphoblastic leukaemia by targeting the glucocorticoid receptor for degradation
- Author
-
Malyukova, A, primary, Brown, S, additional, Papa, R, additional, O'Brien, R, additional, Giles, J, additional, Trahair, T N, additional, Dalla Pozza, L, additional, Sutton, R, additional, Liu, T, additional, Haber, M, additional, Norris, M D, additional, Lock, R B, additional, Sangfelt, O, additional, and Marshall, G M, additional
- Published
- 2012
- Full Text
- View/download PDF
12. Notch signaling induces SKP2 expression and promotes reduction of p27Kip1 in T-cell acute lymphoblastic leukemia cell lines
- Author
-
DOHDA, T, primary, MALJUKOVA, A, additional, LIU, L, additional, HEYMAN, M, additional, GRANDER, D, additional, BRODIN, D, additional, SANGFELT, O, additional, and LENDAHL, U, additional
- Published
- 2007
- Full Text
- View/download PDF
13. Factors Influencing the Response to Interferon Therapy in Chronic Hepatitis C Studies on Viral Genotype and Induction of 2′,5′-Oligoadenylate Synthetase in the Liver and Peripheral Blood Cells
- Author
-
Grandér, D., primary, Hultcrantz, R., additional, Weiland, O., additional, Xu, B., additional, Sangfelt, O., additional, Björklund, A.-C., additional, Befrits, R., additional, Björkholm, M., additional, Gruber, A., additional, Kinnman, N., additional, Reichard, O., additional, Widell, A., additional, and Einhorn, S., additional
- Published
- 1996
- Full Text
- View/download PDF
14. Primary leukemia cells resistant to alpha-interferon in vitro are defective in the activation of the DNA-binding factor interferon- stimulated gene factor 3
- Author
-
Xu, B, primary, Grander, D, additional, Sangfelt, O, additional, and Einhorn, S, additional
- Published
- 1994
- Full Text
- View/download PDF
15. In Vivo Induction of the Interferon-Stimulated Protein 2'5'-Oligoadenylate Synthetase in Tumor and Peripheral Blood Cells During IFN-a Treatment of Metastatic Melanoma
- Author
-
Grander, D., Sangfelt, O., Skoog, L., and Hansson, J.
- Published
- 1998
16. Wild-type p53-induced apoptosis in a Burkill lymphoma cell line is inhibited by interferon gamma
- Author
-
Sangfelt, O., Einhorn, S., Bjorklund, A.C., Wiman, K.G., Okan, I., and Grander, D.
- Subjects
Tumor suppressor genes -- Physiological aspects ,Burkitt's lymphoma -- Development and progression ,Health ,Physiological aspects ,Development and progression - Abstract
According to the authors' abstract of an article published in International Journal of Cancer, 'The tumor suppressor p53 plays a central role in negative growth control, including growth arrest and [...]
- Published
- 1996
17. Retraction Notice to: A FBXO7/EYA2-SCF FBXW7 axis promotes AXL-mediated maintenance of mesenchymal and immune evasion phenotypes of cancer cells.
- Author
-
Shen JZ, Qiu Z, Wu Q, Zhang G, Harris R, Sun D, Rantala J, Barshop WD, Zhao L, Lv D, Won KA, Wohlschlegel J, Sangfelt O, Laman H, Rich JN, and Spruck C
- Published
- 2024
- Full Text
- View/download PDF
18. Sequential drug treatment targeting cell cycle and cell fate regulatory programs blocks non-genetic cancer evolution in acute lymphoblastic leukemia.
- Author
-
Malyukova A, Lahnalampi M, Falqués-Costa T, Pölönen P, Sipola M, Mehtonen J, Teppo S, Akopyan K, Viiliainen J, Lohi O, Hagström-Andersson AK, Heinäniemi M, and Sangfelt O
- Subjects
- Humans, Histone-Lysine N-Methyltransferase genetics, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Cell Cycle Proteins antagonists & inhibitors, Cell Line, Tumor, Pyrimidines pharmacology, Pyrimidines therapeutic use, Pyrimidinones pharmacology, Pyrimidinones therapeutic use, Myeloid-Lymphoid Leukemia Protein genetics, Pyrazoles pharmacology, Pyrazoles therapeutic use, Protein-Tyrosine Kinases antagonists & inhibitors, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Cell Cycle drug effects, Core Binding Factor Alpha 2 Subunit genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
Background: Targeted therapies exploiting vulnerabilities of cancer cells hold promise for improving patient outcome and reducing side-effects of chemotherapy. However, efficacy of precision therapies is limited in part because of tumor cell heterogeneity. A better mechanistic understanding of how drug effect is linked to cancer cell state diversity is crucial for identifying effective combination therapies that can prevent disease recurrence., Results: Here, we characterize the effect of G2/M checkpoint inhibition in acute lymphoblastic leukemia (ALL) and demonstrate that WEE1 targeted therapy impinges on cell fate decision regulatory circuits. We find the highest inhibition of recovery of proliferation in ALL cells with KMT2A-rearrangements. Single-cell RNA-seq and ATAC-seq of RS4;11 cells harboring KMT2A::AFF1, treated with the WEE1 inhibitor AZD1775, reveal diversification of cell states, with a fraction of cells exhibiting strong activation of p53-driven processes linked to apoptosis and senescence, and disruption of a core KMT2A-RUNX1-MYC regulatory network. In this cell state diversification induced by WEE1 inhibition, a subpopulation transitions to a drug tolerant cell state characterized by activation of transcription factors regulating pre-B cell fate, lipid metabolism, and pre-BCR signaling in a reversible manner. Sequential treatment with BCR-signaling inhibitors dasatinib, ibrutinib, or perturbing metabolism by fatostatin or AZD2014 effectively counteracts drug tolerance by inducing cell death and repressing stemness markers., Conclusions: Collectively, our findings provide new insights into the tight connectivity of gene regulatory programs associated with cell cycle and cell fate regulation, and a rationale for sequential administration of WEE1 inhibitors with low toxicity inhibitors of pre-BCR signaling or metabolism., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
19. FBXL12 degrades FANCD2 to regulate replication recovery and promote cancer cell survival under conditions of replication stress.
- Author
-
Brunner A, Li Q, Fisicaro S, Kourtesakis A, Viiliäinen J, Johansson HJ, Pandey V, Mayank AK, Lehtiö J, Wohlschlegel JA, Spruck C, Rantala JK, Orre LM, and Sangfelt O
- Subjects
- Humans, Cell Survival genetics, Chromatin genetics, Cyclin E genetics, Cyclin E metabolism, DNA Damage, DNA Repair, DNA Replication genetics, Fanconi Anemia Complementation Group D2 Protein genetics, Fanconi Anemia Complementation Group D2 Protein metabolism, Fanconi Anemia metabolism, Neoplasms genetics
- Abstract
Fanconi anemia (FA) signaling, a key genomic maintenance pathway, is activated in response to replication stress. Here, we report that phosphorylation of the pivotal pathway protein FANCD2 by CHK1 triggers its FBXL12-dependent proteasomal degradation, facilitating FANCD2 clearance at stalled replication forks. This promotes efficient DNA replication under conditions of CYCLIN E- and drug-induced replication stress. Reconstituting FANCD2-deficient fibroblasts with phosphodegron mutants failed to re-establish fork progression. In the absence of FBXL12, FANCD2 becomes trapped on chromatin, leading to replication stress and excessive DNA damage. In human cancers, FBXL12, CYCLIN E, and FA signaling are positively correlated, and FBXL12 upregulation is linked to reduced survival in patients with high CYCLIN E-expressing breast tumors. Finally, depletion of FBXL12 exacerbated oncogene-induced replication stress and sensitized cancer cells to drug-induced replication stress by WEE1 inhibition. Collectively, our results indicate that FBXL12 constitutes a vulnerability and a potential therapeutic target in CYCLIN E-overexpressing cancers., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
20. Dormant SOX9-Positive Cells Facilitate MYC-Driven Recurrence of Medulloblastoma.
- Author
-
Borgenvik A, Holmberg KO, Bolin S, Zhao M, Savov V, Rosén G, Hutter S, Garancher A, Rahmanto AS, Bergström T, Olsen TK, Mainwaring OJ, Sattanino D, Verbaan AD, Rusert JM, Sundström A, Bravo MB, Dang Y, Wenz AS, Richardson S, Fotaki G, Hill RM, Dubuc AM, Kalushkova A, Remke M, Čančer M, Jernberg-Wiklund H, Giraud G, Chen X, Taylor MD, Sangfelt O, Clifford SC, Schüller U, Wechsler-Reya RJ, Weishaupt H, and Swartling FJ
- Subjects
- Animals, Humans, Mice, Neoplasm Recurrence, Local genetics, Proto-Oncogene Proteins c-myc genetics, Proto-Oncogene Proteins c-myc metabolism, SOX9 Transcription Factor genetics, SOX9 Transcription Factor metabolism, Transcription Factors metabolism, Brain Neoplasms, Cerebellar Neoplasms pathology, Medulloblastoma pathology
- Abstract
Relapse is the leading cause of death in patients with medulloblastoma, the most common malignant pediatric brain tumor. A better understanding of the mechanisms underlying recurrence could lead to more effective therapies for targeting tumor relapses. Here, we observed that SOX9, a transcription factor and stem cell/glial fate marker, is limited to rare, quiescent cells in high-risk medulloblastoma with MYC amplification. In paired primary-recurrent patient samples, SOX9-positive cells accumulated in medulloblastoma relapses. SOX9 expression anti-correlated with MYC expression in murine and human medulloblastoma cells. However, SOX9-positive cells were plastic and could give rise to a MYC high state. To follow relapse at the single-cell level, an inducible dual Tet model of medulloblastoma was developed, in which MYC expression was redirected in vivo from treatment-sensitive bulk cells to dormant SOX9-positive cells using doxycycline treatment. SOX9 was essential for relapse initiation and depended on suppression of MYC activity to promote therapy resistance, epithelial-mesenchymal transition, and immune escape. p53 and DNA repair pathways were downregulated in recurrent tumors, whereas MGMT was upregulated. Recurrent tumor cells were found to be sensitive to treatment with an MGMT inhibitor and doxorubicin. These findings suggest that recurrence-specific targeting coupled with DNA repair inhibition comprises a potential therapeutic strategy in patients affected by medulloblastoma relapse., Significance: SOX9 facilitates therapy escape and recurrence in medulloblastoma via temporal inhibition of MYC/MYCN genes, revealing a strategy to specifically target SOX9-positive cells to prevent tumor relapse., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
21. A FBXO7/EYA2-SCF FBXW7 axis promotes AXL-mediated maintenance of mesenchymal and immune evasion phenotypes of cancer cells.
