Network


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

Hotspot


Dive into the research topics where A. Shvarts is active.

Publication


Featured researches published by A. Shvarts.


The EMBO Journal | 1996

MDMX : A NOVEL P53-BINDING PROTEIN WITH SOME FUNCTIONAL PROPERTIES OF MDM2

A. Shvarts; Wilma T. Steegenga; Nicole Riteco; T. van Laar; Patrick Dekker; M. Bazuine; R. C. A. Van Ham; W. van der Houven van Oordt; G. Hateboer; A.J. van der Eb; Aart G. Jochemsen

Here we report the isolation of a cDNA encoding a new p53‐associating protein. This new protein has been called MDMX on the basis of its structural similarity to MDM2, which is especially notable in the p53‐binding domain. In addition, the putative metal binding domains in the C‐terminal part of MDM2 are completely conserved in MDMX. The middle part of the MDMX and MDM2 proteins shows a low degree of conservation. We can show by co‐immunoprecipitation that the MDMX protein interacts specifically with p53 in vivo. This interaction probably occurs with the N‐terminal part of p53, because the activity of the transcription activation domain of p53 was inhibited by co‐transfection of MDMX. Northern blotting showed that MDMX, like MDM2, is expressed in all tissues tested, and that several mRNAs for MDMX can be detected. Interestingly, the level of MDMX mRNA is unchanged after UV irradiation, in contrast to MDM2 transcription. This observation suggests that MDMX may be a differently regulated modifier of p53 activity in comparison with MDM2. Our study indicates that at least one additional member of the MDM protein family exists which can modulate p53 function.


Journal of Clinical Pathology | 2005

Differential prognostic impact of hypoxia induced and diffuse HIF-1α expression in invasive breast cancer

Marije M. Vleugel; A E Greijer; A. Shvarts; P van der Groep; M van Berkel; Y Aarbodem; H. van Tinteren; Adrian L. Harris; P. J. van Diest; E. van der Wall

Background: Intratumorous hypoxia triggers a broad cellular response mediated by the transcription factor hypoxia inducible factor 1 (HIF-1). HIF-1α concentrations increase during breast carcinogenesis, and are associated with poor prognosis. An earlier study noted two HIF-1α overexpression patterns: diffuse scattered throughout the tissue and confined to perinecrotic cells. Aims: To investigate the prognostic impact of these different HIF-1α overexpression patterns in relation to its downstream effectors carbonic anhydrase (CA) IX and glucose transporter 1 (GLUT-1). Methods: HIF-1α, CA IX, and GLUT-1 expression was studied by immunohistochemistry, including double staining for CA IX and HIF-1α. Clinical data included disease free survival, lymph node status, and tumour size. Results: HIF-1α overexpression (44% of cases) had a perinecrotic (13.5%) or diffuse staining pattern (30.5%). CA IX expression was detectable in 12.5% of breast cancers, whereas GLUT-1 expression was seen in 29%, with both showing perinecrotic membrane staining. Perinecrotic HIF-1α overexpression was highly associated with CA IX and GLUT-1 overexpression, and double staining for HIF-1α and CA IX showed strong expression in the same cells. Diffusely overexpressed HIF-1α was not associated with CA IX or GLUT-1 expression. Patients with diffuse HIF-1α staining had a significantly better prognosis than patients with perinecrotically overexpressed HIF-1α. Conclusions: Different regulation pathways of HIF-1α overexpression exist in breast cancer: (1) hypoxia induced, perinecrotic HIF-1α overexpression with strong expression of hypoxia associated genes (CA IX and GLUT-1), which is associated with a poor prognosis; and (2) diffuse HIF-1α overexpression lacking major hypoxia associated downstream effects, resulting in a more favourable prognosis.


Oncogene | 2008

The TWIST1 oncogene is a direct target of hypoxia-inducible factor-2α

Eelke H. Gort; G van Haaften; I Verlaan; Arjan J. Groot; Ronald H.A. Plasterk; A. Shvarts; Karijn P.M. Suijkerbuijk; T. van Laar; E. van der Wall; Venu Raman; P. J. van Diest; Marcel Tijsterman; Marc Vooijs

Hypoxia-inducible factors (HIFs) are highly conserved transcription factors that play a crucial role in oxygen homeostasis. Intratumoral hypoxia and genetic alterations lead to HIF activity, which is a hallmark of solid cancer and is associated with poor clinical outcome. HIF activity is regulated by an evolutionary conserved mechanism involving oxygen-dependent HIFα protein degradation. To identify novel components of the HIF pathway, we performed a genome-wide RNA interference screen in Caenorhabditis elegans, to suppress HIF-dependent phenotypes, like egg-laying defects and hypoxia survival. In addition to hif-1 (HIFα) and aha-1 (HIFβ), we identified hlh-8, gska-3 and spe-8. The hlh-8 gene is homologous to the human oncogene TWIST1. We show that TWIST1 expression in human cancer cells is enhanced by hypoxia in a HIF-2α-dependent manner. Furthermore, intronic hypoxia response elements of TWIST1 are regulated by HIF-2α, but not HIF-1α. These results identify TWIST1 as a direct target gene of HIF-2α, which may provide insight into the acquired metastatic capacity of hypoxic tumors.


Journal of Clinical Pathology | 2004

Lack of lymphangiogenesis during breast carcinogenesis

Marije M. Vleugel; Rinke Bos; P van der Groep; Astrid E. Greijer; A. Shvarts; H V Stel; E. van der Wall; P. J. van Diest

Background: Recent evidence suggests that functional intratumorous lymph vessels may be absent from some human cancers. This could result from either the failure of tumours to induce lymphangiogenesis, or the collapse of lymph vessels, caused by high interstitial tumour pressure. Methods: To differentiate between these two hypotheses, paraffin wax embedded clinical specimens from normal breast (n  =  13), usual ductal hyperplasia (n  =  11), ductal carcinoma in situ (n  =  21), and invasive breast cancer (n  =  40) were compared for lymphatic and blood vessel density by immunohistochemistry with antibodies to the lymphatic endothelial hyaluronan receptor (LYVE-1) and CD31, respectively. Results: Lymph vessel density was lower than blood vessel density in normal breast tissue. Within breast lobuli, lymph vessels were absent. In premalignant lesions blood microvessel density increased, whereas no increase in lymph vessels could be seen intralesionally. In invasive cancers, lymph vessels were absent in all but a few cases, where probably some pre-existing lymph vessels remained, although blood microvessel density was once again increased. Conclusion: Unlike angiogenesis, lymphangiogenesis is absent during breast carcinogenesis. This, and not rising interstitial pressure caused by an increase in the size of lesions, explains the absence of intratumorous lymph vessels in invasive breast cancer.


Breast Cancer Research | 2004

Expression of hypoxia-inducible factor-1α and cell cycle proteins in invasive breast cancer are estrogen receptor related

Reinhard Bos; Paul J. van Diest; Petra van der Groep; A. Shvarts; Astrid E. Greijer; Elsken van der Wall

BackgroundThe transcription factor hypoxia-inducible factor-1 (HIF-1) is a key regulator of the cellular response to hypoxia. Previous studies showed that concentrations of its subunit HIF-1α, as a surrogate for HIF-1 activity, are increased during breast carcinogenesis and can independently predict prognosis in breast cancer. During carcinogenesis, the cell cycle is progressively deregulated, and proliferation rate is a strong prognostic factor in breast cancer. In this study we undertook a detailed evaluation of the relationships between HIF-1α and cell cycle-associated proteins.MethodsIn a representative estrogen receptor (ER) group of 150 breast cancers, the expression of HIF-1α, vascular endothelial growth factor, the ER, HER-2/neu, Ki-67, cyclin A, cyclin D1, p21, p53, and Bcl-2 was investigated by immunohistochemistry.ResultsHigh concentrations (5% or more) of HIF-1α were associated with increased proliferation as shown by positive correlations with Ki-67 (P < 0.001) and the late S–G2-phase protein cyclin A (P < 0.001), but not with the G1-phase protein cyclin D1. High HIF-1α concentrations were also strongly associated with p53 positivity (P < 0.001) and loss of Bcl-2 expression (P = 0.013). No association was found between p21 and HIF-1α (P = 0.105) in the whole group of patients. However, the subgroup of ER-positive cancers was characterized by a strong positive association between HIF-1α and p21 (P = 0.023), and HIF-1α lacked any relation with proliferation.ConclusionHIF-1α overexpression is associated with increased proliferation, which might explain the adverse prognostic impact of increased concentrations of HIF-1α in invasive breast cancer. In ER-positive tumors, HIF-1α is associated with p21 but not against proliferation. This shows the importance of further functional analysis to unravel the role of HIF-1 in late cell cycle progression, and the link between HIF-1, p21, and ER.


Oncogene | 2006

Hypoxia-inducible factor-1α expression requires PI 3-kinase activity and correlates with Akt1 phosphorylation in invasive breast carcinomas

Eelke H. Gort; Arjan J. Groot; T L P Derks van de Ven; P van der Groep; I Verlaan; T. van Laar; P. J. van Diest; E. van der Wall; A. Shvarts

Hypoxia-inducible factor-1 alpha (HIF-1α) is the regulatory subunit of the heterodimeric transcription factor HIF-1 and the key factor in cellular response to low oxygen tension. Expression of HIF-1α protein is associated with poor patient survival and therapy resistance in many types of solid tumors. Insight into HIF-1α regulation in solid tumors is important for therapeutic strategies. In this study, we determined the pathophysiological relevance of HIF-1α regulation by the oncogenic phosphatidylinositol 3′-kinase (PI 3-kinase)/Akt signaling pathway. We modeled the physiology of hypoxic tumor regions by culturing carcinoma cells under low oxygen tension in the absence of serum. We observed that hypoxic induction of HIF-1α protein was decreased by serum deprivation. Overexpression of dominant-active Akt1 restored HIF-1α expression, whereas inhibition of PI 3-kinase activity reduced hypoxic HIF-1α protein levels to a similar extent as serum deprivation. Immunohistochemical analysis of 95 human breast cancers revealed that lack of Akt1 phosphorylation correlates with low HIF-1α levels. To our knowledge, this is the first reported comparison between HIF-1α expression and Akt phosphorylation in human carcinomas. We conclude that Akt activity is physiologically relevant for HIF-1α expression in breast cancer. This implies that HIF-1α function might be therapeutically targeted by inhibition of the PI 3-kinase/Akt pathway.


Analytical Cellular Pathology | 2005

Hypoxia‐induced acidification causes mitoxantrone resistance not mediated by drug transporters in human breast cancer cells

Astrid E. Greijer; Mc de Jong; George L. Scheffer; A. Shvarts; P. J. van Diest; E. van der Wall

Hypoxia has clinically been associated with resistance to chemotherapy. The aim of this study was to investigate whether hypoxia induces resistance to doxorubicin and mitoxantrone, two common drugs in cancer treatment, in MCF‐7 breast cancer cells, and SW1573 non‐small lung cancer cells. In addition, the role of drug transporters P‐gp, BCRP and MRP1 was analysed. Hypoxia induced resistance in MCF‐7 cells to mitoxantrone shifted the IC50 value from 0.09 μM (±0.01) to 0.54 μM (±0.06) under hypoxia, whereas survival of MCF‐7 and SW1573 cells in the presence of doxorubicin was not altered. Accumulation of mitoxantrone and daunorubicin, a doxorubicin fluorescent homologue, appeared to be 5.3 and 3.2 times lower in MCF‐7 cells, respectively. Cytotoxicity assays showed no increased functionality of the drug transporters P‐gp, BCRP and MRP1 under hypoxia. In addition, protein levels of these drug transporters were not changed. Medium of the MCF‐7 cells became more acidic under hypoxia thereby causing a decreased uptake of mitoxantrone. Hypoxia induces mitoxantrone resistance in MCF‐7 cells not mediated by the three major MDR transporters. Hypoxia‐induced acidification may cause this resistance by decreased cellular uptake together with a lowered cytotoxicity due to pH‐dependent topoisomerase type II activity.


Laboratory Investigation | 2006

Identification by phage display of single-domain antibody fragments specific for the ODD domain in hypoxia-inducible factor 1alpha.

Arjan J. Groot; Peter Verheesen; Elike J Westerlaken; Eelke H. Gort; Petra van der Groep; Niels Bovenschen; Elsken van der Wall; Paul J. van Diest; A. Shvarts

Hypoxia triggers the transcription of genes responsible for cell survival via the key player transcription factor hypoxia-inducible factor 1alpha (HIF-1α). Overexpression of this protein has been implicated in cardiovascular disorders, carcinogenesis and cancer progression. For functional and diagnostic studies on the HIF-1α protein, we have identified single-domain antibody fragments directed against this protein by using a llama-derived nonimmune phage display library. This library displays the variable domains of the heavy-chain antibody subclass, found in these animals. Phage display selection with six recombinant HIF-1α proteins yielded five different antibody fragments. By epitope-mapping, we show that all five antibody fragments bind within the functionally important oxygen-dependent degradation domain of the HIF-1α protein. Two of these antibody fragments were engineered into bivalent antibodies that were able to detect human HIF-1α by immunohistochemistry, Western blotting and immunoprecipitation, and mouse HIF-1α by immunofluorescence and immunoprecipitation. These are the first single-domain antibody fragments that may be used in exploration of HIF-1α as a possible therapeutic target through molecular applications.


Archives of Gynecology and Obstetrics | 2006

BRCA1 and p53 protein expression in cultured ovarian surface epithelial cells derived from women with and without a BRCA1 germline mutation.

Jurgen M. J. Piek; Josephine C. Dorsman; Leon F.A.G. Massuger; Anca C. Ansink; Jitske Weegenaar; A. Shvarts; P. Kenemans; René H.M. Verheijen


The Journal of Pathology | 2004

Identification of hypoxia responsive genes regulated by the hypoxia inducible factor 1 (HIF-1)

Astrid E. Greijer; P van der Groep; D Kemming; A. Shvarts; Gregg L. Semenza; P. J. van Diest; E. van der Wall

Collaboration


Dive into the A. Shvarts's collaboration.

Top Co-Authors

Avatar

E. van der Wall

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Astrid E. Greijer

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aart G. Jochemsen

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge