Network


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

Hotspot


Dive into the research topics where Saeideh Nakhaei-Rad is active.

Publication


Featured researches published by Saeideh Nakhaei-Rad.


Nature Genetics | 2015

Juvenile myelomonocytic leukemia displays mutations in components of the RAS pathway and the PRC2 network

Aurélie Caye; Marion Strullu; Fabien Guidez; Bruno Cassinat; Steven Gazal; Odile Fenneteau; Elodie Lainey; Kazem Nouri; Saeideh Nakhaei-Rad; Radovan Dvorsky; Julie Lachenaud; Sabrina Pereira; Jocelyne Vivent; Emmanuelle Verger; Dominique Vidaud; Claire Galambrun; Capucine Picard; Arnaud Petit; Audrey Contet; Marilyne Poirée; Nicolas Sirvent; Francoise Mechinaud; Dalila Adjaoud; Catherine Paillard; Brigitte Nelken; Yves Reguerre; Yves Bertrand; Dieter Häussinger; Jean-Hugues Dalle; Mohammad Reza Ahmadian

Juvenile myelomonocytic leukemia (JMML) is a rare and severe myelodysplastic and myeloproliferative neoplasm of early childhood initiated by germline or somatic RAS-activating mutations. Genetic profiling and whole-exome sequencing of a large JMML cohort (118 and 30 cases, respectively) uncovered additional genetic abnormalities in 56 cases (47%). Somatic events were rare (0.38 events/Mb/case) and restricted to sporadic (49/78; 63%) or neurofibromatosis type 1 (NF1)-associated (8/8; 100%) JMML cases. Multiple concomitant genetic hits targeting the RAS pathway were identified in 13 of 78 cases (17%), disproving the concept of mutually exclusive RAS pathway mutations and defining new pathways activated in JMML involving phosphoinositide 3-kinase (PI3K) and the mTORC2 complex through RAC2 mutation. Furthermore, this study highlights PRC2 loss (26/78; 33% of sporadic JMML cases) that switches the methylation/acetylation status of lysine 27 of histone H3 in JMML cases with altered RAS and PRC2 pathways. Finally, the association between JMML outcome and mutational profile suggests a dose-dependent effect for RAS pathway activation, distinguishing very aggressive JMML rapidly progressing to acute myeloid leukemia.


Journal of Biological Chemistry | 2014

Functional Cross-talk between Ras and Rho Pathways A Ras-SPECIFIC GTPase-ACTIVATING PROTEIN (p120RasGAP) COMPETITIVELY INHIBITS THE RhoGAP ACTIVITY OF DELETED IN LIVER CANCER (DLC) TUMOR SUPPRESSOR BY MASKING THE CATALYTIC ARGININE FINGER

Mamta Jaiswal; Radovan Dvorsky; Ehsan Amin; Sarah L. Risse; Eyad K. Fansa; Si-Cai Zhang; Mohamed S. Taha; Aziz R. Gauhar; Saeideh Nakhaei-Rad; Claus Kordes; Katja T. Koessmeier; Ion C. Cirstea; Monilola A. Olayioye; Dieter Häussinger; Mohammad Reza Ahmadian

Background: The regulatory mechanism of the DLC1 tumor suppressor protein is unclear. Results: Structure-function analysis revealed determinants for the selectivity, activity, and inhibition of DLC1 RhoGAP function. Conclusion: p120RasGAP competitively and selectively inhibits DLC1 by targeting its catalytic arginine finger. Significance: This mechanistic study emphasizes the importance of the functional inter-relationships of GTPase-activating proteins mediating cross-talk between the Ras and Rho pathways. The three deleted in liver cancer genes (DLC1–3) encode Rho-specific GTPase-activating proteins (RhoGAPs). Their expression is frequently silenced in a variety of cancers. The RhoGAP activity, which is required for full DLC-dependent tumor suppressor activity, can be inhibited by the Src homology 3 (SH3) domain of a Ras-specific GAP (p120RasGAP). Here, we comprehensively investigated the molecular mechanism underlying cross-talk between two distinct regulators of small GTP-binding proteins using structural and biochemical methods. We demonstrate that only the SH3 domain of p120 selectively inhibits the RhoGAP activity of all three DLC isoforms as compared with a large set of other representative SH3 or RhoGAP proteins. Structural and mutational analyses provide new insights into a putative interaction mode of the p120 SH3 domain with the DLC1 RhoGAP domain that is atypical and does not follow the classical PXXP-directed interaction. Hence, p120 associates with the DLC1 RhoGAP domain by targeting the catalytic arginine finger and thus by competitively and very potently inhibiting RhoGAP activity. The novel findings of this study shed light on the molecular mechanisms underlying the DLC inhibitory effects of p120 and suggest a functional cross-talk between Ras and Rho proteins at the level of regulatory proteins.


Journal of Biological Chemistry | 2016

The Role of Embryonic Stem Cell-expressed RAS (ERAS) in the Maintenance of Quiescent Hepatic Stellate Cells

Saeideh Nakhaei-Rad; Hossein Nakhaeizadeh; Silke Götze; Claus Kordes; Iris Sawitza; Michèle J. Hoffmann; Manuel Franke; Wolfgang A. Schulz; Jürgen Scheller; Roland P. Piekorz; Dieter Häussinger; Mohammad Reza Ahmadian

Hepatic stellate cells (HSCs) were recently identified as liver-resident mesenchymal stem cells. HSCs are activated after liver injury and involved in pivotal processes, such as liver development, immunoregulation, regeneration, and also fibrogenesis. To date, several studies have reported candidate pathways that regulate the plasticity of HSCs during physiological and pathophysiological processes. Here we analyzed the expression changes and activity of the RAS family GTPases and thereby investigated the signaling networks of quiescent HSCs versus activated HSCs. For the first time, we report that embryonic stem cell-expressed RAS (ERAS) is specifically expressed in quiescent HSCs and down-regulated during HSC activation via promoter DNA methylation. Notably, in quiescent HSCs, the high level of ERAS protein correlates with the activation of AKT, STAT3, mTORC2, and HIPPO signaling pathways and inactivation of FOXO1 and YAP. Our data strongly indicate that in quiescent HSCs, ERAS targets AKT via two distinct pathways driven by PI3Kα/δ and mTORC2, whereas in activated HSCs, RAS signaling shifts to RAF-MEK-ERK. Thus, in contrast to the reported role of ERAS in tumor cells associated with cell proliferation, our findings indicate that ERAS is important to maintain quiescence in HSCs.


Journal of Biological Chemistry | 2015

The Function of Embryonic Stem Cell-expressed RAS (E-RAS), a Unique RAS Family Member, Correlates with Its Additional Motifs and Its Structural Properties

Saeideh Nakhaei-Rad; Hossein Nakhaeizadeh; Claus Kordes; Ion C. Cirstea; Malte Schmick; Radovan Dvorsky; Philippe I. H. Bastiaens; Dieter Häussinger; Mohammad Reza Ahmadian

Background: E-RAS contains additional motifs and regions with unknown functions. Results: Biochemical analysis reveals that effector selection of E-RAS significantly differs from H-RAS. Conclusion: E-RAS selectivity and consequently cellular outcomes depend on its unique switch and interswitch regions. Significance: E-RAS possesses specific sequence fingerprints and therefore no overlapping function with H-RAS. E-RAS is a member of the RAS family specifically expressed in embryonic stem cells, gastric tumors, and hepatic stellate cells. Unlike classical RAS isoforms (H-, N-, and K-RAS4B), E-RAS has, in addition to striking and remarkable sequence deviations, an extended 38-amino acid-long unique N-terminal region with still unknown functions. We investigated the molecular mechanism of E-RAS regulation and function with respect to its sequence and structural features. We found that N-terminal extension of E-RAS is important for E-RAS signaling activity. E-RAS protein most remarkably revealed a different mode of effector interaction as compared with H-RAS, which correlates with deviations in the effector-binding site of E-RAS. Of all these residues, tryptophan 79 (arginine 41 in H-RAS), in the interswitch region, modulates the effector selectivity of RAS proteins from H-RAS to E-RAS features.


PLOS ONE | 2016

The RAS-Effector Interface: Isoform-Specific Differences in the Effector Binding Regions

Hossein Nakhaeizadeh; Ehsan Amin; Saeideh Nakhaei-Rad; Radovan Dvorsky; Mohammad Reza Ahmadian; László Buday

RAS effectors specifically interact with the GTP-bound form of RAS in response to extracellular signals and link them to downstream signaling pathways. The molecular nature of effector interaction by RAS is well-studied but yet still incompletely understood in a comprehensive and systematic way. Here, structure-function relationships in the interaction between different RAS proteins and various effectors were investigated in detail by combining our in vitro data with in silico data. Equilibrium dissociation constants were determined for the binding of HRAS, KRAS, NRAS, RRAS1 and RRAS2 to both the RAS binding (RB) domain of CRAF and PI3Kα, and the RAS association (RA) domain of RASSF5, RALGDS and PLCε, respectively, using fluorescence polarization. An interaction matrix, constructed on the basis of available crystal structures, allowed identification of hotspots as critical determinants for RAS-effector interaction. New insights provided by this study are the dissection of the identified hotspots in five distinct regions (R1 to R5) in spite of high sequence variability not only between, but also within, RB/RA domain-containing effectors proteins. Finally, we propose that intermolecular β-sheet interaction in R1 is a central recognition region while R3 may determine specific contacts of RAS versus RRAS isoforms with effectors.


Biochemistry and Cell Biology | 2012

New windows to enhance direct reprogramming of somatic cells towards induced pluripotent stem cells

Saeideh Nakhaei-Rad; Ahmad Reza Bahrami; Mahdi Mirahmadi; Maryam Moghaddam Matin

Induced pluripotent stem cells are generated by direct reprogramming of somatic cells with the introduction of defined transcription factors or other means. Clinical applications of induced pluripotent stem cells are the latest of stem cell therapy approaches due to overcoming problems associated with insufficient cells from conventional sources and immune rejections. In practice, this is restricted by 4 major barriers including the use of genetic manipulations for delivering the reprogramming factors, low efficiency of this process, slow kinetics of the direct reprogramming, and potential for tumor development. Here, we review the latest achievements in improving reprogramming efficiency by alternative strategies. These alternatives mainly involve the replacement of genetic reprogramming factors with small molecules or other factors.


Scientific Reports | 2018

IL-2 Inducible Kinase ITK is Critical for HIV-1 Infection of Jurkat T-cells

Anika Hain; Melanie Krämer; René Martin Linka; Saeideh Nakhaei-Rad; Mohammad Reza Ahmadian; Dieter Häussinger; Arndt Borkhardt; Carsten Münk

Successful replication of Human immunodeficiency virus (HIV)-1 depends on the expression of various cellular host factors, such as the interleukin-2 inducible T-cell kinase (ITK), a member of the protein family of TEC-tyrosine kinases. ITK is selectively expressed in T-cells and coordinates signaling pathways downstream of the T-cell receptor and chemokine receptors, including PLC-1 activation, Ca2+-release, transcription factor mobilization, and actin rearrangements. The exact role of ITK during HIV-1 infection is still unknown. We analyzed the function of ITK during HIV-1 replication and showed that attachment, fusion of virions with the cell membrane and entry into Jurkat T-cells was inhibited when ITK was knocked down. In contrast, reverse transcription and provirus expression were not affected by ITK deficiency. Inhibited ITK expression did not affect the CXCR4 receptor on the cell surface, whereas CD4 and LFA-1 integrin levels were slightly enhanced in ITK knockdown cells and heparan sulfate (HS) expression was completely abolished in ITK depleted T-cells. However, neither HS expression nor other attachment factors could explain the impaired HIV-1 binding to ITK-deficient cells, which suggests that a more complex cellular process is influenced by ITK or that not yet discovered molecules contribute to restriction of HIV-1 binding and entry.


PLOS Genetics | 2017

Correction: Aberrant neuronal activity-induced signaling and gene expression in a mouse model of RASopathy

Franziska Altmüller; Santosh Pothula; Anil Annamneedi; Saeideh Nakhaei-Rad; Carolina Montenegro-Venegas; Eneko Pina-Fernández; Claudia Marini; Mónica Santos; Denny Schanze; Dirk Montag; Mohammad Reza Ahmadian; Oliver Stork; Martin Zenker; Anna Fejtova

[This corrects the article DOI: 10.1371/journal.pgen.1006684.].


Cytokine | 2018

Suppression of dsRNA response genes and innate immunity following Oct4, Stella, and Nanos2 overexpression in mouse embryonic fibroblasts.

Moein Farshchian; Maryam Moghaddam Matin; Olivier Armant; Dirk Geerts; Mahtab Dastpak; Saeideh Nakhaei-Rad; Massoumeh Tajeran; Amir Jebelli; Mina Shahriyari; Monireh Bahrami; Ali Fallah; Vesal Yaghoobi; Mahdi Mirahmadi; Mohammad Reza Abbaszadegan; Ahmad Reza Bahrami

&NA; The self‐renewal capacity of germline derived stem cells (GSCs) makes them an ideal source for research and use in clinics. Despite the presence of active gene network similarities between embryonic stem cells (ESCs) and GSCs, there are unanswered questions regarding the roles of evolutionary conserved genes in GSCs. To determine the reprogramming potential of germ cell‐ specific genes, we designed a polycistronic gene cassette expressing Stella, Oct4 and Nanos2 in a lentiviral‐based vector. Deep transcriptome analysis showed the activation of a set of pluripotency and germ‐cell‐specific markers and the downregulation of innate immune system. The global shut down of antiviral genes included MHC class I, interferon response genes and dsRNA 2′‐5′‐oligoadenylate synthetase are critical pathways that has been affected. Individual expression of each factor highlighted suppressive effect of Nanos2 on genes such as Isg15 and Oasl2. Collectively, to our knowledge this is the first report showing that Nanos2 could be considered as an immunosuppressive factor. Furthermore, our results demonstrate suppression of endogenous retrotransposons that harbor immune response but further analysis require to uncover the correlation between transposon suppression and immune response in germ cell development.


Critical Reviews in Biochemistry and Molecular Biology | 2018

Structural fingerprints, interactions, and signaling networks of RAS family proteins beyond RAS isoforms

Saeideh Nakhaei-Rad; Fereshteh Haghighi; Parivash Nouri; Soheila Rezaei Adariani; Jana Lissy; Neda S. Kazemein Jasemi; Radovan Dvorsky; Mohammad Reza Ahmadian

Abstract Among the signaling molecules indirectly linked to many different cell surface receptors, RAS proteins essentially respond to a diverse range of extracellular cues. They control activities of multiple signaling pathways and consequently a wide array of cellular processes, including survival, growth, adhesion, migration, and differentiation. Any dysregulation of these pathway leads, thus, to cancer, developmental disorders, metabolic, and cardiovascular diseases. The biochemistry of RAS family proteins has become multifaceted since the discovery of the first members, more than 40 years ago. Substantial knowledge has been attained about molecular mechanisms underlying post-translational modification, membrane localization, regulation, and signal transduction through diverse effector molecules. However, the increasing complexity of the underlying signaling mechanisms is considerable, in part due to multiple effector pathways, crosstalks between them and eventually feedback mechanisms. Here, we take a broad view of regulatory and signaling networks of all RAS family proteins that extends beyond RAS paralogs. As described in this review, a lot is known but a lot has to be discovered yet. Graphical abstract: The RAS paralogs, KRAS4B, NRAS, and HRAS, are the best investigated members of the RAS family, not only because of their oncogenic capacity. This protein family, however, contains 22 additional isoforms and paralogs, most of which are distantly related, with typically 20–30% amino acid identity, although they share a conserved GTP-binding domain [the color spectrum goes from white (for identical) through yellow and orange (for partially conserved) to red (for highly variable amino acids). RAS family proteins control a wide array of signaling pathways and cellular processes distinct from those controlled by RAS paralogs. This review focuses on common features and differences of RAS family proteins regarding their structure, function, regulation, signaling, and involvement in diseases.

Collaboration


Dive into the Saeideh Nakhaei-Rad's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Radovan Dvorsky

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Claus Kordes

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar

Ehsan Amin

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ion C. Cirstea

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar

Kazem Nouri

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar

Mohamed S. Taha

University of Düsseldorf

View shared research outputs
Researchain Logo
Decentralizing Knowledge