- Author
-
Shen JZ, Qiu Z, Wu Q, Zhang G, Harris R, Sun D, Rantala J, Barshop WD, Zhao L, Lv D, Won KA, Wohlschlegel J, Sangfelt O, Laman H, Rich JN, and Spruck C
- Subjects
- Animals, Cell Line, Tumor, Homeodomain Proteins genetics, Humans, Immune Evasion, Intracellular Signaling Peptides and Proteins metabolism, Mice, Nuclear Proteins metabolism, Phenotype, Protein Tyrosine Phosphatases genetics, Ubiquitin metabolism, F-Box Proteins genetics, F-Box Proteins metabolism, F-Box-WD Repeat-Containing Protein 7 genetics, Neoplasms genetics
- Abstract
A mesenchymal tumor phenotype associates with immunotherapy resistance, although the mechanism is unclear. Here, we identified FBXO7 as a maintenance regulator of mesenchymal and immune evasion phenotypes of cancer cells. FBXO7 bound and stabilized SIX1 co-transcriptional regulator EYA2, stimulating mesenchymal gene expression and suppressing IFNα/β, chemokines CXCL9/10, and antigen presentation machinery, driven by AXL extracellular ligand GAS6. Ubiquitin ligase SCF
FBXW7 antagonized this pathway by promoting EYA2 degradation. Targeting EYA2 Tyr phosphatase activity decreased mesenchymal phenotypes and enhanced cancer cell immunogenicity, resulting in attenuated tumor growth and metastasis, increased infiltration of cytotoxic T and NK cells, and enhanced anti-PD-1 therapy response in mouse tumor models. FBXO7 expression correlated with mesenchymal and immune-suppressive signatures in patients with cancer. An FBXO7-immune gene signature predicted immunotherapy responses. Collectively, the FBXO7/EYA2-SCFFBXW7 axis maintains mesenchymal and immune evasion phenotypes of cancer cells, providing rationale to evaluate FBXO7/EYA2 inhibitors in combination with immune-based therapies to enhance onco-immunotherapy responses., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
22. PTENP1-AS contributes to BRAF inhibitor resistance and is associated with adverse clinical outcome in stage III melanoma.
- Author
-
Vidarsdottir L, Azimi A, Das I, Sigvaldadottir I, Suryo Rahmanto A, Petri A, Kauppinen S, Ingvar C, Jönsson G, Olsson H, Frostvik Stolt M, Tuominen R, Sangfelt O, Pokrovskaja Tamm K, Hansson J, Grandér D, Egyházi Brage S, and Johnsson P
- Subjects
- Humans, Cell Line, Tumor, Neoplasm Staging, Gene Expression Regulation, Neoplastic, Skin Neoplasms drug therapy, Skin Neoplasms genetics, Skin Neoplasms pathology, Skin Neoplasms mortality, Protein Kinase Inhibitors therapeutic use, Protein Kinase Inhibitors pharmacology, RNA, Antisense genetics, Prognosis, Enhancer of Zeste Homolog 2 Protein metabolism, Enhancer of Zeste Homolog 2 Protein genetics, Enhancer of Zeste Homolog 2 Protein antagonists & inhibitors, Promoter Regions, Genetic, Female, Male, Melanoma genetics, Melanoma drug therapy, Melanoma pathology, Melanoma mortality, Melanoma metabolism, Proto-Oncogene Proteins B-raf genetics, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Drug Resistance, Neoplasm genetics, PTEN Phosphohydrolase genetics, PTEN Phosphohydrolase metabolism
- Abstract
BRAF inhibitors (BRAFi) selectively target oncogenic BRAF
V600E/K and are effective in 80% of advanced cutaneous malignant melanoma cases carrying the V600 mutation. However, the development of drug resistance limits their clinical efficacy. Better characterization of the underlying molecular processes is needed to further improve treatments. We previously demonstrated that transcription of PTEN is negatively regulated by the PTEN pseudogene antisense RNA, PTENP1-AS, and here we investigated the impact of this transcript on clinical outcome and BRAFi resistance in melanoma. We observed that increased expression levels of PTENP1-AS in BRAFi resistant cells associated with enrichment of EZH2 and H3K27me3 at the PTEN promoter, consequently reducing the expression levels of PTEN. Further, we showed that targeting of the PTENP1-AS transcript sensitized resistant cells to BRAFi treatment and that high expression of PTENP1-AS in stage III melanoma correlated with poor survival. Collectively, the data presented here show that PTENP1-AS is a promising target for re-sensitizing cells to BRAFi and also a possible prognostic marker for clinical outcome in stage III melanoma.- Published
- 2021
- Full Text
- View/download PDF
23. Ex vivo analysis of DNA repair targeting in extreme rare cutaneous apocrine sweat gland carcinoma.
- Author
-
Mäkelä R, Härmä V, Badra Fajardo N, Wells G, Lygerou Z, Sangfelt O, Kononen J, and Rantala JK
- Abstract
Cutaneous apocrine carcinoma is an extreme rare malignancy derived from a sweat gland. Histologically sweat gland cancers resemble metastatic mammary apocrine carcinomas, but the genetic landscape remains poorly understood. Here, we report a rare metastatic case with a PALB2 aberration identified previously as a familial susceptibility gene for breast cancer in the Finnish population. As PALB2 exhibits functions in the BRCA1/2-RAD51-dependent homologous DNA recombination repair pathway, we sought to use ex vivo functional screening to explore sensitivity of the tumor cells to therapeutic targeting of DNA repair. Drug screening suggested sensitivity of the PALB2 deficient cells to BET-bromodomain inhibition, and modest sensitivity to DNA-PKi, ATRi, WEE1i and PARPi. A phenotypic RNAi screen of 300 DNA repair genes was undertaken to assess DNA repair targeting in more detail. Core members of the HR and MMEJ pathways were identified to be essential for viability of the cells. RNAi inhibition of RAD52-dependent HR on the other hand potentiated the efficacy of a novel BETi ODM-207. Together these results describe the first ever CAC case with a BRCAness genetic background, evaluate combinatorial DNA repair targeting, and provide a data resource for further analyses of DNA repair targeting in PALB2 deficient cancers., Competing Interests: CONFLICTS OF INTEREST Juha K. Rantala is the founder of Misvik Biology Oy. The authors of this manuscript declare that they have no competing interests., (Copyright: © 2021 Mäkelä et al.)
- Published
- 2021
- Full Text
- View/download PDF
24. A novel combination therapy targeting ubiquitin-specific protease 5 in MYCN-driven neuroblastoma.
- Author
-
Cheung BB, Kleynhans A, Mittra R, Kim PY, Holien JK, Nagy Z, Ciampa OC, Seneviratne JA, Mayoh C, Raipuria M, Gadde S, Massudi H, Wong IPL, Tan O, Gong A, Suryano A, Diakiw SM, Liu B, Arndt GM, Liu T, Kumar N, Sangfelt O, Zhu S, Norris MD, Haber M, Carter DR, Parker MW, and Marshall GM
- Subjects
- Animals, Animals, Genetically Modified genetics, Apoptosis drug effects, Cell Proliferation drug effects, Cell Survival drug effects, Drug Evaluation, Preclinical, Gene Expression Regulation, Neoplastic drug effects, Heterografts, Humans, Mice, Neuroblastoma genetics, Neuroblastoma pathology, Small Molecule Libraries pharmacology, Vorinostat pharmacology, Zebrafish genetics, Carcinogenesis drug effects, Histone Deacetylase Inhibitors pharmacology, N-Myc Proto-Oncogene Protein genetics, Neuroblastoma drug therapy, Ubiquitin-Specific Proteases genetics, Zebrafish Proteins genetics
- Abstract
Histone deacetylase (HDAC) inhibitors are effective in MYCN-driven cancers, because of a unique need for HDAC recruitment by the MYCN oncogenic signal. However, HDAC inhibitors are much more effective in combination with other anti-cancer agents. To identify novel compounds which act synergistically with HDAC inhibitor, such as suberanoyl hydroxamic acid (SAHA), we performed a cell-based, high-throughput drug screen of 10,560 small molecule compounds from a drug-like diversity library and identified a small molecule compound (SE486-11) which synergistically enhanced the cytotoxic effects of SAHA. Effects of drug combinations on cell viability, proliferation, apoptosis and colony forming were assessed in a panel of neuroblastoma cell lines. Treatment with SAHA and SE486-11 increased MYCN ubiquitination and degradation, and markedly inhibited tumorigenesis in neuroblastoma xenografts, and, MYCN transgenic zebrafish and mice. The combination reduced ubiquitin-specific protease 5 (USP5) levels and increased unanchored polyubiquitin chains. Overexpression of USP5 rescued neuroblastoma cells from the cytopathic effects of the combination and reduced unanchored polyubiquitin, suggesting USP5 is a therapeutic target of the combination. SAHA and SE486-11 directly bound to USP5 and the drug combination exhibited a 100-fold higher binding to USP5 than individual drugs alone in microscale thermophoresis assays. MYCN bound to the USP5 promoter and induced USP5 gene expression suggesting that USP5 and MYCN expression created a forward positive feedback loop in neuroblastoma cells. Thus, USP5 acts as an oncogenic cofactor with MYCN in neuroblastoma and the novel combination of HDAC inhibitor with SE486-11 represents a novel therapeutic approach for the treatment of MYCN-driven neuroblastoma.
- Published
- 2021
- Full Text
- View/download PDF
25. FBXO44 promotes DNA replication-coupled repetitive element silencing in cancer cells.
- Author
-
Shen JZ, Qiu Z, Wu Q, Finlay D, Garcia G, Sun D, Rantala J, Barshop W, Hope JL, Gimple RC, Sangfelt O, Bradley LM, Wohlschlegel J, Rich JN, and Spruck C
- Subjects
- Adult, Cell Line, Tumor, Cell Proliferation genetics, Cell Survival genetics, DNA Breaks, Double-Stranded, Drug Resistance, Neoplasm, Female, Gene Expression Regulation, Neoplastic, Histones metabolism, Humans, Immune Checkpoint Inhibitors pharmacology, Immune Checkpoint Inhibitors therapeutic use, Immunity, Interferons metabolism, Lysine metabolism, Male, Methylation, Middle Aged, Neoplasm Proteins metabolism, Neoplasms immunology, Nucleosomes metabolism, Signal Transduction, Transcription, Genetic, Treatment Outcome, DNA Replication genetics, F-Box Proteins metabolism, Neoplasms genetics, Repetitive Sequences, Nucleic Acid genetics
- Abstract
Repetitive elements (REs) compose ∼50% of the human genome and are normally transcriptionally silenced, although the mechanism has remained elusive. Through an RNAi screen, we identified FBXO44 as an essential repressor of REs in cancer cells. FBXO44 bound H3K9me3-modified nucleosomes at the replication fork and recruited SUV39H1, CRL4, and Mi-2/NuRD to transcriptionally silence REs post-DNA replication. FBXO44/SUV39H1 inhibition reactivated REs, leading to DNA replication stress and stimulation of MAVS/STING antiviral pathways and interferon (IFN) signaling in cancer cells to promote decreased tumorigenicity, increased immunogenicity, and enhanced immunotherapy response. FBXO44 expression inversely correlated with replication stress, antiviral pathways, IFN signaling, and cytotoxic T cell infiltration in human cancers, while a FBXO44-immune gene signature correlated with improved immunotherapy response in cancer patients. FBXO44/SUV39H1 were dispensable in normal cells. Collectively, FBXO44/SUV39H1 are crucial repressors of RE transcription, and their inhibition selectively induces DNA replication stress and viral mimicry in cancer cells., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
26. K63-linked ubiquitylation induces global sequestration of mitochondria.
- Author
-
Richard TJC, Herzog LK, Vornberger J, Rahmanto AS, Sangfelt O, Salomons FA, and Dantuma NP
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Autophagy, HeLa Cells, Humans, Mitochondrial Proteins genetics, Polyubiquitin genetics, Sequestosome-1 Protein genetics, Ubiquitination genetics, Mitochondria genetics, Mitophagy genetics, RNA-Binding Proteins genetics, Ubiquitin-Protein Ligases genetics
- Abstract
Even though K63-linked polyubiquitin chains do not target proteins for proteasomal degradation, they play nevertheless a complementary protective role in maintaining protein homeostasis by directing malfunctioning proteins and organelles to inclusion bodies or autophagosomes. A paradigm for this process is the sequestration and autophagic degradation of dysfunctional mitochondria. Although studies have shown that K63-ubiquitylation of mitochondrial proteins by the ubiquitin ligase Parkin is important in this process, it is presently not clear if this modification also suffices to initiate this cascade of events. To address this question, we have engineered the ubiquitin ligase ProxE3, which in an inducible manner synthesizes K63-linked ubiquitin chains on the surface of mitochondria. We found that the presence of K63-linked ubiquitin chains on mitochondria resulted in the recruitment of the ubiquitin adaptor p62 and induced a dramatic redistribution of mitochondria, which was reminiscent to the Parkin-facilitated sequestration in response to mitochondrial uncoupler. However, ProxE3 did not induce autophagic degradation of mitochondria. Our data show that K63-linked ubiquitin chains at the mitochondrial membrane are sufficient for the induction of mitochondrial sequestration, but not mitophagy, without the need of extrinsically inflicting mitochondrial dysfunction.
- Published
- 2020
- Full Text
- View/download PDF
27. Single cell characterization of B-lymphoid differentiation and leukemic cell states during chemotherapy in ETV6-RUNX1-positive pediatric leukemia identifies drug-targetable transcription factor activities.
- Author
-
Mehtonen J, Teppo S, Lahnalampi M, Kokko A, Kaukonen R, Oksa L, Bouvy-Liivrand M, Malyukova A, Mäkinen A, Laukkanen S, Mäkinen PI, Rounioja S, Ruusuvuori P, Sangfelt O, Lund R, Lönnberg T, Lohi O, and Heinäniemi M
- Subjects
- Bone Marrow, Cell Line, Tumor, Child, Core Binding Factor Alpha 2 Subunit metabolism, Drug Delivery Systems, Gene Expression Regulation, Neoplastic, Genetic Predisposition to Disease, Genome-Wide Association Study, Humans, Leukemia drug therapy, Proto-Oncogene Proteins c-ets metabolism, Repressor Proteins metabolism, Transcription Factors, Transcriptome, Translocation, Genetic, ETS Translocation Variant 6 Protein, Cell Differentiation genetics, Cell Proliferation, Core Binding Factor Alpha 2 Subunit genetics, Leukemia genetics, Lymphocytes physiology, Proto-Oncogene Proteins c-ets genetics, Repressor Proteins genetics
- Abstract
Background: Tight regulatory loops orchestrate commitment to B cell fate within bone marrow. Genetic lesions in this gene regulatory network underlie the emergence of the most common childhood cancer, acute lymphoblastic leukemia (ALL). The initial genetic hits, including the common translocation that fuses ETV6 and RUNX1 genes, lead to arrested cell differentiation. Here, we aimed to characterize transcription factor activities along the B-lineage differentiation trajectory as a reference to characterize the aberrant cell states present in leukemic bone marrow, and to identify those transcription factors that maintain cancer-specific cell states for more precise therapeutic intervention., Methods: We compared normal B-lineage differentiation and in vivo leukemic cell states using single cell RNA-sequencing (scRNA-seq) and several complementary genomics profiles. Based on statistical tools for scRNA-seq, we benchmarked a workflow to resolve transcription factor activities and gene expression distribution changes in healthy bone marrow lymphoid cell states. We compared these to ALL bone marrow at diagnosis and in vivo during chemotherapy, focusing on leukemias carrying the ETV6-RUNX1 fusion., Results: We show that lymphoid cell transcription factor activities uncovered from bone marrow scRNA-seq have high correspondence with independent ATAC- and ChIP-seq data. Using this comprehensive reference for regulatory factors coordinating B-lineage differentiation, our analysis of ETV6-RUNX1-positive ALL cases revealed elevated activity of multiple ETS-transcription factors in leukemic cells states, including the leukemia genome-wide association study hit ELK3. The accompanying gene expression changes associated with natural killer cell inactivation and depletion in the leukemic immune microenvironment. Moreover, our results suggest that the abundance of G1 cell cycle state at diagnosis and lack of differentiation-associated regulatory network changes during induction chemotherapy represent features of chemoresistance. To target the leukemic regulatory program and thereby overcome treatment resistance, we show that inhibition of ETS-transcription factors reduced cell viability and resolved pathways contributing to this using scRNA-seq., Conclusions: Our data provide a detailed picture of the transcription factor activities characterizing both normal B-lineage differentiation and those acquired in leukemic bone marrow and provide a rational basis for new treatment strategies targeting the immune microenvironment and the active regulatory network in leukemia.
- Published
- 2020
- Full Text
- View/download PDF
28. Ex vivo assessment of targeted therapies in a rare metastatic epithelial-myoepithelial carcinoma.
- Author
-
Mäkelä R, Arjonen A, Suryo Rahmanto A, Härmä V, Lehtiö J, Kuopio T, Helleday T, Sangfelt O, Kononen J, and Rantala JK
- Subjects
- Adult, Antineoplastic Agents therapeutic use, Biomarkers, Tumor genetics, Carcinoma drug therapy, Carcinoma genetics, Carcinoma pathology, DNA Mutational Analysis, Female, Gene Expression Profiling, High-Throughput Screening Assays, Humans, Lung Neoplasms genetics, Lung Neoplasms secondary, Myoepithelioma genetics, Myoepithelioma pathology, Prognosis, Salivary Gland Neoplasms genetics, Salivary Gland Neoplasms pathology, Tumor Cells, Cultured, Biomarkers, Tumor antagonists & inhibitors, Everolimus therapeutic use, Lung Neoplasms drug therapy, Molecular Targeted Therapy, Mutation, Myoepithelioma drug therapy, Salivary Gland Neoplasms drug therapy
- Abstract
Epithelial-myoepithelial carcinoma (EMC) is a rare subtype of salivary gland neoplasms. Since the initial description of the cancer, just over 300 cases have been reported. EMCs occupy a biphasic cellular differentiation-state defined by the constitution of two cell types representing epithelial and myoepithelial lineages, yet the functional consequence of the differentiation-state heterogeneity with respect to therapy resistance of the tumors remains unclear. The reported local recurrence rate of the cases is approximately 30%, and while distant metastases are rare, a significant fraction of these cases are reported to receive no survival benefit from radio- or chemotherapy given in addition to surgery. Moreover, no targeted therapies have been reported for these neoplasms. We report here the first use and application of ex vivo drug screening together with next generation sequencing to assess targeted treatment strategies for a rare metastatic epithelial-myoepithelial carcinoma. Results of the ex vivo drug screen demonstrate significant differential therapeutic sensitivity between the epithelial and myoepithelial intra-tumor cell lineages suggesting that differentiation-state heterogeneity within epithelial-myoepithelial carcinomas may present an outlet to partial therapeutic responses to targeted therapies including MEK and mTOR inhibitors. These results suggest that the intra-tumor lineage composition of EMC could be an important factor to be assessed when novel treatments are being evaluated for management of metastatic EMC., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
29. PTEN and DNA-PK determine sensitivity and recovery in response to WEE1 inhibition in human breast cancer.
- Author
-
Brunner A, Suryo Rahmanto A, Johansson H, Franco M, Viiliäinen J, Gazi M, Frings O, Fredlund E, Spruck C, Lehtiö J, Rantala JK, Larsson LG, and Sangfelt O
- Subjects
- Cell Cycle Proteins metabolism, Cell Line, Tumor, DNA-Activated Protein Kinase metabolism, Female, Gene Expression Profiling, Humans, PTEN Phosphohydrolase metabolism, Protein-Tyrosine Kinases metabolism, Proteome, Antineoplastic Agents pharmacology, Breast Neoplasms drug therapy, Cell Cycle Proteins genetics, DNA-Activated Protein Kinase genetics, PTEN Phosphohydrolase genetics, Protein-Tyrosine Kinases genetics, Pyrazoles pharmacology, Pyrimidinones pharmacology
- Abstract
Inhibition of WEE1 kinase by AZD1775 has shown promising results in clinical cancer trials, but markers predicting AZD1775 response are lacking. Here we analysed AZD1775 response in a panel of human breast cancer (BC) cell lines by global proteome/transcriptome profiling and identified two groups of basal-like BC (BLBCs): 'PTEN low' BLBCs were highly sensitive to AZD1775 and failed to recover following removal of AZD1775, while 'PTEN high' BLBCs recovered. AZD1775 induced phosphorylation of DNA-PK, protecting cells from replication-associated DNA damage and promoting cellular recovery. Deletion of DNA-PK or PTEN, or inhibition of DNA-PK sensitized recovering BLBCs to AZD1775 by abrogating replication arrest, allowing replication despite DNA damage. This was linked to reduced CHK1 activation, increased cyclin E levels and apoptosis. In conclusion, we identified PTEN and DNA-PK as essential regulators of replication checkpoint arrest in response to AZD1775 and defined PTEN as a promising biomarker for efficient WEE1 cancer therapy., Competing Interests: AB, AS, HJ, MF, JV, MG, OF, EF, CS, JL, JR, LL, OS No competing interests declared, (© 2020, Brunner et al.)
- Published
- 2020
- Full Text
- View/download PDF
30. Ablation of Key Oncogenic Pathways by RITA-Reactivated p53 Is Required for Efficient Apoptosis.
- Author
-
Grinkevich VV, Nikulenkov F, Shi Y, Enge M, Bao W, Maljukova A, Gluch A, Kel A, Sangfelt O, and Selivanova G
- Published
- 2017
- Full Text
- View/download PDF
31. Targeting SOX9 for degradation to inhibit chemoresistance, metastatic spread, and recurrence.
- Author
-
Suryo Rahmanto A, Swartling FJ, and Sangfelt O
- Abstract
Cancer cells with stem-like properties are believed to contribute to treatment resistance, dissemination, and recurrence. SOX9 controls stem cell plasticity and its deregulation may provide a basis for tumor progression. Here, we summarize our findings of targeted SOX9 destruction by SCF
FBW7 (Skp1/Cul1/F-box) in medulloblastoma and its potential for therapeutic intervention.- Published
- 2016
- Full Text
- View/download PDF
32. FBW7 suppression leads to SOX9 stabilization and increased malignancy in medulloblastoma.
- Author
-
Suryo Rahmanto A, Savov V, Brunner A, Bolin S, Weishaupt H, Malyukova A, Rosén G, Čančer M, Hutter S, Sundström A, Kawauchi D, Jones DT, Spruck C, Taylor MD, Cho YJ, Pfister SM, Kool M, Korshunov A, Swartling FJ, and Sangfelt O
- Subjects
- Aniline Compounds pharmacology, Animals, Benzamides, Cell Cycle Proteins genetics, Cell Line, Tumor, Cell Movement, Chromones pharmacology, Cisplatin pharmacology, F-Box Proteins genetics, F-Box-WD Repeat-Containing Protein 7, Glycogen Synthase Kinase 3 metabolism, HEK293 Cells, Humans, Medulloblastoma drug therapy, Medulloblastoma genetics, Mice, Nude, Morpholines pharmacology, Phosphatidylinositol 3-Kinases metabolism, Phosphoinositide-3 Kinase Inhibitors, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-akt antagonists & inhibitors, Proto-Oncogene Proteins c-akt metabolism, Pyrimidines pharmacology, Pyrroles pharmacology, SOX9 Transcription Factor genetics, TOR Serine-Threonine Kinases antagonists & inhibitors, TOR Serine-Threonine Kinases metabolism, Ubiquitin-Protein Ligases genetics, Ubiquitination, Cell Cycle Proteins metabolism, F-Box Proteins metabolism, Medulloblastoma metabolism, SOX9 Transcription Factor metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
SOX9 is a master transcription factor that regulates development and stem cell programs. However, its potential oncogenic activity and regulatory mechanisms that control SOX9 protein stability are poorly understood. Here, we show that SOX9 is a substrate of FBW7, a tumor suppressor, and a SCF (SKP1/CUL1/F-box)-type ubiquitin ligase. FBW7 recognizes a conserved degron surrounding threonine 236 (T236) in SOX9 that is phosphorylated by GSK3 kinase and consequently degraded by SCF
FBW 7α Failure to degrade SOX9 promotes migration, metastasis, and treatment resistance in medulloblastoma, one of the most common childhood brain tumors. FBW7 is either mutated or downregulated in medulloblastoma, and in cases where FBW7 mRNA levels are low, SOX9 protein is significantly elevated and this phenotype is associated with metastasis at diagnosis and poor patient outcome. Transcriptional profiling of medulloblastoma cells expressing a degradation-resistant SOX9 mutant reveals activation of pro-metastatic genes and genes linked to cisplatin resistance. Finally, we show that pharmacological inhibition of PI3K/AKT/mTOR pathway activity destabilizes SOX9 in a GSK3/FBW7-dependent manner, rendering medulloblastoma cells sensitive to cytostatic treatment., (© 2016 The Authors. Published under the terms of the CC BY NC ND 4.0 license.)- Published
- 2016
- Full Text
- View/download PDF
33. In β-actin knockouts, epigenetic reprogramming and rDNA transcription inactivation lead to growth and proliferation defects.
- Author
-
Almuzzaini B, Sarshad AA, Rahmanto AS, Hansson ML, Von Euler A, Sangfelt O, Visa N, Farrants AK, and Percipalle P
- Subjects
- Actins genetics, Animals, Cells, Cultured, Chromatin, DNA, Ribosomal genetics, Mice, Myosin Type I genetics, Myosin Type I metabolism, Pol1 Transcription Initiation Complex Proteins physiology, Transcription, Genetic physiology, Actins metabolism, Cellular Reprogramming physiology, DNA, Ribosomal metabolism, Epigenesis, Genetic physiology, Fibroblasts metabolism, Gene Expression Regulation, Developmental physiology
- Abstract
Actin and nuclear myosin 1 (NM1) are regulators of transcription and chromatin organization. Using a genome-wide approach, we report here that β-actin binds intergenic and genic regions across the mammalian genome, associated with both protein-coding and rRNA genes. Within the rDNA, the distribution of β-actin correlated with NM1 and the other subunits of the B-WICH complex, WSTF and SNF2h. In β-actin(-/-) mouse embryonic fibroblasts (MEFs), we found that rRNA synthesis levels decreased concomitantly with drops in RNA polymerase I (Pol I) and NM1 occupancies across the rRNA gene. Reintroduction of wild-type β-actin, in contrast to mutated forms with polymerization defects, efficiently rescued rRNA synthesis underscoring the direct role for a polymerization-competent form of β-actin in Pol I transcription. The rRNA synthesis defects in the β-actin(-/-) MEFs are a consequence of epigenetic reprogramming with up-regulation of the repressive mark H3K4me1 (monomethylation of lys4 on histone H3) and enhanced chromatin compaction at promoter-proximal enhancer (T0 sequence), which disturb binding of the transcription factor TTF1. We propose a novel genome-wide mechanism where the polymerase-associated β-actin synergizes with NM1 to coordinate permissive chromatin with Pol I transcription, cell growth, and proliferation.-Almuzzaini, B., Sarshad, A. A. , Rahmanto, A. S., Hansson, M. L., Von Euler, A., Sangfelt, O., Visa, N., Farrants, A.-K. Ö., Percipalle, P. In β-actin knockouts, epigenetic reprogramming and rDNA transcription inactivation lead to growth and proliferation defects., (© FASEB.)
- Published
- 2016
- Full Text
- View/download PDF
34. CDK-mediated activation of the SCF(FBXO) (28) ubiquitin ligase promotes MYC-driven transcription and tumourigenesis and predicts poor survival in breast cancer.
- Author
-
Cepeda D, Ng HF, Sharifi HR, Mahmoudi S, Cerrato VS, Fredlund E, Magnusson K, Nilsson H, Malyukova A, Rantala J, Klevebring D, Viñals F, Bhaskaran N, Zakaria SM, Rahmanto AS, Grotegut S, Nielsen ML, Szigyarto CA, Sun D, Lerner M, Navani S, Widschwendter M, Uhlén M, Jirström K, Pontén F, Wohlschlegel J, Grandér D, Spruck C, Larsson LG, and Sangfelt O
- Subjects
- Amino Acid Sequence, Breast metabolism, Breast Neoplasms diagnosis, Breast Neoplasms genetics, Breast Neoplasms pathology, Cell Line, Tumor, Cell Proliferation, Female, Gene Expression Regulation, Neoplastic, Humans, Molecular Sequence Data, Phosphorylation, Prognosis, Promoter Regions, Genetic, Proteolysis, SKP Cullin F-Box Protein Ligases analysis, SKP Cullin F-Box Protein Ligases genetics, Signal Transduction, Survival Analysis, Transcriptional Activation, Ubiquitination, Breast pathology, Breast Neoplasms metabolism, CDC2 Protein Kinase metabolism, Cyclin-Dependent Kinase 2 metabolism, Proto-Oncogene Proteins c-myc metabolism, SKP Cullin F-Box Protein Ligases metabolism
- Abstract
SCF (Skp1/Cul1/F-box) ubiquitin ligases act as master regulators of cellular homeostasis by targeting key proteins for ubiquitylation. Here, we identified a hitherto uncharacterized F-box protein, FBXO28 that controls MYC-dependent transcription by non-proteolytic ubiquitylation. SCF(FBXO28) activity and stability are regulated during the cell cycle by CDK1/2-mediated phosphorylation of FBXO28, which is required for its efficient ubiquitylation of MYC and downsteam enhancement of the MYC pathway. Depletion of FBXO28 or overexpression of an F-box mutant unable to support MYC ubiquitylation results in an impairment of MYC-driven transcription, transformation and tumourigenesis. Finally, in human breast cancer, high FBXO28 expression and phosphorylation are strong and independent predictors of poor outcome. In conclusion, our data suggest that SCF(FBXO28) plays an important role in transmitting CDK activity to MYC function during the cell cycle, emphasizing the CDK-FBXO28-MYC axis as a potential molecular drug target in MYC-driven cancers, including breast cancer., (© 2013 The Authors. Published by John Wiley and Sons, Ltd on behalf of EMBO.)
- Published
- 2013
- Full Text
- View/download PDF
35. TRIM13 (RFP2) downregulation decreases tumour cell growth in multiple myeloma through inhibition of NF Kappa B pathway and proteasome activity.
- Author
-
Gatt ME, Takada K, Mani M, Lerner M, Pick M, Hideshima T, Carrasco DE, Protopopov A, Ivanova E, Sangfelt O, Grandér D, Barlogie B, Shaughnessy JD Jr, Anderson KC, and Carrasco DR
- Subjects
- Apoptosis genetics, Cell Cycle genetics, Cell Division genetics, Cell Line, Tumor, Cell Survival genetics, Chromosome Deletion, Chromosomes, Human, Pair 13, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Down-Regulation, Gene Expression Profiling, Gene Knockdown Techniques, Humans, Multiple Myeloma metabolism, NF-kappa B genetics, NF-kappa B metabolism, Proteasome Endopeptidase Complex genetics, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism, DNA-Binding Proteins biosynthesis, Multiple Myeloma genetics, Multiple Myeloma pathology, NF-kappa B antagonists & inhibitors, Proteasome Endopeptidase Complex metabolism, Tumor Suppressor Proteins biosynthesis
- Abstract
Multiple myeloma (MM) is an incurable neoplasm caused by proliferation of malignant plasma cells in the bone marrow (BM). MM is characterized frequently by a complete or partial deletion of chromosome 13q14, seen in more than 50% of patients at diagnosis. Within this deleted region the tripartite motif containing 13 (TRIM13, also termed RFP2) gene product has been proposed to be a tumour suppressor gene (TSG). Here, we show that low expression levels of TRIM13 in MM are associated with chromosome 13q deletion and poor clinical outcome. We present a functional analysis of TRIM13 using a loss-of-function approach, and demonstrate that TRIM13 downregulation decreases tumour cell survival as well as cell cycle progression and proliferation of MM cells. In addition, we provide evidence for the involvement of TRIM13 downregulation in inhibiting the NF kappa B pathway and the activity of the 20S proteasome. Although this data does not support a role of TRIM13 as a TSG, it substantiates important roles of TRIM13 in MM tumour survival and proliferation, underscoring its potential role as a novel target for therapeutic intervention., (© 2013 John Wiley & Sons Ltd.)
- Published
- 2013
- Full Text
- View/download PDF
36. Fbw7α and Fbw7γ collaborate to shuttle cyclin E1 into the nucleolus for multiubiquitylation.
- Author
-
Bhaskaran N, van Drogen F, Ng HF, Kumar R, Ekholm-Reed S, Peter M, Sangfelt O, and Reed SI
- Subjects
- Animals, Cell Cycle Proteins genetics, Cell Line, Cyclin E genetics, F-Box Proteins genetics, F-Box-WD Repeat-Containing Protein 7, Fibroblasts metabolism, HEK293 Cells, Humans, Mice, NIMA-Interacting Peptidylprolyl Isomerase, Nucleoplasmins metabolism, Oncogene Proteins genetics, Peptidylprolyl Isomerase metabolism, Phosphorylation, Proteasome Endopeptidase Complex metabolism, Proteasome Inhibitors pharmacology, Protein Isoforms genetics, Protein Isoforms metabolism, Protein Transport, S Phase, Ubiquitin-Protein Ligases genetics, Ubiquitination, Cell Cycle Proteins metabolism, Cell Nucleolus metabolism, Cyclin E metabolism, F-Box Proteins metabolism, Oncogene Proteins metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
Cyclin E1, an activator of cyclin-dependent kinase 2 (Cdk2) that promotes replicative functions, is normally expressed periodically within the mammalian cell cycle, peaking at the G(1)-S-phase transition. This periodicity is achieved by E2F-dependent transcription in late G(1) and early S phases and by ubiquitin-mediated proteolysis. The ubiquitin ligase that targets phosphorylated cyclin E is SCF(Fbw7) (also known as SCF(Cdc4)), a member of the cullin ring ligase (CRL) family. Fbw7, a substrate adaptor subunit, is expressed as three splice-variant isoforms with different subcellular distributions: Fbw7α is nucleoplasmic but excluded from the nucleolus, Fbw7β is cytoplasmic, and Fbw7γ is nucleolar. Degradation of cyclin E in vivo requires SCF complexes containing Fbw7α and Fbw7γ, respectively. In vitro reconstitution showed that the role of SCF(Fbw7α) in cyclin E degradation, rather than ubiquitylation, is to serve as a cofactor of the prolyl cis-trans isomerase Pin1 in the isomerization of a noncanonical proline-proline bond in the cyclin E phosphodegron. This isomerization is required for subsequent binding and ubiquitylation by SCF(Fbw7γ). Here we show that Pin1-mediated isomerization of the cyclin E phosphodegron and subsequent binding to Fbw7γ drive nucleolar localization of cyclin E, where it is ubiquitylated by SCF(Fbw7γ) prior to its degradation by the proteasome. It is possible that this constitutes a mechanism for rapid inactivation of phosphorylated cyclin E by nucleolar sequestration prior to its multiubiquitylation and degradation.
- Published
- 2013
- Full Text
- View/download PDF
37. Proteomic screen reveals Fbw7 as a modulator of the NF-κB pathway.
- Author
-
Arabi A, Ullah K, Branca RM, Johansson J, Bandarra D, Haneklaus M, Fu J, Ariës I, Nilsson P, Den Boer ML, Pokrovskaja K, Grandér D, Xiao G, Rocha S, Lehtiö J, and Sangfelt O
- Subjects
- Cell Cycle Proteins genetics, Cell Line, Cell Line, Tumor, Computational Biology, F-Box Proteins genetics, F-Box-WD Repeat-Containing Protein 7, Glycogen Synthase Kinase 3 genetics, Glycogen Synthase Kinase 3 metabolism, Humans, Immunoblotting, Immunoprecipitation, NF-kappa B p52 Subunit genetics, Phosphorylation genetics, Phosphorylation physiology, Proteomics, Signal Transduction genetics, Signal Transduction physiology, Tandem Mass Spectrometry, Ubiquitin-Protein Ligases genetics, Cell Cycle Proteins metabolism, F-Box Proteins metabolism, NF-kappa B p52 Subunit metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
Fbw7 is a ubiquitin-ligase that targets several oncoproteins for proteolysis, but the full range of Fbw7 substrates is not known. Here we show that by performing quantitative proteomics combined with degron motif searches, we effectively screened for a more complete set of Fbw7 targets. We identify 89 putative Fbw7 substrates, including several disease-associated proteins. The transcription factor NF-κB2 (p100/p52) is one of the candidate Fbw7 substrates. We show that Fbw7 interacts with p100 via a conserved degron and that it promotes degradation of p100 in a GSK3β phosphorylation-dependent manner. Fbw7 inactivation increases p100 levels, which in the presence of NF-κB pathway stimuli, leads to increased p52 levels and activity. Accordingly, the apoptotic threshold can be increased by loss of Fbw7 in a p100-dependent manner. In conclusion, Fbw7-mediated destruction of p100 is a regulatory component restricting the response to NF-κB2 pathway stimulation.
- Published
- 2012
- Full Text
- View/download PDF
38. MiRNA-27a controls FBW7/hCDC4-dependent cyclin E degradation and cell cycle progression.
- Author
-
Lerner M, Lundgren J, Akhoondi S, Jahn A, Ng HF, Akbari Moqadam F, Oude Vrielink JA, Agami R, Den Boer ML, Grandér D, and Sangfelt O
- Subjects
- 3' Untranslated Regions, Cell Cycle Proteins genetics, Cell Line, Child, Cyclin E genetics, F-Box Proteins genetics, F-Box-WD Repeat-Containing Protein 7, Gene Expression Regulation, Genes, Reporter, Humans, Leukemia, B-Cell genetics, Leukemia, B-Cell metabolism, MicroRNAs genetics, Mutation, Ubiquitin-Protein Ligases genetics, Cell Cycle physiology, Cell Cycle Proteins metabolism, Cyclin E metabolism, F-Box Proteins metabolism, MicroRNAs metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
The F-box protein FBW7/hCDC4 is a tumor suppressor that acts as the substrate recognition component of an SCF ubiquitin ligase that targets numerous oncoproteins for proteasomal degradation. In this study, we investigated whether FBW7 is regulated by microRNAs, using a screen combining bioinformatic analysis, luciferase reporters and microRNA libraries. The ubiquitous miR-27a was identified as a major suppressor of FBW7 and in line with this, miR-27a prohibited ubiquitylation and turnover of the key FBW7 substrate cyclin E. Notably, we found that miR-27a only suppresses FBW7 during specific cell cycle phases, relieving its negative impact at the G1 to S-phase transition, prior to cyclin E protein degradation. We also demonstrate that attenuation of FBW7 by miR-27a overexpression leads to improper cell cycle progression and DNA replication stress, consistent with dysregulation of cyclin E expression. Finally, in the context of human cancer, miR-27a was discovered to be generally overexpressed in pediatric B-ALL and its expression to be inversely correlated with that of FBW7 in hyperdiploid cases of B-ALL. These data provide evidence for microRNA-mediated regulation of FBW7, and highlight the role of miR-27a as a novel factor fine-tuning the periodic events regulating cell cycle progression.
- Published
- 2011
- Full Text
- View/download PDF
39. Inactivation of FBXW7/hCDC4-β expression by promoter hypermethylation is associated with favorable prognosis in primary breast cancer.
- Author
-
Akhoondi S, Lindström L, Widschwendter M, Corcoran M, Bergh J, Spruck C, Grandér D, and Sangfelt O
- Subjects
- Adult, Aged, Aged, 80 and over, Biomarkers, Tumor, Breast Neoplasms diagnosis, Breast Neoplasms physiopathology, Cell Line, Tumor, CpG Islands, F-Box-WD Repeat-Containing Protein 7, Female, Gene Expression, Gene Expression Regulation, Neoplastic, Genes, p53, Humans, Lymphatic Metastasis, Middle Aged, Multivariate Analysis, Polymerase Chain Reaction, Prognosis, RNA, Messenger genetics, Survival, Breast Neoplasms genetics, Cell Cycle Proteins genetics, DNA Methylation, F-Box Proteins genetics, Promoter Regions, Genetic, Ubiquitin-Protein Ligases genetics
- Abstract
Introduction: Mutational inactivation of the FBXW7/hCDC4 tumor suppressor gene (TSG) is common in many cancer types, but infrequent in breast cancers. This study investigates the presence and impact of FBXW7/hCDC4 promoter methylation in breast cancer., Methods: FBXW7/hCDC4-β expression and promoter methylation was assessed in 161 tumors from two independent breast cancer cohorts. Associations between methylation status and clinicopathologic characteristics were assessed by Fisher's exact test. Survival was analyzed using the Kaplan-Meier method in addition to modeling the risk by use of a multivariate proportional hazard (Cox) model adjusting for possible confounders of survival., Results: Methylation of the promoter and loss of mRNA expression was found both in cell lines and primary tumors (43% and 51%, respectively). Using Cox modeling, a trend was found towards decreased hazard ratio (HR) for death in women with methylation of FBXW7/hCDC4-β in both cohorts (HR 0.53 (95% CI 0.23 to 1.23) and HR 0.50 (95% CI 0.23 to 1.08), respectively), despite an association between methylation and high-grade tumors (P = 0.017). Interestingly, in subgroups of patients whose tumors are p53 mutated or lymph-node positive, promoter methylation identified patients with significantly improved survival (P = 0.048 and P = 0.017, respectively)., Conclusions: We demonstrate an alternative mechanism for inactivation of the TSG FBXW7/hCDC4, namely promoter specific methylation. Importantly, in breast cancer, methylation of FBXW7/hCDC4-β is related to favorable prognosis despite its association with poorly differentiated tumors. Future work may define whether FBXW7/hCDC4 methylation is a biomarker of the response to chemotherapy and a target for epigenetic modulation therapy.
- Published
- 2010
- Full Text
- View/download PDF
40. Control of iron homeostasis by an iron-regulated ubiquitin ligase.
- Author
-
Vashisht AA, Zumbrennen KB, Huang X, Powers DN, Durazo A, Sun D, Bhaskaran N, Persson A, Uhlen M, Sangfelt O, Spruck C, Leibold EA, and Wohlschlegel JA
- Subjects
- Cell Line, Cullin Proteins metabolism, Hemerythrin metabolism, Homeostasis, Humans, Iron Regulatory Protein 1 metabolism, Oxygen metabolism, Protein Structure, Tertiary, Recombinant Proteins metabolism, SKP Cullin F-Box Protein Ligases metabolism, Ubiquitin-Protein Ligase Complexes, F-Box Proteins metabolism, Iron metabolism, Iron Regulatory Protein 2 metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
Eukaryotic cells require iron for survival and have developed regulatory mechanisms for maintaining appropriate intracellular iron concentrations. The degradation of iron regulatory protein 2 (IRP2) in iron-replete cells is a key event in this pathway, but the E3 ubiquitin ligase responsible for its proteolysis has remained elusive. We found that a SKP1-CUL1-FBXL5 ubiquitin ligase protein complex associates with and promotes the iron-dependent ubiquitination and degradation of IRP2. The F-box substrate adaptor protein FBXL5 was degraded upon iron and oxygen depletion in a process that required an iron-binding hemerythrin-like domain in its N terminus. Thus, iron homeostasis is regulated by a proteolytic pathway that couples IRP2 degradation to intracellular iron levels through the stability and activity of FBXL5.
- Published
- 2009
- Full Text
- View/download PDF
41. DLEU2, frequently deleted in malignancy, functions as a critical host gene of the cell cycle inhibitory microRNAs miR-15a and miR-16-1.
- Author
-
Lerner M, Harada M, Lovén J, Castro J, Davis Z, Oscier D, Henriksson M, Sangfelt O, Grandér D, and Corcoran MM
- Subjects
- Bone Marrow physiology, Cell Line, Chromatin genetics, Colony-Forming Units Assay, Cyclin D1 genetics, Cyclin E genetics, Cyclins genetics, DNA genetics, DNA Primers, Flow Cytometry, Gene Deletion, Humans, Kidney embryology, MicroRNAs physiology, Polymerase Chain Reaction, RNA genetics, RNA, Long Noncoding, RNA, Small Interfering genetics, Reverse Transcriptase Polymerase Chain Reaction, Transferases, Cell Cycle genetics, MicroRNAs genetics, Neoplasms genetics, Tumor Suppressor Proteins deficiency, Tumor Suppressor Proteins genetics
- Abstract
The microRNAs miR-15a and miR-16-1 are downregulated in multiple tumor types and are frequently deleted in chronic lymphocytic leukemia (CLL), myeloma and mantle cell lymphoma. Despite their abundance in most cells the transcriptional regulation of miR-15a/16-1 remains unclear. Here we demonstrate that the putative tumor suppressor DLEU2 acts as a host gene of these microRNAs. Mature miR-15a/miR-16-1 are produced in a Drosha-dependent process from DLEU2 and binding of the Myc oncoprotein to two alterative DLEU2 promoters represses both the host gene transcript and levels of mature miR-15a/miR-16-1. In line with a functional role for DLEU2 in the expression of the microRNAs, the miR-15a/miR-16-1 locus is retained in four CLL cases that delete both promoters of this gene and expression analysis indicates that this leads to functional loss of mature miR-15a/16-1. We additionally show that DLEU2 negatively regulates the G1 Cyclins E1 and D1 through miR-15a/miR-16-1 and provide evidence that these oncoproteins are subject to miR-15a/miR-16-1-mediated repression under normal conditions. We also demonstrate that DLEU2 overexpression blocks cellular proliferation and inhibits the colony-forming ability of tumor cell lines in a miR-15a/miR-16-1-dependent way. Together the data illuminate how inactivation of DLEU2 promotes cell proliferation and tumor progression through functional loss of miR-15a/miR-16-1.
- Published
- 2009
- Full Text
- View/download PDF
42. SCF(Fbxw7/hCdc4) targets cyclin E2 for ubiquitin-dependent proteolysis.
- Author
-
Klotz K, Cepeda D, Tan Y, Sun D, Sangfelt O, and Spruck C
- Subjects
- Binding Sites, Cell Cycle, Cell Line, Tumor, Cyclin E antagonists & inhibitors, Cyclin E genetics, Cyclin E metabolism, Cyclins chemistry, Cyclins genetics, F-Box-WD Repeat-Containing Protein 7, HeLa Cells, Humans, Mutagenesis, Site-Directed, Neoplasms genetics, Neoplasms metabolism, Neoplasms pathology, Oncogene Proteins antagonists & inhibitors, Oncogene Proteins genetics, Oncogene Proteins metabolism, Phosphorylation, RNA Interference, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Serine chemistry, Substrate Specificity, Threonine chemistry, Cell Cycle Proteins metabolism, Cyclins metabolism, F-Box Proteins metabolism, Ubiquitin metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
E-type cyclins (E1 and E2) regulate the S phase program in the mammalian cell division cycle. Expression of cyclin E1 and E2 is frequently deregulated in a variety of cancer types and a wealth of experimental evidence supports an oncogenic role of these proteins in human tumorigenesis. Although the molecular mechanisms responsible for cyclin E1 deregulation in cancer are well defined, little is known regarding cyclin E2. Here we report that cyclin E2 is targeted for ubiquitin-dependent proteolysis by the ubiquitin ligase SCF(Fbxw7/hCdc4). Ubiquitylation is triggered by phosphorylation of cyclin E2 on residues Thr392 and Ser396, and to a lesser extent Thr74, contained in two consensus Cdc4-phosphodegrons. Furthermore, we found that ectopic expression of cyclin E1 enhances the ubiquitin-dependent proteolysis of cyclin E2 in vivo, suggesting a potential cross-talk in the regulation of E-type cyclin activity. Since SCF(Fbxw7/hCdc4) is functionally inactivated in several human cancer types, alteration of this molecular pathway could contribute to the deregulation of cyclin E2 in tumorigenesis.
- Published
- 2009
- Full Text
- View/download PDF
43. Ablation of key oncogenic pathways by RITA-reactivated p53 is required for efficient apoptosis.
- Author
-
Grinkevich VV, Nikulenkov F, Shi Y, Enge M, Bao W, Maljukova A, Gluch A, Kel A, Sangfelt O, and Selivanova G
- Subjects
- Cell Line, Tumor, Down-Regulation, Furans pharmacology, Humans, Microtubule-Associated Proteins metabolism, Myeloid Cell Leukemia Sequence 1 Protein, Neoplasms pathology, Proto-Oncogene Proteins c-akt metabolism, Proto-Oncogene Proteins c-bcl-2 metabolism, Signal Transduction, Small Molecule Libraries, beta Catenin metabolism, Apoptosis, Neoplasms metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
Targeting "oncogene addiction" is a promising strategy for anticancer therapy. We report a potent inhibition of crucial oncogenes by p53 upon reactivation by small-molecule RITA in vitro and in vivo. RITA-activated p53 unleashes the transcriptional repression of antiapoptotic proteins Mcl-1, Bcl-2, MAP4, and survivin; blocks the Akt pathway on several levels; and downregulates c-Myc, cyclin E, and beta-catenin. p53 ablates c-Myc expression via several mechanisms at the transcriptional and posttranscriptional level. We show that the threshold for p53-mediated transrepression of survival genes is higher than for transactivation of proapoptotic targets. Inhibition of oncogenes by p53 reduces the cell's ability to buffer proapoptotic signals and elicits robust apoptosis. Our study highlights the role of transcriptional repression for p53-mediated tumor suppression.
- Published
- 2009
- Full Text
- View/download PDF
44. The Fbxw7/hCdc4 tumor suppressor in human cancer.
- Author
-
Tan Y, Sangfelt O, and Spruck C
- Subjects
- F-Box-WD Repeat-Containing Protein 7, Humans, Cell Cycle Proteins genetics, F-Box Proteins genetics, Genes, Tumor Suppressor, Neoplasms genetics, Ubiquitin-Protein Ligases genetics
- Abstract
Fbxw7/hCdc4 is a member of the F-box family of proteins, which function as interchangeable substrate recognition components of the SCF ubiquitin ligases. SCF(Fbxw7/hCdc4) targets several important oncoproteins including c-Myc, c-Jun, cyclin E1, and Notch, for ubiquitin-dependent proteolysis. Recent studies have shown that FBXW7/hCDC4 is mutated in a variety of human tumor types, suggesting that it is a general tumor suppressor in human cancer. Alteration of Fbxw7/hCdc4 function is linked to defects in differentiation, cellular proliferation, and genetic instability. In this review, we summarize what is known about Fbxw7/hCdc4-mediated degradation in the regulation of cellular proliferation and discuss how alteration of its function contributes to human tumorigenesis.
- Published
- 2008
- Full Text
- View/download PDF
45. Both SCF(Cdc4alpha) and SCF(Cdc4gamma) are required for cyclin E turnover in cell lines that do not overexpress cyclin E.
- Author
-
Sangfelt O, Cepeda D, Malyukova A, van Drogen F, and Reed SI
- Subjects
- Cell Line, Tumor, DNA Primers genetics, F-Box-WD Repeat-Containing Protein 7, Fluorescent Antibody Technique, Humans, Phosphorylation, Protein Isoforms metabolism, RNA Interference, Reverse Transcriptase Polymerase Chain Reaction, Cell Cycle Proteins metabolism, Cyclin E metabolism, F-Box Proteins metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
The ubiquitin-mediated turnover of cyclin E is regulated by phosphorylation and the activity of the ubiquitin ligase SCF(Cdc4) (also known as SCF(Fbw7)). In 293A cells, SCF complexes containing two different Cdc4 isoforms, alpha and gamma, are required for efficient cyclin E ubiquitylation. Whereas SCF(Cdc4gamma) ubiquitylates cyclin E directly, SCF(Cdc4alpha) serves as a cofactor for Pin1-mediated prolyl isomerization of the cyclin E phosphodegron, essential to potentiate ubiquitylation. In the current study, we show that the requirement for both Cdc4alpha and gamma is general, except in cell lines where cyclin E is expressed at an elevated level. Under these circumstances, Cdc4alpha is sufficient for cyclin E turnover. Furthermore, the requirement for Cdc4gamma can be bypassed by ectopic overexpression of cyclin E.
- Published
- 2008
- Full Text
- View/download PDF
46. FBXW7/hCDC4 is a general tumor suppressor in human cancer.
- Author
-
Akhoondi S, Sun D, von der Lehr N, Apostolidou S, Klotz K, Maljukova A, Cepeda D, Fiegl H, Dafou D, Marth C, Mueller-Holzner E, Corcoran M, Dagnell M, Nejad SZ, Nayer BN, Zali MR, Hansson J, Egyhazi S, Petersson F, Sangfelt P, Nordgren H, Grander D, Reed SI, Widschwendter M, Sangfelt O, and Spruck C
- Subjects
- 5-Methylcytosine metabolism, Amination, Cell Cycle Proteins metabolism, DNA Methylation, Dinucleotide Repeats, F-Box Proteins metabolism, F-Box-WD Repeat-Containing Protein 7, Gene Expression Regulation, Neoplastic, Gene Silencing, Humans, Models, Molecular, Mutation, Neoplasms metabolism, Protein Isoforms, Substrate Specificity, Ubiquitin-Protein Ligases metabolism, Cell Cycle Proteins genetics, F-Box Proteins genetics, Genes, Tumor Suppressor, Neoplasms genetics, Ubiquitin-Protein Ligases genetics
- Abstract
The ubiquitin-proteasome system is a major regulatory pathway of protein degradation and plays an important role in cellular division. Fbxw7 (or hCdc4), a member of the F-box family of proteins, which are substrate recognition components of the multisubunit ubiquitin ligase SCF (Skp1-Cdc53/Cullin-F-box-protein), has been shown to mediate the ubiquitin-dependent proteolysis of several oncoproteins including cyclin E1, c-Myc, c-Jun, and Notch. The oncogenic potential of Fbxw7 substrates, frequent allelic loss in human cancers, and demonstration that mutation of FBXW7 cooperates with p53 in mouse tumorigenesis have suggested that Fbxw7 could function as a tumor suppressor in human cancer. Here, we carry out an extensive genetic screen of primary tumors to evaluate the role of FBXW7 as a tumor suppressor in human tumorigenesis. Our results indicate that FBXW7 is inactivated by mutation in diverse human cancer types with an overall mutation frequency of approximately 6%. The highest mutation frequencies were found in tumors of the bile duct (cholangiocarcinomas, 35%), blood (T-cell acute lymphocytic leukemia, 31%), endometrium (9%), colon (9%), and stomach (6%). Approximately 43% of all mutations occur at two mutational "hotspots," which alter Arg residues (Arg465 and Arg479) that are critical for substrate recognition. Furthermore, we show that Fbxw7Arg465 hotspot mutant can abrogate wild-type Fbxw7 function through a dominant negative mechanism. Our study is the first comprehensive screen of FBXW7 mutations in various human malignancies and shows that FBXW7 is a general tumor suppressor in human cancer.
- Published
- 2007
- Full Text
- View/download PDF
47. The tumor suppressor gene hCDC4 is frequently mutated in human T-cell acute lymphoblastic leukemia with functional consequences for Notch signaling.
- Author
-
Malyukova A, Dohda T, von der Lehr N, Akhoondi S, Corcoran M, Heyman M, Spruck C, Grandér D, Lendahl U, and Sangfelt O
- Subjects
- Base Sequence, Blotting, Western, Cell Cycle Proteins metabolism, Child, DNA Mutational Analysis, F-Box Proteins metabolism, F-Box-WD Repeat-Containing Protein 7, Female, Humans, Male, Mutation, Polymerase Chain Reaction, Polymorphism, Single-Stranded Conformational, Receptor, Notch1 genetics, Ubiquitin-Protein Ligases metabolism, Cell Cycle Proteins genetics, F-Box Proteins genetics, Leukemia-Lymphoma, Adult T-Cell genetics, Receptor, Notch1 metabolism, Signal Transduction physiology, Ubiquitin-Protein Ligases genetics
- Abstract
Notch signaling is of crucial importance in normal T-cell development and Notch 1 is frequently mutated in T-cell acute lymphoblastic leukemias (T-ALL), leading to aberrantly high Notch signaling. In this report, we determine whether T-ALL mutations occur not only in Notch1 but also in the F-box protein hCdc4 (Sel-10, Ago, or Fbxw7), a negative regulator of Notch1. We show that the hCDC4 gene is mutated in leukemic cells from more than 30% of patients with pediatric T-ALL and derived cell lines. Most hCDC4 mutations found were missense substitutions at critical arginine residues (Arg(465), Arg(479), and Arg(505)) localized in the substrate-binding region of hCdc4. Cells inactivated for hCdc4 and T-ALL cells containing hCDC4 mutations exhibited an increased Notch1 protein half-life, consistent with the proposed role of hCdc4 in ubiquitin-dependent proteolysis of Notch1. Furthermore, restoration of wild-type but not mutant hCdc4 in HCT 116 hCDC4-negative cells led to an increased Notch1 ubiquitylation and decreased Notch1 signaling. These results show that hCdc4 mutations interfere with normal Notch1 regulation in vivo. Finally, we found that mutations in hCDC4 and NOTCH1 can occur in the same cancers and that patients carrying hCDC4 and/or NOTCH1 mutations have a favorable overall survival. Collectively, these data show that mutation of hCDC4 is a frequent event in T-ALL and suggest that hCDC4 mutations and gain-of-function mutations in NOTCH1 might synergize in contributing to the development of pediatric T-ALL leukemogenesis.
- Published
- 2007
- Full Text
- View/download PDF
48. The RBCC gene RFP2 (Leu5) encodes a novel transmembrane E3 ubiquitin ligase involved in ERAD.
- Author
-
Lerner M, Corcoran M, Cepeda D, Nielsen ML, Zubarev R, Pontén F, Uhlén M, Hober S, Grandér D, and Sangfelt O
- Subjects
- Adenosine Triphosphatases metabolism, Amino Acid Sequence, Cell Cycle Proteins metabolism, Cell Line, DNA-Binding Proteins antagonists & inhibitors, DNA-Binding Proteins chemistry, Humans, Membrane Proteins antagonists & inhibitors, Membrane Proteins chemistry, Membrane Proteins genetics, Membrane Proteins metabolism, Molecular Sequence Data, Proteasome Endopeptidase Complex metabolism, Protein Structure, Tertiary, RNA, Small Interfering genetics, Recombinant Proteins genetics, Recombinant Proteins metabolism, Transfection, Tumor Suppressor Proteins antagonists & inhibitors, Tumor Suppressor Proteins chemistry, Ubiquitin metabolism, Ubiquitin-Protein Ligases antagonists & inhibitors, Ubiquitin-Protein Ligases chemistry, Valosin Containing Protein, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Endoplasmic Reticulum metabolism, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism
- Abstract
RFP2, a gene frequently lost in various malignancies, encodes a protein with RING finger, B-box, and coiled-coil domains that belongs to the RBCC/TRIM family of proteins. Here we demonstrate that Rfp2 is an unstable protein with auto-polyubiquitination activity in vivo and in vitro, implying that Rfp2 acts as a RING E3 ubiquitin ligase. Consequently, Rfp2 ubiquitin ligase activity is dependent on an intact RING domain, as RING deficient mutants fail to drive polyubiquitination in vitro and are stabilized in vivo. Immunopurification and tandem mass spectrometry enabled the identification of several putative Rfp2 interacting proteins localized to the endoplasmic reticulum (ER), including valosin-containing protein (VCP), a protein indispensable for ER-associated degradation (ERAD). Importantly, we also show that Rfp2 regulates the degradation of the known ER proteolytic substrate CD3-delta, but not the N-end rule substrate Ub-R-YFP (yellow fluorescent protein), establishing Rfp2 as a novel E3 ligase involved in ERAD. Finally, we show that Rfp2 contains a C-terminal transmembrane domain indispensable for its localization to the ER and that Rfp2 colocalizes with several ER-resident proteins as analyzed by high-resolution immunostaining. In summary, these data are all consistent with a function for Rfp2 as an ERAD E3 ubiquitin ligase.
- Published
- 2007
- Full Text
- View/download PDF
49. Ubiquitylation of cyclin E requires the sequential function of SCF complexes containing distinct hCdc4 isoforms.
- Author
-
van Drogen F, Sangfelt O, Malyukova A, Matskova L, Yeh E, Means AR, and Reed SI
- Subjects
- Binding Sites, Cell Cycle Proteins genetics, Cell Line, F-Box Proteins genetics, F-Box-WD Repeat-Containing Protein 7, Gene Silencing physiology, Humans, Isoenzymes physiology, Models, Biological, Mutation, NIMA-Interacting Peptidylprolyl Isomerase, Peptidylprolyl Isomerase physiology, Ubiquitin-Protein Ligases genetics, Cell Cycle Proteins classification, Cell Cycle Proteins physiology, Cyclin E metabolism, F-Box Proteins classification, F-Box Proteins physiology, Isoenzymes classification, SKP Cullin F-Box Protein Ligases physiology, Ubiquitin metabolism, Ubiquitin-Protein Ligases classification, Ubiquitin-Protein Ligases physiology
- Abstract
Cyclin E, an activator of cyclin-dependent kinase 2 (Cdk2), is targeted for proteasomal degradation by phosphorylation-dependent multiubiquitylation via the ubiquitin ligase SCF(hCdc4). SCF ubiquitin ligases are composed of a core of conserved subunits and one variable subunit (an F box protein) involved in substrate recognition. We show here that multiubiquitylation of cyclin E requires the sequential function of two distinct splice variant isoforms of the F box protein hCdc4 known as alpha and gamma. SCF(hCdc4alpha) binds a complex containing cyclin E, Cdk2, and the prolyl cis/trans isomerase Pin1 and promotes the activity of Pin1 without directly ubiquitylating cyclin E. However, due to the action of this SCF(hCdc4alpha)-Pin1 complex, cyclin E becomes an efficient ubiquitylation substrate of SCF(hCdc4gamma). Furthermore, in the context of Cdc4alpha and cyclin E, mutational data suggest that Pin1 isomerizes a noncanonical proline-proline bond, with the possibility that Cdc4alpha may serve as a cofactor for altering the specificity of Pin1.
- Published
- 2006
- Full Text
- View/download PDF
50. The HPV-16 E7 oncogene sensitizes malignant cells to IFN-alpha-induced apoptosis.
- Author
-
Thyrell L, Sangfelt O, Zhivotovsky B, Pokrovskaja K, Wang Y, Einhorn S, and Grandér D
- Subjects
- Animals, Caspases metabolism, Cell Line, Transformed, Cell Line, Tumor, Cell Transformation, Viral, Enzyme Activation drug effects, Flow Cytometry, Membrane Proteins metabolism, Mice, Oncogene Proteins, Viral genetics, Papillomavirus E7 Proteins, Proto-Oncogene Proteins c-bcl-2 metabolism, Retroviridae genetics, Transfection, bcl-2 Homologous Antagonist-Killer Protein, Apoptosis drug effects, Interferon-alpha pharmacology, Oncogene Proteins, Viral metabolism, Papillomaviridae
- Abstract
Interferons (IFNs) exert antitumor effects in several human malignancies, but their mechanism of action is unclear. There is a great variability in sensitivity to IFN treatment depending on both tumor type and the individual patient. The reason for this variable sensitivity is not known. The fact that several IFN-induced anticellular effects are exerted through modulation of proto-oncogenes and tumor suppressor genes may indicate that the malignant genotype may be decisive in the cell's sensitivity to IFN. To determine if a deregulated oncogene could alter the cellular response to IFN, a mouse lymphoma cell line (J3D) was stably transfected with the viral human papillomavirus-16 (HPV-16) E7 oncogene. The E7-transfected cells and their respective mock-transfected sister clones were treated with IFN-alpha and examined for possible IFN-induced anticellular effects. We found that the E7-transfected clones were greatly sensitized to IFN-alpha-induced apoptosis compared with their mock-transfected counterparts. Induction of apoptosis in the transfected cells correlated with the ability of IFN to activate parts of the proapoptotic machinery specifically in these cells, including activation of caspases and the proapoptotic protein Bak. In summary, our data suggest that transfection of malignant cells with the E7 oncogene can sensitize them to IFN-alpha-induced apoptosis. This demonstrates that an oncogenic event may alter the cellular sensitivity to IFN and might also have implications for treatment of HPV-related diseases with IFN.
- Published
- 2005
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.