Network


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

Hotspot


Dive into the research topics where Robert Maidhof is active.

Publication


Featured researches published by Robert Maidhof.


Nature Protocols | 2009

Electrical stimulation systems for cardiac tissue engineering

Nina Tandon; Christopher Cannizzaro; Pen-hsiu Grace Chao; Robert Maidhof; Anna Marsano; Hoi Ting Heidi Au; Milica Radisic; Gordana Vunjak-Novakovic

We describe a protocol for tissue engineering of synchronously contractile cardiac constructs by culturing cardiac cells with the application of pulsatile electrical fields designed to mimic those present in the native heart. Tissue culture is conducted in a customized chamber built to allow for cultivation of (i) engineered three-dimensional (3D) cardiac tissue constructs, (ii) cell monolayers on flat substrates or (iii) cells on patterned substrates. This also allows for analysis of the individual and interactive effects of pulsatile electrical field stimulation and substrate topography on cell differentiation and assembly. The protocol is designed to allow for delivery of predictable electrical field stimuli to cells, monitoring environmental parameters, and assessment of cell and tissue responses. The duration of the protocol is 5 d for two-dimensional cultures and 10 d for 3D cultures.


Journal of Tissue Engineering and Regenerative Medicine | 2011

Optimization of electrical stimulation parameters for cardiac tissue engineering.

Nina Tandon; Anna Marsano; Robert Maidhof; Leo Q. Wan; Hyoungshin Park; Gordana Vunjak-Novakovic

In vitro application of pulsatile electrical stimulation to neonatal rat cardiomyocytes cultured on polymer scaffolds has been shown to improve the functional assembly of cells into contractile engineered cardiac tissues. However, to date, the conditions of electrical stimulation have not been optimized. We have systematically varied the electrode material, amplitude and frequency of stimulation to determine the conditions that are optimal for cardiac tissue engineering. Carbon electrodes, exhibiting the highest charge‐injection capacity and producing cardiac tissues with the best structural and contractile properties, were thus used in tissue engineering studies. Engineered cardiac tissues stimulated at 3 V/cm amplitude and 3 Hz frequency had the highest tissue density, the highest concentrations of cardiac troponin‐I and connexin‐43 and the best‐developed contractile behaviour. These findings contribute to defining bioreactor design specifications and electrical stimulation regime for cardiac tissue engineering. Copyright


Journal of Tissue Engineering and Regenerative Medicine | 2012

Biomimetic perfusion and electrical stimulation applied in concert improved the assembly of engineered cardiac tissue

Robert Maidhof; Nina Tandon; Eun Jung Lee; Jianwen Luo; Yi Duan; Keith Yeager; Elisa E. Konofagou; Gordana Vunjak-Novakovic

Maintenance of normal myocardial function depends intimately on synchronous tissue contraction, driven by electrical activation and on adequate nutrient perfusion in support thereof. Bioreactors have been used to mimic aspects of these factors in vitro to engineer cardiac tissue but, due to design limitations, previous bioreactor systems have yet to simultaneously support nutrient perfusion, electrical stimulation and unconstrained (i.e. not isometric) tissue contraction. To the best of our knowledge, the bioreactor system described herein is the first to integrate these three key factors in concert. We present the design of our bioreactor and characterize its capability in integrated experimental and mathematical modelling studies. We then cultured cardiac cells obtained from neonatal rats in porous, channelled elastomer scaffolds with the simultaneous application of perfusion and electrical stimulation, with controls excluding either one or both of these two conditions. After 8 days of culture, constructs grown with simultaneous perfusion and electrical stimulation exhibited substantially improved functional properties, as evidenced by a significant increase in contraction amplitude (0.23 ± 0.10% vs 0.14 ± 0.05%, 0.13 ± 0.08% or 0.09 ± 0.02% in control constructs grown without stimulation, without perfusion, or either stimulation or perfusion, respectively). Consistently, these constructs had significantly improved DNA contents, cell distribution throughout the scaffold thickness, cardiac protein expression, cell morphology and overall tissue organization compared to control groups. Thus, the simultaneous application of medium perfusion and electrical conditioning enabled by the use of the novel bioreactor system may accelerate the generation of fully functional, clinically sized cardiac tissue constructs. Copyright


Biomaterials | 2013

The effect of controlled expression of VEGF by transduced myoblasts in a cardiac patch on vascularization in a mouse model of myocardial infarction

Anna Marsano; Robert Maidhof; Jianwen Luo; Kana Fujikara; Elisa E. Konofagou; Andrea Banfi; Gordana Vunjak-Novakovic

Key requirements for cardiac tissue engineering include the maintenance of cell viability and function and the establishment of a perfusable vascular network in millimeters thick and compact cardiac constructs upon implantation. We investigated if these requirements can be met by providing an intrinsic vascularization stimulus (via sustained action of VEGF secreted at a controlled rate by transduced myoblasts) to a cardiac patch engineered under conditions of effective oxygen supply (via medium flow through channeled elastomeric scaffolds seeded with neonatal cardiomyocytes). We demonstrate that this combined approach resulted in increased viability, vascularization and functionality of the cardiac patch. After implantation in a mouse model of myocardial infarction, VEGF-expressing patches displayed significantly improved engraftment, survival and differentiation of cardiomyocytes, leading to greatly enhanced contractility as compared to controls not expressing VEGF. Controlled VEGF expression also mediated the formation of mature vascular networks, both within the engineered patches and in the underlying ischemic myocardium. We propose that this combined cell-biomaterial approach can be a promising strategy to engineer cardiac patches with intrinsic and extrinsic vascularization potential.


Biotechnology Progress | 2010

Perfusion seeding of channeled elastomeric scaffolds with myocytes and endothelial cells for cardiac tissue engineering

Robert Maidhof; Anna Marsano; Eun Jung Lee; Gordana Vunjak-Novakovic

The requirements for engineering clinically sized cardiac constructs include medium perfusion (to maintain cell viability throughout the construct volume) and the protection of cardiac myocytes from hydrodynamic shear. To reconcile these conflicting requirements, we proposed the use of porous elastomeric scaffolds with an array of channels providing conduits for medium perfusion, and sized to provide efficient transport of oxygen to the cells, by a combination of convective flow and molecular diffusion over short distances between the channels. In this study, we investigate the conditions for perfusion seeding of channeled constructs with myocytes and endothelial cells without the gel carrier we previously used to lock the cells within the scaffold pores. We first established the flow parameters for perfusion seeding of porous elastomer scaffolds using the C2C12 myoblast line, and determined that a linear perfusion velocity of 1.0 mm/s resulted in seeding efficiency of 87% ± 26% within 2 hours. When applied to seeding of channeled scaffolds with neonatal rat cardiac myocytes, these conditions also resulted in high efficiency (77.2% ± 23.7%) of cell seeding. Uniform spatial cell distributions were obtained when scaffolds were stacked on top of one another in perfusion cartridges, effectively closing off the channels during perfusion seeding. Perfusion seeding of single scaffolds resulted in preferential cell attachment at the channel surfaces, and was employed for seeding scaffolds with rat aortic endothelial cells. We thus propose that these techniques can be utilized to engineer thick and compact cardiac constructs with parallel channels lined with endothelial cells.


Biotechnology Progress | 2010

Scaffold stiffness affects the contractile function of three-dimensional engineered cardiac constructs

Anna Marsano; Robert Maidhof; Leo Q. Wan; Yadong Wang; Jin Gao; Nina Tandon; Gordana Vunjak-Novakovic

We investigated the effects of the initial stiffness of a three‐dimensional elastomer scaffold—highly porous poly(glycerol sebacate)—on functional assembly of cardiomyocytes cultured with perfusion for 8 days. The polymer elasticity varied with the extent of polymer cross‐links, resulting in three different stiffness groups, with compressive modulus of 2.35 ± 0.03 (low), 5.28 ± 0.36 (medium), and 5.99 ± 0.40 (high) kPa. Laminin coating improved the efficiency of cell seeding (from 59 ± 15 to 90 ± 21%), resulting in markedly increased final cell density, construct contractility, and matrix deposition, likely because of enhanced cell interaction and spreading on scaffold surfaces. Compact tissue was formed in the low and medium stiffness groups, but not in the high stiffness group. In particular, the low stiffness group exhibited the greatest contraction amplitude in response to electric field pacing, and had the highest compressive modulus at the end of culture. A mathematical model was developed to establish a correlation between the contractile amplitude and the cell distribution within the scaffold. Taken together, our findings suggest that the contractile function of engineered cardiac constructs positively correlates with low compressive stiffness of the scaffold.


Journal of Tissue Engineering and Regenerative Medicine | 2012

Channelled scaffolds for engineering myocardium with mechanical stimulation.

Ting Zhang; Leo Q. Wan; Zhuo Xiong; Anna Marsano; Robert Maidhof; Miri Park; Yongnian Yan; Gordana Vunjak-Novakovic

The characteristics of the matrix (composition, structure, mechanical properties) and external culture environment (pulsatile perfusion, physical stimulation) of the heart are important characteristics in the engineering of functional myocardial tissue. This study reports on the development of chitosan‐collagen scaffolds with micropores and an array of parallel channels (~ 200 µm in diameter) that were specifically designed for cardiac tissue engineering using mechanical stimulation. The scaffolds were designed to have similar structural and mechanical properties of those of native heart matrix. Scaffolds were seeded with neonatal rat heart cells and subjected to dynamic tensile stretch using a custom designed bioreactor. The channels enhanced oxygen transport and facilitated the establishment of cell connections within the construct. The myocardial patches (14 mm in diameter, 1–2 mm thick) consisted of metabolically active cells that began to contract synchronously after 3 days of culture. Mechanical stimulation with high tensile stress promoted cell alignment, elongation, and expression of connexin‐43 (Cx‐43). This study confirms the importance of scaffold design and mechanical stimulation for the formation of contractile cardiac constructs. Copyright


international conference of the ieee engineering in medicine and biology society | 2008

Engineering of functional contractile cardiac tissues cultured in a perfusion system

Anna Marsano; Robert Maidhof; Nina Tandon; Jin Gao; Yadong Wang; Gordana Vunjak-Novakovic

Overcoming the limitations of diffusional transport in conventional culture systems remains an open issue for successfully generating thick, compact and functional cardiac tissues. Previously, it was shown that perfusion systems enhance the yield and uniformity of cell seeding and cell survival in thick cardiac constructs. The aim of our study was to form highly functional cardiac constructs starting from spatially uniform, high density cell seeded constructs. Disk-shaped elastomeric poly(glycerol sebacate) scaffolds were seeded with neonatal rat cardiomyocytes and cultured for eight days with direct perfusion of culture medium or statically in a six-well plate. In the perfusion experimental group, the integrity of some disks was well maintained, whereas in others a central hole was formed, resulting in ring-shaped constructs. This allowed us to also study the effects of construct geometry and of interstitial flow versus channel perfusion. The ring-shaped constructs appeared to have a denser and more uniform deposition of extracellular matrix. In response to electrical stimulation, the fractional area change of the ring-shaped constructs was 7.3 and 2.7 times higher than for disk-shaped tissues cultured in perfusion or statically, respectively. These findings suggest that a combination of many factors, including scaffold elasticity and geometry and the type of perfusion system applied, need to be considered in order to engineer a cardiac construct with high contractile activity.


Archive | 2011

Tissue Engineering Strategies for Cardiac Regeneration

Amandine Godier-Furnemont; Yi Duan; Robert Maidhof; Gordana Vunjak-Novakovic

Once injured, cardiac muscle does not regenerate. Massive and irreversible loss of cardiomyocytes due to myocardial infarction remains the main cause of heart failure. Cardiac tissue engineering has a potential to reestablish the structure and function of injured myocardium, by (1) injection of cardiogenic cells, (2) transplantation of functional cardiac tissue constructs, or (3) mobilization of endogenous repair cells. Irrespective of the therapeutic strategy, the regeneration depends on both the biological potential of the repair cells and the environment (matrix, signals) to which the cells are subjected. In general, the biological potential of the cells – the actual “tissue engineers” – needs to be mobilized by providing highly controllable “biomimetic” environments designed to enhance cell survival, differentiation, and electromechanical coupling. For cardiac regeneration, some of the key requirements include the establishment of cardiac tissue matrix, electromechanical cell coupling, robust contractile function, and functional vascularization. Engineered tissue constructs can also serve as high-fidelity models to study cardiac development and disease. We review the potential and challenges of cardiac tissue engineering to provide for the development of therapies for heart regeneration.


ASME 2009 Summer Bioengineering Conference, Parts A and B | 2009

Chitosan-Collagen Based Channeled Scaffold for Cardiac Tissue Engineering

Ting Zhang; Leo Q. Wan; Anna Marsano; Robert Maidhof; Yongnian Yan; Jiluan Pan; Gordana Vunjak-Novakovic

Tremendous efforts have been made for engineering cardiac tissue for myocardial infarction therapy [1]. Various materials and forming methods have been explored, but due to the specific physiological properties of cardiac muscle tissue, challenges still exist [2]. One of those is to create biomimetic extracellular materials to support cell function and electromechanical coupling. The scaffold material should be biocompatible, biochemically stable, mechanically strong, and highly extensible, just like native heart tissue [2]. Another challenge is to create vascular networks for oxygen and nutrient supply, much as the capillaries do in natural heart tissue [3]. In this study, chitosan and collagen were chosen to fabricate cardiac constructs with channels as small as 200 μm in diameter. Several factors such as chitosan and crosslinker concentrations and coating proteins were optimized for mechanical strength and cell seeding efficiency. Engineered tissues of significant size (12 mm in diameter × 2 mm thick) were generated in vitro using this method.Copyright

Collaboration


Dive into the Robert Maidhof's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Leo Q. Wan

Rensselaer Polytechnic Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yadong Wang

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge