85 results on '"Wickström M"'
Search Results
2. Body surface area-based omega-3 fatty acids supplementation strongly correlates to blood concentrations in children.
- Author
-
Ljungblad, L., primary, Gleissman, H., additional, Hedberg, G., additional, Wickström, M., additional, Eissler, N., additional, Pickova, J., additional, Johnsen, J.I., additional, Tedroff, K., additional, Strandvik, B., additional, and Kogner, P., additional
- Published
- 2021
- Full Text
- View/download PDF
3. Bandscheibenmorphologie nach dorsalen Spondylodesen bei Frakturen im thorakolumbalen Übergang — Eine kernspintomographische Untersuchung
- Author
-
Hartwig, E., primary, Bauer, G., additional, Wickström, M., additional, Kramer, M., additional, and Kinzl, L., additional
- Published
- 1997
- Full Text
- View/download PDF
4. Vätternrundan. Fjärr-registrering av fysiologiska parametrar under idrottsutövning.
- Author
-
Eneling, Martin, Wickström, M, Johansson, Anders, Hult, Peter, Eneling, Martin, Wickström, M, Johansson, Anders, and Hult, Peter
- Published
- 2006
5. The novel tyrosine kinase inhibitor AKN-028 has significant antileukemic activity in cell lines and primary cultures of acute myeloid leukemia
- Author
-
Eriksson, A, primary, Hermanson, M, additional, Wickström, M, additional, Lindhagen, E, additional, Ekholm, C, additional, Jenmalm Jensen, A, additional, Löthgren, A, additional, Lehmann, F, additional, Larsson, R, additional, Parrow, V, additional, and Höglund, M, additional
- Published
- 2012
- Full Text
- View/download PDF
6. Wig-1, a novel regulator of N-Myc mRNA and N-Myc-driven tumor growth
- Author
-
Vilborg, A, primary, Bersani, C, additional, Wickström, M, additional, Segerström, L, additional, Kogner, P, additional, and Wiman, K G, additional
- Published
- 2012
- Full Text
- View/download PDF
7. 208 The alkylating prodrug J1 inhibits ovarian cancer cell growth, activates proapoptotic signalling and potentiates gemcitabine responsiveness in vitro and in vivo in mice
- Author
-
Viktorsson, K., primary, Gullbo, J., additional, Juntti, T., additional, Wickström, M., additional, Larsson, R., additional, Lewensohn, R., additional, and Spira, J., additional
- Published
- 2010
- Full Text
- View/download PDF
8. 459 Validation of a cell panel for preclinical evaluation of antitumor efficacy and toxicity of anticancer agents
- Author
-
Haglund, C., primary, Åleskog, A., additional, Nygren, P., additional, Gullbo, J., additional, Höglund, M., additional, Wickström, M., additional, Larsson, R., additional, and Lindhagen, E., additional
- Published
- 2010
- Full Text
- View/download PDF
9. In vitro evaluation of clinical activity and toxicity of anticancer drugs using tumor cells from patients and cells representing normal tissues.
- Author
-
Haglund C, Aleskog A, Nygren P, Gullbo J, Höglund M, Wickström M, Larsson R, and Lindhagen E
- Published
- 2012
10. W08.197 Interleukin-1β affects macrophage cholesterol levels and efflux
- Author
-
Persson, J., Nilsson, J., and Wickström, M.
- Published
- 2004
- Full Text
- View/download PDF
11. W01.95 Statin treatment stimulates human macrophage IL-1β secretion
- Author
-
Wickström, M. and Nilsson, J.
- Published
- 2004
- Full Text
- View/download PDF
12. The Cytotoxic Agents NSC-95397, Brefeldin A, Bortezomib and Sanguinarine Induce Apoptosis in Neuroendocrine Tumors In Vitro
- Author
-
Larsson, DE, Wickström, M., Hassan, S., Oberg, K., and Dan Granberg
13. The ROCK-1/2 inhibitor RKI-1447 blocks N-MYC, promotes cell death, and emerges as a synergistic partner for BET inhibitors in neuroblastoma.
- Author
-
Pepich A, Tümmler C, Abu Ajamieh S, Treis D, Boje AS, Vellema Q, Tsea I, Åkerlund E, Seashore-Ludlow B, Shirazi Fard S, Kogner P, Johnsen JI, and Wickström M
- Subjects
- Humans, Animals, Cell Line, Tumor, Xenograft Model Antitumor Assays, N-Myc Proto-Oncogene Protein genetics, N-Myc Proto-Oncogene Protein metabolism, Cell Proliferation drug effects, Protein Kinase Inhibitors pharmacology, Myosin Light Chains metabolism, Phosphorylation drug effects, Signal Transduction drug effects, Actin Depolymerizing Factors metabolism, Mice, Antineoplastic Combined Chemotherapy Protocols pharmacology, Cell Death drug effects, Apoptosis drug effects, Proto-Oncogene Proteins c-myc metabolism, Proto-Oncogene Proteins c-myc genetics, Bromodomain Containing Proteins, Proteins, Cardiac Myosins, rho-Associated Kinases antagonists & inhibitors, rho-Associated Kinases metabolism, Neuroblastoma drug therapy, Neuroblastoma pathology, Neuroblastoma metabolism, Neuroblastoma genetics, Drug Synergism, Zebrafish
- Abstract
High-risk neuroblastoma has a poor prognosis despite intensive treatment, highlighting the need for new therapeutic strategies. Genetic alterations in activators and inactivators of Rho GTPase have been identified in neuroblastoma suggested to activate Rho/Rho-kinase (ROCK) signaling. ROCK has also been implicated in therapy resistance. Therefore, we have explored the efficacy of the dual ROCK inhibitor RKI-1447 in neuroblastoma, emphasizing combination strategies. Treatment with RKI-1447 resulted in decreased growth, increased cell death, and inhibition of N-MYC in vitro and in vivo. A combination screen revealed enhanced effects between RKI-1447 and BET inhibitors. Synergistic effects from RKI-1447 and the BET inhibitor, ABBV-075, were confirmed in various neuroblastoma models, including zebrafish. Interestingly, ABBV-075 increased phosphorylation of both myosin light chain 2 and cofilin, downstream effectors of ROCK, increases that were blocked by adding RKI-1447. The combination treatment also augmented an inhibitory effect on C-MYC and, less pronounced, N-MYC protein expression. BET inhibitors have shown preclinical efficacy against neuroblastoma, but acquired resistance has limited their therapeutic benefit. We reveal that the combination of ROCK and BET inhibitors offers a promising treatment approach that can potentially mitigate resistance to BET inhibitors and reduce toxicity., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
14. SOX11 is a novel binding partner and endogenous inhibitor of SAMHD1 ara-CTPase activity in mantle cell lymphoma.
- Author
-
Abdelrazak Morsy MH, Lilienthal I, Lord M, Merrien M, Wasik AM, Sureda-Gómez M, Amador V, Johansson HJ, Lehtiö J, Garcia-Torre B, Martin-Subero JI, Tsesmetzis N, Tao S, Schinazi RF, Kim B, Sorteberg AL, Wickström M, Sheppard D, Rassidakis GZ, Taylor IA, Christensson B, Campo E, Herold N, and Sander B
- Subjects
- Humans, Animals, Mice, Protein Binding, Cell Line, Tumor, Cytarabine pharmacology, Lymphoma, Mantle-Cell metabolism, Lymphoma, Mantle-Cell pathology, Lymphoma, Mantle-Cell drug therapy, Lymphoma, Mantle-Cell genetics, SAM Domain and HD Domain-Containing Protein 1 metabolism, SAM Domain and HD Domain-Containing Protein 1 genetics, SOXC Transcription Factors metabolism, SOXC Transcription Factors genetics
- Abstract
Abstract: Sterile alpha motif and histidine-aspartate (HD) domain-containing protein 1 (SAMHD1) is a deoxynucleoside triphosphate triphosphohydrolase with ara-CTPase activity that confers cytarabine (ara-C) resistance in several hematological malignancies. Targeting SAMHD1's ara-CTPase activity has recently been demonstrated to enhance ara-C efficacy in acute myeloid leukemia. Here, we identify the transcription factor SRY-related HMG-box containing protein 11 (SOX11) as a novel direct binding partner and first known endogenous inhibitor of SAMHD1. SOX11 is aberrantly expressed not only in mantle cell lymphoma (MCL), but also in some Burkitt lymphomas. Coimmunoprecipitation of SOX11 followed by mass spectrometry in MCL cell lines identified SAMHD1 as the top SOX11 interaction partner, which was validated by proximity ligation assay. In vitro, SAMHD1 bound to the HMG box of SOX11 with low-micromolar affinity. In situ crosslinking studies further indicated that SOX11-SAMHD1 binding resulted in a reduced tetramerization of SAMHD1. Functionally, expression of SOX11 inhibited SAMHD1 ara-CTPase activity in a dose-dependent manner resulting in ara-C sensitization in cell lines and in a SOX11-inducible mouse model of MCL. In SOX11-negative MCL, SOX11-mediated ara-CTPase inhibition could be mimicked by adding the recently identified SAMHD1 inhibitor hydroxyurea. Taken together, our results identify SOX11 as a novel SAMHD1 interaction partner and its first known endogenous inhibitor with potentially important implications for clinical therapy stratification., (© 2024 American Society of Hematology. Published by Elsevier Inc. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
15. Single-cell analyses of metastatic bone marrow in human neuroblastoma reveals microenvironmental remodeling and metastatic signature.
- Author
-
Mei S, Alchahin AM, Embaie BT, Gavriliuc IM, Verhoeven BM, Zhao T, Li X, Jeffries NE, Pepich A, Sarkar H, Olsen TK, Wickström M, Stenman J, Reina-Bedoya O, Kharchenko PV, Saylor PJ, Johnsen JI, Sykes DB, Kogner P, and Baryawno N
- Subjects
- Humans, Child, Single-Cell Analysis, Tumor Microenvironment, Bone Marrow pathology, Neuroblastoma genetics
- Abstract
Neuroblastoma is an aggressive pediatric cancer with a high rate of metastasis to the BM. Despite intensive treatments including high-dose chemotherapy, the overall survival rate for children with metastatic neuroblastoma remains dismal. Understanding the cellular and molecular mechanisms of the metastatic tumor microenvironment is crucial for developing new therapies and improving clinical outcomes. Here, we used single-cell RNA-Seq to characterize immune and tumor cell alterations in neuroblastoma BM metastases by comparative analysis with patients without metastases. Our results reveal remodeling of the immune cell populations and reprogramming of gene expression profiles in the metastatic niche. In particular, within the BM metastatic niche, we observed the enrichment of immune cells, including tumor-associated neutrophils, macrophages, and exhausted T cells, as well as an increased number of Tregs and a decreased number of B cells. Furthermore, we highlighted cell communication between tumor cells and immune cell populations, and we identified prognostic markers in malignant cells that are associated with worse clinical outcomes in 3 independent neuroblastoma cohorts. Our results provide insight into the cellular, compositional, and transcriptional shifts underlying neuroblastoma BM metastases that contribute to the development of new therapeutic strategies.
- Published
- 2024
- Full Text
- View/download PDF
16. Humanin Treatment Protects Against Venetoclax-Induced Bone Growth Retardation in Ex Vivo Cultured Rat Bones.
- Author
-
Velentza L, Wickström M, Kogner P, Ohlsson C, Zaman F, and Sävendahl L
- Abstract
Context: Recent preclinical studies reported that the BCL-2 inhibitor venetoclax can impair bone growth. A strategy to prevent such a side effect of this promising anticancer drug is highly desired. Earlier in vitro and in vivo studies suggested that the mitochondrial peptide humanin has the potential to prevent drug-induced growth impairment., Objective: We hypothesized that co-treatment with the humanin analog HNG may prevent venetoclax-induced bone growth impairment., Methods: Ex vivo studies were performed in fetal rat metatarsal bones and human growth plate samples cultured for 12 and 2 days, respectively, while in vivo studies were performed in young neuroblastoma mice being treated daily for 14 days. The treatment groups included venetoclax, HNG, venetoclax plus HNG, or vehicle. Bone growth was continuously monitored and at the end point, histomorphometric and immunohistochemical analyses were performed in fixed tissues., Results: Venetoclax suppressed metatarsal bone growth and when combined with HNG, bone growth was rescued and all histological parameters affected by venetoclax monotherapy were normalized. Mechanistic studies showed that HNG downregulated the pro-apoptotic proteins Bax and p53 in cultured metatarsals and human growth plate tissues, respectively. The study in a neuroblastoma mouse model confirmed a growth-suppressive effect of venetoclax treatment. In this short-term in vivo study, no significant bone growth-rescuing effect could be verified when testing HNG at a single dose. We conclude that humanin dose-dependently protects ex vivo cultured metatarsal bones from venetoclax-induced bone growth impairment by restoring the growth plate microstructure., (© The Author(s) 2024. Published by Oxford University Press on behalf of the Endocrine Society.)
- Published
- 2024
- Full Text
- View/download PDF
17. The multiple functions of miR-574-5p in the neuroblastoma tumor microenvironment.
- Author
-
Proestler E, Donzelli J, Nevermann S, Breitwieser K, Koch LF, Best T, Fauth M, Wickström M, Harter PN, Kogner P, Lavieu G, Larsson K, and Saul MJ
- Abstract
Neuroblastoma is the most common extracranial solid tumor in childhood and arises from neural crest cells of the developing sympathetic nervous system. Prostaglandin E
2 (PGE2 ) has been identified as a key pro-inflammatory mediator of the tumor microenvironment (TME) that promotes neuroblastoma progression. We report that the interaction between the microRNA miR-574-5p and CUG-binding protein 1 (CUGBP1) induces the expression of microsomal prostaglandin E2 synthase 1 (mPGES-1) in neuroblastoma cells, which contributes to PGE2 biosynthesis. PGE2 in turn specifically induces the sorting of miR-574-5p into small extracellular vesicles (sEV) in neuroblastoma cell lines. sEV are one of the major players in intercellular communication in the TME. We found that sEV-derived miR-574-5p has a paracrine function in neuroblastoma. It acts as a direct Toll-like receptor 7/8 (TLR7/8) ligand and induces α-smooth muscle actin (α-SMA) expression in fibroblasts, contributing to fibroblast differentiation. This is particularly noteworthy as it has an opposite function to that in the TME of lung carcinoma, another PGE2 dependent tumor type. Here, sEV-derived miR-574-5p has an autokrine function that inhibits PGE2 biosynthesis in lung cancer cells. We report that the tetraspanin composition on the surface of sEV is associated with the function of sEV-derived miR-574-5p. This suggests that the vesicles do not only transport miRs, but also appear to influence their mode of action., Competing Interests: TB was employed by Merck KGaA. MF was employed by Prolytic GmbH, a Kymos Company. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Proestler, Donzelli, Nevermann, Breitwieser, Koch, Best, Fauth, Wickström, Harter, Kogner, Lavieu, Larsson and Saul.)- Published
- 2023
- Full Text
- View/download PDF
18. Pharmacological inhibition of BCL-2 with the FDA-approved drug venetoclax impairs longitudinal bone growth.
- Author
-
Velentza L, Wickström M, Kogner P, Ohlsson C, Zaman F, and Sävendahl L
- Subjects
- Animals, Female, Mice, Rats, Cartilage metabolism, Growth Plate metabolism, Proto-Oncogene Proteins c-bcl-2 metabolism, Bone Development, Chondrocytes metabolism
- Abstract
Treatment-related skeletal complications are common in childhood cancer patients and survivors. Venetoclax is a BCL-2 inhibitor that has shown efficacy in hematological malignancies in adults and is being investigated in pediatric cancer clinical trials as a promising therapeutic modality. Venetoclax triggers cell death in cancer cells, but whether it exerts similar effects in normal bone cells, is unknown. Chondrogenic ATDC5 cells, E20 fetal rat metatarsal bones, and human growth plate biopsies were treated with different concentrations of venetoclax. Female NMRI nu/nu mice were treated with venetoclax or vehicle for 15 days. Mice were X-rayed at baseline and at the end of the experiment to assess longitudinal bone growth and body weight was monitored throughout the study. Histomorphometric and immunohistochemical analyses were performed to evaluate treatment effects on the growth plate cartilage. Venetoclax decreased the viability of chondrocytes and impaired the growth of ex vivo cultured metatarsals while reducing the height of the resting/proliferative zone and the hypertrophic cell size. When tested in vivo, venetoclax suppressed bone growth and reduced growth plate height. Our experimental data suggest that venetoclax directly targets growth plate chondrocytes suppressing bone growth and we, therefore, encourage careful monitoring of longitudinal bone growth if treating growing children with venetoclax., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
19. The cyclin dependent kinase inhibitor p21 Cip1/Waf1 is a therapeutic target in high-risk neuroblastoma.
- Author
-
Sorteberg AL, Halipi V, Wickström M, and Shirazi Fard S
- Abstract
Platinum-based chemotherapies such as cisplatin are used as first-line treatment for the paediatric tumour neuroblastoma. Although the majority of neuroblastoma tumours respond to therapy, there is a high fraction of high-risk neuroblastoma patients that eventually relapse with increased resistance. Here, we show that one key determinant of cisplatin sensitivity is phosphorylation of the cyclin-dependent kinase inhibitor p21
Cip1/Waf1 . A panel of eight neuroblastoma cell lines and a TH-MYCN mouse model were investigated for the expression of p21Cip1/Waf1 using RT-qPCR, Western blot, and immunofluorescence. This was followed by investigation of sensitivity towards cisplatin and the p21Cip1/Waf1 inhibitor UC2288. Whereas the cell lines and the mouse model showed low levels of un-phosphorylated p21Cip1/Waf1 , the phosphorylated p21Cip1/Waf1 (Thr145) was highly expressed, which in the cell lines correlated to cisplatin resistance. Furthermore, the neuroblastoma cell lines showed high sensitivity to UC2288, and combination treatment with cisplatin resulted in considerably decreased cell viability and delay in regrowth in the two most resistant cell lines, SK-N-DZ and BE(2)-C. Thus, targeting p21Cip1/Waf1 can offer new treatment strategies and subsequently lead to the design of more efficient combination treatments for high-risk neuroblastoma., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Sorteberg, Halipi, Wickström and Shirazi Fard.)- Published
- 2022
- Full Text
- View/download PDF
20. Omega-3 fatty acids decrease CRYAB, production of oncogenic prostaglandin E 2 and suppress tumor growth in medulloblastoma.
- Author
-
Ljungblad L, Bergqvist F, Tümmler C, Madawala S, Olsen TK, Andonova T, Jakobsson PJ, Johnsen JI, Pickova J, Strandvik B, Kogner P, Gleissman H, and Wickström M
- Subjects
- Animals, Apoptosis drug effects, Carcinogenesis, Cell Line, Tumor, Cell Survival drug effects, Chromatography, Liquid methods, Dinoprostone metabolism, Docosahexaenoic Acids pharmacology, Eicosapentaenoic Acid pharmacology, Fatty Acids metabolism, Fatty Acids, Omega-3 metabolism, Female, Humans, Mice, Mice, Nude, Prostaglandins metabolism, Tandem Mass Spectrometry methods, Tumor Microenvironment, Xenograft Model Antitumor Assays methods, alpha-Crystallin B Chain drug effects, alpha-Crystallin B Chain metabolism, Fatty Acids, Omega-3 pharmacology, Medulloblastoma drug therapy, Medulloblastoma metabolism
- Abstract
Aims: Medulloblastoma (MB) is one of the most common malignant central nervous system tumors of childhood. Despite intensive treatments that often leads to severe neurological sequelae, the risk for resistant relapses remains significant. In this study we have evaluated the effects of the ω3-long chain polyunsaturated fatty acids (ω3-LCPUFA) docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) on MB cell lines and in a MB xenograft model., Main Methods: Effects of ω3-LCPUFA treatment of MB cells were assessed using the following: WST-1 assay, cell death probes, clonogenic assay, ELISA and western blot. MB cells were implanted into nude mice and the mice were randomized to DHA, or a combination of DHA and EPA treatment, or to control group. Treatment effects in tumor tissues were evaluated with: LC-MS/MS, RNA-sequencing and immunohistochemistry, and tumors, erythrocytes and brain tissues were analyzed with gas chromatography., Key Findings: ω3-LCPUFA decreased prostaglandin E2 (PGE
2 ) secretion from MB cells, and impaired MB cell viability and colony forming ability and increased apoptosis in a dose-dependent manner. DHA reduced tumor growth in vivo, and both PGE2 and prostacyclin were significantly decreased in tumor tissue from treated mice compared to control animals. All ω3-LCPUFA and dihomo-γ-linolenic acid increased in tumors from treated mice. RNA-sequencing revealed 10 downregulated genes in common among ω3-LCPUFA treated tumors. CRYAB was the most significantly altered gene and the downregulation was confirmed by immunohistochemistry., Significance: Our findings suggest that addition of DHA and EPA to the standard MB treatment regimen might be a novel approach to target inflammation in the tumor microenvironment., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
21. MYCMI-7: A Small MYC-Binding Compound that Inhibits MYC: MAX Interaction and Tumor Growth in a MYC-Dependent Manner.
- Author
-
Castell A, Yan Q, Fawkner K, Bazzar W, Zhang F, Wickström M, Alzrigat M, Franco M, Krona C, Cameron DP, Dyberg C, Olsen TK, Verschut V, Schmidt L, Lim SY, Mahmoud L, Hydbring P, Lehmann S, Baranello L, Nelander S, Johnsen JI, and Larsson LG
- Subjects
- Animals, Mice, Humans, N-Myc Proto-Oncogene Protein genetics, Cell Line, Tumor, Cell Proliferation, Cell Cycle, Neuroblastoma drug therapy
- Abstract
Deregulated expression of MYC family oncogenes occurs frequently in human cancer and is often associated with aggressive disease and poor prognosis. While MYC is a highly warranted target, it has been considered "undruggable," and no specific anti-MYC drugs are available in the clinic. We recently identified molecules named MYCMIs that inhibit the interaction between MYC and its essential partner MAX. Here we show that one of these molecules, MYCMI-7, efficiently and selectively inhibits MYC:MAX and MYCN:MAX interactions in cells, binds directly to recombinant MYC, and reduces MYC-driven transcription. In addition, MYCMI-7 induces degradation of MYC and MYCN proteins. MYCMI-7 potently induces growth arrest/apoptosis in tumor cells in a MYC/MYCN-dependent manner and downregulates the MYC pathway on a global level as determined by RNA sequencing. Sensitivity to MYCMI-7 correlates with MYC expression in a panel of 60 tumor cell lines and MYCMI-7 shows high efficacy toward a collection of patient-derived primary glioblastoma and acute myeloid leukemia (AML) ex vivo cultures. Importantly, a variety of normal cells become G
1 arrested without signs of apoptosis upon MYCMI-7 treatment. Finally, in mouse tumor models of MYC-driven AML, breast cancer, and MYCN-amplified neuroblastoma, treatment with MYCMI-7 downregulates MYC/MYCN, inhibits tumor growth, and prolongs survival through apoptosis with few side effects. In conclusion, MYCMI-7 is a potent and selective MYC inhibitor that is highly relevant for the development into clinically useful drugs for the treatment of MYC-driven cancer., Significance: Our findings demonstrate that the small-molecule MYCMI-7 binds MYC and inhibits interaction between MYC and MAX, thereby hampering MYC-driven tumor cell growth in culture and in vivo while sparing normal cells., Competing Interests: A. Castell reports a patent to 2118752.1 pending. K. Fawkner reports a patent to 2118752.1 pending. L.-G. Larsson reports a patent to 2118752.1 pending. No disclosures were reported by the other authors., (© 2022 The Authors; Published by the American Association for Cancer Research.)- Published
- 2022
- Full Text
- View/download PDF
22. GIT1 protects against breast cancer growth through negative regulation of Notch.
- Author
-
Zhang S, Miyakawa A, Wickström M, Dyberg C, Louhivuori L, Varas-Godoy M, Kemppainen K, Kanatani S, Kaczynska D, Ellström ID, Elfman L, Kronqvist P, Repo H, Mikoshiba K, Sahlgren C, Johnsen JI, and Uhlén P
- Subjects
- Adaptor Proteins, Signal Transducing metabolism, Breast pathology, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Female, Humans, Neoplasm Recurrence, Local, Receptors, Notch genetics, Receptors, Notch metabolism, Signal Transduction, Breast Neoplasms pathology
- Abstract
Hyperactive Notch signalling is frequently observed in breast cancer and correlates with poor prognosis. However, relatively few mutations in the core Notch signalling pathway have been identified in breast cancer, suggesting that as yet unknown mechanisms increase Notch activity. Here we show that increased expression levels of GIT1 correlate with high relapse-free survival in oestrogen receptor-negative (ER(-)) breast cancer patients and that GIT1 mediates negative regulation of Notch. GIT1 knockdown in ER(-) breast tumour cells increased signalling downstream of Notch and activity of aldehyde dehydrogenase, a predictor of poor clinical outcome. GIT1 interacts with the Notch intracellular domain (ICD) and influences signalling by inhibiting the cytoplasm-to-nucleus transport of the Notch ICD. In xenograft experiments, overexpression of GIT1 in ER(-) cells prevented or reduced Notch-driven tumour formation. These results identify GIT1 as a modulator of Notch signalling and a guardian against breast cancer growth., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
23. PPM1D Is a Therapeutic Target in Childhood Neural Tumors.
- Author
-
Milosevic J, Treis D, Fransson S, Gallo-Oller G, Sveinbjörnsson B, Eissler N, Tanino K, Sakaguchi K, Martinsson T, Wickström M, Kogner P, and Johnsen JI
- Abstract
Childhood medulloblastoma and high-risk neuroblastoma frequently present with segmental gain of chromosome 17q corresponding to aggressive tumors and poor patient prognosis. Located within the 17q-gained chromosomal segments is PPM1D at chromosome 17q23.2. PPM1D encodes a serine/threonine phosphatase, WIP1, that is a negative regulator of p53 activity as well as key proteins involved in cell cycle control, DNA repair and apoptosis. Here, we show that the level of PPM1D expression correlates with chromosome 17q gain in medulloblastoma and neuroblastoma cells, and both medulloblastoma and neuroblastoma cells are highly dependent on PPM1D expression for survival. Comparison of different inhibitors of WIP1 showed that SL-176 was the most potent compound inhibiting medulloblastoma and neuroblastoma growth and had similar or more potent effects on cell survival than the MDM2 inhibitor Nutlin-3 or the p53 activator RITA. SL-176 monotherapy significantly suppressed the growth of established medulloblastoma and neuroblastoma xenografts in nude mice. These results suggest that the development of clinically applicable compounds inhibiting the activity of WIP1 is of importance since PPM1D activating mutations, genetic gain or amplifications and/or overexpression of WIP1 are frequently detected in several different cancers.
- Published
- 2021
- Full Text
- View/download PDF
24. Inhibition of the ubiquitin-proteasome system by an NQO1-activatable compound.
- Author
-
Giovannucci TA, Salomons FA, Haraldsson M, Elfman LHM, Wickström M, Young P, Lundbäck T, Eirich J, Altun M, Jafari R, Gustavsson AL, Johnsen JI, and Dantuma NP
- Subjects
- Animals, Caspases metabolism, Cell Death drug effects, Cell Line, Tumor, Deubiquitinating Enzymes metabolism, Female, High-Throughput Screening Assays, Humans, Mice, Nude, Phenotype, Proteasome Inhibitors chemistry, Proteasome Inhibitors pharmacology, Recombinant Proteins metabolism, Small Molecule Libraries pharmacology, Substrate Specificity drug effects, Ubiquitin metabolism, Xenograft Model Antitumor Assays, Mice, NAD(P)H Dehydrogenase (Quinone) metabolism, Proteasome Endopeptidase Complex metabolism, Ubiquitin antagonists & inhibitors
- Abstract
Malignant cells display an increased sensitivity towards drugs that reduce the function of the ubiquitin-proteasome system (UPS), which is the primary proteolytic system for destruction of aberrant proteins. Here, we report on the discovery of the bioactivatable compound CBK77, which causes an irreversible collapse of the UPS, accompanied by a general accumulation of ubiquitylated proteins and caspase-dependent cell death. CBK77 caused accumulation of ubiquitin-dependent, but not ubiquitin-independent, reporter substrates of the UPS, suggesting a selective effect on ubiquitin-dependent proteolysis. In a genome-wide CRISPR interference screen, we identified the redox enzyme NAD(P)H:quinone oxidoreductase 1 (NQO1) as a critical mediator of CBK77 activity, and further demonstrated its role as the compound bioactivator. Through affinity-based proteomics, we found that CBK77 covalently interacts with ubiquitin. In vitro experiments showed that CBK77-treated ubiquitin conjugates were less susceptible to disassembly by deubiquitylating enzymes. In vivo efficacy of CBK77 was validated by reduced growth of NQO1-proficient human adenocarcinoma cells in nude mice treated with CBK77. This first-in-class NQO1-activatable UPS inhibitor suggests that it may be possible to exploit the intracellular environment in malignant cells for leveraging the impact of compounds that impair the UPS., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
25. Effects of PI3K and FGFR inhibitors alone and in combination, and with/without cytostatics in childhood neuroblastoma cell lines.
- Author
-
Holzhauser S, Lukoseviciute M, Papachristofi C, Vasilopoulou C, Herold N, Wickström M, Kostopoulou ON, and Dalianis T
- Subjects
- Antineoplastic Combined Chemotherapy Protocols therapeutic use, Apoptosis drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Child, Cisplatin pharmacology, Cisplatin therapeutic use, Cytostatic Agents therapeutic use, Doxorubicin pharmacology, Doxorubicin therapeutic use, Drug Resistance, Neoplasm, Drug Synergism, Humans, Neuroblastoma pathology, Phosphoinositide-3 Kinase Inhibitors therapeutic use, Pyrazoles pharmacology, Pyrazoles therapeutic use, Quinoxalines pharmacology, Quinoxalines therapeutic use, Receptors, Fibroblast Growth Factor metabolism, Thiazoles pharmacology, Thiazoles therapeutic use, Vincristine pharmacology, Vincristine therapeutic use, Antineoplastic Combined Chemotherapy Protocols pharmacology, Cytostatic Agents pharmacology, Neuroblastoma drug therapy, Phosphoinositide-3 Kinase Inhibitors pharmacology, Receptors, Fibroblast Growth Factor antagonists & inhibitors
- Abstract
Neuroblastoma (NB) is a heterogenous disease with treatment varying from observation for low‑risk tumors, to extensive therapy with chemotherapy, surgery, radiotherapy, and autologous bone‑marrow‑transplantation and immunotherapy. However, a high frequency of primary‑chemo‑refractory disease and recurrences urgently require novel treatment strategies. The present study therefore investigated the anti‑NB efficacy of the recently FDA‑approved phosphoinositide 3‑kinase (PI3K) and fibroblast growth factor receptor (FGFR) inhibitors, alpelisib (BYL719) and erdafitinib (JNJ‑42756493), alone and in combination with or without cisplatin, vincristine, or doxorubicin on 5 NB cell lines. For this purpose, the NB cell lines, SK‑N‑AS, SK‑N‑BE(2)‑C, SK‑N‑DZ, SK‑N‑FI and SK‑N‑SH (where SK‑N‑DZ had a deletion of PIK3C2G and none had FGFR mutations according to the Cancer Program's Dependency Map, although some were chemoresistant), were tested for their sensitivity to FDA‑approved inhibitors alone or in combination, or together with cytostatic drugs by viability, cytotoxicity, apoptosis and proliferation assays. The results revealed that monotherapy with alpelisib or erdafitinib resulted in a dose‑dependent inhibition of cell viability and proliferation. Notably, the combined use of PI3K and FGFR inhibitors resulted in an enhanced efficacy, while their combined use with the canonical cytotoxic agents, cisplatin, vincristine and doxorubicin, resulted in variable synergistic, additive and antagonistic effects. Collectively, the present study provides pre‑clinical evidence that PI3K and FGFR inhibitors exhibit promising anti‑NB activity. The data presented herein also indicate that the incorporation of these inhibitors into chemotherapeutic regimens requires careful consideration and further research in order to obtain a beneficial efficacy. Nevertheless, the addition of PI3K and FGFR inhibitors to the treatment arsenal might reduce the occurrence of refractory and relapsing disease in NB without FGFR and PI3K mutations.
- Published
- 2021
- Full Text
- View/download PDF
26. How can sexual and reproductive health and rights be enhanced for young people with intellectual disability? - focus group interviews with staff in Sweden.
- Author
-
Wickström M, Larsson M, and Höglund B
- Subjects
- Adolescent, Adult, Aged, Allied Health Personnel, Attitude of Health Personnel, Contraception, Female, Human Rights, Humans, Male, Middle Aged, Reproductive Rights, Sweden, Young Adult, Focus Groups, Intellectual Disability psychology, Reproductive Health, Right to Health, Sexual Health
- Abstract
Background: Different types of staff support individuals with intellectual disability (ID) in their daily life, in schools, leisure activities and in special accommodations. This study aimed to gain a deeper understanding of experiences and perceptions regarding sexual and reproductive health and rights (SRHR) among staff., Methods: Data were collected in mid-Sweden in four focus groups with altogether 20 participants, 18 women and 2 men aged between 18 and 65 years. They had different professions and worked among youth and adults with ID aged 18-40 years in schools, accommodations and with leisure activities. Their working experience varied from 3 years to more than 20 years. Interviews were audio recorded, transcribed and analysed with content analysis., Results: The participants generally described positive attitudes towards sexuality for people with ID, both among themselves and in society. However, many situations such as ensuring privacy, balancing between waiting and acting, issues around contraception and reproduction were difficult to address and participants had hesitations about childbearing. They described different strategies such as showing respect, enhancing self-esteem and decision making ability and using interprofessional support to cope with frustrating situations. They lacked a clear mandate from managers as well as written guidelines and policies. They requested education and support from peers, supervisors and other professionals., Conclusion: Participants in the study were generally open-minded and accepting towards sexuality among young people with ID. They thought it was difficult to deal with reproduction/parenthood and felt unprepared and frustrated in certain situations. The participants requested a clear mandate from managers, organizational guidelines, more education and inter-professional support. We believe these findings can inform the development of policy and support the implementation of SRHR related guidelines to support staff working with young people with ID.
- Published
- 2020
- Full Text
- View/download PDF
27. Targeting Fibroblast Growth Factor Receptor (FGFR) and Phosphoinositide 3-kinase (PI3K) Signaling Pathways in Medulloblastoma Cell Lines.
- Author
-
Holzhauser S, Lukoseviciute M, Andonova T, Ursu RG, Dalianis T, Wickström M, and Kostopoulou ON
- Subjects
- Animals, Antineoplastic Agents pharmacology, Apoptosis drug effects, Caspase 3 metabolism, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Disease Models, Animal, Dose-Response Relationship, Drug, Humans, Medulloblastoma pathology, Protein Kinase Inhibitors pharmacology, Xenograft Model Antitumor Assays, Medulloblastoma metabolism, Phosphatidylinositol 3-Kinase metabolism, Receptors, Fibroblast Growth Factor metabolism, Signal Transduction drug effects
- Abstract
Background/aim: Medulloblastoma (MB) accounts for ~20% of pediatric malignant central nervous system tumors. Treatment strategies, including surgery, radiation therapy and/or chemotherapy, are effective, but recurrence and metastasis frequently occur. Therefore, novel therapies are required. Herein, the effects of fibroblast growth factor receptor (FGFR) and phosphoinositide 3-kinase (PI3K) inhibitors on MB cells lines were evaluated., Materials and Methods: MB cell lines (UW228-3, DAOY, Med8a, D425, D283) were tested for sensitivity to FGFR (AZD4547) and PI3K (BEZ235 and BYL719) inhibitors by viability, cytotoxicity, apoptosis, and proliferation assays., Results: Single treatments with FGFR and PI3K inhibitors decreased viability and proliferation in a dose-dependent pattern in most cell lines. Combinination of the two type of drugs, increased sensitivity, especially of the most resistant cell line UW228-3., Conclusion: Combination treatments with FGFR and PI3K inhibitors were superior to single treatments with FGFR and PI3K inhibitors, especially with BEZ235, for MB cell lines., (Copyright© 2020, International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
28. Inhibition of Rho-Associated Kinase Suppresses Medulloblastoma Growth.
- Author
-
Dyberg C, Andonova T, Olsen TK, Brodin B, Kool M, Kogner P, Johnsen JI, and Wickström M
- Abstract
Medulloblastoma is one of the most common malignant brain tumor types in children, with an overall survival of 70%. Mortality is associated with metastatic relapsed tumors. Rho-associated kinases (ROCKs), important for epithelial-mesenchymal transition (EMT) and proper nervous system development, have previously been identified as a promising drug target to inhibit cancer growth and metastatic spread. Here, we show that ROCKs are expressed in medulloblastoma, with higher ROCK2 mRNA expression in metastatic compared to non-metastatic tumors. By evaluating three ROCK inhibitors in a panel of medulloblastoma cell lines we demonstrated that medulloblastoma cells were sensitive for pharmacological ROCK inhibition. The specific ROCK inhibitor RKI-1447 inhibited the tumorigenicity in medulloblastoma cells as well as impeded cell migration and invasion. Differential gene expression analysis suggested that ROCK inhibition was associated with the downregulation of signaling pathways important in proliferation and metastasis e.g., TNFα via NFκβ, TGFβ, and EMT. Expression of key proteins in these pathways such as RHOA, RHOB, JUN, and vimentin was downregulated in ROCK inhibited cells. Finally, we showed that ROCK inhibition by RKI-1447 suppressed medulloblastoma growth and proliferation in vivo. Collectively, our results suggest that ROCK inhibition presents a potential new therapeutic option in medulloblastoma, especially for children with metastatic disease.
- Published
- 2019
- Full Text
- View/download PDF
29. Analyses of FGFR3 and PIK3CA mutations in neuroblastomas and the effects of the corresponding inhibitors on neuroblastoma cell lines.
- Author
-
Kostopoulou ON, Holzhauser S, Lange BKA, Ohmayer A, Andonova T, Bersani C, Wickström M, and Dalianis T
- Subjects
- Aminopyridines pharmacology, Aminopyridines therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Apoptosis drug effects, Benzamides pharmacology, Benzamides therapeutic use, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Child, Child, Preschool, Class I Phosphatidylinositol 3-Kinases antagonists & inhibitors, DNA Mutational Analysis, Drug Resistance, Neoplasm genetics, Female, Humans, Imidazoles pharmacology, Imidazoles therapeutic use, Infant, Infant, Newborn, Male, Morpholines pharmacology, Morpholines therapeutic use, Mutation, Neuroblastoma drug therapy, Phosphoinositide-3 Kinase Inhibitors therapeutic use, Piperazines pharmacology, Piperazines therapeutic use, Pyrazoles pharmacology, Pyrazoles therapeutic use, Quinolines pharmacology, Quinolines therapeutic use, Receptor, Fibroblast Growth Factor, Type 3 antagonists & inhibitors, Antineoplastic Combined Chemotherapy Protocols pharmacology, Class I Phosphatidylinositol 3-Kinases genetics, Neuroblastoma genetics, Phosphoinositide-3 Kinase Inhibitors pharmacology, Receptor, Fibroblast Growth Factor, Type 3 genetics
- Abstract
Fibroblast growth factor receptor (FGFR)3 and phosphatidylinositol‑4,5‑bisphosphate 3‑kinase, catalytic subunit alpha (PIK3CA) mutations are found in various types of cancer, such as urinary bladder cancer, human papillomavirus‑positive tonsillar and base of the tongue squamous cell carcinoma, breast cancer and some childhood sarcomas. Several drugs can target these genes, some of which have been used for the treatment of urinary bladder cancer. Much less is known about childhood cancer. For this reason, the present study investigated the presence of such mutations in neuroblastomas (NBs) and tested NB cell lines for sensitivity to FGFR and phosphoinositide 3‑kinase (PI3K) inhibitors. In total, 29 NBs were examined for the presence of the three most common FGFR3 and PIK3CA mutations using a competitive allele‑specific TaqMan PCR (CAST‑PCR). Furthermore, the SK‑N‑AS, SK‑N‑BE(2)‑C, SK‑N‑DZ, SK‑N‑FI and SK‑N‑SH NB cell lines (where SK‑N‑DZ had a deletion of PIK3C2G, none had FGFR mutations according to the Cancer Program's Dependency Map, but some were chemoresistant), were tested for sensitivity to FGFR (AZD4547) and PI3K (BEZ235 and BKM120) inhibitors by viability, cytotoxicity, apoptosis and proliferation assays. CAST‑PCR detected one FGFR3 mutation in 1/29 NBs. Following treatment with FGFR and PI3K inhibitors, a decrease in viability and proliferation was observed in the majority, but not all, the cell lines. Following combination treatment with both drugs, the sensitivity of all cell lines was increased. On the whole, the findings of this study demonstrate that FGFR3 and PIK3CA mutations are uncommon in patients with NB. However, certain NB cell lines are rather sensitive to both FGFR and PI3K inhibitors alone, and even more so when the different drugs are used in combination.
- Published
- 2019
- Full Text
- View/download PDF
30. In vitro antitumor effects of FGFR and PI3K inhibitors on human papillomavirus positive and negative tonsillar and base of tongue cancer cell lines.
- Author
-
Holzhauser S, Kostopoulou ON, Ohmayer A, Lange BKA, Ramqvist T, Andonova T, Bersani C, Wickström M, and Dalianis T
- Abstract
Human papillomavirus positive (HPV
+ ) tonsillar and base of tongue squamous cell carcinoma (TSCC/BOTSCC) have better outcomes than corresponding HPV- negative (HPV- ) cancer cases. Our previous study demonstrated that fibroblast growth factor receptor 3 (FGFR3) and phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit a (PIK3CA) are often mutated in HPV+ cancer. To investigate whether targeted therapy is an option for TSCC/BOTSCC, two HPV+ and one HPV- TSCC/BOTSCC cell lines were tested for their sensitivity towards FGFR and PI3K inhibitors. The HPV+ cell lines UM-SCC-47 and UPCI-SCC-154, and the HPV- cell line UT-SSC-60A were tested by competitive allele-specific TaqMan-PCR for presence/absence of frequently occurring FGFR3 and PIK3CA mutations. All cells were then treated with FGFR inhibitor AZD4547 and PI3K inhibitors BEZ235 and BKM120 alone, or with AZD4547 and BEZ235 in combination. Viability was analyzed using a WST-1 assay, cytotoxicity tested by a CellTox Green cytotoxicity assay, apoptosis analyzed by a Caspase Glo 3/7 assay and proliferation examined with the xCELLigence system. HPV+ UM-SCC-47 and UPCI-SCC-154 cells, and HPV- UT-SSC-60A cells, did not exhibit any common FGFR3 or PIK3CA mutations, but were all sensitive to FGFR inhibitor AZD4547 and PI3K inhibitors BEZ235 and BKM120. Notably, HPV+ UPCI-SCC-154 cells were more sensitive than the other two cell lines. Furthermore, when AZD4547 and BEZ235 treatment was combined in HPV+ UPCI-SCC-154 and HPV- UT-SSC-60A cells, potentiated combination effects were observed. HPV+ UM-SCC-47 and UPCI-SCC-154 cells, and HPV- UT-SSC-60A cells had no common FGFR3 or PIK3CA mutations, but were sensitive to FGFR inhibitor AZD4547, and PI3K inhibitors BEZ235 and BKM120. Furthermore, the latter two cell lines were particularly sensitive to combinations of AZD4547 and BEZ235., (Copyright © 2019, Spandidos Publications.)- Published
- 2019
- Full Text
- View/download PDF
31. An intervention using the Parenting Toolkit "Children-What does it involve?" and the Real-Care-Baby simulator among students with intellectual disability-A feasibility study.
- Author
-
Janeslätt G, Larsson M, Wickström M, Springer L, and Höglund B
- Subjects
- Adolescent, Adult, Feasibility Studies, Female, Humans, Infant, Male, Pregnancy, Schools, Young Adult, Child Rearing, Health Education methods, Health Knowledge, Attitudes, Practice, Intellectual Disability rehabilitation, Pregnancy in Adolescence prevention & control, Simulation Training methods, Students
- Abstract
Background: There is limited knowledge about how young people with intellectual disability can be facilitated in their process of deciding about parenthood. This study aimed to evaluate the feasibility of an upcoming trial to evaluate an intervention using the Toolkit "Children-what does it involve?" and the "Real-Care-Baby" (RCB) simulator among students with intellectual disability., Methods: Six students with intellectual disability participated in an intervention with eight educational sessions and a 3-day caring session with the RCB simulator. Data were collected with questionnaires and interviews., Results: The study showed that it is possible to evaluate an intervention using these instruments among students with intellectual disability in order to provide them with further insights about parenthood., Conclusion: It is feasible to evaluate the Toolkit and the RCB in a cluster-randomized study and that such a study could add to our knowledge about possible intervention strategies regarding reproduction and parenting among students with intellectual disability., (© 2018 John Wiley & Sons Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
32. SYK Inhibition Potentiates the Effect of Chemotherapeutic Drugs on Neuroblastoma Cells in Vitro.
- Author
-
Tümmler C, Dumitriu G, Wickström M, Coopman P, Valkov A, Kogner P, Johnsen JI, Moens U, and Sveinbjörnsson B
- Abstract
Neuroblastoma is a malignancy arising from the developing sympathetic nervous system and the most common and deadly cancer of infancy. New therapies are needed to improve the prognosis for high-risk patients and to reduce toxicity and late effects. Spleen tyrosine kinase (SYK) has previously been identified as a promising drug target in various inflammatory diseases and cancers but has so far not been extensively studied as a potential therapeutic target in neuroblastoma. In this study, we observed elevated SYK gene expression in neuroblastoma compared to neural crest and benign neurofibroma. While SYK protein was detected in the majority of examined neuroblastoma tissues it was less frequently observed in neuroblastoma cell lines. Depletion of SYK by siRNA and the use of small molecule SYK inhibitors significantly reduced the cell viability of neuroblastoma cell lines expressing SYK protein. Moreover, SYK inhibition decreased ERK1/2 and Akt phosphorylation. The SYK inhibitor BAY 613606 enhanced the effect of different chemotherapeutic drugs. Transient expression of a constitutive active SYK variant increased the viability of neuroblastoma cells independent of endogenous SYK levels. Collectively, our findings suggest that targeting SYK in combination with conventional chemotherapy should be further evaluated as a treatment option in neuroblastoma., Competing Interests: The authors declare no conflict of interest.
- Published
- 2019
- Full Text
- View/download PDF
33. Neuroblastoma-A Neural Crest Derived Embryonal Malignancy.
- Author
-
Johnsen JI, Dyberg C, and Wickström M
- Abstract
Neuroblastoma is a neural crest derived malignancy of the peripheral nervous system and is the most common and deadliest tumor of infancy. It is characterized by clinical heterogeneity with a disease spectrum ranging from spontaneous regression without any medical intervention to treatment resistant tumors with metastatic spread and poor patient survival. The events that lead to the development of neuroblastoma from the neural crest have not been fully elucidated. Here we discuss factors and processes within the neural crest that when dysregulated have the potential to be initiators or drivers of neuroblastoma development. A more precise biological understanding of neuroblastoma causes and cell of origin is highly warranted. This will give valuable information for the development of medicines that specifically target molecules within neuroblastoma cells and also give hint about the mechanisms behind treatment resistance that is frequently seen in neuroblastoma.
- Published
- 2019
- Full Text
- View/download PDF
34. Molecular mechanisms and therapeutic targets in neuroblastoma.
- Author
-
Johnsen JI, Dyberg C, Fransson S, and Wickström M
- Subjects
- Antineoplastic Agents pharmacology, Child, Drug Discovery methods, Drug Resistance, Neoplasm, Drugs, Investigational pharmacology, Drugs, Investigational therapeutic use, Humans, Neuroblastoma metabolism, Neuroblastoma pathology, Peripheral Nervous System Neoplasms metabolism, Peripheral Nervous System Neoplasms pathology, Quality of Life, Sympathetic Nervous System drug effects, Sympathetic Nervous System metabolism, Sympathetic Nervous System pathology, Antineoplastic Agents therapeutic use, Molecular Targeted Therapy methods, Neuroblastoma drug therapy, Peripheral Nervous System Neoplasms drug therapy
- Abstract
Neuroblastoma is the most common extracranical tumor of childhood and the most deadly tumor of infancy. It is characterized by early age onset and high frequencies of metastatic disease but also the capacity to spontaneously regress. Despite intensive therapy, the survival for patients with high-risk neuroblastoma and those with recurrent or relapsed disease is low. Hence, there is an urgent need to develop new therapies for these patient groups. The molecular pathogenesis based on high-throughput omics technologies of neuroblastoma is beginning to be resolved which have given the opportunity to develop personalized therapies for high-risk patients. Here we discuss the potential of developing targeted therapies against aberrantly expressed molecules detected in sub-populations of neuroblastoma patients and how these selected targets can be drugged in order to overcome treatment resistance, improve survival and quality of life for these patients and also the possibilities to transfer preclinical research into clinical testing., (Copyright © 2018 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
35. Rho-associated kinase is a therapeutic target in neuroblastoma.
- Author
-
Dyberg C, Fransson S, Andonova T, Sveinbjörnsson B, Lännerholm-Palm J, Olsen TK, Forsberg D, Herlenius E, Martinsson T, Brodin B, Kogner P, Johnsen JI, and Wickström M
- Subjects
- Animals, Cell Line, Tumor, Glycogen Synthase Kinase 3 beta metabolism, Humans, Mice, Mice, Nude, N-Myc Proto-Oncogene Protein metabolism, Protein Kinase Inhibitors, Xenograft Model Antitumor Assays, rho-Associated Kinases metabolism, Neuroblastoma drug therapy, Neuroblastoma enzymology, Neuroblastoma pathology, Signal Transduction drug effects, rho-Associated Kinases antagonists & inhibitors
- Abstract
Neuroblastoma is a peripheral neural system tumor that originates from the neural crest and is the most common and deadly tumor of infancy. Here we show that neuroblastoma harbors frequent mutations of genes controlling the Rac/Rho signaling cascade important for proper migration and differentiation of neural crest cells during neuritogenesis. RhoA is activated in tumors from neuroblastoma patients, and elevated expression of Rho-associated kinase (ROCK)2 is associated with poor patient survival. Pharmacological or genetic inhibition of ROCK1 and 2, key molecules in Rho signaling, resulted in neuroblastoma cell differentiation and inhibition of neuroblastoma cell growth, migration, and invasion. Molecularly, ROCK inhibition induced glycogen synthase kinase 3β-dependent phosphorylation and degradation of MYCN protein. Small-molecule inhibition of ROCK suppressed MYCN -driven neuroblastoma growth in TH- MYCN homozygous transgenic mice and MYCN gene-amplified neuroblastoma xenograft growth in nude mice. Interference with Rho/Rac signaling might offer therapeutic perspectives for high-risk neuroblastoma., Competing Interests: The authors declare no conflict of interest.
- Published
- 2017
- Full Text
- View/download PDF
36. Inhibition of chemerin/CMKLR1 axis in neuroblastoma cells reduces clonogenicity and cell viability in vitro and impairs tumor growth in vivo .
- Author
-
Tümmler C, Snapkov I, Wickström M, Moens U, Ljungblad L, Maria Elfman LH, Winberg JO, Kogner P, Johnsen JI, and Sveinbjørnsson B
- Abstract
Pro-inflammatory cells, cytokines, and chemokines are essential in promoting a tumor supporting microenvironment. Chemerin is a chemotactic protein and a natural ligand for the receptors CMKLR1, GPR1, and CCRL2. The chemerin/CMKLR1 axis is involved in immunity and inflammation, and it has also been implicated in obesity and cancer. In neuroblastoma, a childhood tumor of the peripheral nervous system we identified correlations between high CMKLR1 and GPR1 expression and reduced overall survival probability. CMKLR1, GPR1, and chemerin RNA and protein were detected in neuroblastoma cell lines and neuroblastoma primary tumor tissue. Chemerin induced calcium mobilization, increased MMP-2 synthesis as well as MAP-kinase- and Akt-mediated signaling in neuroblastoma cells. Stimulation of neuroblastoma cells with serum, TNFα or IL-1β increased chemerin secretion. The small molecule CMKLR1 antagonist α-NETA reduced the clonogenicity and viability of neuroblastoma cell lines indicating the chemerin/CMKLR1 axis as a promoting factor in neuroblastoma tumorigenesis. Furthermore, nude mice carrying neuroblastoma SK-N-AS cells as xenografts showed impaired tumor growth when treated daily with α-NETA from day 1 after tumor cell injection. This study demonstrates the potential of the chemerin/CMKLR1 axis as a prognostic factor and possible therapeutic target in neuroblastoma., Competing Interests: CONFLICTS OF INTEREST The authors declare no conflicts of interest.
- Published
- 2017
- Full Text
- View/download PDF
37. Melflufen - a peptidase-potentiated alkylating agent in clinical trials.
- Author
-
Wickström M, Nygren P, Larsson R, Harmenberg J, Lindberg J, Sjöberg P, Jerling M, Lehmann F, Richardson P, Anderson K, Chauhan D, and Gullbo J
- Abstract
Aminopeptidases like aminopeptidase N (APN, also known as CD13) play an important role not only in normal cellular functioning but also in the development of cancer, including processes like tumor cell invasion, differentiation, proliferation, apoptosis, motility, and angiogenesis. An increased expression of APN has been described in several types of human malignancies, especially those characterized by fast-growing and aggressive phenotypes, suggesting APN as a potential therapeutic target. Melphalan flufenamide ethyl ester (melflufen, previously denoted J1) is a peptidase-potentiated alkylating agent. Melflufen readily penetrates membranes and an equilibrium is rapidly achieved, followed by enzymatic cleavage in aminopeptidase positive cells, which results in trapping of less lipophilic metabolites. This targeting effect results in very high intracellular concentrations of its metabolite melphalan and subsequent apoptotic cell death. This results in a potency increase (melflufen vs melphalan) ranging from 10- to several 100-fold in different in vitro models. Melflufen triggers a rapid, robust, and an irreversible DNA damage which may account for its ability to overcome melphalan-resistance in multiple myeloma cells. Furthermore, anti-angiogenic properties of melflufen have been described. Consequently, it is hypothesized that melflufen could provide better efficacy but no more toxicity than what is achieved with melphalan, an assumption so far supported by experiences from hollow fiber and xenograft studies in rodents as well as by clinical data from patients with solid tumors and multiple myeloma. This review summarizes the current preclinical and clinical knowledge of melflufen., Competing Interests: CONFLICTS OF INTEREST JG, RL and PN are co-founders and minor shareholders of OncoPeptides AB. DC has received research support from OncoPeptides AB. JH, PS, MJ, PR, JG and FL are consultants of OncoPeptides AB, and JL is CEO of the company. JH, FL, JG, PN, RL and JL have stock options in the company. JL and JH are shareholders.
- Published
- 2017
- Full Text
- View/download PDF
38. Lithium protects hippocampal progenitors, cognitive performance and hypothalamus-pituitary function after irradiation to the juvenile rat brain.
- Author
-
Zhou K, Xie C, Wickström M, Dolga AM, Zhang Y, Li T, Xu Y, Culmsee C, Kogner P, Zhu C, and Blomgren K
- Subjects
- Animals, Anxiety etiology, Cell Line, Tumor, Cell Proliferation drug effects, Cognitive Dysfunction etiology, Disease Models, Animal, Hippocampus drug effects, Hippocampus radiation effects, Humans, Hyperkinesis etiology, Lithium pharmacology, Mice, Neurogenesis drug effects, Neurogenesis radiation effects, Rats, Stem Cells drug effects, Stem Cells radiation effects, Anxiety prevention & control, Cognitive Dysfunction prevention & control, Cranial Irradiation adverse effects, Hippocampus cytology, Hyperkinesis prevention & control, Lithium administration & dosage
- Abstract
Cranial radiotherapy in children typically causes delayed and progressive cognitive dysfunction and there is no effective preventive strategy for radiation-induced cognitive impairments. Here we show that lithium treatment reduced irradiation-induced progenitor cell death in the subgranular zone of the hippocampus, and subsequently ameliorated irradiation-reduced neurogenesis and astrogenesis in the juvenile rat brain. Irradiation-induced memory impairment, motor hyperactivity and anxiety-like behaviour were normalized by lithium treatment. Late-onset irradiation-induced hypopituitarism was prevented by lithium treatment. Additionally, lithium appeared relatively toxic to multiple cultured tumour cell lines, and did not improve viability of radiated DAOY cells in vitro. In summary, our findings demonstrate that lithium can be safely administered to prevent both short- and long-term injury to the juvenile brain caused by ionizing radiation.
- Published
- 2017
- Full Text
- View/download PDF
39. Establishment and characterization of an orthotopic patient-derived Group 3 medulloblastoma model for preclinical drug evaluation.
- Author
-
Sandén E, Dyberg C, Krona C, Gallo-Oller G, Olsen TK, Enríquez Pérez J, Wickström M, Estekizadeh A, Kool M, Visse E, Ekström TJ, Siesjö P, Johnsen JI, and Darabi A
- Subjects
- Animals, Biomarkers, Tumor metabolism, Cerebellar Neoplasms genetics, Cerebellar Neoplasms metabolism, Humans, Medulloblastoma genetics, Medulloblastoma metabolism, Mice, Mice, Inbred NOD, Mice, SCID, Neoplasm Transplantation, Patient-Specific Modeling, Tumor Cells, Cultured, Tumor Microenvironment, Biomarkers, Tumor genetics, Cell Culture Techniques methods, Cerebellar Neoplasms pathology, Medulloblastoma pathology, Spheroids, Cellular cytology
- Abstract
Medulloblastomas comprise a heterogeneous group of tumours and can be subdivided into four molecular subgroups (WNT, SHH, Group 3 and Group 4) with distinct prognosis, biological behaviour and implications for targeted therapies. Few experimental models exist of the aggressive and poorly characterized Group 3 tumours. In order to establish a reproducible transplantable Group 3 medulloblastoma model for preclinical therapeutic studies, we acquired a patient-derived tumour sphere culture and inoculated low-passage spheres into the cerebellums of NOD-scid mice. Mice developed symptoms of brain tumours with a latency of 17-18 weeks. Neurosphere cultures were re-established and serially transplanted for 3 generations, with a negative correlation between tumour latency and numbers of injected cells. Xenografts replicated the phenotype of the primary tumour, including high degree of clustering in DNA methylation analysis, high proliferation, expression of tumour markers, MYC amplification and elevated MYC expression, and sensitivity to the MYC inhibitor JQ1. Xenografts maintained maintained expression of tumour-derived VEGFA and stromal-derived COX-2. VEGFA, COX-2 and c-Myc are highly expressed in Group 3 compared to other medulloblastoma subgroups, suggesting that these molecules are relevant therapeutic targets in Group 3 medulloblastoma.
- Published
- 2017
- Full Text
- View/download PDF
40. Increased risk for mental illness, injuries, and violence in children born to mothers with intellectual disability: A register study in Sweden during 1999-2012.
- Author
-
Wickström M, Höglund B, Larsson M, and Lundgren M
- Subjects
- Adult, Child, Child, Preschool, Cohort Studies, Female, Humans, Infant, Infant, Newborn, Male, Odds Ratio, Registries, Risk Factors, Sweden epidemiology, Child Abuse statistics & numerical data, Child of Impaired Parents psychology, Intellectual Disability epidemiology, Mental Disorders epidemiology, Mothers psychology, Violence statistics & numerical data, Wounds and Injuries epidemiology
- Abstract
Several studies have demonstrated that mothers with intellectual disability (ID) have a higher prevalence of mental health illness, lower socio-economic status, and a higher risk of alcohol and drug use compared to mothers without ID. The children of mothers with ID are over-represented in child protection and legal proceedings but are generally a less studied group than the mothers. The aim of this study was to investigate if children born to mothers with ID had an increased risk of being diagnosed with mental illness, injuries, and violence compared with children of mothers without ID. The study comprised a population-based cohort of children born in Sweden between 1999 and 2005. Data were collected from the Medical Birth Register and linked with two other national registers; ICD-10 codes were used for medical diagnoses, including ID. The children were followed from birth to seven years of age. In total, 478,577 children were included, of whom 2749 were born to mothers with ID. Children of mothers with ID were at a greater risk of having mental health problems (adjusted odds ratio (OR)=2.02; 95% confidence interval (CI)=1.74-2.35) and ID (OR=4.14; CI=2.95-5.82) in early childhood. They had an increased risk for injuries due to falls (OR=1.15; Cl 1.04-1.27). The largest risk related to trauma was violence and child abuse (OR=3.11; CI=1.89-5.12). In conclusion, children of mothers with ID had an increased risk for injuries, violence, and child abuse. We therefore suggest that parents with ID should receive evidence based support so that their children receive the best care and protection., (Copyright © 2017 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
41. In vitro and in vivo activity of melflufen (J1)in lymphoma.
- Author
-
Delforoush M, Strese S, Wickström M, Larsson R, Enblad G, and Gullbo J
- Subjects
- Animals, Cell Line, Tumor, G2 Phase Cell Cycle Checkpoints drug effects, Humans, Lymphoma pathology, Melphalan administration & dosage, Melphalan adverse effects, Mice, Multiple Myeloma pathology, Phenylalanine administration & dosage, Phenylalanine adverse effects, Xenograft Model Antitumor Assays, Cell Proliferation drug effects, Lymphoma drug therapy, Melphalan analogs & derivatives, Multiple Myeloma drug therapy, Phenylalanine analogs & derivatives
- Abstract
Background: Melphalan has been used in the treatment of various hematologic malignancies for almost 60 years. Today it is part of standard therapy for multiple myeloma and also as part of myeloablative regimens in association with autologous allogenic stem cell transplantation. Melflufen (melphalan flufenamide ethyl ester, previously called J1) is an optimized derivative of melphalan providing targeted delivery of active metabolites to cells expressing aminopeptidases. The activity of melflufen has compared favorably with that of melphalan in a series of in vitro and in vivo experiments performed preferentially on different solid tumor models and multiple myeloma. Melflufen is currently being evaluated in a clinical phase I/II trial in relapsed or relapsed and refractory multiple myeloma., Methods: Cytotoxicity of melflufen was assayed in lymphoma cell lines and in primary tumor cells with the Fluorometric Microculture Cytotoxicity Assay and cell cycle analyses was performed in two of the cell lines. Melflufen was also investigated in a xenograft model with subcutaneous lymphoma cells inoculated in mice., Results: Melflufen showed activity with cytotoxic IC50-values in the submicromolar range (0.011-0.92 μM) in the cell lines, corresponding to a mean of 49-fold superiority (p < 0.001) in potency vs. melphalan. In the primary cultures melflufen yielded slightly lower IC50-values (2.7 nM to 0.55 μM) and an increased ratio vs. melphalan (range 13-455, average 108, p < 0.001). Treated cell lines exhibited a clear accumulation in the G2/M-phase of the cell cycle. Melflufen also showed significant activity and no, or minimal side effects in the xenografted animals., Conclusion: This study confirms previous reports of a targeting related potency superiority of melflufen compared to that of melphalan. Melflufen was active in cell lines and primary cultures of lymphoma cells, as well as in a xenograft model in mice and appears to be a candidate for further evaluation in the treatment of this group of malignant diseases.
- Published
- 2016
- Full Text
- View/download PDF
42. Planar cell polarity gene expression correlates with tumor cell viability and prognostic outcome in neuroblastoma.
- Author
-
Dyberg C, Papachristou P, Haug BH, Lagercrantz H, Kogner P, Ringstedt T, Wickström M, and Johnsen JI
- Subjects
- Animals, Cell Movement genetics, Cell Polarity genetics, Cell Survival genetics, Gene Expression Regulation, Neoplastic, Humans, Intracellular Signaling Peptides and Proteins genetics, LIM Domain Proteins genetics, Membrane Proteins genetics, Mice, Neuroblastoma pathology, Tumor Suppressor Proteins genetics, Wnt Signaling Pathway genetics, beta Catenin genetics, rho-Associated Kinases genetics, Carcinogenesis genetics, Intracellular Signaling Peptides and Proteins biosynthesis, LIM Domain Proteins biosynthesis, Membrane Proteins biosynthesis, Neuroblastoma genetics, Tumor Suppressor Proteins biosynthesis
- Abstract
Background: The non-canonical Wnt/Planar cell polarity (PCP) signaling pathway is a major player in cell migration during embryonal development and has recently been implicated in tumorigenesis., Methods: Transfections with cDNA plasmids or siRNA were used to increase and suppress Prickle1 and Vangl2 expression in neuroblastoma cells and in non-tumorigenic cells. Cell viability was measured by trypan blue exclusion and protein expression was determined with western blotting. Transcriptional activity was studied with luciferase reporter assay and mRNA expression with real-time RT-PCR. Immunofluorescence stainings were used to study the effects of Vangl2 overexpression in non-tumorigenic embryonic cells. Statistical significance was tested with t-test or one-way ANOVA., Results: Here we show that high expression of the PCP core genes Prickle1 and Vangl2 is associated with low-risk neuroblastoma, suppression of neuroblastoma cell growth and decreased Wnt/β-catenin signaling. Inhibition of Rho-associated kinases (ROCKs) that are important in mediating non-canonical Wnt signaling resulted in increased expression of Prickle1 and inhibition of β-catenin activity in neuroblastoma cells. In contrast, overexpression of Vangl2 in MYC immortalized neural stem cells induced accumulation of active β-catenin and decreased the neural differentiation marker Tuj1. Similarly, genetically modified mice with forced overexpression of Vangl2 in nestin-positive cells showed decreased Tuj1 differentiation marker during embryonal development., Conclusions: Our experimental data demonstrate that high expression of Prickle1 and Vangl2 reduce the growth of neuroblastoma cells and indicate different roles of PCP proteins in tumorigenic cells compared to normal cells. These results suggest that the activity of the non-canonical Wnt/PCP signaling pathway is important for neuroblastoma development and that manipulation of the Wnt/PCP pathway provides a possible therapy for neuroblastoma.
- Published
- 2016
- Full Text
- View/download PDF
43. Wingless/β-catenin signaling as a modulator of chemoresistance in cancer.
- Author
-
Johnsen JI, Wickström M, and Baryawno N
- Abstract
O6-methylguanine-DNA methyltransferase (MGMT) is a DNA repair enzyme involved in chemoresistance. We have shown that MGMT abundance is regulated by canonical Wingless (Wnt) signaling and that inhibition of Wnt signaling restores chemosensitivity in preclinical cancer models. These findings have direct therapeutic implications for the treatment of cancers with high MGMT expression.
- Published
- 2016
- Full Text
- View/download PDF
44. Wnt/β-catenin pathway regulates MGMT gene expression in cancer and inhibition of Wnt signalling prevents chemoresistance.
- Author
-
Wickström M, Dyberg C, Milosevic J, Einvik C, Calero R, Sveinbjörnsson B, Sandén E, Darabi A, Siesjö P, Kool M, Kogner P, Baryawno N, and Johnsen JI
- Subjects
- Animals, Benzeneacetamides pharmacology, Brain Neoplasms drug therapy, Brain Neoplasms genetics, Camptothecin analogs & derivatives, Camptothecin pharmacology, Celecoxib pharmacology, Cisplatin pharmacology, Colorectal Neoplasms drug therapy, Colorectal Neoplasms genetics, Dacarbazine analogs & derivatives, Dacarbazine pharmacology, Doxorubicin pharmacology, Flow Cytometry, Gene Expression Profiling, Gene Expression Regulation, Neoplastic drug effects, Glioma drug therapy, Glioma genetics, Glucose-6-Phosphate Isomerase, Heterocyclic Compounds, 3-Ring pharmacology, Humans, Immunoblotting, Immunohistochemistry, Irinotecan, Medulloblastoma drug therapy, Medulloblastoma genetics, Mice, Neoplasm Transplantation, Neoplasms drug therapy, Neuroblastoma drug therapy, Neuroblastoma genetics, Pyrans pharmacology, Pyrazines pharmacology, Pyridines pharmacology, Real-Time Polymerase Chain Reaction, Sulfones pharmacology, Temozolomide, Triazoles pharmacology, Vincristine pharmacology, Wnt Proteins genetics, Wnt Proteins metabolism, Wnt Signaling Pathway drug effects, beta Catenin genetics, beta Catenin metabolism, Antineoplastic Agents pharmacology, DNA Modification Methylases genetics, DNA Repair Enzymes genetics, Drug Resistance, Neoplasm genetics, Gene Expression Regulation, Neoplastic genetics, Neoplasms genetics, Tumor Suppressor Proteins genetics, Wnt Signaling Pathway genetics
- Abstract
The DNA repair enzyme O6-methylguanine-DNA methyltransferase (MGMT) is commonly overexpressed in cancers and is implicated in the development of chemoresistance. The use of drugs inhibiting MGMT has been hindered by their haematologic toxicity and inefficiency. As a different strategy to inhibit MGMT we investigated cellular regulators of MGMT expression in multiple cancers. Here we show a significant correlation between Wnt signalling and MGMT expression in cancers with different origin and confirm the findings by bioinformatic analysis and immunofluorescence. We demonstrate Wnt-dependent MGMT gene expression and cellular co-localization between active β-catenin and MGMT. Pharmacological or genetic inhibition of Wnt activity downregulates MGMT expression and restores chemosensitivity of DNA-alkylating drugs in mouse models. These findings have potential therapeutic implications for chemoresistant cancers, especially of brain tumours where the use of temozolomide is frequently used in treatment.
- Published
- 2015
- Full Text
- View/download PDF
45. Healthy late preterm infants and supplementary artificial milk feeds: effects on breast feeding and associated clinical parameters.
- Author
-
Mattsson E, Funkquist EL, Wickström M, Nyqvist KH, and Volgsten H
- Subjects
- Adult, Biomarkers, Female, Humans, Infant, Infant, Newborn, Mothers statistics & numerical data, Pregnancy, Weight Loss, Infant Nutritional Physiological Phenomena standards, Infant, Premature growth & development, Milk, Human
- Abstract
Objectives: to compare the influence of supplementary artificial milk feeds on breast feeding and certain clinical parameters among healthy late preterm infants given regular supplementary artificial milk feeds versus being exclusively breast fed from birth., Design: a comparative study using quantitative methods. Data were collected via a parental diary and medical records., Methods: parents of 77 late preterm infants (34 5/7-36 6/7 weeks), whose mothers intended to breast feed, completed a diary during the infants׳ hospital stay., Findings: infants who received regular supplementary artificial milk feeds experienced a longer delay before initiation of breast feeding, were breast fed less frequently and had longer hospital stays than infants exclusively breast fed from birth. Exclusively breast-fed infants had a greater weight loss than infants with regular artificial milk supplementation. A majority of the mothers (65%) with an infant prescribed artificial milk never expressed their milk and among the mothers who used a breast-pump, milk expression commenced late (10-84 hours after birth). At discharge, all infants were breast fed to some extent, 43% were exclusively breast fed., Key Conclusions: clinical practice and routines influence the initiation of breast feeding among late preterm infants and may act as barriers to the mothers׳ establishment of exclusive breast feeding., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
46. The impact of S6K1 kinase on neuroblastoma cell proliferation is independent of GLI1 signaling.
- Author
-
Diao Y, Rahman MF, Villegas VE, Wickström M, Johnsen JI, and Zaphiropoulos PG
- Subjects
- Cell Line, Tumor, Cell Proliferation drug effects, Gene Expression Regulation, Neoplastic, Gene Knockdown Techniques, Hedgehog Proteins metabolism, Humans, Imidazoles pharmacology, Neuroblastoma metabolism, Phosphorylation, Pyridines pharmacology, Pyrimidines pharmacology, Quinolines pharmacology, RNA, Small Interfering pharmacology, Ribosomal Protein S6 Kinases, 70-kDa antagonists & inhibitors, Signal Transduction drug effects, Transcription Factors antagonists & inhibitors, Zinc Finger Protein GLI1, Neuroblastoma pathology, Ribosomal Protein S6 Kinases, 70-kDa genetics, Ribosomal Protein S6 Kinases, 70-kDa metabolism, Transcription Factors genetics, Transcription Factors metabolism
- Abstract
Background: The crosstalk between Hedgehog (HH) signaling and other signal transduction cascades has been extensively studied in different cancers. In neuroblastoma, mTOR/S6K1 signaling is known to have a role in the development of this disease and recent evidence also implicates the HH pathway. Moreover, S6K1 kinase has been shown to phosphorylate GLI1, the effector of HH signaling, promoting GLI1 transcriptional activity and oncogenic function in esophageal adenocarcinoma. In this study, we examined the possible interplay of S6K1 and GLI1 signaling in neuroblastoma., Methods: siRNA knockdowns were used to suppress S6K1 and GLI1 expression, and the siRNA effects were validated by real-time PCR and Western blotting. Cell proliferation analysis was performed with the EdU incorporation assay. Cytotoxic analysis with increasing concentrations of PI3K/mTOR and GLI inhibitors, individually and in combination, was used to determine drug response., Results: Although knockdown of either S6K1 or GLI1 reduces the cellular proliferation of neuroblastoma cells, there is little effect of S6K1 on the expression of GLI1 mRNA and protein and on the capacity of GLI1 to activate target genes. No detectable phosphorylation of GLI1 is observed prior or following S6K1 knockdown. GLI1 overexpression can not rescue the reduced proliferation elicited by S6K1 knockdown. Moreover, inhibitors of PI3K/mTOR and GLI signaling reduced neuroblastoma cell growth, but no additional growth inhibitory effects were detected when the two classes of drugs were combined., Conclusion: Our results demonstrate that the impact of S6K1 kinase on neuroblastoma cells is not mediated through modulation of GLI1 expression/activity.
- Published
- 2014
- Full Text
- View/download PDF
47. Protective role of humanin on bortezomib-induced bone growth impairment in anticancer treatment.
- Author
-
Eriksson E, Wickström M, Perup LS, Johnsen JI, Eksborg S, Kogner P, and Sävendahl L
- Subjects
- Animals, Antineoplastic Agents administration & dosage, Boronic Acids administration & dosage, Bortezomib, Cell Line, Tumor, Chondrocytes drug effects, Dose-Response Relationship, Drug, Drug Administration Schedule, Femur drug effects, Growth Plate drug effects, Heterografts, Humans, Intracellular Signaling Peptides and Proteins administration & dosage, Male, Metatarsal Bones drug effects, Mice, Mice, Nude, Proteasome Inhibitors administration & dosage, Pyrazines administration & dosage, Time Factors, Antineoplastic Agents adverse effects, Apoptosis drug effects, Bone Development drug effects, Boronic Acids adverse effects, Intracellular Signaling Peptides and Proteins pharmacology, Medulloblastoma drug therapy, Neuroblastoma drug therapy, Proteasome Inhibitors adverse effects, Pyrazines adverse effects
- Abstract
Background: Bortezomib is a proteasome inhibitor currently studied in clinical trials of childhood cancers. So far, no side effects on bone growth have been reported in treated children. However, bortezomib was recently found to induce apoptosis in growth plate chondrocytes and impair linear bone growth in treated mice. We hypothesize that [Gly(14)]-humanin (HNG), a 24-amino acid synthetic antiapoptotic peptide, can prevent bortezomib-induced bone growth impairment., Methods: Mice with human neuroblastoma or medulloblastoma tumor xenografts (9-13 animals/group) received one 2-week cycle (2 injections/week) of bortezomib (0.8 mg/kg or 1.0mg/kg), or HNG (1 µg/mouse), or the combination of HNG/bortezomib, or vehicle. Cultures of human growth plate cartilage, chondrogenic- and cancer cell lines, and immunohistochemistry for detection of proapoptotic proteins were also used. Statistical significance was evaluated by two-sided Mann-Whitney U test or by parametric or nonparametric analysis of variance., Results: Bortezomib efficiently blocked the proteasome and induced pronounced impairment of linear bone growth from day 0 to day 13 (0.09 mm/day, 95% confidence interval [CI] = 0.07 to 0.11 mm/day; vs 0.19 mm/day, 95% CI = 0.15 to 0.23 mm/day in vehicle; P < .001), an effect significantly prevented by the addition of HNG (0.15 mm growth/day, 95% CI = 0.14 to 0.16 mm/day; P < .001 vs bortezomib only; P = 0.03 vs vehicle). Bortezomib was highly toxic when added to cultures of human growth plate cartilage, with markedly increased apoptosis compared with control (P < .001). However, when combining with HNG, bortezomib-induced apoptosis was entirely prevented, as was Bax and PARP activation. Bortezomib delayed tumor growth, and HNG did not interfere with the anticancer effect when studied in human tumor xenografts or cell lines., Conclusions: HNG prevents bortezomib-induced bone growth impairment without interfering with bortezomib's desired anticancer effects.
- Published
- 2014
- Full Text
- View/download PDF
48. The novel alkylating prodrug melflufen (J1) inhibits angiogenesis in vitro and in vivo.
- Author
-
Strese S, Wickström M, Fuchs PF, Fryknäs M, Gerwins P, Dale T, Larsson R, and Gullbo J
- Subjects
- Animals, CD13 Antigens metabolism, Cattle, Chick Embryo, Chorioallantoic Membrane drug effects, Drug Screening Assays, Antitumor methods, Human Umbilical Vein Endothelial Cells drug effects, Melphalan pharmacokinetics, Melphalan pharmacology, Mice, Phenylalanine pharmacology, Prodrugs pharmacology, Alkylating Agents pharmacology, Angiogenesis Inhibitors pharmacology, Melphalan analogs & derivatives, Neovascularization, Pathologic drug therapy, Phenylalanine analogs & derivatives
- Abstract
Aminopeptidase N (APN) has been reported to have a functional role in tumor angiogenesis and repeatedly reported to be over-expressed in human tumors. The melphalan-derived prodrug melphalan-flufenamide (melflufen, previously designated J1) can be activated by APN. This suggests that this alkylating prodrug may exert anti-angiogenic properties, which will possibly contribute to the anti-tumoral activity in vivo. This work presents a series of experiments designed to investigate this effect of melflufen. In a cytotoxicity assay we show that bovine endothelial cells were more than 200 times more sensitive to melflufen than to melphalan, in HUVEC cells the difference was more than 30-fold and accompanied by aminopetidase-mediated accumulation of intracellular melphalan. In the chicken embryo chorioallantoic membrane (CAM) assay it is indicated that both melflufen and melphalan inhibit vessel ingrowth. Two commercially available assays with human endothelial cells co-cultured with fibroblasts (TCS Cellworks AngioKit, and Essen GFP-AngioKit) also illustrate the superior anti-angiogenic effect of melflufen compared to melphalan. Finally, in a commercially available in vivo assay in mice (Cultrex DIVAA angio-reactor assay) melflufen displayed an anti-angiogenic effect, comparable to bevacizumab. In conclusion, this study demonstrates through all methods used, that melphalan-flufenamide besides being an alkylating agent also reveals anti-angiogenic effects in different preclinical models in vitro and in vivo., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
49. Screening for phenotype selective activity in multidrug resistant cells identifies a novel tubulin active agent insensitive to common forms of cancer drug resistance.
- Author
-
Fryknäs M, Gullbo J, Wang X, Rickardson L, Jarvius M, Wickström M, Hassan S, Andersson C, Gustafsson M, Westman G, Nygren P, Linder S, and Larsson R
- Subjects
- Animals, Apoptosis drug effects, Cell Line, Tumor, Drug Resistance, Multiple, Drug Screening Assays, Antitumor, Flow Cytometry, Inhibitory Concentration 50, Mice, Oligonucleotide Array Sequence Analysis, Antineoplastic Agents pharmacology, Drug Resistance, Neoplasm, Hydroxyquinolines pharmacology, Neoplasms, Tubulin drug effects
- Abstract
Background: Drug resistance is a common cause of treatment failure in cancer patients and encompasses a multitude of different mechanisms. The aim of the present study was to identify drugs effective on multidrug resistant cells., Methods: The RPMI 8226 myeloma cell line and its multidrug resistant subline 8226/Dox40 was screened for cytotoxicity in response to 3,000 chemically diverse compounds using a fluorometric cytotoxicity assay (FMCA). Follow-up profiling was subsequently performed using various cellular and biochemical assays., Results: One compound, designated VLX40, demonstrated a higher activity against 8226/Dox40 cells compared to its parental counterpart. VLX40 induced delayed cell death with apoptotic features. Mechanistic exploration was performed using gene expression analysis of drug exposed tumor cells to generate a drug-specific signature. Strong connections to tubulin inhibitors and microtubule cytoskeleton were retrieved. The mechanistic hypothesis of VLX40 acting as a tubulin inhibitor was confirmed by direct measurements of interaction with tubulin polymerization using a biochemical assay and supported by demonstration of G2/M cell cycle arrest. When tested against a broad panel of primary cultures of patient tumor cells (PCPTC) representing different forms of leukemia and solid tumors, VLX40 displayed high activity against both myeloid and lymphoid leukemias in contrast to the reference compound vincristine to which myeloid blast cells are often insensitive. Significant in vivo activity was confirmed in myeloid U-937 cells implanted subcutaneously in mice using the hollow fiber model., Conclusions: The results indicate that VLX40 may be a useful prototype for development of novel tubulin active agents that are insensitive to common mechanisms of cancer drug resistance.
- Published
- 2013
- Full Text
- View/download PDF
50. Alternative cytotoxic effects of the postulated IGF-IR inhibitor picropodophyllin in vitro.
- Author
-
Wu X, Sooman L, Wickström M, Fryknäs M, Dyrager C, Lennartsson J, and Gullbo J
- Subjects
- Carcinoma, Squamous Cell metabolism, Cell Line, Tumor, Dose-Response Relationship, Drug, ErbB Receptors antagonists & inhibitors, ErbB Receptors metabolism, Esophageal Neoplasms metabolism, Humans, Microtubules metabolism, Phosphorylation drug effects, Podophyllotoxin toxicity, Protein Kinase Inhibitors pharmacology, Receptor, IGF Type 1 metabolism, Signal Transduction drug effects, Tubulin Modulators pharmacology, Antineoplastic Agents toxicity, Podophyllotoxin analogs & derivatives, Receptor, IGF Type 1 antagonists & inhibitors
- Abstract
The insulin-like growth factor-1 (IGF-I) and its receptors play an important role in transformation and progression of several malignancies. Inhibitors of this pathway have been developed and evaluated but generally performed poorly in clinical trials, and several drug candidates have been abandoned. The cyclolignan picropodophyllin (PPP) has been described as a potent and selective IGF-IR inhibitor and is currently undergoing clinical trials. We investigated PPP's activity in panels of human cancer cell lines (e.g., esophageal squamous carcinoma cell lines) but found no effects on the phosphorylation or expression of IGF-IR. Nor was the cytotoxic activity of PPP related to the presence or spontaneous phosphorylation of IGF-IR. However, its activity correlated with that of known tubulin inhibitors, and it destabilized microtubule assembly at cytotoxic concentrations also achievable in patients. PPP is a stereoisomer of podophyllotoxin (PPT), a potent tubulin inhibitor, and an equilibrium between the two has previously been described. PPP could thus potentially act as a reservoir for the continuous generation of low doses of PPT. Interestingly, PPP also inhibited downstream signaling from tyrosine kinase receptors, including the serine/threonine kinase Akt. This effect is associated with microtubule-related downregulation of the EGF receptor, rather than the IGF-IR. These results suggest that the cytotoxicity and pAkt inhibition observed following treatment with the cyclolignan PPP in vitro result from microtubule inhibition (directly or indirectly by spontaneous PPT formation), rather than any effect on IGF-IR. It is also suggested that PPT should be used as a reference compound in all future studies on PPP.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.