Research

Our lab focuses on the development and translation of mechanism-based therapies for heart disease. Our approach combines induced pluripotent stem cell biology and disease modeling, engineered heart tissues, development of physiological screening modalities, and therapeutic target and early-stage drug discovery.

iPSC and Disease Modeling

Perea-Gil et al. used high throughput physiological screening to identify small molecule activators of de novo serine biosynthesis might be a therapeutic strategy for familial dilated cardiomyopathy (DCM). doi:10.1093/eurheartj/ehac305 (2022).

Physiological immaturity of iPSC-derived cardiomyocytes limits their fidelity as disease models. Feyen et al. developed a new culture media that increases maturation of ventricular-like hiPSC-CMs in 2D and 3D engineered heart cultures relative to standard protocols. Improved characteristics include a low resting Vm, rapid depolarization, and increased Ca2+ dependence and force generation. Figure from Feyen et al., Cell Reports, 2021.

We have made seminal discoveries that laid the foundation for iPSC modeling of heart disease, including the definition of Wnt inhibitors and other molecules that are used in most efficient protocols to produce iPSC-derived cardiac cells (Schneider and Mercola, Genes Dev, 2000; Willems et al., Circ. Res., 2012). We recently developed a metabolic maturation protocol that confers more mature electrophysiological, structural and mechanical properties of iPSC-derived cardiomyocytes and improves the fidelity of disease modeling (Feyen et al., Cell Reports, 2020). iPSC models can be used to model inherited heart disease, including cardiomyopathies and ion channel disorders. Through the use of high throughput physiology studies (below), we apply these models to understand disease mechanisms and develop mechanism-based therapeutics.

Publications

  1. Perea-Gil, I. et al. Serine biosynthesis as a novel therapeutic target for dilated cardiomyopathy. Eur Heart J. doi: 10.1093/eurheartj/ehac305 (2022).
  2. Hnatiuk, A. P. et al. Human iPSC modeling of heart disease for drug development. Cell Chem Biol 28, 271-282, doi:10.1016/j.chembiol.2021.02.016 (2021).
  3. Feyen, D. A. M. et al. The Unfolded Protein Response as a Compensatory Mechanism and Potential Therapeutic Target in PLN R14del Cardiomyopathy. Circulation, doi:10.1161/CIRCULATIONAHA.120.049844 (2021).
  4. Pei, J., Maas, R. G. C. et al. Transcriptional regulation profiling reveals disrupted lipid metabolism in failing hearts with a pathogenic phospholamban mutation. bioRxiv, 2020.2011.2030.402792, doi:10.1101/2020.11.30.402792 (2020).
  5. Feyen, D. A. M. et al. Metabolic Maturation Media Improve Physiological Function of Human iPSC-Derived Cardiomyocytes. Cell Rep 32, 107925, doi:10.1016/j.celrep.2020.107925 (2020).
  6. Briganti, F., et al. iPSC Modeling of RBM20-Deficient DCM Identifies Upregulation of RBM20 as a Therapeutic Strategy. Cell Rep 32, 108117, doi:10.1016/j.celrep.2020.108117 (2020).
  7. Sharma, A. et al. High-throughput screening of tyrosine kinase inhibitor cardiotoxicity with human induced pluripotent stem cells. Sci Transl Med 9, doi:10.1126/scitranslmed.aaf2584 (2017).
  8. Cai, W. et al. A Nodal-to-TGFbeta cascade exerts biphasic control over cardiopoiesis. Circ Res 111, 876-881, doi:10.1161/CIRCRESAHA.112.270272 (2012).
  9. Willems, E. et al. Small-molecule inhibitors of the Wnt pathway potently promote cardiomyocytes from human embryonic stem cell-derived mesoderm. Circ Res 109, 360-364, doi:10.1161/CIRCRESAHA.111.249540 (2011).
  10. Schneider, V. A. et al. Wnt antagonism initiates cardiogenesis in Xenopus laevis. Genes Dev 15, 304-315, doi:10.1101/gad.855601 (2001).

High throughput physiological screening for drug discovery

Feyen et al. (Circulation, 2021) generated iPSC-derived cardiomyocytes carrying the PLN R14del mutation and isogenic control lines (A). BiX activation of the Unfolded Protein Response (UPR) (B) restores the contractile deficit of PLN R14del mutant iPSC-cardiomyocytes to the same extent as gene correction (C). Figure redrawn from Feyen et al., Circulation, 2021.

We perform high throughput, automated physiological recording to read out voltage, calcium and contractile measurements of heart cells in 384-well plate format. This has enabled quantitative assessment of physiological defects in hiPSC-derived cardiac cells carrying gene variants that cause congenital heart disease. The in vitro physiological defects recapitulate aspects of disease in affected patients, and thus allows us to study the influence of genes, proteins and signaling pathways on disease in high throughput. We are using this platform in large-scale functional genomics studies to probe mechanisms of disease as well as map therapeutic target space. One area of interest is ventricular arrhythmia. An ongoing project is to use our screening technology to comprehensively identify the therapeutic target space for ventricular arrhythmia that occurs in patients with congenital forms of hypertrophic cardiomyopathy (HCM). Myopathies in general are of interest, and a major focus of the lab is to discover mechanism-based therapeutic strategies for different forms of hypertrophic (HCM), dilated (DCM) and restrictive cardiomyopathy (RCM). Recent collaborative publications in this area have explored therapeutic strategies for RBM20 mutant DCM (Briganti et al., Cell Reports, 2020) and PLN mutant DCM (Feyen et al., Circulation, 2020)

Publications

  1. Hnatiuk, A. P. et al. Human iPSC modeling of heart disease for drug development. Cell Chem Biol 28, 271-282, doi:10.1016/j.chembiol.2021.02.016 (2021).
  2. Feyen, D. A. M. et al. The Unfolded Protein Response as a Compensatory Mechanism and Potential Therapeutic Target in PLN R14del Cardiomyopathy. Circulation, doi:10.1161/CIRCULATIONAHA.120.049844 (2021).
  3. Vaskova, E. et al. Sacubitril/Valsartan Improves Cardiac Function and Decreases Myocardial Fibrosis Via Downregulation of Exosomal miR-181a in a Rodent Chronic Myocardial Infarction Model. J Am Heart Assoc 9, e015640, doi:10.1161/JAHA.119.015640 (2020).
  4. Paige, S. L. et al. Patient-Specific Induced Pluripotent Stem Cells Implicate Intrinsic Impaired Contractility in Hypoplastic Left Heart Syndrome. Circulation 142, 1605-1608, doi:10.1161/CIRCULATIONAHA.119.045317 (2020).
  5. Briganti, F. et al. iPSC Modeling of RBM20-Deficient DCM Identifies Upregulation of RBM20 as a Therapeutic Strategy. Cell Rep 32, 108117, doi:10.1016/j.celrep.2020.108117 (2020).

Cardiotoxicity and Drug-reengineering

Hnatiuk, Bruyneel, Tailor and Pandrala et al. (l10.1158/0008-5472.CAN-21-3652 and 10.1021/acs.jmedchem.1c01853) used parallel models of cardiotoxicity and tumor growth to reengineer ponatinib, which is one of the most cardiotoxic of cancer drugs. The new molecules retained anti-tumor efficacy but had reduced cardiovascular toxicity.

McKeithan et al. (Cell Stem Cell, 2021) used large-scale functional screening of hiPSC-cardiomyocytes carrying a mutation that causes an electrophysiological disorder (Long QT syndrome type 3) to direct the chemical optimization of mexiletine, an antiarrhythmic drug used to treat the disease. Figure from McKeithan et al., Cell Stem Cell, 2021.

Many drugs induce arrhythmic and cardiomyopathic effects as an adverse consequence of treatment. The mechanisms can be either on-target or off-target, the latter meaning that the adverse effect is due to interaction with a protein other than the intended target of the drug. Phenotypic physiological assays can be used to determine moieties in the chemical structures of drugs responsible for their undesirable effects. Based on electrophysiological effects in patient hiPSC-derived cardiomyocytes carrying mutation that causes the electrophysiological disorder Long QT type 3 (LQT3), we optimized the chemical structure of an anti-arrhythmic targets the cardiac sodium channel to decrease potential adverse effects and increase therapeutic potential (McKeithan et al., Cell Stem Cell, 2020; Cashman et al., J. Med. Chem., 2020). Cardiotoxicity is a major concern for oncology drugs and about a third of cancer patients suffer from adverse cardiovascular effects due to their cancer treatment. As a class, kinase inhibitor drugs elicit cardiotoxic effects in patients that can be revealed using iPSC-based assays (Sharma et al., Sci. Transl. Med., 2017). A current project uses this technology to map the chemical moieties responsible for cardiotoxicity of the BCR-ABL kinase inhibitor ponatinib, which is a highly cardiotoxic drug used to treat forms of Philadelphia chromosome-positive leukemia. Using assays that recapitulate the vascular and cardiomyocyte toxicities, we are developing cardiosafe analogues that retain anti-tumor efficacy.

Publications

  1. Hnatiuk, A.P. et al. Reengineering Ponatinib to Minimize Cardiovascular Toxicity. Cancer Res. doi: 10.1158/0008-5472.CAN-21-3652 (2022).
  2. Pandrala, M. et al. Designing Novel BCR-ABL Inhibitors for Chronic Myeloid Leukemia with Improved Cardiac Safety. J Med Chem. doi: 10.1021/acs.jmedchem.1c01853 (2022).
  3. Vincent, F. et al. Phenotypic drug discovery: recent successes, lessons learned and new directions. Nat Rev Drug Discov. doi: 10.1038/s41573-022-00472-w (2022).
  4. Abdelsayed, M. et al. Repurposing drugs to treat cardiovascular disease in the era of precision medicine. Nat Rev Cardiol. doi: 10.1038/s41569-022-00717-6 (2022).
  5. Cerignoli, F. et al. High throughput measurement of Ca(2)(+) dynamics for drug risk assessment in human stem cell-derived cardiomyocytes by kinetic image cytometry. J Pharmacol Toxicol Methods 66, 246-256, doi:10.1016/j.vascn.2012.08.167 (2012).
  6. Sharma, A. et al. High-throughput screening of tyrosine kinase inhibitor cardiotoxicity with human induced pluripotent stem cells. Sci Transl Med 9, doi:10.1126/scitranslmed.aaf2584 (2017).
  7. McKeithan, W. L. et al. An Automated Platform for Assessment of Congenital and Drug-Induced Arrhythmia with hiPSC-Derived Cardiomyocytes. Front Physiol 8, 766, doi:10.3389/fphys.2017.00766 (2017).
  8. Cashman, J. R. et al. Antiarrhythmic Hit to Lead Refinement in a Dish Using Patient-Derived iPSC Cardiomyocytes. J Med Chem 64, 5384-5403, doi:10.1021/acs.jmedchem.0c01545 (2021).
  9. McKeithan, W. L. et al. Reengineering an Antiarrhythmic Drug Using Patient hiPSC Cardiomyocytes to Improve Therapeutic Potential and Reduce Toxicity. Cell Stem Cell 27, 813-821 e816, doi:10.1016/j.stem.2020.08.003 (2020).

Chemical Genomics and Drug Discovery

High throughput screening identified a small molecule with dual p53 agonist and Wnt inhibitory activity. Named PAWI, the compounds were used to trace a kinase cascade linking stress signaling to the regulation of TCF/LEF function and the control of Wnt-dependent gene expression. PAWI does not destabilize microtubules at therapeutic doses, but is effective against models of pancreatic and colon cancer. Figure from Cheng, Tsuda et al., Cell Chemical Biology, 2021.

Our chemical genomics projects began over a decade ago using libraries of small molecule ion channel inhibitors to probe processes during early embryogenesis that underlie congenital left-right asymmetry disorders, such as heterotaxia and transposition of the great vessels (Levin et al., Cell, 2002) and to identify classes of compounds to direct cardiomyocyte differentiation from iPSCs (Willems et al., Circulation Research, 2011). Recent studies have focused on developing a highly potent dual p53 agonist and Wnt inhibitor that elucidated a link between cellular stress and Wnt signaling. An optimized analogue has excellent PK properties and is effective against orthotopic pancreatic cancer and xenograft colon cancer models. Additional work has used high throughput physiological assays to develop a first in class NOTCH/RBPJ inhibitor and a first in class TGFb type II receptor inhibitor that has greater selectivity for inhibiting TGFb signaling than do type I receptor kinase active site inhibitors

Publications

  1. Levin, M. et al. Asymmetries in H(+)/K(+)-ATPase and Cell Membrane Potentials Comprise a Very Early Step in Left-Right Patterning. Cell 111, 77-89 (2002).
  2. Cheng, J. et al. Small-molecule probe reveals a kinase cascade that links stress signaling to TCF/LEF and Wnt responsiveness. Cell Chem Biol 28, 1-11, doi:10.1016/j.chembiol.2021.01.001 (2021).
  3. Cashman, J. R. et al. Antiarrhythmic Hit to Lead Refinement in a Dish Using Patient-Derived iPSC Cardiomyocytes. J Med Chem 64, 5384-5403, doi:10.1021/acs.jmedchem.0c01545 (2021).
  4. Hurtado, C. et al. Disruption of NOTCH signaling by a small molecule inhibitor of the transcription factor RBPJ. Sci Rep 9, 10811, doi:10.1038/s41598-019-46948-5 (2019).
  5. Cheng, J. et al. A Novel Inhibitor Targets Both Wnt Signaling and ATM/p53 in Colorectal Cancer. Cancer Res 78, 5072-5083, doi:10.1158/0008-5472.CAN-17-2642 (2018).
  6. Willems, E. et al. Small molecule-mediated TGF-beta type II receptor degradation promotes cardiomyogenesis in embryonic stem cells. Cell Stem Cell 11, 242-252, doi:10.1016/j.stem.2012.04.025 (2012).
  7. Lanier, M. et al. Wnt inhibition correlates with human embryonic stem cell cardiomyogenesis: a structure-activity relationship study based on inhibitors for the Wnt response. J Med Chem 55, 697-708, doi:10.1021/jm2010223 (2012).
  8. Kiselyuk, A. et al. HNF4alpha antagonists discovered by a high-throughput screen for modulators of the human insulin promoter. Chem Biol 19, 806-818, doi:10.1016/j.chembiol.2012.05.014 (2012).
  9. Willems, E. et al. Small-molecule inhibitors of the Wnt pathway potently promote cardiomyocytes from human embryonic stem cell-derived mesoderm. Circ Res 109, 360-364, doi:10.1161/CIRCRESAHA.111.249540 (2011).

Heart Regeneration

Wei et al. (Nature, 2015) found that cells located at the epicardial surface of the heart normally express a hypoglycosylated form of a protein called follistatin-like 1 (Fstl1), which is lost in response to an infarction. Restoration of a recombinant form of hypoglycosylated FSTL1 improved survival and cardiac function in mice after myocardial infarction. Figure from Van Rooij et al., New England Journal of Medicine, 2016.

A collaborative study with Dr. Pilar Ruiz-Lozano led to the discovery that hypoglycosylated forms of Follistatin-like-1 (FSTL1) can regenerate myocardium of mice and pigs when applied following myocardial infarction (Wei et al., Nature, 2015 link to doi:10.1038/nature15372). This discovery led to the founding of Regencor [link to https://www.regencor.com], which is commercializing the technology.

Publications

  1. Diez-Cunado, M. et al. miRNAs that Induce Human Cardiomyocyte Proliferation Converge on the Hippo Pathway. Cell Rep 23, 2168-2174, doi:10.1016/j.celrep.2018.04.049 (2018).
  2. Diaz-Trelles, R. et al. Notch-independent RBPJ controls angiogenesis in the adult heart. Nat Commun 7, 12088, doi:10.1038/ncomms12088 (2016).
  3. Wei, K. et al. Epicardial FSTL1 reconstitution regenerates the adult mammalian heart. Nature 525, 479-485, doi:10.1038/nature15372 (2015).
  4. Wei, K. et al. Developmental origin of age-related coronary artery disease. Cardiovasc Res 107, 287-294, doi:10.1093/cvr/cvv167 (2015).