Network


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

Hotspot


Dive into the research topics where Ilias Mylonis is active.

Publication


Featured researches published by Ilias Mylonis.


Journal of Biological Chemistry | 2006

Identification of MAPK Phosphorylation Sites and Their Role in the Localization and Activity of Hypoxia-inducible Factor-1α

Ilias Mylonis; Georgia Chachami; Martina Samiotaki; George Panayotou; Efrosini Paraskeva; Alkmini Kalousi; Eleni Georgatsou; Sofia Bonanou; George Simos

Hypoxia-inducible factor 1 (HIF-1) controls the expression of most genes induced by hypoxic conditions. Regulation of expression and activity of its inducible subunit, HIF-1α, involves several post-translational modifications. To study HIF-1α phosphorylation, we have used human full-length recombinant HIF-1α as a substrate in kinase assays. We show that at least two different nuclear protein kinases, one of them identified as p42/p44 MAPK, can modify HIF-1α. Analysis of in vitro phosphorylated HIF-1α by mass spectroscopy revealed residues Ser-641 and Ser-643 as possible MAPK phosphorylation sites. Site-directed mutagenesis of these residues reduced significantly the phosphorylation of HIF-1α. When these mutant forms of HIF-1α were expressed in HeLa cells, they exhibited much lower transcriptional activity than the wild-type form. However, expression of the same mutants in yeast revealed that their capacity to stimulate transcription was not significantly compromised. Localization of the green fluorescent protein-tagged HIF-1α mutants in HeLa cells showed their exclusion from the nucleus in contrast to wild-type HIF-1α. Treatment of the cells with leptomycin B, an inhibitor of the major exportin CRM1, reversed this exclusion and led to nuclear accumulation and partial recovery of the activity of the HIF-1α mutants. Moreover, inhibition of the MAPK pathway by PD98059 impaired the phosphorylation, nuclear accumulation, and activity of wild-type GFP-HIF-1α. Overall, these data suggest that phosphorylation of Ser-641/643 by MAPK promotes the nuclear accumulation and transcriptional activity of HIF-1α by blocking its CRM1-dependent nuclear export.


Journal of Cell Science | 2012

Hypoxia causes triglyceride accumulation by HIF-1-mediated stimulation of lipin 1 expression

Ilias Mylonis; Hiroshi Sembongi; Christina Befani; Panagiotis Liakos; Symeon Siniossoglou; George Simos

Summary Adaptation to hypoxia involves hypoxia-inducible transcription factors (HIFs) and requires reprogramming of cellular metabolism that is essential during both physiological and pathological processes. In contrast to the established role of HIF-1 in glucose metabolism, the involvement of HIFs and the molecular mechanisms concerning the effects of hypoxia on lipid metabolism are poorly characterized. Here, we report that exposure of human cells to hypoxia causes accumulation of triglycerides and lipid droplets. This is accompanied by induction of lipin 1, a phosphatidate phosphatase isoform that catalyzes the penultimate step in triglyceride biosynthesis, whereas lipin 2 remains unaffected. Hypoxic upregulation of lipin 1 expression involves predominantly HIF-1, which binds to a single distal hypoxia-responsive element in the lipin 1 gene promoter and causes its activation under low oxygen conditions. Accumulation of hypoxic triglycerides or lipid droplets can be blocked by siRNA-mediated silencing of lipin 1 expression or kaempferol-mediated inhibition of HIF-1. We conclude that direct control of lipin 1 transcription by HIF-1 is an important regulatory feature of lipid metabolism and its adaptation to hypoxia.


Journal of Biological Chemistry | 2008

Atypical CRM1-dependent Nuclear Export Signal Mediates Regulation of Hypoxia-inducible Factor-1α by MAPK

Ilias Mylonis; Georgia Chachami; Efrosyni Paraskeva; George Simos

Hypoxia-inducible factor 1 (HIF-1) is the key transcriptional activator of hypoxia-inducible genes and an important anti-cancer target. Its regulated subunit, HIF-1α, is controlled by oxygen levels and major signaling pathways. We reported previously that phosphorylation of Ser641/643 by p42/44 MAPK is essential for HIF-1α nuclear accumulation and activity. We now show that a fragment of HIF-1α (amino acids 616–658), termed MAPK target domain, contains a nuclear export signal (NES), which has atypical hydrophobic residue spacing. Localization, reporter gene, and co-immunoprecipitation assays demonstrate that the identified NES interacts with CRM1 in a phosphorylation-sensitive manner. Furthermore, disruption of the NES (I637A/L638A/I639A) restores nuclear localization and activity of nonphosphorylated HIF-1α and renders it largely resistant to inhibition of MAPK, an effect reproduced by a phosphomimetic mutation (S641E). As these data predict, overexpression of wild-type or mutant (S641A/S643A) MAPK target domain in HeLa cells modulates the activity and subcellular distribution of endogenous HIF-1α. We suggest that control of HIF-1α nuclear transport represents an important MAPK-dependent regulatory mechanism.


Biochemical and Biophysical Research Communications | 2010

The dietary flavonoid kaempferol effectively inhibits HIF-1 activity and hepatoma cancer cell viability under hypoxic conditions.

Ilias Mylonis; Achillia Lakka; Andreas Tsakalof; George Simos

Hepatocellular carcinoma (HCC) is characterized by high mortality rates and resistance to conventional treatment. HCC tumors usually develop local hypoxia, which stimulates proliferation of cancer cells and renders them resilient to chemotherapy. Adaptation of tumor cells to the hypoxic conditions depends on the hypoxia-inducible factor 1 (HIF-1). Over-expression of its regulated HIF-1alpha subunit, an important target of anti-cancer therapy, is observed in many cancers including HCC and is associated with severity of tumor growth and poor patient prognosis. In this report we investigate the effect of the dietary flavonoid kaempferol on activity, expression levels and localization of HIF-1alpha as well as viability of human hepatoma (Huh7) cancer cells. Treatment of Huh7 cells with kaempferol under hypoxic conditions (1% oxygen) effectively inhibited HIF-1 activity in a dose-dependent manner (IC(50)=5.16microM). The mechanism of this inhibition did not involve suppression of HIF-1alpha protein levels but rather its mislocalization into the cytoplasm due to inactivation of p44/42 MAPK by kaempferol (IC(50)=4.75microM). Exposure of Huh7 cells to 10microM kaempferol caused significant reduction of their viability, which was remarkably more evident under hypoxic conditions. In conclusion, kaempferol, a non-toxic natural food component, inhibits both MAPK and HIF-1 activity at physiologically relevant concentrations (5-10microM) and suppresses hepatocarcinoma cell survival more efficiently under hypoxia. It has, therefore, potential as a therapeutic or chemopreventive anti-HCC agent.


Journal of Cell Science | 2010

Casein kinase 1 regulates human hypoxia-inducible factor HIF-1

Alkmini Kalousi; Ilias Mylonis; Anastasia S. Politou; Georgia Chachami; Efrosyni Paraskeva; George Simos

Hypoxia-inducible factor 1 (HIF-1), a transcriptional activator that mediates cellular response to hypoxia and a promising target of anticancer therapy, is essential for adaptation to low oxygen conditions, embryogenesis and tumor progression. HIF-1 is a heterodimer of HIF-1α, expression of which is controlled by oxygen levels as well as by various oxygen-independent mechanisms, and HIF-1β (or ARNT), which is constitutively expressed. In this work, we investigate the phosphorylation of the N-terminal heterodimerization (PAS) domain of HIF-1α and identify Ser247 as a major site of in vitro modification by casein kinase 1δ (CK1δ). Mutation of this site to alanine, surprisingly, enhanced the transcriptional activity of HIF-1α, a result phenocopied by inhibition or small interfering RNA (siRNA)-mediated silencing of CK1δ under hypoxic conditions. Conversely, overexpression of CK1δ or phosphomimetic mutation of Ser247 to aspartate inhibited HIF-1α activity without affecting its stability or nuclear accumulation. Immunoprecipitation and in vitro binding experiments suggest that CK1-dependent phosphorylation of HIF-1α at Ser247 impairs its association with ARNT, a notion also supported by modeling the structure of the complex between HIF-1α and ARNT PAS-B domains. We suggest that modification of HIF-1α by CK1 represents a novel mechanism that controls the activity of HIF-1 during hypoxia by regulating the interaction between its two subunits.


Cellular Physiology and Biochemistry | 2007

MgcRacGAP interacts with HIF-1alpha and regulates its transcriptional activity.

Aggeliki Lyberopoulou; Emmanouil Venieris; Ilias Mylonis; Georgia Chachami; Ioannis S. Pappas; George Simos; Sofia Bonanou; Eleni Georgatsou

HIF-1α is the inducible subunit of the dimeric transcription factor HIF-1 (Hypoxia Inducible Factor 1). It is induced by hypoxia and hypoxia-mimetics in most cell types, as well as non-hypoxic signals such as growth factors, cytokines and oncogenes, often in a cell specific manner. HIF-1 is present in virtually all cells of higher eukaryotes and its function is of great biomedical relevance since it is highly involved in development, tumor progression and tissue ischemia. Intracellular signaling to HIF-1α, as well as its further action, involves its participation in numerous protein complexes. Using the yeast two-hybrid system we have identified MgcRacGAP (male germ cell Rac GTPase Activating Protein) as a HIF-1α interacting protein. The MgcRacGAP protein is a regulator of Rho proteins, which are principally involved in cytoskeletal organization. We have verified specific binding of HIF-1α and MgcRacGAP in vitro and in vivo in mammalian cells. We have additionally shown that MgcRacGAP overexpression inhibits HIF-1α transcriptional activity, without lowering HIF-1α protein levels, or altering its subcellular localization. Moreover, this inhibition is dependent on the MgcRacGAP domain that interacts with HIF-1α. In conclusion, our findings demonstrate that HIF-1α function is negatively affected by its interaction with MgcRacGAP.


Biochemical and Biophysical Research Communications | 2003

Protein kinase CK2 phosphorylates and activates the SR protein-specific kinase 1.

Ilias Mylonis; Thomas Giannakouros

The serine/arginine subfamily of protein kinases has been conserved throughout evolution and its members are thought to play important roles in the regulation of multiple cellular processes. Mammalian SRPK1 has been considered as a constitutively active kinase that is predominantly expressed in testis. In the present study, recombinant GST-SRPK1 was used as substrate to identify potential protein kinase(s) in testis extracts, involved in phosphorylating and thereby regulating the activity of this enzyme. Using a panel of chromatography media, inhibition by heparin, immunoblot analysis, and phosphopeptide mapping, CK2 was determined to be the major kinase that phosphorylates SRPK1. Phosphorylation of SRPK1 by CK2 occurred mainly at Ser(51) and Ser(555) in vitro, and resulted in approximately 6-fold activation of the enzyme. These findings suggest that SRPK1 may be an important cellular target for CK2 action.


Cellular Signalling | 2015

CK1δ restrains lipin-1 induction, lipid droplet formation and cell proliferation under hypoxia by reducing HIF-1α/ARNT complex formation.

Maria Kourti; Georgia Ikonomou; Nikolaos-Nikiforos Giakoumakis; Maria Anna Rapsomaniki; Ulf Landegren; Symeon Siniossoglou; Zoi Lygerou; George Simos; Ilias Mylonis

Proliferation of cells under hypoxia is facilitated by metabolic adaptation, mediated by the transcriptional activator Hypoxia Inducible Factor-1 (HIF-1). HIF-1α, the inducible subunit of HIF-1 is regulated by oxygen as well as by oxygen-independent mechanisms involving phosphorylation. We have previously shown that CK1δ phosphorylates HIF-1α in its N-terminus and reduces its affinity for its heterodimerization partner ARNT. To investigate the importance of this mechanism for cell proliferation under hypoxia, we visually monitored HIF-1α interactions within the cell nucleus using the in situ proximity ligation assay (PLA) and fluorescence recovery after photobleaching (FRAP). Both methods show that CK1δ-dependent modification of HIF-1α impairs the formation of a chromatin binding HIF-1 complex. This is confirmed by analyzing expression of lipin-1, a direct target of HIF-1 that mediates hypoxic neutral lipid accumulation. Inhibition of CK1δ increases lipid droplet formation and proliferation of both cancer and normal cells specifically under hypoxia and in an HIF-1α- and lipin-1-dependent manner. These data reveal a novel role for CK1δ in regulating lipid metabolism and, through it, cell adaptation to low oxygen conditions.


Journal of Cutaneous Pathology | 2009

Increased HIF-1 alpha immunostaining in psoriasis compared to psoriasiform dermatitides

Maria Ioannou; Fani Sourli; Ilias Mylonis; Sotirios Barbanis; Rodula Papamichali; Evangelos Kouvaras; Efterpi Zafiriou; Eleni Klimi; George Simos; Aggeliki-Victoria Roussaki-Schulze; George K. Koukoulis

Background: Expression of hypoxia inducible factor 1 (HIF‐1) alpha can be linked to inflammation through reciprocal interactions with several cytokines. This finding is in accordance with the previously noted HIF‐1 alpha overexpression in psoriatic lesional keratinocytes.


Journal of Cellular Physiology | 2013

TNFα induces expression of HIF-1α mRNA and protein but inhibits hypoxic stimulation of HIF-1 transcriptional activity in airway smooth muscle cells.

Stamatina Tsapournioti; Ilias Mylonis; Apostolia Hatziefthimiou; Maria Ioannou; Rodopi Stamatiou; Georgios Koukoulis; George Simos; Paschalis-Adam Molyvdas; Efrosyni Paraskeva

Airway smooth muscle cells (ASMCs) participate in tissue remodeling characteristic of airway inflammatory diseases like asthma. Inflammation and hypoxia pathways are often interconnected and the regulatory subunit of the hypoxia inducible factor, HIF‐1α, has been recently shown to be induced by cytokines. Here we investigate the effect of individual or combined treatment of ASMCs with the inflammatory mediator TNFα and/or hypoxia on the expression of HIF‐1α, HIF‐1 targets and inflammation markers. TNFα enhances HIF‐1α protein and mRNA levels, under both normoxia and hypoxia. TNFα‐mediated induction of HIF‐1α gene transcription is repressed by inhibition of the NF‐κB pathway. Despite the up‐regulation of HIF‐1α protein, the transcription of HIF‐1 target genes remains low in the presence of TNFα at normoxia and is even reduced at hypoxia. We show that the reduction in HIF‐1 transcriptional activity by TNFα is due to inhibition of the interaction of HIF‐1α with ARNT and subsequent blocking of its binding to HREs. Comparison between hypoxia and TNFα for their effects on the expression of inflammatory markers shows significant differences: hypoxia up‐regulates the expression of IL‐6, but not RANTES or ICAM, and reduces the induction of VCAM by TNFα. Finally, ex vivo treatment of rabbit trachea strips with TNFα increases HIF‐1α protein levels, but reduces the expression of HIF‐1 targets under hypoxia. Overall, TNFα induces HIF‐1α mRNA synthesis via an NF‐κB dependent pathway but inhibits binding of HIF‐1α to ARNT and DNA, while hypoxia and TNFα have distinct effects on ASMC inflammatory gene expression. J. Cell. Physiol. 228: 1745–1753, 2013.

Collaboration


Dive into the Ilias Mylonis'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
Researchain Logo
Decentralizing Knowledge