Network


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

Hotspot


Dive into the research topics where Ann-Sofie Jemth is active.

Publication


Featured researches published by Ann-Sofie Jemth.


The EMBO Journal | 2009

PARP is activated at stalled forks to mediate Mre11-dependent replication restart and recombination.

Helen E. Bryant; Eva Petermann; Niklas Schultz; Ann-Sofie Jemth; Olga Loseva; Natalia Issaeva; Fredrik Johansson; Serena Fernandez; Peter McGlynn; Thomas Helleday

If replication forks are perturbed, a multifaceted response including several DNA repair and cell cycle checkpoint pathways is activated to ensure faithful DNA replication. Here, we show that poly(ADP‐ribose) polymerase 1 (PARP1) binds to and is activated by stalled replication forks that contain small gaps. PARP1 collaborates with Mre11 to promote replication fork restart after release from replication blocks, most likely by recruiting Mre11 to the replication fork to promote resection of DNA. Both PARP1 and PARP2 are required for hydroxyurea‐induced homologous recombination to promote cell survival after replication blocks. Together, our data suggest that PARP1 and PARP2 detect disrupted replication forks and attract Mre11 for end processing that is required for subsequent recombination repair and restart of replication forks.


Nature | 2014

MTH1 inhibition eradicates cancer by preventing sanitation of the dNTP pool.

Helge Gad; Tobias Koolmeister; Ann-Sofie Jemth; Saeed Eshtad; Sylvain A. Jacques; Cecilia E. Ström; Linda M. Svensson; Niklas Schultz; Thomas Lundbäck; Berglind O. Einarsdottir; Aljona Saleh; Camilla Göktürk; Pawel Baranczewski; Richard Svensson; Ronnie P.-A. Berntsson; Robert Gustafsson; Kia Strömberg; Kumar Sanjiv; Marie-Caroline Jacques-Cordonnier; Matthieu Desroses; Anna-Lena Gustavsson; Roger Olofsson; Fredrik Johansson; Evert Homan; Olga Loseva; Lars Bräutigam; Lars Johansson; Andreas Höglund; Anna Hagenkort; Therese Pham

Cancers have dysfunctional redox regulation resulting in reactive oxygen species production, damaging both DNA and free dNTPs. The MTH1 protein sanitizes oxidized dNTP pools to prevent incorporation of damaged bases during DNA replication. Although MTH1 is non-essential in normal cells, we show that cancer cells require MTH1 activity to avoid incorporation of oxidized dNTPs, resulting in DNA damage and cell death. We validate MTH1 as an anticancer target in vivo and describe small molecules TH287 and TH588 as first-in-class nudix hydrolase family inhibitors that potently and selectively engage and inhibit the MTH1 protein in cells. Protein co-crystal structures demonstrate that the inhibitors bind in the active site of MTH1. The inhibitors cause incorporation of oxidized dNTPs in cancer cells, leading to DNA damage, cytotoxicity and therapeutic responses in patient-derived mouse xenografts. This study exemplifies the non-oncogene addiction concept for anticancer treatment and validates MTH1 as being cancer phenotypic lethal.


Nature | 2014

Stereospecific targeting of MTH1 by ( S )-crizotinib as an anticancer strategy

Kilian Huber; E. Salah; Branka Radic; Manuela Gridling; J.M. Elkins; Alexey Stukalov; Ann-Sofie Jemth; Camilla Göktürk; Kumar Sanjiv; Kia Strömberg; Therese Pham; Ulrika Warpman Berglund; Jacques Colinge; Keiryn L. Bennett; Joanna I. Loizou; Thomas Helleday; Stefan Knapp; Giulio Superti-Furga

Activated RAS GTPase signalling is a critical driver of oncogenic transformation and malignant disease. Cellular models of RAS-dependent cancers have been used to identify experimental small molecules, such as SCH51344, but their molecular mechanism of action remains generally unknown. Here, using a chemical proteomic approach, we identify the target of SCH51344 as the human mutT homologue MTH1 (also known as NUDT1), a nucleotide pool sanitizing enzyme. Loss-of-function of MTH1 impaired growth of KRAS tumour cells, whereas MTH1 overexpression mitigated sensitivity towards SCH51344. Searching for more drug-like inhibitors, we identified the kinase inhibitor crizotinib as a nanomolar suppressor of MTH1 activity. Surprisingly, the clinically used (R)-enantiomer of the drug was inactive, whereas the (S)-enantiomer selectively inhibited MTH1 catalytic activity. Enzymatic assays, chemical proteomic profiling, kinome-wide activity surveys and MTH1 co-crystal structures of both enantiomers provide a rationale for this remarkable stereospecificity. Disruption of nucleotide pool homeostasis via MTH1 inhibition by (S)-crizotinib induced an increase in DNA single-strand breaks, activated DNA repair in human colon carcinoma cells, and effectively suppressed tumour growth in animal models. Our results propose (S)-crizotinib as an attractive chemical entity for further pre-clinical evaluation, and small-molecule inhibitors of MTH1 in general as a promising novel class of anticancer agents.


Journal of Biological Chemistry | 2010

PARP-3 Is a Mono-ADP-ribosylase That Activates PARP-1 in the Absence of DNA

Olga Loseva; Ann-Sofie Jemth; Helen E. Bryant; Herwig Schüler; Lari Lehtiö; Tobias Karlberg; Thomas Helleday

The PARP-3 protein is closely related to the PARP-1 and PARP-2 proteins, which are involved in DNA repair and genome maintenance. Here, we characterized the biochemical properties of human PARP-3. PARP-3 is able to ADP-ribosylate itself as well as histone H1, a previously unknown substrate for PARP-3. PARP-3 is not activated upon binding to DNA and is a mono-ADP-ribosylase, in contrast to PARP-1 and PARP-2. PARP-3 interacts with PARP-1 and activates PARP-1 in the absence of DNA, resulting in synthesis of polymers of ADP-ribose. The N-terminal WGR domain of PARP-3 is involved in this activation. The functional interaction between PARP-3 and PARP-1 suggests that it may have a role in DNA repair. However, here we report that PARP-3 small interfering RNA-depleted cells are not sensitive to the topoisomerase I poison camptothecin, inducing DNA single-strand breaks, and repair these lesions as efficiently as wild-type cells. Altogether, these results suggest that the interaction between PARP-1 and PARP-3 is unrelated to DNA single-strand break repair.


Journal of Medicinal Chemistry | 2009

Structural basis for inhibitor specificity in human poly(ADP-ribose) polymerase-3.

Lari Lehtiö; Ann-Sofie Jemth; R. Collins; Olga Loseva; Andreas Johansson; Natalia Markova; Martin Hammarström; A. Flores; Lovisa Holmberg-Schiavone; Johan Weigelt; Thomas Helleday; Herwig Schüler; Tobias Karlberg

Poly(ADP-ribose) polymerases (PARPs) activate DNA repair mechanisms upon stress- and cytotoxin-induced DNA damage, and inhibition of PARP activity is a lead in cancer drug therapy. We present a structural and functional analysis of the PARP domain of human PARP-3 in complex with several inhibitors. Of these, KU0058948 is the strongest inhibitor of PARP-3 activity. The presented crystal structures highlight key features for potent inhibitor binding and suggest routes for creating isoenzyme-specific PARP inhibitors.


FEBS Letters | 2011

Crystal structure of human MTH1 and the 8‐oxo‐dGMP product complex

Linda M. Svensson; Ann-Sofie Jemth; Matthieu Desroses; Olga Loseva; Thomas Helleday; Martin Högbom; Pål Stenmark

MTH1 hydrolyzes oxidized nucleotide triphosphates, thereby preventing them from being incorporated into DNA. We here present the structures of human MTH1 (1.9 Å) and its complex with the product 8‐oxo‐dGMP (1.8 Å). Unexpectedly MTH1 binds the nucleotide in the anti conformation with no direct interaction between the 8‐oxo group and the protein. We suggest that the specificity depends on the stabilization of an enol tautomer of the 8‐oxo form of dGTP. The binding of the product induces no major structural changes. The structures reveal the mode of nucleotide binding in MTH1 and provide the structural basis for inhibitor design.


Nature Communications | 2015

Crystal structure, biochemical and cellular activities demonstrate separate functions of MTH1 and MTH2.

Megan Carter; Ann-Sofie Jemth; Anna Hagenkort; Brent D. G. Page; Robert Gustafsson; Julia J. Griese; Helge Gad; Nicholas C. K. Valerie; Matthieu Desroses; Johan Boström; Ulrika Warpman Berglund; Thomas Helleday; Pål Stenmark

Deregulated redox metabolism in cancer leads to oxidative damage to cellular components including deoxyribonucleoside triphosphates (dNTPs). Targeting dNTP pool sanitizing enzymes, such as MTH1, is a highly promising anticancer strategy. The MTH2 protein, known as NUDT15, is described as the second human homologue of bacterial MutT with 8-oxo-dGTPase activity. We present the first NUDT15 crystal structure and demonstrate that NUDT15 prefers other nucleotide substrates over 8-oxo-dGTP. Key structural features are identified that explain different substrate preferences for NUDT15 and MTH1. We find that depletion of NUDT15 has no effect on incorporation of 8-oxo-dGTP into DNA and does not impact cancer cell survival in cell lines tested. NUDT17 and NUDT18 were also profiled and found to have far less activity than MTH1 against oxidized nucleotides. We show that NUDT15 is not a biologically relevant 8-oxo-dGTPase, and that MTH1 is the most prominent sanitizer of the cellular dNTP pool known to date.


Nature Medicine | 2017

Targeting SAMHD1 with the Vpx protein to improve cytarabine therapy for hematological malignancies

Nikolas Herold; Sean G. Rudd; Linda Ljungblad; Kumar Sanjiv; Ida Hed Myrberg; Cynthia B.J. Paulin; Yaser Heshmati; Anna Hagenkort; Juliane Kutzner; Brent D. G. Page; José Manuel Calderón-Montaño; Olga Loseva; Ann-Sofie Jemth; Lorenzo Bulli; Hanna Axelsson; Bianca Tesi; Nicholas C. K. Valerie; Andreas Höglund; Julia Bladh; Elisee Wiita; Mikael Sundin; Michael Uhlin; Georgios Rassidakis; Mats Heyman; Katja Pokrovskaja Tamm; Ulrika Warpman-Berglund; Julian Walfridsson; Sören Lehmann; Dan Grandér; Thomas Lundbäck

The cytostatic deoxycytidine analog cytarabine (ara-C) is the most active agent available against acute myelogenous leukemia (AML). Together with anthracyclines, ara-C forms the backbone of AML treatment for children and adults. In AML, both the cytotoxicity of ara-C in vitro and the clinical response to ara-C therapy are correlated with the ability of AML blasts to accumulate the active metabolite ara-C triphosphate (ara-CTP), which causes DNA damage through perturbation of DNA synthesis. Differences in expression levels of known transporters or metabolic enzymes relevant to ara-C only partially account for patient-specific differential ara-CTP accumulation in AML blasts and response to ara-C treatment. Here we demonstrate that the deoxynucleoside triphosphate (dNTP) triphosphohydrolase SAM domain and HD domain 1 (SAMHD1) promotes the detoxification of intracellular ara-CTP pools. Recombinant SAMHD1 exhibited ara-CTPase activity in vitro, and cells in which SAMHD1 expression was transiently reduced by treatment with the simian immunodeficiency virus (SIV) protein Vpx were dramatically more sensitive to ara-C-induced cytotoxicity. CRISPR–Cas9-mediated disruption of the gene encoding SAMHD1 sensitized cells to ara-C, and this sensitivity could be abrogated by ectopic expression of wild-type (WT), but not dNTPase-deficient, SAMHD1. Mouse models of AML lacking SAMHD1 were hypersensitive to ara-C, and treatment ex vivo with Vpx sensitized primary patient-derived AML blasts to ara-C. Finally, we identified SAMHD1 as a risk factor in cohorts of both pediatric and adult patients with de novo AML who received ara-C treatment. Thus, SAMHD1 expression levels dictate patient sensitivity to ara-C, providing proof-of-concept that the targeting of SAMHD1 by Vpx could be an attractive therapeutic strategy for potentiating ara-C efficacy in hematological malignancies.


Cancer Research | 2016

NUDT15 Hydrolyzes 6-Thio-DeoxyGTP to Mediate the Anticancer Efficacy of 6-Thioguanine.

Nicholas C. K. Valerie; Anna Hagenkort; Brent D. G. Page; Geoffrey Masuyer; Daniel Rehling; Megan Carter; Luka Bevc; Patrick Herr; Evert Homan; Nina Gustafsson Sheppard; Pål Stenmark; Ann-Sofie Jemth; Thomas Helleday

Thiopurines are a standard treatment for childhood leukemia, but like all chemotherapeutics, their use is limited by inherent or acquired resistance in patients. Recently, the nucleoside diphosphate hydrolase NUDT15 has received attention on the basis of its ability to hydrolyze the thiopurine effector metabolites 6-thio-deoxyGTP (6-thio-dGTP) and 6-thio-GTP, thereby limiting the efficacy of thiopurines. In particular, increasing evidence suggests an association between the NUDT15 missense variant, R139C, and thiopurine sensitivity. In this study, we elucidated the role of NUDT15 and NUDT15 R139C in thiopurine metabolism. In vitro and cellular results argued that 6-thio-dGTP and 6-thio-GTP are favored substrates for NUDT15, a finding supported by a crystallographic determination of NUDT15 in complex with 6-thio-GMP. We found that NUDT15 R139C mutation did not affect enzymatic activity but instead negatively influenced protein stability, likely due to a loss of supportive intramolecular bonds that caused rapid proteasomal degradation in cells. Mechanistic investigations in cells indicated that NUDT15 ablation potentiated induction of the DNA damage checkpoint and cancer cell death by 6-thioguanine. Taken together, our results defined how NUDT15 limits thiopurine efficacy and how genetic ablation via the R139C missense mutation confers sensitivity to thiopurine treatment in patients. Cancer Res; 76(18); 5501-11. ©2016 AACR.


Annals of Oncology | 2016

Validation and development of MTH1 inhibitors for treatment of cancer

U. Warpman Berglund; Kumar Sanjiv; Helge Gad; Christina Kalderén; Tobias Koolmeister; Therese Pham; Camilla Göktürk; Rozbeh Jafari; Gianluca Maddalo; B. Seashore-Ludlow; A. Chernobrovkin; A. Manoilov; I. S. Pateras; A. Rasti; Ann-Sofie Jemth; Ingrid Almlöf; Olga Loseva; Torkild Visnes; Berglind O. Einarsdottir; Fabienne Z. Gaugaz; Aljona Saleh; B. Platzack; Olov A. Wallner; Karl S. A. Vallin; Martin Henriksson; P. Wakchaure; S. Borhade; P. Herr; Y. Kallberg; Pawel Baranczewski

BACKGROUND Previously, we showed cancer cells rely on the MTH1 protein to prevent incorporation of otherwise deadly oxidised nucleotides into DNA and we developed MTH1 inhibitors which selectively kill cancer cells. Recently, several new and potent inhibitors of MTH1 were demonstrated to be non-toxic to cancer cells, challenging the utility of MTH1 inhibition as a target for cancer treatment. MATERIAL AND METHODS Human cancer cell lines were exposed in vitro to MTH1 inhibitors or depleted of MTH1 by siRNA or shRNA. 8-oxodG was measured by immunostaining and modified comet assay. Thermal Proteome profiling, proteomics, cellular thermal shift assays, kinase and CEREP panel were used for target engagement, mode of action and selectivity investigations of MTH1 inhibitors. Effect of MTH1 inhibition on tumour growth was explored in BRAF V600E-mutated malignant melanoma patient derived xenograft and human colon cancer SW480 and HCT116 xenograft models. RESULTS Here, we demonstrate that recently described MTH1 inhibitors, which fail to kill cancer cells, also fail to introduce the toxic oxidized nucleotides into DNA. We also describe a new MTH1 inhibitor TH1579, (Karonudib), an analogue of TH588, which is a potent, selective MTH1 inhibitor with good oral availability and demonstrates excellent pharmacokinetic and anti-cancer properties in vivo. CONCLUSION We demonstrate that in order to kill cancer cells MTH1 inhibitors must also introduce oxidized nucleotides into DNA. Furthermore, we describe TH1579 as a best-in-class MTH1 inhibitor, which we expect to be useful in order to further validate the MTH1 inhibitor concept.

Collaboration


Dive into the Ann-Sofie Jemth's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Helge Gad

Karolinska Institutet

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge