Academic Appointments


Honors & Awards


  • NHLBI K99/R00 Pathway to Independence Award, NIH (2023/05)
  • American Heart Association Career Development Award (declined due to budget conflict), American Heart Association (2023/04)
  • Tobacco-Related Disease Research Program (TRDRP) Postdoctoral Fellowship, University of California, Office of the President (2019/07-2022/12)

All Publications


  • Studying Long QT Syndrome Caused by NAA10 Genetic Variants Using Patient-Derived Induced Pluripotent Stem Cells. Circulation Belbachir, N., Wu, Y., Shen, M., Zhang, S. L., Zhang, J. Z., Liu, C., Knollmann, B. C., Lyon, G. J., Ma, N., Wu, J. C. 2023; 148 (20): 1598-1601

    View details for DOI 10.1161/CIRCULATIONAHA.122.061864

    View details for PubMedID 37956223

  • Recent advances in regulating the proliferation or maturation of human-induced pluripotent stem cell-derived cardiomyocytes. Stem cell research & therapy Yang, H., Yang, Y., Kiskin, F. N., Shen, M., Zhang, J. Z. 2023; 14 (1): 228

    Abstract

    In the last decade, human-induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM)-based cell therapy has drawn broad attention as a potential therapy for treating injured hearts. However, mass production of hiPSC-CMs remains challenging, limiting their translational potential in regenerative medicine. Therefore, multiple strategies including cell cycle regulators, small molecules, co-culture systems, and epigenetic modifiers have been used to improve the proliferation of hiPSC-CMs. On the other hand, the immaturity of these proliferative hiPSC-CMs could lead to lethal arrhythmias due to their limited ability to functionally couple with resident cardiomyocytes. To achieve functional maturity, numerous methods such as prolonged culture, biochemical or biophysical stimulation, in vivo transplantation, and 3D culture approaches have been employed. In this review, we summarize recent approaches used to promote hiPSC-CM proliferation, and thoroughly review recent advances in promoting hiPSC-CM maturation, which will serve as the foundation for large-scale production of mature hiPSC-CMs for future clinical applications.

    View details for DOI 10.1186/s13287-023-03470-w

    View details for PubMedID 37649113

    View details for PubMedCentralID PMC10469435

  • Statins improve endothelial function via suppression of epigenetic-driven EndMT. Nature cardiovascular research Liu, C., Shen, M., Tan, W. L., Chen, I. Y., Liu, Y., Yu, X., Yang, H., Zhang, A., Liu, Y., Zhao, M. T., Ameen, M., Zhang, M., Gross, E. R., Qi, L. S., Sayed, N., Wu, J. C. 2023; 2 (5): 467-485

    Abstract

    The pleiotropic benefits of statins in cardiovascular diseases that are independent of their lipid-lowering effects have been well documented, but the underlying mechanisms remain elusive. Here we show that simvastatin significantly improves human induced pluripotent stem cell-derived endothelial cell functions in both baseline and diabetic conditions by reducing chromatin accessibility at transcriptional enhanced associate domain elements and ultimately at endothelial-to-mesenchymal transition (EndMT)-regulating genes in a yes-associated protein (YAP)-dependent manner. Inhibition of geranylgeranyltransferase (GGTase) I, a mevalonate pathway intermediate, repressed YAP nuclear translocation and YAP activity via RhoA signaling antagonism. We further identified a previously undescribed SOX9 enhancer downstream of statin-YAP signaling that promotes the EndMT process. Thus, inhibition of any component of the GGTase-RhoA-YAP-SRY box transcription factor 9 (SOX9) signaling axis was shown to rescue EndMT-associated endothelial dysfunction both in vitro and in vivo, especially under diabetic conditions. Overall, our study reveals an epigenetic modulatory role for simvastatin in repressing EndMT to confer protection against endothelial dysfunction.

    View details for DOI 10.1038/s44161-023-00267-1

    View details for PubMedID 37693816

    View details for PubMedCentralID PMC10489108

  • Protocol to generate cardiac pericytes from human induced pluripotent stem cells. STAR protocols Shen, M., Zhao, S. R., Khokhar, Y., Li, L., Zhou, Y., Liu, C., Wu, J. C. 2023; 4 (2): 102256

    Abstract

    Cardiac pericytes are a critical yet enigmatic cell type within the coronary microvasculature. Since primary human cardiac pericytes are not readily accessible, we present a protocol to generate them from human induced pluripotent stem cells (hiPSCs). Our protocol involves several steps, including the generation of intermediate cell types such as mid-primitive streak, lateral plate mesoderm, splanchnic mesoderm, septum transversum, and epicardium, before deriving cardiac pericytes. With hiPSC-derived cardiac pericytes, researchers can decipher the mechanisms underlying coronary microvascular dysfunction. For complete details on the use and execution of this protocol, please refer to Shen et al.1.

    View details for DOI 10.1016/j.xpro.2023.102256

    View details for PubMedID 37119139

  • Stepwise Generation of Human Induced Pluripotent Stem Cell-Derived Cardiac Pericytes to Model Coronary Microvascular Dysfunction. Circulation Shen, M., Liu, C., Zhao, S. R., Manhas, A., Sundaram, L., Ameen, M., Wu, J. C. 2023; 147 (6): 515-518

    View details for DOI 10.1161/CIRCULATIONAHA.122.061770

    View details for PubMedID 36745700

  • Integrative single-cell analysis of cardiogenesis identifies developmental trajectories and non-coding mutations in congenital heart disease. Cell Ameen, M., Sundaram, L., Shen, M., Banerjee, A., Kundu, S., Nair, S., Shcherbina, A., Gu, M., Wilson, K. D., Varadarajan, A., Vadgama, N., Balsubramani, A., Wu, J. C., Engreitz, J. M., Farh, K., Karakikes, I., Wang, K. C., Quertermous, T., Greenleaf, W. J., Kundaje, A. 2022; 185 (26): 4937

    Abstract

    To define the multi-cellular epigenomic and transcriptional landscape of cardiac cellular development, we generated single-cell chromatin accessibility maps of human fetal heart tissues. We identified eight major differentiation trajectories involving primary cardiac cell types, each associated with dynamic transcription factor (TF) activity signatures. We contrasted regulatory landscapes of iPSC-derived cardiac cell types and their invivo counterparts, which enabled optimization of invitro differentiation of epicardial cells. Further, we interpreted sequence based deep learning models of cell-type-resolved chromatin accessibility profiles to decipher underlying TF motif lexicons. De novo mutations predicted to affect chromatin accessibility in arterial endothelium were enriched in congenital heart disease (CHD) cases vs. controls. Invitro studies in iPSCs validated the functional impact of identified variation on the predicted developmental cell types. This work thus defines the cell-type-resolved cis-regulatory sequence determinants of heart development and identifies disruption of cell type-specific regulatory elements in CHD.

    View details for DOI 10.1016/j.cell.2022.11.028

    View details for PubMedID 36563664

  • Generation of two induced pluripotent stem cell lines from dilated cardiomyopathy patients carrying TTN mutations. Stem cell research Zhang, T. T., Zhao, S. R., Alamana, C., Shen, M., Parikh, V., Wheeler, M. T., Wu, J. C. 2022; 65: 102941

    Abstract

    Dilated cardiomyopathy (DCM) is a common heart disease that can lead to heart failure and sudden cardiac death. Mutations in the TTN gene are the most frequent cause of DCM. Here, we generated two human induced pluripotent stem cell (iPSC) lines from the peripheral blood mononuclear cells (PBMCs) of two DCM patients carrying c.94816C>T and c.104188A>G mutations in TTN, respectively. The two lines exhibited a normal morphology, full expression of pluripotency markers, a normal karyotype and the ability of trilineage differentiation. The two lines can serve as useful tools for drug screening and mechanism studies on DCM.

    View details for DOI 10.1016/j.scr.2022.102941

    View details for PubMedID 36270069

  • Technical Applications of Microelectrode Array and Patch Clamp Recordings on Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Journal of visualized experiments : JoVE Zhao, S. R., Mondejar-Parreno, G., Li, D., Shen, M., Wu, J. C. 2022

    Abstract

    Drug-induced cardiotoxicity is the leading cause of drug attrition and withdrawal from the market. Therefore, using appropriate preclinical cardiac safety assessment models is a critical step during drug development. Currently, cardiac safety assessment is still highly dependent on animal studies. However, animal models are plagued by poor translational specificity to humans due to species-specific differences, particularly in terms of cardiac electrophysiological characteristics. Thus, there is an urgent need to develop a reliable, efficient, and human-based model for preclinical cardiac safety assessment. Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have emerged as an invaluable in vitro model for drug-induced cardiotoxicity screening and disease modeling. hiPSC-CMs can be obtained from individuals with diverse genetic backgrounds and various diseased conditions, making them an ideal surrogate to assess drug-induced cardiotoxicity individually. Therefore, methodologies to comprehensively investigate the functional characteristics of hiPSC-CMs need to be established. In this protocol, we detail various functional assays that can be assessed on hiPSC-CMs, including the measurement of contractility, field potential, action potential, and calcium handling. Overall, the incorporation of hiPSC-CMs into preclinical cardiac safety assessment has the potential to revolutionize drug development.

    View details for DOI 10.3791/64265

    View details for PubMedID 35993757

  • Generation of Embryonic Origin-Specific Vascular Smooth Muscle Cells from Human Induced Pluripotent Stem Cells. Methods in molecular biology (Clifton, N.J.) Shen, M., Liu, C., Wu, J. C. 2022; 2429: 233-246

    Abstract

    Vascular smooth muscle cells (VSMCs), a highly mosaic tissue, arise from multiple distinct embryonic origins and populate different regions of our vascular network with defined boundaries. Accumulating evidence has revealed that the heterogeneity of VSMC origins contributes to region-specific vascular diseases such as atherosclerosis and aortic aneurysm. These findings highlight the necessity of taking into account lineage-dependent responses of VSMCs to common vascular risk factors when studying vascular diseases. This chapter describes a reproducible, stepwise protocol for the generation of isogenic VSMC subtypes originated from proepicardium, second heart field, cardiac neural crest, and ventral somite using human induced pluripotent stem cells. By leveraging this robust induction protocol, patient-derived VSMC subtypes of desired embryonic origins can be generated for disease modeling as well as drug screening and development for vasculopathies with regional susceptibility.

    View details for DOI 10.1007/978-1-0716-1979-7_15

    View details for PubMedID 35507165

  • Generation of Vascular Smooth Muscle Cells From Induced Pluripotent Stem Cells: Methods, Applications, and Considerations. Circulation research Shen, M., Quertermous, T., Fischbein, M. P., Wu, J. C. 2021; 128 (5): 670–86

    Abstract

    The developmental origin of vascular smooth muscle cells (VSMCs) has been increasingly recognized as a major determinant for regional susceptibility or resistance to vascular diseases. As a human material-based complement to animal models and human primary cultures, patient induced pluripotent stem cell iPSC-derived VSMCs have been leveraged to conduct basic research and develop therapeutic applications in vascular diseases. However, iPSC-VSMCs (induced pluripotent stem cell VSMCs) derived by most existing induction protocols are heterogeneous in developmental origins. In this review, we summarize signaling networks that govern in vivo cell fate decisions and in vitro derivation of distinct VSMC progenitors, as well as key regulators that terminally specify lineage-specific VSMCs. We then highlight the significance of leveraging patient-derived iPSC-VSMCs for vascular disease modeling, drug discovery, and vascular tissue engineering and discuss several obstacles that need to be circumvented to fully unleash the potential of induced pluripotent stem cells for precision vascular medicine.

    View details for DOI 10.1161/CIRCRESAHA.120.318049

    View details for PubMedID 33818124

  • Generation of three induced pluripotent stem cell lines from hypertrophic cardiomyopathy patients carrying TNNI3 mutations. Stem cell research Zhao, S. R., Shen, M., Lee, C., Zha, Y., Guevara, J. V., Wheeler, M. T., Wu, J. C. 2021; 57: 102597

    Abstract

    Hypertrophic cardiomyopathy (HCM) is a common inherited heart disease with a prevalence of about 0.2%. HCM is typically caused by mutations in genes encoding sarcomere or sarcomere-associated proteins. Here, we characterized induced pluripotent stem cell (iPSC) lines generated from the peripheral blood mononuclear cells of three HCM patients each carrying c.433C > T, c.610C > T, or c.235C > T mutation in the TNNI3 gene by non-integrated Sendai virus. All of the three lines exhibited normal morphology, expression of pluripotent markers, stable karyotype, and the potential of trilineage differentiation. The cardiomyocytes differentiated from these iPSC lines can serve as useful tools to model HCM in vitro.

    View details for DOI 10.1016/j.scr.2021.102597

    View details for PubMedID 34798544

  • The Regulation of Endothelial Function Through Hmgcr/mevalonate Pathway Mediated Yap Activity Liu, C., Liu, Y., Chen, C., Ameen, M., Yang, H., Shen, M., Rhee, J., Chen, I. Y., Sayed, N., Wu, J. C. LIPPINCOTT WILLIAMS & WILKINS. 2020
  • Generation of Quiescent Cardiac Fibroblasts Derived from Human Induced Pluripotent Stem Cells. Methods in molecular biology (Clifton, N.J.) Zhang, H. n., Shen, M. n., Wu, J. C. 2020

    Abstract

    Myocardial fibrosis is a hallmark of cardiac remodeling, which can progressively lead to heart failure, a leading cause of death worldwide. The effector cells of fibrosis in the heart are cardiac fibroblasts (CFs). There is currently no effective therapeutic strategy clinically available to specifically attenuate maladaptive responses of CFs. Large-scale applications such as high-throughput drug screening are difficult due to the limited availability of human primary CFs, thus limiting the development of future treatments. Here, we describe a robust induction protocol that can be used to generate a scalable, consistent, genetically defined source of quiescent CFs from human induced pluripotent stem cells for cardiac fibrosis modeling, drug discovery, and tissue engineering.

    View details for DOI 10.1007/7651_2020_300

    View details for PubMedID 32671814

  • Generation of Quiescent Cardiac Fibroblasts from Human Induced Pluripotent Stem Cells for In Vitro Modeling of Cardiac Fibrosis. Circulation research Zhang, H., Tian, L., Shen, M., Wu, H., Gu, M., Tu, C., Paik, D. T., Wu, J. C. 2019

    Abstract

    RATIONALE: Activated fibroblasts are the major cell type that secrete excessive extracellular matrix in response to injury, contributing to pathological fibrosis and leading to organ failure. Effective anti-fibrotic therapeutic solutions, however, are not available due to the poorly defined characteristics and unavailability of tissue-specific fibroblasts. Recent advances in single-cell RNA-sequencing (scRNA-seq) fill such gaps of knowledge by enabling delineation of the developmental trajectories and identification of regulatory pathways of tissue-specific fibroblasts among different organs.OBJECTIVE: This study aims to define the transcriptome profiles of tissue-specific fibroblasts using recently reported mouse scRNA-seq atlas, and to develop a robust chemically defined protocol to derive cardiac fibroblasts (CFs) from human induced pluripotent stem cells (iPSCs) for in vitro modeling of cardiac fibrosis and drug screening.METHODS AND RESULTS: By analyzing the single-cell transcriptome profiles of fibroblasts from 10 selected mouse tissues, we identified distinct tissue-specific signature genes, including transcription factors that define the identities of fibroblasts in the heart, lungs, trachea, and bladder. We also determined that CFs in large are of the epicardial lineage. We thus developed a robust chemically-defined protocol that generates CFs from human iPSCs. Functional studies confirmed that iPSC-derived CFs preserved a quiescent phenotype and highly resembled primary CFs at the transcriptional, cellular, and functional levels. We demonstrated that this cell-based platform is sensitive to both pro- and anti-fibrosis drugs. Finally, we showed that crosstalk between cardiomyocytes and CFs via the atrial/brain natriuretic peptide-natriuretic peptide receptor 1 pathway is implicated in suppressing fibrogenesis.CONCLUSIONS: This study uncovers unique gene signatures that define tissue-specific identities of fibroblasts. The bona fide quiescent CFs derived from human iPSCs can serve as a faithful in vitro platform to better understand the underlying mechanisms of cardiac fibrosis and to screen anti-fibrotic drugs.

    View details for DOI 10.1161/CIRCRESAHA.119.315491

    View details for PubMedID 31288631

  • Extracellular matrix, regional heterogeneity of the aorta, and aortic aneurysm. Experimental & molecular medicine Jana, S. n., Hu, M. n., Shen, M. n., Kassiri, Z. n. 2019; 51 (12): 160

    Abstract

    Aortic aneurysm is an asymptomatic disease with dire outcomes if undiagnosed. Aortic aneurysm rupture is a significant cause of death worldwide. To date, surgical repair or endovascular repair (EVAR) is the only effective treatment for aortic aneurysm, as no pharmacological treatment has been found effective. Aortic aneurysm, a focal dilation of the aorta, can be formed in the thoracic (TAA) or the abdominal (AAA) region; however, our understanding as to what determines the site of aneurysm formation remains quite limited. The extracellular matrix (ECM) is the noncellular component of the aortic wall, that in addition to providing structural support, regulates bioavailability of an array of growth factors and cytokines, thereby influencing cell function and behavior that ultimately determine physiological or pathological remodeling of the aortic wall. Here, we provide an overview of the ECM proteins that have been reported to be involved in aortic aneurysm formation in humans or animal models, and the experimental models for TAA and AAA and the link to ECM manipulations. We also provide a comparative analysis, where data available, between TAA and AAA, and how aberrant ECM proteolysis versus disrupted synthesis may determine the site of aneurysm formation.

    View details for DOI 10.1038/s12276-019-0286-3

    View details for PubMedID 31857579