Umeå University's logo

umu.sePublications
Planned maintenance
A system upgrade is planned for 10/12-2024, at 12:00-13:00. During this time DiVA will be unavailable.
Change search
Refine search result
12 1 - 50 of 87
CiteExportLink to result list
Permanent link
Cite
Citation style
  • apa
  • ieee
  • modern-language-association-8th-edition
  • vancouver
  • Other style
More styles
Language
  • de-DE
  • en-GB
  • en-US
  • fi-FI
  • nn-NO
  • nn-NB
  • sv-SE
  • Other locale
More languages
Output format
  • html
  • text
  • asciidoc
  • rtf
Rows per page
  • 5
  • 10
  • 20
  • 50
  • 100
  • 250
Sort
  • Standard (Relevance)
  • Author A-Ö
  • Author Ö-A
  • Title A-Ö
  • Title Ö-A
  • Publication type A-Ö
  • Publication type Ö-A
  • Issued (Oldest first)
  • Issued (Newest first)
  • Created (Oldest first)
  • Created (Newest first)
  • Last updated (Oldest first)
  • Last updated (Newest first)
  • Disputation date (earliest first)
  • Disputation date (latest first)
  • Standard (Relevance)
  • Author A-Ö
  • Author Ö-A
  • Title A-Ö
  • Title Ö-A
  • Publication type A-Ö
  • Publication type Ö-A
  • Issued (Oldest first)
  • Issued (Newest first)
  • Created (Oldest first)
  • Created (Newest first)
  • Last updated (Oldest first)
  • Last updated (Newest first)
  • Disputation date (earliest first)
  • Disputation date (latest first)
Select
The maximal number of hits you can export is 250. When you want to export more records please use the Create feeds function.
  • 1. Antonsson, Andreas
    et al.
    Persson, Jenny L.
    Department of Laboratory Medicine, Division of Experimental Cancer Research, Clinical Research Center, Lund University, Malmö, Sweden.
    Induction of apoptosis by staurosporine involves the inhibition of expression of the major cell cycle proteins at the G(2)/m checkpoint accompanied by alterations in Erk and Akt kinase activities2009In: Anticancer Research, ISSN 0250-7005, E-ISSN 1791-7530, Vol. 29, no 8, p. 2893-2898Article in journal (Refereed)
    Abstract [en]

    BACKGROUND: Staurosporine is a therapeutic agent that inhibits tumor cell growth by inducing cell death via intrinsic apoptotic pathways. Our previous studies in clinical settings have suggested that certain subpopulations of patients with acute myeloid leukemia (AML) had poor response to chemotherapy.

    MATERIALS AND METHODS: The effect of staurosporine on apoptosis and cell cycle distribution in human leukemic cell line U-937 cells was determined. U-937 cells were treated with staurosporine at 0.5 microM for 18 hours or 1 microM for 24 hours. Analyses of cell cycle distribution and apoptosis were performed using flow cytometric analysis. The effects of staurosporine on the targeted proteins were assessed by immunoblot analysis.

    RESULTS: A blockade of the cell cycle at the G(2)/M phase was observed in U-937 cells treated with staurosporine. A concomitant induction of apoptosis and activation of caspase-3 in U-937 cells was also achieved. Treatment of U-937 cells with staurosporine at 1 microM for 24 hours, compared with 0.5 microM for 18 hours, appeared to kill the leukemic more efficiently cells and this dose and duration may specifically target p27, Erk and Akt pathways that are important for cancer cell survival and resistance to treatment. We also show that the effects of stauroporine on cell cycle progression and apoptosis in U-937 cells are closely linked.

    CONCLUSION: Our results suggest that induction of apoptosis and inhibitory proliferation and survival pathways are important events induced by staurosporine. Understanding the conditions under which staurosporine shows high specificity and low toxicity in treatment of leukemic cells is of great importance for improving the efficacy of targeted therapeutics and overcoming resistance to chemotherapeutic agents.

  • 2. Atiomo, William
    et al.
    Shafiee, Mohamad Nasir
    Chapman, Caroline
    Metzler, Veronika M.
    Abouzeid, Jad
    Latif, Ayşe
    Chadwick, Amy
    Kitson, Sarah
    Sivalingam, Vanitha N.
    Stratford, Ian J.
    Rutland, Catrin S.
    Persson, Jenny L.
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine). Clinical Research Center, Lund University, Malmö, Sweden.
    Ødum, Niels
    Fuentes-Utrilla, Pablo
    Jeyapalan, Jennie N.
    Heery, David M.
    Crosbie, Emma J.
    Mongan, Nigel P.
    Expression of NAD(P)H quinone dehydrogenase 1 (NQO1) is increased in the endometrium of women with endometrial cancer and women with polycystic ovary syndrome2017In: Clinical Endocrinology, ISSN 0300-0664, E-ISSN 1365-2265, Vol. 87, no 5, p. 557-565Article in journal (Refereed)
    Abstract [en]

    Objective: Women with a prior history of polycystic ovary syndrome (PCOS) have an increased risk of endometrial cancer (EC). Aim: To investigate whether the endometrium of women with PCOS possesses gene expression changes similar to those found in EC. Design and Methods: Patients with EC, PCOS and control women unaffected by either PCOS or EC were recruited into a cross-sectional study at the Nottingham University Hospital, UK. For RNA sequencing, representative individual endometrial biopsies were obtained from women with EC, PCOS and a woman unaffected by PCOS or EC. Expression of a subset of differentially expressed genes identified by RNA sequencing, including NAD(P)H quinone dehydrogenase 1 (NQO1), was validated by quantitative reverse transcriptase PCR validation (n = 76) and in the cancer genome atlas UCEC (uterine corpus endometrioid carcinoma) RNA sequencing data set (n = 381). The expression of NQO1 was validated by immunohistochemistry in EC samples from a separate cohort (n = 91) comprised of consecutive patients who underwent hysterectomy at St Mary's Hospital, Manchester, between 2011 and 2013. A further 6 postmenopausal women with histologically normal endometrium who underwent hysterectomy for genital prolapse were also included. Informed consent and local ethics approval were obtained for the study. Results: We show for the first that NQO1 expression is significantly increased in the endometrium of women with PCOS and EC. Immunohistochemistry confirms significantly increased NQO1 protein expression in EC relative to nonmalignant endometrial tissue (P < .0001). Conclusions: The results obtained here support a previously unrecognized molecular link between PCOS and EC involving NQO1.

    Download full text (pdf)
    fulltext
  • 3.
    Beyer, Sarah
    et al.
    Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden.
    Kimani, Martha
    Chemical and Optical Sensing Division, Bundesanstalt für Materialforschung und-prüfung (BAM), Richard-Willstätter Straße 11, Berlin, Germany.
    Zhang, Yuecheng
    Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden; Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden.
    Verhassel, Alejandra
    Institute of Biomedicine, University of Turku, Turku, Finland; FICAN West Cancer Centre, Turku University Hospital, Turku, Finland.
    Sternbæk, Louise
    Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden; Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden; Phase Holographic Imaging AB, Lund, Sweden.
    Wang, Tianyan
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine).
    Persson, Jenny L.
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine). Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden; Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden.
    Härkönen, Pirkko
    Institute of Biomedicine, University of Turku, Turku, Finland; FICAN West Cancer Centre, Turku University Hospital, Turku, Finland.
    Johansson, Emil
    Umeå University, Faculty of Science and Technology, Department of Chemistry. Umeå University, Faculty of Medicine, Umeå Centre for Microbial Research (UCMR).
    Caraballo, Remi
    Umeå University, Faculty of Science and Technology, Department of Chemistry. Umeå University, Faculty of Medicine, Umeå Centre for Microbial Research (UCMR).
    Elofsson, Mikael
    Umeå University, Faculty of Science and Technology, Department of Chemistry. Umeå University, Faculty of Medicine, Umeå Centre for Microbial Research (UCMR).
    Gawlitza, Kornelia
    Chemical and Optical Sensing Division, Bundesanstalt für Materialforschung und-prüfung (BAM), Richard-Willstätter Straße 11, Berlin, Germany.
    Rurack, Knut
    Chemical and Optical Sensing Division, Bundesanstalt für Materialforschung und-prüfung (BAM), Richard-Willstätter Straße 11, Berlin, Germany.
    Ohlsson, Lars
    Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden; Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden.
    El-Schich, Zahra
    Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden; Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden.
    Wingren, Anette Gjörloff
    Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden; Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden.
    Stollenwerk, Maria M.
    Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden; Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden.
    Fluorescent Molecularly Imprinted Polymer Layers against Sialic Acid on Silica-Coated Polystyrene Cores — Assessment of the Binding Behavior to Cancer Cells2022In: Cancers, ISSN 2072-6694, Vol. 14, no 8, article id 1875Article in journal (Refereed)
    Abstract [en]

    Sialic acid (SA) is a monosaccharide usually linked to the terminus of glycan chains on the cell surface. It plays a crucial role in many biological processes, and hypersialylation is a common feature in cancer. Lectins are widely used to analyze the cell surface expression of SA. However, these protein molecules are usually expensive and easily denatured, which calls for the development of alternative glycan-specific receptors and cell imaging technologies. In this study, SA-imprinted fluorescent core-shell molecularly imprinted polymer particles (SA-MIPs) were employed to recognize SA on the cell surface of cancer cell lines. The SA-MIPs improved suspensibility and scattering properties compared with previously used core-shell SA-MIPs. Although SA-imprinting was performed using SA without preference for the α2,3-and α2,6-SA forms, we screened the cancer cell lines analyzed using the lectins Maackia Amurensis Lectin I (MAL I, α2,3-SA) and Sambucus Nigra Lectin (SNA, α2,6-SA). Our results show that the selected cancer cell lines in this study presented a varied binding behavior with the SA-MIPs. The binding pattern of the lectins was also demonstrated. Moreover, two different pentavalent SA conjugates were used to inhibit the binding of the SA-MIPs to breast, skin, and lung cancer cell lines, demonstrating the specificity of the SA-MIPs in both flow cytometry and confocal fluorescence microscopy. We concluded that the synthesized SA-MIPs might be a powerful future tool in the diagnostic analysis of various cancer cells.

    Download full text (pdf)
    fulltext
  • 4. Blumel, Edda
    et al.
    Willerslev-Olsen, Andreas
    Gluud, Maria
    Lindahl, Lise M.
    Fredholm, Simon
    Nastasi, Claudia
    Krejsgaard, Thorbjorn
    Surewaard, Bas G. J.
    Koralov, Sergei B.
    Hu, Tengpeng
    Persson, Jenny L.
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine). Clinical Research Center, Lund University, Sweden.
    Bonefeld, Charlotte Menne
    Geisler, Carsten
    Iversen, Lars
    Becker, Juergen C.
    Andersen, Mads Hald
    Woetmann, Anders
    Buus, Terkild Brink
    Odum, Niels
    Staphylococcal alpha-toxin tilts the balance between malignant and non-malignant CD4+ T cells in cutaneous T-cell lymphoma2019In: Oncoimmunology, ISSN 2162-4011, E-ISSN 2162-402X, Vol. 8, no 11, article id e1641387Article in journal (Refereed)
    Abstract [en]

    Staphylococcus aureus is implicated in disease progression in cutaneous T-cell lymphoma (CTCL). Here, we demonstrate that malignant T cell lines derived from CTCL patients as well as primary malignant CD4+ T cells from Sézary syndrome patients are considerably more resistant to alpha-toxin-induced cell death than their non-malignant counterparts. Thus, in a subset of Sézary syndrome patients the ratio between malignant and non-malignant CD4+ T cells increases significantly following exposure to alpha-toxin. Whereas toxin-induced cell death is ADAM10 dependent in healthy CD4+ T cells, resistance to alpha-toxin in malignant T cells involves both downregulation of ADAM10 as well as other resistance mechanisms. In conclusion, we provide first evidence that Staphylococcus aureus derived alpha-toxin can tilt the balance between malignant and non-malignant CD4+ T cells in CTCL patients. Consequently, alpha-toxin may promote disease progression through positive selection of malignant CD4+ T cells, identifying alpha-toxin as a putative drug target in CTCL.

    Download full text (pdf)
    fulltext
  • 5. Buus, Terkild Brink
    et al.
    Willerslev-Olsen, Andreas
    Fredholm, Simon
    Blumel, Edda
    Nastasi, Claudia
    Gluud, Maria
    Hu, Tengpeng
    Lindahl, Lise M.
    Iversen, Lars
    Fogh, Hanne
    Gniadecki, Robert
    Litvinov, Ivan V.
    Persson, Jenny L.
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine). Clinical Research Center, Lund University, Malmö, Sweden.
    Bonefeld, Charlotte Menne
    Geisler, Carsten
    Christensen, Jan Praysgaard
    Krejsgaard, Thorbjorn
    Litman, Thomas
    Woetmann, Anders
    Odum, Niels
    Single-cell heterogeneity in Sézary syndrome2018In: Blood Advances, ISSN 2473-9529 , E-ISSN 2473-9537, Vol. 2, no 16, p. 2115-2126Article in journal (Refereed)
    Abstract [en]

    Sezary syndrome (SS) is an aggressive leukemic variant of cutaneous T-cell lymphoma (CTCL) with a median life expectancy of less than 4 years. Although initial treatment responses are often good, the vast majority of patients with SS fail to respond to ongoing therapy. We hypothesize that malignant T cells are highly heterogeneous and harbor subpopulations of SS cells that are both sensitive and resistant to treatment. Here, we investigate the presence of single-cell heterogeneity and resistance to histone deacetylase inhibitors (HDACi) within primary malignant T cells from patients with SS. Using single-cell RNA sequencing and flow cytometry, we find that malignant T cells from all investigated patients with SS display a high degree of single-cell heterogeneity at both the mRNA and protein levels. We show that this heterogeneity divides the malignant cells into distinct subpopulations that can be isolated by their expression of different surface antigens. Finally, we show that treatment with HDACi (suberanilohydroxamic acid and romidepsin) selectively eliminates some subpopulations while leaving other subpopulations largely unaffected. In conclusion, we show that patients with SS display a high degree of single-cell heterogeneity within the malignant T-cell population, and that distinct subpopulations of malignant T cells carry HDACi resistance. Our data point to the importance of understanding the heterogeneous nature of malignant SS cells in each individual patient to design combinational and new therapies to counter drug resistance and treatment failure.

  • 6. de Brot, Simone
    et al.
    Ntekim, Atara
    Cardenas, Ryan
    James, Victoria
    Allegrucci, Cinzia
    Heery, David M.
    Bates, David O.
    Ødum, Niels
    Persson, Jenny L.
    Clinical Research Center, Lund University, Malmö, Sweden.
    Mongan, Nigel P
    Regulation of vascular endothelial growth factor in prostate cancer2015In: Endocrine-Related Cancer, ISSN 1351-0088, E-ISSN 1479-6821, Vol. 22, no 3, p. R107-R123Article in journal (Refereed)
    Abstract [en]

    Prostate cancer (PCa) is the most common malignancy affecting men in the western world. Although radical prostatectomy and radiation therapy can successfully treat PCa in the majority of patients, up to ~30% will experience local recurrence or metastatic disease. Prostate carcinogenesis and progression is typically an androgen-dependent process. For this reason, therapies for recurrent PCa target androgen biosynthesis and androgen receptor function. Such androgen deprivation therapies (ADT) are effective initially, but the duration of response is typically ≤24 months. Although ADT and taxane-based chemotherapy have delivered survival benefits, metastatic PCa remains incurable. Therefore, it is essential to establish the cellular and molecular mechanisms that enable localized PCas to invade and disseminate. It has long been accepted that metastases require angiogenesis. In the present review, we examine the essential role for angiogenesis in PCa metastases, and we focus in particular on the current understanding of the regulation of vascular endothelial growth factor (VEGF) in localized and metastatic PCa. We highlight recent advances in understanding the role of VEGF in regulating the interaction of cancer cells with tumor-associated immune cells during the metastatic process of PCa. We summarize the established mechanisms of transcriptional and post-transcriptional regulation of VEGF in PCa cells and outline the molecular insights obtained from preclinical animal models of PCa. Finally, we summarize the current state of anti-angiogenesis therapies for PCa and consider how existing therapies impact VEGF signaling.

  • 7. Diffner, Eva
    et al.
    Gauffin, Fredrika
    Anagnostaki, Lola
    Nordgren, Ann
    Gustafsson, Bertil
    Sander, Birgitta
    Gustafsson, Britt
    Persson, Jenny Liao
    Laboratory Medicine, Clinical Research Center, Lund University.
    Expression of VEGF and VEGF Receptors in Childhood Precursor B-cell Acute Lymphoblastic Leukemia Evaluated by Immunohistochemistry2009In: Journal of pediatric hematology/oncology (Print), ISSN 1077-4114, E-ISSN 1536-3678, Vol. 31, no 9, p. 696-701Article in journal (Refereed)
    Abstract [en]

    Perturbation in the expression and signaling pathways of vascular endothelial growth factor (VEGF) has been linked to pathogenesis of hematologic malignancies. We investigated the expression and clinical importance of VEGF and two of its receptors, VEGFR-1 and VEGFR-2, in childhood precursor B-cell acute lymphoblastic leukemia (pre-B ALL) by using immunohistochemistry. These angiogenic proteins were expressed in the majority of leukemic bone marrow samples. Notably, pre-B ALL patients had significantly increased expression of VEGFR-1 compared with no expression in the nonmalignant group, indicating a link between VEGFR-1 protein expression and pre-B ALL. These novel findings suggest that VEGFR-1 may have clinical importance in childhood pre-B ALL.

  • 8.
    Dongre, Mitesh
    et al.
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine). Umeå University, Faculty of Medicine, Molecular Infection Medicine Sweden (MIMS).
    Singh, Bhupender
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine). Umeå University, Faculty of Medicine, Molecular Infection Medicine Sweden (MIMS).
    Aung, Kyaw Min
    Umeå University, Faculty of Medicine, Molecular Infection Medicine Sweden (MIMS). Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine).
    Larsson, Per
    Umeå University, Faculty of Medicine, Molecular Infection Medicine Sweden (MIMS). Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine).
    Miftakhova, Regina R.
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine). Umeå University, Faculty of Medicine, Molecular Infection Medicine Sweden (MIMS).
    Persson, Karina
    Umeå University, Faculty of Science and Technology, Department of Chemistry.
    Askarian, Fatemeh
    Johannessen, Mona
    von Hofsten, Jonas
    Umeå University, Faculty of Medicine, Umeå Centre for Molecular Medicine (UCMM). Umeå University, Faculty of Medicine, Department of Integrative Medical Biology (IMB).
    Persson, Jenny L.
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine). Umeå University, Faculty of Medicine, Molecular Infection Medicine Sweden (MIMS).
    Erhardt, Marc
    Tuck, Simon
    Umeå University, Faculty of Medicine, Umeå Centre for Molecular Medicine (UCMM).
    Uhlin, Bernt Eric
    Umeå University, Faculty of Medicine, Molecular Infection Medicine Sweden (MIMS). Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine).
    Wai, Sun Nyunt
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine). Umeå University, Faculty of Medicine, Molecular Infection Medicine Sweden (MIMS).
    Flagella-mediated secretion of a novel Vibrio cholerae cytotoxin affecting both vertebrate and invertebrate hosts2018In: Communications Biology, E-ISSN 2399-3642, Vol. 1, article id 59Article in journal (Refereed)
    Abstract [en]

    Using Caenorhabditis elegans as an infection host model for Vibrio cholerae predator interactions, we discovered a bacterial cytotoxin, MakA, whose function as a virulence factor relies on secretion via the flagellum channel in a proton motive force-dependent manner. The MakA protein is expressed from the polycistronic makDCBA (motility-associated killing factor) operon. Bacteria expressing makDCBA induced dramatic changes in intestinal morphology leading to a defecation defect, starvation and death in C. elegans. The Mak proteins also promoted V. cholerae colonization of the zebrafish gut causing lethal infection. A structural model of purified MakA at 1.9 Å resolution indicated similarities to members of a superfamily of bacterial toxins with unknown biological roles. Our findings reveal an unrecognized role for V. cholerae flagella in cytotoxin export that may contribute both to environmental spread of the bacteria by promoting survival and proliferation in encounters with predators, and to pathophysiological effects during infections.

    Download full text (pdf)
    fulltext
  • 9. Ekberg, Jenny
    et al.
    Holm, Caroline
    Jalili, Sara
    Richter, Johan
    Anagnostaki, Lola
    Landberg, Göran
    Persson, Jenny L.
    Division of Pathology, Department of LaboratoryMedicine, Lund University, University Hospital, Malmö, Sweden.
    Expression of cyclin A1 and cell cycle proteins in hematopoietic cells and acute myeloid leukemia and links to patient outcome2005In: European Journal of Haematology, ISSN 0902-4441, E-ISSN 1600-0609, Vol. 75, no 2, p. 106-115Article in journal (Refereed)
    Abstract [en]

    Abnormal expression of several key regulators essential for G1/S transitions has been implicated in tumorigenesis. A critical role of cyclin A1 in the development of acute myeloid leukemia (AML) has previously been demonstrated in transgenic mice. Our present study focused on the expression and prognostic significance of cyclin A1 and a panel of cell cycle regulatory proteins including cyclin A2, cyclin B1, cyclin E, CDK1, CDK2, p21 and p27 in bone marrow samples from 40 patients with AML. Freshly isolated CD34+ hematopoietic cells and bone marrow samples from 10 healthy donors were also assessed for cell type- and subcellular-specific expression of the cell cycle regulatory proteins. The level of cyclin A1 expression was the only factor that showed a significant correlation with patient outcome. In log-rank test stratified by levels of cyclin A1 expression, patients with high levels of cyclin A1 had significantly worse overall survival (OS) (P = 0.012) compared to those with low levels. Further, patients with high levels of cyclin A1 had significantly lower disease-free survival (DFS) (P = 0.028). Multivariate analysis indicated that cyclin A1 protein expression was an independent prognostic factor for predicting DFS (P = 0.035) and OS (P = 0.045). No correlation between cyclin A1 expression and age was found. However, expression of cyclin A2, cyclin B1, cyclin E, CDK1, CDK2, p21 and p27 did not show prognostic significance in these AML patients.

  • 10. Ekberg, Jenny
    et al.
    Landberg, Göran
    Holm, Caroline
    Richter, Johan
    Wolgemuth, Debra J
    Persson, Jenny L.
    Division of Pathology, Department of Laboratory Medicine, Lund University, University Hospital, Malmö, Sweden.
    Regulation of the cyclin A1 protein is associated with its differential subcellular localization in hematopoietic and leukemic cells2004In: Oncogene, ISSN 0950-9232, E-ISSN 1476-5594, Vol. 23, no 56, p. 9082-9089Article in journal (Refereed)
    Abstract [en]

    An important role of the cell cycle regulatory protein cyclin A1 in the development of acute myeloid leukemia (AML) was previously demonstrated in a transgenic mouse model. We have now turned our attention to study specific aspects of the activity and subcellular distribution of cyclin A1 using bone marrow samples from normal donors and patients with AML, as well as leukemic cell lines. We show that the localization of cyclin A1 in normal hematopoietic cells is nuclear, whereas in leukemic cells from AML patients and cell lines, it is predominantly cytoplasmic. In leukemic cell lines treated with all-trans retinoic acid (ATRA), cyclin A1 localized to the nucleus. Further, there was a direct interaction between cyclin A1 and cyclin-dependent kinase 1, as well as a major ATRA receptor, RARalpha, in ATRA-treated cells but not in untreated leukemic cells. Our results indicate that the altered intracellular distribution of cyclin A1 in leukemic cells correlates with the status of the leukemic phenotype.

  • 11. Ekberg, Jenny
    et al.
    Persson, Jenny L.
    Post-translational modification of cyclin A1 is associated with staurosporine and TNFalpha induced apoptosis in leukemic cells.2009In: Molecular and Cellular Biochemistry, ISSN 0300-8177, E-ISSN 1573-4919, Vol. 320, no 1-2, p. 115-24Article in journal (Refereed)
    Abstract [en]

    Understanding of molecular mechanisms underlying the effects of cell cycle proteins in response to the chemotherapeutic agents is of great importance for improving the efficacy of targeted therapeutics and overcoming resistance to chemotherapeutic agents. Staurosporine and tumor necrosis factor alpha (TNFalpha) are the therapeutic agents that inhibit tumor cell growth by inducing cell death. Staurosporine induces apoptosis through the intrinsic pathway, while TNFalpha trigger the cell death via the extrinsic apoptotic pathway. We have previously demonstrated that the cell cycle regulatory protein, cyclin A1 played an important role in the development of acute myeloid leukemia (AML), and cyclin A1 expression correlated with disease characteristics and patient outcome in leukemia. However, it remains unknown how cyclin A1 expression is regulated in leukemic cells treated with the therapeutic agents. Here, we demonstrate that cyclin A1 protein is regulated by proteasome-mediated ubiquitination and degradation in untreated U-937 cells. Interestingly, ubiquitination- and proteasomal-mediated degradation of cyclin A1 is prevented in cells treated with staurosporine or TNFalpha. Induction of apoptosis in U-937 cells by staurosporine or TNFalpha resulted in an increase in cyclin A1 protein expression, which correlated well with cyclin A1 protein modification and the activation of caspase-3. Blocking caspases activity by Z-VAD-FMK had no effect on the increased cyclin A1 expression, suggesting that cyclin A1 might be regulated by caspase-3 independent pathways. We further propose that CDC25C may be associated with cyclin A1 protein modification in response to staurosporine or TNFalpha treatment. Our results suggest that cyclin A1 protein is stabilized via post-transcriptional modification in response to apoptosis induced by staurosporine or TNFalpha.

  • 12. El-Schich, Zahra
    et al.
    Janicke, Birgit
    Alm, Kersti
    Dizeyi, Nishtman
    Persson, Jenny L.
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine). Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden; Biofilms-Research Center for Biointerfaces, Malmö University, Sweden.
    Gjorloff Wingren, Anette
    Discrimination between Breast Cancer Cells and White Blood Cells by Non-Invasive Measurements: Implications for a Novel In Vitro-Based Circulating Tumor Cell Model Using Digital Holographic Cytometry2020In: Applied Sciences, E-ISSN 2076-3417, Vol. 10, no 14, article id 4854Article in journal (Refereed)
    Abstract [en]

    Breast cancer is the second most common cancer worldwide. Metastasis is the main reason for death in breast cancer, and today, there is a lack of methods to detect and isolate circulating tumor cells (CTCs), mainly due to their heterogeneity and rarity. There are some systems that are designed to detect rare epithelial cancer cells in whole blood based on the most common marker used today, the epithelial cell adhesion molecule (EpCAM). It has been shown that aggressive breast cancer metastases are of non-epithelial origin and are therefore not always detected using EpCAM as a marker. In the present study, we used an in vitro-based circulating tumor cell model comprising a collection of six breast cancer cell lines and white blood cell lines. We used digital holographic cytometry (DHC) to characterize and distinguish between the different cell types by area, volume and thickness. Here, we present significant differences in cell size-related parameters observed when comparing white blood cells and breast cancer cells by using DHC. In conclusion, DHC can be a powerful diagnostic tool for the characterization of CTCs in the blood.

    Download full text (pdf)
    fulltext
  • 13.
    El-Schich, Zahra
    et al.
    Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden; Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden.
    Zhang, Yuecheng
    Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden; Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden.
    Göransson, Tommy
    Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden.
    Dizeyi, Nishtman
    Department of Translational Medicine, Lund University, Malmö, Sweden.
    Persson, Jenny L.
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine). Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö; Biofilms-Research Center for Biointerfaces, Malmö University.
    Johansson, Emil
    Umeå University, Faculty of Science and Technology, Department of Chemistry. Umeå University, Faculty of Medicine, Umeå Centre for Microbial Research (UCMR).
    Caraballo, Remi
    Umeå University, Faculty of Science and Technology, Department of Chemistry. Umeå University, Faculty of Medicine, Umeå Centre for Microbial Research (UCMR).
    Elofsson, Mikael
    Umeå University, Faculty of Science and Technology, Department of Chemistry. Umeå University, Faculty of Medicine, Umeå Centre for Microbial Research (UCMR).
    Shinde, Sudhirkumar
    School of Consciousness, Vishwanath Karad Maharashtra Institute of Technology—World Peace University, Kothrud, Pune, India.
    Sellergren, Börje
    Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden; Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden.
    Wingren, Anette Gjörloff
    Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden; Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden.
    Sialic acid as a biomarker studied in breast cancer cell lines in vitro using fluorescent molecularly imprinted polymers2021In: Applied Sciences, E-ISSN 2076-3417, Vol. 11, no 7, article id 3256Article in journal (Refereed)
    Abstract [en]

    Sialylations are post-translational modifications of proteins and lipids that play important roles in many cellular events, including cell-cell interactions, proliferation, and migration. Tumor cells express high levels of sialic acid (SA), which are often associated with the increased invasive potential in clinical tumors, correlating with poor prognosis. To overcome the lack of natural SA-receptors, such as antibodies and lectins with high enough specificity and sensitivity, we have used molecularly imprinted polymers (MIPs), or “plastic antibodies”, as nanoprobes. Because high expression of epithelial cell adhesion molecule (EpCAM) in primary tumors is often associated with proliferation and a more aggressive phenotype, the expression of EpCAM and CD44 was initially analyzed. The SA-MIPs were used for the detection of SA on the cell surface of breast cancer cells. Lectins that specifically bind to the a-2,3 SA and a-2,6 SA variants were used for analysis of SA expression, with both flow cytometry and confocal microscopy. Here we show a correlation of EpCAM and SA expression when using the SA-MIPs for detection of SA. We also demonstrate the binding pattern of the SA-MIPs on the breast cancer cell lines using confocal microscopy. Pre-incubation of the SA-MIPs with SA-derivatives as inhibitors could reduce the binding of the SA-MIPs to the tumor cells, indicating the specificity of the SA-MIPs. In conclusion, the SA-MIPs may be a new powerful tool in the diagnostic analysis of breast cancer cells.

    Download full text (pdf)
    fulltext
  • 14.
    Feith, Marek
    et al.
    Department of Pathophysiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic; Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden.
    Zhang, Yuecheng
    Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden; Biofilms Research Center for Biointerfaces, Malmö University, Malmö, Sweden; College of Chemistry and Chemical Engineering, Yan’an University, Yan’an, China.
    Persson, Jenny L.
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine). Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden; Biofilms Research Center for Biointerfaces, Malmö University, Malmö, Sweden.
    Balvan, Jan
    Department of Pathophysiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.
    El-Schich, Zahra
    Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden; Biofilms Research Center for Biointerfaces, Malmö University, Malmö, Sweden.
    Wingren, Anette Gjörloff
    Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden; Biofilms Research Center for Biointerfaces, Malmö University, Malmö, Sweden.
    Circulating tumor cell models mimicking metastasizing cells in vitro: discrimination of colorectal cancer cells and white blood cells using digital holographic cytometry2022In: Photonics, ISSN 2304-6732, Vol. 9, no 12, article id 955Article in journal (Refereed)
    Abstract [en]

    Colorectal cancer (CRC) is the second most metastatic disease with the majority of cases detected in Western countries. Metastases are formed by circulating altered phenotype tumor cells causing 20% of CRC related deaths. Metastatic cells may show higher expression of surface molecules such as CD44, and changes in morphological properties are associated with increased invasiveness and poor prognosis. In this study, we intended to mimic the environment for metastasizing cells. Here, we used digital holographic cytometry (DHC) analysis to determine cellular morphological properties of three metastatic and two non-metastatic colorectal cancer cell lines to show differences in morphology between the CRC cells and peripheral blood mononuclear cells (PBMCs). By establishing differences in cell area, cell thickness, cell volume, and cell irregularity even when the CRC cells were in minority (5% out of PBMCs), DHC does discriminate between CRC cells and the PBMCs in vitro. We also analyzed the epithelial marker EpCAM and migration marker CD44 using flow cytometry and demonstrate that the CRC cell lines and PBMC cells differ in EpCAM and CD44 expression. Here, we present DHC as a new powerful tool in discriminating cells of different sizes in suspension together with a combination of biomarkers.

    Download full text (pdf)
    fulltext
  • 15.
    Flodbring Larsson, Per
    et al.
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine).
    Karlsson, Richard
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine). Division of Experimental Cancer Research, Department of Translational Medicine, Clinical Research Centre, Lund University, Malmö, Sweden.
    Sarwar, Martuza
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine).
    Miftakhova, Regina R.
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine).
    Wang, Tianyan
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine).
    Khaja, Azharuddin Sajid Syed
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine).
    Semenas, Julius
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine).
    Chen, Sa
    Umeå University, Faculty of Science and Technology, Department of Molecular Biology (Faculty of Science and Technology).
    Hedblom, Andreas
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine). Division of Experimental Cancer Research, Department of Translational Medicine, Clinical Research Centre, Lund University, Malmö, Sweden.
    Amjad, Ali
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine).
    Ekström-Holka, Kristina
    Simoulis, Athanasios
    Kumar, Anjani
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine).
    Gjörloff Wingren, Anette
    Robinson, Brian
    Wai, Sun Nyunt
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine). Umeå University, Faculty of Medicine, Umeå Centre for Microbial Research (UCMR).
    Mongan, Nigel P.
    Heery, David M.
    Öhlund, Daniel
    Umeå University, Faculty of Medicine, Wallenberg Centre for Molecular Medicine at Umeå University (WCMM). Umeå University, Faculty of Medicine, Department of Radiation Sciences.
    Grundström, Thomas
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine).
    Ødum, Niels
    Persson, Jenny L.
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine). Division of Experimental Cancer Research, Department of Translational Medicine, Clinical Research Centre, Lund University, Malmö, Sweden; Department of Biomedical Sciences, Malmö University, Malmö, Sweden.
    FcγRIIIa receptor interacts with androgen receptor and PIP5K1α to promote growth and metastasis of prostate cancer2022In: Molecular Oncology, ISSN 1574-7891, E-ISSN 1878-0261Article in journal (Refereed)
    Abstract [en]

    Low-affinity immunoglobulin gamma Fc region receptor III-A (FcγRIIIa) is a cell surface protein that belongs to a family of Fc receptors that facilitate the protective function of the immune system against pathogens. However, the role of FcγRIIIa in prostate cancer (PCa) progression remained unknown. In this study, we found that FcγRIIIa expression was present in PCa cells and its level was significantly higher in metastatic lesions than in primary tumors from the PCa cohort (P = 0.006). PCa patients with an elevated level of FcγRIIIa expression had poorer biochemical recurrence (BCR)-free survival compared with those with lower FcγRIIIa expression, suggesting that FcγRIIIa is of clinical importance in PCa. We demonstrated that overexpression of FcγRIIIa increased the proliferative ability of PCa cell line C4-2 cells, which was accompanied by the upregulation of androgen receptor (AR) and phosphatidylinositol-4-phosphate 5-kinase alpha (PIP5Kα), which are the key players in controlling PCa progression. Conversely, targeted inhibition of FcγRIIIa via siRNA-mediated knockdown or using its inhibitory antibody suppressed growth of xenograft PC-3 and PC-3M prostate tumors and reduced distant metastasis in xenograft mouse models. We further showed that elevated expression of AR enhanced FcγRIIIa expression, whereas inhibition of AR activity using enzalutamide led to a significant downregulation of FcγRIIIa protein expression. Similarly, inhibition of PIP5K1α decreased FcγRIIIa expression in PCa cells. FcγRIIIa physically interacted with PIP5K1α and AR via formation of protein-protein complexes, suggesting that FcγRIIIa is functionally associated with AR and PIP5K1α in PCa cells. Our study identified FcγRIIIa as an important factor in promoting PCa growth and invasion. Further, the elevated activation of FcγRIII and AR and PIP5K1α pathways may cooperatively promote PCa growth and invasion. Thus, FcγRIIIa may serve as a potential new target for improved treatment of metastatic and castration-resistant PCa.

    Download full text (pdf)
    fulltext
  • 16. Fredholm, Simon
    et al.
    Litvinov, Ivan V
    Mongan, Nigel P
    Schiele, Sarah
    Willerslev-Olsen, Andreas
    Petersen, David Leander
    Krejsgaard, Thorbjørn
    Sibbesen, Nina
    Nastasi, Claudia
    Bonefeld, Charlotte M
    Persson, Jenny L.
    Clinical Research Center, Lund University, Malmö, Sweden.
    Straten, Per Thor
    Andersen, Mads Hald
    Koralov, Sergei B
    Wasik, Mariusz M
    Geisler, Carsten
    Sasseville, Denis
    Woetmann, Anders
    Ødum, Niels
    The Expression of IL-21 Is Promoted by MEKK4 in Malignant T Cells and Associated with Increased Progression Risk in Cutaneous T-Cell Lymphoma2016In: Journal of Investigative Dermatology, ISSN 0022-202X, E-ISSN 1523-1747, Vol. 136, no 4, p. 866-869Article in journal (Refereed)
  • 17. Fredholm, Simon
    et al.
    Willerslev-Olsen, Andreas
    Met, Özcan
    Kubat, Linda
    Gluud, Maria
    Mathiasen, Sarah L.
    Friese, Christina
    Blümel, Edda
    Petersen, David L.
    Hu, Tengpeng
    Nastasi, Claudia
    Lindahl, Lise M.
    Buus, Terkild B.
    Krejsgaard, Thorbjørn
    Wasik, Mariusz A.
    Kopp, Katharina L.
    Koralov, Sergei B.
    Persson, Jenny L.
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine). Division of Experimental Cancer Research, Department of Translational Medicine, Lund University, Clinical Research Centre, Malmö, Sweden.
    Bonefeld, Charlotte M.
    Geisler, Carsten
    Woetmann, Anders
    Iversen, Lars
    Becker, Jürgen C.
    Odum, Niels
    SATB1 in Malignant T Cells2018In: Journal of Investigative Dermatology, ISSN 0022-202X, E-ISSN 1523-1747, Vol. 138, no 8, p. 1805-1815Article in journal (Refereed)
    Abstract [en]

    Deficient expression of SATB1 hampers thymocyte development and results in inept T-cell lineages. Recent data implicate dysregulated SATB1 expression in the pathogenesis of mycosis fungoides, the most frequent variant of cutaneous T-cell lymphoma. Here, we report on a disease stage-associated decrease of SATB1 expression and an inverse expression of STAT5 and SATB1 in situ. STAT5 inhibited SATB1 expression through induction of microRNA-155. Decreased SATB1 expression triggered enhanced expression of IL-5 and IL-9 (but not IL-6 and IL-32), whereas increased SATB1 expression had the opposite effect, indicating that the microRNA-155 target SATB1 is a repressor of IL-5 and IL-9 in malignant T cells. In accordance, inhibition of STAT5 and its upstream activator JAK3 triggered increased SATB1 expression and a concomitant suppression of IL-5 and IL-9 expression in malignant T cells. In conclusion, we provide a mechanistic link between the proto-oncogenic JAK3/STAT5/microRNA-155 pathway, SATB1, and cytokines linked to CTCL severity and progression, indicating that SATB1 dysregulation is involved in cutaneous T-cell lymphoma pathogenesis.

  • 18. Fridberg, Marie
    et al.
    Kjellström, Sofia
    Anagnostaki, Lola
    Skogvall, Ingela
    Mustelin, Tomas
    Wiebe, Thomas
    Persson, Jenny L.
    Department of Pathology, Lund University, Malmö University Hospital, Malmö, Sweden.
    Dictor, Michael
    Wingren, Anette Gjörloff
    Immunohistochemical analyses of phosphatases in childhood B-cell lymphoma: lower expression of PTEN and HePTP and higher number of positive cells for nuclear SHP2 in B-cell lymphoma cases compared to controls2008In: Pediatric Hematology & Oncology, ISSN 0888-0018, E-ISSN 1521-0669, Vol. 25, no 6, p. 528-540Article in journal (Refereed)
    Abstract [en]

    Although many pediatric B-cell lymphoma patients are being cured today, much is still unknown about the pathogenesis of this disease. Protein tyrosine phosphatases are involved in the control of survival, growth, and differentiation of cells. The authors have analyzed 26 pediatric B-cell lymphoma cases for the expression of a panel of phosphatases and report a statistically significant lower expression intensity of PTEN and HePTP and higher nuclear SHP2 expression in B-cell lymphoma cases compared to lymphoid tissue. Knowledge about the expression of key regulatory proteins in pediatric B-cell lymphomas is necessary for revealing the complex molecular background of this disease.

  • 19. Fridberg, Marie
    et al.
    Servin, Anna
    Anagnostaki, Lola
    Linderoth, Johan
    Berglund, Mattias
    Söderberg, Ola
    Enblad, Gunilla
    Rosén, Anders
    Mustelin, Tomas
    Jerkeman, Mats
    Persson, Jenny L.
    Department of Pathology, Lund University, Malmö University Hospital, Malmö, Sweden.
    Wingren, Anette Gjörloff
    Protein expression and cellular localization in two prognostic subgroups of diffuse large B-cell lymphoma: Higher expression of ZAP70 and PKC-β II in the non-germinal center group and poor survival in patients deficient in nuclear PTEN2007In: Leukemia and Lymphoma, ISSN 1042-8194, E-ISSN 1029-2403, Vol. 48, no 11, p. 2221-2232Article in journal (Refereed)
    Abstract [en]

    Patients diagnosed with diffuse large B-cell lymphoma (DLBCL) show varying responses to conventional therapy, and this might be contributed to the differentiation stage of the tumor B-cells. The aim of the current study was to evaluate a panel of kinases (ZAP70, PKC-beta I and II and phosphorylated PKB/Akt) and phosphatases (PTEN, SHP1 and SHP2) known to be frequently deregulated in lymphoid malignancies. De novo DLBCL cases were divided into two subgroups, the germinal center (GC) group (14/28) and the non-germinal center (non-GC) or activated B-cell (ABC) group (14/28). ZAP70 and PKC-beta II were expressed in a significantly higher percentage of tumor cells in the clinically more aggressive non-GC group compared with the prognostically favourable GC group. Also, the subcellular localization of PKC-beta I and II differed in DLBCL cells, with the PKC-beta I isoform being expressed in both the cytoplasm and nucleus, while PKC-beta II was found exclusively in the cytoplasm. Loss of nuclear PTEN correlated with poor survival in cases from both subgroups. In addition, five cell lines of DLBCL origin were analyzed for protein expression and for mRNA levels of PTEN and SHP1. For the first time, we show that ZAP70 is expressed in a higher percentage of tumor cells in the aggressive non-GC subgroup of DLBCL and that PKC-beta I and II are differently distributed in the two prognostic subgroups of de novo DLBCL.

  • 20. Fulton, Joel
    et al.
    Mazumder, Bismoy
    Whitchurch, Jonathan B.
    Monteiro, Cintia J.
    Collins, Hilary M.
    Chan, Chun M.
    Clemente, Maria P.
    Hernandez-Quiles, Miguel
    Stewart, Elizabeth A.
    Amoaku, Winfried M.
    Moran, Paula M.
    Mongan, Nigel P.
    Persson, Jenny L.
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine). Division of Experimental Cancer Research, Department of Translational Medicine, Lund University, Clinical Research Centre, Malmö, Sweden.
    Ali, Simak
    Heery, David M.
    Heterodimers of photoreceptor-specific nuclear receptor (PNR/NR2E3) and peroxisome proliferator-activated receptor-gamma (PPAR gamma) are disrupted by retinal disease-associated mutations2017In: Cell Death and Disease, E-ISSN 2041-4889, Vol. 8, article id e2677Article in journal (Refereed)
    Abstract [en]

    Photoreceptor-specific nuclear receptor (PNR/NR2E3) and Tailless homolog (TLX/NR2E1) are human orthologs of the NR2E group, a subgroup of phylogenetically related members of the nuclear receptor (NR) superfamily of transcription factors. We assessed the ability of these NRs to form heterodimers with other members of the human NRs representing all major subgroups. The TLX ligand-binding domain (LBD) did not appear to form homodimers or interact directly with any other NR tested. The PNR LBD was able to form homodimers, but also exhibited robust interactions with the LBDs of peroxisome proliferator-activated receptor-gamma (PPAR gamma)/NR1C3 and thyroid hormone receptor b (TRb) TR beta/NR1A2. The binding of PNR to PPAR. was specific for this paralog, as no interaction was observed with the LBDs of PPAR alpha/NR1C1 or PPAR delta/NR1C2. In support of these findings, PPAR. and PNR were found to be co-expressed in human retinal tissue extracts and could be co-immunoprecipitated as a native complex. Selected sequence variants in the PNR LBD associated with human retinopathies, or a mutation in the dimerization region of PPAR. LBD associated with familial partial lipodystrophy type 3, were found to disrupt PNR/PPAR gamma complex formation. Wild-type PNR, but not a PNR309G mutant, was able to repress PPAR gamma-mediated transcription in reporter assays. In summary, our results reveal novel heterodimer interactions in the NR superfamily, suggesting previously unknown functional interactions of PNR with PPAR. and TR beta that have potential importance in retinal development and disease.

    Download full text (pdf)
    fulltext
  • 21. Gauffin, Fredrika
    et al.
    Diffner, Eva
    Gustafsson, Bertil
    Nordgren, Ann
    Wingren, Anette Gjörloff
    Sander, Birgitta
    Persson, Jenny L.
    Gustafsson, Britt
    Expression of PTEN and SHP1, investigated from tissue microarrays in pediatric acute lymphoblastic, leukemia2009In: Pediatric Hematology & Oncology, ISSN 0888-0018, E-ISSN 1521-0669, Vol. 26, no 1, p. 48-56Article in journal (Refereed)
    Abstract [en]

    PTEN and SHP1 are tumor suppressor genes involved in the regulation of cell cycle control and apoptosis. The authors investigated the protein expression of PTEN and SHP1, by immunohistochemistry in tissue microarrays from bone marrow samples in children, diagnosed with acute lymphoblastic leukaemia and nonmalignant controls. PTEN was overexpressed in diagnostic ALL samples, while SHP1 showed a low expression. Both proteins showed a significant difference in expression compared to nonmalignant controls. The roles of PTEN and SHP1 are not well investigated in pediatric leukemia and could in the future play a role as prognostic factors.

  • 22. Gluud, Maria
    et al.
    Willerslev-Olsen, Andreas
    Gjerdrum, Lise Mette Rahbek
    Lindahl, Lise M.
    Buus, Terkild B.
    Andersen, Mads Hald
    Bonefeld, Charlotte Menne
    Krejsgaard, Thorbjorn
    Litvinov, Ivan V.
    Iversen, Lars
    Becker, Jürgen C.
    Persson, Jenny L.
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine).
    Koralov, Sergei B.
    Litman, Thomas
    Geisler, Carsten
    Woetmann, Anders
    Odum, Niels
    MicroRNAs in the Pathogenesis, Diagnosis, Prognosis and Targeted Treatment of Cutaneous T-Cell Lymphomas2020In: Cancers, ISSN 2072-6694, Vol. 12, no 5Article, review/survey (Refereed)
    Abstract [en]

    Cutaneous T-cell lymphoma (CTCL) represents a heterogeneous group of potentially devastating primary skin malignancies. Despite decades of intense research efforts, the pathogenesis is still not fully understood. In the early stages, both clinical and histopathological diagnosis is often difficult due to the ability of CTCL to masquerade as benign skin inflammatory dermatoses. Due to a lack of reliable biomarkers, it is also difficult to predict which patients will respond to therapy or progress towards severe recalcitrant disease. In this review, we discuss recent discoveries concerning dysregulated microRNA (miR) expression and putative pathological roles of oncogenic and tumor suppressive miRs in CTCL. We also focus on the interplay between miRs, histone deacetylase inhibitors, and oncogenic signaling pathways in malignant T cells as well as the impact of miRs in shaping the inflammatory tumor microenvironment. We highlight the potential use of miRs as diagnostic and prognostic markers, as well as their potential as therapeutic targets. Finally, we propose that the combined use of miR-modulating compounds with epigenetic drugs may provide a novel avenue for boosting the clinical efficacy of existing anti-cancer therapies in CTCL.

    Download full text (pdf)
    fulltext
  • 23. Gomzikova, M. O.
    et al.
    Aimaletdinov, A. M.
    Bondar, O. V.
    Starostina, I. G.
    Gorshkova, N. V.
    Neustroeva, O. A.
    Kletukhina, S. K.
    Kurbangaleeva, S. V.
    Vorobev, V. V.
    Garanina, E. E.
    Persson, Jenny L.
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine).
    Jeyapalan, J.
    Mongan, N. P.
    Khaiboullina, S. F.
    Rizvanov, A. A.
    Immunosuppressive properties of cytochalasin B-induced membrane vesicles of mesenchymal stem cells: comparing with extracellular vesicles derived from mesenchymal stem cells2020In: Scientific Reports, E-ISSN 2045-2322, Vol. 10, no 1, article id 10740Article in journal (Refereed)
    Abstract [en]

    Extracellular vesicles derived from mesenchymal stem cells (MSCs) represent a novel approach for regenerative and immunosuppressive therapy. Recently, cytochalasin B-induced microvesicles (CIMVs) were shown to be effective drug delivery mediators. However, little is known about their immunological properties. We propose that the immunophenotype and molecular composition of these vesicles could contribute to the therapeutic efficacy of CIMVs. To address this issue, CIMVs were generated from murine MSC (CIMVs-MSCs) and their cytokine content and surface marker expression determined. For the first time, we show that CIMVs-MSCs retain parental MSCs phenotype (Sca-1(+), CD49e(+), CD44(+), CD45(-)). Also, CIMVs-MSCs contained a cytokine repertoire reflective of the parental MSCs, including IL-1 beta, IL-2, IL-3, IL-4, IL-5, IL-6, IL-9, IL-10, IL-12(p40), IL-13, IL-17, CCL2, CCL3, CCL4, CCL5, CCL11, G-CSF, GM-CSF and TNF-alpha. Next, we evaluated the immune-modulating properties of CIMVs-MSCs in vivo using standard preclinical tests. MSCs and CIMVs-MSCs reduced serum levels of anti-sheep red blood cell antibody and have limited effects on neutrophil and peritoneal macrophage activity. We compared the immunomodulatory effect of MSCs, CIMVs and EVs. We observed no immunosuppression in mice pretreated with natural EVs, whereas MSCs and CIMVs-MSCs suppressed antibody production in vivo. Additionally, we have investigated the biodistribution of CIMVs-MSCs in vivo and demonstrated that CIMVs-MSCs localized in liver, lung, brain, heart, spleen and kidneys 48 h after intravenous injection and can be detected 14 days after subcutaneous and intramuscular injection. Collectively our data demonstrates immunomodulatory efficacy of CIMVs and supports their further preclinical testing as an effective therapeutic delivery modality.

    Download full text (pdf)
    fulltext
  • 24. Gomzikova, Marina O.
    et al.
    Zhuravleva, Margarita N.
    Miftakhova, Regina R.
    Arkhipova, Svetlana S.
    Evtugin, Vladimir G.
    Khaiboullina, Svetlana F.
    Kiyasov, Andrey P.
    Persson, Jenny L.
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine).
    Mongan, Nigel P.
    Pestell, Richard G.
    Rizvanov, Albert A.
    Cytochalasin B-induced membrane vesicles convey angiogenic activity of parental cells2017In: Oncotarget, E-ISSN 1949-2553, Vol. 8, no 41, p. 70496-70507Article in journal (Refereed)
    Abstract [en]

    Naturally occurring extracellular vesicles (EVs) play essential roles in intracellular communication and delivery of bioactive molecules. Therefore it has been suggested that EVs could be used for delivery of therapeutics. However, to date the therapeutic application of EVs has been limited by number of factors, including limited yield and full understanding of their biological activities. To address these issues, we analyzed the morphology, molecular composition, fusion capacity and biological activity of Cytochalasin B-induced membrane vesicles (CIMVs). The size of these vesicles was comparable to that of naturally occurring EVs. In addition, we have shown that CIMVs from human SH-SY5Y cells contain elevated levels of VEGF as compared to the parental cells, and stimulate angiogenesis in vitro and in vivo.

    Download full text (pdf)
    fulltext
  • 25.
    Guo, Jinan
    et al.
    Department of Urology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China; Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People’s Hospital, Shenzhen, China; Shenzhen Urology Minimally Invasive Engineering Center, Shenzhen, China; Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosis, Clinical Medicine Research Centre, Shenzhen, China; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China.
    Gu, Liangyou
    Department of Urology, The Third Medical Centre, Chinese PLA General Hospital, Beijing, China.
    Johnson, Heather
    Olympia Diagnostics, Inc., CA, Sunnyvale, United States.
    Gu, Di
    Department of Urology, The First affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
    Lu, Zhenquan
    The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
    Luo, Binfeng
    The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
    Yuan, Qian
    Department of Urology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China.
    Zhang, Xuhui
    Department of Bio-diagnosis, Institute of Basic Medical Sciences, Beijing, China.
    Xia, Taolin
    Department of Urology, Foshan First People’s Hospital, Foshan, China.
    Zeng, Qingsong
    Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
    Wu, Alan H. B.
    Clinical Laboratories, San Francisco General Hospital, CA, San Francisco, United States.
    Johnson, Allan
    Kinetic Reality, Santa Clara, United States.
    Dizeyi, Nishtman
    Department of Translational Medicine, Lund University, Clinical Research Centre, Malmö, Sweden.
    Abrahamsson, Per-Anders
    Department of Translational Medicine, Lund University, Clinical Research Centre, Malmö, Sweden.
    Zhang, Heqiu
    Department of Bio-diagnosis, Institute of Basic Medical Sciences, Beijing, China.
    Chen, Lingwu
    Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
    Xiao, Kefeng
    Department of Urology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China.
    Zou, Chang
    Department of Urology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China; Shenzhen Urology Minimally Invasive Engineering Center, Shenzhen, China; Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosis, Clinical Medicine Research Centre, Shenzhen, China; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China; Key Laboratory of Medical Electrophysiology of Education Ministry, School of Pharmacy, Southwest Medical University, Luzhou, China.
    Persson, Jenny L.
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine). Department of Biomedical Sciences, Malmö University, Malmö, Sweden.
    A non-invasive 25-Gene PLNM-Score urine test for detection of prostate cancer pelvic lymph node metastasis2024In: Prostate Cancer and Prostatic Diseases, ISSN 1365-7852, E-ISSN 1476-5608Article in journal (Refereed)
    Abstract [en]

    Background: Prostate cancer patients with pelvic lymph node metastasis (PLNM) have poor prognosis. Based on EAU guidelines, patients with >5% risk of PLNM by nomograms often receive pelvic lymph node dissection (PLND) during prostatectomy. However, nomograms have limited accuracy, so large numbers of false positive patients receive unnecessary surgery with potentially serious side effects. It is important to accurately identify PLNM, yet current tests, including imaging tools are inaccurate. Therefore, we intended to develop a gene expression-based algorithm for detecting PLNM.

    Methods: An advanced random forest machine learning algorithm screening was conducted to develop a classifier for identifying PLNM using urine samples collected from a multi-center retrospective cohort (n = 413) as training set and validated in an independent multi-center prospective cohort (n = 243). Univariate and multivariate discriminant analyses were performed to measure the ability of the algorithm classifier to detect PLNM and compare it with the Memorial Sloan Kettering Cancer Center (MSKCC) nomogram score.

    Results: An algorithm named 25 G PLNM-Score was developed and found to accurately distinguish PLNM and non-PLNM with AUC of 0.93 (95% CI: 0.85–1.01) and 0.93 (95% CI: 0.87–0.99) in the retrospective and prospective urine cohorts respectively. Kaplan–Meier plots showed large and significant difference in biochemical recurrence-free survival and distant metastasis-free survival in the patients stratified by the 25 G PLNM-Score (log rank P < 0.001 and P < 0.0001, respectively). It spared 96% and 80% of unnecessary PLND with only 0.51% and 1% of PLNM missing in the retrospective and prospective cohorts respectively. In contrast, the MSKCC score only spared 15% of PLND with 0% of PLNM missing.

    Conclusions: The novel 25 G PLNM-Score is the first highly accurate and non-invasive machine learning algorithm-based urine test to identify PLNM before PLND, with potential clinical benefits of avoiding unnecessary PLND and improving treatment decision-making.

    Download full text (pdf)
    fulltext
  • 26.
    Guo, Jinan
    et al.
    Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen Urology Minimally Invasive Engineering Centre, Shenzhen, China; Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosis, Clinical Medical Research Centre, Shenzhen, China.
    Johnson, Heather
    Olympia Diagnostics, Inc., Sunnyvale, California.
    Zhang, Xuhui
    Department of Bio-Diagnosis, Institute of Basic Medical Sciences, Beijing, China.
    Feng, Xiaoyan
    Department of Bio-Diagnosis, Institute of Basic Medical Sciences, Beijing, China.
    Zhang, Heqiu
    Department of Bio-Diagnosis, Institute of Basic Medical Sciences, Beijing, China.
    Simoulis, Athanasios
    Department of Clinical Pathology and Cytology, Skåne University Hospital, Malmö, Sweden.
    Wu, Alan HB
    Clinical Laboratories, San Francisco General Hospital, San Francisco, California.
    Xia, Taolin
    Department of Urology, Foshan First People's Hospital, Foshan, China.
    Li, Fei
    Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
    Tan, Wanlong
    Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
    Johnson, Allan
    Kinetic Reality, Santa Clara, California.
    Dizeyi, Nishtman
    Department of Translational Medicine, Lund University, Clinical Research Centre, Malmö, Sweden.
    Abrahamsson, Per-Anders
    Department of Translational Medicine, Lund University, Clinical Research Centre, Malmö, Sweden.
    Kenner, Lukas
    Department of Experimental Pathology, Medical University Vienna & Unit of Laboratory Animal Pathology, University of Veterinary Medicine, Vienna, Austria.
    Chen, Lingwu
    Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
    Zhong, Wanmei
    Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
    Xiao, Kefeng
    Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen Urology Minimally Invasive Engineering Centre, Shenzhen, China; Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosis, Clinical Medical Research Centre, Shenzhen, China.
    Persson, Jenny L.
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine). Department of Biomedical Sciences, Malmö University, Malmö, Sweden; Division of Experimental Cancer Research, Department of Translational Medicine, Lund University, Malmö, Sweden.
    Zou, Chang
    Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen Urology Minimally Invasive Engineering Centre, Shenzhen, China; Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosis, Clinical Medical Research Centre, Shenzhen, China.
    A 23-Gene Classifier urine test for prostate cancer prognosis2021In: Clinical and translational medicine, ISSN 2001-1326, Vol. 11, no 3, article id e340Article in journal (Other academic)
    Download full text (pdf)
    fulltext
  • 27. Guo, Jinan
    et al.
    Liu, Dale
    Zhang, Xuhui
    Johnson, Heather
    Feng, Xiaoyan
    Zhang, Heqiu
    Wu, Alan H. B.
    Chen, Lingwu
    Fang, Jiequn
    Xiao, Zhangang
    Xiao, Kefeng
    Persson, Jenny L.
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine). Division of Experimental Cancer Research, Department of Translational Medicine, Lund University, Malmö, Sweden.
    Zou, Chang
    Establishing a Urine-Based Biomarker Assay for Prostate Cancer Risk Stratification2020In: Frontiers in Cell and Developmental Biology, E-ISSN 2296-634X, Vol. 8, article id 597961Article in journal (Refereed)
    Abstract [en]

    One of the major features of prostate cancer (PCa) is its heterogeneity, which often leads to uncertainty in cancer diagnostics and unnecessary biopsies as well as overtreatment of the disease. Novel non-invasive tests using multiple biomarkers that can identify clinically high-risk cancer patients for immediate treatment and monitor patients with low-risk cancer for active surveillance are urgently needed to improve treatment decision and cancer management. In this study, we identified 14 promising biomarkers associated with PCa and tested the performance of these biomarkers on tissue specimens and pre-biopsy urinary sediments. These biomarkers showed differential gene expression in higher- and lower-risk PCa. The 14-Gene Panel urine test (PMP22, GOLM1, LMTK2, EZH2, GSTP1, PCA3, VEGFA, CST3, PTEN, PIP5K1A, CDK1, TMPRSS2, ANXA3, and CCND1) was assessed in two independent prospective and retrospective urine study cohorts and showed high diagnostic accuracy to identify higher-risk PCa patients with the need for treatment and lower-risk patients for surveillance. The AUC was 0.897 (95% CI 0.939–0.855) in the prospective cohort (n = 202), and AUC was 0.899 (95% CI 0.964–0.834) in the retrospective cohort (n = 97). In contrast, serum PSA and Gleason score had much lower accuracy in the same 202 patient cohorts [AUC was 0.821 (95% CI 0.879–0.763) for PSA and 0.860 (95% CI 0.910–0.810) for Gleason score]. In addition, the 14-Gene Panel was more accurate at risk stratification in a subgroup of patients with Gleason scores 6 and 7 in the prospective cohort (n = 132) with AUC of 0.923 (95% CI 0.968–0.878) than PSA [AUC of 0.773 (95% CI 0.852–0.794)] and Gleason score [AUC of 0.776 (95% CI 0.854–0.698)]. Furthermore, the 14-Gene Panel was found to be able to accurately distinguish PCa from benign prostate with AUC of 0.854 (95% CI 0.892–0.816) in a prospective urine study cohort (n = 393), while PSA had lower accuracy with AUC of 0.652 (95% CI 0.706–0.598). Taken together, the 14-Gene Panel urine test represents a promising non-invasive tool for detection of higher-risk PCa to aid treatment decision and lower-risk PCa for active surveillance.

    Download full text (pdf)
    fulltext
  • 28. Guo, Jinan
    et al.
    Yang, Jianggen
    Zhang, Xuhui
    Feng, Xiaoyan
    Zhang, Heqiu
    Chen, Lingwu
    Johnson, Heather
    Persson, Jenny L.
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine). Department of Translational Medicine, Lund University, Malmö, Sweden.
    Xiao, Kefeng
    A Panel of Biomarkers for Diagnosis of Prostate Cancer Using Urine Samples2018In: Anticancer Research, ISSN 0250-7005, E-ISSN 1791-7530, Vol. 38, no 3, p. 1471-1477Article in journal (Refereed)
    Abstract [en]

    Background/Aim: Prostate cancer (PCa) diagnosis using patient urine samples represents a non-invasive and more convenient method than the conventional biopsy and prostate-specific antigen (PSA) test. This study intended to identify a biomarker panel to distinguish PCa from benign prostate using urine samples. Materials and Methods: We identified six biomarkers with differential gene expression in 154 PCa and benign prostate specimens. We then determined mRNA expression signature and the diagnostic performance of the 6-biomarker panel in 156 urine samples from patients with PCa and benign disease. Results: The 6-biomarker panel distinguished PCa from benign prostate cases with sensitivity of 80.6%, specificity of 62.9% and area under the curve (AUC) of 0.803 (p<0.0001), whereas serum PSA at 4 ng/ml cutoff had sensitivity of 95.5%, specificity of 20.2% and AUC of 0.521 (p<0.0001). Conclusion: The 6-biomarker panel for use in urine samples was able to distinguish PCa from benign prostate with higher specificity and accuracy than PSA and may be useful in clinical settings.

  • 29. Guo, Jinan
    et al.
    Zhang, Xuhui
    Xia, Taolin
    Johnson, Heather
    Feng, Xiaoyan
    Simoulis, Athanasios
    Wu, Alan H. B.
    Li, Fei
    Tan, Wanlong
    Johnson, Allan
    Dizeyi, Nishtman
    Abrahamsson, Per-Anders
    Kenner, Lukas
    Department of Experimental Pathology, Medical University Vienna & Unit of Laboratory Animal Pathology, University of Veterinary Medicine, Vienna, Austria.
    Xiao, Kefeng
    Zhang, Heqiu
    Chen, Lingwu
    Zou, Chang
    Persson, Jenny L.
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine). Department of Biomedical Sciences, Malmö University, Malmö, Sweden; Division of Experimental Cancer Research, Department of Translational Medicine, Lund University, Malmö, Sweden.
    Non-invasive urine test for molecular classification of clinical significance in newly diagnosed prostate cancer patients2021In: Frontiers in Medicine, E-ISSN 2296-858X, Vol. 8, article id 721554Article in journal (Refereed)
    Abstract [en]

    Objective: To avoid over-treatment of low-risk prostate cancer patients, it is important to identify clinically significant and insignificant cancer for treatment decision-making. However, no accurate test is currently available.

    Methods: To address this unmet medical need, we developed a novel gene classifier to distinguish clinically significant and insignificant cancer, which were classified based on the National Comprehensive Cancer Network risk stratification guidelines. A non-invasive urine test was developed using quantitative mRNA expression data of 24 genes in the classifier with an algorithm to stratify the clinical significance of the cancer. Two independent, multicenter, retrospective and prospective studies were conducted to assess the diagnostic performance of the 24-Gene Classifier and the current clinicopathological measures by univariate and multivariate logistic regression and discriminant analysis. In addition, assessments were performed in various Gleason grades/ISUP Grade Groups.

    Results: The results showed high diagnostic accuracy of the 24-Gene Classifier with an AUC of 0.917 (95% CI 0.892-0.942) in the retrospective cohort (n = 520), AUC of 0.959 (95% CI 0.935-0.983) in the prospective cohort (n = 207), and AUC of 0.930 (95% 0.912-CI 0.947) in the combination cohort (n = 727). Univariate and multivariate analysis showed that the 24-Gene Classifier was more accurate than cancer stage, Gleason score, and PSA, especially in the low/intermediate-grade/ISUP Grade Group 1-3 cancer subgroups.

    Conclusions: The 24-Gene Classifier urine test is an accurate and non-invasive liquid biopsy method for identifying clinically significant prostate cancer in newly diagnosed cancer patients. It has the potential to improve prostate cancer treatment decisions and active surveillance.

    Download full text (pdf)
    fulltext
  • 30.
    Haigh, Daisy B.
    et al.
    Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom; School of Veterinary Medicine and Sciences, University of Nottingham, Sutton Bonington, United Kingdom.
    Woodcock, Corinne L.
    Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom; School of Veterinary Medicine and Sciences, University of Nottingham, Sutton Bonington, United Kingdom.
    Lothion-Roy, Jennifer
    Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom; School of Veterinary Medicine and Sciences, University of Nottingham, Sutton Bonington, United Kingdom.
    Harris, Anna E.
    Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom; School of Veterinary Medicine and Sciences, University of Nottingham, Sutton Bonington, United Kingdom.
    Metzler, Veronika M.
    Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom; School of Veterinary Medicine and Sciences, University of Nottingham, Sutton Bonington, United Kingdom.
    Persson, Jenny L.
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine). Department of Biomedical Sciences, Malmö Universitet, Malmö, Sweden.
    Robinson, Brian D.
    Department of Pathology, Weill Cornell Medicine, NY, New York, United States.
    Khani, Francesca
    Department of Pathology, Weill Cornell Medicine, NY, New York, United States.
    Alsaleem, Mansour
    Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom; School of Medicine, University of Nottingham, Nottingham, United Kingdom; Department of Applied Medical Science, Applied College, Qassim University, Qassim, Unayzah, Saudi Arabia.
    Ntekim, Atara
    School of Veterinary Medicine and Sciences, University of Nottingham, Sutton Bonington, United Kingdom; Department of Radiation Oncology, University College Hospital, University of Ibadan, Ibadan, Nigeria.
    Madhusudan, Srinivasan
    Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom; School of Medicine, University of Nottingham, Nottingham, United Kingdom.
    Davis, Melissa B.
    Department of Surgery, Weill Cornell Medicine, NY, New York, United States.
    Laursen, Kristian B.
    Department of Pharmacology, Weill Cornell Medicine, NY, New York, United States.
    Gudas, Lorraine J.
    Department of Pharmacology, Weill Cornell Medicine, NY, New York, United States.
    Rutland, Catrin S.
    School of Veterinary Medicine and Sciences, University of Nottingham, Sutton Bonington, United Kingdom.
    Toss, Michael S.
    Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom; School of Medicine, University of Nottingham, Nottingham, United Kingdom.
    Archer, Nathan
    School of Veterinary Medicine and Sciences, University of Nottingham, Sutton Bonington, United Kingdom.
    Bodi, Zsuzsanna
    School of Biosciences, University of Nottingham, Sutton Bonington, United Kingdom.
    Rakha, Emad A.
    School of Medicine, University of Nottingham, Nottingham, United Kingdom.
    Fray, Rupert G.
    School of Biosciences, University of Nottingham, Sutton Bonington, United Kingdom.
    Jeyapalan, Jennie N.
    Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom; School of Veterinary Medicine and Sciences, University of Nottingham, Sutton Bonington, United Kingdom.
    Mongan, Nigel P.
    Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom; School of Veterinary Medicine and Sciences, University of Nottingham, Sutton Bonington, United Kingdom; Department of Pharmacology, Weill Cornell Medicine, NY, New York, United States.
    The METTL3 RNA Methyltransferase Regulates Transcriptional Networks in Prostate Cancer2022In: Cancers, ISSN 2072-6694, Vol. 14, no 20, article id 5148Article in journal (Refereed)
    Abstract [en]

    Prostate cancer (PCa) is a leading cause of cancer-related deaths and is driven by aberrant androgen receptor (AR) signalling. For this reason, androgen deprivation therapies (ADTs) that suppress androgen-induced PCa progression either by preventing androgen biosynthesis or via AR signalling inhibition (ARSi) are common treatments. The N6-methyladenosine (m6A) RNA modification is involved in regulating mRNA expression, translation, and alternative splicing, and through these mechanisms has been implicated in cancer development and progression. RNA-m6A is dynamically regulated by the METTL3 RNA methyltransferase complex and the FTO and ALKBH5 demethylases. While there is evidence supporting a role for aberrant METTL3 in many cancer types, including localised PCa, the wider contribution of METTL3, and by inference m6A, in androgen signalling in PCa remains poorly understood. Therefore, the aim of this study was to investigate the expression of METTL3 in PCa patients and study the clinical and functional relevance of METTL3 in PCa. It was found that METTL3 is aberrantly expressed in PCa patient samples and that siRNA-mediated METTL3 knockdown or METTL3-pharmacological inhibition significantly alters the basal and androgen-regulated transcriptome in PCa, which supports targeting m6A as a novel approach to modulate androgen signalling in PCa.

    Download full text (pdf)
    fulltext
  • 31. Hao, Wende
    et al.
    Zhang, Xuhui
    Xiu, Bingshui
    Yang, Xiqin
    Hu, Shuofeng
    Liu, Zhiqiang
    Duan, Cuimi
    Jin, Shujuan
    Ying, Xiaomin
    Zhao, Yanfeng
    Han, Xiaowei
    Hao, Xiaopeng
    Fan, Yawen
    Johnson, Heather
    Meng, Di
    Persson, Jenny L.
    Division of Experimental Cancer Research, Department of Laboratory Medicine in Malmö, Clinical Research Center, Lund University, Malmö, Sweden.
    Zhang, Heqiu
    Feng, XiaoYan
    Huang, Yan
    Vitronectin: a promising breast cancer serum biomarker for early diagnosis of breast cancer in patients2016In: Tumor Biology, ISSN 1010-4283, E-ISSN 1423-0380, Vol. 37, no 7, p. 8909-8916Article in journal (Refereed)
    Abstract [en]

    Breast cancer is the most common cancer in women worldwide, identification of new biomarkers for early diagnosis and detection will improve the clinical outcome of breast cancer patients. In the present study, we determined serum levels of vitronectin (VN) in 93 breast cancer patients, 30 benign breast lesions, 9 precancerous lesions, and 30 healthy individuals by enzyme-linked immunosorbent assays. Serum VN level was significantly higher in patients with stage 0-I primary breast cancer than in healthy individuals, patients with benign breast lesion or precancerous lesions, as well as those with breast cancer of higher stages. Serum VN level was significantly and negatively correlated with tumor size, lymph node status, and clinical stage (p < 0.05 in all cases). In addition, VN displayed higher area under curve (AUC) value (0.73, 95 % confidence interval (CI) [0.62-0.84]) than carcinoembryonic antigen (CEA) (0.64, 95 % CI [0.52-0.77]) and cancer antigen 15-3 (CA 15-3) (0.69, 95 % CI [0.58-0.81]) when used to distinguish stage 0-I cancer and normal control. Importantly, the combined use of three biomarkers yielded an improvement in receiver operating characteristic curve with an AUC of 0.83, 95 % CI [0.74-0.92]. Taken together, our current study showed for the first time that serum VN is a promising biomarker for early diagnosis of breast cancer when combined with CEA and CA15-3.

  • 32.
    Harris, Anna E.
    et al.
    University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom.
    Metzler, Veronika M.
    University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom.
    Lothion-Roy, Jennifer
    University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom.
    Varun, Dhruvika
    University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom.
    Woodcock, Corinne L.
    University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom.
    Haigh, Daisy B.
    University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom.
    Endeley, Chantelle
    University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom.
    Haque, Maria
    University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom.
    Toss, Michael S.
    University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom.
    Alsaleem, Mansour
    University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom; Department of Applied Medical Science, Applied College, Qassim University, Qassim, Saudi Arabia.
    Persson, Jenny L.
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine). Department of Biomedical Sciences, Malmö Universitet, Malmö, Sweden.
    Gudas, Lorraine J.
    Department of Pharmacology, Weill Cornell Medicine, NY, New York, United States.
    Rakha, Emad
    University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom.
    Robinson, Brian D.
    Department of Urology, Weill Cornell Medicine, NY, New York, United States.
    Khani, Francesca
    Department of Urology, Weill Cornell Medicine, NY, New York, United States.
    Martin, Laura M.
    Englander Institute for Precision Medicine, Weill Cornell Medicine, NY, New York, United States.
    Moyer, Jenna E.
    Englander Institute for Precision Medicine, Weill Cornell Medicine, NY, New York, United States.
    Brownlie, Juliette
    University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom.
    Madhusudan, Srinivasan
    University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom.
    Allegrucci, Cinzia
    University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom.
    James, Victoria H.
    University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom.
    Rutland, Catrin S.
    University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom.
    Fray, Rupert G.
    School of Biosciences, University of Nottingham, Nottingham, United Kingdom.
    Ntekim, Atara
    Department of Oncology, University Hospital Ibadan, Ibadan, Nigeria.
    de Brot, Simone
    Comparative Pathology Platform (COMPATH), Institute of Animal Pathology, University of Bern, Bern, Switzerland.
    Mongan, Nigel P.
    University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom; Department of Pharmacology, Weill Cornell Medicine, NY, New York, United States.
    Jeyapalan, Jennie N.
    University of Nottingham Biodiscovery Institute, University of Nottingham, University Park, Nottingham, United Kingdom.
    Exploring anti-androgen therapies in hormone dependent prostate cancer and new therapeutic routes for castration resistant prostate cancer2022In: Frontiers in Endocrinology, E-ISSN 1664-2392, Vol. 13, article id 1006101Article, review/survey (Refereed)
    Abstract [en]

    Androgen deprivation therapies (ADTs) are important treatments which inhibit androgen-induced prostate cancer (PCa) progression by either preventing androgen biosynthesis (e.g. abiraterone) or by antagonizing androgen receptor (AR) function (e.g. bicalutamide, enzalutamide, darolutamide). A major limitation of current ADTs is they often remain effective for limited durations after which patients commonly progress to a lethal and incurable form of PCa, called castration-resistant prostate cancer (CRPC) where the AR continues to orchestrate pro-oncogenic signalling. Indeed, the increasing numbers of ADT-related treatment-emergent neuroendocrine-like prostate cancers (NePC), which lack AR and are thus insensitive to ADT, represents a major therapeutic challenge. There is therefore an urgent need to better understand the mechanisms of AR action in hormone dependent disease and the progression to CRPC, to enable the development of new approaches to prevent, reverse or delay ADT-resistance. Interestingly the AR regulates distinct transcriptional networks in hormone dependent and CRPC, and this appears to be related to the aberrant function of key AR-epigenetic coregulator enzymes including the lysine demethylase 1 (LSD1/KDM1A). In this review we summarize the current best status of anti-androgen clinical trials, the potential for novel combination therapies and we explore recent advances in the development of novel epigenetic targeted therapies that may be relevant to prevent or reverse disease progression in patients with advanced CRPC.

    Download full text (pdf)
    fulltext
  • 33.
    Hedblom, Andreas
    et al.
    Umeå University, Faculty of Medicine, Department of Molecular Biology (Faculty of Medicine). Department of Surgery, Cancer Research Institute and Transplant Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Department of Translational Medicine, Lund University, Lund, Sweden.
    Hejazi, Seyed M.