Network


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

Hotspot


Dive into the research topics where Zdenka Matijasevic is active.

Publication


Featured researches published by Zdenka Matijasevic.


Molecular and Cellular Biology | 2008

MdmX Promotes Bipolar Mitosis To Suppress Transformation and Tumorigenesis in p53-Deficient Cells and Mice

Zdenka Matijasevic; Heather Anne Steinman; Kathleen Hoover; Stephen N. Jones

ABSTRACT Mdm2 and MdmX are structurally related p53-binding proteins that function as critical negative regulators of p53 activity in embryonic and adult tissue. The overexpression of Mdm2 or MdmX inhibits p53 tumor suppressor functions in vitro, and the amplification of Mdm2 or MdmX is observed in human cancers retaining wild-type p53. We now demonstrate a surprising role for MdmX in suppressing tumorigenesis that is distinct from its oncogenic ability to inhibit p53. The deletion of MdmX induces multipolar mitotic spindle formation and the loss of chromosomes from hyperploid p53-null cells. This reduction in chromosome number, not observed in p53-null cells with Mdm2 deleted, correlates with increased cell proliferation and the spontaneous transformation of MdmX/p53-null mouse embryonic fibroblasts in vitro and with an increased rate of spontaneous tumorigenesis in MdmX/p53-null mice in vivo. These results indicate that MdmX has a p53-independent role in suppressing oncogenic cell transformation, proliferation, and tumorigenesis by promoting centrosome clustering and bipolar mitosis.


Cell Cycle | 2008

MdmX regulates transformation and chromosomal stability in p53-deficient cells

Zdenka Matijasevic; Anna Krzywicka-Racka; Greenfield Sluder; Stephen N. Jones

The cellular homologues Mdm2 and MdmX play critical roles in regulating the activity of the p53 tumor suppressor in damaged and non-damaged cells and during development in mice. Recently, we have utilized genetically defined primary cells and mice to reveal that endogenous levels of MdmX can also suppress multipolar mitosis and transformation in hyperploid p53-deficient cells and tumorigenesis in p53-deficient mice. These MdmX functions are not shared by Mdm2, and are distinct from the well-established ability of MdmX to complex with and inhibit p53 activity. Here we discuss some of the ramifications of MdmX loss in p53-deficient cells and mice, and we explore further the fate of MdmX/p53-double null embryonic fibroblasts undergoing multi-polar cell division using time-lapse video microscopy. We also discuss the relationship between chromosomal loss, cell proliferation, and the tumorigenic potential of p53-deficient cells lacking MdmX.


Mutation Research Letters | 1985

Mutagenicity of pyrene in Salmonella.

Zdenka Matijasevic; Errol Zeiger

Pyrene was tested for mutagenicity in Salmonella typhimurium strains TA97, TA98, TA100 and TA1537. Mutagenicity was seen in all strains when S9 was present.


PLOS ONE | 2014

Dicer Cooperates with p53 to Suppress DNA Damage and Skin Carcinogenesis in Mice

Stephen Lyle; Kathleen Hoover; Cansu Colpan; Zhiqing Zhu; Zdenka Matijasevic; Stephen N. Jones

Dicer is required for the maturation of microRNA, and loss of Dicer and miRNA processing has been found to alter numerous biological events during embryogenesis, including the development of mammalian skin and hair. We have previously examined the role of miRNA biogenesis in mouse embryonic fibroblasts and found that deletion of Dicer induces cell senescence regulated, in part, by the p53 tumor suppressor. Although Dicer and miRNA molecules are thought to have either oncogenic or tumor suppressing roles in various types of cancer, a role for Dicer and miRNAs in skin carcinogenesis has not been established. Here we show that perinatal ablation of Dicer in the skin of mice leads to loss of fur in adult mice, increased epidermal cell proliferation and apoptosis, and the accumulation of widespread DNA damage in epidermal cells. Co-ablation of Dicer and p53 did not alter the timing or extent of fur loss, but greatly reduced survival of Dicer-skin ablated mice, as these mice developed multiple and highly aggressive skin carcinomas. Our results describe a new mouse model for spontaneous basal and squamous cell tumorigenesis. Furthermore, our findings reveal that loss of Dicer in the epidermis induces extensive DNA damage, activation of the DNA damage response and p53-dependent apoptosis, and that Dicer and p53 cooperate to suppress mammalian skin carcinogenesis.


Oncogenesis | 2016

The Zn-finger domain of MdmX suppresses cancer progression by promoting genome stability in p53-mutant cells

Zdenka Matijasevic; Anna Krzywicka-Racka; Greenfield Sluder; Judith Gallant; Stephen N. Jones

The MDMX (MDM4) oncogene is amplified or overexpressed in a significant percentage of human tumors. MDMX is thought to function as an oncoprotein by binding p53 tumor suppressor protein to inhibit p53-mediated transcription, and by complexing with MDM2 oncoprotein to promote MDM2-mediated degradation of p53. However, down-regulation or loss of functional MDMX has also been observed in a variety of human tumors that are mutated for p53, often correlating with more aggressive cancers and a worse patient prognosis. We have previously reported that endogenous levels of MdmX can suppress proliferation and promote pseudo-bipolar mitosis in primary and tumor cells derived from p53-deficient mice, and that MdmX-p53 double deficient mice succumb to spontaneously formed tumors more rapidly than p53-deficient mice. These results suggest that the MdmX oncoprotein may act as a tumor-suppressor in cancers with compromised p53 function. By using orthotopic transplantation and lung colonization assays in mice we now establish a p53-independent anti-oncogenic role for MdmX in tumor progression. We also demonstrate that the roles of MdmX in genome stability and in proliferation are two distinct functions encoded by the separate MdmX protein domains. The central Zn-finger domain suppresses multipolar mitosis and chromosome loss, whereas the carboxy-terminal RING domain suppresses proliferation of p53-deficient cells. Furthermore, we determine that it is the maintenance of genome stability that underlies MdmX role in suppression of tumorigenesis in hyperploid p53 mutant tumors. Our results offer a rationale for the increased metastatic potential of p53 mutant human cancers with aberrant MdmX function and provide a caveat for the application of anti-MdmX treatment of tumors with compromised p53 activity.


Journal of Bacteriology | 1994

Induction of the Escherichia coli aidB gene under oxygen-limiting conditions requires a functional rpoS (katF) gene.

Michael R. Volkert; Laurel I. Hajec; Zdenka Matijasevic; Ferric C. Fang; Robert Prince


Proceedings of the National Academy of Sciences of the United States of America | 1992

Release of N2,3-ethenoguanine from chloroacetaldehyde-treated DNA by Escherichia coli 3-methyladenine DNA glycosylase II.

Zdenka Matijasevic; Mutsuo Sekiguchi; David B. Ludlum


Oncology Research | 1998

Hypothermia causes a reversible, p53-mediated cell cycle arrest in cultured fibroblasts.

Zdenka Matijasevic; Snyder Je; David B. Ludlum


Proceedings of the National Academy of Sciences of the United States of America | 1993

Protection against chloroethylnitrosourea cytotoxicity by eukaryotic 3-methyladenine DNA glycosylase

Zdenka Matijasevic; M Boosalis; W Mackay; L Samson; David B. Ludlum


Carcinogenesis | 2001

Repair of sulfur mustard-induced DNA damage in mammalian cells measured by a host cell reactivation assay.

Zdenka Matijasevic; Melissa Lynn Precopio; Jennifer E. Snyder; David B. Ludlum

Collaboration


Dive into the Zdenka Matijasevic's collaboration.

Top Co-Authors

Avatar

David B. Ludlum

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Michael R. Volkert

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Stephen N. Jones

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Allen Stering

University of Massachusetts Amherst

View shared research outputs
Top Co-Authors

Avatar

Errol Zeiger

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Greenfield Sluder

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Kathleen Hoover

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Laurel I. Hajec

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Paula Austin-Ritchie

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Qiong Li

University of Massachusetts Medical School

View shared research outputs
Researchain Logo
Decentralizing Knowledge