Network


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

Hotspot


Dive into the research topics where Myriam Maoz is active.

Publication


Featured researches published by Myriam Maoz.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Identification of tissue-specific cell death using methylation patterns of circulating DNA

Roni Lehmann-Werman; Daniel Neiman; Hai Zemmour; Joshua Moss; Judith Magenheim; Adi Vaknin-Dembinsky; Sten Rubertsson; Bengt Nellgård; Kaj Blennow; Henrik Zetterberg; Kirsty L. Spalding; Michael J. Haller; Clive Wasserfall; Desmond A. Schatz; Carla J. Greenbaum; Craig Dorrell; Markus Grompe; Aviad Zick; Ayala Hubert; Myriam Maoz; Volker Fendrich; Detlef K. Bartsch; Talia Golan; Shmuel Ben Sasson; Gideon Zamir; Aharon Razin; Howard Cedar; A. M. James Shapiro; Benjamin Glaser; Ruth Shemer

Significance We describe a blood test for detection of cell death in specific tissues based on two principles: (i) dying cells release fragmented DNA to the circulation, and (ii) each cell type has a unique DNA methylation pattern. We have identified tissue-specific DNA methylation markers and developed a method for sensitive detection of these markers in plasma or serum. We demonstrate the utility of the method for identification of pancreatic β-cell death in type 1 diabetes, oligodendrocyte death in relapsing multiple sclerosis, brain cell death in patients after traumatic or ischemic brain damage, and exocrine pancreas cell death in pancreatic cancer or pancreatitis. The approach allows minimally invasive monitoring of tissue dynamics in humans in multiple physiological and pathological conditions. Minimally invasive detection of cell death could prove an invaluable resource in many physiologic and pathologic situations. Cell-free circulating DNA (cfDNA) released from dying cells is emerging as a diagnostic tool for monitoring cancer dynamics and graft failure. However, existing methods rely on differences in DNA sequences in source tissues, so that cell death cannot be identified in tissues with a normal genome. We developed a method of detecting tissue-specific cell death in humans based on tissue-specific methylation patterns in cfDNA. We interrogated tissue-specific methylome databases to identify cell type-specific DNA methylation signatures and developed a method to detect these signatures in mixed DNA samples. We isolated cfDNA from plasma or serum of donors, treated the cfDNA with bisulfite, PCR-amplified the cfDNA, and sequenced it to quantify cfDNA carrying the methylation markers of the cell type of interest. Pancreatic β-cell DNA was identified in the circulation of patients with recently diagnosed type-1 diabetes and islet-graft recipients; oligodendrocyte DNA was identified in patients with relapsing multiple sclerosis; neuronal/glial DNA was identified in patients after traumatic brain injury or cardiac arrest; and exocrine pancreas DNA was identified in patients with pancreatic cancer or pancreatitis. This proof-of-concept study demonstrates that the tissue origins of cfDNA and thus the rate of death of specific cell types can be determined in humans. The approach can be adapted to identify cfDNA derived from any cell type in the body, offering a minimally invasive window for diagnosing and monitoring a broad spectrum of human pathologies as well as providing a better understanding of normal tissue dynamics.


The FASEB Journal | 2003

Oncogenic transformation induces tumor angiogenesis: a role for PAR1 activation

Yong-Jun Yin; Zaidoun Salah; Myriam Maoz; Sharona Cohen Even Ram; Shalom Ochayon; Gera Neufeld; Shulamit Katzav; Rachel Bar-Shavit

The formation of new blood vessels is a critical determinant of tumor progression. We find that Par1 gene expression plays a central role in blood vessel recruitment in animal models. By in vivo injection of either Matrigel plugs containing Par1‐expressing cells or of rat prostatic carcinoma cells transfected with tetracycline‐inducible Par1 expression vectors, we show that Par1 significantly enhances both angiogenesis and tumor growth. Several vascular endothelial growth factor (VEGF) splice forms are induced in cells expressing Par1. Activation of PAR1 markedly augments the expression of VEGF mRNAs and of functional VEGFs as determined by in vitro assays for endothelial tube alignment and bovine aortic endothelial cell proliferation. Because neutralizing anti‐VEGF antibodies potently inhibited Par1‐induced endothelial cell proliferation, we conclude that Par1‐induced angiogenesis requires VEGF. Specific inhibitors of protein kinase C (PKC), Src, and phosphatidylinositol 3‐kinase (PI3K) inhibit Par1‐induced VEGF expression, suggesting the participation of these kinases in the process. We also show that oncogenic transformation by genes known to be part of PAR1 signaling machinery is sufficient to increase VEGF expression in NIH 3T3 cells. These data support the novel notion that initiation of cell signaling either by activating PAR1 or by the activated forms of oncogenes is sufficient to induce VEGF and hence angiogenesis. Yin, Y.‐J., Salah, Z., Maoz, M., Cohen Even Ram, S., Ochayon, S., Neufeld, G., Katzav, S., Bar‐Shavit, R. Oncogenic transformation induces tumor angiogenesis: a role for PAR1 activation. FASEB J. 17, 163–174 (2003)


International Journal of Cancer | 2005

Differential expression of Protease activated receptor 1 (Par1) and pY397FAK in benign and malignant human ovarian tissue samples

Sorina Grisaru-Granovsky; Zaidoun Salah; Myriam Maoz; Diana Pruss; Uziel Beller; Rachel Bar-Shavit

Protease activated receptors (PAR) form a family of G‐protein coupled receptors (GPCR) encoding their own ligands and uniquely activated via proteolytic cleavage. Although proteases in general have been implicated in the remodeling of the extracellular tumor microenvironment, the role of cell surface receptors activated by proteolysis is now emerging. In our present study we investigated the expression pattern of protease activated receptor 1 hPar1 in ovarian carcinoma tissue samples. Abundant hPar1 mRNA and protein were detected in “low malignant potential” and in invasive carcinomas, regardless of the histological subtype. In contrast, no hPar1 expression was detected on the cell surface of normal ovarian epithelium. The differential expression pattern of hPar1 was shown by in situ hybridization, immunohistochemistry and semi‐quantitative RT‐PCR analyses. In early stages of ovarian carcinoma (Ia), the contra lateral normal ovary showed strong PAR1 expression as opposed to the lack of expression in the ovarian epithelium obtained from normal individuals. In parallel, we analyzed the expression pattern of αvβ5 integrin and of activated focal adhesion kinase (FAK), a major focal contact protein, in these tissues. Although abundant expression of αvβ5 integrin was observed in all tissues specimens examined, regardless of either normal or malignant, the level of activated FAK was differentially expressed. Phosphorylated FAK was seen in invasive ovarian carcinoma, but not in the normal ovarian epithelium. The abundant hPar1 levels in pathological malignant ovarian carcinoma is likely to transmit signals leading to the phosphorylation of FAK and thereby alterations in the integrin functional state. Altogether our data suggest that hPar1 and FAK cooperate to promote ovarian cancer malignancy.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2003

Human Protease-Activated Receptor 1 Expression in Malignant Epithelia: A Role in Invasiveness

Yong-Jun Yin; Zaidoun Salah; Sorina Grisaru-Granovsky; Irit Cohen; Sharona Even-Ram; Myriam Maoz; Beatrice Uziely; Tamar Peretz; Rachel Bar-Shavit

While protease-activated receptors (PARs) play a traditional role in vascular biology, they emerge with surprisingly new assignments in tumor biology. PAR1 expression correlates with the invasion properties of breast carcinoma, whereas human PAR1 antisense reduces their ability to migrate through Matrigel. Part of the molecular mechanism of PAR1 invasion involves the formation of focal contact complexes on PAR1 activation. PAR1 induces angiogenesis in animal models in vivo and exhibits an oncogenic phenotype of enhanced ductal complexity when overexpressed in mouse mammary glands.


Cancer Research | 2007

Transcriptional Regulation of Human Protease-Activated Receptor 1: A Role for the Early Growth Response-1 Protein in Prostate Cancer

Zaidoun Salah; Myriam Maoz; Gallina Pizov; Rachel Bar-Shavit

Transcriptional regulation plays a central role in the molecular pathways underlying preferential cancer growth and metastasis. In the present study, we investigated the regulation of human protease-activated receptor 1 (hPar1) gene overexpression in the malignant androgen hormone-resistant phase. We found increased hPar1 RNA chain elongation and no change in message stability in cells with high levels of PAR1 expression, indicating that increased transcription is largely responsible for the overexpression of hPar1 in prostate tumor progression. Enforced expression of early growth response-1 (Egr-1) plasmid markedly enhanced luciferase activity driven by the hPar1 promoter. The neuroendocrine peptide bombesin significantly induced hPar1 expression and increased the ability of the cells to invade Matrigel, an effect abolished by expression of hPar1 small interfering RNA, showing the importance of hPAR1 in invasion. Bombesin also markedly enhanced Egr-1 binding to the hPar1 promoter in vivo and in vitro. These data suggest that bombesin enhances Egr-1 expression leading to increased hPar1 transcription, thereby increasing PAR1 expression and function. Immunohistostaining of prostate tissue biopsy specimens revealed a direct correlation between the degree of prostate cancer malignancy, PAR1 expression, and EGR-1 expression. Altogether, we show that transcriptional regulation of hPar1 in the aggressive hormone-resistant prostate cancer stage is controlled in part by the transcription factor Egr-1 and may play a central role in invasiveness, an important indicator of malignancy.


Journal of Cellular Physiology | 2009

Protease activated receptor-1, PAR1, promotes placenta trophoblast invasion and β-catenin stabilization

Sorina Grisaru-Granovsky; Myriam Maoz; Oded Barzilay; Yong-Jun Yin; Diana Prus; Rachel Bar-Shavit

Despite extensive efforts toward elucidation of the molecular pathway controlling cytotrophoblast (CTB) invasion to the uterine decidua, it remains poorly defined. There are striking similarities between tumor cell invasion and cytotrophoblast implantation to the deciduas whereby the role of Protease Activated Receptors (PARs) and wnt signaling is well recognized. We examine here consequences of modulation of PAR1 and PAR2 expression and function on CTB invasion and β‐catenin stabilization. Toward this end, we utilized a model system of extravillous trophoblast (EVT) organ culture and various placenta cell lines (e.g., JAR and HTR‐8/Svneo). Activation of PAR1 induces EVT invasion while hPar1‐SiRNA and PAR1 antagonist SCH79797—effectively inhibited it. In parallel, the Wnt inhibitor Dickkopf‐1 (Dkk1) similarly inhibited it. Nuclear localization of β‐catenin is seen only after PAR1 activation, and is markedly reduced following the application of hPar1‐SiRNA construct and PAR1 antagonist in CTBs. In contrast, PAR2 elicited a low cytoplasmic β‐catenin level as also proliferation and invasion. In the non‐activated CTBs in‐comparison, β‐catenin appeared limited to the membrane pools. Concomitantly, a temporal regulated pattern of Wnt‐4, 5a, 7b, 10a, 10b expression is seen along PAR1 appearance. Enforced expression of Wnt antagonists, Secreted Frizzled Related Proteins; SFRP2 & 5; into HTR‐8/Svneo, resulted with a markedly reduced nuclear β‐catenin levels, similar to the effect obtained by hPar1‐SiRNA treatment. Identification of PAR1 downstream target/s may nonetheless contribute to the formation of a future platform system for eliciting a firm placenta‐uterus interactions and to the definition of late pregnancy outcomes. J. Cell. Physiol. 218: 512–521, 2009.


International Journal of Molecular Sciences | 2016

G Protein-Coupled Receptors in Cancer

Rachel Bar-Shavit; Myriam Maoz; Arun Kancharla; Jeetendra Kumar Nag; Daniel Agranovich; Sorina Grisaru-Granovsky; Beatrice Uziely

Despite the fact that G protein-coupled receptors (GPCRs) are the largest signal-conveying receptor family and mediate many physiological processes, their role in tumor biology is underappreciated. Numerous lines of evidence now associate GPCRs and their downstream signaling targets in cancer growth and development. Indeed, GPCRs control many features of tumorigenesis, including immune cell-mediated functions, proliferation, invasion and survival at the secondary site. Technological advances have further substantiated GPCR modifications in human tumors. Among these are point mutations, gene overexpression, GPCR silencing by promoter methylation and the number of gene copies. At this point, it is imperative to elucidate specific signaling pathways of “cancer driver” GPCRs. Emerging data on GPCR biology point to functional selectivity and “biased agonism”; hence, there is a diminishing enthusiasm for the concept of “one drug per GPCR target” and increasing interest in the identification of several drug options. Therefore, determining the appropriate context-dependent conformation of a functional GPCR as well as the contribution of GPCR alterations to cancer development remain significant challenges for the discovery of dominant cancer genes and the development of targeted therapeutics.


Molecular Cancer Research | 2007

Protease-Activated Receptor-1 (hPar1), A Survival Factor Eliciting Tumor Progression

Zaidoun Salah; Myriam Maoz; Elisheva Pokroy; Michal Lotem; Rachel Bar-Shavit; Beatrice Uziely

Although ample evidence point to the central involvement of protease activated receptor-1 (PAR1) in tumor progression, little is known about the fate of the tumor when hPar1 is being silenced. We observed that hPar1 antisense clones exhibit low PAR1 levels, attenuated cell proliferation and invasion in vitro, and tumor formation in vivo. These clones showed noticeably reduced paxillin phosphorylation compared with the parental A375SM cells, whereas no change in the integrin levels was noticed. Antisense clones injected into the mice resulted in very few and only occasional small tumors, whereas advanced and vascularized tumors were observed in A375SM cells. The antisense-derived tumor sections expressed active caspase-3, increased terminal deoxynucleotidyl transferase–mediated nick-end labeling staining, and a markedly reduced proliferating cell nuclear antigen level compared with A375SM cell–derived tissue sections. Likewise, ablation of the hPar1 gene in a tetracycline-inducible hPar1 system leads to apoptosis in immature blood vessels, whereas mature vessels were unaffected. The activation of PAR1-induced pAkt/protein kinase B abrogated serum-deprived BimEL induction and also markedly inhibited Bax levels. On the other hand, small interfering RNA silencing of the hPar1 gene induced the expression of BimEL, a direct substrate of Akt/protein kinase B and also induced expression of active caspase-9 and caspase-3. These results altogether identify PAR1 as a survival factor that protects cells from undergoing apoptosis. We conclude that whereas PAR1 gene expression correlates with tumor progression, its neutralization effectively initiates an apoptotic pathway leading at least in part to significantly reduced tumor formation. (Mol Cancer Res 2007;5(3):229–40)


The FASEB Journal | 2005

Identification of a novel functional androgen response element within hPar1 promoter: implications to prostate cancer progression

Zaidoun Salah; Myriam Maoz; Irit Cohen; Galina Pizov; Dov Pode; Marschall S. Runge; Rachel Bar-Shavit

Human protease‐activated receptor‐1 (hPar1) plays a role in malignant and physiological invasion processes. We have identified a functional androgen response element (ARE) located in the hPar1 promoter upstream of the transcription start site at ‐1791 to ‐1777. Dihydrotestosterone treatment of the prostate cancer cell line LNCaP increased endogenous hPar1 mRNA levels, consistent with the threefold increase in promoter activity of hPar1‐luciferase reporter con‐ struct. Specific binding of the hPar1‐derived ARE to LNCaP nuclear extracts was demonstrated by electro‐ phoretic mobility shift assay. This binding was abro‐ gated by antiandrogen receptor (anti‐AR) antibodies or excess cold oligonucleotide but not by a mutated oligonucleotide. Moreover, using chromatin immuno‐ precipitation assays, we confirm the in vivo interaction between the AR and ARE domain of the hPar1 pro‐ moter. In parallel, we show that hormone ablation therapy markedly reduces the otherwise high hPar1 expression levels in prostate cancer biopsy specimens. We suggest that the hPar1 gene is regulated transcrip‐ tionally by androgens, representing one of several target genes effectively reduced during hormone abla‐ tion therapy. A major limitation of hormonal depriva‐ tion is that it causes only a temporary remission, and the cancer eventually reappears in a more malignant, androgen‐independent form. hPar1 is also overex‐ pressed in CL1 cells, an aggressively metastasizing, hormone‐independent subclone of LNCaP, and in PC3 prostate adenocarcinoma lacking AR in a mechanism yet to be fully elucidated. These data may imply that hPar1 expression correlates with prostate cancer pro‐ gression in androgen‐dependent and ‐independent phases and therefore, provides an instrumental, thera‐ peutic target for treatment in prostate cancer. FASEBJ. 19, 62‐72 (2005)


Journal of Biological Chemistry | 2010

Protease-activated Receptor-1 (PAR1) Acts via a Novel Gα13-Dishevelled Axis to Stabilize β-Catenin Levels

Hagit Turm; Myriam Maoz; Vered Katz; Yong-Jun Yin; Steffan Offermanns; Rachel Bar-Shavit

We have previously shown a novel link between hPar-1 (human protease-activated receptor-1) and β-catenin stabilization. Although it is well recognized that Wnt signaling leads to β-catenin accumulation, the role of PAR1 in the process is unknown. We provide here evidence that PAR1 induces β-catenin stabilization independent of Wnt, Fz (Frizzled), and the co-receptor LRP5/6 (low density lipoprotein-related protein 5/6) and identify selective mediators of the PAR1-β-catenin axis. Immunohistological analyses of hPar1-transgenic (TG) mouse mammary tissues show the expression of both Gα12 and Gα13 compared with age-matched control counterparts. However, only Gα13 was found to be actively involved in PAR1-induced β-catenin stabilization. Indeed, a dominant negative form of Gα13 inhibited both PAR1-induced Matrigel invasion and Lef/Tcf (lymphoid enhancer factor/T cell factor) transcription activity. PAR1-Gα13 association is followed by the recruitment of DVL (Dishevelled), an upstream Wnt signaling protein via the DIX domain. Small interfering RNA-Dvl silencing leads to a reduction in PAR1-induced Matrigel invasion, inhibition of Lef/Tcf transcription activity, and decreased β-catenin accumulation. It is of note that PAR1 also promotes the binding of β-arrestin-2 to DVL, suggesting a role for β-arrestin-2 in PAR1-induced DVL phosphorylation dynamics. Although infection of small interfering RNA-LRP5/6 or the use of the Wnt antagonists, SFRP2 (soluble Frizzled-related protein 2) or SFRP5 potently reduced Wnt3A-mediated β-catenin accumulation, no effect was observed on PAR1-induced β-catenin stabilization. Collectively, our data show that PAR1 mediates β-catenin stabilization independent of Wnt. We propose here a novel cascade of PAR1-induced Gα13-DVL axis in cancer and β-catenin stabilization.

Collaboration


Dive into the Myriam Maoz's collaboration.

Top Co-Authors

Avatar

Rachel Bar-Shavit

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Beatrice Uziely

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zaidoun Salah

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Hagit Turm

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Irit Cohen

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Aviad Zick

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Jeetendra Kumar Nag

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Tamar Peretz

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Arun Kancharla

Hebrew University of Jerusalem

View shared research outputs
Researchain Logo
Decentralizing Knowledge