Network


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

Hotspot


Dive into the research topics where Elena Serena is active.

Publication


Featured researches published by Elena Serena.


Experimental Cell Research | 2009

Electrical stimulation of human embryonic stem cells: Cardiac differentiation and the generation of reactive oxygen species

Elena Serena; Elisa Figallo; Nina Tandon; Christopher Cannizzaro; Sharon Gerecht; Nicola Elvassore; Gordana Vunjak-Novakovic

Exogenous electric fields have been implied in cardiac differentiation of mouse embryonic stem cells and the generation of reactive oxygen species (ROS). In this work, we explored the effects of electrical field stimulation on ROS generation and cardiogenesis in embryoid bodies (EBs) derived from human embryonic stem cells (hESC, line H13), using a custom-built electrical stimulation bioreactor. Electrical properties of the bioreactor system were characterized by electrochemical impedance spectroscopy (EIS) and analysis of electrical currents. The effects of the electrode material (stainless steel, titanium-nitride-coated titanium, titanium), length of stimulus (1 and 90 s) and age of EBs at the onset of electrical stimulation (4 and 8 days) were investigated with respect to ROS generation. The amplitude of the applied electrical field was 1 V/mm. The highest rate of ROS generation was observed for stainless steel electrodes, for signal duration of 90 s and for 4-day-old EBs. Notably, comparable ROS generation was achieved by incubation of EBs with 1 nM H(2)O(2). Cardiac differentiation in these EBs was evidenced by spontaneous contractions, expression of troponin T and its sarcomeric organization. These results imply that electrical stimulation plays a role in cardiac differentiation of hESCs, through mechanisms associated with the intracellular generation of ROS.


Current Opinion in Biotechnology | 2014

Human-on-chip for therapy development and fundamental science.

Camilla Luni; Elena Serena; Nicola Elvassore

Organ-on-chip systems integrate microfluidic technology and living cells to study human physiology and pathophysiology. These human in vitro models are promising substitutes for animal testing, and their small scale enables precise control of culture conditions and high-throughput experiments, which would not be economically sustainable on a macroscopic level. Multiple sources of biological material are used in the development of organ-on-chips, from biopsies to stem cells. Each source has its own peculiarities and technical requirements for integration into microfluidic chips, and is suitable for specific applications. While a biopsy is the tissue of choice for the biomimetic response to ageing, induced pluripotent stem cells hold great promise for the study of genetic-related disease pathogenesis, and primary cultures can fill the gap.


Neurological Research | 2008

Electrophysiologic stimulation improves myogenic potential of muscle precursor cells grown in a 3D collagen scaffold

Elena Serena; Marina Flaibani; Silvia Carnio; Luisa Boldrin; Libero Vitiello; Paolo De Coppi; Nicola Elvassore

Abstract The production of engineered three-dimensional (3D) skeletal muscle grafts holds promise for treatment of several diseases. An important factor in the development of such approach involves the capability of preserving myogenicity and regenerative potential during ex vivo culturing. We have previously shown that electrical stimulation of myogenic cells grown in monolayer could improve the differentiation process. Here we investigated the effect of exogenous electrical field, specifically designed to mimic part of the neuronal activity, on muscle precursor cells (MPCs) cultured within 3D collagen scaffolds. Our data showed that electric stimulation did not affect cell viability and increased by 65.6% the release rate of NOx, an early molecular activator of satellite cells in vivo. NOx release rate was decreased by an inhibitor of NO synthase, both in stimulated and non-stimulated cultures, confirming the endocrine origin of the measured NOx. Importantly, electrical stimulation also increased the expression of two myogenic markers, MyoD and desmin. We also carried out some preliminary experiments aimed at determining the biocompatibility of our seeded collagen scaffolds, implanting them in the tibialis anterior muscles of syngeneic mice. Ten days after transplantation, we could observe the formation of new myofibers both inside the scaffold and at the scaffold/muscle interface. Altogether, our findings indicate that electrical stimulation could be a new strategy for the effective 3D expansion of muscle precursor cells in vitro without losing myogenic potential and that 3D collagen matrices could be a promising tool for delivering myogenic cells in recipient muscles.


Langmuir | 2012

Micropatterning Topology on Soft Substrates Affects Myoblast Proliferation and Differentiation

Susi Zatti; Alice Zoso; Elena Serena; Camilla Luni; Elisa Cimetta; Nicola Elvassore

Micropatterning techniques and substrate engineering are becoming useful tools to investigate several aspects of cell-cell interaction biology. In this work, we rationally study how different micropatterning geometries can affect myoblast behavior in the early stage of in vitro myogenesis. Soft hydrogels with physiological elastic modulus (E = 15 kPa) were micropatterned in parallel lanes (100, 300, and 500 μm width) resulting in different local and global myoblast densities. Proliferation and differentiation into multinucleated myotubes were evaluated for murine and human myoblasts. Wider lanes showed a decrease in murine myoblast proliferation: (69 ± 8)% in 100 μm wide lanes compared to (39 ± 7)% in 500 μm lanes. Conversely, fusion index increased in wider lanes: from (46 ± 7)% to (66 ± 7)% for murine myoblasts, and from (15 ± 3)% to (36 ± 2)% for human primary myoblasts, using a patterning width of 100 and 500 μm, respectively. These results are consistent with both computational modeling data and conditioned medium experiments, which demonstrated that wider lanes favor the accumulation of endogenous secreted factors. Interestingly, human primary myoblast proliferation is not affected by patterning width, which may be because the high serum content of their culture medium overrides the effect of secreted factors. These data highlight the role of micropatterning in shaping the cellular niche through secreted factor accumulation, and are of paramount importance in rationally understanding myogenesis in vitro for the correct design of in vitro skeletal muscle models.


International Journal of Artificial Organs | 2007

Enhancement of viability of muscle precursor cells on 3D scaffold in a perfusion bioreactor.

Elisa Cimetta; Marina Flaibani; M. Mella; Elena Serena; Luisa Boldrin; P De Coppi; Nicola Elvassore

The aim of this study was to develop a methodology for the in vitro expansion of skeletal-muscle precursor cells (SMPC) in a three-dimensional (3D) environment in order to fabricate a cellularized artificial graft characterized by high density of viable cells and uniform cell distribution over the entire 3D domain. Cell seeding and culture within 3D porous scaffolds by conventional static techniques can lead to a uniform cell distribution only on the scaffold surface, whereas dynamic culture systems have the potential of allowing a uniform growth of SMPCs within the entire scaffold structure. In this work, we designed and developed a perfusion bioreactor able to ensure long-term culture conditions and uniform flow of medium through 3D collagen sponges. A mathematical model to assist the design of the experimental setup and of the operative conditions was developed. The effects of dynamic vs static culture in terms of cell viability and spatial distribution within 3D collagen scaffolds were evaluated at 1, 4 and 7 days and for different flow rates of 1, 2, 3.5 and 4.5 ml/min using C2C12 muscle cell line and SMPCs derived from satellite cells. C2C12 cells, after 7 days of culture in our bioreactor, perfused applying a 3.5 ml/min flow rate, showed a higher viability resulting in a three-fold increase when compared with the same parameter evaluated for cultures kept under static conditions. In addition, dynamic culture resulted in a more uniform 3D cell distribution. The 3.5 ml/min flow rate in the bioreactor was also applied to satellite cell-derived SMPCs cultured on 3D collagen scaffolds. The dynamic culture conditions improved cell viability leading to higher cell density and uniform distribution throughout the entire 3D collagen sponge for both C2C12 and satellite cells.


Biotechnology and Bioengineering | 2012

Confined 3D microenvironment regulates early differentiation in human pluripotent stem cells.

Giovanni Giuseppe Giobbe; Monica Zagallo; M. Riello; Elena Serena; Giulia Masi; Luisa Barzon; Barbara Di Camillo; Nicola Elvassore

The therapeutic potential of human pluripotent stem (hPS) cells is threatened, among various problems, by the difficulty to homogenously direct cell differentiation into specific lineages. The transition from hPSC into committed differentiated cells is accompanied by secretome activity, remodeling of extracellular matrix and self‐organization into germ layers. In this work, we aimed to investigate how different three‐dimensional microenvironments regulate the early differentiation of the three germ layers in human embryonic stem (hES) cells derived embryoid bodies. In particular, a permeable, biocompatible, hydrogel microwell array was specifically designed for recreating a confined niche in which EB secreted molecules accumulate in accordance with hydrogel diffusional cut‐off. Fluorescence recovery after photobleaching technique was performed to accurately evaluate hydrogel permeability, mesh size and diffusional cutoff for soluble molecules. Three different culture conditions of EB culture were analyzed: suspension, confinement in microwells of width/depth ratio 1:1 and 1:2. Results show that EBs cultured in microwells are viable and have comparable average size after 8 days culture. Whole genome microarrays show that significative differential gene expression was observed between suspension and confined EBs culture. In particular, EBs culture in microwells promotes the expression of genes involved in pattern specification processes, brain development, ectoderm and endoderm differentiation. On the contrary, suspension EBs express instead genes involved in mesoderm specification and heart development. These results suggest that local accumulation of EBs secreted molecules drives differentiation patterns, as confirmed by immunofluorescence of germ layer markers, in hydrogel confined EB culture from both hES cells and human induced pluripotent stem (hiPS) cells. Our findings highlight an additional potential role of biomaterial in controlling hPSC differentiation through secreted factor niche specification. Biotechnol. Bioeng. 2012; 109: 3119–3132.


Nature Methods | 2016

High-efficiency cellular reprogramming with microfluidics

Camilla Luni; Stefano Giulitti; Elena Serena; Luca Ferrari; Alessandro Zambon; Onelia Gagliano; Giovanni Giuseppe Giobbe; Federica Michielin; Sebastian Knöbel; Andreas Bosio; Nicola Elvassore

We report that the efficiency of reprogramming human somatic cells to induced pluripotent stem cells (hiPSCs) can be dramatically improved in a microfluidic environment. Microliter-volume confinement resulted in a 50-fold increase in efficiency over traditional reprogramming by delivery of synthetic mRNAs encoding transcription factors. In these small volumes, extracellular components of the TGF-β and other signaling pathways exhibited temporal regulation that appears critical to acquisition of pluripotency. The high quality and purity of the resulting hiPSCs (μ-hiPSCs) allowed direct differentiation into functional hepatocyte- and cardiomyocyte-like cells in the same platform without additional expansion.


PLOS ONE | 2012

Micro-Arrayed Human Embryonic Stem Cells-Derived Cardiomyocytes for In Vitro Functional Assay

Elena Serena; Elisa Cimetta; Susi Zatti; Tania Zaglia; Monica Zagallo; Gordon Keller; Nicola Elvassore

Introduction The heart is one of the least regenerative organs in the body and any major insult can result in a significant loss of heart cells. The development of an in vitro-based cardiac tissue could be of paramount importance for many aspects of the cardiology research. In this context, we developed an in vitro assay based on human cardiomyocytes (hCMs) and ad hoc micro-technologies, suitable for several applications: from pharmacological analysis to physio-phatological studies on transplantable hCMs. We focused on the development of an assay able to analyze not only hCMs viability, but also their functionality. Methods hCMs were cultured onto a poly-acrylamide hydrogel with tunable tissue-like mechanical properties and organized through micropatterning in a 20×20 array. Arrayed hCMs were characterized by immunofluorescence, GAP-FRAP analyses and live and dead assay. Their functionality was evaluated monitoring the excitation-contraction coupling. Results Micropatterned hCMs maintained the expression of the major cardiac markers (cTnT, cTnI, Cx43, Nkx2.5, α-actinin) and functional properties. The spontaneous contraction frequency was (0.83±0.2) Hz, while exogenous electrical stimulation lead to an increase up to 2 Hz. As proof of concept that our device can be used for screening the effects of pathological conditions, hCMs were exposed to increasing levels of H2O2. Remarkably, hCMs viability was not compromised with exposure to 0.1 mM H2O2, but hCMs contractility was dramatically suppressed. As proof of concept, we also developed a microfluidic platform to selectively treat areas of the cell array, in the perspective of performing multi-parametric assay. Conclusions Such system could be a useful tool for testing the effects of multiple conditions on an in vitro cell model representative of human heart physiology, thus potentially helping the processes of therapy and drug development.


Molecular therapy. Methods & clinical development | 2014

Complete restoration of multiple dystrophin isoforms in genetically corrected Duchenne muscular dystrophy patient–derived cardiomyocytes

Susi Zatti; Sebastian Martewicz; Elena Serena; Narumi Uno; Giovanni Giuseppe Giobbe; Yasuhiro Kazuki; Mitsuo Oshimura; Nicola Elvassore

Duchenne muscular dystrophy (DMD)–associated cardiac diseases are emerging as a major cause of morbidity and mortality in DMD patients, and many therapies for treatment of skeletal muscle failed to improve cardiac function. The reprogramming of patients’ somatic cells into pluripotent stem cells, combined with technologies for correcting the genetic defect, possesses great potential for the development of new treatments for genetic diseases. In this study, we obtained human cardiomyocytes from DMD patient–derived, induced pluripotent stem cells genetically corrected with a human artificial chromosome carrying the whole dystrophin genomic sequence. Stimulation by cytokines was combined with cell culturing on hydrogel with physiological stiffness, allowing an adhesion-dependent maturation and a proper dystrophin expression. The obtained cardiomyocytes showed remarkable sarcomeric organization of cardiac troponin T and α-actinin, expressed cardiac-specific markers, and displayed electrically induced calcium transients lasting less than 1 second. We demonstrated that the human artificial chromosome carrying the whole dystrophin genomic sequence is stably maintained throughout the cardiac differentiation process and that multiple promoters of the dystrophin gene are properly activated, driving expression of different isoforms. These dystrophic cardiomyocytes can be a valuable source for in vitro modeling of DMD-associated cardiac disease. Furthermore, the derivation of genetically corrected, patient-specific cardiomyocytes represents a step toward the development of innovative cell and gene therapy approaches for DMD.


Journal of Tissue Engineering and Regenerative Medicine | 2011

Three‐dimensional porous scaffold allows long‐term wild‐type cell delivery in dystrophic muscle

Silvia Carnio; Elena Serena; Carlo Alberto Rossi; Paolo De Coppi; Nicola Elvassore; Libero Vitiello

Duchenne muscular dystrophy (DMD) is caused by the lack of dystrophin; affected muscles are characterized by continuous bouts of muscle degeneration, eventually leading to the exhaustion of the endogenous satellite cell pool. At present, only palliative treatments are available, although several gene and cell therapy‐based approaches are being studied. In this study we proposed to overcome the limitations hampering intramuscular cell injection by using a biomaterial‐based strategy. In particular, we used a three‐dimensional (3D) collagen porous scaffold to deliver myogenic precursor cells (MPCs) in vivo in the mdx murine model of DMD. MPCs, derived from single fibres of wild‐type donors, were expanded in vitro, seeded onto collagen scaffolds and implanted into the tibialis anterior muscles of normal and mdx mice. As a control, cells were delivered via direct intramuscular cell injection in the contralateral muscles. Scaffold‐delivered MPCs displayed lower apoptosis and higher proliferation than injected cells; in terms of dystrophin restoration, collagen scaffolds yielded better results than direct injections. Importantly, time‐course experiments indicated that the scaffolds acted as a cell reservoir, although cell migration was mostly contained within 400 µm from the scaffold–host tissue interface. Copyright

Collaboration


Dive into the Elena Serena'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

Paolo De Coppi

University College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge