Network


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

Hotspot


Dive into the research topics where Ewa Zajac is active.

Publication


Featured researches published by Ewa Zajac.


American Journal of Pathology | 2011

Tumor-recruited neutrophils and neutrophil TIMP-free MMP-9 regulate coordinately the levels of tumor angiogenesis and efficiency of malignant cell intravasation.

Erin M. Bekes; Bernhard Schweighofer; Tatyana A. Kupriyanova; Ewa Zajac; Veronica C. Ardi; James P. Quigley; Elena I. Deryugina

Tumor-associated neutrophils contribute to neovascularization by supplying matrix metalloproteinase-9 (MMP-9), a protease that has been genetically and biochemically linked to induction of angiogenesis. Specific roles of inflammatory neutrophils and their distinct proMMP-9 in the coordinate regulation of tumor angiogenesis and tumor cell dissemination, however, have not been addressed. We demonstrate that the primary tumors formed by highly disseminating variants of human fibrosarcoma and prostate carcinoma recruit elevated levels of infiltrating MMP-9-positive neutrophils and concomitantly exhibit enhanced levels of angiogenesis and intravasation. Specific inhibition of neutrophil influx by interleukin 8 (IL-8) neutralization resulted in the coordinated diminishment of tumor angiogenesis and intravasation, both of which were rescued by purified neutrophil proMMP-9. However, if neutrophil proMMP-9, naturally devoid of tissue inhibitor of metalloproteinases (TIMP), was delivered in complex with TIMP-1 or in a mixture with TIMP-2, the protease failed to rescue the inhibitory effects of anti-IL8 therapy, indicating that the TIMP-free status of proMMP-9 is critical for facilitating tumor angiogenesis and intravasation. Our findings directly link tumor-associated neutrophils and their TIMP-free proMMP-9 with the ability of aggressive tumor cells to induce the formation of new blood vessels that serve as conduits for tumor cell dissemination. Thus, treatment of cancers associated with neutrophil infiltration may benefit from specific targeting of neutrophil MMP-9 at early stages to prevent ensuing tumor angiogenesis and tumor metastasis.


Blood | 2013

Angiogenic capacity of M1- and M2-polarized macrophages is determined by the levels of TIMP-1 complexed with their secreted proMMP-9

Ewa Zajac; Bernhard Schweighofer; Tatyana A. Kupriyanova; Anna Juncker-Jensen; Petra Minder; James P. Quigley; Elena I. Deryugina

A proangiogenic function of tissue-infiltrating monocytes/macrophages has long been attributed to their matrix metalloproteinase-9 zymogen (proMMP-9). Herein, we evaluated the capacity of human monocytes, mature M0 macrophages, and M1- and M2-polarized macrophages to induce proMMP-9-mediated angiogenesis. Only M2 macrophages induced angiogenesis at levels comparable with highly angiogenic neutrophils previously shown to release their proMMP-9 in a unique form, free of tissue inhibitor of metalloproteinases-1 (TIMP-1). Macrophage differentiation was accompanied by induction of low-angiogenic, TIMP-1-encumbered proMMP-9. However, polarization toward the M2, but not the M1 phenotype, caused a substantial downregulation of TIMP-1 expression, resulting in production of angiogenic, TIMP-deficient proMMP-9. Correspondingly, the angiogenic potency of M2 proMMP-9 was lost after its complexing with TIMP-1, whereas TIMP-1 silencing in M0/M1 macrophages rendered them both angiogenic. Similar to human cells, murine bone marrow-derived M2 macrophages also shut down their TIMP-1 expression and produced proMMP-9 unencumbered by TIMP-1. Providing proof that angiogenic capacity of murine M2 macrophages depended on their TIMP-free proMMP-9, Mmp9-null M2 macrophages were nonangiogenic, although their TIMP-1 was severely downregulated. Our study provides a unifying molecular mechanism for high angiogenic capacity of TIMP-free proMMP-9 that would be uniquely produced in a pathophysiological microenvironment by influxing neutrophils and/or M2 polarized macrophages.


Neoplasia | 2014

Tissue-infiltrating neutrophils constitute the major in vivo source of angiogenesis-inducing MMP-9 in the tumor microenvironment.

Elena I. Deryugina; Ewa Zajac; Anna Juncker-Jensen; Tatyana A. Kupriyanova; Lisa Welter; James P. Quigley

According to established notion, one of the major angiogenesis-inducing factors, pro-matrix metalloproteinase-9 (proMMP-9), is supplied to the tumor microenvironment by tumor-associated macrophages (TAMs). Accumulated evidence, however, indicates that tumor-associated neutrophils (TANs) are also critically important for proMMP-9 delivery, especially at early stages of tumor development. To clarify how much angiogenic proMMP-9 is actually contributed by TAMs and TANs, we quantitatively evaluated TAMs and TANs from different tumor types, including human xenografts and syngeneic murine tumors grown in wild-type and Mmp9-knockout mice. Whereas host MMP-9 competence was required for full angiogenic potential of both normal and tumor-associated leukocytes, direct comparisons of neutrophils versus macrophages and TANs versus TAMs demonstrated that macrophages and TAMs secrete 40- to 50-fold less proMMP-9 than the same numbers of neutrophils or TANs. Correspondingly, the levels of MMP-9–mediated in vivo angiogenesis induced by neutrophils and TANs substantially exceeded those induced by macrophages and TAMs. MMP-9–delivering TANs were also required for development of metastasis-supporting intratumoral vasculature, characterized by ≥ 11-μm size lumens and partial coverage with stabilizing pericytes. Importantly, MMP-9–producing TAMs exhibit M2-skewed phenotype but do not express tissue inhibitor of metalloproteinases-1 (TIMP-1), a novel characteristic allowing them to secrete TIMP-1–free, neutrophil-like MMP-9 zymogen unencumbered by its natural inhibitor. Together, our findings support the notion whereby TANs, capable of immediate release of their pre-stored cargo, are the major contributors of highly angiogenic MMP-9, whereas tumor-influxing precursors of macrophages require time to differentiate, polarize into M2-skewed TAMs, shut down their TIMP-1 expression, and only then, initiate relatively low-level production of TIMP-free MMP-9 zymogen.


Cancer Research | 2013

Tumor MMP-1 Activates Endothelial PAR1 to Facilitate Vascular Intravasation and Metastatic Dissemination

Anna Juncker-Jensen; Elena I. Deryugina; Ivo Rimann; Ewa Zajac; Tatyana A. Kupriyanova; Lars H. Engelholm; James P. Quigley

Intravasation, the active entry of primary tumor cells into the vasculature, remains the least studied step in the metastatic cascade. Protease-mediated escape and stromal invasion of tumor cells represent widely accepted processes leading up to the intravasation step. However, molecular factors that contribute directly to tumor cell vascular penetration have not been identified. In this study, the in vivo role of the collagenolytic protease, MMP-1, in cancer cell intravasation and metastasis was analyzed by using a highly disseminating variant of human HEp3 epidermoid carcinoma, HEp3-hi/diss. Although naturally acquired or experimentally induced MMP-1 deficiency substantially suppressed HEp3-hi/diss intravasation, supplementation of recombinant MMP-1 to MMP-1-silenced primary tumors restored their impaired vascular dissemination. Surprisingly, abrogation of MMP-1 production and activity did not significantly affect HEp3-hi/diss migration or matrix invasion, suggesting noncollagenolytic mechanisms underlying MMP-1-dependent cell intravasation. In support of such noncollagenolytic mechanisms, MMP-1 silencing in HEp3-hi/diss cells modulated the microarchitecture and integrity of the angiogenic vasculature in a novel microtumor model. Concomitantly, MMP-1 deficiency led to decreased levels of intratumoral vascular permeability, tumor cell intravasation, and metastatic dissemination. Taking advantage of PAR1 deficiency of HEp3-hi/diss cells, we further show that endothelial PAR1 is a putative nontumor-cell/nonmatrix target, activation of which by carcinoma-produced MMP-1 regulates endothelial permeability and transendothelial migration. The inhibitory effects of specific PAR1 antagonists in live animals have also indicated that the mechanisms of MMP-1-dependent vascular permeability in tumors involve endothelial PAR1 activation. Together, our findings mechanistically underscore the contribution of a tumor MMP-1/endothelial PAR1 axis to actual intravasation events manifested by aggressive carcinoma cells.


Neoplasia | 2015

EGFR Regulates the Development and Microarchitecture of Intratumoral Angiogenic Vasculature Capable of Sustaining Cancer Cell Intravasation

Petra Minder; Ewa Zajac; James P. Quigley; Elena I. Deryugina

Many malignant characteristics of cancer cells are regulated through pathways induced by the tyrosine kinase activity of the epidermal growth factor receptor (EGFR). Herein, we show that besides directly affecting the biology of cancer cells per se, EGFR also regulates the primary tumor microenvironment. Specifically, our findings demonstrate that both the expression and signaling activity of EGFR are required for the induction of a distinct intratumoral vasculature capable of sustaining tumor cell intravasation, a critical rate-limiting step in the metastatic cascade. An intravasation-sustaining mode of intratumoral angiogenic vessels depends on high levels of tumor cell EGFR and the interplay between EGFR-regulated production of interleukin 8 by tumor cells, interleukin-8–induced influx of tumor-infiltrating neutrophils delivering their unique matrix metalloproteinase-9, and neutrophil matrix metalloproteinase-9–dependent release of the vascular permeability and endothelial growth factor, VEGF. Our data indicate that through VEGF-mediated disruption of endothelial layer integrity and increase of intratumoral vasculature permeability, EGFR activity significantly facilitates active intravasation of cancer cells. Therefore, this study unraveled an important but overlooked function of EGFR in cancer, namely, its ability to create an intravasation-sustaining microenvironment within the developing primary tumor by orchestrating several interrelated processes required for the initial steps of cancer metastasis through vascular routes. Our findings also suggest that EGFR-targeted therapies might be more effective when implemented in cancer patients with early-staged primary tumors containing a VEGF-dependent angiogenic vasculature. Accordingly, early EGFR inhibition combined with various anti-VEGF approaches could synergistically suppress tumor cell intravasation through inhibiting the highly permeable angiogenic vasculature induced by EGFR-overexpressing aggressive cancer cells.


Oncogene | 2018

LTBP3 promotes early metastatic events during cancer cell dissemination

Elena I. Deryugina; Ewa Zajac; Lior Zilberberg; Tomoki Muramatsu; Grishma Joshi; Branka Dabovic; Daniel B. Rifkin; James P. Quigley

Latent transforming growth factor β (TGFβ)-binding proteins (LTBPs) are important for the secretion, activation, and function of mature TGFβ, especially so in cancer cell physiology. However, specific roles of the LTBPs remain understudied in the context of the primary tumor microenvironment. Herein, we investigated the role of LTBP3 in the distinct processes involved in cancer metastasis. By using three human tumor cell lines of different tissue origin (epidermoid HEp-3 and prostate PC-3 carcinomas and HT-1080 fibrosarcoma) and several metastasis models conducted in both mammalian and avian settings, we show that LTBP3 is involved in the early dissemination of primary cancer cells, namely in the intravasation step of the metastatic cascade. Knockdown of LTBP3 in all tested cell lines led to significant inhibition of tumor cell intravasation, but did not affect primary tumor growth. LTBP3 was dispensable in the late steps of carcinoma cell metastasis that follow tumor cell intravasation, including vascular arrest, extravasation, and tissue colonization. However, LTBP3 depletion diminished the angiogenesis-inducing potential of HEp-3 cells in vivo, which was restorable by exogenous delivery of LTBP3 protein. A similar compensatory approach rescued the dampened intravasation of LTBP3-deficient HEp-3 cells, suggesting that LTBP3 regulates the induction of the intravasation-supporting angiogenic vasculature within developing primary tumors. Using our recently developed microtumor model, we confirmed that LTBP3 loss resulted in the development of intratumoral vessels with an abnormal microarchitecture incompatible with efficient intravasation of HEp-3 carcinoma cells. Collectively, these findings demonstrate that LTBP3 represents a novel oncotarget that has distinctive functions in the regulation of angiogenesis-dependent tumor cell intravasation, a critical process during early cancer dissemination. Our experimental data are also consistent with the survival prognostic value of LTBP3 expression in early-stage head and neck squamous cell carcinomas, further indicating a specific role for LTBP3 in cancer progression toward metastatic disease.


Cancer Research | 2013

Abstract C18: Matrix metalloproteinase-1 (MMP-1) facilitates intravasation and vascular dissemination of human carcinoma cells by regulating the microarchitecture and permeability of intratumoral vasculature

Anna Juncker-Jensen; Berta Casar; Ivo Rimann; Ewa Zajac; Tatyana A. Kupriyanova; Elena I. Deryugina; James P. Quigley

Patients with squamous cell carcinomas (SCCs) often present with advanced stage disease and face one of the lowest 5-year post-therapeutic survival rates. Among 25 signature genes, Mmp1 has been found to be third best SCC tumor predictor, suggesting a critical role of MMP-1 in SCC progression. Functional role of MMP-1 in SCCs was analyzed in this study by employing a highly-disseminating variant of human epidermoid HEp-3 carcinoma, HEp3-hi/diss. The dissemination capacity of HEp3-hi/diss cells in chick embryo spontaneous metastasis models positively correlated with their high levels of MMP-1 expression. MMP-1-deficiency induced by RNA interference resulted in 70-95% inhibition of HEp3-hi/diss intravasation, concomitant with collapse of the intravasation-sustaining microarchitecture of intratumoral vasculature. Furthermore, we have demonstrated the mechanism whereby HEp3-hi/diss-produced MMP-1 contributes to tumor cell intravasation by proteolytically inducing vascular endothelial permeability and increasing rates of carcinoma cells crossing over vascular barriers during the intravasation step of the metastatic cascade. Citation Format: Anna Juncker-Jensen, Berta Casar, Ivo Rimann, Ewa Zajac, Tatyana A. Kupriyanova, Elena I. Deryugina, James P. Quigley. Matrix metalloproteinase-1 (MMP-1) facilitates intravasation and vascular dissemination of human carcinoma cells by regulating the microarchitecture and permeability of intratumoral vasculature. [abstract]. In: Proceedings of the AACR Special Conference on Tumor Invasion and Metastasis; Jan 20-23, 2013; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2013;73(3 Suppl):Abstract nr C18.


Cancer Research | 2013

Abstract PR10: High angiogenic capacity of inflammatory neutrophils and polarized M2 macrophages is determined by production of MMP-9 unencumbered by its natural inhibitor TIMP-1

Ewa Zajac; Bernhard Schweighofer; Berta Casar; Tatyana A. Kupriyanova; Ivo Rimann; Anna Juncker-Jensen; Elena I. Deryugina; James P. Quigley

By delivering a potent matrix metalloproteinase, MMP-9, inflammatory neutrophils and macrophages play an important role in tumor angiogenesis. We have previously demonstrated that high angiogenic capacity of neutrophil MMP-9 (nMMP-9) is determined by its TIMP1-free status. In this study, we compared biochemically and functionally nMMP-9 with the MMP-9 produced by monocytes and macrophages of innate (M0), tumor-suppressing (M1) and tumor-promoting (M2) phenotypes. The angiogenic potential of a specific leukocyte type correlated with amount of TIMP-1 complexed with secreted proMMP-9. Similar to neutrophils, M2-macrophages produce proMMP-9 complexed with very low amounts of TIMP-1, rendering its high angiogenic capacity. In contrast, M0 macrophages and M1-polarized macrophages secrete non-angiogenic proMMP-9 complexed with TIMP-1 at near stoichiometric ratios. However, silencing of TIMP-1 in M0- and M1-macrophages results in induction of their angiogenic potential, thereby supporting a biochemical mechanism for unique angiogenic capacity of proMMP-9 delivered by neutrophils and a distinct subset of pro-tumorigenic macrophages. This abstract is also presented as Poster B89. Citation Format: Ewa Zajac, Bernhard Schweighofer, Berta Casar, Tatyana Kupriyanova, Ivo Rimann, Anna Juncker-Jensen, Elena Deryugina, James P. Quigley. High angiogenic capacity of inflammatory neutrophils and polarized M2 macrophages is determined by production of MMP-9 unencumbered by its natural inhibitor TIMP-1. [abstract]. In: Proceedings of the AACR Special Conference on Tumor Invasion and Metastasis; Jan 20-23, 2013; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2013;73(3 Suppl):Abstract nr PR10.


Neoplasia | 2011

Activation of Pro-uPA Is Critical for Initial Escape from the Primary Tumor and Hematogenous Dissemination of Human Carcinoma Cells

Erin M. Bekes; Elena I. Deryugina; Tatyana A. Kupriyanova; Ewa Zajac; Kenneth A. Botkjaer; Peter A. Andreasen; James P. Quigley


Biochemical Journal | 2015

Circular trimers of gelatinase B/matrix metalloproteinase-9 constitute a distinct population of functional enzyme molecules differentially regulated by tissue inhibitor of metalloproteinases-1.

Jennifer Vandooren; Benjamin Born; Inna Solomonov; Ewa Zajac; Radka Saldova; Michael Senske; Estefanía Ugarte-Berzal; Erik Martens; Philippe E. Van den Steen; Jo Van Damme; Angeles García-Pardo; Matheus Froeyen; Elena I. Deryugina; James P. Quigley; Søren K. Moestrup; Pauline M. Rudd; Irit Sagi; Ghislain Opdenakker

Collaboration


Dive into the Ewa Zajac's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

James P. Quigley

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ivo Rimann

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Berta Casar

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Erin M. Bekes

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Lisa Welter

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Petra Minder

Scripps Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge