Network


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

Hotspot


Dive into the research topics where Francesca Stillitano is active.

Publication


Featured researches published by Francesca Stillitano.


Stem Cells Translational Medicine | 2014

Small Molecule-Mediated Directed Differentiation of Human Embryonic Stem Cells Toward Ventricular Cardiomyocytes

Ioannis Karakikes; Grant Senyei; Jens Hansen; Chi Wing Kong; Evren U. Azeloglu; Francesca Stillitano; Deborah K. Lieu; Jiaxian Wang; Lihuan Ren; Jean Sebastien Hulot; Ravi Iyengar; Ronald A. Li; Roger J. Hajjar

The generation of human ventricular cardiomyocytes from human embryonic stem cells and/or induced pluripotent stem cells could fulfill the demand for therapeutic applications and in vitro pharmacological research; however, the production of a homogeneous population of ventricular cardiomyocytes remains a major limitation. By combining small molecules and growth factors, we developed a fully chemically defined, directed differentiation system to generate ventricular‐like cardiomyocytes (VCMs) from human embryonic stem cells and induced pluripotent stem cells with high efficiency and reproducibility. Molecular characterization revealed that the differentiation recapitulated the developmental steps of cardiovascular fate specification. Electrophysiological analyses further illustrated the generation of a highly enriched population of VCMs. These chemically induced VCMs exhibited the expected cardiac electrophysiological and calcium handling properties as well as the appropriate chronotropic responses to cardioactive compounds. In addition, using an integrated computational and experimental systems biology approach, we demonstrated that the modulation of the canonical Wnt pathway by the small molecule IWR‐1 plays a key role in cardiomyocyte subtype specification. In summary, we developed a reproducible and efficient experimental platform that facilitates a chemical genetics‐based interrogation of signaling pathways during cardiogenesis that bypasses the limitations of genetic approaches and provides a valuable source of ventricular cardiomyocytes for pharmacological screenings as well as cell replacement therapies.


Stem Cell Research & Therapy | 2014

Considerations for pre-clinical models and clinical trials of pluripotent stem cell-derived cardiomyocytes

Jean-Sébastien Hulot; Francesca Stillitano; Joe Elie Salem; Jason C. Kovacic; Valentin Fuster; Roger J. Hajjar

Pluripotent stem cells (PSCs) represent an appealing source from which to develop cell replacement therapies. Different initiatives have been launched to promote their development toward clinical applications. This article will review the main questions that should be considered before translating PSC-derived cardiomyocytes into clinical investigations, including the development of good manufacturing practice-level PSC lines, the development of efficient protocols to generate pure populations of cardiac myocytes, and the development of techniques to improve the retention and survival rate of transplanted cells.


Nature Communications | 2015

Correction of human phospholamban R14del mutation associated with cardiomyopathy using targeted nucleases and combination therapy

Ioannis Karakikes; Francesca Stillitano; Mathieu Nonnenmacher; Christos Tzimas; Despina Sanoudou; Vittavat Termglinchan; Chi Wing Kong; Stephanie N. Rushing; Jens Hansen; Delaine K. Ceholski; Fotis Kolokathis; Dimitrios Th. Kremastinos; Alexandros Katoulis; Lihuan Ren; Ninette Cohen; Johannes M.I.H. Gho; Dimitrios Tsiapras; Aryan Vink; Joseph C. Wu; Folkert W. Asselbergs; Ronald A. Li; Jean Sebastien Hulot; Evangelia G. Kranias; Roger J. Hajjar

A number of genetic mutations is associated with cardiomyopathies. A mutation in the coding region of the phospholamban (PLN) gene (R14del) is identified in families with hereditary heart failure. Heterozygous patients exhibit left ventricular dilation and ventricular arrhythmias. Here we generate induced pluripotent stem cells (iPSCs) from a patient harbouring the PLN R14del mutation and differentiate them into cardiomyocytes (iPSC-CMs). We find that the PLN R14del mutation induces Ca2+ handling abnormalities, electrical instability, abnormal cytoplasmic distribution of PLN protein and increases expression of molecular markers of cardiac hypertrophy in iPSC-CMs. Gene correction using transcription activator-like effector nucleases (TALENs) ameliorates the R14del-associated disease phenotypes in iPSC-CMs. In addition, we show that knocking down the endogenous PLN and simultaneously expressing a codon-optimized PLN gene reverses the disease phenotype in vitro. Our findings offer novel strategies for targeting the pathogenic mutations associated with cardiomyopathies.


eLife | 2017

Modeling susceptibility to drug-induced long QT with a panel of subject-specific induced pluripotent stem cells

Francesca Stillitano; Jens Hansen; Chi-Wing Kong; Ioannis Karakikes; Christian Funck-Brentano; Lin Geng; Stuart A. Scott; Stephan Reynier; Ma Wu; Yannick Valogne; Carole Desseaux; Joe-Elie Salem; Dorota Jeziorowska; Noël Zahr; Ronald A. Li; Ravi Iyengar; Roger J. Hajjar; Jean-Sébastien Hulot

A large number of drugs can induce prolongation of cardiac repolarization and life-threatening cardiac arrhythmias. The prediction of this side effect is however challenging as it usually develops in some genetically predisposed individuals with normal cardiac repolarization at baseline. Here, we describe a platform based on a genetically diverse panel of induced pluripotent stem cells (iPSCs) that reproduces susceptibility to develop a cardiotoxic drug response. We generated iPSC-derived cardiomyocytes from patients presenting in vivo with extremely low or high changes in cardiac repolarization in response to a pharmacological challenge with sotalol. In vitro, the responses to sotalol were highly variable but strongly correlated to the inter-individual differences observed in vivo. Transcriptomic profiling identified dysregulation of genes (DLG2, KCNE4, PTRF, HTR2C, CAMKV) involved in downstream regulation of cardiac repolarization machinery as underlying high sensitivity to sotalol. Our findings offer novel insights for the development of iPSC-based screening assays for testing individual drug reactions. DOI: http://dx.doi.org/10.7554/eLife.19406.001


Diabetes | 2017

Functional Human Beige Adipocytes from Induced Pluripotent Stem Cells

Anne-Claire Guénantin; Nolwenn Briand; Emilie Capel; Florent Dumont; Romain Morichon; Claire Provost; Francesca Stillitano; Dorota Jeziorowska; Jean-Pierre Siffroi; Roger J. Hajjar; Bruno Fève; Jean-Sébastien Hulot; Philippe Collas; Jacqueline Capeau; Corinne Vigouroux

Activation of thermogenic beige adipocytes has recently emerged as a promising therapeutic target in obesity and diabetes. Relevant human models for beige adipocyte differentiation are essential to implement such therapeutic strategies. We report a straightforward and efficient protocol to generate functional human beige adipocytes from human induced pluripotent stem cells (hiPSCs). Without overexpression of exogenous adipogenic genes, our method recapitulates an adipogenic developmental pathway through successive mesodermal and adipogenic progenitor stages. hiPSC-derived adipocytes are insulin sensitive and display beige-specific markers and functional properties, including upregulation of thermogenic genes, increased mitochondrial content, and increased oxygen consumption upon activation with cAMP analogs. Engraftment of hiPSC-derived adipocytes in mice produces well-organized and vascularized adipose tissue, capable of β-adrenergic–responsive glucose uptake. Our model of human beige adipocyte development provides a new and scalable tool for disease modeling and therapeutic screening.


Circulation Research | 2018

Exosomal microRNA-21-5p Mediates Mesenchymal Stem Cell Paracrine Effects on Human Cardiac Tissue Contractility

Joshua Mayourian; Delaine K. Ceholski; Przemyslaw Gorski; Prabhu Mathiyalagan; Jack F. Murphy; Sophia I Salazar; Francesca Stillitano; Joshua M. Hare; Susmita Sahoo; Roger J. Hajjar; Kevin D. Costa

Rationale: The promising clinical benefits of delivering human mesenchymal stem cells (hMSCs) for treating heart disease warrant a better understanding of underlying mechanisms of action. hMSC exosomes increase myocardial contractility; however, the exosomal cargo responsible for these effects remains unresolved. Objective: This study aims to identify lead cardioactive hMSC exosomal microRNAs to provide a mechanistic basis for optimizing future stem cell-based cardiotherapies. Methods and Results: Integrating systems biology and human engineered cardiac tissue (hECT) technologies, partial least squares regression analysis of exosomal microRNA profiling data predicted microRNA-21-5p (miR-21-5p) levels positively correlate with contractile force and calcium handling gene expression responses in hECTs treated with conditioned media from multiple cell types. Furthermore, miR-21-5p levels were significantly elevated in hECTs treated with the exosome-enriched fraction of the hMSC secretome (hMSC-exo) versus untreated controls. This motivated experimentally testing the human-specific role of miR-21-5p in hMSC-exo–mediated increases of cardiac tissue contractility. Treating hECTs with miR-21-5p alone was sufficient to recapitulate effects observed with hMSC-exo on hECT developed force and expression of associated calcium handling genes (eg, SERCA2a and L-type calcium channel). Conversely, knockdown of miR-21-5p in hMSCs significantly diminished exosomal procontractile and associated calcium handling gene expression effects on hECTs. Western blots supported miR-21-5p effects on calcium handling gene expression at the protein level, corresponding to significantly increased calcium transient amplitude and decreased decay time constant in comparison to miR-scramble control. Mechanistically, cotreating with miR-21-5p and LY294002, a PI3K inhibitor, suppressed these effects. Finally, mathematical simulations predicted the translational capacity for miR-21-5p treatment to restore calcium handling in mature ischemic adult human cardiomyocytes. Conclusions: miR-21-5p plays a key role in hMSC-exo–mediated effects on cardiac contractility and calcium handling, likely via PI3K signaling. These findings may open new avenues of research to harness the role of miR-21-5p in optimizing future stem cell-based cardiotherapies.


Molecular therapy. Methods & clinical development | 2015

Effectiveness of gene delivery systems for pluripotent and differentiated cells

Kleopatra Rapti; Francesca Stillitano; Ioannis Karakikes; Mathieu Nonnenmacher; Thomas Weber; Jean-Sebastian Hulot; Roger J. Hajjar

Human embryonic stem cells (hESC) and induced pluripotent stem cells (hiPSC) assert a great future for the cardiovascular diseases, both to study them and to explore therapies. However, a comprehensive assessment of the viral vectors used to modify these cells is lacking. In this study, we aimed to compare the transduction efficiency of recombinant adeno-associated vectors (AAV), adenoviruses and lentiviral vectors in hESC, hiPSC, and the derived cardiomyocytes. In undifferentiated cells, adenoviral and lentiviral vectors were superior, whereas in differentiated cells AAV surpassed at least lentiviral vectors. We also tested four AAV serotypes, 1, 2, 6, and 9, of which 2 and 6 were superior in their transduction efficiency. Interestingly, we observed that AAVs severely diminished the viability of undifferentiated cells, an effect mediated by induction of cell cycle arrest genes and apoptosis. Furthermore, we show that the transduction efficiency of the different viral vectors correlates with the abundance of their respective receptors. Finally, adenoviral delivery of the calcium-transporting ATPase SERCA2a to hESC and hiPSC-derived cardiomyocytes successfully resulted in faster calcium reuptake. In conclusion, adenoviral vectors prove to be efficient for both differentiated and undifferentiated lines, whereas lentiviral vectors are more applicable to undifferentiated cells and AAVs to differentiated cells.


Methods of Molecular Biology | 2017

Gene Transfer in Cardiomyocytes Derived from ES and iPS Cells

Francesca Stillitano; Ioannis Karakikes; Roger J. Hajjar

The advent of human induced pluripotent stem cell (hiPSC) technology has produced patient-specific hiPSC derived cardiomyocytes (hiPSC-CMs) that can be used as a platform to study cardiac diseases and to explore new therapies.The ability to genetically manipulate hiPSC-CMs not only is essential for identifying the structural and/or functional role of a protein but can also provide valuable information regarding therapeutic applications. In this chapter, we describe protocols for culture, maintenance, and cardiac differentiation of hiPSCs. Then, we provide a basic procedure to transduce hiPSC-CMs.


Archive | 2018

Cardiac Tissue Engineering Models of Inherited and Acquired Cardiomyopathies

Irene C. Turnbull; Joshua Mayourian; Jack F. Murphy; Francesca Stillitano; Delaine K. Ceholski; Kevin D. Costa

The lack of biomimetic in vitro models of the human heart has posed a critical barrier to progress in the field of modeling cardiac disease. Human engineered cardiac tissues (hECTs)-autonomous, beating structures that recapitulate key aspects of native cardiac muscle physiology-offer an attractive alternative to traditional in vitro models. Here we describe the use of hECTs to advance our understanding and modeling of cardiac diseases in order to test therapeutic interventions, with a focus on contractile dysfunction in the setting of inherited and acquired forms of cardiomyopathies. Four major procedures are discussed in this chapter: (1) preparation of hECTs from human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) on single-tissue and multitissue bioreactors; (2) data acquisition of hECT contractile function on both of these platforms; (3) hECT modeling of hereditary phospholamban-R14 deletion-dilated cardiomyopathy; and (4) cryo-injury and doxorubicin-induced hECT models of acquired cardiomyopathy.


Journal of Molecular and Cellular Cardiology | 2018

Functional and transcriptomic insights into pathogenesis of R9C phospholamban mutation using human induced pluripotent stem cell-derived cardiomyocytes

Delaine K. Ceholski; Irene C. Turnbull; Chi-Wing Kong; Simon Koplev; Joshua Mayourian; Przemek A. Gorski; Francesca Stillitano; Angelos A. Skodras; Mathieu Nonnenmacher; Ninette Cohen; Johan Björkegren; Daniel R. Stroik; Razvan L. Cornea; David D. Thomas; Ronald A. Li; Kevin D. Costa; Roger J. Hajjar

Dilated cardiomyopathy (DCM) can be caused by mutations in the cardiac protein phospholamban (PLN). We used CRISPR/Cas9 to insert the R9C PLN mutation at its endogenous locus into a human induced pluripotent stem cell (hiPSC) line from an individual with no cardiovascular disease. R9C PLN hiPSC-CMs display a blunted β-agonist response and defective calcium handling. In 3D human engineered cardiac tissues (hECTs), a blunted lusitropic response to β-adrenergic stimulation was observed with R9C PLN. hiPSC-CMs harboring the R9C PLN mutation showed activation of a hypertrophic phenotype, as evidenced by expression of hypertrophic markers and increased cell size and capacitance of cardiomyocytes. RNA-seq suggests that R9C PLN results in an altered metabolic state and profibrotic signaling, which was confirmed by gene expression analysis and picrosirius staining of R9C PLN hECTs. The expression of several miRNAs involved in fibrosis, hypertrophy, and cardiac metabolism were also perturbed in R9C PLN hiPSC-CMs. This study contributes to better understanding of the pathogenic mechanisms of the hereditary R9C PLN mutation in the context of human cardiomyocytes.

Collaboration


Dive into the Francesca Stillitano's collaboration.

Top Co-Authors

Avatar

Roger J. Hajjar

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kevin D. Costa

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Delaine K. Ceholski

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Mathieu Nonnenmacher

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Ronald A. Li

University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Irene C. Turnbull

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Jens Hansen

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Joshua Mayourian

Icahn School of Medicine at Mount Sinai

View shared research outputs
Researchain Logo
Decentralizing Knowledge