Network


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

Hotspot


Dive into the research topics where Antonia Marazioti is active.

Publication


Featured researches published by Antonia Marazioti.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Hydrogen sulfide is an endogenous stimulator of angiogenesis

Andreas Papapetropoulos; Anastasia Pyriochou; Zaid Altaany; Guangdong Yang; Antonia Marazioti; Zongmin Zhou; Mark G. Jeschke; Ludwik K. Branski; David N. Herndon; Rui Wang; Csaba Szabó

The goal of the current study was to investigate the role of exogenous and endogenous hydrogen sulfide (H2S) on neovascularization and wound healing in vitro and in vivo. Incubation of endothelial cells (ECs) with H2S enhanced their angiogenic potential, evidenced by accelerated cell growth, migration, and capillary morphogenesis on Matrigel. Treatment of chicken chorioallantoic membranes (CAMS) with H2S increased vascular length. Exposure of ECs to H2S resulted in increased phosphorylation of Akt, ERK, and p38. The KATP channel blocker glibenclamide or the p38 inhibitor SB203580 abolished H2S-induced EC motility. Since glibenclamide inhibited H2S-triggered p38 phosphorylation, we propose that KATP channels lay upstream of p38 in this process. When CAMs were treated with H2S biosynthesis inhibitors dl-propylargylglycine or beta-cyano-L-alanine, a reduction in vessel length and branching was observed, indicating that H2S serves as an endogenous stimulator of the angiogenic response. Stimulation of ECs with vascular endothelial growth factor (VEGF) increased H2S release, while pharmacological inhibition of H2S production or KATP channels or silencing of cystathionine gamma-lyase (CSE) attenuated VEGF signaling and migration of ECs. These results implicate endothelial H2S synthesis in the pro-angiogenic action of VEGF. Aortic rings isolated from CSE knockout mice exhibited markedly reduced microvessel formation in response to VEGF when compared to wild-type littermates. Finally, in vivo, topical administration of H2S enhanced wound healing in a rat model, while wound healing was delayed in CSE−/− mice. We conclude that endogenous and exogenous H2S stimulates EC-related angiogenic properties through a KATP channel/MAPK pathway.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2011

Inhibition of nitric oxide-stimulated vasorelaxation by carbon monoxide-releasing molecules

Antonia Marazioti; Mariarosaria Bucci; Ciro Coletta; Valentina Vellecco; Padmamalini Baskaran; Csaba Szabó; Giuseppe Cirino; Ana R. Marques; Bruno Guerreiro; Ana M. L. Gonçalves; João Seixas; Annie Beuve; Carlos C. Romão; Andreas Papapetropoulos

Objective—Carbon monoxide (CO) is a weak soluble guanylyl cyclase stimulator, leading to transient increases in cGMP and vasodilation. The aim of the present work was to measure the effect of CO-releasing molecules (CORMs) on the cGMP/nitric oxide (NO) pathway and to evaluate how selected CORMs affect NO-induced vasorelaxation. Methods and Results—Incubation of smooth muscle cells with some but not all of the CORMs caused a minor increase in cGMP levels. Concentration-response curves were bell-shaped, with higher CORMs concentrations producing lower increases in cGMP levels. Although exposure of cells to CORM-2 enhanced cGMP formation, we observed that the compound inhibited NO-stimulated cGMP accumulation in cells and NO-stimulated soluble guanylyl cyclase activity that could be reversed by superoxide anion scavengers. Reactive oxygen species generation from CORMs was confirmed using luminol-induced chemiluminescence and electron spin resonance. Furthermore, we observed that NO is scavenged by CORM-2. When used alone CORM-2 relaxed vessels through a cGMP-mediated pathway but attenuated NO donor-stimulated vasorelaxation. Conclusion—We conclude that the CORMs examined have context-dependent effects on vessel tone, as they can directly dilate blood vessels, but also block NO-induced vasorelaxation.


Journal of Clinical Investigation | 2015

Mast cells mediate malignant pleural effusion formation

Anastasios D. Giannou; Antonia Marazioti; Magda Spella; Nikolaos Kanellakis; Hara Apostolopoulou; Ioannis Psallidas; Zeljko M. Prijovich; Malamati Vreka; Dimitra Zazara; Ioannis Lilis; Vassilios Papaleonidopoulos; Chrysoula A. Kairi; Alexandra L. Patmanidi; Ioanna Giopanou; Nikolitsa Spiropoulou; Vaggelis Harokopos; Vassilis Aidinis; Dionisios Spyratos; Stamatia Teliousi; Helen Papadaki; Stavros Taraviras; Linda A. Snyder; Oliver Eickelberg; Dimitrios Kardamakis; Yoichiro Iwakura; Thorsten B. Feyerabend; Hans Reimer Rodewald; Ioannis Kalomenidis; Timothy S. Blackwell; Theodora Agalioti

Mast cells (MCs) have been identified in various tumors; however, the role of these cells in tumorigenesis remains controversial. Here, we quantified MCs in human and murine malignant pleural effusions (MPEs) and evaluated the fate and function of these cells in MPE development. Evaluation of murine MPE-competent lung and colon adenocarcinomas revealed that these tumors actively attract and subsequently degranulate MCs in the pleural space by elaborating CCL2 and osteopontin. MCs were required for effusion development, as MPEs did not form in mice lacking MCs, and pleural infusion of MCs with MPE-incompetent cells promoted MPE formation. Once homed to the pleural space, MCs released tryptase AB1 and IL-1β, which in turn induced pleural vasculature leakiness and triggered NF-κB activation in pleural tumor cells, thereby fostering pleural fluid accumulation and tumor growth. Evaluation of human effusions revealed that MCs are elevated in MPEs compared with benign effusions. Moreover, MC abundance correlated with MPE formation in a human cancer cell-induced effusion model. Treatment of mice with the c-KIT inhibitor imatinib mesylate limited effusion precipitation by mouse and human adenocarcinoma cells. Together, the results of this study indicate that MCs are required for MPE formation and suggest that MC-dependent effusion formation is therapeutically addressable.


PLOS ONE | 2013

Beneficial Impact of CCL2 and CCL12 Neutralization on Experimental Malignant Pleural Effusion

Antonia Marazioti; Chrysoula A. Kairi; Magda Spella; Anastasios D. Giannou; Sophia Magkouta; Ioanna Giopanou; Vassilios Papaleonidopoulos; Ioannis Kalomenidis; Linda A. Snyder; Dimitrios Kardamakis; Georgios T. Stathopoulos

Using genetic interventions, we previously determined that C-C motif chemokine ligand 2 (CCL2) promotes malignant pleural effusion (MPE) formation in mice. Here we conducted preclinical studies aimed at assessing the specific therapeutic potential of antibody-mediated CCL2 blockade against MPE. For this, murine MPEs or skin tumors were generated in C57BL/6 mice by intrapleural or subcutaneous delivery of lung (LLC) or colon (MC38) adenocarcinoma cells. Human lung adenocarcinoma cells (A549) were used to induce MPEs in severe combined immunodeficient mice. Intraperitoneal antibodies neutralizing mouse CCL2 and/or CCL12, a murine CCL2 ortholog, were administered at 10 or 50 mg/kg every three days. We found that high doses of CCL2/12 neutralizing antibody treatment (50 mg/kg) were required to limit MPE formation by LLC cells. CCL2 and CCL12 blockade were equally potent inhibitors of MPE development by LLC cells. Combined CCL2 and CCL12 neutralization was also effective against MC38-induced MPE and prolonged the survival of mice in both syngeneic models. Mouse-specific CCL2-blockade limited A549-caused xenogeneic MPE, indicating that host-derived CCL2 also contributes to MPE precipitation in mice. The impact of CCL2/12 antagonism was associated with inhibition of immune and vascular MPE-related phenomena, such as inflammation, new blood vessel assembly and plasma extravasation into the pleural space. We conclude that CCL2 and CCL12 blockade are effective against experimental MPE induced by murine and human adenocarcinoma in mice. These results suggest that CCL2-targeted therapies may hold promise for future use against human MPE.


PLOS ONE | 2015

Comprehensive Evaluation of Nuclear Factor-κΒ Expression Patterns in Non-Small Cell Lung Cancer.

Ioanna Giopanou; Ioannis Lilis; Vassilios Papaleonidopoulos; Antonia Marazioti; Magda Spella; Malamati Vreka; Helen Papadaki; Georgios T. Stathopoulos

Nuclear factor (NF)-κB signalling is required for lung adenocarcinoma development in mice, and both of its subunits RelA and RelB were independently reported to be highly expressed in human non-small cell lung cancer (NSCLC). To comprehensively examine NF-κB expression in NSCLC, we analyzed serial sections of primary tumor samples from 77 well-documented patients (36 adenocarcinomas, 40 squamous cell carcinomas and 3 large cell carcinomas) for immunoreactivity of RelA, RelB, P50, and P52/P100. Tumor and intratumoral stroma areas were discriminated based on proliferating cell nuclear antigen immunoreactivity and inflammatory infiltration was assessed in intratumoral stroma areas. NF-κB immunoreactivity was quantified by intensity, extent, and nuclear localization and was cross-examined with tumor cell proliferation, inflammatory infiltration, and clinical-pathologic data. We found that the expression of the different NF-κB subunits was not concordant, warranting our integral approach. Overall, RelA, RelB, and P50 were expressed at higher levels compared with P52/P100. However, RelA and P50 were predominantly expressed in intratumoral stroma, but RelB in tumor cells. Importantly, tumor area RelA expression was correlated with the intensity of inflammatory infiltration, whereas RelB expression was identified in proliferating tumor cells. Using multiple logistic regression, we identified that tumor RelB expression was an independent predictor of lymph node metastasis, and tumor P50 was an independent predictor of TNM6 stage IIB or higher, whereas tumor RelA was an independent predictor of inflammatory infiltration. We conclude that pathologic studies of NF-κB expression in cancer should include multiple pathway components. Utilizing such an approach, we identified intriguing associations between distinct NF-κB subunits and clinical and pathologic features of NSCLC.


Nature Communications | 2017

Mutant KRAS promotes malignant pleural effusion formation

Theodora Agalioti; Anastasios D. Giannou; Anthi Krontira; Nikolaos Kanellakis; Danai Kati; Malamati Vreka; Mario Pepe; Magda Spella; Ioannis Lilis; Dimitra Zazara; Eirini Nikolouli; Nikolitsa Spiropoulou; Andreas Papadakis; Konstantina Papadia; Apostolos Voulgaridis; Vaggelis Harokopos; Panagiota Stamou; Silke Meiners; Oliver Eickelberg; Linda A. Snyder; Sophia G. Antimisiaris; Dimitrios Kardamakis; Ioannis Psallidas; Antonia Marazioti; Georgios T. Stathopoulos

Malignant pleural effusion (MPE) is the lethal consequence of various human cancers metastatic to the pleural cavity. However, the mechanisms responsible for the development of MPE are still obscure. Here we show that mutant KRAS is important for MPE induction in mice. Pleural disseminated, mutant KRAS bearing tumour cells upregulate and systemically release chemokine ligand 2 (CCL2) into the bloodstream to mobilize myeloid cells from the host bone marrow to the pleural space via the spleen. These cells promote MPE formation, as indicated by splenectomy and splenocyte restoration experiments. In addition, KRAS mutations are frequently detected in human MPE and cell lines isolated thereof, but are often lost during automated analyses, as indicated by manual versus automated examination of Sanger sequencing traces. Finally, the novel KRAS inhibitor deltarasin and a monoclonal antibody directed against CCL2 are equally effective against an experimental mouse model of MPE, a result that holds promise for future efficient therapies against the human condition.


Embo Molecular Medicine | 2017

NRAS destines tumor cells to the lungs

Anastasios D. Giannou; Antonia Marazioti; Nikolaos Kanellakis; Ioanna Giopanou; Ioannis Lilis; Dimitra Zazara; Giannoula Ntaliarda; Danai Kati; Vasileios Armenis; Georgia Giotopoulou; Anthi Krontira; Marina Lianou; Theodora Agalioti; Malamati Vreka; Maria Papageorgopoulou; Sotirios Fouzas; Dimitrios Kardamakis; Ioannis Psallidas; Magda Spella; Georgios T. Stathopoulos

The lungs are frequently affected by cancer metastasis. Although NRAS mutations have been associated with metastatic potential, their exact role in lung homing is incompletely understood. We cross‐examined the genotype of various tumor cells with their ability for automatic pulmonary dissemination, modulated NRAS expression using RNA interference and NRAS overexpression, identified NRAS signaling partners by microarray, and validated them using Cxcr1‐ and Cxcr2‐deficient mice. Mouse models of spontaneous lung metastasis revealed that mutant or overexpressed NRAS promotes lung colonization by regulating interleukin‐8‐related chemokine expression, thereby initiating interactions between tumor cells, the pulmonary vasculature, and myeloid cells. Our results support a model where NRAS‐mutant, chemokine‐expressing circulating tumor cells target the CXCR1‐expressing lung vasculature and recruit CXCR2‐expressing myeloid cells to initiate metastasis. We further describe a clinically relevant approach to prevent NRAS‐driven pulmonary metastasis by inhibiting chemokine signaling. In conclusion, NRAS promotes the colonization of the lungs by various tumor types in mouse models. IL‐8‐related chemokines, NRAS signaling partners in this process, may constitute an important therapeutic target against pulmonary involvement by cancers of other organs.


European Respiratory Journal | 2016

“Scar-cinoma”: viewing the fibrotic lung mesenchymal cell in the context of cancer biology

Jeffrey C. Horowitz; John J. Osterholzer; Antonia Marazioti; Georgios T. Stathopoulos

Lung cancer and pulmonary fibrosis are common, yet distinct, pathological processes that represent urgent unmet medical needs. Striking clinical and mechanistic parallels exist between these distinct disease entities. The goal of this article is to examine lung fibrosis from the perspective of cancer-associated phenotypic hallmarks, to discuss areas of mechanistic overlap and distinction, and to highlight profibrotic mechanisms that contribute to carcinogenesis. Ultimately, we speculate that such comparisons might identify opportunities to leverage our current understanding of the pathobiology of each disease process in order to advance novel therapeutic approaches for both. We anticipate that such “outside the box” concepts could be translated to a more precise and individualised approach to fibrotic diseases of the lung. Lung fibrosis and cancer are distinct, but share a number of cell signalling and phenotypic features http://ow.ly/Za9YB


Journal of Medicinal Chemistry | 2013

Insights into soluble guanylyl cyclase activation derived from improved heme-mimetics

Margarete von Wantoch Rekowski; Vijay Kumar; Zongmin Zhou; Johann Moschner; Antonia Marazioti; Marina Bantzi; Georgios A. Spyroulias; Focco van den Akker; Athanassios Giannis; Andreas Papapetropoulos

Recently, the structure of BAY 58-2667 bound to the Nostoc sp. H-NOX domain was published. On the basis of this structural information, we designed BAY 58-2667 derivatives and tested their effects on soluble guanylyl cyclase (sGC) activity. Derivative 20 activated sGC 4.8-fold more than BAY 58-2667. Co-crystallization of 20 with the Ns H-NOX domain revealed that the increased conformational distortion at the C-terminal region of αF helix containing 110-114 residues contributes to the higher activation triggered by 20.


Cancer Research | 2018

ΙκΒ kinase α is required for development and progression of KRAS-mutant lung adenocarcinoma.

Malamati Vreka; Ioannis Lilis; Maria Papageorgopoulou; Georgia Giotopoulou; Marina Lianou; Ioanna Giopanou; Nikolaos Kanellakis; Magda Spella; Theodora Agalioti; Vasileios Armenis; Torsten Goldmann; Sebastian Marwitz; Fiona E. Yull; Timothy S. Blackwell; Manolis Pasparakis; Antonia Marazioti; Georgios T. Stathopoulos

Although oncogenic activation of NFκB has been identified in various tumors, the NFκB-activating kinases (inhibitor of NFκB kinases, IKK) responsible for this are elusive. In this study, we determined the role of IKKα and IKKβ in KRAS-mutant lung adenocarcinomas induced by the carcinogen urethane and by respiratory epithelial expression of oncogenic KRASG12D Using NFκB reporter mice and conditional deletions of IKKα and IKKβ, we identified two distinct early and late activation phases of NFκB during chemical and genetic lung adenocarcinoma development, which were characterized by nuclear translocation of RelB, IκBβ, and IKKα in tumor-initiated cells. IKKα was a cardinal tumor promoter in chemical and genetic KRAS-mutant lung adenocarcinoma, and respiratory epithelial IKKα-deficient mice were markedly protected from the disease. IKKα specifically cooperated with mutant KRAS for tumor induction in a cell-autonomous fashion, providing mutant cells with a survival advantage in vitro and in vivo IKKα was highly expressed in human lung adenocarcinoma, and a heat shock protein 90 inhibitor that blocks IKK function delivered superior effects against KRAS-mutant lung adenocarcinoma compared with a specific IKKβ inhibitor. These results demonstrate an actionable requirement for IKKα in KRAS-mutant lung adenocarcinoma, marking the kinase as a therapeutic target against this disease.Significance: These findings report a novel requirement for IKKα in mutant KRAS lung tumor formation, with potential therapeutic applications. Cancer Res; 78(11); 2939-51. ©2018 AACR.

Collaboration


Dive into the Antonia Marazioti's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andreas Papapetropoulos

National and Kapodistrian University of Athens

View shared research outputs
Researchain Logo
Decentralizing Knowledge