Network


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

Hotspot


Dive into the research topics where Margareta Berggren is active.

Publication


Featured researches published by Margareta Berggren.


Molecular Cancer Therapeutics | 2005

The phosphatidylinositol-3-kinase inhibitor PX-866 overcomes resistance to the epidermal growth factor receptor inhibitor gefitinib in A-549 human non–small cell lung cancer xenografts

Nathan T. Ihle; Gillian Paine-Murrieta; Margareta Berggren; Amanda F. Baker; Wendy R. Tate; Peter Wipf; Robert T. Abraham; D. Lynn Kirkpatrick; Garth Powis

Epidermal growth factor receptor (EGFR) inhibitors such as gefitinib show antitumor activity in a subset of non–small cell lung cancer (NSCLC) patients having mutated EGFR. Recent work shows that phosphatidylinositol-3-kinase (PI3-K) is coupled to the EGFR only in NSCLC cell lines expressing ErbB-3 and that EGFR inhibitors do not inhibit PI3-K signaling in these cells. The central role PI3-K plays in cell survival suggests that a PI3-K inhibitor offers a strategy to increase the antitumor activity of EGFR inhibitors in resistant NSCL tumors that do not express ErbB-3. We show that PX-866, a PI3-K inhibitor with selectivity for p110α, potentiates the antitumor activity of gefitinib against even large A-549 NSCL xenografts giving complete tumor growth control in the early stages of treatment. A-549 xenograft phospho-Akt was inhibited by PX-866 but not by gefitinib. A major toxicity of PX-866 administration was hyperglycemia with decreased glucose tolerance, which was reversed upon cessation of treatment. The decreased glucose tolerance caused by PX-866 was insensitive to the AMP-activated protein kinase inhibitor metformin but reversed by insulin and by the peroxisome proliferator-activated receptor-γ activator pioglitazone. Prolonged PX-866 administration also caused increased neutrophil counts. Thus, PX-866, by inhibiting PI3-K signaling, may have clinical use in increasing the response to EGFR inhibitors such as gefitinib in patients with NSCLC and possibly in other cancers who do not respond to EGFR inhibition.


Biochemical Pharmacology | 1998

Mechanisms of Inhibition of the Thioredoxin Growth Factor System by Antitumor 2-Imidazolyl Disulfides

D. Lynn Kirkpatrick; Miles Kuperus; Marla Dowdeswell; Noelle Potier; Lynda J. Donald; Mark Kunkel; Margareta Berggren; Miguel Angulo; Garth Powis

The interactions of a series of 2-imidazolyl disulfide antitumor compounds with the thioredoxin reductase(TR)/thioredoxin (hTrx) redox system have been studied. Disulfides III-2 (n-butyl 2-mercaptoimidazolyl disulfide) and VI-2 (ethyl 2-mercaptoimidazolyl disulfide) were substrates for reduction by TR with Km values of 43 and 48 microM. Disulfides IV-2 (1-methylpropyl 2-mercaptoimidazolyl disulfide) and DLK-36 (benzyl 2-mercaptoimidazolyl disulfide) were competitive inhibitors of the reduction of hTrx by TR with Ki values of 31 microM. None of the disulfides were substrates for reduction by human glutathione reductase. The disulfides caused reversible thioalkylation of hTrx at the redox catalytic site as shown by the fact that there was no thioalkylation of a mutant hTrx where both the catalytic site Cys32 and Cys35 residues were replaced by Ser. In addition, the disulfides caused a slower irreversible inactivation of hTrx as a substrate for reduction by TR, with half-lives for III-2 of 30 min, for IV-2 of 4 hr, and for IX-2 (t-butyl 2-mercaptoimidazolyl disulfide) of 24 hr. This irreversible inactivation of hTrx occurred at concentrations of the disulfides an order of magnitude below those that inhibited TR, and involved the Cys73 of hTrx, which is outside the conserved redox catalytic site, as shown by the resistance to inactivation of a mutant hTrx where Cys73 was replaced by Ser. Electrophoretic and mass spectral analyses of the products of the reaction between the disulfides and hTrx show that modification of 1-3 Cys residues of the protein occurred in a concentration-dependent fashion. The disulfides inhibited the hTrx-dependent proliferation of MCF-7 breast cancer cells with IC50 values for III-2 and IV-2 of 0.2 and 1.2 microM, respectively. The results show that although the catalytic sites of TR and hTrx are reversibly inhibited by the 2-imidazolyl disulfides, it is the irreversible thioalkylation of Cys73 of hTrx by the disulfides that most probably accounts for the inhibition of thioredoxin-dependent cell growth by the disulfides.


Biochemical Pharmacology | 1996

Oxidative inactivation of thioredoxin as a cellular growth factor and protection by a Cys73 → Ser mutation

John R. Gasdaska; D. Lynn Kirkpatrick; William R. Montfort; Miles Kuperus; Simon R. Hill; Margareta Berggren; Garth Powis

Thioredoxin (Trx) is a widely distributed redox protein that regulates several intracellular redox-dependent processes and stimulates the proliferation of both normal and tumor cells. We have found that when stored in the absence of reducing agents, human recombinant Trx undergoes spontaneous oxidation, losing its ability to stimulate cell growth, but is still a substrate for NADPH-dependent reduction by human thioredoxin reductase. There is a slower spontaneous conversion of Trx to a homodimer that is not a substrate for reduction by thioredoxin reductase and that does not stimulate cell proliferation. Both conversions can be induced by chemical oxidants and are reversible by treatment with the thiol reducing agent dithiothreitol. SDS-PAGE suggests that Trx undergoes oxidation to monomeric form(s) preceding dimer formation. We have recently shown by X-ray crystallography that Trx forms a dimer that is stabilized by an intermolecular Cys73-Cys73 disulfide bond. A Cys73-->Ser mutant Trx (C73S) was prepared to determine the role of Cys73 in oxidative stability and growth stimulation. C73S was as effective as Trx in stimulating cell growth and was a comparable substrate for thioredoxin reductase. C73S did not show spontaneous or oxidant-induced loss of activity and did not form a dimer. The results suggest that Trx can exist in monomeric forms, some of which are mediated by Cys73 that do not stimulate cell proliferation but can be reduced by thioredoxin reductase. Cys73 is also involved in formation of an enzymatically inactive homodimer, which occurs on long term storage or by chemical oxidation. Thus, although clearly involved in protein inactivation, Cys73 is not necessary for the growth stimulating activity of Trx.


Molecular Cancer Therapeutics | 2008

Discovery of a novel class of AKT pleckstrin homology domain inhibitors

Daruka Mahadevan; Garth Powis; Eugene A. Mash; Benjamin George; Vijay Gokhale; Shuxing Zhang; Kishore Shakalya; Lei Du-Cuny; Margareta Berggren; M. Ahad Ali; Umasish Jana; Nathan T. Ihle; Sylvestor A. Moses; Chloe Franklin; Satya Narayan; Nikhil V. Shirahatti; Emmanuelle J. Meuillet

AKT, a phospholipid-binding serine/threonine kinase, is a key component of the phosphoinositide 3-kinase cell survival signaling pathway that is aberrantly activated in many human cancers. Many attempts have been made to inhibit AKT; however, selectivity remains to be achieved. We have developed a novel strategy to inhibit AKT by targeting the pleckstrin homology (PH) domain. Using in silico library screening and interactive molecular docking, we have identified a novel class of non–lipid-based compounds that bind selectively to the PH domain of AKT, with “in silico” calculated KD values ranging from 0.8 to 3.0 μmol/L. In order to determine the selectivity of these compounds for AKT, we used surface plasmon resonance to measure the binding characteristics of the compounds to the PH domains of AKT1, insulin receptor substrate-1, and 3-phosphoinositide–dependent protein kinase 1. There was excellent correlation between predicted in silico and measured in vitro KDs for binding to the PH domain of AKT, which were in the range 0.4 to 3.6 μmol/L. Some of the compounds exhibited PH domain–binding selectivity for AKT compared with insulin receptor substrate-1 and 3-phosphoinositide–dependent protein kinase 1. The compounds also inhibited AKT in cells, induced apoptosis, and inhibited cancer cell proliferation. In vivo, the lead compound failed to achieve the blood concentrations required to inhibit AKT in cells, most likely due to rapid metabolism and elimination, and did not show antitumor activity. These results show that these compounds are the first small molecules selectively targeting the PH domain of AKT. [Mol Cancer Ther 2008;7(9):2621–32]


Bioorganic & Medicinal Chemistry Letters | 2001

3-Deoxy-3-substituted-d-myo-inositol imidazolyl ether lipid phosphates and carbonate as inhibitors of the phosphatidylinositol 3-kinase pathway and cancer cell growth

Youhong Hu; Emmanuelle J. Meuillet; Margareta Berggren; Garth Powis; Alan P. Kozikowski

3-Modified D-myo-inositol imidazolyl ether lipid phosphates and a carbonate were synthesized and evaluated as inhibitors of P13-K and Akt. These data are presented along with IC50 values for the inhibition of the growth of three cancer cell lines.


Nutrition and Cancer | 2009

Sodium Selenite Increases the Activity of the Tumor Suppressor Protein, PTEN, in DU-145 Prostate Cancer Cells

Margareta Berggren; Sivanandane Sittadjody; Zuohe Song; Jean Louis Samira; Randy Burd; Emmanuelle J. Meuillet

Epidemiological and clinical data suggest that selenium may prevent prostate cancer; however, the cellular effects of selenium in malignant prostate cells are not well understood. We previously reported that the activity of the tumor suppressor PTEN is modulated by thioredoxin (Trx) in a RedOx-dependent manner. In this study, we demonstrated that the activity of Trx reductase (TR) is increased by sevenfold in the human prostate cancer cell line, DU-145, after 5 days of sodium selenite (Se) treatment. The treatment of DU-145 cells with increasing concentrations of Se induced an increase in PTEN lipid phosphatase activity by twofold, which correlated with a decrease in phospho-ser 473 -Akt, and an increase in phospho-Ser 370 -PTEN levels. Se also increased casein kinase-2 (CK2) activity; and the use of apigenin, an inhibitor of CK2, revealed that the regulation of the tumor suppressor PTEN by Se may be achieved via both the Trx-TR system and the RedOx control of the kinase involved in the regulation of PTEN activity.


Tetrahedron Letters | 2002

Synthesis and biological activity of 3-hydroxy(phosphono)methyl-bearing phosphatidylinositol ether lipid analogues

Haiying Sun; Gaddam Bapu Reddy; Clifford George; Emmanuelle J. Meuillet; Margareta Berggren; Garth Powis; Alan P. Kozikowski

Abstract Two 3-hydroxy(phosphono)methyl-bearing phosphatidylinositol ether lipid analogues were synthesized and shown to be inhibitors of Akt and PI3-K. These compounds were also shown to inhibit the growth of HT-29 human colon cancer cells and MCF-7 human breast cancer cells.


FEBS Letters | 1994

Biochemical, structural, and biological properties of human thioredoxin active site peptides

John E. Oblong; Margareta Berggren; Garth Powis

The human redox protein thioredoxin is an autocrine growth factor for some cancer cells. Redox activity is essential for this function but other required structural features of thioredoxin are not known. Two 8‐mer peptides (I and II) and one 14‐mer peptide (III) were designed based on the amino acid sequence of the redox active site of thioredoxin. Peptide I and peptide III contained the wild‐type sequence of thioredoxin while peptide II contained serine residues in place of the catalytically active cysteines. Circular dichroism spectroscopy indicated that all three peptides were comprised mainly of random coil, with peptide III containing slightly more ordered secondary structure. Peptides I and III were substrates for thioredoxin reductase with K M values of 890 and 265 μM, respectively. The redox inactive peptide II could not compete with thioredoxin for reduction by thioredoxin reductase in a coupled insulin reduction assay. However, peptide II was a competitive inhibitor for the reduction of 5,5‐dithiobis‐(2‐nitrobenzoic acid) by thioredoxin reductase. All three peptides gave only background levels of stimulation of the proliferation of Swiss 3T3 murine fibroblasts when compared to the stimulation caused by thioredoxin. These results suggest that while the ability of thioredoxin to stimulate cellular proliferation is redox‐dependent, more information than that contained in the redox active site domain alone defined by 14 amino acids is required.


Advances in Experimental Medicine and Biology | 2001

DNA microarray reveals increased expression of thioredoxin peroxidase in thioredoxin-1 transfected cells and its functional consequences.

Bryan Husbeck; Margareta Berggren; Garth Powis

The mammalian thioredoxins are a family of small redox proteins that undergo NADPH dependent reduction by thioredoxin reductase. Reduced thioredoxins reduce oxidized cysteine groups on proteins including transcription factors to increase their binding to DNA, and is a source of reducing equivalents for enzymes such as thioredoxin peroxidase which removes H2O2 and alkyl peroxides. Thioredoxin-1 is over expressed in many human tumors where it is associated with aggressive tumor growth, inhibited apoptosis and decreased patient survival. Transfection of cells with thioredoxin-1 has been shown to increase cell growth and inhibit apoptosis. We have used DNA micro array to investigate the effects of thioredoxin-1 transfection on the expression of a panel of 520 redox, apoptosis and cell growth related genes in MCF-7 human breast cancer cells. One of the genes whose expression was increased as a result of thioredoxin-1 over expression was thioredoxin peroxidase-2. This increase was confirmed by Northern blotting. Transfection of mouse WEHI7.2 thymoma cells with human thioredoxin peroxidase-2 was found to protect the cells from apoptosis induced by H2O2 but not from apoptosis induced by dexamethasone, doxorubicin or etoposide. Thus, increased thioredoxin peroxidase-2 expression does not explain the widespread antiapoptotic effects of thioredoxin-1.


Molecular Cancer Therapeutics | 2004

Molecular pharmacology and antitumor activity of PX-866, a novel inhibitor of phosphoinositide-3-kinase signaling

Nathan T. Ihle; Ryan Williams; Sherry Chow; Wade Chew; Margareta Berggren; Gillian Paine-Murrieta; Daniel J. Minion; Robert J. Halter; Peter Wipf; Robert T. Abraham; Lynn Kirkpatrick; Garth Powis

Collaboration


Dive into the Margareta Berggren's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alan P. Kozikowski

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nathan T. Ihle

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Peter Wipf

University of Pittsburgh

View shared research outputs
Researchain Logo
Decentralizing Knowledge