Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Sara Bolin is active.

Publication


Featured researches published by Sara Bolin.


Clinical Cancer Research | 2014

BET Bromodomain Inhibition of MYC-Amplified Medulloblastoma

Pratiti Bandopadhayay; Guillaume Bergthold; Brian Nguyen; Simone Schubert; Sharareh Gholamin; Yujie Tang; Sara Bolin; Steven E. Schumacher; Rhamy Zeid; Sabran Masoud; Furong Yu; Nujsaubnusi Vue; William J. Gibson; Brenton R. Paolella; Siddhartha Mitra; Samuel H. Cheshier; Jun Qi; Kun-Wei Liu; Robert J. Wechsler-Reya; William A. Weiss; Fredrik J. Swartling; Mark W. Kieran; James E. Bradner; Rameen Beroukhim; Yoon-Jae Cho

Purpose: MYC-amplified medulloblastomas are highly lethal tumors. Bromodomain and extraterminal (BET) bromodomain inhibition has recently been shown to suppress MYC-associated transcriptional activity in other cancers. The compound JQ1 inhibits BET bromodomain-containing proteins, including BRD4. Here, we investigate BET bromodomain targeting for the treatment of MYC-amplified medulloblastoma. Experimental Design: We evaluated the effects of genetic and pharmacologic inhibition of BET bromodomains on proliferation, cell cycle, and apoptosis in established and newly generated patient- and genetically engineered mouse model (GEMM)-derived medulloblastoma cell lines and xenografts that harbored amplifications of MYC or MYCN. We also assessed the effect of JQ1 on MYC expression and global MYC-associated transcriptional activity. We assessed the in vivo efficacy of JQ1 in orthotopic xenografts established in immunocompromised mice. Results: Treatment of MYC-amplified medulloblastoma cells with JQ1 decreased cell viability associated with arrest at G1 and apoptosis. We observed downregulation of MYC expression and confirmed the inhibition of MYC-associated transcriptional targets. The exogenous expression of MYC from a retroviral promoter reduced the effect of JQ1 on cell viability, suggesting that attenuated levels of MYC contribute to the functional effects of JQ1. JQ1 significantly prolonged the survival of orthotopic xenograft models of MYC-amplified medulloblastoma (P < 0.001). Xenografts harvested from mice after five doses of JQ1 had reduced the expression of MYC mRNA and a reduced proliferative index. Conclusion: JQ1 suppresses MYC expression and MYC-associated transcriptional activity in medulloblastomas, resulting in an overall decrease in medulloblastoma cell viability. These preclinical findings highlight the promise of BET bromodomain inhibitors as novel agents for MYC-amplified medulloblastoma. Clin Cancer Res; 20(4); 912–25. ©2013 AACR.


Journal of Biological Chemistry | 2014

Mast Cell Chymase Degrades the Alarmins Heat Shock Protein 70, Biglycan, HMGB1, and Interleukin-33 (IL-33) and Limits Danger-induced Inflammation

Ananya Roy; Goutham Ganesh; Helena Sippola; Sara Bolin; Osama Sawesi; Anders Dagälv; Susan M. Schlenner; Thorsten B. Feyerabend; Hans Reimer Rodewald; Lena Kjellén; Lars Hellman; Magnus Åbrink

Background: Mast cell chymase may be both pro-inflammatory and anti-inflammatory during infection and tissue damage. Results: Human and mouse chymases modulate extracellular levels of the alarmins Hsp70, biglycan, HMGB1, and IL-33. Conclusion: Mast cell chymase degrades alarmins and may limit inflammation. Significance: Identifying the physiological chymase substrates is crucial for understanding the role of chymase in immune responses and could aid in drug development. During infection and tissue damage, virulence factors and alarmins are pro-inflammatory and induce activation of various immune cells including macrophages and mast cells (MCs). Activated MCs instantly release preformed inflammatory mediators, including several proteases. The chymase mouse mast cell protease (MCPT)-4 is thought to be pro-inflammatory, whereas human chymase also degrades pro-inflammatory cytokines, suggesting that chymase instead limits inflammation. Here we explored the contribution of MCPT4 and human chymase to the control of danger-induced inflammation. We found that protein extracts from wild type (WT), carboxypeptidase A3-, and MCPT6-deficient mice and MCs and recombinant human chymase efficiently degrade the Trichinella spiralis virulence factor heat shock protein 70 (Hsp70) as well as endogenous Hsp70. MC-(Wsash)-, serglycin-, NDST2-, and MCPT4-deficient extracts lacked this capacity, indicating that chymase is responsible for the degradation. Chymase, but not MC tryptase, also degraded other alarmins, i.e. biglycan, HMGB1, and IL-33, a degradation that was efficiently blocked by the chymase inhibitor chymostatin. IL-7, IL-22, GM-CSF, and CCL2 were resistant to chymase degradation. MCPT4-deficient conditions ex vivo and in vivo showed no reduction in added Hsp70 and only minor reduction of IL-33. Peritoneal challenge with Hsp70 resulted in increased neutrophil recruitment and TNF-α levels in the MCPT4-deficient mice, whereas IL-6 and CCL2 levels were similar to the levels found in WT mice. The rapid and MC chymase-specific degradation of virulence factors and alarmins may depend on the presence of accessible extended recognition cleavage sites in target substrates and suggests a protective and regulatory role of MC chymase during danger-induced inflammation.


The EMBO Journal | 2016

FBW7 suppression leads to SOX9 stabilization and increased malignancy in medulloblastoma

Aldwin Suryo Rahmanto; Vasil Savov; Andrä Brunner; Sara Bolin; Holger Weishaupt; Alena Malyukova; Gabriela Rosén; Matko Čančer; Sonja Hutter; Anders Sundström; Daisuke Kawauchi; David T. W. Jones; Charles Spruck; Michael D. Taylor; Yoon-Jae Cho; Stefan M. Pfister; Marcel Kool; Andrey Korshunov; Fredrik J. Swartling; Olle Sangfelt

SOX9 is a master transcription factor that regulates development and stem cell programs. However, its potential oncogenic activity and regulatory mechanisms that control SOX9 protein stability are poorly understood. Here, we show that SOX9 is a substrate of FBW7, a tumor suppressor, and a SCF (SKP1/CUL1/F‐box)‐type ubiquitin ligase. FBW7 recognizes a conserved degron surrounding threonine 236 (T236) in SOX9 that is phosphorylated by GSK3 kinase and consequently degraded by SCFFBW7α. Failure to degrade SOX9 promotes migration, metastasis, and treatment resistance in medulloblastoma, one of the most common childhood brain tumors. FBW7 is either mutated or downregulated in medulloblastoma, and in cases where FBW7 mRNA levels are low, SOX9 protein is significantly elevated and this phenotype is associated with metastasis at diagnosis and poor patient outcome. Transcriptional profiling of medulloblastoma cells expressing a degradation‐resistant SOX9 mutant reveals activation of pro‐metastatic genes and genes linked to cisplatin resistance. Finally, we show that pharmacological inhibition of PI3K/AKT/mTOR pathway activity destabilizes SOX9 in a GSK3/FBW7‐dependent manner, rendering medulloblastoma cells sensitive to cytostatic treatment.


Experimental Neurology | 2014

Signals that regulate the oncogenic fate of neural stem cells and progenitors

Fredrik J. Swartling; Sara Bolin; Joanna J. Phillips; Anders Persson

Brain tumors have frequently been associated with a neural stem cell (NSC) origin and contain stem-like tumor cells, so-called brain tumor stem cells (BTSCs) that share many features with normal NSCs. A stem cell state of BTSCs confers resistance to radiotherapy and treatment with alkylating agents. It is also a hallmark of aggressive brain tumors and is maintained by transcriptional networks that are also active in embryonic stem cells. Advances in reprogramming of somatic cells into induced pluripotent stem (iPS) cells have further identified genes that drive stemness. In this review, we will highlight the possible drivers of stemness in medulloblastoma and glioma, the most frequent types of primary malignant brain cancer in children and adults, respectively. Signals that drive expansion of developmentally defined neural precursor cells are also active in corresponding brain tumors. Transcriptomal subgroups of human medulloblastoma and glioma match features of NSCs but also more restricted progenitors. Lessons from genetically-engineered mouse (GEM) models show that temporally and regionally defined NSCs can give rise to distinct subgroups of medulloblastoma and glioma. We will further discuss how acquisition of stem cell features may drive brain tumorigenesis from a non-NSC origin. Genetic alterations, signaling pathways, and therapy-induced changes in the tumor microenvironment can drive reprogramming networks and induce stemness in brain tumors. Finally, we propose a model where dysregulation of microRNAs (miRNAs) that normally provide barriers against reprogramming plays an integral role in promoting stemness in brain tumors.


European Journal of Pharmaceutical Sciences | 2009

Hepatitis C virus NS3 protease inhibitors : large, flexible molecules of peptide origin show satisfactory permeability across Caco-2 cells

Christel A. S. Bergström; Sara Bolin; Per Artursson; Robert Rönn; Anja Sandström

The purpose of this study was to investigate the intestinal absorption of tripeptide-based compounds intended for treatment of hepatitis C virus (HCV) infection. The intestinal permeability of 11 HCV NS3 protease inhibitors (Mw 687-841, ClogD(pH 7.4) 1.2-7.3 and 10-13 hydrogen bond donors/acceptors) was measured using Caco-2 cells. Each compound was investigated in the apical to basolateral (a-b) and basolateral to apical (b-a) direction at pH 7.4. For compounds displaying efflux the experiment was repeated in the presence of 1 microM GF120918 to investigate possible involvement of P-glycoprotein (Pgp; ABCB1). All compounds displayed intermediate to high permeability. Seven of them showed extensive efflux, with 31-114-fold higher permeability in the b-a direction than the a-b direction. Addition of the Pgp inhibitor GF120918 reduced the b-a transport rate for the effluxed compounds. However, for inhibitors with a C-terminal carboxylic acid and the acidic bioisosteres thereof the efflux was still significant. Hence, the negative charge resulted in efflux by other ABC-transporters than Pgp. From this study it can be concluded that small changes in the overall structure can lead to a large variation in permeability and efflux as shown by the inhibitors herein, properties that also may influence the resulting inhibition potency of the compounds when performing cell-based pharmacological assays.


Genes | 2017

Modeling and Targeting MYC Genes in Childhood Brain Tumors

Sonja Hutter; Sara Bolin; Holger Weishaupt; Fredrik J. Swartling

Brain tumors are the second most common group of childhood cancers, accounting for about 20%–25% of all pediatric tumors. Deregulated expression of the MYC family of transcription factors, particularly c-MYC and MYCN genes, has been found in many of these neoplasms, and their expression levels are often correlated with poor prognosis. Elevated c-MYC/MYCN initiates and drives tumorigenesis in many in vivo model systems of pediatric brain tumors. Therefore, inhibition of their oncogenic function is an attractive therapeutic target. In this review, we explore the roles of MYC oncoproteins and their molecular targets during the formation, maintenance, and recurrence of childhood brain tumors. We also briefly summarize recent progress in the development of therapeutic approaches for pharmacological inhibition of MYC activity in these tumors.


Oncogene | 2018

Combined BET-bromodomain and CDK2 inhibition in MYC-driven medulloblastoma

Sara Bolin; Anna Borgenvik; Camilla U. Persson; Anders Sundström; Jun Qi; James E. Bradner; William A. Weiss; Yoon Jae Cho; Holger Weishaupt; Fredrik J. Swartling

Medulloblastoma (MB) is the most common malignant brain tumor in children. MYC genes are frequently amplified and correlate with poor prognosis in MB. BET bromodomains recognize acetylated lysine residues and often promote and maintain MYC transcription. Certain cyclin-dependent kinases (CDKs) are further known to support MYC stabilization in tumor cells. In this report, MB cells were suppressed by combined targeting of MYC expression and MYC stabilization using BET bromodomain inhibition and CDK2 inhibition, respectively. Such combination treatment worked synergistically and caused cell cycle arrest as well as massive apoptosis. Immediate transcriptional changes from this combined MYC blockade were found using RNA-Seq profiling and showed remarkable similarities to changes in MYC target gene expression when MYCN was turned off with doxycycline in our MYCN-inducible animal model for Group 3 MB. In addition, the combination treatment significantly prolonged survival as compared to single-agent therapy in orthotopically transplanted human Group 3 MB with MYC amplifications. Our data suggest that dual inhibition of CDK2 and BET bromodomains can be a novel treatment approach for suppressing MYC-driven cancer.


iScience | 2018

Microglia Induce Pdgfrb Expression In Glioma Cells To Enhance Their Migratory Capacity

Tatjana Wallmann; Xing-Mei Zhang; Majken Wallerius; Sara Bolin; Anne-Laure Joly; Caroline Sobocki; Lina Leiss; Yiwen Jiang; Jonas Bergh; Eric C. Holland; Per Øyvind Enger; John Andersson; Fredrik J. Swartling; Hrvoje Miletic; Lene Uhrbom; Robert A. Harris; Charlotte Rolny

Summary 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.


Cancer Research | 2016

Abstract 2473: Combined BET-bromodomain and CDK2 inhibition in MYC-driven medulloblastoma

Sara Bolin; Anna Borgenvik; Camilla Persson; Gabriela Rosén; Anders Sundström; Jun Qi; James E. Bradner; William A. Weiss; Yoon-Jae Cho; Holger Weishaupt; Fredrik J. Swartling

Misexpression of MYC genes (MYC and MYCN) occurs commonly in medulloblastoma (MB), the most frequent malignant childhood brain tumor. We previously showed that tumors are addicted to MYCN and that MYCN stabilization is required for MB development in mice (Swartling et al, Genes & Dev, 2010; Cancer Cell, 2012). Targeted MYCN suppression completely depleted MYCN-driven MB cells in vivo. Immediate transcriptional changes from such MYCN blockade were found by RNA-Seq and showed similarities to changes that occurred after CDK2 suppression or when inhibiting BET bromodomains. CDK2 and BET inhibitors both inhibited MYC protein expression and effectively induced cell cycle arrest or apoptosis. Compared with either agent alone a sustained combination treatment over 7-10 days displayed synergy and effectively abolished tumor cell proliferation in vitro. The combined treatment further reduced tumor growth in orthotopical MB transplants and significantly prolonged survival as compared to single agent therapy. Our data suggest that dual inhibition of CDK2 and BET Bromodomains could be a novel treatment approach in suppressing medulloblastoma by targeting MYC proteins. Citation Format: Sara Bolin, Anna Borgenvik, Camilla Persson, Gabriela Rosen, Anders Sundstrom, Jun Qi, James E. Bradner, William A. Weiss, Yoon-Jae Cho, Holger Weishaupt, Fredrik J. Swartling. Combined BET-bromodomain and CDK2 inhibition in MYC-driven medulloblastoma. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2473.


Cancer Research | 2016

Abstract 2688: A forward genetics screen of murine brain tumors identifies novel candidate genes involved in gliomagenesis

Matko Čančer; Holger Weishaupt; Gabriela Rosén; Ignas Bunikis; Yiwen Jiang; Smitha Sreedharan; Sara Bolin; Ulf Gyllensten; Oren J. Becher; Lene Uhrbom; Adam Ameur; Fredrik J. Swartling

Glioma is the most frequent malignant brain tumor in adults. Platelet-derived growth factor (PDGF) signaling is commonly activated in glioma. We have used a retrovirus-driven PDGFB-induced murine glioma model that causes tumors that closely resemble human gliomas of various grades. Knowing that retroviruses have a capacity to induce insertional mutagenesis, we have employed whole genome sequencing to identify potential genes that, together with PDGFB, drive glioma development. Gliomas were induced by RCAS virus injection into the brains of mice expressing the RCAS retroviral receptor from specific promoters. Genomic DNA from tumor cell lines was probed for retroviral tags and sequenced to identify genomic targets of the retrovirus. A streamlined analysis pipeline was developed for retrovirus integration detection and mapping to the reference mouse genome. Integration sites were analyzed and a common integration site (CIS) label was assigned to a gene, given that it was either tagged by a retrovirus more than once within a discovery set or found within the Retroviral Tagged Cancer Gene Database (RTCGD). In a small discovery subset of 15 murine gliomas, we have identified 40 CIS, of which 37 were validated by Sanger sequencing. When compared with previously identified CIS in RTCGD, 5.5% of them were shared with our older screen, where we overexpressed PDGFB from another retrovirus in order to induce glioma. Less CIS genes were shared with other published tumor models induced by viruses driven by other cancer genes/viruses. The majority of genes identified in our screen were tagged twice. However, Nfic, Cuecd1, Thra, Foxj1 and Nrxn1 were tagged three times, Ppfibp1 and Rhbg four times, and Mir29a/29b-1 seven times. As compared to control tumor lines, two top candidate genes, Mir29a and Ppfibp1, demonstrated significantly increased expression in tumor lines in were they were respectively tagged. Mir29a is often found downregulated in human tumors including gliomas, still high levels of Mir29a are sometimes found in certain aggressive cancers and in metastases. Interestingly, we found that specific PDGFR inhibition negatively regulates Mir29a, indicating a possible role for PDGF signaling in Mir29a regulation. Ppfibp1 has not been extensively studied in cancer. However, Ppfibp1 seems to have a subgroup-specific expression in human glioblastoma, making it an interesting candidate for further analysis. Here we present a new screening method that can be employed to identify genes involved in PDGFB-driven gliomagenesis. So far, we have identified 37 candidate genes by whole genome sequencing. Two of the most frequently tagged candidates, Mir29a and Ppfibp1 were upregulated as a consequence of retroviral mutagenesis. Their precise role in driving glioma formation in collaboration with PDGF is currently explored. Citation Format: Matko Cancer, Holger Weishaupt, Gabriela Rosen, Ignas Bunikis, Yiwen Jiang, Smitha Sreedharan, Sara Bolin, Ulf Gyllensten, Oren J. Becher, Lene Uhrbom, Adam Ameur, Fredrik J. Swartling. A forward genetics screen of murine brain tumors identifies novel candidate genes involved in gliomagenesis. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2688.

Collaboration


Dive into the Sara Bolin's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sonja Hutter

German Cancer Research Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge