Network


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

Hotspot


Dive into the research topics where Nathaniel Huebsch is active.

Publication


Featured researches published by Nathaniel Huebsch.


Scientific Reports | 2015

Human iPSC-based Cardiac Microphysiological System For Drug Screening Applications

Anurag Mathur; Peter Loskill; Kaifeng Shao; Nathaniel Huebsch; SoonGweon Hong; Sivan G. Marcus; Natalie C. Marks; Mohammad A. Mandegar; Bruce R. Conklin; Luke P. Lee; Kevin E. Healy

Drug discovery and development are hampered by high failure rates attributed to the reliance on non-human animal models employed during safety and efficacy testing. A fundamental problem in this inefficient process is that non-human animal models cannot adequately represent human biology. Thus, there is an urgent need for high-content in vitro systems that can better predict drug-induced toxicity. Systems that predict cardiotoxicity are of uppermost significance, as approximately one third of safety-based pharmaceutical withdrawals are due to cardiotoxicty. Here, we present a cardiac microphysiological system (MPS) with the attributes required for an ideal in vitro system to predict cardiotoxicity: i) cells with a human genetic background; ii) physiologically relevant tissue structure (e.g. aligned cells); iii) computationally predictable perfusion mimicking human vasculature; and, iv) multiple modes of analysis (e.g. biological, electrophysiological, and physiological). Our MPS is able to keep human induced pluripotent stem cell derived cardiac tissue viable and functional over multiple weeks. Pharmacological studies using the cardiac MPS show half maximal inhibitory/effective concentration values (IC50/EC50) that are more consistent with the data on tissue scale references compared to cellular scale studies. We anticipate the widespread adoption of MPSs for drug screening and disease modeling.


Cell Stem Cell | 2016

CRISPR Interference Efficiently Induces Specific and Reversible Gene Silencing in Human iPSCs

Mohammad A. Mandegar; Nathaniel Huebsch; Frolov Eb; Shin E; Annie Truong; Michael P. Olvera; Amanda H. Chan; Yuichiro Miyaoka; Holmes K; Spencer Ci; Luke M. Judge; David E. Gordon; Tilde Eskildsen; Jacqueline E. Villalta; Max A. Horlbeck; Luke A. Gilbert; Nevan J. Krogan; Søren Paludan Sheikh; Jonathan S. Weissman; Lei S. Qi; Po-Lin So; Bruce R. Conklin

Developing technologies for efficient and scalable disruption of gene expression will provide powerful tools for studying gene function, developmental pathways, and disease mechanisms. Here, we develop clustered regularly interspaced short palindromic repeat interference (CRISPRi) to repress gene expression in human induced pluripotent stem cells (iPSCs). CRISPRi, in which a doxycycline-inducible deactivated Cas9 is fused to a KRAB repression domain, can specifically and reversibly inhibit gene expression in iPSCs and iPSC-derived cardiac progenitors, cardiomyocytes, and T lymphocytes. This gene repression system is tunable and has the potential to silence single alleles. Compared with CRISPR nuclease (CRISPRn), CRISPRi gene repression is more efficient and homogenous across cell populations. The CRISPRi system in iPSCs provides a powerful platform to perform genome-scale screens in a wide range of iPSC-derived cell types, dissect developmental pathways, and model disease.


Biomaterials | 2014

Three-dimensional filamentous human diseased cardiac tissue model.

Zhen Ma; Sangmo Koo; Micaela A. Finnegan; Peter Loskill; Nathaniel Huebsch; Natalie C. Marks; Bruce R. Conklin; Costas P. Grigoropoulos; Kevin E. Healy

A human in vitro cardiac tissue model would be a significant advancement for understanding, studying, and developing new strategies for treating cardiac arrhythmias and related cardiovascular diseases. We developed an in vitro model of three-dimensional (3D) human cardiac tissue by populating synthetic filamentous matrices with cardiomyocytes derived from healthy wild-type volunteer (WT) and patient-specific long QT syndrome type 3 (LQT3) induced pluripotent stem cells (iPS-CMs) to mimic the condensed and aligned human ventricular myocardium. Using such a highly controllable cardiac model, we studied the contractility malfunctions associated with the electrophysiological consequences of LQT3 and their response to a panel of drugs. By varying the stiffness of filamentous matrices, LQT3 iPS-CMs exhibited different level of contractility abnormality and susceptibility to drug-induced cardiotoxicity.


Scientific Reports | 2016

Miniaturized iPS-Cell-Derived Cardiac Muscles for Physiologically Relevant Drug Response Analyses

Nathaniel Huebsch; Peter Loskill; Nikhil Deveshwar; C. Ian Spencer; Luke M. Judge; Mohammad A. Mandegar; Cade B. Fox; Tamer M.A. Mohamed; Zhen Ma; Anurag Mathur; Alice M. Sheehan; Annie Truong; Mike Saxton; Jennie Yoo; Deepak Srivastava; Tejal A. Desai; Po Lin So; Kevin E. Healy; Bruce R. Conklin

Tissue engineering approaches have the potential to increase the physiologic relevance of human iPS-derived cells, such as cardiomyocytes (iPS-CM). However, forming Engineered Heart Muscle (EHM) typically requires >1 million cells per tissue. Existing miniaturization strategies involve complex approaches not amenable to mass production, limiting the ability to use EHM for iPS-based disease modeling and drug screening. Micro-scale cardiospheres are easily produced, but do not facilitate assembly of elongated muscle or direct force measurements. Here we describe an approach that combines features of EHM and cardiospheres: Micro-Heart Muscle (μHM) arrays, in which elongated muscle fibers are formed in an easily fabricated template, with as few as 2,000 iPS-CM per individual tissue. Within μHM, iPS-CM exhibit uniaxial contractility and alignment, robust sarcomere assembly, and reduced variability and hypersensitivity in drug responsiveness, compared to monolayers with the same cellular composition. μHM mounted onto standard force measurement apparatus exhibited a robust Frank-Starling response to external stretch, and a dose-dependent inotropic response to the β-adrenergic agonist isoproterenol. Based on the ease of fabrication, the potential for mass production and the small number of cells required to form μHM, this system provides a potentially powerful tool to study cardiomyocyte maturation, disease and cardiotoxicology in vitro.


Nature Communications | 2015

Self-organizing human cardiac microchambers mediated by geometric confinement

Zhen Ma; Jason J. Wang; Peter Loskill; Nathaniel Huebsch; Sangmo Koo; Felicia L. Svedlund; Natalie C. Marks; Ethan W. Hua; Costas P. Grigoropoulos; Bruce R. Conklin; Kevin E. Healy

Tissue morphogenesis and organ formation are the consequences of biochemical and biophysical cues that lead to cellular spatial patterning in development. To model such events in vitro, we use PEG-patterned substrates to geometrically confine human pluripotent stem cell colonies and spatially present mechanical stress. Modulation of the WNT/β-catenin pathway promotes spatial patterning via geometric confinement of the cell condensation process during epithelial–mesenchymal transition, forcing cells at the perimeter to express an OCT4+ annulus, which is coincident with a region of higher cell density and E-cadherin expression. The biochemical and biophysical cues synergistically induce self-organizing lineage specification and creation of a beating human cardiac microchamber confined by the pattern geometry. These highly defined human cardiac microchambers can be used to study aspects of embryonic spatial patterning, early cardiac development and drug-induced developmental toxicity.


JCI insight | 2017

A BAG3 chaperone complex maintains cardiomyocyte function during proteotoxic stress

Luke M. Judge; Juan A. Perez-Bermejo; Annie Truong; Alexandre J.S. Ribeiro; Jennie C. Yoo; Christina L. Jensen; Mohammad A. Mandegar; Nathaniel Huebsch; Robyn M. Kaake; Po-Lin So; Deepak Srivastava; Beth L. Pruitt; Nevan J. Krogan; Bruce R. Conklin

Molecular chaperones regulate quality control in the human proteome, pathways that have been implicated in many diseases, including heart failure. Mutations in the BAG3 gene, which encodes a co-chaperone protein, have been associated with heart failure due to both inherited and sporadic dilated cardiomyopathy. Familial BAG3 mutations are autosomal dominant and frequently cause truncation of the coding sequence, suggesting a heterozygous loss-of-function mechanism. However, heterozygous knockout of the murine BAG3 gene did not cause a detectable phenotype. To model BAG3 cardiomyopathy in a human system, we generated an isogenic series of human induced pluripotent stem cells (iPSCs) with loss-of-function mutations in BAG3. Heterozygous BAG3 mutations reduced protein expression, disrupted myofibril structure, and compromised contractile function in iPSC-derived cardiomyocytes (iPS-CMs). BAG3-deficient iPS-CMs were particularly sensitive to further myofibril disruption and contractile dysfunction upon exposure to proteasome inhibitors known to cause cardiotoxicity. We performed affinity tagging of the endogenous BAG3 protein and mass spectrometry proteomics to further define the cardioprotective chaperone complex that BAG3 coordinates in the human heart. Our results establish a model for evaluating protein quality control pathways in human cardiomyocytes and their potential as therapeutic targets and susceptibility factors for cardiac drug toxicity.


bioRxiv | 2018

Inversion and computational maturation of drug response using human stem cell derived cardiomyocytes in microphysiological systems

Aslak Tveito; Karoline H. Jæger; Nathaniel Huebsch; Berenice Charrez; Andrew G. Edwards; Samuel T. Wall; Kevin E. Healy

While cardiomyocytes differentiated from human induced pluripotent stems cells (hiPSCs) hold great promise for drug screening, the electrophys-iological properties of these cells can be variable and immature, producing results that are significantly different from their human adult counterparts. Here, we describe a computational framework to address this limitation, and show how in silico methods, applied to measurements on immature cardiomyocytes, can be used to both identify drug action and to predict its effect in mature cells. Our synthetic and experimental results indicate that optically obtained waveforms of voltage and calcium from microphysiological systems can be inverted into information on drug ion channel blockage, and then, through assuming functional invariance of proteins during maturation, this data can be used to predict drug induced changes in mature ventricular cells. Together, this pipeline of measurements and computational analysis could significantly improve the ability of hiPSC derived cardiomycocytes to predict dangerous drug side effects.


Nature Biomedical Engineering | 2018

Contractile deficits in engineered cardiac microtissues as a result of MYBPC3 deficiency and mechanical overload

Zhen Ma; Nathaniel Huebsch; Sangmo Koo; Mohammad A. Mandegar; Brian Siemons; Steven Boggess; Bruce R. Conklin; Costas P. Grigoropoulos; Kevin E. Healy

The integration of in vitro cardiac tissue models, human induced pluripotent stem cells (hiPSCs) and genome-editing tools allows for the enhanced interrogation of physiological phenotypes and recapitulation of disease pathologies. Here, using a cardiac tissue model consisting of filamentous three-dimensional matrices populated with cardiomyocytes derived from healthy wild-type (WT) hiPSCs (WT hiPSC-CMs) or isogenic hiPSCs deficient in the sarcomere protein cardiac myosin-binding protein C (MYBPC3–/– hiPSC-CMs), we show that the WT microtissues adapted to the mechanical environment with increased contraction force commensurate to matrix stiffness, whereas the MYBPC3–/– microtissues exhibited impaired force development kinetics regardless of matrix stiffness and deficient contraction force only when grown on matrices with high fibre stiffness. Under mechanical overload, the MYBPC3–/– microtissues had a higher degree of calcium transient abnormalities, and exhibited an accelerated decay of calcium dynamics as well as calcium desensitization, which accelerated when contracting against stiffer fibres. Our findings suggest that MYBPC3 deficiency and the presence of environmental stresses synergistically lead to contractile deficits in cardiac tissues.Cardiac tissue engineered to enable the modulation of mechanical resistance to tissue contraction facilitates the modelling of genetic pathologies associated with the absence of a thick-filament accessory protein found in striated heart muscle.


Biotechnology and Bioengineering | 2018

Quantitatively Characterizing Drug-Induced Arrhythmic Contractile Motions of Human Stem Cell-Derived Cardiomyocytes.

Plansky Hoang; Nathaniel Huebsch; Shin Hyuk Bang; Brian Siemons; Bruce R. Conklin; Kevin E. Healy; Zhen Ma; Sabir Jacquir

Quantification of abnormal contractile motions of cardiac tissue has been a noteworthy challenge and significant limitation in assessing and classifying the drug-induced arrhythmias (i.e. Torsades de pointes). To overcome these challenges, researchers have taken advantage of computational image processing tools to measure contractile motion from cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs). However, the amplitude and frequency analysis of contractile motion waveforms doesnt produce sufficient information to objectively classify the degree of variations between two or more sets of cardiac contractile motions. In this paper, we generated contractile motion data from beating hiPSC-CMs using motion tracking software based on optical flow analysis, and then implemented a computational algorithm, phase space reconstruction (PSR), to derive parameters (embedding, regularity, and fractal dimensions) to further characterize the dynamic nature of the cardiac contractile motions. Application of drugs known to cause cardiac arrhythmia induced significant changes to these resultant dimensional parameters calculated from PSR analysis. Integrating this new computational algorithm with the existing analytical toolbox of cardiac contractile motions will allow us to expand current assessments of cardiac tissue physiology into an automated, high-throughput, and quantifiable manner which will allow more objective assessments of drug-induced proarrhythmias. This article is protected by copyright. All rights reserved.


Tissue Engineering Part C-methods | 2015

Automated Video-Based Analysis of Contractility and Calcium Flux in Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes Cultured over Different Spatial Scales

Nathaniel Huebsch; Peter Loskill; Mohammad A. Mandegar; Natalie C. Marks; Alice Sheehan; Zhen Ma; Anurag Mathur; Trieu N. Nguyen; Jennie C. Yoo; Luke M. Judge; C. Ian Spencer; Anand C. Chukka; Caitlin R. Russell; Po Lin So; Bruce R. Conklin; Kevin E. Healy

Collaboration


Dive into the Nathaniel Huebsch's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kevin E. Healy

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Luke M. Judge

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Peter Loskill

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

C. Ian Spencer

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge