SwePub
Sök i SwePub databas

  Extended search

Träfflista för sökning "WFRF:(Jarvius Malin) "

Search: WFRF:(Jarvius Malin)

  • Result 1-10 of 47
Sort/group result
   
EnumerationReferenceCoverFind
1.
  • Leuchowius, Karl-Johan, et al. (author)
  • High content screening for inhibitors of protein interactions and post-translational modifications in primary cells by proximity ligation
  • 2010
  • In: Molecular & Cellular Proteomics. - 1535-9476 .- 1535-9484. ; 9:1, s. 178-183
  • Journal article (peer-reviewed)abstract
    • The cost of developing new drugs is a major obstacle for pharmaceutical companies and academia with many drugs identified in the drug discovery process failing approval for clinical use due to lack of intended effect or because of severe side effects. Since the early 1990 s, high throughput screening of drug compounds has increased enormously in capacity but has not resulted in a higher success rate of the identified drugs. Thus, there is a need for methods that can identify biologically relevant compounds and more accurately predict in vivo effects early in the drug discovery process. To address this, we developed a proximity ligation-based assay for high content screening of drug effects on signaling pathways. As a proof of concept, we used the assay to screen through a library of previously identified kinase inhibitors, including six clinically used tyrosine kinase inhibitors, to identify compounds that inhibited the platelet-derived growth factor (PDGF) receptor beta signaling pathway in stimulated primary human fibroblasts. Thirteen of the 80 compounds were identified as hits, and the dose responses of these compounds were measured. The assay exhibited a very high Z' factor (0.71) and signal to noise ratio (11.7), demonstrating excellent ability to identify compounds interfering with the specific signaling event. A comparison with regular immunofluorescence detection of phosphorylated PDGF receptor demonstrated a far superior ability by the in situ proximity ligation assay to reveal inhibition of receptor phosphorylation. In addition, inhibitor-induced perturbation of protein-protein interactions of the PDGF signaling pathway could be quantified, further demonstrating the usefulness of the assay in drug discovery.
  •  
2.
  • Andersson, Claes, et al. (author)
  • Mebendazole is unique among tubulin-active drugs in activating the MEK-ERK pathway
  • 2020
  • In: Scientific Reports. - : Springer Science and Business Media LLC. - 2045-2322. ; 10:1
  • Journal article (peer-reviewed)abstract
    • We recently showed that the anti-helminthic compound mebendazole (MBZ) has immunomodulating activity in monocyte/macrophage models and induces ERK signalling. In the present study we investigated whether MBZ induced ERK activation is shared by other tubulin binding agents (TBAs) and if it is observable also in other human cell types. Curated gene signatures for a panel of TBAs in the LINCS Connectivity Map (CMap) database showed a unique strong negative correlation of MBZ with MEK/ERK inhibitors indicating ERK activation also in non-haematological cell lines. L1000 gene expression signatures for MBZ treated THP-1 monocytes also connected negatively to MEK inhibitors. MEK/ERK phosphoprotein activity testing of a number of TBAs showed that only MBZ increased the activity in both THP-1 monocytes and PMA differentiated macrophages. Distal effects on ERK phosphorylation of the substrate P90RSK and release of IL1B followed the same pattern. The effect of MBZ on MEK/ERK phosphorylation was inhibited by RAF/MEK/ERK inhibitors in THP-1 models, CD3/IL2 stimulated PBMCs and a MAPK reporter HEK-293 cell line. MBZ was also shown to increase ERK activity in CD4+ T-cells from lupus patients with known defective ERK signalling. Given these mechanistic features MBZ is suggested suitable for treatment of diseases characterized by defective ERK signalling, notably difficult to treat autoimmune diseases.
  •  
3.
  • Blom, Kristin, et al. (author)
  • Mebendazole-induced M1 polarisation of THP-1 macrophages may involve DYRK1B inhibition
  • 2019
  • In: BMC Research Notes. - : Springer Nature. - 1756-0500. ; 12:1
  • Journal article (peer-reviewed)abstract
    • Objective: We recently showed that the anti-helminthic compound mebendazole (MBZ) has immunomodulating activity by inducing a M2 to M1 phenotype switch in monocyte/macrophage models. In the present study we investigated the potential role of protein kinases in mediating this effect.Results: MBZ potently binds and inhibits Dual specificity tyrosine-phosphorylation-regulated kinase 1B (DYRK1B) with a Kd and an IC50 of 7 and 360 nM, respectively. The specific DYRK1B inhibitor AZ191 did not mimic the cytokine release profile of MBZ in untreated THP-1 monocytes. However, in THP-1 cells differentiated into macrophages, AZ191 strongly induced a pro-inflammatory cytokine release pattern similar to MBZ and LPS/IFNγ. Furthermore, like MBZ, AZ191 increased the expression of the M1 marker CD80 and decreased the M2 marker CD163 in THP-1 macrophages. In this model, AZ191 also increased phospho-ERK activity although to a lesser extent compared to MBZ. Taken together, the results demonstrate that DYRK1B inhibition could, at least partly, recapitulate immune responses induced by MBZ. Hence, DYRK1B inhibition induced by MBZ may be part of the mechanism of action to switch M2 to M1 macrophages.
  •  
4.
  • Blom, Kristin, et al. (author)
  • The anticancer effect of mebendazole may be due to M1 monocyte/macrophage activation via ERK1/2 and TLR8-dependent inflammasome activation
  • 2017
  • In: Immunopharmacology and immunotoxicology. - : Informa UK Limited. - 0892-3973 .- 1532-2513. ; 39:4, s. 199-210
  • Journal article (peer-reviewed)abstract
    • Mebendazole (MBZ), a drug commonly used for helminitic infections, has recently gained substantial attention as a repositioning candidate for cancer treatment. However, the mechanism of action behind its anticancer activity remains unclear. To address this problem, we took advantage of the curated MBZ-induced gene expression signatures in the LINCS Connectivity Map (CMap) database. The analysis revealed strong negative correlation with MEK/ERK1/2 inhibitors. Moreover, several of the most upregulated genes in response to MBZ exposure were related to monocyte/macrophage activation. The MBZ-induced gene expression signature in the promyeloblastic HL-60 cell line was strongly enriched in genes involved in monocyte/macrophage pro-inflammatory (M1) activation. This was subsequently validated using MBZ-treated THP-1 monocytoid cells that demonstrated gene expression, surface markers and cytokine release characteristic of the M1 phenotype. At high concentrations MBZ substantially induced the release of IL-1 beta and this was further potentiated by lipopolysaccharide (LPS). At low MBZ concentrations, cotreatment with LPS was required for MBZ-stimulated IL-1 beta secretion to occur. Furthermore, we show that the activation of protein kinase C, ERK1/2 and NF-kappaB were required for MBZ-induced IL-1 release. MBZ-induced IL-1 release was found to be dependent on NLRP3 inflammasome activation and to involve TLR8 stimulation. Finally, MBZ induced tumor-suppressive effects in a coculture model with differentiated THP-1 macrophages and HT29 colon cancer cells. In summary, we report that MBZ induced a pro-inflammatory (M1) phenotype of monocytoid cells, which may, at least partly, explain MBZ's anticancer activity observed in animal tumor models and in the clinic.
  •  
5.
  • Fryknäs, Mårten, et al. (author)
  • Screening for phenotype selective activity in multidrug resistant cells identifies a novel tubulin active agent insensitive to common forms of cancer drug resistance
  • 2013
  • In: BMC Cancer. - : Springer Science and Business Media LLC. - 1471-2407. ; 13, s. 374-
  • Journal article (peer-reviewed)abstract
    • Background: Drug resistance is a common cause of treatment failure in cancer patients and encompasses a multitude of different mechanisms. The aim of the present study was to identify drugs effective on multidrug resistant cells. Methods: The RPMI 8226 myeloma cell line and its multidrug resistant subline 8226/Dox40 was screened for cytotoxicity in response to 3,000 chemically diverse compounds using a fluorometric cytotoxicity assay (FMCA). Follow-up profiling was subsequently performed using various cellular and biochemical assays. Results: One compound, designated VLX40, demonstrated a higher activity against 8226/Dox40 cells compared to its parental counterpart. VLX40 induced delayed cell death with apoptotic features. Mechanistic exploration was performed using gene expression analysis of drug exposed tumor cells to generate a drug-specific signature. Strong connections to tubulin inhibitors and microtubule cytoskeleton were retrieved. The mechanistic hypothesis of VLX40 acting as a tubulin inhibitor was confirmed by direct measurements of interaction with tubulin polymerization using a biochemical assay and supported by demonstration of G2/M cell cycle arrest. When tested against a broad panel of primary cultures of patient tumor cells (PCPTC) representing different forms of leukemia and solid tumors, VLX40 displayed high activity against both myeloid and lymphoid leukemias in contrast to the reference compound vincristine to which myeloid blast cells are often insensitive. Significant in vivo activity was confirmed in myeloid U-937 cells implanted subcutaneously in mice using the hollow fiber model. Conclusions: The results indicate that VLX40 may be a useful prototype for development of novel tubulin active agents that are insensitive to common mechanisms of cancer drug resistance.
  •  
6.
  •  
7.
  • Segerman, Anna, et al. (author)
  • Clonal Variation in Drug and Radiation Response among Glioma-Initiating Cells Is Linked to Proneural-Mesenchymal Transition
  • 2016
  • In: Cell Reports. - : Elsevier BV. - 2211-1247. ; 17:11, s. 2994-3009
  • Journal article (peer-reviewed)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 continuumof 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.
  •  
8.
  • Selvin, Tove, et al. (author)
  • Immuno-oncological effects of standard anticancer agents and commonly used concomitant drugs : an in vitro assessment
  • 2024
  • In: BMC Pharmacology & Toxicology. - : BioMed Central (BMC). - 2050-6511. ; 25:1
  • Journal article (peer-reviewed)abstract
    • BackgroundIt has become evident in the field of oncology that the outcome of medical treatment is influenced by the combined effect exerted on both cancer- and immune cells. Therefore, we evaluated potential immunological effects of 46 standard anticancer agents and 22 commonly administered concomitant non-cancer drugs.MethodsWe utilized a miniaturized in vitro model system comprised of fluorescently labeled human colon and lung cancer cell lines grown as monocultures and co-cultured with activated peripheral blood mononuclear cells (PBMCs). The Bliss Independence Model was then applied to detect antagonism and synergy between the drugs and activated immune cells.ResultsAmong the standard anticancer agents, tyrosine kinase inhibitors (TKIs) stood out as the top inducers of both antagonism and synergy. Ruxolitinib and dasatinib emerged as the most notably antagonistic substances, exhibiting the lowest Bliss scores, whereas sorafenib was shown to synergize with activated PBMCs. Most concomitant drugs did not induce neither antagonism nor synergy. However, the statins mevastatin and simvastatin were uniquely shown to synergize with activated PBMC at all tested drug concentrations in the colon cancer model.ConclusionWe utilized a miniaturized tumor-immune model to enable time and cost-effective evaluation of a broad panel of drugs in an immuno-oncology setting in vitro. Using this approach, immunomodulatory effects exerted by TKIs and statins were identified.
  •  
9.
  • Selvin, Tove, et al. (author)
  • Phenotypic screening platform identifies statins as enhancers of immune cell-induced cancer cell death.
  • 2023
  • In: BMC cancer. - : Springer Science and Business Media LLC. - 1471-2407. ; 23:1
  • Journal article (peer-reviewed)abstract
    • High-throughput screening (HTS) of small molecule drug libraries has greatly facilitated the discovery of new cancer drugs. However, most phenotypic screening platforms used in the field of oncology are based solely on cancer cell populations and do not allow for the identification of immunomodulatory agents.We developed a phenotypic screening platform based on a miniaturized co-culture system with human colorectal cancer- and immune cells, providing a model that recapitulates part of the tumor immune microenvironment (TIME) complexity while simultaneously being compatible with a simple image-based readout. Using this platform, we screened 1,280 small molecule drugs, all approved by the Food and Drug Administration (FDA), and identified statins as enhancers of immune cell-induced cancer cell death.The lipophilic statin pitavastatin had the most potent anti-cancer effect. Further analysis demonstrated that pitavastatin treatment induced a pro-inflammatory cytokine profile as well as an overall pro-inflammatory gene expression profile in our tumor-immune model.Our study provides an in vitro phenotypic screening approach for the identification of immunomodulatory agents and thus addresses a critical gap in the field of immuno-oncology. Our pilot screen identified statins, a drug family gaining increasing interest as repurposing candidates for cancer treatment, as enhancers of immune cell-induced cancer cell death. We speculate that the clinical benefits described for cancer patients receiving statins are not simply caused by a direct effect on the cancer cells but rather are dependent on the combined effect exerted on both cancer and immune cells.
  •  
10.
  • Selvin, Tove, et al. (author)
  • The Immuno-Oncology Hollow Fiber Assay
  • Other publication (other academic/artistic)abstract
    • In order to facilitate the translation of novel immunotherapies from bench to bedside, continued development of predictive preclinical models is essential. Herein, we developed the immuno-oncology hollow fiber assay (HFA) to bridge the gap between cell based in vitro assays and more complex mouse models for evaluation of immuno-oncological agents. The colorectal cancer (CRC) cell line HCT116-GFP and human peripheral blood mononuclear cells (PBMCs) were co-cultured inside semipermeable hollow fibers. As a proof of concept, aCD3 and IL-2 was used to induce immune cell-mediated cancer cell death. During in vitro characterization of the model system, an enhanced effect of aCD3 and IL-2 was observed in the HFA compared to conventional monolayers. Further investigation demonstrated that increased cell proximity alone is sufficient to augment immune cell activation and effector function. To assess the functionality of the assay in vivo, a pilot study was performed using nude mice. Hollow fibers were surgically implanted intraperitoneally (i.p.) and the mice received local injections of aCD3 at the time of implantation and/ or systemic IL-2 via i.p. injection once daily for 3 consecutive days. Compared to untreated mice and mice receiving IL-2 alone, the combination of aCD3 and IL-2 resulted in a significant decrease in cancer cell viability. Traditional in vivo models often necessitate lengthy observation periods to monitor tumor growth and treatment response. We have developed a simplified model system that enables initial in vivo evaluation of immunological agents on cancer and immune cells of human origin within a matter of days. 
  •  
Skapa referenser, mejla, bekava och länka
  • Result 1-10 of 47
Type of publication
journal article (34)
other publication (4)
research review (3)
book chapter (3)
doctoral thesis (2)
licentiate thesis (1)
show more...
show less...
Type of content
peer-reviewed (34)
other academic/artistic (12)
pop. science, debate, etc. (1)
Author/Editor
Jarvius, Malin (43)
Larsson, Rolf (26)
Fryknäs, Mårten (23)
Nygren, Peter (12)
Söderberg, Ola (12)
Andersson, Claes (9)
show more...
Landegren, Ulf (8)
Selvin, Tove (7)
Berglund, Malin (7)
Uhrbom, Lene (7)
Leuchowius, Karl-Joh ... (7)
Jarvius, Jonas (6)
Rickardson, Linda (6)
Gullberg, Mats (6)
Blom, Kristin (5)
Lenhammar, Lena (5)
Parrow, Vendela (5)
Sundström, Anders (4)
Gustafsson, Mats G. (4)
Gullbo, Joachim (4)
Senkowski, Wojciech (4)
Weibrecht, Irene (4)
Höglund, Martin (3)
Alafuzoff, Irina (3)
Kamali-Moghaddam, Ma ... (3)
Dotti, Gianpietro (3)
Hesselager, Göran (3)
Svensson, Emma (3)
Östman, Arne (3)
Nelander, Sven (3)
Rubin, Jenny (3)
Maturi, Naga Prathyu ... (3)
Olsson-Strömberg, Ul ... (3)
Paulsson, Janna (3)
Jiang, Yiwen (3)
Gustafsson, Mats (2)
Nilsson, Mats (2)
Wickström, Malin (2)
Larsson, Lars-Gunnar (2)
Schallmeiner, Edith (2)
Westermark, Bengt (2)
Linder, Stig (2)
Loskog, Angelica S., ... (2)
Olofsson, Sara (2)
Eriksson, Anna (2)
Marinescu, Voichita ... (2)
Bahram, Fuad (2)
Bergström, Tobias, 1 ... (2)
Hassan, Sadia Bashir (2)
Dang, Yonglong (2)
show less...
University
Uppsala University (46)
Karolinska Institutet (12)
Stockholm University (2)
Lund University (2)
The Swedish School of Sport and Health Sciences (2)
University of Gothenburg (1)
show more...
Royal Institute of Technology (1)
Chalmers University of Technology (1)
show less...
Language
English (47)
Research subject (UKÄ/SCB)
Medical and Health Sciences (30)
Natural sciences (6)

Year

Kungliga biblioteket hanterar dina personuppgifter i enlighet med EU:s dataskyddsförordning (2018), GDPR. Läs mer om hur det funkar här.
Så här hanterar KB dina uppgifter vid användning av denna tjänst.

 
pil uppåt Close

Copy and save the link in order to return to this view