Network


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

Hotspot


Dive into the research topics where Praveen Shukla is active.

Publication


Featured researches published by Praveen Shukla.


Nature Methods | 2014

Chemically defined generation of human cardiomyocytes

Paul W. Burridge; Elena Matsa; Praveen Shukla; Ziliang C Lin; Jared M. Churko; Antje D. Ebert; Feng Lan; Sebastian Diecke; Bruno C. Huber; Nicholas M. Mordwinkin; Jordan R. Plews; Oscar J. Abilez; Bianxiao Cui; Joseph D. Gold; Joseph C. Wu

Existing methods for human induced pluripotent stem cell (hiPSC) cardiac differentiation are efficient but require complex, undefined medium constituents that hinder further elucidation of the molecular mechanisms of cardiomyogenesis. Using hiPSCs derived under chemically defined conditions on synthetic matrices, we systematically developed an optimized cardiac differentiation strategy, using a chemically defined medium consisting of just three components: the basal medium RPMI 1640, L-ascorbic acid 2-phosphate and rice-derived recombinant human albumin. Along with small molecule–based induction of differentiation, this protocol produced contractile sheets of up to 95% TNNT2+ cardiomyocytes at a yield of up to 100 cardiomyocytes for every input pluripotent cell and was effective in 11 hiPSC lines tested. This chemically defined platform for cardiac specification of hiPSCs will allow the elucidation of cardiomyocyte macromolecular and metabolic requirements and will provide a minimal system for the study of maturation and subtype specification.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 ricederived 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 Users may view, print, copy, and download text and data-mine the content in such documents, for the purposes of academic research, subject always to the full Conditions of use:http://www.nature.com/authors/editorial_policies/license.html#terms Addresses for Correspondence: Joseph C. Wu, MD, PhD, Stanford University School of Medicine, Lorry I. Lokey Stem Cell Research Building, 265 Campus Drive, Room G1120B, Stanford, CA 94305-5454. [email protected] or Paul W. Burridge, PhD, [email protected]. Author Contributions P.W.B. conceived, performed, and interpreted the experiments and wrote the manuscript; E.M. performed cardiomyocyte immunofluorescence, single-cell RT-PCR, and electrophysiology data assessment; P.S., Z.L., and A.J.O. performed electrophysiology experiments and assessed data; S.D. provided CoMiP reprogrammed cells; B.H. performed teratoma assay; J.M.C. A.D.E, F.L., N.M.M., and J.R.P tested differentiation; B.C., J.D.G. provided experimental advice; and J.C.W. provided experimental advice, manuscript writing, and funding support. Competing Financial Interests JCW is a co-founder of Stem Cell Theranostics. Other authors declare no competing financial interests. HHS Public Access Author manuscript Nat Methods. Author manuscript; available in PMC 2015 February 01. Published in final edited form as: Nat Methods. 2014 August ; 11(8): 855–860. doi:10.1038/nmeth.2999. A uhor M anscript


Advanced Drug Delivery Reviews | 2016

Engineered heart tissues and induced pluripotent stem cells: Macro- and microstructures for disease modeling, drug screening, and translational studies.

Evangeline Tzatzalos; Oscar J. Abilez; Praveen Shukla; Joseph C. Wu

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.


Science Translational Medicine | 2017

High-throughput screening of tyrosine kinase inhibitor cardiotoxicity with human induced pluripotent stem cells

Arun Sharma; Paul W. Burridge; Wesley L. McKeithan; Ricardo Serrano; Praveen Shukla; Nazish Sayed; Jared M. Churko; Tomoya Kitani; Haodi Wu; Alexandra Holmström; Elena Matsa; Yuan Zhang; Anusha Kumar; Alice C. Fan; Juan C. del Álamo; Sean M. Wu; Javid Moslehi; Mark Mercola; Joseph C. Wu

High-throughput screening of drugs with human induced pluripotent stem cell–derived cardiomyocytes reveals a “cardiac safety index.” Failing fast for tyrosine kinase inhibitors Discovery early in its life cycle that an anticancer drug causes heart damage (a common side effect) can halt development—saving money, time, and perhaps lives. To this end, Sharma and colleagues derived heart cells from human induced pluripotent stem cells and then examined how a battery of anticancer tyrosine kinase inhibitors altered their physiology. By measuring cell death, contraction, excitability, calcium dynamics, and signal transduction and integrating the results, they calculated a drug-specific “cardiac safety index.” This index proved highly informative, with low values corresponding to those drugs known to cause heart problems in patients. The analysis even revealed that VEGFR2-inhibiting drugs caused cells to try to compensate for the toxic effects by up-regulating protective insulin/IGF pathways, prompting the authors to devise a combination treatment that may limit the toxicity of this class of drug. This screening method is expected to reveal early on whether potential anticancer drugs are cardiotoxic. Tyrosine kinase inhibitors (TKIs), despite their efficacy as anticancer therapeutics, are associated with cardiovascular side effects ranging from induced arrhythmias to heart failure. We used human induced pluripotent stem cell–derived cardiomyocytes (hiPSC-CMs), generated from 11 healthy individuals and 2 patients receiving cancer treatment, to screen U.S. Food and Drug Administration–approved TKIs for cardiotoxicities by measuring alterations in cardiomyocyte viability, contractility, electrophysiology, calcium handling, and signaling. With these data, we generated a “cardiac safety index” to reflect the cardiotoxicities of existing TKIs. TKIs with low cardiac safety indices exhibit cardiotoxicity in patients. We also derived endothelial cells (hiPSC-ECs) and cardiac fibroblasts (hiPSC-CFs) to examine cell type–specific cardiotoxicities. Using high-throughput screening, we determined that vascular endothelial growth factor receptor 2 (VEGFR2)/platelet-derived growth factor receptor (PDGFR)–inhibiting TKIs caused cardiotoxicity in hiPSC-CMs, hiPSC-ECs, and hiPSC-CFs. With phosphoprotein analysis, we determined that VEGFR2/PDGFR-inhibiting TKIs led to a compensatory increase in cardioprotective insulin and insulin-like growth factor (IGF) signaling in hiPSC-CMs. Up-regulating cardioprotective signaling with exogenous insulin or IGF1 improved hiPSC-CM viability during cotreatment with cardiotoxic VEGFR2/PDGFR-inhibiting TKIs. Thus, hiPSC-CMs can be used to screen for cardiovascular toxicities associated with anticancer TKIs, and the results correlate with clinical phenotypes. This approach provides unexpected insights, as illustrated by our finding that toxicity can be alleviated via cardioprotective insulin/IGF signaling.


Stem Cell Research | 2015

Development of a scalable suspension culture for cardiac differentiation from human pluripotent stem cells

Vincent C. Chen; Jingjing Ye; Praveen Shukla; Giau Hua; Danlin Chen; Ziguang Lin; Jian-chang Liu; Jing Chai; Joseph D. Gold; Joseph C. Wu; David Hsu; Larry A. Couture

To meet the need of a large quantity of hPSC-derived cardiomyocytes (CM) for pre-clinical and clinical studies, a robust and scalable differentiation system for CM production is essential. With a human pluripotent stem cells (hPSC) aggregate suspension culture system we established previously, we developed a matrix-free, scalable, and GMP-compliant process for directing hPSC differentiation to CM in suspension culture by modulating Wnt pathways with small molecules. By optimizing critical process parameters including: cell aggregate size, small molecule concentrations, induction timing, and agitation rate, we were able to consistently differentiate hPSCs to >90% CM purity with an average yield of 1.5 to 2×10(9) CM/L at scales up to 1L spinner flasks. CM generated from the suspension culture displayed typical genetic, morphological, and electrophysiological cardiac cell characteristics. This suspension culture system allows seamless transition from hPSC expansion to CM differentiation in a continuous suspension culture. It not only provides a cost and labor effective scalable process for large scale CM production, but also provides a bioreactor prototype for automation of cell manufacturing, which will accelerate the advance of hPSC research towards therapeutic applications.


Stem Cells | 2017

Comparison of Non‐Coding RNAs in Exosomes and Functional Efficacy of Human Embryonic Stem Cell‐ versus Induced Pluripotent Stem Cell‐Derived Cardiomyocytes

Won Hee Lee; Wen Yi Chen; Ning Yi Shao; Dan Xiao; Xulei Qin; Natalie Baker; Hye Ryeong Bae; Tzu Tang Wei; Yongjun Wang; Praveen Shukla; Haodi Wu; Kazuki Kodo; Sang Ging Ong; Joseph C. Wu

Both human embryonic stem cell‐derived cardiomyocytes (ESC‐CMs) and human induced pluripotent stem cell‐derived CMs (iPSC‐CMs) can serve as unlimited cell sources for cardiac regenerative therapy. However, the functional equivalency between human ESC‐CMs and iPSC‐CMs for cardiac regenerative therapy has not been demonstrated. Here, we performed a head‐to‐head comparison of ESC‐CMs and iPSC‐CMs in their ability to restore cardiac function in a rat myocardial infarction (MI) model as well as their exosomal secretome. Human ESCs and iPSCs were differentiated into CMs using small molecule inhibitors. Fluorescence‐activated cell sorting analysis confirmed ∼85% and ∼83% of CMs differentiated from ESCs and iPSCs, respectively, were positive for cardiac troponin T. At a single‐cell level, both cell types displayed similar calcium handling and electrophysiological properties, with gene expression comparable with the human fetal heart marked by striated sarcomeres. Sub‐acute transplantation of ESC‐CMs and iPSC‐CMs into nude rats post‐MI improved cardiac function, which was associated with increased expression of angiogenic genes in vitro following hypoxia. Profiling of exosomal microRNAs (miRs) and long non‐coding RNAs (lncRNAs) revealed that both groups contain an identical repertoire of miRs and lncRNAs, including some that are known to be cardioprotective. We demonstrate that both ESC‐CMs and iPSC‐CMs can facilitate comparable cardiac repair. This is advantageous because, unlike allogeneic ESC‐CMs used in therapy, autologous iPSC‐CMs could potentially avoid immune rejection when used for cardiac cell transplantation in the future. Stem Cells 2017;35:2138–2149


Circulation Research | 2017

Paracrine Effects of the Pluripotent Stem Cell-Derived Cardiac Myocytes Salvage the Injured MyocardiumNovelty and Significance

Atsushi Tachibana; Michelle R. Santoso; Morteza Mahmoudi; Praveen Shukla; Lei Wang; Mihoko V. Bennett; Andrew B. Goldstone; Mouer Wang; Masahiro Fukushi; Antje D. Ebert; Y. Joseph Woo; Eric Rulifson; Phillip C. Yang

Rationale: Cardiac myocytes derived from pluripotent stem cells have demonstrated the potential to mitigate damage of the infarcted myocardium and improve left ventricular ejection fraction. However, the mechanism underlying the functional benefit is unclear. Objective: To evaluate whether the transplantation of cardiac-lineage differentiated derivatives enhance myocardial viability and restore left ventricular ejection fraction more effectively than undifferentiated pluripotent stem cells after a myocardial injury. Herein, we utilize novel multimodality evaluation of human embryonic stem cells (hESCs), hESC-derived cardiac myocytes (hCMs), human induced pluripotent stem cells (iPSCs), and iPSC-derived cardiac myocytes (iCMs) in a murine myocardial injury model. Methods and Results: Permanent ligation of the left anterior descending coronary artery was induced in immunosuppressed mice. Intramyocardial injection was performed with (1) hESCs (n=9), (2) iPSCs (n=8), (3) hCMs (n=9), (4) iCMs (n=14), and (5) PBS control (n=10). Left ventricular ejection fraction and myocardial viability, measured by cardiac magnetic resonance imaging and manganese-enhanced magnetic resonance imaging, respectively, was significantly improved in hCM- and iCM-treated mice compared with pluripotent stem cell- or control-treated mice. Bioluminescence imaging revealed limited cell engraftment in all treated groups, suggesting that the cell secretions may underlie the repair mechanism. To determine the paracrine effects of the transplanted cells, cytokines from supernatants from all groups were assessed in vitro. Gene expression and immunohistochemistry analyses of the murine myocardium demonstrated significant upregulation of the promigratory, proangiogenic, and antiapoptotic targets in groups treated with cardiac lineage cells compared with pluripotent stem cell and control groups. Conclusions: This study demonstrates that the cardiac phenotype of hCMs and iCMs salvages the injured myocardium effectively than undifferentiated stem cells through their differential paracrine effects.


Stem Cells | 2018

Passive Stretch Induces Structural and Functional Maturation of Engineered Heart Muscle as Predicted by Computational Modeling

Oscar J. Abilez; Evangeline Tzatzalos; Huaxiao Yang; Ming Tao Zhao; Gwanghyun Jung; Alexander M. Zöllner; Malte Tiburcy; Johannes Riegler; Elena Matsa; Praveen Shukla; Yan Zhuge; Tony Chour; Vincent Cw Chen; Paul W. Burridge; Ioannis Karakikes; Ellen Kuhl; Daniel Bernstein; Larry A. Couture; Joseph D. Gold; Wolfram H. Zimmermann; Joseph C. Wu

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. Stem Cells 2018;36:265–277


Scientific Reports | 2017

Partial Reprogramming of Pluripotent Stem Cell-Derived Cardiomyocytes into Neurons

Wenpo Chuang; Arun Sharma; Praveen Shukla; Guang Li; Moritz Mall; Kuppusamy Rajarajan; Oscar J. Abilez; Ryoko Hamaguchi; Joseph C. Wu; Marius Wernig; Sean M. Wu

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.


Stem cell reports | 2018

Comparison of Non-human Primate versus Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes for Treatment of Myocardial Infarction

Xin Zhao; Haodong Chen; Dan Xiao; Huaxiao Yang; Ilanit Itzhaki; Xulei Qin; Tony Chour; Aitor Aguirre; Kim A. Lehmann; Youngkyun Kim; Praveen Shukla; Alexandra Holmström; Joe Z. Zhang; Yan Zhuge; Babacar Ndoye; Ming-Tao Zhao; Evgenios Neofytou; Wolfram-Hubertus Zimmermann; Mohit Jain; Joseph C. Wu

Summary Non-human primates (NHPs) can serve as a human-like model to study cell therapy using induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). However, whether the efficacy of NHP and human iPSC-CMs is mechanistically similar remains unknown. To examine this, RNU rats received intramyocardial injection of 1 × 107 NHP or human iPSC-CMs or the same number of respective fibroblasts or PBS control (n = 9–14/group) at 4 days after 60-min coronary artery occlusion-reperfusion. Cardiac function and left ventricular remodeling were similarly improved in both iPSC-CM-treated groups. To mimic the ischemic environment in the infarcted heart, both cultured NHP and human iPSC-CMs underwent 24-hr hypoxia in vitro. Both cells and media were collected, and similarities in transcriptomic as well as metabolomic profiles were noted between both groups. In conclusion, both NHP and human iPSC-CMs confer similar cardioprotection in a rodent myocardial infarction model through relatively similar mechanisms via promotion of cell survival, angiogenesis, and inhibition of hypertrophy and fibrosis.


European Heart Journal | 2016

Cardiac subtype characterization using all-optical action potential imaging

Praveen Shukla; Joseph C. Wu

This editorial refers to ‘Subtype-specific promoter-driven action potential imaging for precise disease modelling and drug testing in human induced pluripotent stem cell-derived cardiomyocytes’, by Z. Chen et al. doi: 10.1093/eurheartj/ehw189. Cellular modelling of human heart disease has been hampered by several limitations, including difficult accessibility of human cardiac tissues and low proliferative capacity of freshly dissociated human cardiomyocytes (CMs). The advent of human pluripotent stem cells (hPSCs), which include both human embryonic stem cells (ESCs) and human induced pluripotent stem cells (iPSCs), has provided an unprecedented opportunity to generate potentially limitless quantities of hPSC-derived somatic cell types (including human CMs) for studying disease mechanisms, identifying novel drug targets, and accelerating drug screening.1 However, two major limitations must be overcome before such wide-ranging application of iPSC-CM technology is possible. The first problem is that current cardiac differentiation methods produce a heterogeneous population of ventricular-, atrial-, and nodal-like cells.2–4 Despite efforts to produce a more enriched population of a specific cardiac phenotype to model specific cardiac diseases (e.g. atrial fibrillation, ventricular arrhythmias, etc.), no effective and validated strategies are yet available. A second limitation is that iPSC-CMs are assessed by low-throughput functional assays, primarily the labour-intensive patch-clamp electrophysiology, which considerably slows the pace of identification and characterization of the disease and/or drug-induced cellular phenotypes. Therefore, there is an urgent need to develop sensitive, scalable, and high-throughput functional assays to capitalize fully on our ability to generate iPSC-CMs at an industrial scale.5,6 Over the past two decades, membrane voltage-sensitive dyes (VSDs) have been used as an alternative to patch-clamp electrophysiology to record both cardiac action potential (AP) and calcium transients. However, the use of VSDs in AP recordings is limited by the indiscriminate cell staining and dye-mediated acute phototoxicity, especially in the setting of repeated single-cell measurement. …

Collaboration


Dive into the Praveen Shukla's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jay N. Sharma

University of Louisville

View shared research outputs
Researchain Logo
Decentralizing Knowledge