Network


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

Hotspot


Dive into the research topics where Harsha D. Devalla is active.

Publication


Featured researches published by Harsha D. Devalla.


Embo Molecular Medicine | 2015

Atrial-like cardiomyocytes from human pluripotent stem cells are a robust preclinical model for assessing atrial-selective pharmacology

Harsha D. Devalla; Verena Schwach; John W Ford; James Milnes; Said El-Haou; Claire Jackson; Konstantinos Gkatzis; David A. Elliott; Susana Lopes; Arie O. Verkerk; Robert Passier

Drugs targeting atrial‐specific ion channels, Kv1.5 or Kir3.1/3.4, are being developed as new therapeutic strategies for atrial fibrillation. However, current preclinical studies carried out in non‐cardiac cell lines or animal models may not accurately represent the physiology of a human cardiomyocyte (CM). In the current study, we tested whether human embryonic stem cell (hESC)‐derived atrial CMs could predict atrial selectivity of pharmacological compounds. By modulating retinoic acid signaling during hESC differentiation, we generated atrial‐like (hESC‐atrial) and ventricular‐like (hESC‐ventricular) CMs. We found the expression of atrial‐specific ion channel genes, KCNA5 (encoding Kv1.5) and KCNJ3 (encoding Kir 3.1), in hESC‐atrial CMs and further demonstrated that these ion channel genes are regulated by COUP‐TF transcription factors. Moreover, in response to multiple ion channel blocker, vernakalant, and Kv1.5 blocker, XEN‐D0101, hESC‐atrial but not hESC‐ventricular CMs showed action potential (AP) prolongation due to a reduction in early repolarization. In hESC‐atrial CMs, XEN‐R0703, a novel Kir3.1/3.4 blocker restored the AP shortening caused by CCh. Neither CCh nor XEN‐R0703 had an effect on hESC‐ventricular CMs. In summary, we demonstrate that hESC‐atrial CMs are a robust model for pre‐clinical testing to assess atrial selectivity of novel antiarrhythmic drugs.


Nature Biotechnology | 2015

Expansion and patterning of cardiovascular progenitors derived from human pluripotent stem cells

Matthew J. Birket; Marcelo C. Ribeiro; Arie O. Verkerk; Dorien Ward; Ana Rita Leitoguinho; Sabine C. Den Hartogh; Valeria V. Orlova; Harsha D. Devalla; Verena Schwach; Milena Bellin; Robert Passier

The inability of multipotent cardiovascular progenitor cells (CPCs) to undergo multiple divisions in culture has precluded stable expansion of precursors of cardiomyocytes and vascular cells. This contrasts with neural progenitors, which can be expanded robustly and are a renewable source of their derivatives. Here we use human pluripotent stem cells bearing a cardiac lineage reporter to show that regulated MYC expression enables robust expansion of CPCs with insulin-like growth factor-1 (IGF-1) and a hedgehog pathway agonist. The CPCs can be patterned with morphogens, recreating features of heart field assignment, and controllably differentiated to relatively pure populations of pacemaker-like or ventricular-like cardiomyocytes. The cells are clonogenic and can be expanded for >40 population doublings while retaining the ability to differentiate into cardiomyocytes and vascular cells. Access to CPCs will allow precise recreation of elements of heart development in vitro and facilitate investigation of the molecular basis of cardiac fate determination. This technology is applicable for cardiac disease modeling, toxicology studies and tissue engineering.


Circulation-arrhythmia and Electrophysiology | 2011

Molecular analysis of patterning of conduction tissues in the developing human heart.

Aleksander Sizarov; Harsha D. Devalla; Robert H. Anderson; Robert Passier; Vincent M. Christoffels; Antoon F. M. Moorman

Background— Recent studies in experimental animals have revealed some molecular mechanisms underlying the differentiation of the myocardium making up the conduction system. To date, lack of gene expression data for the developing human conduction system has precluded valid extrapolations from experimental studies to the human situation. Methods and Results— We performed immunohistochemical analyses of the expression of key transcription factors, such as ISL1, TBX3, TBX18, and NKX2–5, ion channel HCN4, and connexins in the human embryonic heart. We supplemented our molecular analyses with 3-dimensional reconstructions of myocardial TBX3 expression. TBX3 is expressed in the developing conduction system and in the right venous valve, atrioventricular ring bundles, and retro-aortic nodal region. TBX3-positive myocardium, with exception of the top of the ventricular septum, is devoid of fast-conducting connexin40 and connexin43 and hence identifies slowly conducting pathways. In the early embryonic heart, we found wide expression of the pacemaker channel HCN4 at the venous pole, including the atrial chambers. HCN4 expression becomes confined during later developmental stages to the components of the conduction system. Patterns of expression of transcription factors, known from experimental studies to regulate the development of the sinus node and atrioventricular conduction system, are similar in the human and mouse developing hearts. Conclusions— Our findings point to the comparability of mechanisms governing the development of the cardiac conduction patterning in human and mouse, which provide a molecular basis for understanding the functioning of the human developing heart before formation of a discrete conduction system.


Cell Reports | 2015

Contractile Defect Caused by Mutation in MYBPC3 Revealed under Conditions Optimized for Human PSC-Cardiomyocyte Function

Matthew J. Birket; Marcelo C. Ribeiro; Georgios Kosmidis; Dorien Ward; Ana Rita Leitoguinho; Vera van de Pol; Cheryl Dambrot; Harsha D. Devalla; Richard P. Davis; Pier G. Mastroberardino; Douwe E. Atsma; Robert Passier

Summary Maximizing baseline function of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) is essential for their effective application in models of cardiac toxicity and disease. Here, we aimed to identify factors that would promote an adequate level of function to permit robust single-cell contractility measurements in a human induced pluripotent stem cell (hiPSC) model of hypertrophic cardiomyopathy (HCM). A simple screen revealed the collaborative effects of thyroid hormone, IGF-1 and the glucocorticoid analog dexamethasone on the electrophysiology, bioenergetics, and contractile force generation of hPSC-CMs. In this optimized condition, hiPSC-CMs with mutations in MYBPC3, a gene encoding myosin-binding protein C, which, when mutated, causes HCM, showed significantly lower contractile force generation than controls. This was recapitulated by direct knockdown of MYBPC3 in control hPSC-CMs, supporting a mechanism of haploinsufficiency. Modeling this disease in vitro using human cells is an important step toward identifying therapeutic interventions for HCM.


Stem cell reports | 2015

KeyGenes, a Tool to Probe Tissue Differentiation Using a Human Fetal Transcriptional Atlas

Matthias S Roost; Liesbeth van Iperen; Yavuz Ariyurek; Henk P. J. Buermans; Wibowo Arindrarto; Harsha D. Devalla; Robert Passier; Françoise Carlotti; Eelco J.P. de Koning; Erik W. van Zwet; Jelle J. Goeman; Susana Lopes

Summary Differentiated derivatives of human pluripotent stem cells in culture are generally phenotypically immature compared to their adult counterparts. Their identity is often difficult to determine with certainty because little is known about their human fetal equivalents in vivo. Cellular identity and signaling pathways directing differentiation are usually determined by extrapolating information from either human adult tissue or model organisms, assuming conservation with humans. To resolve this, we generated a collection of human fetal transcriptional profiles at different developmental stages. Moreover, we developed an algorithm, KeyGenes, which uses this dataset to quantify the extent to which next-generation sequencing or microarray data resemble specific cell or tissue types in the human fetus. Using KeyGenes combined with the human fetal atlas, we identified multiple cell and tissue samples unambiguously on a limited set of features. We thus provide a flexible and expandable platform to monitor and evaluate the efficiency of differentiation in vitro.


Embo Molecular Medicine | 2016

TECRL, a new life‐threatening inherited arrhythmia gene associated with overlapping clinical features of both LQTS and CPVT

Harsha D. Devalla; Roselle Gélinas; Elhadi H. Aburawi; Abdelaziz Beqqali; Philippe Goyette; Christian Freund; Marie-A. Chaix; Rafik Tadros; Hui Jiang; Antony Le Béchec; Jantine Monshouwer-Kloots; Tom Zwetsloot; Georgios Kosmidis; Frédéric Latour; Azadeh Alikashani; Maaike Hoekstra; Jurg Schlaepfer; Brian J. Stevenson; Zoltán Kutalik; Antoine A.F. de Vries; Lena Rivard; Arthur A.M. Wilde; Mario Talajic; Arie O. Verkerk; Lihadh Al-Gazali; John D. Rioux; Zahurul A. Bhuiyan; Robert Passier

Genetic causes of many familial arrhythmia syndromes remain elusive. In this study, whole‐exome sequencing (WES) was carried out on patients from three different families that presented with life‐threatening arrhythmias and high risk of sudden cardiac death (SCD). Two French Canadian probands carried identical homozygous rare variant in TECRL gene (p.Arg196Gln), which encodes the trans‐2,3‐enoyl‐CoA reductase‐like protein. Both patients had cardiac arrest, stress‐induced atrial and ventricular tachycardia, and QT prolongation on adrenergic stimulation. A third patient from a consanguineous Sudanese family diagnosed with catecholaminergic polymorphic ventricular tachycardia (CPVT) had a homozygous splice site mutation (c.331+1G>A) in TECRL. Analysis of intracellular calcium ([Ca2+]i) dynamics in human induced pluripotent stem cell‐derived cardiomyocytes (hiPSC‐CMs) generated from this individual (TECRLHom‐hiPSCs), his heterozygous but clinically asymptomatic father (TECRLHet‐hiPSCs), and a healthy individual (CTRL‐hiPSCs) from the same Sudanese family, revealed smaller [Ca2+]i transient amplitudes as well as elevated diastolic [Ca2+]i in TECRLHom‐hiPSC‐CMs compared with CTRL‐hiPSC‐CMs. The [Ca2+]i transient also rose markedly slower and contained lower sarcoplasmic reticulum (SR) calcium stores, evidenced by the decreased magnitude of caffeine‐induced [Ca2+]i transients. In addition, the decay phase of the [Ca2+]i transient was slower in TECRLHom‐hiPSC‐CMs due to decreased SERCA and NCX activities. Furthermore, TECRLHom‐hiPSC‐CMs showed prolonged action potentials (APs) compared with CTRL‐hiPSC‐CMs. TECRL knockdown in control human embryonic stem cell‐derived CMs (hESC‐CMs) also resulted in significantly longer APs. Moreover, stimulation by noradrenaline (NA) significantly increased the propensity for triggered activity based on delayed afterdepolarizations (DADs) in TECRLHom‐hiPSC‐CMs and treatment with flecainide, a class Ic antiarrhythmic drug, significantly reduced the triggered activity in these cells. In summary, we report that mutations in TECRL are associated with inherited arrhythmias characterized by clinical features of both LQTS and CPVT. Patient‐specific hiPSC‐CMs recapitulated salient features of the clinical phenotype and provide a platform for drug screening evidenced by initial identification of flecainide as a potential therapeutic. These findings have implications for diagnosis and treatment of inherited cardiac arrhythmias.


Stem cell reports | 2017

A COUP-TFII Human Embryonic Stem Cell Reporter Line to Identify and Select Atrial Cardiomyocytes

Verena Schwach; Arie O. Verkerk; Mervyn P.H. Mol; Jantine Monshouwer-Kloots; Harsha D. Devalla; Valeria V. Orlova; Konstantinos Anastassiadis; Christine L. Mummery; Richard P. Davis; Robert Passier

Summary Reporter cell lines have already proven valuable in identifying, tracking, and purifying cardiac subtypes and progenitors during differentiation of human pluripotent stem cells (hPSCs). We previously showed that chick ovalbumin upstream promoter transcription factor II (COUP-TFII) is highly enriched in human atrial cardiomyocytes (CMs), but not ventricular. Here, we targeted mCherry to the COUP-TFII genomic locus in hPSCs expressing GFP from the NKX2.5 locus. This dual atrial NKX2.5EGFP/+-COUP-TFIImCherry/+ reporter line allowed identification and selection of GFP+ (G+)/mCherry+ (M+) CMs following cardiac differentiation. These cells exhibited transcriptional and functional properties of atrial CMs, whereas G+/M− CMs displayed ventricular characteristics. Via CRISPR/Cas9-mediated knockout, we demonstrated that COUP-TFII is not required for atrial specification in hPSCs. This new tool allowed selection of human atrial and ventricular CMs from mixed populations, of relevance for studying cardiac specification, developing human atrial disease models, and examining distinct effects of drugs on the atrium versus ventricle.


Nature Reviews Cardiology | 2018

Transcriptional regulation of the cardiac conduction system

Vincent van Eif; Harsha D. Devalla; Gerard J.J. Boink; Vincent M. Christoffels

The rate and rhythm of heart muscle contractions are coordinated by the cardiac conduction system (CCS), a generic term for a collection of different specialized muscular tissues within the heart. The CCS components initiate the electrical impulse at the sinoatrial node, propagate it from atria to ventricles via the atrioventricular node and bundle branches, and distribute it to the ventricular muscle mass via the Purkinje fibre network. The CCS thereby controls the rate and rhythm of alternating contractions of the atria and ventricles. CCS function is well conserved across vertebrates from fish to mammals, although particular specialized aspects of CCS function are found only in endotherms (mammals and birds). The development and homeostasis of the CCS involves transcriptional and regulatory networks that act in an embryonic-stage-dependent, tissue-dependent, and dose-dependent manner. This Review describes emerging data from animal studies, stem cell models, and genome-wide association studies that have provided novel insights into the transcriptional networks underlying CCS formation and function. How these insights can be applied to develop disease models and therapies is also discussed.In this Review, Christoffels and colleagues detail the transcriptional networks that control development and homeostasis of the cardiac conduction system. The pathophysiological consequences of aberrations in these networks are also discussed, with potential insights into the generation of biological pacemakers.Key pointsThe cardiac conduction system (CCS) and atrial and ventricular working myocardium are derived from shared precursor cells that diverge during heart formation owing to localized signalling cues.Differentiation of CCS components is controlled by a network of core cardiac transcription factors and CCS-specific transcription factors; the latter also maintain phenotypic homeostasis of the adult CCS.CCS-specific transcription factors suppress the working myocardial gene programme, maintain embryonic myocardial properties, and activate a pacemaker gene programme.The atrioventricular bundle and bundle branches acquire fast-conducting properties during cardiac development, on top of their pacemaker-like properties.The Purkinje fibre network is derived from the embryonic trabecular chamber myocardium, which acquires fast-conducting properties from the onset of its development.Insights from developmental biology are being applied to develop novel cardiac disease models and additional translational efforts aimed at regeneration of the CCS.


Science Translational Medicine | 2018

Cardiac differentiation of pluripotent stem cells and implications for modeling the heart in health and disease

Harsha D. Devalla; Robert Passier

Differentiation of human pluripotent stem cells into cardiomyocytes and other cardiac cells enables modeling of heart function and elucidation of disease pathogenesis. Cellular models comprising cardiac cell types derived from human pluripotent stem cells are valuable for studying heart development and disease. We discuss transcriptional differences that define cellular identity in the heart, current methods for generating different cardiomyocyte subtypes, and implications for disease modeling, tissue engineering, and regenerative medicine.


Archive | 2018

Cells to repair the infarcted myocardium

Daniela Salvatori; Harsha D. Devalla; Robert Passier

Collaboration


Dive into the Harsha D. Devalla's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Verena Schwach

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Georgios Kosmidis

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jantine Monshouwer-Kloots

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Marcelo C. Ribeiro

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Matthew J. Birket

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Richard P. Davis

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Susana Lopes

Leiden University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge