Our research is focused on human cardiomyocytes derived from human pluripotent stem cells, which hold promise for cardiac cell therapy, disease modeling, drug discovery, and the study of developmental biology. We are also collaborating with investigators at Georgia Tech, Emory University, and Children’s Healthcare of Atlanta, exploring the application of nanotechnology and tissue engineering in stem cell research. 

Highlights of Recent Research

Metabolic regulation enhances maturation of hiPSC-cardiomyocytes and enables pathological modeling

Cardiac pathological outcome of metabolic remodeling is difficult to model using cardiomyocytes derived from human-induced pluripotent stem cells (hiPSC-CMs) due to low metabolic maturation. Metabolic regulation through activation of AMPK improves maturation of hiPSC-derived cardiomyocytes and allows the cells to respond to pathological stimuli, which is not achievable in immature cells.

To read more, please see:

Li D, Armand LC, Sun F, Hwang H, Wolfson D, Rampoldi A, Liu R, Forghani P, Hu X, Yu W-M, Qu C-K, Jones DP, Wu R, Cho HC, Maxwell JT, Xu C. AMPK activator-treated human cardiac spheres enhance maturation and enable pathological modeling. Stem Cell Res Ther. 14:322 (2023).

Space microgravity increases cardiac proliferation

In microgravity, cells undergo profound changes in their properties. We have found that cryopreserved 3D cardiac progenitors differentiated into highly enriched cardiomyocytes on the International Space Station (ISS). Compared with ISS 1G cells, ISS microgravity cells had increased proliferation and improved Ca2+ transients. Microgravity also increased the expression of genes associated with proliferation and cardiac differentiation and contraction.

To read more, please see:

Hwang H, Rampoldi1 A, Forghani1 P, Li D, Fite J, Boland G, Maher K, Xu C. Space microgravity increases expression of genes associated with proliferation and differentiation in human cardiac spheres. npj Microgravity. 9:88 (2023).

Rampoldi A, Forghani F, Li D, Hwang H, Armand LC, Fite J, Boland G, Maxwell J, Maher K, Xu C. Space microgravity improves proliferation of human iPSC-derived cardiomyocytes. Stem Cell Rep. 17:2272-2285. (2022).

Cancer therapy-induced cardiotoxicity in human stem cell-cardiomyocytes

Cancer treatment with proteasome inhibitors can lead to cardiotoxicity, but the underlying mechanisms are not fully understood. We have found that treatment of human stem cell-derived cardiomyocytes with carfilzomib, a proteasome inhibitor, induces oxidative stress, mitochondrial dysfunction, and cell death. The carfilzomib treatment also negatively affects cardiac function and gene expression.

To read more, please see:

Forghani P, Rashid A, Sun F, Liu R, Li D, Lee MR, Hwang H, Maxwell1 JT, Mandawat A, Wu R, Salaita K, Xu C. Carfilzomib treatment causes molecular and functional alterations of human induced pluripotent stem cell-derived cardiomyocytes. J Am Heart Assoc 7:e022247 (2021).

 

Chronic ethanol exposure induces deleterious changes in human stem cell-cardiomyocytes

Chronic alcohol consumption can induce cardiomyopathy, arrhythmias, and heart failure. We have found that human stem cell-derived cardiomyocytes can be a novel model for the study of cardiotoxicity induced by chronic alcohol exposure. Long-term exposure of stem cell-derived cardiomyocytes with ethanol results in cell death, oxidative stress, intracellular Ca2+ handling dysfunction, altered membrane potential, impaired cardiomyocyte contractility, suppressed cardiac structure development, and altered the expression of proteins, genes and metabolites.

To read more, please see:

Hwang H, Liu R, Eldridge R, Hu X, Forghani P, Jones DP, Xu C. Chronic ethanol exposure induces mitochondrial dysfunction and alters gene expression and metabolism in human cardiac spheroids. Alcohol Clin Exp Res. 47: 643-658 (2023).

Liu R, Sun F, Armand LC, Wu R, Xu C. Chronic ethanol exposure induces deleterious changes in cardiomyocytes derived from human induced pluripotent stem cells. Stem Cell Rev Rep 17:2314-2331 (2021).

 

Cryopreservation and CO2-independent culture of 3D cardiac progenitors for spaceflight experiments

Space experimentation of cardiomyocyte differentiation from human induced pluripotent stem cells offers an exciting opportunity to explore the potential of these cells for disease modeling, drug discovery and regenerative medicine. We have developed methods of cryopreservation and CO2-independent culture of 3D cardiac progenitors for space experiments. Our cryopreserved cardiac progenitor spheres were successfully cultivated in a spaceflight culture module without CO2 for 3 weeks aboard the International Space Station. Beating cardiomyocytes were generated and returned to the earth for further study.

To read more, please see:

Rampoldi A, Jha R, Fite J, Boland G and Xu C. Cryopreservation and CO2-independent culture of 3D cardiac progenitors for spaceflight experiments. Biomaterials 269:120673 (2021). 

 

Proinflammatory cytokine TNF-α induces molecule and functional alterations in  human cardiomyocytes 

Proinflammatory TNF-α is predominantly elevated in cytokine storm as well as worsening cardiac function. TNF-α treatment of stem cell-cardiomyocytes resulted in dysregulation of cardiomyocyte function with respect to the increased abnormal Ca2+ handling, Ca2+ wave propagation between cells and excitation-contraction coupling, which are consistent with clinical pathological events. In addition, TNF-α treatment up-regulated IL-32 (a human specific cytokine, not present in rodents) and down-regulated glutamate receptors and cardiomyocyte contractile proteins. Our study provides a model to incorporate inflammatory factors into stem cell-based studies to evaluate mechanistic aspects of heart disease.

To read more, please see:

Saraf A, Rampoldi A, Chao M, Li D, Armand L, Hwang H, Liu R, Jha R, Fu H, Maxwell JT, Xu C. Functional and molecular effects of TNF-α on human iPSC-derived cardiomyocytes. Stem Cell Res 52:102218 (2021).

 

Chemotherapeutic drug melphalan induces cardiotoxicity through oxidative stress

Chemotherapeutic drugs such as melphalan could induce cardiotoxicity including severe arrhythmias and heart failure. Using a human cell model of cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs), we have found that melphalan treatment of hiPSC-CMs induced oxidative stress, caused Ca2+-handling defects and dysfunctional contractility, altered global transcriptomic and proteomic profiles, and resulted in apoptosis and cell death. This study opens up new clinical mechanism-based targets to prevent and treat melphalan-induced cardiotoxicity.

To read more, please see:

Liu R, Li D, Sun F, Rampoldi A, Maxwell JT, Wu R, Fischbach P, Castellino SM, Du Y, Fu H, Mandawat A, Xu C. Melphalan induces cardiotoxicity through oxidative stress in cardiomyocytes derived from human induced pluripotent stem cells. Stem Cell Res Ther 11(1):470 (2020). 

 

Machine learning identifies abnormal Ca2+ transients

Identifying abnormal Ca2+ transients is crucial for evaluating cardiomyocyte function that requires labor-intensive manual effort. Therefore, we have developed an analytical pipeline for automatic assessment of Ca2+ transient abnormality, by employing advanced machine learning methods, and achieved 88% training accuracy and 87% test accuracy. Further, we provide a free R package to implement our pipeline for high-throughput analysis of cardiomyocyte Ca2+ transients.

To read more, please see:

Hwang H, Liu R, Maxwell JT, Yang J, Xu C. Machine learning identifies abnormal Ca2+ transients in human induced pluripotent stem cell-derived cardiomyocytes. Sci Rep 10:16977 (2020).

 

A long non-coding RNA GATA6-AS1 is required for cardiomyocyte differentiation

Long noncoding RNAs (lncRNAs) are crucial in many cellular processes, yet relative few have been shown to regulate human cardiomyocyte differentiation. Through genetic modification of human stem cells, we have demonstrated a previously unrecognized and functional role of lncRNA GATA6-AS1 adjacent to GATA6 in controlling human cardiomyocyte differentiation.

To read more, please see:

Jha R, Li D, Wu Q, Ferguson KE, Forghani P, Gibson GC, Xu C. A long non-coding RNA adjacent to GATA6 is required for cardiomyocyte differentiation from human pluripotent stem cells. FASEB J 34:14336-14352 (2020). 

 

Proteomic profiling of alcohol-induced cardiotoxicity

Alcohol use in pregnancy increases the risk of abnormal cardiac development, and excessive alcohol consumption in adults can induce cardiomyopathy, contractile dysfunction, and arrhythmias. We have performed proteomic and bioinformatic analysis to examine protein alterations globally and quantitatively in cardiomyocytes derived from human pluripotent stem cells (hiPSC-CMs) treated with ethanol. Our findings highlight the involvement of oxidative stress and dysregulation of Ca2+ handling and contraction while also implicating potential novel targets in alcohol-induced cardiotoxicity.

To read more, please see:

Liu R, Sun F, Forghani P, Armand LC, Rampoldi A, Li D, Wu R, Xu C. Proteomic profiling reveals roles of stress response, Ca2+ transient dysregulation and novel signaling pathways in alcohol-induced cardiotoxicity. Alcohol Clin Exp Res 44:2187-2199 (2020).

 

Targeting HIF-1α promotes the metabolic maturation of cardiomyocytes

Immature phenotypes of cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs) limit the utility of these cells in clinical application and basic research. During cardiac development, postnatal cardiomyocytes experience high oxygen tension along with a concomitant downregulation of hypoxia-inducible factor 1α (HIF-1α), leading to increased fatty acid oxidation. We have found that targeting HIF-1α in combination with other metabolic regulators significantly improves the metabolic maturation of hiPSC-CMs.

To read more, please see:

Gentillon C, Li D, Duan M, Yu W-M, Preininger MK, Jha R, Rampoldi A, Saraf A, Gibson GC, Qu C-K, Brown L, Xu C. Targeting HIF-1α in combination with PPAR activation and postnatal factors promotes the metabolic maturation of human induced pluripotent stem cell-derived cardiomyocytes. J Mol Cell Cardiol 132: 20-135 (2019). 

 

Modeling alcohol-induced cardiotoxicity in stem cell-derived cardiomyocytes

Alcohol use prior to and during pregnancy remains a significant societal problem and can lead to developmental fetal abnormalities including compromised myocardia function and increased risk for heart disease later in life.  We have found that human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) could serve as a useful tool to study alcohol-induced cardiac defects and/or toxicity.  hiPSC-CMs had increased irregular Ca2+ transients and contractility when the cells were treated with ethanol even at a low dose.

To read more, please see:

Rampoldi A, Singh M, Wu Q, Duan M, Jha R, Maxwell JT, Bradner JM, Zhang X, Saraf A, Miller GW, Gibson G, Brown LA, Xu C. Cardiac toxicity from ethanol exposure in human induced pluripotent stem cell-derived cardiomyocytes. Toxicol Sci 169: 280-292 (2019).

Fetal alcohol cardiac toxicity in a dish. March 2019. Lab Land the Emory Health Science Blog

Alcohol and fetal heart development. Spring 2019. Emory Medicine Magazine

 

VLPs for specific elimination of tumorigenic human pluripotent stem cells

 

Human pluripotent stem cell (hPSC) derivatives are a promising cell source for regenerative medicine, and transgenic expression of suicide genes is a leading strategy for the selective elimination of potentially tumorigenic hPSCs in differentiation cultures before transplantation. However, transgenic modification poses safety risks such as deleterious mutagenesis. We have established an alternative method (without transgenic modification) by delivering suicide-inducing molecules explicitly to hPSCs using virus-like particles (VLPs), and demonstrate its use in eliminating undifferentiated hPSCs without affecting neighboring non-stem cells.

To read more, please see:

Targeted elimination of tumorigenic human pluripotent stem cells using suicide-inducing virus-like particles. ACS Chem Biol. 13: 2324-2338 (2018).

Stem cells driven into selective suicide. July 2018.  Lab Land the Emory Health Science Blog.

 

A novel role of LGR5 in cardiomyocyte and endothelial cell differentiation from human pluripotent stem cells

Understanding molecules involved in the differentiation of human pluripotent stem cells (hPSCs) into cardiomyocytes and endothelial cells is important in advancing hPSCs for cell therapy and drug testing.  In a study published in Stem Cell Reports, we have demonstrated that LGR5 (a leucine-rich repeat-containing G-protein-coupled receptor) plays a critical role in cardiomyocyte and endothelial cell differentiation from hPSCs.  Expression of LGR5 is transiently upregulated during the early stage of cardiomyocyte differentiation, and knockdown of LGR5 results in reduced cardiomyocyte differentiation but increased endothelial differentiation.

To read more, please see

Downregulation of LGR5 expression inhibits cardiomyocyte differentiation and potentiates endothelial differentiation from human pluripotent stem cells. Stem Cell Reports. 9:513-527 (2017).

Blood vessels and cardiac muscle cells off the shelf. August, 2017. Lab Land the Emory Health Science Blog.

 

Cardiac disease model in a dish using human induced pluripotent stem cells

Induced pluripotent stem cells (iPSCs) derived from patient’s somatic cells could present an auspicious platform for investigating precision medicine and drug discovery. In a study published in Disease Models & Mechanisms, we demonstrated that clinically observed drug responses can be recapitulated in iPSC-cardiomyocytes derived from an individual with catecholaminergic polymorphic ventricular tachycardia.

To read more, please see:

A stem cell model of catecholaminergic polymorphic ventricular tachycardia recapitulates patient-specific drug responses. Dis Model Mech. 9:927-939 (2016)

Cardiac 'disease in a dish' models advance arrhythmia research. September, 2016.  Emory News Center.

Stem cell stories that caught our eye: improving heart care, fixing sickle cell disease, stem cells & sugar. September, 2016. The Stem Cellar  ̶  The Official Blog of CIRM, California's Stem Cell Agency.

 

A novel nanotechnology tool for quality control of stem cell products

Derivatives from human pluripotent stem cells (hPSCs) have great potential for regenerative medicine; however, residual stem cells in final cell products could be tumorigenic. To ensure the safety of stem cell-based therapies, we have established a new nanotechnology to detect residual stem cells in stem cell products at the sensitivity of several thousand-fold higher compared with that of current assays. The results were published in Biomaterials.

To read more, please see:

Novel surface-enhanced Raman scattering-based assays for ultra-sensitive detection of human pluripotent stem cells.  Biomaterials. 105:66-76 (2016).

Stay out, stray stem cells. August, 2016. Lab Land the Emory Health Science Blog.

Better, faster quality control for stem cell-based therapies. August, 2016. The Stem Cellar  ̶  The Official Blog of CIRM, California's Stem Cell Agency.

Using Raman spectroscopy to identify cell types. December, 2016. The Scientist.

 

Efficient generation of cardiac muscle cells from stem cells using simulated microgravity and 3D culture

Cardiac muscle cells derived from human pluripotent stem cells (hPSCs) could provide an unlimited supply of cells to treat heart diseases in children or adults.  In a study published in Scientific Reports, we found that simulated microgravity and suspension culture of 3D cardiac tissue constructs can increase the efficiency in generating functional cardiac muscle cells from hPSCs.

To read more, please see:

Simulated microgravity and 3D culture enhance induction, viability, proliferation and differentiation of cardiac progenitors from human pluripotent stem cells. Sci Rep. 6:30956 (2016).

Microgravity means more cardiac muscle cells. August, 2016. Lab Land the Emory Health Science Blog.

Pure of heart: How microgravity is improving cardiac cell quality. November, 2017. Upward, magazine of the ISS National Lab


Current Members


Chunhui Xu, PhD, Professor

Email: chunhui.xu@emory.edu; Tel:404-727-7774; Faculty page

 

Antonio Rampoldi, PhD, Postdoctoral Fellow  

Email: antonio.rampoldi@emory.edu; Tel: 404-727-7129; LinkedIn

I completed my PhD in Animal Genetics at the Department of Agricultural Sciences of the Swiss Federal Institute of Technology (ETH) Zurich. I was initially a Postdoctoral Fellow at the Department of Microbiology, Biochemistry & Immunology at Morehouse School of Medicine, Atlanta, Georgia. I am now a member of the Cardiomyocyte Stem Cell Laboratory to gain experience in handling stem cells.

 

 

Parvin Forghani, PhD, Postdoctoral Fellow

Email: parvin.forghani.esfahani@emory.edu; Tel: 404-727-7129; NCBI

I received my PhD in Immunology from Tehran University.  During my project I felt a great sense of pride to work under the supervision of Ned Waller in Winship cancer institute. My research has been focused on the modulation of breast tumor microenvironment, specifically Myeloid Derived Suppressor Cells using anti- and pro- inflammatory mediators. My first year of postdoctoral fellowship lead me to stay at the border of immunology and biology. Now as a new member of the cardiomyocyte stem cell lab, I found the opportunity to continue my ideas with great interest to modulate microenvironment toward maturation of cardiomyocytes. I do believe there is common language between cardio progenitor cells  and cancer cells!

 

Dong Li, PhD, Postdoctoral Fellow

Email: dong.li@emory.edu; Tel: 404-727-7129; LinkedIn

I received my PhD in Cell Biology from University of Copenhagen, Denmark. My dissertation was focused on generating transgene-free human and porcine iPSCs and investigating the mechanisms underlying the iPSC reprogramming. I obtained significant training on neurogenesis of human PSCs and adult stem cells before joining Emory University. My current research focus is on exploring the signaling pathways guiding the differentiation of human PSCs into mature cardiomyocytes and analyzing the protein modifications during maturation using advanced proteomics techniques. 

 

Anita Saraf, MD, PhD, Cardiology Fellow 

Email: anita.saraf@emory.edu; Tel: 404-727-7129; LinkedIn

 

 

 

 

 

 

 

 

Rui Liu (Lemon), MD, Visting Scholar

Email: rui.liu@emory.edu; Tel: 404-789-1524; LinkedIn

I received my MD from Central South University School of Medicine, China. My clinic and research direction is pediatric cardiovascular diseases. At present I am a visiting scholar at the Cardiomyocyte Stem Cell Laboratory to perform scientific research for two years. My current research focus is on screening small molecules to promote the proliferation of cardiomyocytes derived from human pluripotent stem cells through high-throughput screening method.

 

 

Megan Lee, Student 

Email: mlee487@gatech.edu; Linkedin

I am currently a third year student at Georgia Tech majoring in biology with a minor in health and medical sciences. After college I plan on attending medical school in hopes of becoming a pediatrician. I joined the Cardiomyocyte Stem Cell Laboratory to improve my laboratory skills and gain valuable research experience. Since joining Dr. Xu’s lab this past summer, I have learned about immunostaining, calcium imaging, culturing stem cells, and MATLAB.

 

 

Hui Qi Loo, Student

Email: hui.qi.loo@emory.edu; Linkedin

I am currently a sophomore at Emory University majoring in Biology. In addition to working in Dr. Xu’s lab, I am part of the Emory Biotech Club and a STEM mentor in after school science programs. My future goal is to become a biomedical researcher.

 

 

 

 

Nicole Lin, Student

Emial: nicole.lin@emory.edu

 


Past Members


Rajneesh Jha, PhD, Research Associate

Email: rajneesh.jha@emory.edu; Tel: 404-727-7129; ResearchGate; Linkedin

I received my PhD in Biological Sciences from Birla Institute of Technology and Sciences, Pilani, and National Institute of Pathology-ICMR, India. I have gained significant training in stem cell cardiac tissue engineering and expertise in handling human pluripotent stem cell culture, differentiation and characterization of cardiomyocytes using different cellular, molecular and electrophysiological techniques.  My research interests are focused on regulation of cell signaling pathways during the differentiation of human pluripotent stem cells to cardiomyocytes with particular interest to subtype specification and maturation.

 

Cinsley Gentillon, MS, Research Specialist

Email: cinsley.gentillon@emory.edu; Tel: 404-727-7129; LinkedIn

I recently earned my Master of Science degree in Biological Sciences at the University of Delaware. I am extremely grateful to be a member of the Cardiomyocyte Stem Cell Laboratory and look forward to gain expertise in handling human cardiomyocytes derived from pluripotent stem cells. I am interested in attending medical school and hope to utilize my research experience throughout my medical career.

 

 

 

 

Sydney Sharp, Student

Email: sydneybsharp@gmail.comLinkedIn

I am currently a rising sophomore at the University of Maryland, College Park double majoring in Cellular Biology & Molecular Genetics and African American Studies. I am from Silver Spring, MD and graduated from Montgomery Blair High School’s Mathematics, Science, and Computer Science magnet program. Being an intern in the Emory Summer Undergraduate Research Experience (SURE) program will give me the opportunity to gain more research experience by working with cell culture in Dr. Xu’s Lab. My prior research experience includes working at the National Institute of Health (NIH) where I assisted with a research project focused on the development of novel tissue biomarker screening techniques in the rat brain following ischemic brain injury. My plan is to attend medical school in hopes of becoming a pediatrician.

Sameer Sarma, Student

Email: sameersarma0@gmail.com; LinkedIn

I am a rising senior at Tufts University, pursuing an undergraduate major in Biomedical Engineering with a minor in Religion. I am also planning on pursuing a career in medicine. My research experience began in tissue engineering, specifically constructing 3D silk scaffold for culturing and studying primary neuronal cells. My research focus now is shifting to organ regeneration, primarily cardiac regeneration. I am look forward to gaining experience working in the Cardiomyocyte Stem Cell Laboratory, and learning techniques and methods to help me with my own research back at Tufts.

 

Monalisa Singh, MD, PhD, Postdoctoral Fellow   

Email: monalisa.singh@emory.edu; ResearchGate


I'm a physician scientist by training, I received my MD from Indira Gandhi Government Medical College, India and my PhD in pharmaceutical sciences from Mercer University, Atlanta, Georgia. Over the past few years I have gained expertise in bioprocessing of stem cells for tissue engineering and cardiac regenerative medicine applications. My area of research is focused on studying the effects of environmental toxins on stem cell-derived cardiomyocytes and utilizing this platform for potential drug screenings.

 

Makenna Pierce, Student

Email: makenna920@gmail.com

I'm a junior at Druid Hills high school and interested in attending college for biomedical research.

Selected Publications

Forghani P, Rashid A, Armanda LC, Wolfson D, Liu R, Cho HC, Maxwell JT, Jo H, Salaita K, Xu C. Simulated microgravity improves maturation of cardiomyocytes derived from human induced pluripotent stem cells. Sci Rep. In press. doi.10.1038/s41598-024-52453-1.

Hwang H, Rampoldi1 A, Forghani1 P, Li D, Fite J, Boland G, Maher K, Xu C. Space microgravity increases expression of genes associated with proliferation and differentiation in human cardiac spheres. NPJ Microgravity. 9:88 (2023)

Li D, Armand LC, Sun F, Hwang H, Wolfson D, Rampoldi A, Liu R, Forghani P, Hu X, Yu W-M, Qu C-K, Jones DP, Wu R, Cho HC, Maxwell JT, Xu C. AMPK activator-treated human cardiac spheres enhance maturation and enable pathological modeling. Stem Cell Res Ther. 14:322 (2023).

Zheng H, Li Q, Li S, Li Z, Brotto M, Weiss D, Prosdocimo D, Xu C, Reddy A, Puchowicz M, Zhao X, Weitzmann MN, Jain MK, Qu CK. Loss of Ptpmt1 limits mitochondrial utilization of carbohydrates and leads to muscle atrophy and heart failure in tissue-specific knockout mice. Elife. 6;12:RP86944 (2023)

Mandracchia B, Liu W, Hua X, Forghani P, Lee S, Hou J, Nie S, Xu C, Jia S. Optimal sparsity allows reliable system-aware restoration of fluorescence microscopy images. Sci Adv. 9(35): eadg9245 (2023).

Hwang H, Liu R, Eldridge R, Hu X, Forghani P, Jones DP, Xu C. Chronic ethanol exposure induces mitochondrial dysfunction and alters gene expression and metabolism in human cardiac spheroids. Alcohol Clin Exp Res. 47: 643-658 (2023).

Rampoldi A, Forghani F, Li D, Hwang H, Armand LC, Fite J, Boland G, Maxwell J, Maher K, Xu C. Space microgravity improves proliferation of human iPSCderived cardiomyocytes. Stem Cell Rep17:2272-2285(2022)

A 3D Bioprinted in vitro Model of Neuroblastoma Recapitulates Dynamic Tumor-Endothelial Cell Interactions Contributing to Solid Tumor Aggressive Behavior.
Ning L, Shim J, Tomov ML, Liu R, Mehta R, Mingee A, Hwang B, Jin L, Mantalaris A, Xu C, Mahmoudi M, Goldsmith KC, Serpooshan V. Adv Sci. 9:e2200244 (2022)

Forghani P, Rashid A, Sun F, Liu R, Li D, Lee MR, Hwang H, Maxwell1 JT, Mandawat A, Wu R, Salaita K, Xu C. Carfilzomib treatment causes molecular and functional alterations of human induced pluripotent stem cell-derived cardiomyocytes. J Am Heart Assoc. 7:e022247 (2021).

Liu R, Sun F, Armand LC, Wu R, Xu C. Chronic ethanol exposure induces cardiac dysfunction in cardiomyocytes derived from human induced pluripotent stem cells. Stem Cell Rev Rep. 17:2314-2331 (2021).

Rampoldi A, Jha R, Fite J, Boland G, Xu C. Cryopreservation and CO2-independent culture of 3D cardiac progenitors for spaceflight experiments. Biomaterials. 269:120673 (2021). 

Saraf A, Rampoldi A, Chao M, Li D, Armand L, Hwang H, Liu R, Jha R, Fu H, Maxwell JT, Xu C. Functional and molecular effects of TNF-α on human iPSC-derived cardiomyocytes. Stem Cell Res .52:102218 (2021). 

Liu R, Li D, Sun F, Rampoldi A, Maxwell1 JT, Wu R, Fischbach P, Castellino SM, Du Y, Fu H, Mandawat A, Xu C. Melphalan induces cardiotoxicity through oxidative stress in cardiomyocytes derived from human induced pluripotent stem cells. Stem Cell Res Ther. 11:470 (2020)

Hwang H, Liu R, Maxwell JT, Yang J, Xu C. Machine learning identifies abnormal Ca2+ transients in human induced pluripotent stem cell-derived cardiomyocytes. Sci Rep10:16977 (2020)

Liu R, Sun F, Forghani P, Armand LC, Rampoldi A, Li D, Wu R, Xu C. Proteomic profiling reveals roles of stress response, Ca2+ transient dysregulation and novel signaling pathways in alcohol-induced cardiotoxicity. Alcohol Clin Exp Res. 44:2187-2199 (2020)

Jha R, Li D, Wu Q, Ferguson KE, Forghani P, Gibson GC, Xu C. A long non-coding RNA GATA6-AS1 adjacent to GATA6 is required for cardiomyocyte differentiation from human pluripotent stem cells. FASEB J34:14336-14352 (2020)

Gentillon C, Li D, Duan M, Yu W-M, Preininger MK, Jha R, Rampoldi A, Saraf A, Gibson GC, Qu C-K, Brown L, Xu C. Targeting HIF-1a in combination with PPARα activation and postnatal factors promotes the metabolic maturation of human induced pluripotent stem cell-derived cardiomyocytes. J Mol Cell Cardiol. 132, 120-135 (2019)

Rampoldi A, Singh M, Wu Q, Duan M, Jha R, Maxwell JT, Bradner JM, Zhang X, Saraf A, Miller GW, Gibson G, Brown LA, Xu C. Cardiac toxicity from ethanol exposure in human induced pluripotent stem cell-derived cardiomyocytes. Toxicol Sci. 169, 280-292 (2019)

Trac D, Maxwell JT, Brown ME, Xu C, Davis ME. Aggregation of child cardiac progenitor cells into spheres activates notch signaling and improves treatment of right ventricular heart failure. Circ Res. 124, 526-538 (2019)

Rampoldi A, Crooke SN, Preininger MK, Jha R, Maxwell J, Ding L, Spearman P, Finn MG, Xu C. Targeted elimination of tumorigenic human pluripotent stem cells using suicide-inducing virus-like particles. ACS Chem Biol. 13, 2329-2338 (2018)

Maxwell JT, Xu C. Stem-cell-derived cardiomyocytes grow up: start young and train harder. Cell Stem Cell. 22, 790-791 (2018)

Nachlas ALY, Li S, Jha R, Singh M, Xu C, Davis ME. Human iPSC-derived mesenchymal stem cells matured into valve interstitial-like cells using PEGDA hydrogels. Acta Biomater. 71, 235-246 (2018)

Jha R, Singh M, Wu Q, Gentillon C, Preininger MK, Xu C. Downregulation of LGR5 expression inhibits cardiomyocyte differentiation and potentiates endothelial differentiation from human pluripotent stem cells. Stem Cell Reports. 9, 513-527 (2017)

Bylund J, Awgulewitsch CP, Trinh LT, Paik DT, Jetter C, Jha R, Zhang J, Nolan K, Xu C, Thompson T, Kamp TJ, Hatzopoulos AK. Coordinated proliferation and differentiation of human iPS cell-derived cardiac progenitor cells depends on BMP signaling regulation by Gremlin 2. Stem Cells Dev. 26, 678-693 (2017)

Preininger MK, Jha R, Maxwell JT, Wu Q, Singh M, Wang B, Dalal A, Mceachin ZT, Rossoll W, Hales CM, Fischbach P, Wagner MB, Xu C. A stem cell model of catecholaminergic polymorphic ventricular tachycardia recapitulates patient-specific drug responses. Dis Model Mech. 9:927-939 (2016)

Jha R, Wu Q, Singh M, Preininger MK, Han P, Ding G, Cho H, Jo H, Maher KO, Wagner MB, Xu C. Simulated microgravity and 3D culture enhance induction, viability, proliferation and differentiation of cardiac progenitors from human pluripotent stem cells. Sci Rep. 6:30956 (2016)

Han J, Qian X, Wu Q, Jha R, Duan J, Yang Z, Maher KO, Nie S, Xu C. Novel surface-enhanced Raman scattering-based assays for ultra-sensitive detection of human pluripotent stem cells. Biomaterials. 105:66-76 (2016)

Han J, Wu Q, Xia Y, Wagner MB and Xu C. Cell alignment induced by anisotropic electrospun fibrous alone has limited effect on cardiomyocyte maturation. Stem Cell Res. 16, 740-750 (2016)

Jha R, Wile B, Wu Q, Morris AH, Maher KO, Wagner MB, Bao G, Xu C. Molecular beacon-based detection and isolation of working-type cardiomyocytes derived from human pluripotent stem cells. Biomaterials. 50, 176-185 (2015)

Jha R, Xu R.H., Xu C. Efficient differentiation of cardiomyocytes from human pluripotent stem cells with growth factors. Methods Mol Biol. 1299:115-31 (2015)

Nguyen DC, Hookway TA, Wu Q, Jha R, Preininger MK, Chen X, Easley CA, Spearman P, Deshpande SR, Maher K, Wagner MB, McDevitt TC, Xu C. Microscale generation of cardiospheres promotes robust enrichment of cardiomyocytes derived from human pluripotent stem cells. Stem Cell Reports. 3, 260-268 (2014)

Maher KO, Xu C. Marching towards regenerative cardiac therapy with human pluripotent stem cells. Discov Med. 15, 349–356 (2013)

Xu C. Differentiation and enrichment of cardiomyocytes from human pluripotent stem cells. J Mol Cell Cardiol. 52, 1203–1212 (2012)

 

Complete List of Published Work in PubMed and Google Scholar

2023

Engineered cardiac tissues to fly on Northrup Grumman resupply mission to advance treatments for heart disease. July 2023. ISS National Laboratory Press Release.

NG-19 Research: Emory University. July 2023. ISS National Laboratory.

 

2022

Pharma goes to space (Chunhui Xu quoted).  November 2022. Chemical & Engineering News.

Emory heart disease experiment goes to space. November 2022. FOX 5 Atlanta.

Engineered cardiac tissues to fly on NG-18 to advance treatments for heart disease. October 2022. ISS National Laboratory, Center for the Advancement of Science in Space.

Emory Professor working with International Space Station to grow cardiac cells in space. October 2022. Atlanta News First.

 

2021

Finding a key to the heart: stem cells in microgravity could improve cardiac repair. August 2021. ISS National Laboratory, Center for the Advancement of Science in Space.

Cardiac cells that beat in space offer hope for better heart treatment on Earth. July 2021. Emory News Center.

These beating heart cells went to space. This is how they could save patientsJune 2021. CNN.

Researchers develop better ways to culture living heart cells on the International Space Station. May 2021. NASA Space Station Research & Technology.

 

2020

Stem cells & the heart: towards safe and effective stem cell therapies.  2020. Featured innovations, Emory Office of Technology Transfer

Beating heart cells catch a ride to the International Space Station. March 2020. CNN.

SpaceX's CRS-20 Mission to the Space Station: What's On Board. March 2020. NASA

 

2019

Dr. Xu receives Emory I3 Venture Award. May 2019. Lab Land the Emory Health Science Blog.

Alcohol and fetal heart development. Spring 2019. Emory Medicine Magazine.

Fetal alcohol cardiac toxicity in a dish. March 2019. Lab Land the Emory Health Science Blog.

 

2018

Stem cells driven into selective suicide. July 2018.  Lab Land the Emory Health Science Blog.

Dr. Rajneesh Jha gave an oral presentation at the ISS R&D Conference. July 2018.

Exciting time for women in science. May 2018. CASIS.

 

2017

Pure of heart: How microgravity is improving cardiac cell quality. November, 2017. Upward, magazine of the ISS National Lab

Space station will host stem cells for heart research. October, 2017. Emory News Center.

Dr. Xu receives Emory Synergy Award. October, 2017. Emory News Center.

Blood vessels and cardiac muscle cells off the shelf. August, 2017. Lab Land the Emory Health Science Blog.

Cinsley Gentillon won the 2nd Place Poster Award at the Southeast Pediatric Research Conference. June, 2017. News of the Emory / Children's / GT Pediatric Research Alliance.

Dr. Rajneesh Jha gave an oral presentation at the Southeast Pediatric Research Conference. June, 2017. News of the Emory / Children's / GT Pediatric Research Alliance.

 

2016

Dr. Xu received the MilliPub Award.  December, 2016. Celebration of Faculty Excellence Ceremony, Emory University School of Medicine

Using Raman spectroscopy to identify cell types. December, 2016. The Scientist.

Cardiac 'disease in a dish' models advance arrhythmia research. September, 2016. Emory News Center.

Stem cell stories that caught our eye: improving heart care, fixing sickle cell disease, stem cells & sugar. September, 2016. The Stem Cellar  ̶  The Official Blog of CIRM, California's Stem Cell Agency.

Stay out, stray stem cells. August, 2016. Lab Land the Emory Health Science Blog.

Better, faster quality control for stem cell-based therapies. August, 2016. The Stem Cellar  ̶  The Official Blog of CIRM, California's Stem Cell Agency.

Microgravity means more cardiac muscle cells. August, 2016. Lab Land the Emory Health Science Blog.

 

2015

Marcela Preininger won the 1st Place Poster Award at the 5th Pediatric Research Conference. June, 2015. News of the Emory / Children's / GT Pediatric Research Alliance.

Dr. Rajneesh Jha won the Oral Presentation Award at the Postdoctoral Research Symposium at Emory University. May, 2015. News of Emory University School of Medicine.

Faculty recognition photos - Millipub or Emory 1% Awards. March, 2015. News of the Department of Pediatrics, Emory University School of Medicine.

Dr. Xu received the Millipub Award. March, 2015. MilliPub Club, Emory University School of Medicine.

 

2014

Dr. Rajneesh Jha won the 2nd Place Poster Award at the 4th Pediatric Research Conference. April, 2014. News of the Emory / Children's / GT Pediatric Research Alliance.

A CRISPR way to edit DNA.  April, 2014. Emory News Center.

Addendum on CRISPR. April, 2014. Lab Land the Emory Health Science Blog.

 

2013

Dr. Xu has been awarded one of seven grants from the Center for the Advancement of Science in Space (CASIS). October, 2013. CASIS Press releases.

Dr. Doan Nguyen won the 1st place Poster Award at the 3rd Pediatric Research Conference. June, 2013. News of the Emory / Children's / GT Pediatric Research Alliance.

Postdoctoral Fellowship in Stem Cells

We are seeking a postdoctoral fellow to work on human pluripotent stem cells. The research is focused on differentiation and applications of cardiomyocytes from pluripotent stem cells and will be conducted in collaboration with investigators in Children's Healthcare of Atlanta, Georgia Tech, and Emory University.  The candidate should have a recent Ph.D. preferably in Cell Biology or related disciplines with strong cellular and molecular biology expertise including cell culture, immunocytochemistry, flow cytometry analysis, qRT-PCR, generation of expression vectors and animal models. The candidate will conduct independent research and collaborate with members in the group and is expected to actively participate in experimental planning, data analysis, and manuscript writing.

To apply please send curriculum vitae, the names of 3 references, and a cover letter describing your research experience and interests to Dr. Chunhui Xu via email at chunhui.xu@emory.edu.