Network


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

Hotspot


Dive into the research topics where Ara Parlakian is active.

Publication


Featured researches published by Ara Parlakian.


Nature Cell Biology | 2010

Identification and characterization of a non-satellite cell muscle resident progenitor during postnatal development

Kathryn J. Mitchell; Alice Pannérec; Bruno Cadot; Ara Parlakian; Vanessa Besson; Edgar R. Gomes; Giovanna Marazzi; David Sassoon

Satellite cells are resident myogenic progenitors in postnatal skeletal muscle involved in muscle postnatal growth and adult regenerative capacity. Here, we identify and describe a population of muscle-resident stem cells, which are located in the interstitium, that express the cell stress mediator PW1 but do not express other markers of muscle stem cells such as Pax7. PW1+/Pax7− interstitial cells (PICs) are myogenic in vitro and efficiently contribute to skeletal muscle regeneration in vivo as well as generating satellite cells and PICs. Whereas Pax7 mutant satellite cells show robust myogenic potential, Pax7 mutant PICs are unable to participate in myogenesis and accumulate during postnatal growth. Furthermore, we found that PICs are not derived from a satellite cell lineage. Taken together, our findings uncover a new and anatomically identifiable population of muscle progenitors and define a key role for Pax7 in a non-satellite cell population during postnatal muscle growth.


Molecular and Cellular Biology | 2004

Targeted inactivation of serum response factor in the developing heart results in myocardial defects and embryonic lethality

Ara Parlakian; David Tuil; Ghislaine Hamard; Geneviève Tavernier; Danièle Hentzen; Jean-Paul Concordet; Denise Paulin; Zhenlin Li; Dominique Daegelen

ABSTRACT Serum response factor (SRF) is at the confluence of multiple signaling pathways controlling the transcription of immediate-early response genes and muscle-specific genes. There are active SRF target sequences in more than 50 genes expressed in the three muscle lineages including normal and diseased hearts. However, the role of SRF in heart formation has not been addressed in vivo thus far due to the early requirement of SRF for mesoderm formation. We have generated a conditional mutant of SRF by using Cre-LoxP strategy that will be extremely useful to study the role of SRF in embryonic and postnatal cardiac functions, as well as in other tissues. This report shows that heart-specific deletion of SRF in the embryo by using a new βMHC-Cre transgenic mouse line results in lethal cardiac defects between embryonic day 10.5 (E10.5) and E13.5, as evidenced by abnormally thin myocardium, dilated cardiac chambers, poor trabeculation, and a disorganized interventricular septum. At E9.5, we found a marked reduction in the expression of essential regulators of heart development, including Nkx2.5, GATA4, myocardin, and the SRF target gene c-fos prior to overt maldevelopment. We conclude that SRF is crucial for cardiac differentiation and maturation, acting as a global regulator of multiple developmental genes.


Circulation | 2005

Temporally Controlled Onset of Dilated Cardiomyopathy Through Disruption of the SRF Gene in Adult Heart

Ara Parlakian; Claude L. Charvet; Brigitte Escoubet; Mathias Mericskay; Jeffery D. Molkentin; Guillaume Gary-Bobo; Leon J. De Windt; Marie-Aline Ludosky; Denise Paulin; Dominique Daegelen; David Tuil; Zhenlin Li

Background— Serum response factor (SRF) is a cardiac transcription factor involved in cell growth and differentiation. We have shown, using the Cre/loxP system, that cardiac-specific disruption of SRF gene in the embryonic heart results in lethal cardiac defects. The role of SRF in adult heart is unknown. Methods and Results— We disrupted SRF in the adult heart using a heart-specific tamoxifen-inducible Cre recombinase. This disruption led to impaired left ventricular function with reduced contractility, subsequently progressing to dilated cardiomyopathy, as demonstrated by serial echocardiography, including tissue Doppler imaging. The cytoarchitecture of cardiomyocytes was altered in the intercalated disks. All mutant mice died from heart failure 10 weeks after treatment. These functional and structural defects were preceded by early alterations in the cardiac gene expression program: major decreases in mRNA levels for cardiac &agr;-actin, muscle creatine kinase, and calcium-handling genes. Conclusions— SRF is crucial for adult cardiac function and integrity. We suggest that the rapid progression to heart failure in SRF mutant mice results primarily from decreased expression of proteins involved in force generation and transmission, low levels of polymerized actin, and changes in cytoarchitecture, without hypertrophic compensation. These cardiac-specific SRF-deficient mice have the morphological and clinical features of acquired dilated cardiomyopathy in humans and may therefore be used as an inducible model of this disorder.


Molecular and Cellular Biology | 2006

New Role for Serum Response Factor in Postnatal Skeletal Muscle Growth and Regeneration via the Interleukin 4 and Insulin-Like Growth Factor 1 Pathways

Claude L. Charvet; Christophe Houbron; Ara Parlakian; Julien Giordani; Charlotte Lahoute; Anne Bertrand; Athanassia Sotiropoulos; Laure Renou; Alain Schmitt; Judith Melki; Zhenlin Li; Dominique Daegelen; David Tuil

ABSTRACT Serum response factor (SRF) is a crucial transcriptional factor for muscle-specific gene expression. We investigated SRF function in adult skeletal muscles, using mice with a postmitotic myofiber-targeted disruption of the SRF gene. Mutant mice displayed severe skeletal muscle mass reductions due to a postnatal muscle growth defect resulting in highly hypotrophic adult myofibers. SRF-depleted myofibers also failed to regenerate following injury. Muscles lacking SRF had very low levels of muscle creatine kinase and skeletal alpha-actin (SKA) transcripts and displayed other alterations to the gene expression program, indicating an overall immaturity of mutant muscles. This loss of SKA expression, together with a decrease in beta-tropomyosin expression, contributed to myofiber growth defects, as suggested by the extensive sarcomere disorganization found in mutant muscles. However, we observed a downregulation of interleukin 4 (IL-4) and insulin-like growth factor 1 (IGF-1) expression in mutant myofibers which could also account for their defective growth and regeneration. Indeed, our demonstration of SRF binding to interleukin 4 and IGF-1 promoters in vivo suggests a new crucial role for SRF in pathways involved in muscle growth and regeneration.


Developmental Cell | 2008

Serum Response Factor Is Required for Sprouting Angiogenesis and Vascular Integrity

Claudio A. Franco; Mathias Mericskay; Ara Parlakian; Guillaume Gary-Bobo; Jacqueline Gao-Li; Denise Paulin; Erika Gustafsson; Zhenlin Li

Serum response factor (SRF) is a transcription factor that controls the expression of cytoskeletal proteins and immediate early genes in different cell types. Here, we found that SRF expression is restricted to endothelial cells (ECs) of small vessels such as capillaries in the mouse embryo. EC-specific Srf deletion led to aneurysms and hemorrhages from 11.5 days of mouse development (E11.5) and lethality at E14.5. Mutant embryos presented a reduced capillary density and defects in EC migration, with fewer numbers of filopodia in tip cells and ECs showing defects in actin polymerization and intercellular junctions. We show that SRF is essential for the expression of VE-cadherin and beta-actin in ECs both in vivo and in vitro. Moreover, knockdown of SRF in ECs impaired VEGF- and FGF-induced in vitro angiogenesis. Taken together, our results demonstrate that SRF plays an important role in sprouting angiogenesis and small vessel integrity in the mouse embryo.


Proceedings of the National Academy of Sciences of the United States of America | 2013

CTIP2 is a negative regulator of P-TEFb.

Thomas Cherrier; Valentin Le Douce; Sebastian Eilebrecht; Raphael Riclet; Céline Marban; Franck Dequiedt; Yannick Goumon; Jean-Christophe Paillart; Mathias Mericskay; Ara Parlakian; Pedro Bausero; Wasim Abbas; Georges Herbein; Siavash K. Kurdistani; Xavier Graña; Benoît Van Driessche; Christian Schwartz; Ermanno Candolfi; Arndt Benecke; Carine Van Lint; Olivier Rohr

The positive transcription elongation factor b (P-TEFb) is involved in physiological and pathological events including inflammation, cancer, AIDS, and cardiac hypertrophy. The balance between its active and inactive form is tightly controlled to ensure cellular integrity. We report that the transcriptional repressor CTIP2 is a major modulator of P-TEFb activity. CTIP2 copurifies and interacts with an inactive P-TEFb complex containing the 7SK snRNA and HEXIM1. CTIP2 associates directly with HEXIM1 and, via the loop 2 of the 7SK snRNA, with P-TEFb. In this nucleoprotein complex, CTIP2 significantly represses the Cdk9 kinase activity of P-TEFb. Accordingly, we show that CTIP2 inhibits large sets of P-TEFb- and 7SK snRNA-sensitive genes. In hearts of hypertrophic cardiomyopathic mice, CTIP2 controls P-TEFb-sensitive pathways involved in the establishment of this pathology. Overexpression of the β-myosin heavy chain protein contributes to the pathological cardiac wall thickening. The inactive P-TEFb complex associates with CTIP2 at the MYH7 gene promoter to repress its activity. Taken together, our results strongly suggest that CTIP2 controls P-TEFb function in physiological and pathological conditions.


Circulation Research | 2001

Chimeric Smooth Muscle–Specific Enhancer/Promoters: Valuable Tools for Adenovirus-Mediated Cardiovascular Gene Therapy

Sébastien Ribault; Pascal Neuville; Agnès Méchine-Neuville; Fabrice Augé; Ara Parlakian; Giulio Gabbiani; Denise Paulin; Valerie Calenda

Abstract— Gene transfer with adenoviral vectors is an attractive approach for the treatment of atherosclerosis and restenosis. However, because expression of a therapeutic gene in nontarget tissues may have deleterious effects, artery-specific expression is desirable. Although expression vectors containing transcriptional regulatory elements of genes expressed solely in smooth muscle cells (SMCs) have proved efficient to restrict expression of the transgene, their use in the clinical setting can be limited by their reduced strength. In the present study, we show that low levels of transgene expression are obtained with the smooth muscle (SM)-specific SM22&agr; promoter compared with the viral cytomegalovirus (CMV) enhancer/promoter. We have generated chimeric transcriptional cassettes containing either a SM (SM-myosin heavy chain) or a skeletal muscle (creatine kinase) enhancer combined with the SM22&agr; promoter. With both constructs we observed significantly stronger expression that remains SM-specific. In vivo, reporter gene expression was restricted to arterial SMCs with no detectable signal at remote sites. Moreover, when interferon-&ggr; expression was driven by one of these two chimeras, SMC growth was inhibited as efficiently as with the CMV promoter. Finally, we demonstrate that neointima formation in the rat carotid balloon injury model was reduced to the same extent by adenoviral gene transfer of interferon-&ggr; driven either by the SM-myosin heavy chain enhancer/SM22&agr; promoter or the CMV promoter. These results indicate that such vectors can be useful for the treatment of hyperproliferative vascular disorders.


Journal of Cell Science | 2014

Synemin acts as a regulator of signalling molecules during skeletal muscle hypertrophy

Zhenlin Li; Ara Parlakian; Dario Coletti; Sonia Alonso-Martin; Christophe Hourdé; Pierre Joanne; Jacqueline Gao-Li; Jocelyne Blanc; Arnaud Ferry; Denise Paulin; Zhigang Xue; Onnik Agbulut

ABSTRACT Synemin, a type IV intermediate filament (IF) protein, forms a bridge between IFs and cellular membranes. As an A-kinase-anchoring protein, it also provides temporal and spatial targeting of protein kinase A (PKA). However, little is known about its functional roles in either process. To better understand its functions in muscle tissue, we generated synemin-deficient (Synm−/−) mice. Synm−/− mice displayed normal development and fertility but showed a mild degeneration and regeneration phenotype in myofibres and defects in sarcolemma membranes. Following mechanical overload, Synm−/− mice muscles showed a higher hypertrophic capacity with increased maximal force and fatigue resistance compared with control mice. At the molecular level, increased remodelling capacity was accompanied by decreased myostatin (also known as GDF8) and atrogin (also known as FBXO32) expression, and increased follistatin expression. Furthermore, the activity of muscle-mass control molecules (the PKA RII&agr; subunit, p70S6K and CREB1) was increased in mutant mice. Finally, analysis of muscle satellite cell behaviour suggested that the absence of synemin could affect the balance between self-renewal and differentiation of these cells. Taken together, our results show that synemin is necessary to maintain membrane integrity and regulates signalling molecules during muscle hypertrophy.


European Journal of Heart Failure | 2008

Mosaic inactivation of the serum response factor gene in the myocardium induces focal lesions and heart failure

Guillaume Gary-Bobo; Ara Parlakian; Brigitte Escoubet; Claudio A. Franco; Sophie Clément; Patrick Bruneval; David Tuil; Dominique Daegelen; Denise Paulin; Zhenlin Li; Mathias Mericskay

Regional alterations in ventricular mechanical functions are a primary determinant for the risk of myocardial injuries in various cardiomyopathies. The serum response factor (SRF) is a transcription factor regulating contractile and cytoskeletal genes and may play an important role in the remodelling of myocardium at the cellular level.


The FASEB Journal | 2014

Endothelial mineralocorticoid receptor activation enhances endothelial protein C receptor and decreases vascular thrombosis in mice

Jérémy Lagrange; Zhenlin Li; Céline Fassot; Mustapha Bourhim; Huguette Louis; Aurélie Nguyen Dinh Cat; Ara Parlakian; Denis Wahl; Patrick Lacolley; Frederic Jaisser; Véronique Regnault

Previous studies have shown that aldosterone, which activates the mineralocorticoid receptor (MR), promotes thrombosis in animal models. Our objective was to determine whether MR activation/expression in the vascular endothelium could modify thrombotic risk in vivo and to examine thrombin generation at the surface of aortic endothelial cells (HAECs). MR was conditionally overexpressed in vivo in vascular endothelial cells in mice (MR‐EC mice) or stimulated with aldosterone in HAECs. Thrombosis after ferric chloride injury was delayed in MR‐EC mice compared with controls as well as in wild‐type FVB/NRj mice treated with aldosterone (60 μg/kg/d for 21 d). Thrombin generation in platelet‐poor plasma did not differ between MR‐EC mice and controls. In MR‐EC mice, aortic endothelial cell protein C receptor (EPCR) expression was increased. Aldosterone (10–8 M) attenuated thrombin generation at the surface of cultured HAECs, and this effect was associated with up‐regula‐tion of expression of EPCR, which promotes formation of activated protein C. Aldosterone increases EPCR expression via a transcriptional mechanism involving interaction of MR with the specificity protein 1 site. These findings demonstrate that MR activation acts on endothelial cells to protect against thrombosis in physiological conditions and that MR‐mediated EPCR overexpression drives this antithrombotic property through enhancing protein C activation.—Lagrange, J., Li, Z., Fassot, C., Bourhim, M., Louis, H., Nguyen Dinh Cat, A., Parlakian, A., Wahl, D., Lacolley, P., Jaisser, F., Regnault, V. Endothelial mineralocorticoid receptor activation enhances endothelial protein C receptor and decreases vascular thrombosis in mice. FASEB J. 28, 2062–2072 (2014). www.fasebj.org

Collaboration


Dive into the Ara Parlakian's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dario Coletti

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Olivier Rohr

University of Strasbourg

View shared research outputs
Top Co-Authors

Avatar

Raphael Riclet

University of Strasbourg

View shared research outputs
Top Co-Authors

Avatar

Jocelyne Blanc

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge