85 results on '"Uhrbom L"'
Search Results
2. Sox5 can suppress platelet-derived growth factor B-induced glioma development in Ink4a-deficient mice through induction of acute cellular senescence
- Author
-
Tchougounova, E, Jiang, Y, Bråsäter, D, Lindberg, N, Kastemar, M, Asplund, A, Westermark, B, and Uhrbom, L
- Published
- 2009
- Full Text
- View/download PDF
3. Loss of Arf causes tumor progression of PDGFB-induced oligodendroglioma
- Author
-
Tchougounova, E, Kastemar, M, Bråsäter, D, Holland, E C, Westermark, B, and Uhrbom, L
- Published
- 2007
- Full Text
- View/download PDF
4. Snail depletes the tumorigenic potential of glioblastoma
- Author
-
Savary, K, Caglayan, D, Caja, L, Tzavlaki, K, Nayeem, S, Bergström, Bin T, Jiang, Y, Uhrbom, L, Forsberg-Nilsson, K, Westermark, B, Heldin, C-H, Ferletta, M, and Moustakas, A
- Published
- 2013
- Full Text
- View/download PDF
5. CB-09 * THE CELL OF ORIGIN FOR GLIOBLASTOMA CONTRIBUTES TO THE PHENOTYPIC HETEROGENEITY OF GLIOMA STEM CELLS
- Author
-
Jiang, Y., primary, Marinescu, V. D., additional, Xie, Y., additional, Haglund, C., additional, Jarvius, M., additional, Lindberg, N., additional, Olofsson, T., additional, Hesselager, G., additional, Alafuzoff, I., additional, Fryknas, M., additional, Larsson, R., additional, Nelander, S., additional, and Uhrbom, L., additional
- Published
- 2014
- Full Text
- View/download PDF
6. Comparative drug pair screening across multiple glioblastoma cell lines reveals novel drug-drug interactions
- Author
-
Schmidt, L., primary, Kling, T., additional, Monsefi, N., additional, Olsson, M., additional, Hansson, C., additional, Baskaran, S., additional, Lundgren, B., additional, Martens, U., additional, Haggblad, M., additional, Westermark, B., additional, Forsberg Nilsson, K., additional, Uhrbom, L., additional, Karlsson-Lindahl, L., additional, Gerlee, P., additional, and Nelander, S., additional
- Published
- 2013
- Full Text
- View/download PDF
7. Glioma-derived plasminogen activator inhibitor-1 (PAI-1) regulates the recruitment of LRP1 positive mast cells
- Author
-
Roy A, Coum A, Vd, Marinescu, Põlajeva J, Smits A, Nelander S, Uhrbom L, Bengt Westermark, Forsberg-Nilsson K, Pontén F, and Tchougounova E
8. Induction of brain tumors in mice using a recombinant platelet-derived growth factor B-chain retrovirus
- Author
-
Uhrbom L, Hesselager G, Nistér M, and Bengt Westermark
- Subjects
Male ,Platelet-Derived Growth Factor ,Receptor, Platelet-Derived Growth Factor alpha ,Brain Neoplasms ,3T3 Cells ,Proto-Oncogene Proteins c-sis ,Recombinant Proteins ,Clone Cells ,Disease Models, Animal ,Mice ,Proto-Oncogene Proteins ,Animals ,Female ,Receptors, Platelet-Derived Growth Factor ,RNA, Messenger ,Moloney murine leukemia virus - Abstract
In existing mouse models for malignant brain tumors, genes with no proven pathogenical relevance for humans have been used. Coexpression of platelet-derived growth factor (PDGF) and PDGF receptors suggests an autocrine mechanism of growth factor stimulation in the development of brain tumors in man. A murine retrovirus coding for the PDGF B-chain was, therefore, used to induce brain tumors in mice. Of 35 mice who received injections, 15 developed brain tumors of oligo- or monoclonal origin. They coexpressed PDGF B-chain and alpha-receptor mRNA, as expected, from an autocrine mechanism of transformation. Most tumors displayed characteristics of glioblastoma multiforme or of a primitive neuroectodermal tumor, and the consistent expression of nestin suggested that they were all derived from an immature neuroglial progenitor. The results show that an autocrine mechanism of transformation may be an initial or early event in neuro-oncogenesis. The present model provides an ideal system for studies of genetic mechanisms involved in the development of brain tumors.
9. miRNA-21 is developmentally regulated in mouse brain and is co-expressed with SOX2 in glioma
- Author
-
Põlajeva Jelena, Swartling Fredrik J, Jiang Yiwen, Singh Umashankar, Pietras Kristian, Uhrbom Lene, Westermark Bengt, and Roswall Pernilla
- Subjects
miRNA ,miR-21 ,Glioma ,PDGF-BB ,SOX2 ,Imatinib (Gleevec) ,RCAS/tv-a ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background MicroRNAs (miRNAs) and their role during tumor development have been studied in great detail during the last decade, albeit their expression pattern and regulation during normal development are however not so well established. Previous studies have shown that miRNAs are differentially expressed in solid human tumors. Platelet-derived growth factor (PDGF) signaling is known to be involved in normal development of the brain as well as in malignant primary brain tumors, gliomas, but the complete mechanism is still lacking. We decided to investigate the expression of the oncogenic miR-21 during normal mouse development and glioma, focusing on PDGF signaling as a potential regulator of miR-21. Methods We generated mouse glioma using the RCAS/tv-a system for driving PDGF-BB expression in a cell-specific manner. Expression of miR-21 in mouse cell cultures and mouse brain were assessed using Northern blot analysis and in situ hybridization. Immunohistochemistry and Western blot analysis were used to investigate SOX2 expression. LNA-modified siRNA was used for irreversible depletion of miR-21. For inhibition of PDGF signaling Gleevec (imatinib mesylate), Rapamycin and U0126, as well as siRNA were used. Statistical significance was calculated using double-sided unpaired Student´s t-test. Results We identified miR-21 to be highly expressed during embryonic and newborn brain development followed by a gradual decrease until undetectable at postnatal day 7 (P7), this pattern correlated with SOX2 expression. Furthermore, miR-21 and SOX2 showed up-regulation and overlapping expression pattern in RCAS/tv-a generated mouse brain tumor specimens. Upon irreversible depletion of miR-21 the expression of SOX2 was strongly diminished in both mouse primary glioma cultures and human glioma cell lines. Interestingly, in normal fibroblasts the expression of miR-21 was induced by PDGF-BB, and inhibition of PDGF signaling in mouse glioma primary cultures resulted in suppression of miR-21 suggesting that miR-21 is indeed regulated by PDGF signaling. Conclusions Our data show that miR-21 and SOX2 are tightly regulated already during embryogenesis and define a distinct population with putative tumor cell of origin characteristics. Furthermore, we believe that miR-21 is a mediator of PDGF-driven brain tumors, which suggests miR-21 as a promising target for treatment of glioma.
- Published
- 2012
- Full Text
- View/download PDF
10. GABA-A channel subunit expression in cell lines derived from human glioblastoma.
- Author
-
Babateen, O., Jin, Z., Bhandage, A., Westermark, B., Forsberg-Nilsson, K., Uhrbom, L., Smits, A., and Birnir, B.
- Subjects
GLIOMAS ,AMINOBUTYRIC acid ,CELL lines - Abstract
Gliomas are the most common form of primary brain tumors with an overall incidence of about 4 - 5 per 105 persons. There is growing evidence for a widespread role of the neurotransmitter γ-aminobutyric acid (GABA) in the growth regulation of many cell types, including neuronal stem cells and perhaps tumor stem cells. Recently we showed that human brain tumors express GABA-A receptors subunits that are generally down-regulated in glioblastomas (Smits et al, 2012). We further showed that there was a correlation between GABA-A channel subunits and patient survival. Whether tumor cells that are equipped with functional GABA-A channels maintain proliferative activity upon in physiological concentrations of GABA may depend on which subtype of GABA-A channels are activated. We have now examined 8 glioblastoma derived celllines and examined which GABA-A subunits are expressed and then, selected one cell line to study in detail the functional and pharmacological properties of the GABA-A channels expressed. We used the method of RT-PCR for determining the subunits expressed, immunocytochemistry to identify cellular location of subunits and the whole-cell patch-clamp technique to record GABA-activated currents. Our results show that the eight different cell lines all express GABA-A receptor subunits but the type of subunits that are expressed varies. Cell line U3047 expressed α1, α2, α3, α5, β2, β3, δ, γ3 and θ GABA-A receptors subunits mRNAs and was selected for studies of properties of the GABA-A receptors expressed in glial/tumor cells. Immunocytochemistry demonstrated expression of the α3 and β3 proteins. GABA concentrations ranging from 1 to 1000 μM were applied to the cells and gave a half-maximal concentration of current activation (EC50) of 10 μM. The current response to 10 μM GABA was enhanced by 1 μM diazepam and 50 μM propofol, modulators of GABA-A channels. The currents were inhibited by the GABA-A channels antagonists SR-95531 (100 μM) and picrotoxin (100 μM). Our results show that the GABA-A receptors in the glioblastoma cell line U3047 are functional and respond to classical agonists and antagonists of GABA-A receptors. The profile of the subunits expressed is similar to what we reported in glioblastoma tumors from humans (Smits et al., 2012). We propose that the cell line can be used as a model system to study how GABA signaling affects tumor development. [ABSTRACT FROM AUTHOR]
- Published
- 2013
11. Electrode plate for lead accumulators
- Author
-
Uhrbom, L
- Published
- 1980
12. TAK1 inhibition leads to RIPK1-dependent apoptosis in immune-activated cancers.
- Author
-
Damhofer H, Tatar T, Southgate B, Scarneo S, Agger K, Shlyueva D, Uhrbom L, Morrison GM, Hughes PF, Haystead T, Pollard SM, and Helin K
- Subjects
- Humans, Cytokines, MAP Kinase Kinase Kinases antagonists & inhibitors, MAP Kinase Kinase Kinases metabolism, Signal Transduction, Transforming Growth Factor beta, Tumor Necrosis Factor-alpha, Apoptosis genetics, Glioblastoma genetics, Glioblastoma immunology, Glioblastoma metabolism, Glioblastoma pathology, Glioma genetics, Glioma immunology, Glioma metabolism, Glioma pathology, Receptor-Interacting Protein Serine-Threonine Kinases metabolism
- Abstract
Poor survival and lack of treatment response in glioblastoma (GBM) is attributed to the persistence of glioma stem cells (GSCs). To identify novel therapeutic approaches, we performed CRISPR/Cas9 knockout screens and discovered TGFβ activated kinase (TAK1) as a selective survival factor in a significant fraction of GSCs. Loss of TAK1 kinase activity results in RIPK1-dependent apoptosis via Caspase-8/FADD complex activation, dependent on autocrine TNFα ligand production and constitutive TNFR signaling. We identify a transcriptional signature associated with immune activation and the mesenchymal GBM subtype to be a characteristic of cancer cells sensitive to TAK1 perturbation and employ this signature to accurately predict sensitivity to the TAK1 kinase inhibitor HS-276. In addition, exposure to pro-inflammatory cytokines IFNγ and TNFα can sensitize resistant GSCs to TAK1 inhibition. Our findings reveal dependency on TAK1 kinase activity as a novel vulnerability in immune-activated cancers, including mesenchymal GBMs that can be exploited therapeutically., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
13. Live Detection of Neural Progenitors and Glioblastoma Cells by an Oligothiophene Derivative.
- Author
-
Ilkhanizadeh S, Gracias A, Åslund AKO, Bäck M, Simon R, Kavanagh E, Migliori B, Neofytou C, Nelander S, Westermark B, Uhrbom L, Forsberg-Nilsson K, Konradsson P, Teixeira AI, Uhlén P, Joseph B, Hermanson O, and Nilsson KPR
- Subjects
- Adult, Humans, Brain, Adapalene, Glioblastoma diagnosis, Brain Neoplasms diagnosis, Neural Stem Cells
- Abstract
There is an urgent need for simple and non-invasive identification of live neural stem/progenitor cells (NSPCs) in the developing and adult brain as well as in disease, such as in brain tumors, due to the potential clinical importance in prognosis, diagnosis, and treatment of diseases of the nervous system. Here, we report a luminescent conjugated oligothiophene (LCO), named p-HTMI, for non-invasive and non-amplified real-time detection of live human patient-derived glioblastoma (GBM) stem cell-like cells and NSPCs. While p-HTMI stained only a small fraction of other cell types investigated, the mere addition of p-HTMI to the cell culture resulted in efficient detection of NSPCs or GBM cells from rodents and humans within minutes. p-HTMI is functionalized with a methylated imidazole moiety resembling the side chain of histidine/histamine, and non-methylated analogues were not functional. Cell sorting experiments of human GBM cells demonstrated that p-HTMI labeled the same cell population as CD271, a proposed marker for stem cell-like cells and rapidly migrating cells in glioblastoma. Our results suggest that the LCO p-HTMI is a versatile tool for immediate and selective detection of neural and glioma stem and progenitor cells.
- Published
- 2023
- Full Text
- View/download PDF
14. Tailoring vascular phenotype through AAV therapy promotes anti-tumor immunity in glioma.
- Author
-
Ramachandran M, Vaccaro A, van de Walle T, Georganaki M, Lugano R, Vemuri K, Kourougkiaouri D, Vazaios K, Hedlund M, Tsaridou G, Uhrbom L, Pietilä I, Martikainen M, van Hooren L, Olsson Bontell T, Jakola AS, Yu D, Westermark B, Essand M, and Dimberg A
- Subjects
- Mice, Animals, Phenotype, Brain, Tumor Microenvironment, Glioma genetics, Glioma therapy, Brain Neoplasms genetics, Brain Neoplasms therapy, Brain Neoplasms blood supply, Glioblastoma genetics
- Abstract
Glioblastomas are aggressive brain tumors that are largely immunotherapy resistant. This is associated with immunosuppression and a dysfunctional tumor vasculature, which hinder T cell infiltration. LIGHT/TNFSF14 can induce high endothelial venules (HEVs) and tertiary lymphoid structures (TLS), suggesting that its therapeutic expression could promote T cell recruitment. Here, we use a brain endothelial cell-targeted adeno-associated viral (AAV) vector to express LIGHT in the glioma vasculature (AAV-LIGHT). We found that systemic AAV-LIGHT treatment induces tumor-associated HEVs and T cell-rich TLS, prolonging survival in αPD-1-resistant murine glioma. AAV-LIGHT treatment reduces T cell exhaustion and promotes TCF1
+ CD8+ stem-like T cells, which reside in TLS and intratumoral antigen-presenting niches. Tumor regression upon AAV-LIGHT therapy correlates with tumor-specific cytotoxic/memory T cell responses. Our work reveals that altering vascular phenotype through vessel-targeted expression of LIGHT promotes efficient anti-tumor T cell responses and prolongs survival in glioma. These findings have broader implications for treatment of other immunotherapy-resistant cancers., Competing Interests: Declaration of interests M.R., A.V., T.v.d.W., M.E., and A.D. have filed a patent application for using an AAV to deliver the LIGHT gene to tumor endothelial cells. The patent application includes examples from the article., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
15. Wnt signaling regulates MFSD2A-dependent drug delivery through endothelial transcytosis in glioma.
- Author
-
Xie Y, He L, Zhang Y, Huang H, Yang F, Chao M, Cao H, Wang J, Li Y, Zhang L, Xin L, Xiao B, Shi X, Zhang X, Tang J, Uhrbom L, Dimberg A, Wang L, and Zhang L
- Subjects
- Mice, Animals, Wnt Signaling Pathway, Endothelial Cells metabolism, Temozolomide therapeutic use, Transcytosis, Cell Line, Tumor, Drug Resistance, Neoplasm, Glioma drug therapy, Glioma metabolism, Glioblastoma pathology, Brain Neoplasms pathology, Symporters metabolism, Symporters therapeutic use
- Abstract
Background: Systemic delivery of anti-tumor therapeutic agents to brain tumors is thwarted by the blood-brain barrier (BBB), an organotypic specialization of brain endothelial cells (ECs). A failure of pharmacological compounds to cross BBB is one culprit for the dismal prognosis of glioblastoma (GBM) patients. Identification of novel vascular targets to overcome the challenges posed by the BBB in tumors for GBM treatment is urgently needed., Methods: Temozolomide (TMZ) delivery was investigated in CT2A and PDGFB-driven RCAS/tv-a orthotopic glioma models. Transcriptome analysis was performed on ECs from murine gliomas. Mfsd2a deficient, Cav1 deficient, and Mfsd2a EC-specific inducible mice were developed to study the underlying molecular mechanisms., Results: We demonstrated that inhibiting Wnt signaling by LGK974 could increase TMZ delivery and sensitize glioma to chemotherapy in both murine glioma models. Transcriptome analysis of ECs from murine gliomas revealed that Wnt signaling inhibition enhanced vascular transcytosis as indicated by the upregulation of PLVAP and downregulation of MFSD2A. Mfsd2a deficiency in mice enhances TMZ delivery in tumors, whereas constitutive expression of Mfsd2a in ECs suppresses the enhanced TMZ delivery induced by Wnt pathway inhibition in murine glioma. In addition, Wnt signaling inhibition enhanced caveolin-1 (Cav1)-positive caveolae-mediated transcytosis in tumor ECs. Moreover, Wnt signaling inhibitor or Mfsd2a deficiency fails to enhance TMZ penetration in tumors from Cav1-deficient mice., Conclusions: These results demonstrated that Wnt signaling regulates MFSD2A-dependent TMZ delivery through a caveolae-mediated EC transcytosis pathway. Our findings identify Wnt signaling as a promising therapeutic target to improve drug delivery for GBM treatment., (© The Author(s) 2023. Published by Oxford University Press on behalf of the Society for Neuro-Oncology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
16. Glioblastoma stem cells express non-canonical proteins and exclusive mesenchymal-like or non-mesenchymal-like protein signatures.
- Author
-
Babačić H, Galardi S, Umer HM, Hellström M, Uhrbom L, Maturi N, Cardinali D, Pellegatta S, Michienzi A, Trevisi G, Mangiola A, Lehtiö J, Ciafrè SA, and Pernemalm M
- Subjects
- Humans, Proteomics, Neoplastic Stem Cells metabolism, Cell Line, Tumor, Glioblastoma genetics, Glioblastoma metabolism, Brain Neoplasms metabolism
- Abstract
Glioblastoma (GBM) cancer stem cells (GSCs) contribute to GBM's origin, recurrence, and resistance to treatment. However, the understanding of how mRNA expression patterns of GBM subtypes are reflected at global proteome level in GSCs is limited. To characterize protein expression in GSCs, we performed in-depth proteogenomic analysis of patient-derived GSCs by RNA-sequencing and mass-spectrometry. We quantified > 10 000 proteins in two independent GSC panels and propose a GSC-associated proteomic signature characterizing two distinct phenotypic conditions; one defined by proteins upregulated in proneural and classical GSCs (GPC-like), and another by proteins upregulated in mesenchymal GSCs (GM-like). The GM-like protein set in GBM tissue was associated with necrosis, recurrence, and worse overall survival. Through proteogenomics, we discovered 252 non-canonical peptides in the GSCs, i.e., protein sequences that are variant or derive from genome regions previously considered non-protein-coding, including variants of the heterogeneous ribonucleoproteins implicated in RNA splicing. In summary, GSCs express two protein sets that have an inverse association with clinical outcomes in GBM. The discovery of non-canonical protein sequences questions existing gene models and pinpoints new protein targets for research in GBM., (© 2022 The Authors. Molecular Oncology published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)
- Published
- 2023
- Full Text
- View/download PDF
17. Novel cancer gene discovery using a forward genetic screen in RCAS-PDGFB-driven gliomas.
- Author
-
Weishaupt H, Čančer M, Rosén G, Holmberg KO, Häggqvist S, Bunikis I, Jiang Y, Sreedharan S, Gyllensten U, Becher OJ, Uhrbom L, Ameur A, and Swartling FJ
- Subjects
- Animals, Humans, Mice, Genetic Association Studies, Oncogenes, Proto-Oncogene Proteins c-sis genetics, Brain Neoplasms pathology, Glioma pathology
- Abstract
Background: Malignant gliomas, the most common malignant brain tumors in adults, represent a heterogeneous group of diseases with poor prognosis. Retroviruses can cause permanent genetic alterations that modify genes close to the viral integration site., Methods: Here we describe the use of a high-throughput pipeline coupled to the commonly used tissue-specific retroviral RCAS-TVA mouse tumor model system. Utilizing next-generation sequencing, we show that retroviral integration sites can be reproducibly detected in malignant stem cell lines generated from RCAS-PDGFB-driven glioma biopsies., Results: A large fraction of common integration sites contained genes that have been dysregulated or misexpressed in glioma. Others overlapped with loci identified in previous glioma-related forward genetic screens, but several novel putative cancer-causing genes were also found. Integrating retroviral tagging and clinical data, Ppfibp1 was highlighted as a frequently tagged novel glioma-causing gene. Retroviral integrations into the locus resulted in Ppfibp1 upregulation, and Ppfibp1-tagged cells generated tumors with shorter latency on orthotopic transplantation. In human gliomas, increased PPFIBP1 expression was significantly linked to poor prognosis and PDGF treatment resistance., Conclusions: Altogether, the current study has demonstrated a novel approach to tagging glioma genes via forward genetics, validating previous results, and identifying PPFIBP1 as a putative oncogene in gliomagenesis., (© The Author(s) 2022. Published by Oxford University Press on behalf of the Society for Neuro-Oncology.)
- Published
- 2023
- Full Text
- View/download PDF
18. Cell-lineage controlled epigenetic regulation in glioblastoma stem cells determines functionally distinct subgroups and predicts patient survival.
- Author
-
Lu X, Maturi NP, Jarvius M, Yildirim I, Dang Y, Zhao L, Xie Y, Tan EJ, Xing P, Larsson R, Fryknäs M, Uhrbom L, and Chen X
- Subjects
- Cell Lineage genetics, Chromatin genetics, Epigenesis, Genetic, Humans, Neoplastic Stem Cells, Glioblastoma genetics
- Abstract
There is ample support for developmental regulation of glioblastoma stem cells. To examine how cell lineage controls glioblastoma stem cell function, we present a cross-species epigenome analysis of mouse and human glioblastoma stem cells. We analyze and compare the chromatin-accessibility landscape of nine mouse glioblastoma stem cell cultures of three defined origins and 60 patient-derived glioblastoma stem cell cultures by assay for transposase-accessible chromatin using sequencing. This separates the mouse cultures according to cell of origin and identifies three human glioblastoma stem cell clusters that show overlapping characteristics with each of the mouse groups, and a distribution along an axis of proneural to mesenchymal phenotypes. The epigenetic-based human glioblastoma stem cell clusters display distinct functional properties and can separate patient survival. Cross-species analyses reveals conserved epigenetic regulation of mouse and human glioblastoma stem cells. We conclude that epigenetic control of glioblastoma stem cells primarily is dictated by developmental origin which impacts clinically relevant glioblastoma stem cell properties and patient survival., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
19. FACT-seq: profiling histone modifications in formalin-fixed paraffin-embedded samples with low cell numbers.
- Author
-
Zhao L, Xing P, Polavarapu VK, Zhao M, Valero-Martínez B, Dang Y, Maturi N, Mathot L, Neves I, Yildirim I, Swartling FJ, Sjöblom T, Uhrbom L, and Chen X
- Subjects
- Animals, Cell Line, Humans, Mice, Protein Processing, Post-Translational, Staphylococcal Protein A metabolism, Transposases metabolism, Chromatin metabolism, Epigenesis, Genetic, Histones analysis
- Abstract
The majority of biopsies in both basic research and translational cancer studies are preserved in the format of archived formalin-fixed paraffin-embedded (FFPE) samples. Profiling histone modifications in archived FFPE tissues is critically important to understand gene regulation in human disease. The required input for current genome-wide histone modification profiling studies from FFPE samples is either 10-20 tissue sections or whole tissue blocks, which prevents better resolved analyses. But it is desirable to consume a minimal amount of FFPE tissue sections in the analysis as clinical tissues of interest are limited. Here, we present FFPE tissue with antibody-guided chromatin tagmentation with sequencing (FACT-seq), the first highly sensitive method to efficiently profile histone modifications in FFPE tissues by combining a novel fusion protein of hyperactive Tn5 transposase and protein A (T7-pA-Tn5) transposition and T7 in vitro transcription. FACT-seq generates high-quality chromatin profiles from different histone modifications with low number of FFPE nuclei. We proved a very small piece of FFPE tissue section containing ∼4000 nuclei is sufficient to decode H3K27ac modifications with FACT-seq. H3K27ac FACT-seq revealed disease-specific super enhancers in the archived FFPE human colorectal and human glioblastoma cancer tissue. In summary, FACT-seq allows decoding the histone modifications in archival FFPE tissues with high sensitivity and help researchers to better understand epigenetic regulation in cancer and human disease., (© The Author(s) 2021. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2021
- Full Text
- View/download PDF
20. Key molecular alterations in endothelial cells in human glioblastoma uncovered through single-cell RNA sequencing.
- Author
-
Xie Y, He L, Lugano R, Zhang Y, Cao H, He Q, Chao M, Liu B, Cao Q, Wang J, Jiao Y, Hu Y, Han L, Zhang Y, Huang H, Uhrbom L, Betsholtz C, Wang L, Dimberg A, and Zhang L
- Subjects
- Biological Variation, Population, Brain pathology, Drug Delivery Systems methods, Drug Discovery, Humans, Sequence Analysis, RNA methods, Single-Cell Analysis methods, Biological Transport genetics, Blood-Brain Barrier metabolism, Blood-Brain Barrier physiopathology, Brain Neoplasms drug therapy, Brain Neoplasms genetics, Brain Neoplasms pathology, Carrier Proteins genetics, Cell Membrane Permeability genetics, Endothelial Cells metabolism, Endothelial Cells pathology, Glioblastoma drug therapy, Glioblastoma genetics, Glioblastoma pathology
- Abstract
Passage of systemically delivered pharmacological agents into the brain is largely blocked by the blood-brain-barrier (BBB), an organotypic specialization of brain endothelial cells (ECs). Tumor vessels in glioblastoma (GBM), the most common malignant brain tumor in humans, are abnormally permeable, but this phenotype is heterogeneous and may differ between the tumor's center and invasive front. Here, through single-cell RNA sequencing (scRNA-seq) of freshly isolated ECs from human glioblastoma and paired tumor peripheral tissues, we have constructed a molecular atlas of human brain ECs providing unprecedented molecular insight into the heterogeneity of the human BBB and its molecular alteration in glioblastoma. We identified 5 distinct EC phenotypes representing different states of EC activation and BBB impairment, and associated with different anatomical locations within and around the tumor. This unique data resource provides key information for designing rational therapeutic regimens and optimizing drug delivery.
- Published
- 2021
- Full Text
- View/download PDF
21. A Patient-Derived Cell Atlas Informs Precision Targeting of Glioblastoma.
- Author
-
Johansson P, Krona C, Kundu S, Doroszko M, Baskaran S, Schmidt L, Vinel C, Almstedt E, Elgendy R, Elfineh L, Gallant C, Lundsten S, Ferrer Gago FJ, Hakkarainen A, Sipilä P, Häggblad M, Martens U, Lundgren B, Frigault MM, Lane DP, Swartling FJ, Uhrbom L, Nestor M, Marino S, and Nelander S
- Subjects
- Animals, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Bortezomib pharmacology, Cell Proliferation drug effects, Cyclin-Dependent Kinase Inhibitor p15 genetics, Cyclin-Dependent Kinase Inhibitor p16 genetics, Gene Regulatory Networks drug effects, Genetic Heterogeneity, Genome, Human, Glioblastoma genetics, Humans, Mice, Inbred BALB C, Mutation genetics, Proteasome Inhibitors pharmacology, Tumor Cells, Cultured, Tumor Suppressor Protein p53 metabolism, Glioblastoma drug therapy, Glioblastoma pathology, Molecular Targeted Therapy, Precision Medicine
- Abstract
Glioblastoma (GBM) is a malignant brain tumor with few therapeutic options. The disease presents with a complex spectrum of genomic aberrations, but the pharmacological consequences of these aberrations are partly unknown. Here, we report an integrated pharmacogenomic analysis of 100 patient-derived GBM cell cultures from the human glioma cell culture (HGCC) cohort. Exploring 1,544 drugs, we find that GBM has two main pharmacological subgroups, marked by differential response to proteasome inhibitors and mutually exclusive aberrations in TP53 and CDKN2A/B. We confirm this trend in cell and in xenotransplantation models, and identify both Bcl-2 family inhibitors and p53 activators as potentiators of proteasome inhibitors in GBM cells. We can further predict the responses of individual cell cultures to several existing drug classes, presenting opportunities for drug repurposing and design of stratified trials. Our functionally profiled biobank provides a valuable resource for the discovery of new treatments for GBM., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
22. A molecularly distinct subset of glioblastoma requires serum-containing media to establish sustainable bona fide glioblastoma stem cell cultures.
- Author
-
Maturi NP, Tan EJ, Xie Y, Sundström A, Bergström T, Jiang Y, and Uhrbom L
- Subjects
- Animals, Cell Culture Techniques methods, Cell Line, Tumor, Cell Proliferation physiology, Mice, Transgenic, Brain Neoplasms pathology, Glioblastoma pathology, Neoplastic Stem Cells pathology, Neural Stem Cells pathology
- Abstract
Glioblastoma (GBM) is the most frequent and deadly primary malignant brain tumor. Hallmarks are extensive intra-tumor and inter-tumor heterogeneity and highly invasive growth, which provide great challenges for treatment. Efficient therapy is lacking and the majority of patients survive less than 1 year from diagnosis. GBM progression and recurrence is caused by treatment-resistant glioblastoma stem cells (GSCs). GSC cultures are considered important models in target identification and drug screening studies. The current state-of-the-art method, to isolate and maintain GSC cultures that faithfully mimic the primary tumor, is to use serum-free (SF) media conditions developed for neural stem cells (NSCs). Here we have investigated the outcome of explanting 218 consecutively collected GBM patient samples under both SF and standard, serum-containing media conditions. The frequency of maintainable SF cultures (SFCs) was most successful, but for a subgroup of GBM specimens, a viable culture could only be established in serum-containing media, called exclusive serum culture (ESC). ESCs expressed nestin and SOX2, and displayed all functional characteristics of a GSC, that is, extended proliferation, sustained self-renewal and orthotopic tumor initiation. Once adapted to the in vitro milieu they were also sustainable in SF media. Molecular analyses showed that ESCs formed a discrete group that was most related to the mesenchymal GBM subtype. This distinct subgroup of GBM that would have evaded modeling in SF conditions only provide unique cell models of GBM inter-tumor heterogeneity., (© 2019 Wiley Periodicals, Inc.)
- Published
- 2020
- Full Text
- View/download PDF
23. BET and Aurora Kinase A inhibitors synergize against MYCN-positive human glioblastoma cells.
- Author
-
Čančer M, Drews LF, Bengtsson J, Bolin S, Rosén G, Westermark B, Nelander S, Forsberg-Nilsson K, Uhrbom L, Weishaupt H, and Swartling FJ
- Subjects
- Adult, Aged, Azepines administration & dosage, Azepines pharmacology, Brain Neoplasms genetics, Brain Neoplasms metabolism, Brain Neoplasms pathology, Cell Line, Tumor, Drug Synergism, Female, Glioblastoma drug therapy, Humans, Male, Middle Aged, N-Myc Proto-Oncogene Protein biosynthesis, N-Myc Proto-Oncogene Protein metabolism, Temozolomide administration & dosage, Temozolomide pharmacology, Triazoles administration & dosage, Triazoles pharmacology, Antineoplastic Combined Chemotherapy Protocols pharmacology, Aurora Kinase A antagonists & inhibitors, Brain Neoplasms drug therapy, N-Myc Proto-Oncogene Protein genetics, Proteins antagonists & inhibitors
- Abstract
Glioblastoma multiforme (GBM) is the most common primary malignant brain tumor in adults. Patients usually undergo surgery followed by aggressive radio- and chemotherapy with the alkylating agent temozolomide (TMZ). Still, median survival is only 12-15 months after diagnosis. Many human cancers including GBMs demonstrate addiction to MYC transcription factor signaling and can become susceptible to inhibition of MYC downstream genes. JQ1 is an effective inhibitor of BET Bromodomains, a class of epigenetic readers regulating expression of downstream MYC targets. Here, we show that BET inhibition decreases viability of patient-derived GBM cell lines. We propose a distinct expression signature of MYCN-elevated GBM cells that correlates with significant sensitivity to BET inhibition. In tumors showing JQ1 sensitivity, we found enrichment of pathways regulating cell cycle, DNA damage response and repair. As DNA repair leads to acquired chemoresistance to TMZ, JQ1 treatment in combination with TMZ synergistically inhibited proliferation of MYCN-elevated cells. Bioinformatic analyses further showed that the expression of MYCN correlates with Aurora Kinase A levels and Aurora Kinase inhibitors indeed showed synergistic efficacy in combination with BET inhibition. Collectively, our data suggest that BET inhibitors could potentiate the efficacy of either TMZ or Aurora Kinase inhibitors in GBM treatment.
- Published
- 2019
- Full Text
- View/download PDF
24. Human Mesenchymal glioblastomas are characterized by an increased immune cell presence compared to Proneural and Classical tumors.
- Author
-
Kaffes I, Szulzewsky F, Chen Z, Herting CJ, Gabanic B, Velázquez Vega JE, Shelton J, Switchenko JM, Ross JL, McSwain LF, Huse JT, Westermark B, Nelander S, Forsberg-Nilsson K, Uhrbom L, Maturi NP, Cimino PJ, Holland EC, Kettenmann H, Brennan CW, Brat DJ, and Hambardzumyan D
- Abstract
Glioblastoma (GBM) is the most aggressive malignant primary brain tumor in adults, with a median survival of 14.6 months. Recent efforts have focused on identifying clinically relevant subgroups to improve our understanding of pathogenetic mechanisms and patient stratification. Concurrently, the role of immune cells in the tumor microenvironment has received increasing attention, especially T cells and tumor-associated macrophages (TAM). The latter are a mixed population of activated brain-resident microglia and infiltrating monocytes/monocyte-derived macrophages, both of which express ionized calcium-binding adapter molecule 1 (IBA1). This study investigated differences in immune cell subpopulations among distinct transcriptional subtypes of GBM. Human GBM samples were molecularly characterized and assigned to Proneural, Mesenchymal or Classical subtypes as defined by NanoString nCounter Technology. Subsequently, we performed and analyzed automated immunohistochemical stainings for TAM as well as specific T cell populations. The Mesenchymal subtype of GBM showed the highest presence of TAM, CD8
+ , CD3+ and FOXP3+ T cells, as compared to Proneural and Classical subtypes. High expression levels of the TAM-related gene AIF1 , which encodes the TAM-specific protein IBA1, correlated with a worse prognosis in Proneural GBM, but conferred a survival benefit in Mesenchymal tumors. We used our data to construct a mathematical model that could reliably identify Mesenchymal GBM with high sensitivity using a combination of the aforementioned cell-specific IHC markers. In conclusion, we demonstrated that molecularly distinct GBM subtypes are characterized by profound differences in the composition of their immune microenvironment, which could potentially help to identify tumors amenable to immunotherapy., (© 2019 The Author(s). Published by Taylor & Francis.)- Published
- 2019
- Full Text
- View/download PDF
25. LGR5 promotes tumorigenicity and invasion of glioblastoma stem-like cells and is a potential therapeutic target for a subset of glioblastoma patients.
- Author
-
Xie Y, Sundström A, Maturi NP, Tan EJ, Marinescu VD, Jarvius M, Tirfing M, Jin C, Chen L, Essand M, Swartling FJ, Nelander S, Jiang Y, and Uhrbom L
- Subjects
- Animals, Brain Neoplasms genetics, Brain Neoplasms pathology, Brain Neoplasms therapy, Cell Self Renewal, Drug Resistance, Neoplasm, Gene Expression Regulation, Neoplastic, Glioblastoma genetics, Glioblastoma pathology, Glioblastoma therapy, Humans, Mice, Inbred NOD, Mice, SCID, Neoplasm Invasiveness, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells pathology, Neoplastic Stem Cells radiation effects, Oligodendrocyte Transcription Factor 2 genetics, Oligodendrocyte Transcription Factor 2 metabolism, Phenotype, Radiation Tolerance, Receptors, G-Protein-Coupled genetics, Signal Transduction, Tumor Cells, Cultured, Brain Neoplasms metabolism, Cell Movement drug effects, Cell Movement radiation effects, Cell Proliferation drug effects, Cell Proliferation radiation effects, Glioblastoma metabolism, Neoplastic Stem Cells metabolism, Receptors, G-Protein-Coupled metabolism
- Abstract
Glioblastoma (GBM) is the most common and lethal primary malignant brain tumor which lacks efficient treatment and predictive biomarkers. Expression of the epithelial stem cell marker Leucine-rich repeat-containing G-protein coupled receptor 5 (LGR5) has been described in GBM, but its functional role has not been conclusively elucidated. Here, we have investigated the role of LGR5 in a large repository of patient-derived GBM stem cell (GSC) cultures. The consequences of LGR5 overexpression or depletion have been analyzed using in vitro and in vivo methods, which showed that, among those with highest LGR5 expression (LGR5
high ), there were two phenotypically distinct groups: one that was dependent on LGR5 for its malignant properties and another that was unaffected by changes in LGR5 expression. The LGR5-responding cultures could be identified by their significantly higher self-renewal capacity as measured by extreme limiting dilution assay (ELDA), and these LGR5high -ELDAhigh cultures were also significantly more malignant and invasive compared to the LGR5high -ELDAlow cultures. This showed that LGR5 expression alone would not be a strict marker of LGR5 responsiveness. In a search for additional biomarkers, we identified LPAR4, CCND2, and OLIG2 that were significantly upregulated in LGR5-responsive GSC cultures, and we found that OLIG2 together with LGR5 were predictive of GSC radiation and drug response. Overall, we show that LGR5 regulates the malignant phenotype in a subset of patient-derived GSC cultures, which supports its potential as a predictive GBM biomarker. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd., (Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.)- Published
- 2019
- Full Text
- View/download PDF
26. Microglia Induce PDGFRB Expression in Glioma Cells to Enhance Their Migratory Capacity.
- Author
-
Wallmann T, Zhang XM, Wallerius M, Bolin S, Joly AL, Sobocki C, Leiss L, Jiang Y, Bergh J, Holland EC, Enger PØ, Andersson J, Swartling FJ, Miletic H, Uhrbom L, Harris RA, and Rolny C
- Abstract
High-grade gliomas (HGGs) are the most aggressive and invasive primary brain tumors. The platelet-derived growth factor (PDGF) signaling pathway drives HGG progression, and enhanced expression of PDGF receptors (PDGFRs) is a well-established aberration in a subset of glioblastomas (GBMs). PDGFRA is expressed in glioma cells, whereas PDGFRB is mostly restricted to the glioma-associated stroma. Here we show that the spatial location of TAMMs correlates with the expansion of a subset of tumor cells that have acquired expression of PDGFRB in both mouse and human low-grade glioma and HCGs. Furthermore, M2-polarized microglia but not bone marrow (BM)-derived macrophages (BMDMs) induced PDGFRB expression in glioma cells and stimulated their migratory capacity. These findings illustrate a heterotypic cross-talk between microglia and glioma cells that may enhance the migratory and invasive capacity of the latter by inducing PDGFRB., (Copyright © 2018 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
27. Snail regulates BMP and TGFβ pathways to control the differentiation status of glioma-initiating cells.
- Author
-
Caja L, Tzavlaki K, Dadras MS, Tan EJ, Hatem G, Maturi NP, Morén A, Wik L, Watanabe Y, Savary K, Kamali-Moghaddan M, Uhrbom L, Heldin CH, and Moustakas A
- Subjects
- Animals, Bone Morphogenetic Proteins metabolism, Brain Neoplasms genetics, Cell Differentiation, Cell Line, Tumor, Gene Expression Regulation, Neoplastic, Glioblastoma genetics, Humans, Mice, Neoplasm Transplantation, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, Snail Family Transcription Factors metabolism, Transforming Growth Factor beta metabolism, Brain Neoplasms metabolism, Gene Expression Profiling methods, Glioblastoma metabolism, Neoplastic Stem Cells cytology, Signal Transduction
- Abstract
Glioblastoma multiforme is a brain malignancy characterized by high heterogeneity, invasiveness, and resistance to current therapies, attributes related to the occurrence of glioma stem cells (GSCs). Transforming growth factor β (TGFβ) promotes self-renewal and bone morphogenetic protein (BMP) induces differentiation of GSCs. BMP7 induces the transcription factor Snail to promote astrocytic differentiation in GSCs and suppress tumor growth in vivo. We demonstrate that Snail represses stemness in GSCs. Snail interacts with SMAD signaling mediators, generates a positive feedback loop of BMP signaling and transcriptionally represses the TGFB1 gene, decreasing TGFβ1 signaling activity. Exogenous TGFβ1 counteracts Snail function in vitro, and in vivo promotes proliferation and re-expression of Nestin, confirming the importance of TGFB1 gene repression by Snail. In conclusion, novel insight highlights mechanisms whereby Snail differentially regulates the activity of the opposing BMP and TGFβ pathways, thus promoting an astrocytic fate switch and repressing stemness in GSCs.
- Published
- 2018
- Full Text
- View/download PDF
28. Retraction Notice to: Vulnerability of Glioblastoma Cells to Catastrophic Vacuolization and Death Induced by a Small Molecule.
- Author
-
Kitambi SS, Toledo EM, Usoskin D, Wee S, Harisankar A, Svensson R, Sigmundsson K, Kalderén C, Niklasson M, Kundu S, Aranda S, Westermark B, Uhrbom L, Andäng M, Damberg P, Nelander S, Arenas E, Artursson P, Walfridsson J, Nilsson KF, Hammarström LGJ, and Ernfors P
- Published
- 2017
- Full Text
- View/download PDF
29. Mast Cell Infiltration in Human Brain Metastases Modulates the Microenvironment and Contributes to the Metastatic Potential.
- Author
-
Roy A, Libard S, Weishaupt H, Gustavsson I, Uhrbom L, Hesselager G, Swartling FJ, Pontén F, Alafuzoff I, and Tchougounova E
- Abstract
Metastatic brain tumors continue to be a clinical problem, despite new therapeutic advances in cancer treatment. Brain metastases (BMs) are among the most common mass lesions in the brain that are resistant to chemotherapies, have a very poor prognosis, and currently lack any efficient diagnostic tests. Predictions estimate that about 40% of lung and breast cancer patients will develop BM. Despite this, very little is known about the immunological and genetic aberrations that drive tumorigenesis in BM. In this study, we demonstrate the infiltration of mast cells (MCs) in a large cohort of human BM samples with different tissues of origin for primary cancer. We applied patient-derived BM cell models to the study of BM cell-MC interactions. BM cells when cocultured with MCs demonstrate enhanced growth and self-renewal capacity. Gene set enrichment analyses indicate increased expression of signal transduction and transmembrane proteins related genes in the cocultured BM cells. MCs exert their effect by release of mediators such as IL-8, IL-10, matrix metalloprotease 2, and vascular endothelial growth factor, thereby permitting metastasis. In conclusion, we provide evidence for a role of MCs in BM. Our findings indicate MCs' capability of modulating gene expression in BM cells and suggest that MCs can serve as a new target for drug development against metastases in the brain.
- Published
- 2017
- Full Text
- View/download PDF
30. Glioblastoma Cell Malignancy and Drug Sensitivity Are Affected by the Cell of Origin.
- Author
-
Jiang Y, Marinescu VD, Xie Y, Jarvius M, Maturi NP, Haglund C, Olofsson S, Lindberg N, Olofsson T, Leijonmarck C, Hesselager G, Alafuzoff I, Fryknäs M, Larsson R, Nelander S, and Uhrbom L
- Published
- 2017
- Full Text
- View/download PDF
31. Membrane-Depolarizing Channel Blockers Induce Selective Glioma Cell Death by Impairing Nutrient Transport and Unfolded Protein/Amino Acid Responses.
- Author
-
Niklasson M, Maddalo G, Sramkova Z, Mutlu E, Wee S, Sekyrova P, Schmidt L, Fritz N, Dehnisch I, Kyriatzis G, Krafcikova M, Carson BB, Feenstra JM, Marinescu VD, Segerman A, Haraldsson M, Gustavsson AL, Hammarström LG, Jenmalm Jensen A, Uhrbom L, Altelaar AF, Linnarsson S, Uhlén P, Trantirek L, Vincent CT, Nelander S, Enger PØ, and Andäng M
- Subjects
- Animals, Biological Transport, Brain Neoplasms metabolism, Brain Neoplasms pathology, Cell Death, Cell Line, Tumor, Dihydropyridines pharmacology, Glioma metabolism, Glioma pathology, Humans, Mice, Mycotoxins pharmacology, Neoplastic Stem Cells pathology, Proteomics, Sodium metabolism, Amino Acids metabolism, Brain Neoplasms drug therapy, Calcium Channel Blockers pharmacology, Calcium Channels, T-Type physiology, Glioma drug therapy, Potassium Channels, Calcium-Activated antagonists & inhibitors, Unfolded Protein Response drug effects
- Abstract
Glioma-initiating cells (GIC) are considered the underlying cause of recurrences of aggressive glioblastomas, replenishing the tumor population and undermining the efficacy of conventional chemotherapy. Here we report the discovery that inhibiting T-type voltage-gated Ca
2+ and KCa channels can effectively induce selective cell death of GIC and increase host survival in an orthotopic mouse model of human glioma. At present, the precise cellular pathways affected by the drugs affecting these channels are unknown. However, using cell-based assays and integrated proteomics, phosphoproteomics, and transcriptomics analyses, we identified the downstream signaling events these drugs affect. Changes in plasma membrane depolarization and elevated intracellular Na+ , which compromised Na+ -dependent nutrient transport, were documented. Deficits in nutrient deficit acted in turn to trigger the unfolded protein response and the amino acid response, leading ultimately to nutrient starvation and GIC cell death. Our results suggest new therapeutic targets to attack aggressive gliomas. Cancer Res; 77(7); 1741-52. ©2017 AACR ., (©2017 American Association for Cancer Research.)- Published
- 2017
- Full Text
- View/download PDF
32. Mouse Models of Pediatric Supratentorial High-grade Glioma Reveal How Cell-of-Origin Influences Tumor Development and Phenotype.
- Author
-
Sreedharan S, Maturi NP, Xie Y, Sundström A, Jarvius M, Libard S, Alafuzoff I, Weishaupt H, Fryknäs M, Larsson R, Swartling FJ, and Uhrbom L
- Subjects
- Adult, Animals, Cell Lineage, Child, Disease Models, Animal, Gene Expression Profiling, Humans, Mice, Mice, Transgenic, Oligonucleotide Array Sequence Analysis, Phenotype, Principal Component Analysis, Transcriptome, Glioma pathology, Neural Stem Cells pathology, Neurons pathology, Oligodendroglia pathology, Supratentorial Neoplasms pathology
- Abstract
High-grade glioma (HGG) is a group of primary malignant brain tumors with dismal prognosis. Whereas adult HGG has been studied extensively, childhood HGG, a relatively rare disease, is less well-characterized. Here, we present two novel platelet-derived growth factor (PDGF)-driven mouse models of pediatric supratentorial HGG. Tumors developed from two different cells of origin reminiscent of neural stem cells (NSC) or oligodendrocyte precursor cells (OPC). Cross-species transcriptomics showed that both models are closely related to human pediatric HGG as compared with adult HGG. Furthermore, an NSC-like cell-of-origin enhanced tumor incidence, malignancy, and the ability of mouse glioma cells (GC) to be cultured under stem cell conditions as compared with an OPC-like cell. Functional analyses of cultured GC from these tumors showed that cells of NSC-like origin were more tumorigenic, had a higher rate of self-renewal and proliferation, and were more sensitive to a panel of cancer drugs compared with GC of a more differentiated origin. These two mouse models relevant to human pediatric supratentorial HGG propose an important role of the cell-of-origin for clinicopathologic features of this disease. Cancer Res; 77(3); 802-12. ©2016 AACR., (©2016 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
33. Clonal Variation in Drug and Radiation Response among Glioma-Initiating Cells Is Linked to Proneural-Mesenchymal Transition.
- Author
-
Segerman A, Niklasson M, Haglund C, Bergström T, Jarvius M, Xie Y, Westermark A, Sönmez D, Hermansson A, Kastemar M, Naimaie-Ali Z, Nyberg F, Berglund M, Sundström M, Hesselager G, Uhrbom L, Gustafsson M, Larsson R, Fryknäs M, Segerman B, and Westermark B
- Subjects
- Cell Line, Tumor, DNA Methylation drug effects, DNA Methylation radiation effects, Epithelial-Mesenchymal Transition drug effects, Epithelial-Mesenchymal Transition radiation effects, Gene Expression Regulation, Neoplastic drug effects, Gene Expression Regulation, Neoplastic radiation effects, Glioblastoma drug therapy, Glioblastoma pathology, Glioblastoma radiotherapy, Humans, Neoplastic Stem Cells pathology, Promoter Regions, Genetic, DNA Methylation genetics, Glioblastoma genetics, Neoplasm Proteins genetics, Neoplastic Stem Cells metabolism
- Abstract
Intratumoral heterogeneity is a hallmark of glioblastoma multiforme and thought to negatively affect treatment efficacy. Here, we establish libraries of glioma-initiating cell (GIC) clones from patient samples and find extensive molecular and phenotypic variability among clones, including a range of responses to radiation and drugs. This widespread variability was observed as a continuum of multitherapy resistance phenotypes linked to a proneural-mesenchymal shift in the transcriptome. Multitherapy resistance was associated with a semi-stable cell state that was characterized by an altered DNA methylation pattern at promoter regions of mesenchymal master regulators and enhancers. The gradient of cell states within the GIC compartment constitutes a distinct form of heterogeneity. Our findings may open an avenue toward the development of new therapeutic rationales designed to reverse resistant cell states., (Copyright © 2016 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
34. Pleiotrophin enhances PDGFB-induced gliomagenesis through increased proliferation of neural progenitor cells.
- Author
-
Zhang L, Laaniste L, Jiang Y, Alafuzoff I, Uhrbom L, and Dimberg A
- Subjects
- Animals, Avian Proteins genetics, Avian Sarcoma Viruses genetics, Brain Neoplasms genetics, Brain Neoplasms pathology, Carrier Proteins genetics, Cells, Cultured, Chromosomes, Human, Pair 7, Cyclin-Dependent Kinase Inhibitor p16 genetics, Cytokines genetics, Gene Amplification, Gene Expression Regulation, Neoplastic, Genetic Predisposition to Disease, Glioblastoma genetics, Glioblastoma pathology, Humans, Mice, Transgenic, Neoplasm Grading, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, Neural Stem Cells metabolism, Neural Stem Cells pathology, Phenotype, Proto-Oncogene Proteins c-akt metabolism, Receptors, Virus genetics, Signal Transduction drug effects, Spheroids, Cellular, Transfection, Brain Neoplasms metabolism, Carrier Proteins metabolism, Cell Proliferation drug effects, Cytokines metabolism, Glioblastoma metabolism, Neoplastic Stem Cells drug effects, Neural Stem Cells drug effects, Proto-Oncogene Proteins c-sis pharmacology
- Abstract
Pleiotrophin (PTN) augments tumor growth by increasing proliferation of tumor cells and promoting vascular abnormalization, but its role in early gliomagenesis has not been evaluated. Through analysis of publically available datasets, we demonstrate that increased PTN mRNA expression is associated with amplification of chromosome 7, identified as one of the earliest steps in glioblastoma development. To elucidate the role of PTN in tumor initiation we employed the RCAS/tv-a model that allows glioma induction by RCAS-virus mediated expression of oncogenes in neural progenitor cells. Intracranial injection of RCAS-PTN did not induce glioma formation when administrated alone, but significantly enhanced RCAS-platelet derived growth factor (PDGF)B-induced gliomagenesis. PTN co-treatment augmented PDGFB-induced Akt activation in neural progenitor cells in vitro, and enhanced neural sphere size associated with increased proliferation. Our data indicates that PTN expression is associated with chromosome 7 gain, and that PTN enhances PDGFB-induced gliomagenesis by stimulating proliferation of neural progenitor cells.
- Published
- 2016
- Full Text
- View/download PDF
35. Case-specific potentiation of glioblastoma drugs by pterostilbene.
- Author
-
Schmidt L, Baskaran S, Johansson P, Padhan N, Matuszewski D, Green LC, Elfineh L, Wee S, Häggblad M, Martens U, Westermark B, Forsberg-Nilsson K, Uhrbom L, Claesson-Welsh L, Andäng M, Sintorn IM, Lundgren B, Lönnstedt I, Krona C, and Nelander S
- Subjects
- Aged, Aged, 80 and over, Antineoplastic Agents, Phytogenic therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Brain Neoplasms drug therapy, Brain Neoplasms pathology, Cell Cycle drug effects, Cell Line, Tumor, Cell Proliferation drug effects, DNA Copy Number Variations, Drug Synergism, Female, Gefitinib, Gene Expression Profiling, Gene Knockdown Techniques, Glioblastoma drug therapy, Glioblastoma pathology, Humans, MAP Kinase Signaling System drug effects, Male, Middle Aged, Mutation, Phenotype, Protein Kinase Inhibitors pharmacology, Quinazolines pharmacology, Stilbenes therapeutic use, Transcriptome, Antineoplastic Agents, Phytogenic pharmacology, Stilbenes pharmacology
- Abstract
Glioblastoma multiforme (GBM, astrocytoma grade IV) is the most common malignant primary brain tumor in adults. Addressing the shortage of effective treatment options for this cancer, we explored repurposing of existing drugs into combinations with potent activity against GBM cells. We report that the phytoalexin pterostilbene is a potentiator of two drugs with previously reported anti-GBM activity, the EGFR inhibitor gefitinib and the antidepressant sertraline. Combinations of either of these two compounds with pterostilbene suppress cell growth, viability, sphere formation and inhibit migration in tumor GBM cell (GC) cultures. The potentiating effect of pterostilbene was observed to a varying degree across a panel of 41 patient-derived GCs, and correlated in a case specific manner with the presence of missense mutation of EGFR and PIK3CA and a focal deletion of the chromosomal region 1p32. We identify pterostilbene-induced cell cycle arrest, synergistic inhibition of MAPK activity and induction of Thioredoxin interacting protein (TXNIP) as possible mechanisms behind pterostilbene's effect. Our results highlight a nontoxic stilbenoid compound as a modulator of anticancer drug response, and indicate that pterostilbene might be used to modulate two anticancer compounds in well-defined sets of GBM patients.
- Published
- 2016
- Full Text
- View/download PDF
36. ABCG2 regulates self-renewal and stem cell marker expression but not tumorigenicity or radiation resistance of glioma cells.
- Author
-
Wee B, Pietras A, Ozawa T, Bazzoli E, Podlaha O, Antczak C, Westermark B, Nelander S, Uhrbom L, Forsberg-Nilsson K, Djaballah H, Michor F, and Holland EC
- Subjects
- Animals, Biomarkers, Tumor metabolism, Brain Neoplasms genetics, Brain Neoplasms metabolism, Brain Neoplasms radiotherapy, Cell Line, Tumor, Gene Expression Regulation, Neoplastic radiation effects, Glioma genetics, Glioma metabolism, Glioma radiotherapy, Humans, Mice, Neoplasm Transplantation, Neoplastic Stem Cells radiation effects, Radiation Tolerance, Receptors, Notch metabolism, Signal Transduction radiation effects, Up-Regulation, ATP Binding Cassette Transporter, Subfamily G, Member 2 metabolism, Biomarkers, Tumor genetics, Brain Neoplasms pathology, Glioma pathology, Neoplasm Proteins metabolism, Neoplastic Stem Cells metabolism
- Abstract
Glioma cells with stem cell traits are thought to be responsible for tumor maintenance and therapeutic failure. Such cells can be enriched based on their inherent drug efflux capability mediated by the ABC transporter ABCG2 using the side population assay, and their characteristics include increased self-renewal, high stem cell marker expression and high tumorigenic capacity in vivo. Here, we show that ABCG2 can actively drive expression of stem cell markers and self-renewal in glioma cells. Stem cell markers and self-renewal was enriched in cells with high ABCG2 activity, and could be specifically inhibited by pharmacological and genetic ABCG2 inhibition. Importantly, despite regulating these key characteristics of stem-like tumor cells, ABCG2 activity did not affect radiation resistance or tumorigenicity in vivo. ABCG2 effects were Notch-independent and mediated by diverse mechanisms including the transcription factor Mef. Our data demonstrate that characteristics of tumor stem cells are separable, and highlight ABCG2 as a potential driver of glioma stemness.
- Published
- 2016
- Full Text
- View/download PDF
37. Glioma-derived plasminogen activator inhibitor-1 (PAI-1) regulates the recruitment of LRP1 positive mast cells.
- Author
-
Roy A, Coum A, Marinescu VD, Põlajeva J, Smits A, Nelander S, Uhrbom L, Westermark B, Forsberg-Nilsson K, Pontén F, and Tchougounova E
- Subjects
- Brain Neoplasms mortality, Cell Line, Tumor, Cell Movement drug effects, Cell Proliferation, Glioblastoma mortality, Humans, Low Density Lipoprotein Receptor-Related Protein-1 antagonists & inhibitors, Phosphorylation genetics, STAT3 Transcription Factor metabolism, Tumor Microenvironment physiology, Brain Neoplasms pathology, Glioblastoma pathology, Low Density Lipoprotein Receptor-Related Protein-1 metabolism, Mast Cells metabolism, Neoplasm Invasiveness pathology, Plasminogen Activator Inhibitor 1 metabolism
- Abstract
Glioblastoma (GBM) is a high-grade glioma with a complex microenvironment, including various inflammatory cells and mast cells (MCs) as one of them. Previously we had identified glioma grade-dependent MC recruitment. In the present study we investigated the role of plasminogen activator inhibitor 1 (PAI-1) in MC recruitment.PAI-1, a primary regulator in the fibrinolytic cascade is capable of forming a complex with fibrinolytic system proteins together with low-density lipoprotein receptor-related protein 1 (LRP1). We found that neutralizing PAI-1 attenuated infiltration of MCs. To address the potential implication of LRP1 in this process, we used a LRP1 antagonist, receptor-associated protein (RAP), and demonstrated the attenuation of MC migration. Moreover, a positive correlation between the number of MCs and the level of PAI-1 in a large cohort of human glioma samples was observed. Our study demonstrated the expression of LRP1 in human MC line LAD2 and in MCs in human high-grade glioma. The activation of potential PAI-1/LRP1 axis with purified PAI-1 promoted increased phosphorylation of STAT3 and subsequently exocytosis in MCs.These findings indicate the influence of the PAI-1/LRP1 axis on the recruitment of MCs in glioma. The connection between high-grade glioma and MC infiltration could contribute to patient tailored therapy and improve patient stratification in future therapeutic trials.
- Published
- 2015
- Full Text
- View/download PDF
38. The Human Glioblastoma Cell Culture Resource: Validated Cell Models Representing All Molecular Subtypes.
- Author
-
Xie Y, Bergström T, Jiang Y, Johansson P, Marinescu VD, Lindberg N, Segerman A, Wicher G, Niklasson M, Baskaran S, Sreedharan S, Everlien I, Kastemar M, Hermansson A, Elfineh L, Libard S, Holland EC, Hesselager G, Alafuzoff I, Westermark B, Nelander S, Forsberg-Nilsson K, and Uhrbom L
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Biomarkers, Tumor, Cell Line, Tumor, Cell Proliferation, Cell Transformation, Neoplastic, Cluster Analysis, DNA Copy Number Variations, Disease Models, Animal, Gene Expression Profiling, Genomic Instability, Glioblastoma genetics, Glioblastoma mortality, Glioblastoma surgery, Heterografts, Humans, Kaplan-Meier Estimate, Mice, Middle Aged, Neoplasm Grading, Neoplasm Staging, Prognosis, Tumor Cells, Cultured, Young Adult, Biological Specimen Banks, Glioblastoma pathology
- Abstract
Glioblastoma (GBM) is the most frequent and malignant form of primary brain tumor. GBM is essentially incurable and its resistance to therapy is attributed to a subpopulation of cells called glioma stem cells (GSCs). To meet the present shortage of relevant GBM cell (GC) lines we developed a library of annotated and validated cell lines derived from surgical samples of GBM patients, maintained under conditions to preserve GSC characteristics. This collection, which we call the Human Glioblastoma Cell Culture (HGCC) resource, consists of a biobank of 48 GC lines and an associated database containing high-resolution molecular data. We demonstrate that the HGCC lines are tumorigenic, harbor genomic lesions characteristic of GBMs, and represent all four transcriptional subtypes. The HGCC panel provides an open resource for in vitro and in vivo modeling of a large part of GBM diversity useful to both basic and translational GBM research.
- Published
- 2015
- Full Text
- View/download PDF
39. Etomidate, propofol and diazepam potentiate GABA-evoked GABAA currents in a cell line derived from human glioblastoma.
- Author
-
Babateen O, Jin Z, Bhandage A, Korol SV, Westermark B, Forsberg Nilsson K, Uhrbom L, Smits A, and Birnir B
- Subjects
- Cell Line, Tumor, Drug Synergism, Etomidate pharmacology, Gene Expression Regulation, Neoplastic drug effects, Humans, Ion Channel Gating drug effects, Propofol pharmacology, Protein Subunits genetics, Protein Subunits metabolism, Protein Transport drug effects, Receptors, GABA-A genetics, Anesthetics, General pharmacology, Diazepam pharmacology, Electrophysiological Phenomena drug effects, Glioblastoma pathology, Isoxazoles pharmacology, Receptors, GABA-A metabolism, gamma-Aminobutyric Acid pharmacology
- Abstract
GABAA receptors are pentameric chloride ion channels that are opened by GABA. We have screened a cell line derived from human glioblastoma, U3047MG, for expression of GABAA receptor subunit isoforms and formation of functional ion channels. We identified GABAA receptors subunit α2, α3, α5, β1, β2, β3, δ, γ3, π, and θ mRNAs in the U3047MG cell line. Whole-cell GABA-activated currents were recorded and the half-maximal concentration (EC₅₀) for the GABA-activated current was 36 μM. The currents were activated by THIP (4,5,6,7-tetrahydroisoxazolo[5,4-c]pyridin-3-ol) and enhanced by the benzodiazepine diazepam (1 μM) and the general anesthetics etomidate and propofol (50 μM). In line with the expressed GABAA receptors containing at least the α3β3θ subunits, the receptors were highly sensitive to etomidate (EC₅₀=55 nM). Immunocytochemistry identified expression of the α3 and β3 subunit proteins. Our results show that the GABAA receptors in the glial cell line are functional and are modulated by classical GABAA receptor drugs. We propose that the U3047MG cell line may be used as a model system to study GABAA receptors function and pharmacology in glial cells., (Copyright © 2014 Elsevier B.V. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
40. Selective calcium sensitivity in immature glioma cancer stem cells.
- Author
-
Wee S, Niklasson M, Marinescu VD, Segerman A, Schmidt L, Hermansson A, Dirks P, Forsberg-Nilsson K, Westermark B, Uhrbom L, Linnarsson S, Nelander S, and Andäng M
- Subjects
- Blotting, Western, Brain Neoplasms genetics, Brain Neoplasms pathology, Cell Differentiation drug effects, Cell Proliferation drug effects, Fluorescent Antibody Technique, Gene Expression Profiling, Glioma genetics, Glioma pathology, Humans, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, Neural Stem Cells metabolism, Neural Stem Cells pathology, Oligonucleotide Array Sequence Analysis, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Signal Transduction drug effects, Tumor Cells, Cultured, Biomarkers, Tumor genetics, Brain Neoplasms drug therapy, Calcium pharmacology, Gene Expression Regulation, Neoplastic drug effects, Glioma drug therapy, Neoplastic Stem Cells drug effects, Neural Stem Cells drug effects
- Abstract
Tumor-initiating cells are a subpopulation in aggressive cancers that exhibit traits shared with stem cells, including the ability to self-renew and differentiate, commonly referred to as stemness. In addition, such cells are resistant to chemo- and radiation therapy posing a therapeutic challenge. To uncover stemness-associated functions in glioma-initiating cells (GICs), transcriptome profiles were compared to neural stem cells (NSCs) and gene ontology analysis identified an enrichment of Ca2+ signaling genes in NSCs and the more stem-like (NSC-proximal) GICs. Functional analysis in a set of different GIC lines regarding sensitivity to disturbed homeostasis using A23187 and Thapsigargin, revealed that NSC-proximal GICs were more sensitive, corroborating the transcriptome data. Furthermore, Ca2+ drug sensitivity was reduced in GICs after differentiation, with most potent effect in the NSC-proximal GIC, supporting a stemness-associated Ca2+ sensitivity. NSCs and the NSC-proximal GIC line expressed a larger number of ion channels permeable to potassium, sodium and Ca2+. Conversely, a higher number of and higher expression levels of Ca2+ binding genes that may buffer Ca2+, were expressed in NSC-distal GICs. In particular, expression of the AMPA glutamate receptor subunit GRIA1, was found to associate with Ca2+ sensitive NSC-proximal GICs, and decreased as GICs differentiated along with reduced Ca2+ drug sensitivity. The correlation between high expression of Ca2+ channels (such as GRIA1) and sensitivity to Ca2+ drugs was confirmed in an additional nine novel GIC lines. Calcium drug sensitivity also correlated with expression of the NSC markers nestin (NES) and FABP7 (BLBP, brain lipid-binding protein) in this extended analysis. In summary, NSC-associated NES+/FABP7+/GRIA1+ GICs were selectively sensitive to disturbances in Ca2+ homeostasis, providing a potential target mechanism for eradication of an immature population of malignant cells.
- Published
- 2014
- Full Text
- View/download PDF
41. Oncogenic signaling is dominant to cell of origin and dictates astrocytic or oligodendroglial tumor development from oligodendrocyte precursor cells.
- Author
-
Lindberg N, Jiang Y, Xie Y, Bolouri H, Kastemar M, Olofsson T, Holland EC, and Uhrbom L
- Subjects
- Animals, Astrocytoma metabolism, Brain Neoplasms metabolism, Cell Lineage, Mice, Mice, Transgenic, Neural Stem Cells metabolism, Oligodendroglia metabolism, Oligodendroglioma metabolism, Proto-Oncogene Proteins p21(ras) metabolism, Vimentin metabolism, Astrocytoma pathology, Brain Neoplasms pathology, Neural Stem Cells pathology, Oligodendroglia pathology, Oligodendroglioma pathology
- Abstract
Stem cells, believed to be the cellular origin of glioma, are able to generate gliomas, according to experimental studies. Here we investigated the potential and circumstances of more differentiated cells to generate glioma development. We and others have shown that oligodendrocyte precursor cells (OPCs) can also be the cell of origin for experimental oligodendroglial tumors. However, the question of whether OPCs have the capacity to initiate astrocytic gliomas remains unanswered. Astrocytic and oligodendroglial tumors represent the two most common groups of glioma and have been considered as distinct disease groups with putatively different origins. Here we show that mouse OPCs can give rise to both types of glioma given the right circumstances. We analyzed tumors induced by K-RAS and AKT and compared them to oligodendroglial platelet-derived growth factor B-induced tumors in Ctv-a mice with targeted deletions of Cdkn2a (p16(Ink4a-/-), p19(Arf-/-), Cdkn2a(-/-)). Our results showed that glioma can originate from OPCs through overexpression of K-RAS and AKT when combined with p19(Arf) loss, and these tumors displayed an astrocytic histology and high expression of astrocytic markers. We argue that OPCs have the potential to develop both astrocytic and oligodendroglial tumors given loss of p19(Arf), and that oncogenic signaling is dominant to cell of origin in determining glioma phenotype. Our mouse data are supported by the fact that human astrocytoma and oligodendroglioma display a high degree of overlap in global gene expression with no clear distinctions between the two diagnoses., (Copyright © 2014 the authors 0270-6474/14/3314644-08$15.00/0.)
- Published
- 2014
- Full Text
- View/download PDF
42. RETRACTED: Vulnerability of glioblastoma cells to catastrophic vacuolization and death induced by a small molecule.
- Author
-
Kitambi SS, Toledo EM, Usoskin D, Wee S, Harisankar A, Svensson R, Sigmundsson K, Kalderén C, Niklasson M, Kundu S, Aranda S, Westermark B, Uhrbom L, Andäng M, Damberg P, Nelander S, Arenas E, Artursson P, Walfridsson J, Forsberg Nilsson K, Hammarström LGJ, and Ernfors P
- Subjects
- Animals, Cell Death drug effects, Heterografts, Humans, Hydroxyquinolines pharmacology, MAP Kinase Kinase 4 metabolism, Mice, Neoplasm Transplantation, Pinocytosis drug effects, Vacuoles metabolism, Zebrafish, Brain Neoplasms drug therapy, Brain Neoplasms pathology, Glioblastoma drug therapy, Glioblastoma pathology, Piperidines pharmacology, Quinolines pharmacology, Small Molecule Libraries pharmacology
- Abstract
Glioblastoma multiforme (GBM) is the most aggressive form of brain cancer with marginal life expectancy. Based on the assumption that GBM cells gain functions not necessarily involved in the cancerous process, patient-derived glioblastoma cells (GCs) were screened to identify cellular processes amenable for development of targeted treatments. The quinine-derivative NSC13316 reliably and selectively compromised viability. Synthetic chemical expansion reveals delicate structure-activity relationship and analogs with increased potency, termed Vacquinols. Vacquinols stimulate death by membrane ruffling, cell rounding, massive macropinocytic vacuole accumulation, ATP depletion, and cytoplasmic membrane rupture of GCs. The MAP kinase MKK4, identified by a shRNA screen, represents a critical signaling node. Vacquinol-1 displays excellent in vivo pharmacokinetics and brain exposure, attenuates disease progression, and prolongs survival in a GBM animal model. These results identify a vulnerability to massive vacuolization that can be targeted by small molecules and point to the possible exploitation of this process in the design of anticancer therapies., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
43. Adenovirus serotype 5 vectors with Tat-PTD modified hexon and serotype 35 fiber show greatly enhanced transduction capacity of primary cell cultures.
- Author
-
Yu D, Jin C, Ramachandran M, Xu J, Nilsson B, Korsgren O, Le Blanc K, Uhrbom L, Forsberg-Nilsson K, Westermark B, Adamson R, Maitland N, Fan X, and Essand M
- Subjects
- Adenoviridae immunology, Animals, Cell Line, Gene Order, Genetic Vectors administration & dosage, Genetic Vectors pharmacokinetics, Humans, Mice, Primary Cell Culture, T-Lymphocytes immunology, Tissue Distribution, Adenoviridae genetics, Capsid Proteins genetics, Genetic Vectors genetics, Transduction, Genetic, tat Gene Products, Human Immunodeficiency Virus genetics
- Abstract
Recombinant adenovirus serotype 5 (Ad5) vectors represent one of the most efficient gene delivery vectors in life sciences. However, Ad5 is dependent on expression of the coxsackievirus-adenovirus-receptor (CAR) on the surface of target cell for efficient transduction, which limits it's utility for certain cell types. Herein we present a new vector, Ad5PTDf35, which is an Ad5 vector having serotype 35 fiber-specificity and Tat-PTD hexon-modification. This vector shows dramatically increased transduction capacity of primary human cell cultures including T cells, monocytes, macrophages, dendritic cells, pancreatic islets and exocrine cells, mesenchymal stem cells and tumor initiating cells. Biodistribution in mice following systemic administration (tail-vein injection) show significantly reduced uptake in the liver and spleen of Ad5PTDf35 compared to unmodified Ad5. Therefore, replication-competent viruses with these modifications may be further developed as oncolytic agents for cancer therapy. User-friendly backbone plasmids containing these modifications were developed for compatibility to the AdEasy-system to facilitate the development of surface-modified adenoviruses for gene delivery to difficult-to-transduce cells in basic, pre-clinical and clinical research.
- Published
- 2013
- Full Text
- View/download PDF
44. On the origin of glioma.
- Author
-
Jiang Y and Uhrbom L
- Subjects
- Animals, Astrocytes pathology, Brain Neoplasms genetics, Cell Lineage, Disease Models, Animal, Glioma genetics, Humans, Mice, Mutation, Brain Neoplasms pathology, Glioma pathology
- Abstract
Glioma is the most frequent primary brain tumor of adults that has a presumably glial origin. Although our knowledge regarding molecular mechanisms and signaling pathways involved in gliomagenesis has increased immensely during the past decade, high-grade glioma remains a lethal disease with dismal prognosis. The failure of current therapies has to a large extent been ascribed the functional heterogeneity of glioma cells. One reason for this heterogeneity is most certainly the large number of variations in genetic alterations that can be found in high-grade gliomas. Another factor that may influence glioma heterogeneity could be the cell type from which the glioma is initiated. The cell of origin for glioma is still undefined, and additional knowledge about this issue may prove critical for a more complete understanding of glioma biology. Based on information from patients, developmental biology, and experimental glioma models, the most putative target cells include astrocytes, neural stem cells, and oligodendrocyte precursor cells, which are all discussed in more detail in this article. Animal modeling of glioma suggests that these three cell types have the capability to be the origin of glioma, and we have reason to believe that, depending on the initiating cell type, prognosis and response to therapy may be significantly different. Thus, it is essential to explore further the role of cellular origin in glioma.
- Published
- 2012
- Full Text
- View/download PDF
45. RAD51 can inhibit PDGF-B-induced gliomagenesis and genomic instability.
- Author
-
Westermark UK, Lindberg N, Roswall P, Bråsäter D, Helgadottir HR, Hede SM, Zetterberg A, Jasin M, Nistér M, and Uhrbom L
- Subjects
- ADP-Ribosylation Factor 1 physiology, Animals, Blotting, Western, Brain cytology, Brain metabolism, Brain Neoplasms genetics, Brain Neoplasms pathology, Cell Differentiation, Cell Proliferation, Cells, Cultured, Chickens, DNA Damage, DNA Repair, DNA Replication, Fibroblasts cytology, Fibroblasts metabolism, Flow Cytometry, Glioma genetics, Glioma pathology, Green Fluorescent Proteins genetics, Homologous Recombination, Humans, Hyperplasia genetics, Hyperplasia pathology, Immunoenzyme Techniques, Mice, Mice, Knockout, Mice, Transgenic, Proto-Oncogene Proteins c-sis genetics, Rad51 Recombinase genetics, Stem Cells metabolism, Brain Neoplasms prevention & control, Genomic Instability, Glioma prevention & control, Hyperplasia prevention & control, Proto-Oncogene Proteins c-sis metabolism, Rad51 Recombinase metabolism
- Abstract
Faithful replication and DNA repair are vital for maintenance of genome integrity. RAD51 is a central protein in homologous recombination repair and during replication, when it protects and restarts stalled replication forks. Aberrant RAD51 expression occurs in glioma, and high expression has been shown to correlate with prolonged survival. Furthermore, genes involved in DNA damage response (DDR) are mutated or deleted in human glioblastomas, corroborating the importance of proper DNA repair to suppress gliomagenesis. We have analyzed DDR and genomic instability in PDGF-B-induced gliomas and investigated the role of RAD51 in glioma development. We show that PDGF-B-induced gliomas display genomic instability and that co-expression of RAD51 can suppress PDGF-B-induced tumorigenesis and prolong survival. Expression of RAD51 inhibited proliferation and genomic instability of tumor cells independent of Arf status. Our results demonstrate that the RAD51 pathway can prevent glioma initiation and maintain genome integrity of induced tumors, suggesting reactivation of the RAD51 pathway as a potential therapeutic avenue.
- Published
- 2011
- Full Text
- View/download PDF
46. CGGBP1 regulates cell cycle in cancer cells.
- Author
-
Singh U, Roswall P, Uhrbom L, and Westermark B
- Subjects
- Animals, Cell Cycle Proteins genetics, Cell Line, Tumor, Cyclin-Dependent Kinase Inhibitor p16 genetics, Cyclin-Dependent Kinase Inhibitor p21 genetics, DNA-Binding Proteins genetics, G1 Phase, GPI-Linked Proteins genetics, Humans, Mice, Neoplasms genetics, Promoter Regions, Genetic, RNA Interference, RNA, Small Interfering genetics, Resting Phase, Cell Cycle, S Phase, Tumor Cells, Cultured, Tumor Suppressor Protein p53 genetics, Cell Cycle, DNA-Binding Proteins metabolism, Gene Expression Regulation, Neoplastic, Neoplasms metabolism, Neoplasms pathology
- Abstract
Background: CGGBP1 is a CGG-triplet repeat binding protein, which affects transcription from CGG-triplet-rich promoters such as the FMR1 gene and the ribosomal RNA gene clusters. Earlier, we reported some previously unknown functions of CGGBP1 in gene expression during heat shock stress response. Recently we had found CGGBP1 to be a cell cycle regulatory midbody protein required for normal cytokinetic abscission in normal human fibroblasts, which have all the cell cycle regulatory mechanisms intact., Results: In this study we explored the role of CGGBP1 in the cell cycle in various cancer cell lines. CGGBP1 depletion by RNA interference in tumor-derived cells caused an increase in the cell population at G0/G1 phase and reduced the number of cells in the S phase. CGGBP1 depletion also increased the expression of cell cycle regulatory genes CDKN1A and GAS1, associated with reductions in histone H3 lysine 9 trimethylation in their promoters. By combining RNA interference and genetic mutations, we found that the role of CGGBP1 in cell cycle involves multiple mechanisms, as single deficiencies of CDKN1A, GAS1 as well as TP53, INK4A or ARF failed to rescue the G0/G1 arrest caused by CGGBP1 depletion., Conclusions: Our results show that CGGBP1 expression is important for cell cycle progression through multiple parallel mechanisms including the regulation of CDKN1A and GAS1 levels.
- Published
- 2011
- Full Text
- View/download PDF
47. Forced expression of Sox21 inhibits Sox2 and induces apoptosis in human glioma cells.
- Author
-
Ferletta M, Caglayan D, Mokvist L, Jiang Y, Kastemar M, Uhrbom L, and Westermark B
- Subjects
- Apoptosis, Base Sequence, Blotting, Western, Brain Neoplasms pathology, Cell Line, Tumor, Cell Proliferation, DNA Primers, Fluorescent Antibody Technique, Glial Fibrillary Acidic Protein genetics, Glioma pathology, Humans, Immunohistochemistry, Polymerase Chain Reaction, Brain Neoplasms genetics, Gene Expression Regulation, Neoplastic, Glioma genetics, SOXB1 Transcription Factors genetics, SOXB2 Transcription Factors genetics
- Abstract
Numerous studies support a role for Sox2 to keep stem cells and progenitor cells in an immature and proliferative state. Coexpression of Sox2 and GFAP has been found in regions of the adult brain where neural stem cells are present and in human glioma cells. In our study, we have investigated the roles of Sox2 and its counteracting partner Sox21 in human glioma cells. We show for the first time that Sox21 is expressed in both primary glioblastoma and in human glioma cell lines. We found that coexpression of Sox2, GFAP and Sox21 was mutually exclusive with expression of fibronectin. Our result suggests that glioma consists of at least two different cell populations: Sox2(+) /GFAP(+) /Sox21(+) /FN(-) and Sox2(-) /GFAP(-) /Sox21(-) /FN(+) . Reduction of Sox2 expression by using siRNA against Sox2 or by overexpressing Sox21 using a tetracycline-regulated expression system (Tet-on) caused decreased GFAP expression and a reduction in cell number due to induction of apoptosis. We suggest that Sox21 can negatively regulate Sox2 in glioma. Our findings imply that Sox2 and Sox21 may be interesting targets for the development of novel glioma therapy., (Copyright © 2010 UICC.)
- Published
- 2011
- Full Text
- View/download PDF
48. PDGF-B Can sustain self-renewal and tumorigenicity of experimental glioma-derived cancer-initiating cells by preventing oligodendrocyte differentiation.
- Author
-
Jiang Y, Boije M, Westermark B, and Uhrbom L
- Subjects
- Animals, Animals, Newborn, Apoptosis drug effects, Cell Proliferation drug effects, Cells, Cultured, Glial Fibrillary Acidic Protein, Glioma genetics, Glioma pathology, Immunohistochemistry, Mice, Mice, Transgenic, Mitogens pharmacology, Neoplasm Transplantation, Neoplastic Stem Cells pathology, Nerve Tissue Proteins metabolism, Oligodendroglia pathology, Proto-Oncogene Proteins c-sis genetics, Proto-Oncogene Proteins c-sis pharmacology, RNA Interference, Tubulin metabolism, Tumor Suppressor Protein p14ARF genetics, Tumor Suppressor Protein p14ARF metabolism, Cell Differentiation, Glioma metabolism, Neoplastic Stem Cells metabolism, Oligodendroglia metabolism, Proto-Oncogene Proteins c-sis metabolism
- Abstract
According to the cancer stem cell (CSC)/cancer-initiating cell hypothesis, glioma development is driven by a subpopulation of cells with unique tumor-regenerating capacity. We have characterized sphere-cultured glioma-derived cancer-initiating cells (GICs) from experimental gliomas induced by platelet-derived growth factor-B (PDGF-B) in neonatal Gtv-a Arf(-/-) mice. We found that the GICs can maintain their stem cell-like characteristics in absence of exogenous epidermal growth factor and fibroblast growth factor 2 and that this culture condition was highly selective for tumor-initiating cells where as few as five GICs could induce secondary tumor formation after orthotopic transplantation. Addition of FBS to the medium caused the GICs to differentiate into cells coexpressing glial fibrillary acidic protein and Tuj1, and this differentiation process was reversible, suggesting that the GICs are highly plastic and able to adapt to different environments without losing their tumorigenic properties. On inhibition of virally transduced PDGF-B by small interfering RNA treatment, the GICs stopped proliferating, lost their self-renewal ability, and started to uniformly express CNPase, a marker of oligodendrocyte precursor cells and mature oligodendrocytes. Most importantly, PDGF-B depletion completely abrogated the tumor-initiating capacity of the GICs. Our findings suggest that interfering with PDGF-controlled differentiation could be a therapeutic avenue for patients diagnosed with the PDGF-driven proneural subtype of human glioblastoma.
- Published
- 2011
- Full Text
- View/download PDF
49. Mast cell accumulation in glioblastoma with a potential role for stem cell factor and chemokine CXCL12.
- Author
-
Põlajeva J, Sjösten AM, Lager N, Kastemar M, Waern I, Alafuzoff I, Smits A, Westermark B, Pejler G, Uhrbom L, and Tchougounova E
- Subjects
- Adult, Aged, Animals, Blotting, Western, Brain Neoplasms immunology, Brain Neoplasms pathology, Cells, Cultured, Cyclin-Dependent Kinase Inhibitor p16 physiology, Female, Fluorescent Antibody Technique, Glioblastoma immunology, Glioblastoma pathology, Humans, Immunoenzyme Techniques, Male, Mast Cells metabolism, Mice, Mice, Knockout, Middle Aged, Proto-Oncogene Proteins c-akt metabolism, Proto-Oncogene Proteins p21(ras) metabolism, Brain Neoplasms metabolism, Chemokine CXCL12 metabolism, Glioblastoma metabolism, Mast Cells pathology, Receptors, CXCR4 metabolism, Stem Cell Factor metabolism
- Abstract
Glioblastoma multiforme (GBM) is the most common and malignant form of glioma with high mortality and no cure. Many human cancers maintain a complex inflammatory program triggering rapid recruitment of inflammatory cells, including mast cells (MCs), to the tumor site. However, the potential contribution of MCs in glioma has not been addressed previously. Here we report for the first time that MCs infiltrate KRas+Akt-induced gliomas, using the RCAS/TV-a system, where KRas and Akt are transduced by RCAS into the brains of neonatal Gtv-a- or Ntv-a transgenic mice lacking Ink4a or Arf. The most abundant MC infiltration was observed in high-grade gliomas of Arf-/- mice. MC accumulation could be localized to the vicinity of glioma-associated vessels but also within the tumor mass. Importantly, proliferating MCs were detected, suggesting that the MC accumulation was caused by local expansion of the MC population. In line with these findings, strong expression of stem cell factor (SCF), i.e. the main MC growth factor, was detected, in particular around tumor blood vessels. Further, glioma cells expressed the MC chemotaxin CXCL12 and MCs expressed the corresponding receptor, i.e. CXCR4, suggesting that MCs could be attracted to the tumor through the CXCL12/CXCR4 axis. Supporting a role for MCs in glioma, strong MC infiltration was detected in human glioma, where GBMs contained significantly higher MC numbers than grade II tumors did. Moreover, human GBMs were positive for CXCL12 and the infiltrating MCs were positive for CXCR4. In conclusion, we provide the first evidence for a role for MCs in glioma.
- Published
- 2011
- Full Text
- View/download PDF
50. Histidine-rich glycoprotein can prevent development of mouse experimental glioblastoma.
- Author
-
Kärrlander M, Lindberg N, Olofsson T, Kastemar M, Olsson AK, and Uhrbom L
- Subjects
- Animals, Cell Line, Cell Proliferation drug effects, Chickens, Genetic Vectors genetics, Glioblastoma blood supply, Glioblastoma chemically induced, Humans, Injections, Mice, Neovascularization, Pathologic pathology, Neuroglia cytology, Neuroglia drug effects, Neuroglia metabolism, Proto-Oncogene Proteins c-sis pharmacology, Transduction, Genetic, Histidine-Rich Glycoprotein, Glioblastoma pathology, Glioblastoma prevention & control, Proteins metabolism
- Abstract
Extensive angiogenesis, formation of new capillaries from pre-existing blood vessels, is an important feature of malignant glioma. Several antiangiogenic drugs targeting vascular endothelial growth factor (VEGF) or its receptors are currently in clinical trials as therapy for high-grade glioma and bevacizumab was recently approved by the FDA for treatment of recurrent glioblastoma. However, the modest efficacy of these drugs and emerging problems with anti-VEGF treatment resistance welcome the development of alternative antiangiogenic therapies. One potential candidate is histidine-rich glycoprotein (HRG), a plasma protein with antiangiogenic properties that can inhibit endothelial cell adhesion and migration. We have used the RCAS/TV-A mouse model for gliomas to investigate the effect of HRG on brain tumor development. Tumors were induced with platelet-derived growth factor-B (PDGF-B), in the presence or absence of HRG. We found that HRG had little effect on tumor incidence but could significantly inhibit the development of malignant glioma and completely prevent the occurrence of grade IV tumors (glioblastoma).
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.