Honors & Awards


  • Andrew Olson Scientific Image Award, Stanford (2022)
  • UC San Diego Future Faculty of Cardiovascular Sciences (FOCUS) Summer Institute Scholar, Stanford (2020-2021)
  • Stanford MCHRI Transdisciplinary Initiatives Program (TIP) Award (Co-I), Stanford (2020-2022)
  • Stanford-Penn Cardiovascular Meeting-1st Place Award, Stanford (2019)
  • NIH NHLBI K01 Career Development Award (PI), Stanford (2016-2022)
  • Siebel Scholar, Stanford (2011-2012)
  • New York Stem Cell Foundation Meeting-1st Place Award, Stanford (2011)
  • Advanced Residency Program at Stanford (ARTS), Stanford (2007-2011)
  • International Endovascular Fellows' Research Award, 1st Place, Stanford (2006)
  • Ethicon Endosurgery Fellowship, Stanford (2005-2006)
  • Dean's Fellowship, Stanford (2005)
  • Franklyn Ellenbogen Prize in Hematology/Oncology, Cornell (2002)
  • US-European Medical Education Exchange (US-EUMEE) Fellowship, Cornell (2002)
  • Max Kade Foundation Fellowship, Cornell (2002)
  • Pi Tau Sigma Engineering Honor Society, University of Texas (1991)

Boards, Advisory Committees, Professional Organizations


  • Member, International Society for Stem Cell Research (ISSCR) (2020 - Present)
  • Member, Society for Biomaterials (SFB) (2020 - Present)
  • Member, International Society for Applied Cardiovascular Biology (ISACB) (2020 - Present)

Professional Education


  • Instructor, Stanford University (2013-2021), Cardiovascular Medicine (2013)
  • PhD, Stanford University, Bioengineering (2012)
  • Resident, Stanford University Medical Center, Surgery (2004)
  • Intern, Stanford University Medical Center, Surgery (2003)
  • MD, Cornell University-Weill Medical College, Medicine (2002)
  • BS, University of Texas at Austin, Mechanical Engineering (1992)

Current Research and Scholarly Interests


Dr. Abilez' lab combines human pluripotent stem cell (hPSC) biology, systems biology, developmental biology, bioengineering (systems developmental bioengineering), biotechnology development (optogenetics, microscopes, cell sorters, biomechanical devices), computational modeling, and tissue/organoid engineering to model and control the earliest stages of cardiac development and vascularization. We are also using the same approaches to vascularize neural and hepatic tissue.

We primarily study hPSC directed differentiated into 2D and 3D aggregates, as engineered- and self-organized cardiovascular tissues and organoids. These tissues and organoids exhibit fascinating levels of patterning, self-organization, and function. By collecting high-content, high-throughput, and comprehensive functional data, such as live cell time-lapse fluorescence imaging, single-cell RNA-sequencing (scRNA-seq), and optogenetics-based microscopy, we quantify gene expression, signaling pathways, signaling networks, cell-cell communication, and functions across different cell types at spatiotemporal scales that span several orders of magnitude. We use our established approaches to ask how cells in complex systems make decisions about cell fate, respond to changes in applied external biophysical stimuli, and communicate to produce higher-order functions such as coordinated cardiomyocyte contractions at the tissue level to cardiac pumping at the organ level. In future studies we aim to perform gain/loss-of-function studies (through signaling pathway activation/inhibition and CRISPR gene-editing), epi-genetic regulation studies (scATAC-seq), metabolomics, and proteomics for dissecting gene regulatory networks and enabling mechanistic studies.

Our combined hPSC biology, systems developmental bioengineering, biotechnology development, computational modeling, and tissue/organoid engineering approaches endeavor to provide an increased understanding of complex cellular systems and to realize applications that include modeling developmental processes, modeling diseases, screening drugs, and creating transplantable vascularized tissues.

2023-24 Courses


Stanford Advisees


Work Experience


  • Postdoctoral Scholar, Stanford University (7/1/2004 - 9/30/2007)

    Location

    Clark Center E350, 318 W Campus Dr, Stanford, CA 94305

  • Postdoctoral Scholar, Stanford University (7/1/2012 - 3/31/2013)

    Location

    Clark Center E250, 318 W Campus Dr, Stanford, CA 94305

  • Instructor, Stanford University (4/1/2013 - 7/31/2021)

    Location

    Clark Center E250, 318 W Campus Dr, Stanford, CA 94305

All Publications


  • Tachycardia-induced metabolic rewiring as a driver of contractile dysfunction. Nature biomedical engineering Tu, C., Caudal, A., Liu, Y., Gorgodze, N., Zhang, H., Lam, C. K., Dai, Y., Zhang, A., Wnorowski, A., Wu, M. A., Yang, H., Abilez, O. J., Lyu, X., Narayan, S. M., Mestroni, L., Taylor, M. R., Recchia, F. A., Wu, J. C. 2023

    Abstract

    Prolonged tachycardia-a risk factor for cardiovascular morbidity and mortality-can induce cardiomyopathy in the absence of structural disease in the heart. Here, by leveraging human patient data, a canine model of tachycardia and engineered heart tissue generated from human induced pluripotent stem cells, we show that metabolic rewiring during tachycardia drives contractile dysfunction by promoting tissue hypoxia, elevated glucose utilization and the suppression of oxidative phosphorylation. Mechanistically, a metabolic shift towards anaerobic glycolysis disrupts the redox balance of nicotinamide adenine dinucleotide (NAD), resulting in increased global protein acetylation (and in particular the acetylation of sarcoplasmic/endoplasmic reticulum Ca2+-ATPase), a molecular signature of heart failure. Restoration of NAD redox by NAD+ supplementation reduced sarcoplasmic/endoplasmic reticulum Ca2+-ATPase acetylation and accelerated the functional recovery of the engineered heart tissue after tachycardia. Understanding how metabolic rewiring drives tachycardia-induced cardiomyopathy opens up opportunities for therapeutic intervention.

    View details for DOI 10.1038/s41551-023-01134-x

    View details for PubMedID 38012305

    View details for PubMedCentralID 5336809

  • Micropatterned Organoids Enable Modeling of the Earliest Stages of Human Cardiac Vascularization bioRxiv Abilez, O. J., et al 2022
  • Transcriptome Analysis of Non-Human Primate Induced Pluripotent Stem Cell-Derived Cardiomyocytes in 2D Monolayer Culture versus 3D Engineered Heart Tissue. Cardiovascular research Yang, H. n., Shao, N. n., Holmström, A. n., Zhao, X. n., Chour, T. n., Chen, H. n., Itzhaki, I. n., Wu, H. n., Ameen, M. n., Cunningham, N. J., Tu, C. n., Zhao, M. T., Tarantal, A. F., Abilez, O. J., Wu, J. C. 2020

    Abstract

    Stem cell therapy has shown promise for treating myocardial infarction (MI) via re-muscularization and paracrine signaling in both small and large animals. Non-human primates (NHPs), such as rhesus macaques (Macaca mulatta), are primarily utilized in preclinical trials due to their similarity to humans, both genetically and physiologically. Currently, induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) are delivered into the infarcted myocardium by either direct cell injection or an engineered tissue patch. Although both approaches have advantages in terms of sample preparation, cell-host interaction, and engraftment, how the iPSC-CMs respond to ischemic conditions in the infarcted heart under these two different delivery approaches remain unclear. Here we aim to gain a better understanding of the effects of hypoxia on iPSC-CMs at the transcriptome level.NHP iPSC-CMs in both monolayer culture (2 D) and engineered heart tissue (EHT) (3 D) format were exposed to hypoxic conditions to serve as surrogates of direct cell injection and tissue implantation in vivo, respectively. Outcomes were compared at the transcriptome level. We found the 3 D EHT model was more sensitive to ischemic conditions and similar to the native in vivo myocardium in terms of cell-extracellular matrix/cell-cell interactions, energy metabolism, and paracrine signaling.By exposing NHP iPSC-CMs to different culture conditions, transcriptome profiling improves our understanding of the mechanism of ischemic injury.Stem cell therapy has shown promise for treating ischemic heart tissue. However, how stem cells respond following different delivery method is unclear. Here hypoxic conditioning was applied to non-human primate iPSC-CMs in 2 D monolayer culture and 3 D engineered heart tissue to model cell injection versus patch implantation, respectively, in an ischemic milieu. The differential transcriptome of hypoxic effects on iPSC-CMs show upregulation of ECM-cell/cell-cell interactions (COL9A1, ITGB6, CTSV, and EPHA1), energy metabolism/hypoxia (ALDOC, ENO2, PFKFB4, CA3, and CA9), and paracrine signaling (WNT, PDGF, FGFR, EGFR, PI3K, and VEGF) in the 3 D format, which suggest engineered heart tissue as more suitable model for evaluating cardiac regenerative therapy.

    View details for DOI 10.1093/cvr/cvaa281

    View details for PubMedID 33002105

  • Endogenous Retrovirus-Derived lncRNA BANCR Promotes Cardiomyocyte Migration in Humans and Non-human Primates. Developmental cell Wilson, K. D., Ameen, M. n., Guo, H. n., Abilez, O. J., Tian, L. n., Mumbach, M. R., Diecke, S. n., Qin, X. n., Liu, Y. n., Yang, H. n., Ma, N. n., Gaddam, S. n., Cunningham, N. J., Gu, M. n., Neofytou, E. n., Prado, M. n., Hildebrandt, T. B., Karakikes, I. n., Chang, H. Y., Wu, J. C. 2020

    Abstract

    Transposable elements (TEs) comprise nearly half of the human genome and are often transcribed or exhibit cis-regulatory properties with unknown function in specific processes such as heart development. In the case of endogenous retroviruses (ERVs), a TE subclass, experimental interrogation is constrained as many are primate-specific or human-specific. Here, we use primate pluripotent stem-cell-derived cardiomyocytes that mimic fetal cardiomyocytes in vitro to discover hundreds of ERV transcripts from the primate-specific MER41 family, some of which are regulated by the cardiogenic transcription factor TBX5. The most significant of these are located within BANCR, a long non-coding RNA (lncRNA) exclusively expressed in primate fetal cardiomyocytes. Functional studies reveal that BANCR promotes cardiomyocyte migration in vitro and ventricular enlargement in vivo. We conclude that recently evolved TE loci such as BANCR may represent potent de novo developmental regulatory elements that can be interrogated with species-matching pluripotent stem cell models.

    View details for DOI 10.1016/j.devcel.2020.07.006

    View details for PubMedID 32763147

  • Passive Stretch Induces Structural and Functional Maturation of Engineered Heart Muscle as Predicted by Computational Modeling. Stem cells (Dayton, Ohio) Abilez, O. J., Tzatzalos, E. n., Yang, H. n., Zhao, M. T., Jung, G. n., Zöllner, A. M., Tiburcy, M. n., Riegler, J. n., Matsa, E. n., Shukla, P. n., Zhuge, Y. n., Chour, T. n., Chen, V. C., Burridge, P. W., Karakikes, I. n., Kuhl, E. n., Bernstein, D. n., Couture, L. A., Gold, J. D., Zimmermann, W. H., Wu, J. C. 2017

    Abstract

    The ability to differentiate human pluripotent stem cells (hPSCs) into cardiomyocytes (CMs) makes them an attractive source for repairing injured myocardium, disease modeling, and drug testing. Although current differentiation protocols yield hPSC-CMs to >90% efficiency, hPSC-CMs exhibit immature characteristics. With the goal of overcoming this limitation, we tested the effects of varying passive stretch on engineered heart muscle (EHM) structural and functional maturation, guided by computational modeling.Human embryonic stem cells (hESCs, H7 line) or human induced pluripotent stem cells (hiPSCs, IMR-90 line) were differentiated to human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) in vitro using a small molecule based protocol. hPSC-CMs were characterized by troponin(+) flow cytometry as well as electrophysiological measurements. Afterwards, 1.2 x 10(6) hPSC-CMs were mixed with 0.4 x 10(6) human fibroblasts (IMR-90 line) (3:1 ratio) and Type-I collagen. The blend was cast into custom-made 12-mm long polydimethylsiloxane (PDMS) reservoirs to vary nominal passive stretch of EHMs to 5, 7, or 9 mm. EHM characteristics were monitored for up to 50 days, with EHMs having a passive stretch of 7 mm giving the most consistent formation. Based on our initial macroscopic observations of EHM formation, we created a computational model that predicts the stress distribution throughout EHMs, which is a function of cellular composition, cellular ratio, and geometry. Based on this predictive modeling, we show cell alignment by immunohistochemistry and coordinated calcium waves by calcium imaging. Furthermore, coordinated calcium waves and mechanical contractions were apparent throughout entire EHMs. The stiffness and active forces of hPSC-derived EHMs are comparable to rat neonatal cardiomyocyte-derived EHMs. Three-dimensional EHMs display increased expression of mature cardiomyocyte genes including sarcomeric protein troponin-T, calcium and potassium ion channels, β-adrenergic receptors, and t-tubule protein caveolin-3.Passive stretch affects the structural and functional maturation of EHMs. Based on our predictive computational modeling, we show how to optimize cell alignment and calcium dynamics within EHMs. These findings provide a basis for the rational design of EHMs, which enables future scale-up productions for clinical use in cardiovascular tissue engineering. This article is protected by copyright. All rights reserved.

    View details for PubMedID 29086457

  • Stem cell reprogramming: A 3D boost. Nature materials Abilez, O. J., Wu, J. C. 2016; 15 (3): 259-261

    View details for DOI 10.1038/nmat4583

    View details for PubMedID 26906959

  • Chemically defined generation of human cardiomyocytes. Nature methods Burridge, P. W., Matsa, E., Shukla, P., Lin, Z. C., Churko, J. M., Ebert, A. D., Lan, F., Diecke, S., Huber, B., Mordwinkin, N. M., Plews, J. R., Abilez, O. J., Cui, B., Gold, J. D., Wu, J. C. 2014; 11 (8): 855-860

    Abstract

    Existing methods for human induced pluripotent stem cell (hiPSC) cardiac differentiation are efficient but require complex, undefined medium constituents that hinder further elucidation of the molecular mechanisms of cardiomyogenesis. Using hiPSCs derived under chemically defined conditions on synthetic matrices, we systematically developed an optimized cardiac differentiation strategy, using a chemically defined medium consisting of just three components: the basal medium RPMI 1640, L-ascorbic acid 2-phosphate and rice-derived recombinant human albumin. Along with small molecule-based induction of differentiation, this protocol produced contractile sheets of up to 95% TNNT2(+) cardiomyocytes at a yield of up to 100 cardiomyocytes for every input pluripotent cell and was effective in 11 hiPSC lines tested. This chemically defined platform for cardiac specification of hiPSCs will allow the elucidation of cardiomyocyte macromolecular and metabolic requirements and will provide a minimal system for the study of maturation and subtype specification.

    View details for DOI 10.1038/nmeth.2999

    View details for PubMedID 24930130

  • Prospective isolation of human embryonic stem cell-derived cardiovascular progenitors that integrate into human fetal heart tissue PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Ardehali, R., Ali, S. R., Inlay, M. A., Abilez, O. J., Chen, M. Q., Blauwkamp, T. A., Yazawa, M., Gong, Y., Nusse, R., Drukker, M., Weissman, I. L. 2013; 110 (9): 3405-3410

    Abstract

    A goal of regenerative medicine is to identify cardiovascular progenitors from human ES cells (hESCs) that can functionally integrate into the human heart. Previous studies to evaluate the developmental potential of candidate hESC-derived progenitors have delivered these cells into murine and porcine cardiac tissue, with inconclusive evidence regarding the capacity of these human cells to physiologically engraft in xenotransplantation assays. Further, the potential of hESC-derived cardiovascular lineage cells to functionally couple to human myocardium remains untested and unknown. Here, we have prospectively identified a population of hESC-derived ROR2(+)/CD13(+)/KDR(+)/PDGFRα(+) cells that give rise to cardiomyocytes, endothelial cells, and vascular smooth muscle cells in vitro at a clonal level. We observed rare clusters of ROR2(+) cells and diffuse expression of KDR and PDGFRα in first-trimester human fetal hearts. We then developed an in vivo transplantation model by transplanting second-trimester human fetal heart tissues s.c. into the ear pinna of a SCID mouse. ROR2(+)/CD13(+)/KDR(+)/PDGFRα(+) cells were delivered into these functioning fetal heart tissues: in contrast to traditional murine heart models for cell transplantation, we show structural and functional integration of hESC-derived cardiovascular progenitors into human heart.

    View details for DOI 10.1073/pnas.1220832110

    View details for Web of Science ID 000315841900046

    View details for PubMedID 23391730

    View details for PubMedCentralID PMC3587189

  • Abnormal Calcium Handling Properties Underlie Familial Hypertrophic Cardiomyopathy Pathology in Patient-Specific Induced Pluripotent Stem Cells CELL STEM CELL Lan, F., Lee, A. S., Liang, P., Sanchez-Freire, V., Nguyen, P. K., Wang, L., Han, L., Yen, M., Wang, Y., Sun, N., Abilez, O. J., Hu, S., Ebert, A. D., Navarrete, E. G., Simmons, C. S., Wheeler, M., Pruitt, B., Lewis, R., Yamaguchi, Y., Ashley, E. A., Bers, D. M., Robbins, R. C., Longaker, M. T., Wu, J. C. 2013; 12 (1): 101-113

    Abstract

    Familial hypertrophic cardiomyopathy (HCM) is a prevalent hereditary cardiac disorder linked to arrhythmia and sudden cardiac death. While the causes of HCM have been identified as genetic mutations in the cardiac sarcomere, the pathways by which sarcomeric mutations engender myocyte hypertrophy and electrophysiological abnormalities are not understood. To elucidate the mechanisms underlying HCM development, we generated patient-specific induced pluripotent stem cell cardiomyocytes (iPSC-CMs) from a ten-member family cohort carrying a hereditary HCM missense mutation (Arg663His) in the MYH7 gene. Diseased iPSC-CMs recapitulated numerous aspects of the HCM phenotype including cellular enlargement and contractile arrhythmia at the single-cell level. Calcium (Ca(2+)) imaging indicated dysregulation of Ca(2+) cycling and elevation in intracellular Ca(2+) ([Ca(2+)](i)) are central mechanisms for disease pathogenesis. Pharmacological restoration of Ca(2+) homeostasis prevented development of hypertrophy and electrophysiological irregularities. We anticipate that these findings will help elucidate the mechanisms underlying HCM development and identify novel therapies for the disease.

    View details for DOI 10.1016/j.stem.2012.10.010

    View details for Web of Science ID 000313839500014

    View details for PubMedID 23290139

    View details for PubMedCentralID PMC3638033

  • Robust pluripotent stem cell expansion and cardiomyocyte differentiation via geometric patterning INTEGRATIVE BIOLOGY Myers, F. B., Silver, J. S., Yan Zhuge, Z. G., Beygui, R. E., Zarins, C. K., Lee, L. P., Abilez, O. J. 2013; 5 (12): 1495-1506

    Abstract

    Geometric factors including the size, shape, density, and spacing of pluripotent stem cell colonies play a significant role in the maintenance of pluripotency and in cell fate determination. These factors are impossible to control using standard tissue culture methods. As such, there can be substantial batch-to-batch variability in cell line maintenance and differentiation yield. Here, we demonstrate a simple, robust technique for pluripotent stem cell expansion and cardiomyocyte differentiation by patterning cell colonies with a silicone stencil. We have observed that patterning human induced pluripotent stem cell (hiPSC) colonies improves the uniformity and repeatability of their size, density, and shape. Uniformity of colony geometry leads to improved homogeneity in the expression of pluripotency markers SSEA4 and Nanog as compared with conventional clump passaging. Patterned cell colonies are capable of undergoing directed differentiation into spontaneously beating cardiomyocyte clusters with improved yield and repeatability over unpatterned cultures seeded either as cell clumps or uniform single cell suspensions. Circular patterns result in a highly repeatable 3D ring-shaped band of cardiomyocytes which electrically couple and lead to propagating contraction waves around the ring. Because of these advantages, geometrically patterning stem cells using stencils may offer greater repeatability from batch-to-batch and person-to-person, an increase in differentiation yield, a faster experimental workflow, and a simpler protocol to communicate and follow. Furthermore, the ability to control where cardiomyocytes arise across a culture well during differentiation could greatly aid the design of electrophysiological assays for drug-screening.

    View details for DOI 10.1039/c2ib20191g

    View details for Web of Science ID 000327260600009

    View details for PubMedCentralID PMC3918460

  • Multiscale Computational Models for Optogenetic Control of Cardiac Function BIOPHYSICAL JOURNAL Abilez, O. J., Wong, J., Prakash, R., Deisseroth, K., Zarins, C. K., Kuhl, E. 2011; 101 (6): 1326-1334

    Abstract

    The ability to stimulate mammalian cells with light has significantly changed our understanding of electrically excitable tissues in health and disease, paving the way toward various novel therapeutic applications. Here, we demonstrate the potential of optogenetic control in cardiac cells using a hybrid experimental/computational technique. Experimentally, we introduced channelrhodopsin-2 into undifferentiated human embryonic stem cells via a lentiviral vector, and sorted and expanded the genetically engineered cells. Via directed differentiation, we created channelrhodopsin-expressing cardiomyocytes, which we subjected to optical stimulation. To quantify the impact of photostimulation, we assessed electrical, biochemical, and mechanical signals using patch-clamping, multielectrode array recordings, and video microscopy. Computationally, we introduced channelrhodopsin-2 into a classic autorhythmic cardiac cell model via an additional photocurrent governed by a light-sensitive gating variable. Upon optical stimulation, the channel opens and allows sodium ions to enter the cell, inducing a fast upstroke of the transmembrane potential. We calibrated the channelrhodopsin-expressing cell model using single action potential readings for different photostimulation amplitudes, pulse widths, and frequencies. To illustrate the potential of the proposed approach, we virtually injected channelrhodopsin-expressing cells into different locations of a human heart, and explored its activation sequences upon optical stimulation. Our experimentally calibrated computational toolbox allows us to virtually probe landscapes of process parameters, and identify optimal photostimulation sequences toward pacing hearts with light.

    View details for DOI 10.1016/j.bpj.2011.08.004

    View details for Web of Science ID 000295197300006

    View details for PubMedID 21943413

    View details for PubMedCentralID PMC3177076

  • Treatment of volumetric muscle loss in mice using nanofibrillar scaffolds enhances vascular organization and integration. Communications biology Nakayama, K. H., Quarta, M., Paine, P., Alcazar, C., Karakikes, I., Garcia, V., Abilez, O. J., Calvo, N. S., Simmons, C. S., Rando, T. A., Huang, N. F. 2019; 2: 170

    Abstract

    Traumatic skeletal muscle injuries cause irreversible tissue damage and impaired revascularization. Engineered muscle is promising for enhancing tissue revascularization and regeneration in injured muscle. Here we fabricated engineered skeletal muscle composed of myotubes interspersed with vascular endothelial cells using spatially patterned scaffolds that induce aligned cellular organization, and then assessed their therapeutic benefit for treatment of murine volumetric muscle loss. Murine skeletal myoblasts co-cultured with endothelial cells in aligned nanofibrillar scaffolds form endothelialized and aligned muscle with longer myotubes, more synchronized contractility, and more abundant secretion of angiogenic cytokines, compared to endothelialized engineered muscle formed from randomly-oriented scaffolds. Treatment of traumatically injured muscle with endothelialized and aligned skeletal muscle promotes the formation of highly organized myofibers and microvasculature, along with greater vascular perfusion, compared to treatment of muscle derived from randomly-oriented scaffolds. This work demonstrates the potential of endothelialized and aligned engineered skeletal muscle to promote vascular regeneration following transplantation.

    View details for DOI 10.1038/s42003-019-0416-4

    View details for PubMedID 31098403

    View details for PubMedCentralID PMC6505043

  • An in Vivo miRNA Delivery System for Restoring Infarcted Myocardium. ACS nano Yang, H. n., Qin, X. n., Wang, H. n., Zhao, X. n., Liu, Y. n., Wo, H. T., Liu, C. n., Nishiga, M. n., Chen, H. n., Ge, J. n., Sayed, N. n., Abilez, O. J., Ding, D. n., Heilshorn, S. C., Li, K. n. 2019

    Abstract

    A major challenge in myocardial infarction (MI)-related heart failure treatment using microRNA is the efficient and sustainable delivery of miRNAs into myocardium to achieve functional improvement through stimulation of intrinsic myocardial restoration. In this study, we established an in vivo delivery system using polymeric nanoparticles to carry miRNA (miNPs) for localized delivery within a shear-thinning injectable hydrogel. The miNPs triggered proliferation of human embryonic stem cell-derived cardiomyocytes and endothelial cells (hESC-CMs and hESC-ECs) and promoted angiogenesis in hypoxic conditions, showing significantly lower cytotoxicity than Lipofectamine. Furthermore, one injected dose of hydrogel/miNP in MI rats demonstrated significantly improved cardiac functions: increased ejection fraction from 45% to 64%, reduced scar size from 20% to 10%, and doubled capillary density in the border zone compared to the control group at 4 weeks. As such, our results indicate that this injectable hydrogel/miNP composite can deliver miRNA to restore injured myocardium efficiently and safely.

    View details for DOI 10.1021/acsnano.9b03343

    View details for PubMedID 31149806

  • Big bottlenecks in cardiovascular tissue engineering. Communications biology Huang, N. F., Serpooshan, V., Morris, V. B., Sayed, N., Pardon, G., Abilez, O. J., Nakayama, K. H., Pruitt, B. L., Wu, S. M., Yoon, Y., Zhang, J., Wu, J. C. 2018; 1: 199

    Abstract

    Although tissue engineering using human-induced pluripotent stem cells is a promising approach for treatment of cardiovascular diseases, some limiting factors include the survival, electrical integration, maturity, scalability, and immune response of three-dimensional (3D) engineered tissues. Here we discuss these important roadblocks facing the tissue engineering field and suggest potential approaches to overcome these challenges.

    View details for PubMedID 30480100

  • Partial Reprogramming of Pluripotent Stem Cell-Derived Cardiomyocytes into Neurons SCIENTIFIC REPORTS Chuang, W., Sharma, A., Shukla, P., Li, G., Mall, M., Rajarajan, K., Abilez, O. J., Hamaguchi, R., Wu, J. C., Wernig, M., Wu, S. M. 2017; 7

    Abstract

    Direct reprogramming of somatic cells has been demonstrated, however, it is unknown whether electrophysiologically-active somatic cells derived from separate germ layers can be interconverted. We demonstrate that partial direct reprogramming of mesoderm-derived cardiomyocytes into neurons is feasible, generating cells exhibiting structural and electrophysiological properties of both cardiomyocytes and neurons. Human and mouse pluripotent stem cell-derived CMs (PSC-CMs) were transduced with the neurogenic transcription factors Brn2, Ascl1, Myt1l and NeuroD. We found that CMs adopted neuronal morphologies as early as day 3 post-transduction while still retaining a CM gene expression profile. At week 1 post-transduction, we found that reprogrammed CMs expressed neuronal markers such as Tuj1, Map2, and NCAM. At week 3 post-transduction, mature neuronal markers such as vGlut and synapsin were observed. With single-cell qPCR, we temporally examined CM gene expression and observed increased expression of neuronal markers Dcx, Map2, and Tubb3. Patch-clamp analysis confirmed the neuron-like electrophysiological profile of reprogrammed CMs. This study demonstrates that PSC-CMs are amenable to partial neuronal conversion, yielding a population of cells exhibiting features of both neurons and CMs.

    View details for DOI 10.1038/srep44840

    View details for Web of Science ID 000396983300001

    View details for PubMedID 28327614

    View details for PubMedCentralID PMC5361100

  • Optophysiology of cardiomyocytes: characterizing cellular motion with quantitative phase imaging. Biomedical optics express Cordeiro, C. n., Abilez, O. J., Goetz, G. n., Gupta, T. n., Zhuge, Y. n., Solgaard, O. n., Palanker, D. n. 2017; 8 (10): 4652–62

    Abstract

    Quantitative phase imaging enables precise characterization of cellular shape and motion. Variation of cell volume in populations of cardiomyocytes can help distinguish their types, while changes in optical thickness during beating cycle identify contraction and relaxation periods and elucidate cell dynamics. Parameters such as characteristic cycle shape, beating frequency, duration and regularity can be used to classify stem-cell derived cardiomyocytes according to their health and, potentially, cell type. Unlike classical patch-clamp based electrophysiological characterization of cardiomyocytes, this interferometric approach enables rapid and non-destructive analysis of large populations of cells, with longitudinal follow-up, and applications to tissue regeneration, personalized medicine, and drug testing.

    View details for PubMedID 29082092

    View details for PubMedCentralID PMC5654807

  • Anisotropic microfibrous scaffolds enhance the organization and function of cardiomyocytes derived from induced pluripotent stem cells. Biomaterials science Wanjare, M. n., Hou, L. n., Nakayama, K. H., Kim, J. J., Mezak, N. P., Abilez, O. J., Tzatzalos, E. n., Wu, J. C., Huang, N. F. 2017; 5 (8): 1567–78

    Abstract

    Engineering of myocardial tissue constructs is a promising approach for treatment of coronary heart disease. To engineer myocardial tissues that better mimic the highly ordered physiological arrangement and function of native cardiomyocytes, we generated electrospun microfibrous polycaprolactone scaffolds with either randomly oriented (14 μm fiber diameter) or parallel-aligned (7 μm fiber diameter) microfiber arrangement and co-seeded the scaffolds with human induced pluripotent stem cell-derived cardiomyocytes (iCMs) and endothelial cells (iECs) for up to 12 days after iCM seeding. Here we demonstrated that aligned microfibrous scaffolds induced iCM alignment along the direction of the aligned microfibers after 2 days of iCM seeding, as well as promoted greater iCM maturation by increasing the sarcomeric length and gene expression of myosin heavy chain adult isoform (MYH7), in comparison to randomly oriented scaffolds. Furthermore, the benefit of scaffold anisotropy was evident in the significantly higher maximum contraction velocity of iCMs on the aligned scaffolds, compared to randomly oriented scaffolds, at 12 days of culture. Co-seeding of iCMs with iECs led to reduced contractility, compared to when iCMs were seeded alone. These findings demonstrate a dominant role of scaffold anisotropy in engineering cardiovascular tissues that maintain iCM organization and contractile function.

    View details for PubMedID 28715029

    View details for PubMedCentralID PMC5567776

  • iPSC-derived cardiomyocytes reveal abnormal TGF-ß signalling in left ventricular non-compaction cardiomyopathy. Nature cell biology Kodo, K., Ong, S., Jahanbani, F., Termglinchan, V., Hirono, K., Inanloorahatloo, K., Ebert, A. D., Shukla, P., Abilez, O. J., Churko, J. M., Karakikes, I., Jung, G., Ichida, F., Wu, S. M., Snyder, M. P., Bernstein, D., Wu, J. C. 2016; 18 (10): 1031-1042

    Abstract

    Left ventricular non-compaction (LVNC) is the third most prevalent cardiomyopathy in children and its pathogenesis has been associated with the developmental defect of the embryonic myocardium. We show that patient-specific induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) generated from LVNC patients carrying a mutation in the cardiac transcription factor TBX20 recapitulate a key aspect of the pathological phenotype at the single-cell level and this was associated with perturbed transforming growth factor beta (TGF-β) signalling. LVNC iPSC-CMs have decreased proliferative capacity due to abnormal activation of TGF-β signalling. TBX20 regulates the expression of TGF-β signalling modifiers including one known to be a genetic cause of LVNC, PRDM16, and genome editing of PRDM16 caused proliferation defects in iPSC-CMs. Inhibition of TGF-β signalling and genome correction of the TBX20 mutation were sufficient to reverse the disease phenotype. Our study demonstrates that iPSC-CMs are a useful tool for the exploration of pathological mechanisms underlying poorly understood cardiomyopathies including LVNC.

    View details for DOI 10.1038/ncb3411

    View details for PubMedID 27642787

  • Engineered heart tissues and induced pluripotent stem cells: Macro- and microstructures for disease modeling, drug screening, and translational studies. Advanced drug delivery reviews Tzatzalos, E., Abilez, O. J., Shukla, P., Wu, J. C. 2016; 96: 234-244

    Abstract

    Engineered heart tissue has emerged as a personalized platform for drug screening. With the advent of induced pluripotent stem cell (iPSC) technology, patient-specific stem cells can be developed and expanded into an indefinite source of cells. Subsequent developments in cardiovascular biology have led to efficient differentiation of cardiomyocytes, the force-producing cells of the heart. iPSC-derived cardiomyocytes (iPSC-CMs) have provided potentially limitless quantities of well-characterized, healthy, and disease-specific CMs, which in turn has enabled and driven the generation and scale-up of human physiological and disease-relevant engineered heart tissues. The combined technologies of engineered heart tissue and iPSC-CMs are being used to study diseases and to test drugs, and in the process, have advanced the field of cardiovascular tissue engineering into the field of precision medicine. In this review, we will discuss current developments in engineered heart tissue, including iPSC-CMs as a novel cell source. We examine new research directions that have improved the function of engineered heart tissue by using mechanical or electrical conditioning or the incorporation of non-cardiomyocyte stromal cells. Finally, we discuss how engineered heart tissue can evolve into a powerful tool for therapeutic drug testing.

    View details for DOI 10.1016/j.addr.2015.09.010

    View details for PubMedID 26428619

  • CD13 and ROR2 Permit Isolation of Highly Enriched Cardiac Mesoderm from Differentiating Human Embryonic Stem Cells STEM CELL REPORTS Skelton, R. J., Brady, B., Khoja, S., Sahoo, D., Engel, J., Arasaratnam, D., Saleh, K. K., Abilez, O. J., Zhao, P., Stanley, E. G., Elefanty, A. G., Kwon, M., Elliott, D. A., Ardehali, R. 2016; 6 (1): 95-108

    Abstract

    The generation of tissue-specific cell types from human embryonic stem cells (hESCs) is critical for the development of future stem cell-based regenerative therapies. Here, we identify CD13 and ROR2 as cell-surface markers capable of selecting early cardiac mesoderm emerging during hESC differentiation. We demonstrate that the CD13+/ROR2+ population encompasses pre-cardiac mesoderm, which efficiently differentiates to all major cardiovascular lineages. We determined the engraftment potential of CD13+/ROR2+ in small (murine) and large (porcine) animal models, and demonstrated that CD13+/ROR2+ progenitors have the capacity to differentiate toward cardiomyocytes, fibroblasts, smooth muscle, and endothelial cells in vivo. Collectively, our data show that CD13 and ROR2 identify a cardiac lineage precursor pool that is capable of successful engraftment into the porcine heart. These markers represent valuable tools for further dissection of early human cardiac differentiation, and will enable a detailed assessment of human pluripotent stem cell-derived cardiac lineage cells for potential clinical applications.

    View details for DOI 10.1016/j.stemcr.2015.11.006

    View details for Web of Science ID 000368099500011

  • Human Engineered Heart Muscles Engraft and Survive Long Term in a Rodent Myocardial Infarction Model. Circulation research Riegler, J., Tiburcy, M., Ebert, A., Tzatzalos, E., Raaz, U., Abilez, O. J., Shen, Q., Kooreman, N. G., Neofytou, E., Chen, V. C., Wang, M., Meyer, T., Tsao, P. S., Connolly, A. J., Couture, L. A., Gold, J. D., Zimmermann, W. H., Wu, J. C. 2015; 117 (8): 720-730

    Abstract

    Tissue engineering approaches may improve survival and functional benefits from human embryonic stem cell-derived cardiomyocyte transplantation, thereby potentially preventing dilative remodeling and progression to heart failure.Assessment of transport stability, long-term survival, structural organization, functional benefits, and teratoma risk of engineered heart muscle (EHM) in a chronic myocardial infarction model.We constructed EHMs from human embryonic stem cell-derived cardiomyocytes and released them for transatlantic shipping following predefined quality control criteria. Two days of shipment did not lead to adverse effects on cell viability or contractile performance of EHMs (n=3, P=0.83, P=0.87). One month after ischemia/reperfusion injury, EHMs were implanted onto immunocompromised rat hearts to simulate chronic ischemia. Bioluminescence imaging showed stable engraftment with no significant cell loss between week 2 and 12 (n=6, P=0.67), preserving ≤25% of the transplanted cells. Despite high engraftment rates and attenuated disease progression (change in ejection fraction for EHMs, -6.7±1.4% versus control, -10.9±1.5%; n>12; P=0.05), we observed no difference between EHMs containing viable and nonviable human cardiomyocytes in this chronic xenotransplantation model (n>12; P=0.41). Grafted cardiomyocytes showed enhanced sarcomere alignment and increased connexin 43 expression at 220 days after transplantation. No teratomas or tumors were found in any of the animals (n=14) used for long-term monitoring.EHM transplantation led to high engraftment rates, long-term survival, and progressive maturation of human cardiomyocytes. However, cell engraftment was not correlated with functional improvements in this chronic myocardial infarction model. Most importantly, the safety of this approach was demonstrated by the lack of tumor or teratoma formation.

    View details for DOI 10.1161/CIRCRESAHA.115.306985

    View details for PubMedID 26291556

  • Effect of human donor cell source on differentiation and function of cardiac induced pluripotent stem cells. Journal of the American College of Cardiology Sanchez-Freire, V., Lee, A. S., Hu, S., Abilez, O. J., Liang, P., Lan, F., Huber, B. C., Ong, S., Hong, W. X., Huang, M., Wu, J. C. 2014; 64 (5): 436-448

    Abstract

    Human-induced pluripotent stem cells (iPSCs) are a potentially unlimited source for generation of cardiomyocytes (iPSC-CMs). However, current protocols for iPSC-CM derivation face several challenges, including variability in somatic cell sources and inconsistencies in cardiac differentiation efficiency.This study aimed to assess the effect of epigenetic memory on differentiation and function of iPSC-CMs generated from somatic cell sources of cardiac versus noncardiac origins.Cardiac progenitor cells (CPCs) and skin fibroblasts from the same donors were reprogrammed into iPSCs and differentiated into iPSC-CMs via embryoid body and monolayer-based differentiation protocols.Differentiation efficiency was found to be higher in CPC-derived iPSC-CMs (CPC-iPSC-CMs) than in fibroblast-derived iPSC-CMs (Fib-iPSC-CMs). Gene expression analysis during cardiac differentiation demonstrated up-regulation of cardiac transcription factors in CPC-iPSC-CMs, including NKX2-5, MESP1, ISL1, HAND2, MYOCD, MEF2C, and GATA4. Epigenetic assessment revealed higher methylation in the promoter region of NKX2-5 in Fib-iPSC-CMs compared with CPC-iPSC-CMs. Epigenetic differences were found to dissipate with increased cell passaging, and a battery of in vitro assays revealed no significant differences in their morphological and electrophysiological properties at early passage. Finally, cell delivery into a small animal myocardial infarction model indicated that CPC-iPSC-CMs and Fib-iPSC-CMs possess comparable therapeutic capabilities in improving functional recovery in vivo.This is the first study to compare differentiation of iPSC-CMs from human CPCs versus human fibroblasts from the same donors. The authors demonstrate that although epigenetic memory improves differentiation efficiency of cardiac versus noncardiac somatic cell sources in vitro, it does not contribute to improved functional outcome in vivo.

    View details for DOI 10.1016/j.jacc.2014.04.056

    View details for PubMedID 25082575

  • Human pluripotent stem cell tools for cardiac optogenetics Conf Proc IEEE Eng Med Biol Soc Zhuge, Y., Patlolla, B., Ramakrishnan, C., Beygui, R. E., Zarins, C. K., Deisseroth, K., Kuhl, E., Abilez, O. J. 2014: 6171-4
  • Human pluripotent stem cells (hPSCs) for heart regeneration CARDIAC REGENERATION AND REPAIR, VOL 1: PATHOLOGY AND THERAPIES Abilez, O. J., Wu, J. C., Li, R. K., Weisel, R. D. 2014; 71: 297–324
  • Multi-cellular interactions sustain long-term contractility of human pluripotent stem cell-derived cardiomyocytes. American journal of translational research Burridge, P. W., Metzler, S. A., Nakayama, K. H., Abilez, O. J., Simmons, C. S., Bruce, M. A., Matsuura, Y., Kim, P., Wu, J. C., Butte, M., Huang, N. F., Yang, P. C. 2014; 6 (6): 724-735

    Abstract

    Therapeutic delivery of cardiomyocytes derived from human pluripotent stem cells (hPSC-CMs) represents a novel clinical approach to regenerate the injured myocardium. However, poor survival and contractility of these cells are a significant bottleneck to their clinical use. To better understand the role of cell-cell communication in enhancing the phenotype and contractile properties of hPSC-CMs, we developed a three-dimensional (3D) hydrogel composed of hPSC-CMs, human pluripotent stem cell-derived endothelial cells (hPSC-ECs), and/or human amniotic mesenchymal stem cells (hAMSCs). The objective of this study was to examine the role of multi-cellular interactions among hPSC-ECs and hAMSCs on the survival and long-term contractile phenotype of hPSC-CMs in a 3D hydrogel. Quantification of spontaneous contractility of hPSC-CMs in tri-culture demonstrated a 6-fold increase in the area of contractile motion after 6 weeks with characteristic rhythmic contraction frequency, when compared to hPSC-CMs alone (P < 0.05). This finding was supported by a statistically significant increase in cardiac troponin T protein expression in the tri-culture hydrogel construct at 6 weeks, when compared to hPSC-CMs alone (P < 0.001). The sustained hPSC-CM survival and contractility in tri-culture was associated with a significant upregulation in the gene expression of L-type Ca(2+) ion channel, Cav1.2, and the inward-rectifier potassium channel, Kir2.1 (P < 0.05), suggesting a role of ion channels in mediating these processes. These findings demonstrate that multi-cellular interactions modulate hPSC-CM phenotype, function, and survival, and they will have important implications in engineering cardiac tissues for treatment of cardiovascular diseases.

    View details for PubMedID 25628783

  • Optogenetic LED array for perturbing cardiac electrophysiology. Conference proceedings : ... Annual International Conference of the IEEE Engineering in Medicine and Biology Society. IEEE Engineering in Medicine and Biology Society. Conference Abilez, O. J. 2013; 2013: 1619-1622

    Abstract

    Optogenetics is the targeted genetic introduction of light-sensitive channels, such as Channelrhodopsin, and pumps, such as Halorhodopsin, into electrically-excitable cells that enables high spatiotemporal electrical stimulation and inhibition by optical actuation. Technologies for inducing optogenetically-based electrical stimulation for investigating in vitro and in vivo neural perturbations have been described. However, modification of existing technologies or creation of new ones has not been described for chronic cardiac applications. Here, an LED array system for optogenetically perturbing cardiac electrophysiology is described. The overall layout of the system consists of an LED holder containing six LED's that deliver pulsed ∼470 nm light to pluripotent stem cell-derived cardiomyocytes cultured in a 6-well tissue culture plate. The response of the cardiomyocytes is monitored by microscopy and the system is enclosed within a standard incubator. This system is relatively simple to create and uses mostly off-the-shelf components. The overall function of the system is to deliver chronic light stimulation over days to weeks to differentiating stem cell-derived cardiomyocytes in order to investigate perturbations in their electrophysiology.

    View details for DOI 10.1109/EMBC.2013.6609826

    View details for PubMedID 24110013

  • Stem cell isolation: Differential stickiness. Nature materials Abilez, O. J., Wu, J. C. 2013; 12 (6): 474-476

    View details for DOI 10.1038/nmat3664

    View details for PubMedID 23695740

  • Label-free electrophysiological cytometry for stem cell-derived cardiomyocyte clusters LAB ON A CHIP Myers, F. B., Zarins, C. K., Abilez, O. J., Lee, L. P. 2013; 13 (2): 220-228

    Abstract

    Stem cell therapies hold great promise for repairing tissues damaged due to disease or injury. However, a major obstacle facing this field is the difficulty in identifying cells of a desired phenotype from the heterogeneous population that arises during stem cell differentiation. Conventional fluorescence flow cytometry and magnetic cell purification require exogenous labeling of cell surface markers which can interfere with the performance of the cells of interest. Here, we describe a non-genetic, label-free cell cytometry method based on electrophysiological response to stimulus. As many of the cell types relevant for regenerative medicine are electrically-excitable (e.g. cardiomyocytes, neurons, smooth muscle cells), this technology is well-suited for identifying cells from heterogeneous stem cell progeny without the risk and expense associated with molecular labeling or genetic modification. Our label-free cell cytometer is capable of distinguishing clusters of undifferentiated human induced pluripotent stem cells (iPSC) from iPSC-derived cardiomyocyte (iPSC-CM) clusters. The system utilizes a microfluidic device with integrated electrodes for both electrical stimulation and recording of extracellular field potential (FP) signals from suspended cells in flow. The unique electrode configuration provides excellent rejection of field stimulus artifact while enabling sensitive detection of FPs with a noise floor of 2 μV(rms). Cells are self-aligned to the recording electrodes via hydrodynamic flow focusing. Based on automated analysis of these extracellular signals, the system distinguishes cardiomyocytes from non-cardiomyocytes. This is an entirely new approach to cell cytometry, in which a cell's functionality is assessed rather than its expression profile or physical characteristics.

    View details for DOI 10.1039/c2lc40905d

    View details for Web of Science ID 000312219100006

    View details for PubMedID 23207961

    View details for PubMedCentralID PMC3556464

  • Laparoscopic aortic surgery today NEZHAT'S VIDEO-ASSISTED AND ROBOTIC-ASSISTED LAPAROSCOPY AND HYSTEROSCOPY, 4TH EDITION Picquet, J., Abilez, O. J., Cau, J., Zarins, C. K., Goeau-Brissoniere, O., Nezhat, C., Nezhat, F., Nezhat, C. 2013: 581–85
  • Complications in laparoscopy NEZHAT'S VIDEO-ASSISTED AND ROBOTIC-ASSISTED LAPAROSCOPY AND HYSTEROSCOPY, 4TH EDITION Abilez, O. J., Carpenter, J. E., Picquet, J., Zarins, C. K., Nezhat, C., Nezhat, F., Nezhat, C. 2013: 642–62
  • Stretching Skeletal Muscle: Chronic Muscle Lengthening through Sarcomerogenesis PLOS ONE Zoellner, A. M., Abilez, O. J., Boel, M., Kuhl, E. 2012; 7 (10)

    Abstract

    Skeletal muscle responds to passive overstretch through sarcomerogenesis, the creation and serial deposition of new sarcomere units. Sarcomerogenesis is critical to muscle function: It gradually re-positions the muscle back into its optimal operating regime. Animal models of immobilization, limb lengthening, and tendon transfer have provided significant insight into muscle adaptation in vivo. Yet, to date, there is no mathematical model that allows us to predict how skeletal muscle adapts to mechanical stretch in silico. Here we propose a novel mechanistic model for chronic longitudinal muscle growth in response to passive mechanical stretch. We characterize growth through a single scalar-valued internal variable, the serial sarcomere number. Sarcomerogenesis, the evolution of this variable, is driven by the elastic mechanical stretch. To analyze realistic three-dimensional muscle geometries, we embed our model into a nonlinear finite element framework. In a chronic limb lengthening study with a muscle stretch of 1.14, the model predicts an acute sarcomere lengthening from 3.09[Formula: see text]m to 3.51[Formula: see text]m, and a chronic gradual return to the initial sarcomere length within two weeks. Compared to the experiment, the acute model error was 0.00% by design of the model; the chronic model error was 2.13%, which lies within the rage of the experimental standard deviation. Our model explains, from a mechanistic point of view, why gradual multi-step muscle lengthening is less invasive than single-step lengthening. It also explains regional variations in sarcomere length, shorter close to and longer away from the muscle-tendon interface. Once calibrated with a richer data set, our model may help surgeons to prevent muscle overstretch and make informed decisions about optimal stretch increments, stretch timing, and stretch amplitudes. We anticipate our study to open new avenues in orthopedic and reconstructive surgery and enhance treatment for patients with ill proportioned limbs, tendon lengthening, tendon transfer, tendon tear, and chronically retracted muscles.

    View details for DOI 10.1371/journal.pone.0045661

    View details for Web of Science ID 000309388500010

    View details for PubMedID 23049683

    View details for PubMedCentralID PMC3462200

  • Computational optogenetics: A novel continuum framework for the photoelectrochemistry of living systems JOURNAL OF THE MECHANICS AND PHYSICS OF SOLIDS Wong, J., Abilez, O. J., Kuhl, E. 2012; 60 (6): 1158-1178

    Abstract

    Electrical stimulation is currently the gold standard treatment for heart rhythm disorders. However, electrical pacing is associated with technical limitations and unavoidable potential complications. Recent developments now enable the stimulation of mammalian cells with light using a novel technology known as optogenetics. The optical stimulation of genetically engineered cells has significantly changed our understanding of electrically excitable tissues, paving the way towards controlling heart rhythm disorders by means of photostimulation. Controlling these disorders, in turn, restores coordinated force generation to avoid sudden cardiac death. Here, we report a novel continuum framework for the photoelectrochemistry of living systems that allows us to decipher the mechanisms by which this technology regulates the electrical and mechanical function of the heart. Using a modular multiscale approach, we introduce a non-selective cation channel, channelrhodopsin-2, into a conventional cardiac muscle cell model via an additional photocurrent governed by a light-sensitive gating variable. Upon optical stimulation, this channel opens and allows sodium ions to enter the cell, inducing electrical activation. In side-by-side comparisons with conventional heart muscle cells, we show that photostimulation directly increases the sodium concentration, which indirectly decreases the potassium concentration in the cell, while all other characteristics of the cell remain virtually unchanged. We integrate our model cells into a continuum model for excitable tissue using a nonlinear parabolic second order partial differential equation, which we discretize in time using finite differences and in space using finite elements. To illustrate the potential of this computational model, we virtually inject our photosensitive cells into different locations of a human heart, and explore its activation sequences upon photostimulation. Our computational optogenetics tool box allows us to virtually probe landscapes of process parameters, and to identify optimal photostimulation sequences with the goal to pace human hearts with light and, ultimately, to restore mechanical function.

    View details for DOI 10.1016/j.jmps.2012.02.004

    View details for Web of Science ID 000303285600007

    View details for PubMedCentralID PMC3388516

  • Patient-Specific Induced Pluripotent Stem Cells as a Model for Familial Dilated Cardiomyopathy SCIENCE TRANSLATIONAL MEDICINE Sun, N., Yazawa, M., Liu, J., Han, L., Sanchez-Freire, V., Abilez, O. J., Navarrete, E. G., Hu, S., Wang, L., Lee, A., Pavlovic, A., Lin, S., Chen, R., Hajjar, R. J., Snyder, M. P., Dolmetsch, R. E., Butte, M. J., Ashley, E. A., Longaker, M. T., Robbins, R. C., Wu, J. C. 2012; 4 (130)

    Abstract

    Characterized by ventricular dilatation, systolic dysfunction, and progressive heart failure, dilated cardiomyopathy (DCM) is the most common form of cardiomyopathy in patients. DCM is the most common diagnosis leading to heart transplantation and places a significant burden on healthcare worldwide. The advent of induced pluripotent stem cells (iPSCs) offers an exceptional opportunity for creating disease-specific cellular models, investigating underlying mechanisms, and optimizing therapy. Here, we generated cardiomyocytes from iPSCs derived from patients in a DCM family carrying a point mutation (R173W) in the gene encoding sarcomeric protein cardiac troponin T. Compared to control healthy individuals in the same family cohort, cardiomyocytes derived from iPSCs from DCM patients exhibited altered regulation of calcium ion (Ca(2+)), decreased contractility, and abnormal distribution of sarcomeric α-actinin. When stimulated with a β-adrenergic agonist, DCM iPSC-derived cardiomyocytes showed characteristics of cellular stress such as reduced beating rates, compromised contraction, and a greater number of cells with abnormal sarcomeric α-actinin distribution. Treatment with β-adrenergic blockers or overexpression of sarcoplasmic reticulum Ca(2+) adenosine triphosphatase (Serca2a) improved the function of iPSC-derived cardiomyocytes from DCM patients. Thus, iPSC-derived cardiomyocytes from DCM patients recapitulate to some extent the morphological and functional phenotypes of DCM and may serve as a useful platform for exploring disease mechanisms and for drug screening.

    View details for DOI 10.1126/scitranslmed.3003552

    View details for Web of Science ID 000303045900004

    View details for PubMedID 22517884

    View details for PubMedCentralID PMC3657516

  • Pressure-related lateral displacement of anastomosed arteries and prosthetic grafts in an in vitro model: Implications for neointimal hyperplasia formation Assar, A. N., Abilez, O. J., Xu, C., Zarins, C. K. WILEY-BLACKWELL. 2012: 46
  • Cardiac Optogenetics 34th Annual International Conference of the IEEE Engineering-in-Medicine-and-Biology-Society (EMBS) Abilez, O. J. IEEE. 2012: 1386–1389

    Abstract

    For therapies based on human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CM) to be effective, arrhythmias must be avoided. Towards achieving this goal, light-activated channelrhodopsin-2 (ChR2), a cation channel activated with 480 nm light, and a first generation halorhodopsin (NpHR1.0), an anion pump activated by 580 nm light, have been introduced into hiPSC. By using in vitro approaches, hiPSC-CM are able to be optogenetically activated and inhibited. ChR2 and NpHR1.0 are stably transduced into undifferentiated hiPSC via a lentiviral vector. Via directed differentiation, both wildtype hiPSC-CM (hiPSC(WT)-CM) and hiPSC(ChR2/NpHR)-CM are produced and subjected to both electrical and optical stimulation. Both hiPSC(WT)-CM and hiPSC(ChR2/NpHR)-CM respond to traditional electrical stimulation and produce similar contractility features but only hiPSC(ChR2/NpHR)-CM can be synchronized and inhibited by optical stimulation. Here it is shown that light sensitive proteins can enable in vitro optical control of hiPSC-CM. For future therapy, in vivo optical stimulation could allow precise and specific synchronization of implanted hiPSC-CM with patient cardiac rates and rhythms.

    View details for Web of Science ID 000313296501161

    View details for PubMedID 23366158

  • COMPUTATIONAL MODELLING OF OPTOGENETICS IN CARDIAC CELLS ASME Summer Bioengineering Conference (SBC) Wong, J., Abilez, O., Kuhl, E. AMER SOC MECHANICAL ENGINEERS. 2012: 355–356
  • IN VITRO/IN SILICO CHARACTERIZATION OF ACTIVE AND PASSIVE STRESSES IN CARDIAC MUSCLE INTERNATIONAL JOURNAL FOR MULTISCALE COMPUTATIONAL ENGINEERING Boel, M., Abilez, O. J., Assar, A. N., Zarins, C. K., Kuhl, E. 2012; 10 (2): 171-188
  • Computational modeling of growth: systemic and pulmonary hypertension in the heart BIOMECHANICS AND MODELING IN MECHANOBIOLOGY Rausch, M. K., Dam, A., Goktepe, S., Abilez, O. J., Kuhl, E. 2011; 10 (6): 799-811

    Abstract

    We introduce a novel constitutive model for growing soft biological tissue and study its performance in two characteristic cases of mechanically induced wall thickening of the heart. We adopt the concept of an incompatible growth configuration introducing the multiplicative decomposition of the deformation gradient into an elastic and a growth part. The key feature of the model is the definition of the evolution equation for the growth tensor which we motivate by pressure-overload-induced sarcomerogenesis. In response to the deposition of sarcomere units on the molecular level, the individual heart muscle cells increase in diameter, and the wall of the heart becomes progressively thicker. We present the underlying constitutive equations and their algorithmic implementation within an implicit nonlinear finite element framework. To demonstrate the features of the proposed approach, we study two classical growth phenomena in the heart: left and right ventricular wall thickening in response to systemic and pulmonary hypertension.

    View details for DOI 10.1007/s10237-010-0275-x

    View details for Web of Science ID 000296634000001

    View details for PubMedID 21188611

    View details for PubMedCentralID PMC3235798

  • Identification of Cardiovascular Progenitors From Human Embryonic Stem Cells Scientific Sessions of the American-Heart-Association/Resuscitation Science Symposium Ardehali, R., Ali, S., Drukker, M., Abilez, O., Blauwkamp, T., Nusse, R., Weissman, I. LIPPINCOTT WILLIAMS & WILKINS. 2011
  • Vascular anastomosis using controlled phase transitions in poloxamer gels NATURE MEDICINE Chang, E. I., Galvez, M. G., Glotzbach, J. P., Hamou, C. D., El-Ftesi, S., Rappleye, C. T., Sommer, K., Rajadas, J., Abilez, O. J., Fuller, G. G., Longaker, M. T., Gurtner, G. C. 2011; 17 (9): 1147-U160

    Abstract

    Vascular anastomosis is the cornerstone of vascular, cardiovascular and transplant surgery. Most anastomoses are performed with sutures, which are technically challenging and can lead to failure from intimal hyperplasia and foreign body reaction. Numerous alternatives to sutures have been proposed, but none has proven superior, particularly in small or atherosclerotic vessels. We have developed a new method of sutureless and atraumatic vascular anastomosis that uses US Food and Drug Administration (FDA)-approved thermoreversible tri-block polymers to temporarily maintain an open lumen for precise approximation with commercially available glues. We performed end-to-end anastomoses five times more rapidly than we performed hand-sewn controls, and vessels that were too small (<1.0 mm) to sew were successfully reconstructed with this sutureless approach. Imaging of reconstructed rat aorta confirmed equivalent patency, flow and burst strength, and histological analysis demonstrated decreased inflammation and fibrosis at up to 2 years after the procedure. This new technology has potential for improving efficiency and outcomes in the surgical treatment of cardiovascular disease.

    View details for DOI 10.1038/nm.2424

    View details for Web of Science ID 000294605100038

    View details for PubMedID 21873986

  • Stretchable microelectrode array using room-temperature liquid alloy interconnects JOURNAL OF MICROMECHANICS AND MICROENGINEERING Wei, P., Taylor, R., Ding, Z., Chung, C., Abilez, O. J., Higgs, G., Pruitt, B. L., Ziaie, B. 2011; 21 (5)
  • Stimulation and Artifact-Free Extracellular Electrophysiological Recording of Cells in Suspension 33rd Annual International Conference of the IEEE Engineering-in-Medicine-and-Biology-Society (EMBS) Myers, F. B., Abilez, O. J., Zarins, C. K., Lee, L. P. IEEE. 2011: 4030–4033

    Abstract

    We have developed instrumentation which stimulates and records electrophysiological signals from populations of suspended cells in microfluidic channels. We are employing this instrumentation in a new approach to cell sorting and flow cytometry which distinguishes cells based on their electrophysiology. This label-free approach is ideal for applications where labeling or genetic modification of cells is undesirable, such as in purifying cells for tissue replacement therapies. Electrophysiology is a powerful indicator of phenotype for electrically-excitable cells such as myocytes and neurons. However, extracellular field potential signals are notoriously weak and large stimulus artifacts can easily obscure these signals if care is not taken to suppress them. This is particularly true for suspended cells. Here, we describe a novel microelectrode configuration and the associated instrumentation for suppressing stimulus artifacts and faithfully recovering the extracellular field potential signal. We show that the device is capable of distinguishing cardiomyocytes from non-cardiomyocytes derived from the same stem cell population. Finally, we explain the relationship between extracellular field potentials and the more familiar transmembrane action potential signal, noting the physiologically important features of these signals.

    View details for Web of Science ID 000298810003093

    View details for PubMedID 22255224

  • Localized control of exsanguinating arterial hemorrhage: an experimental model. Polski przeglad chirurgiczny Haick, M. B., Abilez, O. J., Johnson, B. L., Xu, C., Taylor, C. A., Rich, N. M., Zarins, C. K. 2011; 83 (1): 1-9

    Abstract

    To develop an arterial injury model for testing hemostatic devices at well-defined high and low bleeding rates.A side-hole arterial injury was created in the carotid artery of sheep. Shed blood was collected in a jugular venous reservoir and bleeding rate at the site of arterial injury was controlled by regulating outflow resistance from the venous reservoir. Two models were studied: uncontrolled exsanguinating hemorrhage and bleeding at controlled rates with blood return to maintain hemodynamic stability. Transcutaneous Duplex ultrasound was used to characterize ultrasound signatures at various bleeding rates.A 2.5 mm arterial side-hole resulted in exsanguinating hemorrhage with an initial bleeding rate of 400 ml/min which, without resuscitation, decreased to below 100 ml/min in 5 minutes. After 17 minutes, bleeding from the injury site stopped and the animal had lost 60% of total blood volume. Reinfusion of shed blood maintained normal hemodynamics and both high and low bleeding rates could be maintained without hemorrhagic shock. Bleeding rate at the arterial injury site was held at 395±78 ml/min for 8 minutes, 110±11 ml/min for 15 minutes, and 12±1 ml/min for 12 minutes. Doppler flow signatures at the site of injury were characterized by high peak and end-diastolic flow velocities at the bleeding site which varied with the rate of hemorrhage.We have developed a hemodynamically stable model of acute arterial injury which can be used to evaluate diagnostic and treatment methods focused on control of the arterial injury site.

    View details for DOI 10.2478/v10035-011-0001-0

    View details for PubMedID 22166236

  • A matrix micropatterning platform for cell localization and stem cell fate determination ACTA BIOMATERIALIA Huang, N. F., Patlolla, B., Abilez, O., Sharma, H., Rajadas, J., Beygui, R. E., Zarins, C. K., Cooke, J. P. 2010; 6 (12): 4614-4621

    Abstract

    To study the role of cell-extracellular matrix (ECM) interactions, microscale approaches provide the potential to perform high throughput assessment of the effect of the ECM microenvironment on cellular function and phenotype. Using a microscale direct writing (MDW) technique, we characterized the generation of multicomponent ECM microarrays for cellular micropatterning, localization and stem cell fate determination. ECMs and other biomolecules of various geometries and sizes were printed onto epoxide-modified glass substrates to evaluate cell attachment by human endothelial cells. The endothelial cells displayed strong preferential attachment to the ECM patterned regions and aligned their cytoskeleton along the direction of the micropatterns. We next generated ECM microarrays that contained one or more ECM components (namely gelatin, collagen IV and fibronectin) and then cultured murine embryonic stem cell (ESCs) on the microarrays. The ESCs selectively attached to the micropatterned features and expressed markers associated with a pluripotent phenotype, such as E-cadherin and alkaline phosphatase, when maintained in growth medium containing leukemia inhibitory factor. In the presence of the soluble factors retinoic acid and bone morphogenetic protein-4 the ESCs differentiated towards the ectodermal lineage on the ECM microarray with differential ECM effects. The ESCs cultured on gelatin showed significantly higher levels of pan cytokeratin expression, when compared with cells cultured on collagen IV or fibronectin, suggesting that gelatin preferentially promotes ectodermal differentiation. In summary, our results demonstrate that MDW is a versatile approach to print ECMs of diverse geometries and compositions onto surfaces, and it is amenable to the generation of multicomponent ECM microarrays for stem cell fate determination.

    View details for DOI 10.1016/j.actbio.2010.06.033

    View details for Web of Science ID 000284385300018

    View details for PubMedID 20601236

    View details for PubMedCentralID PMC2957527

  • Power Law as a Method for Ultrasound Detection of Internal Bleeding: In Vivo Rabbit Validation IEEE TRANSACTIONS ON BIOMEDICAL ENGINEERING Wang, A. S., Abilez, O. J., Zarins, C. K., Taylor, C. A., Liang, D. H. 2010; 57 (12): 2870-2875

    Abstract

    New detection methods for vascular injuries can augment the usability of an ultrasound (US) imager in trauma settings. The goal of this study was to evaluate a potential-detection strategy for internal bleeding that employs a well-established theoretical biofluid model, the power law. This law characterizes normal blood-flow rates through an arterial tree by its bifurcation geometry. By detecting flows that deviate from the model, we hypothesized that vascular abnormalities could be localized. We devised a bleed metric, flow-split deviation (FSD), that quantified the difference between patient and model blood flows at vessel bifurcations. Femoral bleeds were introduced into ten rabbits (∼5 kg) using a cannula attached to a variable pump. Different bleed rates (0% as control, 5%, 10%, 15%, 20%, 25%, and 30% of descending aortic flow) were created at two physiological states (rest and elevated state with epinephrine). FSDs were found by US imaging the iliac arteries. Our bleed metric demonstrated good sensitivity and specificity at moderate bleed rates; area under receiver-operating characteristic curves were greater than 0.95 for bleed rates 20% and higher. Thus, FSD was a good indicator of bleed severity and may serve as an additional tool in the US bleed detection.

    View details for DOI 10.1109/TBME.2010.2058803

    View details for Web of Science ID 000284360100009

    View details for PubMedID 20639172

  • Dynamic MicroRNA Expression Programs During Cardiac Differentiation of Human Embryonic Stem Cells Role for miR-499 CIRCULATION-CARDIOVASCULAR GENETICS Wilson, K. D., Hu, S., Venkatasubrahmanyam, S., Fu, J., Sun, N., Abilez, O. J., Baugh, J. J., Jia, F., Ghosh, Z., Li, R. A., Butte, A. J., Wu, J. C. 2010; 3 (5): 426-U97

    Abstract

    MicroRNAs (miRNAs) are a newly discovered endogenous class of small, noncoding RNAs that play important posttranscriptional regulatory roles by targeting messenger RNAs for cleavage or translational repression. Human embryonic stem cells are known to express miRNAs that are often undetectable in adult organs, and a growing body of evidence has implicated miRNAs as important arbiters of heart development and disease.To better understand the transition between the human embryonic and cardiac "miRNA-omes," we report here the first miRNA profiling study of cardiomyocytes derived from human embryonic stem cells. Analyzing 711 unique miRNAs, we have identified several interesting miRNAs, including miR-1, -133, and -208, that have been previously reported to be involved in cardiac development and disease and that show surprising patterns of expression across our samples. We also identified novel miRNAs, such as miR-499, that are strongly associated with cardiac differentiation and that share many predicted targets with miR-208. Overexpression of miR-499 and -1 resulted in upregulation of important cardiac myosin heavy-chain genes in embryoid bodies; miR-499 overexpression also caused upregulation of the cardiac transcription factor MEF2C.Taken together, our data give significant insight into the regulatory networks that govern human embryonic stem cell differentiation and highlight the ability of miRNAs to perturb, and even control, the genes that are involved in cardiac specification of human embryonic stem cells.

    View details for DOI 10.1161/CIRCGENETICS.109.934281

    View details for Web of Science ID 000283163100006

    View details for PubMedID 20733065

    View details for PubMedCentralID PMC3057038

  • A generic approach towards finite growth with examples of athlete's heart, cardiac dilation, and cardiac wall thickening JOURNAL OF THE MECHANICS AND PHYSICS OF SOLIDS Goktepe, S., Abilez, O. J., Kuhl, E. 2010; 58 (10): 1661-1680
  • A multiscale model for eccentric and concentric cardiac growth through sarcomerogenesis JOURNAL OF THEORETICAL BIOLOGY Goktepe, S., Abilez, O. J., Parker, K. K., Kuhl, E. 2010; 265 (3): 433-442

    Abstract

    We present a novel computational model for maladaptive cardiac growth in which kinematic changes of the cardiac chambers are attributed to alterations in cytoskeletal architecture and in cellular morphology. We adopt the concept of finite volume growth characterized through the multiplicative decomposition of the deformation gradient into an elastic part and a growth part. The functional form of its growth tensor is correlated to sarcomerogenesis, the creation and deposition of new sarcomere units. In response to chronic volume-overload, an increased diastolic wall strain leads to the addition of sarcomeres in series, resulting in a relative increase in cardiomyocyte length, associated with eccentric hypertrophy and ventricular dilation. In response to chronic pressure-overload, an increased systolic wall stress leads to the addition of sacromeres in parallel, resulting in a relative increase in myocyte cross sectional area, associated with concentric hypertrophy and ventricular wall thickening. The continuum equations for both forms of maladaptive growth are discretized in space using a nonlinear finite element approach, and discretized in time using the implicit Euler backward scheme. We explore a generic bi-ventricular heart model in response to volume- and pressure-overload to demonstrate how local changes in cellular morphology translate into global alterations in cardiac form and function.

    View details for DOI 10.1016/j.jtbi.2010.04.023

    View details for Web of Science ID 000280374100023

    View details for PubMedID 20447409

  • IN VITRO ASSESSMENT OF RAT HEART FORCE GENERATION: A QUANTITATIVE APPROACH FOR PREDICTING OUTCOMES FROM PLURIPOTENT STEM CELL-DERIVED THERAPY FOR MYOCARDIAL INFARCTION 12th ASME Summer Bioengineering Conference Guillou, L., Abilez, O. J., Baugh, J., Billakanti, G., Zarins, C. K., Kuhl, E. AMER SOC MECHANICAL ENGINEERS. 2010: 717–718
  • Outcome of open versus endovascular revascularization for chronic mesenteric ischemia: review of comparative studies JOURNAL OF CARDIOVASCULAR SURGERY Assar, A. N., Abilez, O. J., Zarins, C. K. 2009; 50 (4): 509-514

    Abstract

    Chronic mesenteric ischemia is a rare disorder that has traditionally been treated with open surgical revascularization (OR). Endovascular revascularization (ER) has recently gained popularity as an alternative modality of treatment; however, OR is still predominantly used. This study aimed at comparing the outcomes of these two treatment modalities. The literature was searched using the MEDLINE database through the PubMed search engine for relevant articles that compared the outcomes after OR and ER for chronic mesenteric ischemia. Review of the selected articles revealed that patients had lower postoperative mortality and morbidity, and shorter intensive care unit and hospital stay after ER. However, early and long-term symptomatic relief and significantly lower restenosis rate were characteristic of OR. Although no level 1 evidence governs the treatment of chronic mesenteric ischemia, the durability and efficacy of OR is such that this modality should remain the procedure of choice for patients who are fit or whose fitness could be improved before surgery. For unfit patients, or those with short life expectancy, ER is preferable owing to its minimally invasive nature and reduced postoperative mortality and morbidity. Randomized controlled studies are needed to compare the long-term durability and efficacy of ER to those of OR.

    View details for Web of Science ID 000271955000010

    View details for PubMedID 19455085

  • First report of an ilio-popliteal bypass through the greater sciatic foramen - Case report JOURNAL OF CARDIOVASCULAR SURGERY Picquet, J., Thouveny, F., Abilez, O., Pegis, J. D., Blin, V., Enon, B. 2008; 49 (3): 341-343

    Abstract

    A 47 year-old man, who had a history of pelvic radiotherapy for the treatment of testicular tumour 30 years ago, was referred with minor tissue loss of the right lower extremity, grade III, category 5 of the Rutherford classification. His groin region presented with severe radiation damage. Arteriography demonstrated the occlusion of external iliac and femoral arteries. Revascula-risation was performed in the lateral decubitus position, with a ringed polytetrafluoroethylene (PTFE) graft bypass between normal common iliac and popliteal arteries through the greater sciatic foramen. Quick healing was observed. Patient is well 6 months postoperatively. Immediate and 6 month postoperative imaging demonstrated the good patency of the graft. A duplex ultrasound performed 6 month postoperatively showed no significant compression while the patient was in the sitting position.

    View details for Web of Science ID 000257038800005

    View details for PubMedID 18446119

  • Lateral movement of endografts within the aneurysm sac is an indicator of stent-graft instability 21st International Congress on Endovascular Interventions Rafii, B. Y., Abilez, O. J., Benharash, P., Zarins, C. K. ALLIANCE COMMUNICATIONS GROUP DIVISION ALLEN PRESS. 2008: 335–43

    Abstract

    To determine if lateral movement of an aortic endograft 1 year following endovascular abdominal aortic aneurysm (AAA) repair is an indicator of endograft instability and can serve as a predictor of late adverse events.The records of 60 high-risk AAA patients (52 men, 8 women; mean age 74 years) who were treated with infrarenal (n = 38) or suprarenal (n = 22) endografts and had serial computed tomograms (CT) over > or =12 months were analyzed. Postimplantation and 1-year CT scans were compared, and changes in endograft position within the aneurysm sac [lateral movement (LM) versus no lateral movement (NM)] were measured using a vertebral body reference point. Longitudinal endograft movement was measured with respect to the superior mesenteric artery along the aortic centerline axis. Long-term adverse event rates (endoleaks, secondary procedures, conversion, rupture, and death) were assessed.One year after endograft implantation, LM > or =5 mm was present in 16 (27%) patients; 44 (73%) endografts demonstrated no lateral movement. LM patients had larger aneurysms (6.5+/-1.5 versus 5.6+/-0.9 cm, p = 0.02) and a longer endograft-to-hypogastric artery length (p = 0.01) than NM patients. There were no significant differences between patients treated with infrarenal and suprarenal endografts. At 1 year, longitudinal migration > or =10 mm occurred in 5 (31%) of the LM patients versus 2 (5%) in the NM cohort (p<0.0001). There were no significant differences in adverse event rates between LM and NM at 1 year. However, during long-term follow-up (mean 54+/-26 months, range 12-102), 8 (50%) LM patients developed a type I endoleak versus 8 (18%) NM patients (p = 0.02), and 12 (75%) LM patients required a secondary procedure versus 9 (20%) NM patients (p = 0.0002). One (6%) LM patient experienced aneurysm rupture and 2 (13%) other LM patients underwent conversion to open repair.Lateral endograft movement within the aneurysm sac at 1 year is associated with increased risk of late adverse events and was at least as good a predictor of these complications as was longitudinal migration.

    View details for Web of Science ID 000257093100012

    View details for PubMedID 18540708

  • COMPLICATIONS IN LAPAROSCOPY Section 20.1. Major Vascular Injury NEZHAT'S OPERATIVE GYNECOLOGIC LAPAROSCOPY AND HYSTEROSCOPY, 3RD EDITION Abilez, O. J., Picquet, J., Zarins, C. K., Nezhat, C., Nezhat, F., Nezhat, C. 2008: 520–26
  • LAPAROSCOPIC VASCULAR SURGERY IN 2007 NEZHAT'S OPERATIVE GYNECOLOGIC LAPAROSCOPY AND HYSTEROSCOPY, 3RD EDITION Picquet, J., Abilez, O. J., Cau, J., Goeau-Brissonniere, O., Zarins, C. K., Nezhat, C., Nezhat, F., Nezhat, C. 2008: 516–19
  • In vivo imaging and evaluation of different biomatrices for improvement of stem cell survival JOURNAL OF TISSUE ENGINEERING AND REGENERATIVE MEDICINE Cao, F., Rafie, A. H., Abilez, O. J., Wang, H., Blundo, J. T., Pruitt, B., Zarins, C., Wu, J. C. 2007; 1 (6): 465-468

    Abstract

    Therapeutic effects from injection of stem cells are often hampered by acute donor cell death as well as migration away from damaged areas. This is likely due to the fact that injected cells do not have the physical and biochemical cues for ordered engrafment. Here we evaluate 3 common biomatrices (Matrigel, Collagen I, Purmatrix) that has the potential of providing suitable scaffolds needed to enhance stem cell survival. The longitudinal fate of transplanted stem cells was monitored by reporter imaging techniques.

    View details for DOI 10.1002/term.55

    View details for Web of Science ID 000256520300008

    View details for PubMedID 18163533

    View details for PubMedCentralID PMC3657504

  • Iliac fixation inhibits migration of both suprarenal and infrarenal aortic endografts 60th Annual Meeting of the Society-for-Vascular-Surgery Benharash, P., Lee, J. T., Abilez, O. J., Crabtree, T., Bloch, D. A., Zarins, C. K. MOSBY-ELSEVIER. 2007: 250–57

    Abstract

    To evaluate the role of iliac fixation in preventing migration of suprarenal and infrarenal aortic endografts.Quantitative image analysis was performed in 92 patients with infrarenal aortic aneurysms (76 men and 16 women) treated with suprarenal (n = 36) or infrarenal (n = 56) aortic endografts from 2000 to 2004. The longitudinal centerline distance from the superior mesenteric artery to the top of the stent graft was measured on preoperative, postimplantation, and 1-year three-dimensional computed tomographic scans, with movement more than 5 mm considered to be significant. Aortic diameters were measured perpendicular to the centerline axis. Proximal and distal fixation lengths were defined as the lengths of stent-graft apposition to the aortic neck and the common iliac arteries, respectively.There were no significant differences in age, comorbidities, or preoperative aneurysm size (suprarenal, 6.0 cm; infrarenal, 5.7 cm) between the suprarenal and infrarenal groups. However, the suprarenal group had less favorable aortic necks with a shorter length (13 vs 25 mm; P < .0001), a larger diameter (27 vs 24 mm; P < .0001), and greater angulation (19 degrees vs 11 degrees ; P = .007) compared with the infrarenal group. The proximal aortic fixation length was greater in the suprarenal than in the infrarenal group (22 vs 16 mm; P < .0001), with the top of the device closer to the superior mesenteric artery (8 vs 21 mm; P < .0001) as a result of the 15-mm uncovered suprarenal stent. There was no difference in iliac fixation length between the suprarenal and infrarenal groups (26 vs 25 mm; P = .8). Longitudinal centerline stent graft movement at 1 year was similar in the suprarenal and infrarenal groups (4.3 +/- 4.4 mm vs 4.8 +/- 4.3 mm; P = .6). Patients with longitudinal centerline movement of more than 5 mm at 1 year or clinical evidence of migration at any time during the follow-up period comprised the respective migrator groups. Suprarenal migrators had a shorter iliac fixation length (17 vs 29 mm; P = .006) and a similar aortic fixation length (23 vs 22 mm; P > .999) compared with suprarenal nonmigrators. Infrarenal migrators had a shorter iliac fixation length (18 vs 30 mm; P < .0001) and a similar aortic fixation length (14 vs 17 mm; P = .1) compared with infrarenal nonmigrators. Nonmigrators had closer device proximity to the hypogastric arteries in both the suprarenal (7 vs 17 mm; P = .009) and infrarenal (8 vs 24 mm; P < .0001) groups. No migration occurred in either group in patients with good iliac fixation. Multivariate logistic regression analysis revealed that iliac fixation, as evidenced by iliac fixation length (P = .004) and the device to hypogastric artery distance (P = .002), was a significant independent predictor of migration, whereas suprarenal or infrarenal treatment was not a significant predictor of migration. During a clinical follow-up period of 45 +/- 22 months (range, 12-70 months), there have been no aneurysm ruptures, abdominal aortic aneurysm-related deaths, or surgical conversions in either group.Distal iliac fixation is important in preventing migration of both suprarenal and infrarenal aortic endografts that have longitudinal columnar support. Secure iliac fixation minimizes the risk of migration despite suboptimal proximal aortic neck anatomy. Extension of both iliac limbs to cover the entire common iliac artery to the iliac bifurcation seems to prevent endograft migration.

    View details for DOI 10.1016/j.jvs.2006.09.061

    View details for PubMedID 17263997

  • Biomems platform for electromechanical stimulation of cell culture ASME Summer Bioengineering Conference Blundo, J., Chua, G., Abilez, O., Park, Y., Rastegar, A., Cao, F., Zairins, C., Wu, J., Pruitt, B. AMER SOC MECHANICAL ENGINEERS. 2007: 63–64
  • Pulsatile pressure system for cellular mechanical stimulation ASME Summer Bioengineering Conference Taylor, R., Abilez, O., Cao, F., Wu, J., Xu, C., Zarins, C., Pruitt, B. AMER SOC MECHANICAL ENGINEERS. 2007: 1009–1010
  • P19 progenitor cells progress to organized contracting myocytes after chemical and electrical stimulation: Implications for vascular tissue engineering JOURNAL OF ENDOVASCULAR THERAPY Abilez, O., Benharash, P., Miyamoto, E., Gale, A., Xu, C., Zarins, C. K. 2006; 13 (3): 377-388

    Abstract

    To test the hypothesis that a level of chemical and electrical stimulation exists that allows differentiation of progenitor cells into organized contracting myocytes.A custom-made bioreactor with the capability of delivering electrical pulses of varying field strengths, widths, and frequencies was constructed. Individual chambers of the bioreactor allowed continuous electrical stimulation of cultured cells under microscopic observation. On day 0, 1% dimethylsulfoxide (DMSO), known to differentiate cells into myocytes, was added to P19 progenitor cells. Additionally, for the next 22 days, electrical pulses of varying field strengths (0-3 V/cm), widths (2-40 ms), and frequencies (10-25 Hz) were continuously applied. On day 5, the medium containing DMSO was exchanged with regular medium, and the electrical stimulation was continued. From days 6-22, the cells were visually assessed for signs of viability, contractility, and organization.P19 cells remained viable with pulsed electrical fields <3 V/cm, pulse widths <40 ms, and pulse frequencies from 10 to 25 Hz. On day 12, the first spontaneous contractions were observed. For individual colonies, local synchronization and organization occurred; multiple colonies were synchronized with externally applied electrical fields.P19 progenitor cells progress to organized contracting myocytes after chemical and electrical stimulation. Incorporation of such cells into existing methods of producing endothelial cells, fibroblasts, and scaffolds may allow production of improved tissue-engineered vascular grafts.

    View details for Web of Science ID 000238334300015

    View details for PubMedID 16784327

  • A novel culture system shows that stem cells can be grown in 3D and under physiologic pulsatile conditions for tissue engineering of vascular grafts 39th Annual Meeting of the Association-for-Academic-Surgery Abilez, O., Benharash, P., Mehrotra, M., Miyamoto, E., Gale, A., Picquet, J., Xu, C. P., Zarins, C. ACADEMIC PRESS INC ELSEVIER SCIENCE. 2006: 170–78

    Abstract

    Currently available vascular grafts have been limited by variable patency rates, material availability, and immunological rejection. The creation of a tissue-engineered vascular graft (TEVG) from autologous stem cells would potentially overcome these limitations. As a first step in creating a completely autologous TEVG, our objective was to develop a novel system for culturing undifferentiated mouse embryonic stem cells (mESC) in a three-dimensional (3D) configuration and under physiological pulsatile flow and pressure conditions.A bioreactor was created to provide pulsatile conditions to a specially modified four-well Labtek Chamber-Slide culture system. Undifferentiated mESC were either suspended in a 3D Matrigel matrix or suspended only in cell-culture media within the culture system. Pulsatile conditions were applied to the suspended cells and visualized by video microscopy.Undifferentiated mESC were successfully embedded in a 3D Matrigel matrix and could withstand physiological pulsatile conditions. Video microscopy demonstrated that the mESC in the 3D matrix were constrained to the wells of the culture system, moved in unison with the applied flows, and were not washed downstream; this was in contrast to the mESC suspended in media alone.Undifferentiated mESC can be grown in 3D and under pulsatile conditions. We will use these results to study the effects of long-term pulsatile conditions on the differentiation of mESC into endothelial cells, smooth muscle cells, and fibroblast cells with the long-term goal of creating a completely autologous TEVG.

    View details for DOI 10.1016/j.jss.2006.2.017

    View details for Web of Science ID 000237337000005

    View details for PubMedID 16542683

  • Superficial femoral artery transposition repair for isolated superior mesenteric artery dissection JOURNAL OF VASCULAR SURGERY Picquet, J., Abilez, O., Penard, J., Jousset, Y., Rousselet, M. C., Enon, B. 2005; 42 (4): 788-791

    Abstract

    Isolated dissection of the superior mesenteric artery is an uncommon event, but many new cases have been reported recently, reflecting the progress of imaging and suggesting that this pathology is not as rare as previously thought. Here we report a case of superior mesenteric artery dissection where we performed, after failure of conservative medical management, an original surgical technique for mesenteric revascularization using a superficial femoral artery transposition. To the best of our knowledge, this is the first report of the use of this technique for complex mesenteric revascularization.

    View details for DOI 10.1016/j.jvs.2005.05.048

    View details for Web of Science ID 000232609300033

    View details for PubMedID 16242570

  • Adaptative media remodeling of the uterine artery during pregnancy. Abilez, O., Alsac, J., Heikkinen, M., Nezhat, C. H., Zarin, C., Nezhat, C. ELSEVIER SCIENCE INC. 2005: S399
  • Bone marrow-derived dendritic cells from CD36 null mice are less efficient than those derived from wild-type mice in the phagocytosis of apoptotic cells Abilez, O. J., Berger, D. H., Febbraio, M., Silverstein, R. L. AMER SOC CELL BIOLOGY. 1999: 189A