76 results on '"Oscar J. Abilez"'
Search Results
2. Blood vessels in a dish: the evolution, challenges, and potential of vascularized tissues and organoids
- Author
-
Peter N. Nwokoye and Oscar J. Abilez
- Subjects
vascularized tissues ,pluripotent stem cells ,3D bioprinting ,microfluidics ,bioinks ,vascular pathology ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Vascular pathologies are prevalent in a broad spectrum of diseases, necessitating a deeper understanding of vascular biology, particularly in overcoming the oxygen and nutrient diffusion limit in tissue constructs. The evolution of vascularized tissues signifies a convergence of multiple scientific disciplines, encompassing the differentiation of human pluripotent stem cells (hPSCs) into vascular cells, the development of advanced three-dimensional (3D) bioprinting techniques, and the refinement of bioinks. These technologies are instrumental in creating intricate vascular networks essential for tissue viability, especially in thick, complex constructs. This review provides broad perspectives on the past, current state, and advancements in key areas, including the differentiation of hPSCs into specific vascular lineages, the potential and challenges of 3D bioprinting methods, and the role of innovative bioinks mimicking the native extracellular matrix. We also explore the integration of biophysical cues in vascularized tissues in vitro, highlighting their importance in stimulating vessel maturation and functionality. In this review, we aim to synthesize these diverse yet interconnected domains, offering a broad, multidisciplinary perspective on tissue vascularization. Advancements in this field will help address the global organ shortage and transform patient care.
- Published
- 2024
- Full Text
- View/download PDF
3. CD13 and ROR2 Permit Isolation of Highly Enriched Cardiac Mesoderm from Differentiating Human Embryonic Stem Cells
- Author
-
Rhys J.P. Skelton, Bevin Brady, Suhail Khoja, Debashis Sahoo, James Engel, Deevina Arasaratnam, Kholoud K. Saleh, Oscar J. Abilez, Peng Zhao, Edouard G. Stanley, Andrew G. Elefanty, Murray Kwon, David A. Elliott, and Reza Ardehali
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - 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.
- Published
- 2016
- Full Text
- View/download PDF
4. Human pluripotent stem cell tools for cardiac optogenetics.
- Author
-
Yan Zhuge, Bhagat Patlolla, Charu Ramakrishnan, Ramin E. Beygui, Christopher K. Zarins, Karl Deisseroth, Ellen Kuhl, and Oscar J. Abilez
- Published
- 2014
- Full Text
- View/download PDF
5. Optogenetic LED array for perturbing cardiac electrophysiology.
- Author
-
Oscar J. Abilez
- Published
- 2013
- Full Text
- View/download PDF
6. Cardiac optogenetics.
- Author
-
Oscar J. Abilez
- Published
- 2012
- Full Text
- View/download PDF
7. Stimulation and artifact-free extracellular electrophysiological recording of cells in suspension.
- Author
-
Frank B. Myers, Oscar J. Abilez, Christopher K. Zarins, and Luke P. Lee
- Published
- 2011
- Full Text
- View/download PDF
8. Micropatterned Organoids Enable Modeling of the Earliest Stages of Human Cardiac Vascularization
- Author
-
Oscar J. Abilez, Huaxiao Yang, Lei Tian, Kitchener D. Wilson, Evan H. Lyall, Mengcheng Shen, Rahulkumar Bhoi, Yan Zhuge, Fangjun Jia, Hung Ta Wo, Gao Zhou, Yuan Guan, Bryan Aldana, Detlef Obal, Gary Peltz, Christopher K. Zarins, and Joseph C. Wu
- Abstract
Although model organisms have provided insight into the earliest stages of cardiac vascularization, we know very little about this process in humans. Here we show that spatially micropatterned human pluripotent stem cells (hPSCs) enablein vitromodeling of this process, corresponding to the first three weeks ofin vivohuman development. Using four hPSC fluorescent reporter lines, we create cardiac vascular organoids (cVOs) by identifying conditions that simultaneously give rise to spatially organized and branched vascular networks within endocardial, myocardial, and epicardial cells. Using single-cell transcriptomics, we show that the cellular composition of cVOs resembles that of a 6.5 post-conception week (PCW) human heart. We find that NOTCH and BMP pathways are upregulated in cVOs, and their inhibition disrupts vascularization. Finally, using the same vascular-inducing factors to create cVOs, we produce hepatic vascular organoids (hVOs). This suggests there is a conserved developmental program for creating vasculature within different organ systems.Graphic Abstract
- Published
- 2022
- Full Text
- View/download PDF
9. Power Law as a Method for Ultrasound Detection of Internal Bleeding: In Vivo Rabbit Validation.
- Author
-
Aaron S. Wang, Oscar J. Abilez, Christopher K. Zarins, Charles A. Taylor, and David H. Liang
- Published
- 2010
- Full Text
- View/download PDF
10. An in Vivo miRNA Delivery System for Restoring Infarcted Myocardium
- Author
-
Haodong Chen, Xin Zhao, Kai Li, Masataka Nishiga, Yonggang Liu, Sarah C. Heilshorn, Hung-Ta Wo, Huaxiao Yang, Jing Ge, Xulei Qin, Oscar J. Abilez, Chun Liu, Dan Ding, Nazish Sayed, and Huiyuan Wang
- Subjects
Ejection fraction ,business.industry ,Angiogenesis ,General Engineering ,General Physics and Astronomy ,02 engineering and technology ,Gene delivery ,Pharmacology ,010402 general chemistry ,021001 nanoscience & nanotechnology ,medicine.disease ,01 natural sciences ,Embryonic stem cell ,0104 chemical sciences ,Lipofectamine ,In vivo ,Heart failure ,medicine ,General Materials Science ,Myocardial infarction ,0210 nano-technology ,business - 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.
- Published
- 2019
- Full Text
- View/download PDF
11. Transcriptome analysis of non human primate-induced pluripotent stem cell-derived cardiomyocytes in 2D monolayer culture vs. 3D engineered heart tissue
- Author
-
Ning-Yi Shao, Haodong Chen, Mohamed Ameen, Joseph C. Wu, Haodi Wu, Alice F. Tarantal, Chengyi Tu, Nathan J. Cunningham, Xin Zhao, Alexandra Holmström, Tony Chour, Oscar J. Abilez, Ming-Tao Zhao, Ilanit Itzhaki, and Huaxiao Yang
- Subjects
Male ,Cell signaling ,Physiology ,medicine.medical_treatment ,Cell ,Induced Pluripotent Stem Cells ,Paracrine Communication ,Myocardial Ischemia ,Mice, SCID ,Biology ,Cell-Matrix Junctions ,Transcriptome ,Paracrine signalling ,In vivo ,Heart Rate ,Physiology (medical) ,medicine ,Animals ,Gene Regulatory Networks ,Myocytes, Cardiac ,Induced pluripotent stem cell ,Cells, Cultured ,Tissue Engineering ,Gene Expression Profiling ,Editorials ,Cell Differentiation ,Stem-cell therapy ,Macaca mulatta ,Cell Hypoxia ,Cell biology ,medicine.anatomical_structure ,Phenotype ,Cardiology and Cardiovascular Medicine ,Energy Metabolism - Abstract
Aims Stem cell therapy has shown promise for treating myocardial infarction via re-muscularization and paracrine signalling 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 ischaemic conditions in the infarcted heart under these two different delivery approaches remains unclear. Here, we aim to gain a better understanding of the effects of hypoxia on iPSC-CMs at the transcriptome level. Methods and results NHP iPSC-CMs in both monolayer culture (2D) and engineered heart tissue (EHT) (3D) 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 3D EHT model was more sensitive to ischaemic conditions and similar to the native in vivo myocardium in terms of cell–extracellular matrix/cell–cell interactions, energy metabolism, and paracrine signalling. Conclusion By exposing NHP iPSC-CMs to different culture conditions, transcriptome profiling improves our understanding of the mechanism of ischaemic injury.
- Published
- 2020
12. Passive Stretch Induces Structural and Functional Maturation of Engineered Heart Muscle as Predicted by Computational Modeling
- Author
-
Evangeline Tzatzalos, Larry A. Couture, Tony Chour, Elena Matsa, Joseph D. Gold, Paul W. Burridge, Yan Zhuge, Gwanghyun Jung, Ellen Kuhl, Huaxiao Yang, Johannes Riegler, Vincent C. Chen, Praveen K. Shukla, Ioannis Karakikes, Malte Tiburcy, Wolfram H. Zimmermann, Oscar J. Abilez, Daniel Bernstein, Alexander M. Zöllner, Joseph C. Wu, and Ming-Tao Zhao
- Subjects
Pluripotent Stem Cells ,0301 basic medicine ,chemistry.chemical_element ,Calcium ,Biology ,Article ,Cell Line ,Flow cytometry ,03 medical and health sciences ,Calcium imaging ,Tissue engineering ,medicine ,Humans ,Myocytes, Cardiac ,Induced pluripotent stem cell ,Tissue Engineering ,medicine.diagnostic_test ,Myocardium ,Computational Biology ,Cell Biology ,Anatomy ,Flow Cytometry ,Embryonic stem cell ,Potassium channel ,030104 developmental biology ,chemistry ,Biophysics ,Molecular Medicine ,Stem cell ,Developmental Biology - 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 (IMR-90 line) were differentiated to hPSC-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 × 106 hPSC-CMs were mixed with 0.4 × 106 human fibroblasts (IMR-90 line) (3:1 ratio) and type-I collagen. The blend was cast into custom-made 12-mm long polydimethylsiloxane 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 with 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.
- Published
- 2017
- Full Text
- View/download PDF
13. Anisotropic microfibrous scaffolds enhance the organization and function of cardiomyocytes derived from induced pluripotent stem cells
- Author
-
Joseph J. Kim, Joseph C. Wu, Ngan F. Huang, Nicholas P. Mezak, Karina H. Nakayama, Evangeline Tzatzalos, Maureen Wanjare, Oscar J. Abilez, and Luqia Hou
- Subjects
0301 basic medicine ,Scaffold ,business.product_category ,Induced Pluripotent Stem Cells ,Biomedical Engineering ,Biocompatible Materials ,Nanotechnology ,02 engineering and technology ,Article ,Contractility ,03 medical and health sciences ,chemistry.chemical_compound ,Tissue engineering ,Myosin ,Microfiber ,Humans ,Myocyte ,Myocytes, Cardiac ,General Materials Science ,Induced pluripotent stem cell ,reproductive and urinary physiology ,Tissue Engineering ,Tissue Scaffolds ,Chemistry ,021001 nanoscience & nanotechnology ,Cell biology ,030104 developmental biology ,embryonic structures ,Polycaprolactone ,Anisotropy ,0210 nano-technology ,business ,Porosity - 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 an dominant role of scaffold anisotropy in engineering cardiovascular tissues that maintain iCM organization and contractile function.
- Published
- 2017
- Full Text
- View/download PDF
14. Endogenous Retrovirus-Derived lncRNA BANCR Promotes Cardiomyocyte Migration in Humans and Non-human Primates
- Author
-
Yonggang Liu, Oscar J. Abilez, Evgenios Neofytou, Joseph C. Wu, Sadhana Gaddam, Thomas B. Hildebrandt, Xulei Qin, Huaxiao Yang, Howard Y. Chang, Maricela Prado, Maxwell R. Mumbach, Mohamed Ameen, Mingxia Gu, Hongchao Guo, Ning Ma, Lei Tian, Ioannis Karakikes, Kitchener D. Wilson, Nathan J. Cunningham, and Sebastian Diecke
- Subjects
Transposable element ,Primates ,Endogenous retrovirus ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Evolution, Molecular ,03 medical and health sciences ,0302 clinical medicine ,Species Specificity ,Animals ,Humans ,Myocytes, Cardiac ,Induced pluripotent stem cell ,Molecular Biology ,Transcription factor ,030304 developmental biology ,0303 health sciences ,Heart development ,Genome, Human ,Endogenous Retroviruses ,RNA ,Cell Biology ,Cell biology ,Gene Expression Regulation ,DNA Transposable Elements ,Human genome ,RNA, Long Noncoding ,030217 neurology & neurosurgery ,Function (biology) ,Developmental Biology ,Transcription Factors - 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.
- Published
- 2019
15. An
- Author
-
Huaxiao, Yang, Xulei, Qin, Huiyuan, Wang, Xin, Zhao, Yonggang, Liu, Hung-Ta, Wo, Chun, Liu, Masataka, Nishiga, Haodong, Chen, Jing, Ge, Nazish, Sayed, Oscar J, Abilez, Dan, Ding, Sarah C, Heilshorn, and Kai, Li
- Subjects
Cell Death ,Human Embryonic Stem Cells ,Gene Transfer Techniques ,Myocardial Infarction ,Hydrogels ,Cell Hypoxia ,Injections ,Rats ,MicroRNAs ,Gene Expression Regulation ,Reperfusion Injury ,Animals ,Humans ,Nanoparticles ,Myocytes, Cardiac ,Cell Proliferation - 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
- Published
- 2019
16. Treatment of volumetric muscle loss in mice using nanofibrillar scaffolds enhances vascular organization and integration
- Author
-
Ioannis Karakikes, Oscar J. Abilez, Nicholas S. Calvo, Karina H. Nakayama, Chelsey S. Simmons, Thomas A. Rando, Patrick Paine, Victor Garcia, Cynthia Alcazar, Marco Quarta, and Ngan F. Huang
- Subjects
Myoblasts, Skeletal ,Muscle Fibers, Skeletal ,Nanofibers ,Medicine (miscellaneous) ,02 engineering and technology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cell Line ,Contractility ,03 medical and health sciences ,Mice ,Tissue engineering ,medicine ,Myocyte ,Regeneration ,Animals ,Humans ,Muscle, Skeletal ,lcsh:QH301-705.5 ,030304 developmental biology ,0303 health sciences ,Cell therapies ,Tissue Engineering ,Tissue Scaffolds ,Chemistry ,Myogenesis ,Regeneration (biology) ,Skeletal muscle ,Endothelial Cells ,021001 nanoscience & nanotechnology ,Coculture Techniques ,Cell biology ,Transplantation ,Tissues ,medicine.anatomical_structure ,lcsh:Biology (General) ,Musculoskeletal models ,Cytokines ,0210 nano-technology ,General Agricultural and Biological Sciences ,Perfusion - 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., Karina Nakayama et al. bioengineer endothelialized mouse skeletal muscle using parallel-aligned nanofibrillar scaffolds. They find that muscle produced using the aligned scaffolds developed a spatially patterned structure and showed improved endothelial interaction compared to muscle engineered with randomly-oriented scaffolds.
- Published
- 2019
17. Analyzing the effects of engineering cardiomyocyte shape: quantitative phase imaging reveals differences in morphology and function (Conference Presentation)
- Author
-
Christine Cordeiro, Jenny M. Vo-Phamhi, Olav Solgaard, Oscar J. Abilez, Alison K. Schroer, Aleksandra K. Denisin, and Elizabeth L. Pruitt
- Subjects
Materials science ,Phase imaging ,Morphology (biology) ,Function (mathematics) ,Presentation (obstetrics) ,Biomedical engineering - Published
- 2019
- Full Text
- View/download PDF
18. iPSC-derived cardiomyocytes reveal abnormal TGFβ signaling in left ventricular non-compaction cardiomyopathy
- Author
-
Joseph C. Wu, Michael Snyder, Jared M. Churko, Keiichi Hirono, Fereshteh Jahanbani, Fukiko Ichida, Gwanghyun Jung, Daniel Bernstein, Antje D. Ebert, Oscar J. Abilez, Kazuki Kodo, Vittavat Termglinchan, Sean M. Wu, Kolsoum InanlooRahatloo, Ioannis Karakikes, Praveen K. Shukla, and Sang-Ging Ong
- Subjects
0301 basic medicine ,Heart Defects, Congenital ,TBX20 ,Heart Ventricles ,Induced Pluripotent Stem Cells ,Cardiomyopathy ,030204 cardiovascular system & hematology ,Biology ,Article ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Transforming Growth Factor beta ,medicine ,Humans ,Myocytes, Cardiac ,Induced pluripotent stem cell ,Induced stem cells ,Cell Biology ,Transforming growth factor beta ,medicine.disease ,Embryonic stem cell ,Phenotype ,3. Good health ,Cell biology ,030104 developmental biology ,Mutation ,biology.protein ,Cardiomyopathies ,T-Box Domain Proteins ,Signal Transduction - 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.
- Published
- 2016
19. Engineered heart tissues and induced pluripotent stem cells: Macro- and microstructures for disease modeling, drug screening, and translational studies
- Author
-
Oscar J. Abilez, Praveen K. Shukla, Evangeline Tzatzalos, and Joseph C. Wu
- Subjects
0301 basic medicine ,Drug ,Stromal cell ,media_common.quotation_subject ,Induced Pluripotent Stem Cells ,Cell ,Drug Evaluation, Preclinical ,Pharmaceutical Science ,Disease ,Bioinformatics ,Article ,Translational Research, Biomedical ,03 medical and health sciences ,Tissue engineering ,medicine ,Animals ,Humans ,Myocytes, Cardiac ,Induced pluripotent stem cell ,media_common ,Tissue Engineering ,business.industry ,Models, Cardiovascular ,Heart ,Precision medicine ,Cardiotoxicity ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Cardiovascular Diseases ,Stem cell ,business - 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.
- Published
- 2016
- Full Text
- View/download PDF
20. Big bottlenecks in cardiovascular tissue engineering
- Author
-
Joseph C. Wu, Young Sup Yoon, Jianyi Zhang, Vahid Serpooshan, Gaspard Pardon, Karina H. Nakayama, Ngan F. Huang, Beth L. Pruitt, Nazish Sayed, Oscar J. Abilez, Viola B. Morris, and Sean M. Wu
- Subjects
0301 basic medicine ,Computer science ,Comment ,Medicine (miscellaneous) ,Limiting ,General Biochemistry, Genetics and Molecular Biology ,3. Good health ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Tissue engineering ,Risk analysis (engineering) ,General Agricultural and Biological Sciences ,Induced pluripotent stem cell ,030217 neurology & neurosurgery - 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., In this Comment, Ngan Huang et al. discuss recent advances in cardiovascular tissue engineering and some of the main challenges that remain in translating these advances to the clinic. The authors propose future direction for the field to focus research efforts.
- Published
- 2018
- Full Text
- View/download PDF
21. A 3D boost
- Author
-
Joseph C. Wu and Oscar J. Abilez
- Subjects
0301 basic medicine ,Somatic cell ,Mechanical Engineering ,General Chemistry ,Biology ,Condensed Matter Physics ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,Mechanics of Materials ,Cell culture ,General Materials Science ,Stem cell ,Induced pluripotent stem cell ,Reprogramming - Abstract
Biophysical factors in an optimized three-dimensional microenvironment enhance the reprogramming efficiency of human somatic cells into pluripotent stem cells when compared to traditional cell-culture substrates.
- Published
- 2016
- Full Text
- View/download PDF
22. Optophysiology of cardiomyocytes: characterizing cellular motion with quantitative phase imaging
- Author
-
Christine Cordeiro, Oscar J. Abilez, Georges Goetz, Tushar Gupta, Yan Zhuge, Olav Solgaard, and Daniel Palanker
- Subjects
0301 basic medicine ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030204 cardiovascular system & hematology ,Atomic and Molecular Physics, and Optics ,Article ,Biotechnology - 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.
- Published
- 2017
23. Characterizing Cardiomyocytes Motion with Quantitative Phase Imaging
- Author
-
Oscar J. Abilez, Olav Solgaard, Tushar Gupta, Georges Goetz, Christine Cordeiro, and Daniel Palanker
- Subjects
Materials science ,Noise reduction ,Phase contrast microscopy ,Motion (geometry) ,Speckle noise ,Nanotechnology ,030204 cardiovascular system & hematology ,01 natural sciences ,law.invention ,010309 optics ,03 medical and health sciences ,0302 clinical medicine ,law ,0103 physical sciences ,Phase imaging ,Patch clamp ,Biomedical engineering - Abstract
Characterizing cardiomyocytes' activity is important for drug development, but traditional patch clamping analysis is destructive and slow. A label-free method for extracting timing and motion characteristics of cardiomyocytes using quantitative phase imaging is presented.
- Published
- 2017
- Full Text
- View/download PDF
24. Effect of Human Donor Cell Source on Differentiation and Function of Cardiac Induced Pluripotent Stem Cells
- Author
-
Andrew S. Lee, Sang-Ging Ong, Shijun Hu, Bruno C. Huber, Mei Huang, Feng Lan, Wan Xing Hong, Ping Liang, Joseph C. Wu, Oscar J. Abilez, and Veronica Sanchez-Freire
- Subjects
Donor cell ,Cardiac differentiation ,Cellular differentiation ,Induced Pluripotent Stem Cells ,cardiac differentiation ,health services administration ,Humans ,Medicine ,Myocytes, Cardiac ,Induced pluripotent stem cell ,Cells, Cultured ,Embryonic Stem Cells ,health care economics and organizations ,Genetics ,business.industry ,epigenetic memory ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Fibroblasts ,Embryonic stem cell ,Tissue Donors ,Cell biology ,DNA methylation ,pluripotent stem cells ,Cardiology and Cardiovascular Medicine ,business ,Function (biology) ,Stem Cell Transplantation - Abstract
BackgroundHuman-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.ObjectivesThis 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.MethodsCardiac 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.ResultsDifferentiation 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.ConclusionsThis 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.
- Published
- 2014
- Full Text
- View/download PDF
25. Prospective isolation of human embryonic stem cell-derived cardiovascular progenitors that integrate into human fetal heart tissue
- Author
-
Michael Q. Chen, Irving L. Weissman, Roeland Nusse, Yongquan Gong, Oscar J. Abilez, Matthew A. Inlay, Reza Ardehali, Masayuki Yazawa, Shah R. Ali, Micha Drukker, and Timothy A. Blauwkamp
- Subjects
Receptor, Platelet-Derived Growth Factor alpha ,Primitive Streak ,Cellular differentiation ,Xenotransplantation ,medicine.medical_treatment ,Myocytes, Smooth Muscle ,Cell Culture Techniques ,Cell Separation ,Biology ,Receptor Tyrosine Kinase-like Orphan Receptors ,Mesoderm ,Mice ,Fetus ,medicine ,Animals ,Humans ,Cell Lineage ,Myocytes, Cardiac ,Progenitor cell ,Cells, Cultured ,Embryonic Stem Cells ,Tissue Survival ,Multidisciplinary ,Heart development ,Gene Expression Profiling ,Multipotent Stem Cells ,Myocardium ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Heart ,Kinase insert domain receptor ,Biological Sciences ,Vascular Endothelial Growth Factor Receptor-2 ,Embryonic stem cell ,Cell biology ,Transplantation ,embryonic structures ,Immunology ,Endothelium, Vascular ,Stem cell ,Biomarkers ,Stem Cell Transplantation - 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.
- Published
- 2013
- Full Text
- View/download PDF
26. Label-free electrophysiological cytometry for stem cell-derived cardiomyocyte clusters
- Author
-
Frank B. Myers, Oscar J. Abilez, Christopher K. Zarins, and Luke P. Lee
- Subjects
Cell type ,Cellular differentiation ,Induced Pluripotent Stem Cells ,Biomedical Engineering ,Bioengineering ,Biology ,Biochemistry ,Regenerative medicine ,Article ,Flow cytometry ,Automation ,medicine ,Humans ,Myocytes, Cardiac ,Electrodes ,Extracellular field potential ,Cells, Cultured ,medicine.diagnostic_test ,Cluster of differentiation ,Cell Differentiation ,General Chemistry ,Microfluidic Analytical Techniques ,Flow Cytometry ,Cell biology ,Stem cell ,Cytometry ,Biomedical engineering - 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.
- Published
- 2013
- Full Text
- View/download PDF
27. Abnormal Calcium Handling Properties Underlie Familial Hypertrophic Cardiomyopathy Pathology in Patient-Specific Induced Pluripotent Stem Cells
- Author
-
Euan A. Ashley, Chelsey S. Simmons, Donald M. Bers, Shijun Hu, Feng Lan, Veronica Sanchez-Freire, Ping Liang, Michael T. Longaker, Richard S. Lewis, Patricia K. Nguyen, Beth L. Pruitt, Li Wang, Andrew S. Lee, Michelle Yen, Joseph C. Wu, Enrique G. Navarrete, Oscar J. Abilez, Leng Han, Matthew T. Wheeler, Ning Sun, Antje D. Ebert, Yoshinori Yamaguchi, Robert C. Robbins, and Yongming Wang
- Subjects
Pathology ,medicine.medical_specialty ,Induced Pluripotent Stem Cells ,Mutation, Missense ,Disease ,030204 cardiovascular system & hematology ,Biology ,Article ,Muscle hypertrophy ,Sudden cardiac death ,03 medical and health sciences ,0302 clinical medicine ,Cardiomyopathy, Hypertrophic, Familial ,Genetics ,medicine ,Humans ,Missense mutation ,cardiovascular diseases ,Induced pluripotent stem cell ,Cells, Cultured ,030304 developmental biology ,0303 health sciences ,Myosin Heavy Chains ,Cell Biology ,medicine.disease ,Phenotype ,3. Good health ,cardiovascular system ,Molecular Medicine ,Calcium ,MYH7 ,Cardiac Myosins ,Homeostasis - Abstract
Summary 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.
- Published
- 2013
- Full Text
- View/download PDF
28. Partial Reprogramming of Pluripotent Stem Cell-Derived Cardiomyocytes into Neurons
- Author
-
Ryoko Hamaguchi, Kuppusamy Rajarajan, Guang Li, Arun Sharma, Sean M. Wu, Praveen K. Shukla, Marius Wernig, Moritz Mall, Oscar J. Abilez, Joseph C. Wu, and Wenpo Chuang
- Subjects
0301 basic medicine ,Pluripotent Stem Cells ,Doublecortin Protein ,Somatic cell ,Cellular differentiation ,Population ,Fluorescent Antibody Technique ,Gene Expression ,Biology ,Article ,Cell Line ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Genes, Reporter ,Transduction, Genetic ,health services administration ,Animals ,Myocytes, Cardiac ,Induced pluripotent stem cell ,education ,health care economics and organizations ,Embryonic Stem Cells ,NeuroD ,Neurons ,education.field_of_study ,Multidisciplinary ,Cell Differentiation ,Cellular Reprogramming ,Embryonic stem cell ,Cell biology ,Electrophysiological Phenomena ,ASCL1 ,030104 developmental biology ,Phenotype ,nervous system ,Reprogramming ,030217 neurology & neurosurgery ,Biomarkers - 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.
- Published
- 2016
29. Stem cell reprogramming: A 3D boost
- Author
-
Oscar J, Abilez and Joseph C, Wu
- Subjects
Pluripotent Stem Cells ,Cellular Microenvironment ,Cell Culture Techniques ,Animals ,Humans ,Cell Differentiation ,Epithelial Cells ,Mesenchymal Stem Cells - Published
- 2016
30. CD13 and ROR2 Permit Isolation of Highly Enriched Cardiac Mesoderm from Differentiating Human Embryonic Stem Cells
- Author
-
Edouard G. Stanley, Oscar J. Abilez, Rhys J.P. Skelton, David A. Elliott, Murray Kwon, Andrew G. Elefanty, Reza Ardehali, Deevina Arasaratnam, Bevin Brady, Debashis Sahoo, Kholoud K. Saleh, James L. Engel, Suhail Khoja, and Peng Zhao
- Subjects
0301 basic medicine ,Time Factors ,Swine ,Cellular differentiation ,Human Embryonic Stem Cells ,Fluorescent Antibody Technique ,Cardiovascular ,Regenerative Medicine ,Biochemistry ,Mesoderm ,Mice ,Stem Cell Research - Nonembryonic - Human ,Myocytes, Cardiac ,Induced pluripotent stem cell ,lcsh:QH301-705.5 ,Stem cell transplantation for articular cartilage repair ,lcsh:R5-920 ,Heterologous ,Reverse Transcriptase Polymerase Chain Reaction ,Cell Differentiation ,Cell biology ,medicine.anatomical_structure ,Heart Disease ,Muscle ,Stem Cell Research - Nonembryonic - Non-Human ,Smooth ,Stem cell ,Development of treatments and therapeutic interventions ,lcsh:Medicine (General) ,Cardiac ,Pluripotent Stem Cells ,Cell type ,Transplantation, Heterologous ,Clinical Sciences ,Biology ,CD13 Antigens ,Receptor Tyrosine Kinase-like Orphan Receptors ,Article ,Cell Line ,03 medical and health sciences ,Genetics ,medicine ,Animals ,Humans ,Cell Lineage ,Progenitor cell ,Stem Cell Research - Embryonic - Human ,Myocytes ,Transplantation ,5.2 Cellular and gene therapies ,Myocardium ,Gene Expression Profiling ,Endothelial Cells ,Muscle, Smooth ,Cell Biology ,Fibroblasts ,Stem Cell Research ,Embryonic stem cell ,body regions ,030104 developmental biology ,lcsh:Biology (General) ,Immunology ,Biochemistry and Cell Biology ,Developmental Biology ,Stem Cell Transplantation - Abstract
Summary 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., Highlights • CD13 and ROR2 separate hESC-derived MIXL1+ mesoderm from MIXL1+ endoderm • CD13 and ROR2 select for a population of highly enriched pre-cardiac mesoderm • CD13+/ROR2+ cells derived from hESCs engraft into porcine, but not murine hearts • CD13+/ROR2+ cells differentiate to all major cardiac lineages in the pig heart, CD13 and ROR2 mark pre-cardiac mesoderm precursors of definitive cardiac cell types. Ardehali and colleagues show that upon transplantation in a pig's heart, these cells engraft and differentiate to cardiomyocytes, endothelium, smooth muscle, and fibroblasts.
- Published
- 2016
- Full Text
- View/download PDF
31. IN VITRO/IN SILICO CHARACTERIZATION OF ACTIVE AND PASSIVE STRESSES IN CARDIAC MUSCLE
- Author
-
Ellen Kuhl, Oscar J. Abilez, Ahmed N Assar, Christopher K. Zarins, and Markus Boel
- Subjects
Cardiac function curve ,Area fraction ,Materials science ,Computer Networks and Communications ,In silico ,Computational Mechanics ,Cardiac muscle ,Isometric exercise ,Bioinformatics ,In vitro ,medicine.anatomical_structure ,Active force ,Control and Systems Engineering ,medicine ,Cardiac muscle tissue ,Biomedical engineering - Abstract
We propose a novel, robust, and easily reproducible, in vitro/in silico model system to characterize active and passive stresses in electro-active cardiac muscle using a hybrid experimental/computational approach. We explore active and passive stresses in healthy explanted heart slices in vitro, design a virtual test-bed to simulate the in vitro measured stresses in silico, and predict altered active force generation in infarcted hearts in silico. For the in vitro model, explanted rat heart tissue slices are mounted on a force transducer and stimulated electrically through biphasic pulses. Isometric forces are recorded and translated into active circumferential stress. For the in silico model, stresses are additively decomposed into passive and active contributions, with the latter being related to the measured isometric force. A hierarchical finite element model for cardiac muscle tissue is developed based on passive tetrahedral unit cells, representing a network of interconnected polymeric chains, and active trusses, representing the contracting muscle fibers. First we calibrate the model against our experiments with healthy explanted rat heart slices. Then we predict acute and chronic alterations in active stress generation in infarcted hearts. We virtually explore isometric forces generation for different infarct area fractions and infarct locations. This approach has the potential to precisely quantify global loss of cardiac function for a given infarct area fraction.
- Published
- 2012
- Full Text
- View/download PDF
32. Multiscale Computational Models for Optogenetic Control of Cardiac Function
- Author
-
Oscar J. Abilez, Jonathan Wong, Rohit Prakash, Ellen Kuhl, Karl Deisseroth, and Christopher K. Zarins
- Subjects
Rhodopsin ,Computational model ,Light ,Optical Phenomena ,Cellular differentiation ,Finite Element Analysis ,Biophysics ,Action Potentials ,Cell Differentiation ,Heart ,Nanotechnology ,Video microscopy ,Gating ,Multielectrode array ,Biology ,Optogenetics ,Photostimulation ,Directed differentiation ,Genetic Techniques ,Humans ,Cellular Biophysics and Electrophysiology ,Computer Simulation ,Myocytes, Cardiac ,Embryonic Stem Cells - 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.
- Published
- 2011
- Full Text
- View/download PDF
33. Power Law as a Method for Ultrasound Detection of Internal Bleeding: In Vivo Rabbit Validation
- Author
-
Charles A. Taylor, Christopher K. Zarins, Aaron S. Wang, David Liang, and Oscar J. Abilez
- Subjects
medicine.medical_specialty ,Internal bleeding ,Biomedical Engineering ,Hemodynamics ,Hemorrhage ,Image Processing, Computer-Assisted ,medicine ,Animals ,Ultrasonography ,Vascular disease ,business.industry ,Ultrasound ,Models, Cardiovascular ,Reproducibility of Results ,Blood flow ,Bleed ,medicine.disease ,Cannula ,Arterial tree ,Femoral Artery ,ROC Curve ,Regional Blood Flow ,Area Under Curve ,Rabbits ,Radiology ,medicine.symptom ,business ,Algorithms ,Blood Flow Velocity - 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.
- Published
- 2010
- Full Text
- View/download PDF
34. A generic approach towards finite growth with examples of athlete's heart, cardiac dilation, and cardiac wall thickening
- Author
-
Serdar Göktepe, Ellen Kuhl, and Oscar J. Abilez
- Subjects
Continuum (topology) ,Quantitative Biology::Tissues and Organs ,Mechanical Engineering ,Physics::Medical Physics ,Mathematical analysis ,Work (physics) ,Kinematics ,Condensed Matter Physics ,Finite element method ,Mechanics of Materials ,Finite strain theory ,Tensor (intrinsic definition) ,Calculus ,Dilation (morphology) ,Ansatz ,Mathematics - Abstract
The objective of this work is to establish a generic continuum-based computational concept for finite growth of living biological tissues. The underlying idea is the introduction of an incompatible growth configuration which naturally introduces a multiplicative decomposition of the deformation gradient into an elastic and a growth part. The two major challenges of finite growth are the kinematic characterization of the growth tensor and the identification of mechanical driving forces for its evolution. Motivated by morphological changes in cell geometry, we illustrate a micromechani- cally motivated ansatz for the growth tensor for cardiac tissue that can capture both strain-driven ventricular dilation and stress-driven wall thickening. Guided by clinical observations, we explore three distinct pathophysiological cases: athlete's heart, cardiac dilation, and cardiac wall thickening. We demonstrate the computational solution of finite growth within a fully implicit incremental iterative Newton-Raphson based finite element solution scheme. The features of the proposed approach are illustrated and compared for the three different growth pathologies in terms of a generic bi-ventricular heart model.
- Published
- 2010
- Full Text
- View/download PDF
35. Iliac fixation inhibits migration of both suprarenal and infrarenal aortic endografts
- Author
-
Oscar J. Abilez, Tami Crabtree, Peyman Benharash, Christopher K. Zarins, Jason T. Lee, and Daniel A. Bloch
- Subjects
Male ,medicine.medical_specialty ,Time Factors ,medicine.medical_treatment ,Prosthesis Design ,Iliac Artery ,Blood Vessel Prosthesis Implantation ,Fixation (surgical) ,Imaging, Three-Dimensional ,Renal Artery ,Aneurysm ,Foreign-Body Migration ,Predictive Value of Tests ,Risk Factors ,medicine.artery ,Image Interpretation, Computer-Assisted ,Odds Ratio ,Humans ,Medicine ,Prospective Studies ,Superior mesenteric artery ,Aged ,Aorta ,business.industry ,Angioplasty ,Stent ,medicine.disease ,Common iliac artery ,Surgery ,Connecticut ,Apposition ,Logistic Models ,Treatment Outcome ,medicine.anatomical_structure ,Female ,Stents ,business ,Cardiology and Cardiovascular Medicine ,Tomography, Spiral Computed ,Aortic Aneurysm, Abdominal ,Follow-Up Studies ,Artery - Abstract
ObjectiveTo evaluate the role of iliac fixation in preventing migration of suprarenal and infrarenal aortic endografts.MethodsQuantitative 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.ResultsThere 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° vs 11°; 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.ConclusionsDistal 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.
- Published
- 2007
- Full Text
- View/download PDF
36. Human Engineered Heart Muscles Engraft and Survive Long-Term in a Rodent Myocardial Infarction Model
- Author
-
Malte Tiburcy, Tim Meyer, Antje D. Ebert, Joseph D. Gold, Vincent C. Chen, Qi Shen, Joseph C. Wu, Evgenios Neofytou, Mouer Wang, Oscar J. Abilez, Nigel G. Kooreman, Larry A. Couture, Andrew J. Connolly, Uwe Raaz, Philip S. Tsao, Evangeline Tzatzalos, Wolfram H. Zimmermann, and Johannes Riegler
- Subjects
Male ,Time Factors ,Physiology ,medicine.medical_treatment ,Nude ,Myocardial Infarction ,030204 cardiovascular system & hematology ,Cardiorespiratory Medicine and Haematology ,Cardiovascular ,Regenerative Medicine ,Rats, Sprague-Dawley ,0302 clinical medicine ,Myocytes, Cardiac ,Myocardial infarction ,Heart transplantation ,cardiac function tests ,0303 health sciences ,Graft Survival ,Cell Differentiation ,Papillary Muscles ,3. Good health ,myocardial ischemia ,Heart Disease ,Cardiology ,Heterografts ,Cardiology and Cardiovascular Medicine ,Cardiac ,Immunosuppressive Agents ,medicine.medical_specialty ,Cell Survival ,Clinical Sciences ,Ischemia ,Transfection ,Article ,Cell Line ,03 medical and health sciences ,Rats, Nude ,Internal medicine ,medicine ,cardiac MRI ,Bioluminescence imaging ,Animals ,Humans ,Heart Disease - Coronary Heart Disease ,Embryonic Stem Cells ,030304 developmental biology ,Myocytes ,Transplantation ,Tissue Engineering ,business.industry ,Animal ,Stroke Volume ,medicine.disease ,Stem Cell Research ,Embryonic stem cell ,Myocardial Contraction ,Surgery ,Rats ,Disease Models, Animal ,Cardiovascular System & Hematology ,Heart failure ,Connexin 43 ,Disease Models ,Heart Transplantation ,Sprague-Dawley ,business ,Reperfusion injury ,Biomarkers - Abstract
Rationale: 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. Objective: 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. Methods and Results: 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. Conclusions: 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.
- Published
- 2015
37. Abstract 248: Aberrant TGFβ Signaling as an Etiology of Left Ventricular Non-compaction Cardiomyopathy
- Author
-
Kazuki Kodo, Sang-Ging Ong, Fereshteh Jahanbani, Vittavat Termglinchan, Kolsoum InanlooRahatloo, Antje D Ebert, Praveen Shukla, Oscar J Abilez, Jared M Churko, Ioannis Karakikes, Gwanghyun Jung, Michael P Snyder, Daniel Bernstein, and Joseph C Wu
- Subjects
Physiology ,Cardiology and Cardiovascular Medicine - Abstract
Left ventricular non-compaction (LVNC) is the third most prevalent cardiomyopathy in children and has a unique phenotype with characteristically extensive hypertrabeculation of the left ventricle, similar to the embryonic left ventricle, suggesting a developmental defect of the embryonic myocardium. However, studying this disease has been challenging due to the lack of an animal model that can faithfully recapitulate the clinical phenotype of LVNC. To address this, we showed that patient-specific induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) generated from a family with LVNC history recapitulated a developmental defect consistent with the LVNC phenotype at the single-cell level. We then utilized hiPSC-CMs to show that increased transforming growth factor beta (TGFβ) signaling is one of the central mechanisms underlying the pathogenesis of LVNC. LVNC hiPSC-CMs demonstrated decreased proliferative capacity due to abnormal activation of TGFβ signaling (Figs A-B). Exome sequencing demonstrated a mutation in TBX20, which regulates TGFβ signaling through upregulation of ITGAV, contributing to the LVNC phenotype. Inhibition of abnormal TGFβ signaling or genetic correction of the TBX20 mutation (Figs C-D) using TALEN reversed the proliferation defects seen in LVNC hiPSC-CMs. Our results demonstrate that hiPSC-CMs are a useful tool for the exploration of novel mechanisms underlying poorly understood cardiomyopathies such as LVNC. Here we provide the first evidence of activation of TGFβ signaling as playing a role in the pathogenesis of LVNC.
- Published
- 2015
- Full Text
- View/download PDF
38. A Novel Culture System Shows that Stem Cells Can be Grown in 3D and Under Physiologic Pulsatile Conditions for Tissue Engineering of Vascular Grafts
- Author
-
Emiko Miyamoto, Chengpei Xu, Adrian Gale, Peyman Benharash, Oscar J. Abilez, Jean Picquet, Christopher K. Zarins, and Mahncy Mehrotra
- Subjects
Myocytes, Smooth Muscle ,Cell Culture Techniques ,Pulsatile flow ,Video microscopy ,Matrix (biology) ,Mice ,Bioreactors ,Tissue engineering ,Pressure ,Animals ,Matrigel ,Tissue Engineering ,Chemistry ,Stem Cells ,Endothelial Cells ,Cell Differentiation ,Anatomy ,Fibroblasts ,Embryo, Mammalian ,Embryonic stem cell ,Cell biology ,Drug Combinations ,Cell culture ,Pulsatile Flow ,Blood Vessels ,Proteoglycans ,Surgery ,Collagen ,Laminin ,Stem cell ,Rheology - Abstract
Background. Currently available vascular grafts have been limited by variable patency rates, material avail- ability, and immunological rejection. The creation of a tissue-engineered vascular graft (TEVG) from autolo- gous stem cells would potentially overcome these limi- tations. As a first step in creating a completely autolo- gous 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. Materials and methods. A bioreactor was created to provide pulsatile conditions to a specially modified four-well Labtek Chamber-Slide culture system. Un- differentiated mESC were either suspended in a 3D Matrigel matrix or suspended only in cell-culture me- dia within the culture system. Pulsatile conditions were applied to the suspended cells and visualized by video microscopy. Results. 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 con- strained 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 sus- pended in media alone. Conclusions. Undifferentiated mESC can be grown in 3D and under pulsatile conditions. We will use these results to study the effects of long-term pulsatile con- ditions on the differentiation of mESC into endothelial cells, smooth muscle cells, and fibroblast cells with the long-term goal of creating a completely autologous
- Published
- 2006
- Full Text
- View/download PDF
39. Differential stickiness
- Author
-
Joseph C. Wu and Oscar J. Abilez
- Subjects
Adhesion strength ,Cell type ,Stem Cell Isolation ,Mechanics of Materials ,Mechanical Engineering ,General Materials Science ,General Chemistry ,Biology ,Condensed Matter Physics ,Induced pluripotent stem cell ,Molecular biology ,Cell biology - Abstract
Technologies to isolate colonies of human pluripotent stem cells from other cell types in a high-throughput manner are lacking. A microfluidic-based approach that exploits differences in the adhesion strength between these cells and a substrate may soon fill the gap.
- Published
- 2013
- Full Text
- View/download PDF
40. Human pluripotent stem cells (hPSCs) for heart regeneration
- Author
-
J.C. Wu and Oscar J. Abilez
- Subjects
Directed differentiation ,health services administration ,Regeneration (biology) ,Cellular differentiation ,Potential source ,Optogenetics ,Cell sorting ,Biology ,Induced pluripotent stem cell ,Mechanical force ,Neuroscience ,health care economics and organizations ,Biomedical engineering - Abstract
Cardiovascular disease is the number one cause of mortality in the USA. Because the regenerative capacity of cardiac tissue is limited, human pluripotent stem cells (hPSCs) have emerged as a potential source for cellular-based therapies. However, for these therapies to be effective, sufficient numbers of differentiated cells must be produced and properly sorted, arrhythmias must be avoided, and mechanical force must be produced. In this chapter, we describe directed differentiation of hPSCs into cardiomyocytes (hPSC-CMs), cell sorting of hPSC-CMs, and electrical, optogenetic and mechanical stimulation of hPSC-CMs for improving their function. Finally, we discuss using hPSC-CMs for disease modeling.
- Published
- 2014
- Full Text
- View/download PDF
41. Robust pluripotent stem cell expansion and cardiomyocyte differentiation via geometric patterning
- Author
-
Oscar J. Abilez, Frank B. Myers, Yan Zhuge, Ramin E. Beygui, Christopher K. Zarins, Jason S. Silver, and Luke P. Lee
- Subjects
Homeobox protein NANOG ,Pluripotent Stem Cells ,Stage-Specific Embryonic Antigens ,General Science & Technology ,Cellular differentiation ,Cell ,Biophysics ,Cell Culture Techniques ,Biology ,Cardiovascular ,Regenerative Medicine ,Biochemistry ,Article ,Fluorescence ,Medicinal and Biomolecular Chemistry ,Directed differentiation ,medicine ,Humans ,Myocytes, Cardiac ,Stem Cell Research - Embryonic - Human ,Induced pluripotent stem cell ,Homeodomain Proteins ,Myocytes ,Microscopy ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Stem Cell Research - Induced Pluripotent Stem Cell ,Nanog Homeobox Protein ,Cell Differentiation ,Anatomy ,Pharmacology and Pharmaceutical Sciences ,Stem Cell Research ,Cell biology ,medicine.anatomical_structure ,Microscopy, Fluorescence ,Cell culture ,Biochemistry and Cell Biology ,Stem cell ,Cardiac - 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.
- Published
- 2013
42. Chemically defined generation of human cardiomyocytes
- Author
-
Elena Matsa, Nicholas M. Mordwinkin, Paul W. Burridge, Jared M. Churko, Ziliang C Lin, Sebastian Diecke, Bruno C. Huber, Antje D. Ebert, Joseph C. Wu, Joseph D. Gold, Praveen K. Shukla, Bianxiao Cui, Feng Lan, Jordan R. Plews, and Oscar J. Abilez
- Subjects
Cellular differentiation ,Cardiac differentiation ,Induced Pluripotent Stem Cells ,small molecule ,cardiomyocyte ,heart ,Biology ,Biochemistry ,Article ,03 medical and health sciences ,0302 clinical medicine ,Myocyte ,Humans ,Myocytes, Cardiac ,chemically defined medium ,Induced pluripotent stem cell ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,business.industry ,Cell Differentiation ,Cell Biology ,differentiation ,Cell biology ,Biotechnology ,Culture Media ,business ,030217 neurology & neurosurgery ,Human induced pluripotent stem cell - Abstract
Existing methodologies for human induced pluripotent stem cell (hiPSC) cardiac differentiation are efficient but require the use of 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 a highly optimized cardiac differentiation strategy, employing 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 differentiation induction, 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 is the first fully chemically defined platform for cardiac specification of hiPSCs, and allows the elucidation of cardiomyocyte macromolecular and metabolic requirements whilst providing a minimally complex system for the study of maturation and subtype specification.
- Published
- 2013
43. Computational Optogenetics: A Novel Continuum Framework for the Photoelectrochemistry of Living Systems
- Author
-
Oscar J. Abilez, Jonathan Wong, and Ellen Kuhl
- Subjects
Materials science ,Mechanical Engineering ,Photoelectrochemistry ,Nanotechnology ,Gating ,Optogenetics ,Condensed Matter Physics ,Article ,Photostimulation ,Living systems ,Nonlinear system ,medicine.anatomical_structure ,Mechanics of Materials ,medicine ,Myocyte ,Neuroscience ,Cardiac muscle cell - 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.
- Published
- 2012
44. Computational Modelling of Optogenetics in Cardiac Cells
- Author
-
Oscar J. Abilez, Ellen Kuhl, and Jonathan Wong
- Subjects
Engineering ,business.industry ,fungi ,Optogenetics ,Cardiac cell ,Finite element method ,Electrophysiology ,nervous system ,Optical stimulation ,Myocyte ,Engineering simulation ,business ,Ion channel ,Biomedical engineering - Abstract
Channelrhodopsin-2 (ChR2) is a light-activated ion channel that can allow scientists to electrically activate cells via optical stimulation. Using a combination of existing computational electrophysiological and mechanical cardiac cell models with a novel ChR2 ion channel model, we created a model for ChR2-transduced cardiac myocytes. We implemented this model into a commonly available finite element platform and simulated both the single cell and the tissue electromechanical response. Our simulations show that it is possible to stimulate cardiac tissue optically with ChR2-transduced cells.Copyright © 2012 by ASME
- Published
- 2012
- Full Text
- View/download PDF
45. Patient-specific induced pluripotent stem cells as a model for familial dilated cardiomyopathy
- Author
-
Shin Lin, Euan A. Ashley, Aleksandra Pavlovic, Michael Snyder, Masayuki Yazawa, Veronica Sanchez-Freire, Oscar J. Abilez, Joseph C. Wu, Andrew S. Lee, Leng Han, Ning Sun, Roger J. Hajjar, Robert C. Robbins, Rui Chen, Manish J. Butte, Jianwei Liu, Enrique G. Navarrete, Michael T. Longaker, Li Wang, Shijun Hu, and Ricardo E. Dolmetsch
- Subjects
Cardiomyopathy, Dilated ,medicine.medical_specialty ,medicine.medical_treatment ,Adrenergic beta-Antagonists ,Induced Pluripotent Stem Cells ,Cardiomyopathy ,Biology ,Article ,Sarcoplasmic Reticulum Calcium-Transporting ATPases ,Troponin T ,Internal medicine ,medicine ,Myocyte ,Humans ,Myocytes, Cardiac ,cardiovascular diseases ,Induced pluripotent stem cell ,Cells, Cultured ,Calcium metabolism ,Heart transplantation ,Dilated cardiomyopathy ,General Medicine ,Adrenergic beta-Agonists ,medicine.disease ,musculoskeletal system ,Heart failure ,Cardiology ,cardiovascular system ,Calcium - 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.
- Published
- 2012
46. Localized control of exsanguinating arterial hemorrhage: an experimental model
- Author
-
Bonnie L. Johnson, Oscar J. Abilez, Charles A. Taylor, M. Haick, Christopher K. Zarins, Norman M. Rich, and Chengpei Xu
- Subjects
Resuscitation ,Carotid Artery, Common ,Carotid arteries ,Hemodynamics ,Blood volume ,Postoperative Hemorrhage ,Injury Site ,Medicine ,Animals ,Military Medicine ,Blood Specimen Collection ,Sheep ,business.industry ,Experimental model ,Hemostatic Techniques ,Operative Blood Salvage ,Ultrasound ,General Medicine ,Hemostasis, Surgical ,Disease Models, Animal ,Hemostasis ,Anesthesia ,Surgery ,business ,Carotid Artery Injuries - Abstract
UNLABELLED To develop an arterial injury model for testing hemostatic devices at well-defined high and low bleeding rates. MATERIAL AND METHOD 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. RESULTS 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. CONCLUSION 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.
- Published
- 2011
47. Computational modeling of growth: systemic and pulmonary hypertension in the heart
- Author
-
Manuel K. Rausch, Oscar J. Abilez, Ellen Kuhl, A. Dam, Serdar Göktepe, and OpenMETU
- Subjects
Hypertension, Pulmonary ,Constitutive equation ,Finite elements ,Cardiomegaly ,Growth ,Sarcomere ,Article ,Sarcomerogenesis ,medicine ,Humans ,Computer Simulation ,Biomechanics ,Tensor ,Mathematics ,business.industry ,Mechanical Engineering ,Myocardium ,Models, Cardiovascular ,Heart ,Mechanics ,Structural engineering ,Hypertrophy ,medicine.disease ,Pulmonary hypertension ,Finite element method ,Remodeling ,Modeling and Simulation ,Finite strain theory ,Hypertension ,business ,Biotechnology - 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.
- Published
- 2011
48. Stretchable microelectrode array using room-temperature liquid alloy interconnects
- Author
-
Babak Ziaie, Pinghung Wei, Oscar J. Abilez, Rebecca E. Taylor, Zhenwen Ding, G. Higgs, Cindy Chung, and Beth L. Pruitt
- Subjects
Materials science ,Polydimethylsiloxane ,Biocompatibility ,business.industry ,Mechanical Engineering ,Electrical engineering ,chemistry.chemical_element ,Fusible alloy ,Multielectrode array ,Tungsten ,equipment and supplies ,Electronic, Optical and Magnetic Materials ,Nanoscience and Nanotechnology ,chemistry.chemical_compound ,Microelectrode ,chemistry ,Mechanics of Materials ,Electrode ,DIFFERENTIATION ,CIRCUITS ,Electrode array ,Electrical and Electronic Engineering ,Composite material ,business - Abstract
In this paper, we present a stretchable microelectrode array for studying cell behavior under mechanical strain. The electrode array consists of gold-plated nail-head pins (250 mu m tip diameter) or tungsten micro-wires (25.4 mu m in diameter) inserted into a polydimethylsiloxane (PDMS) platform (25.4 x 25.4 mm(2)). Stretchable interconnects to the outside were provided by fusible indium-alloy-filled microchannels. The alloy is liquid at room temperature, thus providing the necessary stretchability and electrical conductivity. The electrode platform can withstand strains of up to 40% and repeated (100 times) strains of up to 35% did not cause any failure in the electrodes or the PDMS substrate. We confirmed biocompatibility of short-term culture, and using the gold pin device, we demonstrated electric field pacing of adult murine heart cells. Further, using the tungsten microelectrode device, we successfully measured depolarizations of differentiated murine heart cells from embryoid body clusters.
- Published
- 2011
49. Electrophysiological Modeling of Channelrhodophsin-2 in Cardiac Cells
- Author
-
Rohit Prakash, Oscar J. Abilez, Karl Deisseroth, Ellen Kuhl, Jonathan Wong, and Christopher K. Zarins
- Subjects
Computer science ,fungi ,Cell ,Biophysics ,Nanotechnology ,Ion current ,Embryonic stem cell ,Photostimulation ,Electrophysiology ,medicine.anatomical_structure ,nervous system ,medicine ,Ventricular cell ,Neuron ,Neuroscience ,Ion channel - Abstract
Purpose: With the recent interest in Channelrhodopshin-2 (Chr2) in neurological experiments, researchers have begun to investigate the utility of light-activated ion channels in other electrically active cell types, including human embryonic stem cell-derived cardiomyocytes (Abilez et al. 2010). However, the impact of Chr2 in action potential synchronization in cardiac cells is not yet fully realized, as neuronal and cardiac cells differ in electrical behavior. In the past, baseline electrophysiological models for normal neuronal and cardiac cells have been developed and recent attempts have been made to characterize Chr2 in neuron excitation control. However, these approaches do not capture the resulting ion channel current, nor have they been adapted for cardiac cells. By characterizing Chr2 currents within existing cell models, simulations can be conducted concurrently with experiments for principle validation and experiment optimization.Methods: A kinetic model for Chr2 activation (Nikolic et al. 2006) was extended to an ion current formulation from current-voltage comparisons in the literature. This current was introduced into a ventricular cell model (ten Tusscher et al. 2003) and embedded in an implicit non-linear finite element framework (Wong et al. 2010) to perform simulations at cellular, tissue, and organ levels.Results: To illustrate the features of our novel light-activated cell model, we present selected examples to show the benefits of concurrent modeling. At the cellular level, we explore the impact of photostimulation strength, duration, and frequency in Chr2-manipulated ventricular cells. At the tissue level, we evaluate the feasibility of using such manipulated cells as pacemakers in the heart.Conclusion: By “transducing” cell models with Chr2, we can not only virtually probe characteristics of light-activated functional cells for novel applications of Chr2 in cardiac cells and other electrically active cells, but also optimize experiments by qualitatively predicting experimental results.
- Published
- 2011
- Full Text
- View/download PDF
50. In vitro and In silico Optogenetic Control of Differentiated Human Pluripotent Stem Cells
- Author
-
Karl Deisseroth, Rohit Prakash, Jonathan Wong, Ellen Kuhl, Christopher K. Zarins, and Oscar J. Abilez
- Subjects
Yellow fluorescent protein ,biology ,medicine.diagnostic_test ,In silico ,fungi ,Biophysics ,Video microscopy ,Embryonic stem cell ,Molecular biology ,Photostimulation ,Cell biology ,Flow cytometry ,Directed differentiation ,biology.protein ,medicine ,Induced pluripotent stem cell - Abstract
Purpose: Despite the use of a variety of differentiation protocols, no method exists to prospectively generate specific phenotypes of human embryonic stem cell (hESC)-derived cardiomyocytes (CM) such as pacemaker cells. We introduced light-activated channelrhodopsin-2 (ChR2) into hESC, and by using in vitro and in silico approaches, we were able to optogenetically synchronize hESC-CM, both experimentally and computationally.Methods: We experimentally introduced ChR2 coupled to yellow fluorescent protein (YFP) into undifferentiatedhESC via a lentiviral vector and tested for expression via PCR, flow cytometry (FC), and immunocytochemistry (ICC). hESCChR2+ were sorted, expanded, and tested for pluripotency. Via directed differentiation, wildtype hESC-CM and ChR2-CM were produced and subjected to both electrical and optical stimulation. Electrical, biochemical, and mechanical signals were then assessed by patch clamping, multielectrode arrays (MEAs), and video microscopy. To complement our in vitro approach with in silico analyses, we introduced ChR2 into an ionic cardiac cell model.Results: ChR2 was stably transduced into undifferentiated hESC and the resulting hESCChR2+ pluripotent line could be differentiated into CM, all confirmed by PCR, FC, ICC, and electrophysiological methods. Both WT-CM and ChR2-CM responded to traditional electrical stimulation and produced similar calcium and contractility features but only ChR2-CM could be synchronized by optical stimulation. In addition, by calibrating our ionic cell model with single cell action potential (AP) readings, we were able to virtually probe the impact of photostimulation stimulus amplitude, pulse width, and frequency on overall AP characteristics.Conclusions: Here we show for the first time that ChR2 can enable in vitro and in silico optical control of hESC-CM. The long-term application of optical stimulation could potentially lead to specific synchronous phenotypes of hESC-CM. This, in turn, would contribute significantly towards creating effective therapies for cardiovascular disease.
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.