392 results on '"Joseph C"'
Search Results
2. Sex-Specific Cardiovascular Risks of Cancer and Its Therapies
- Author
-
Wilcox, Nicholas S, Rotz, Seth J, Mullen, McKay, Song, Evelyn J, Hamilton, Betty Ky, Moslehi, Javid, Armenian, Saro H, Wu, Joseph C, Rhee, June-Wha, and Ky, Bonnie
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Cardiovascular ,Regenerative Medicine ,Immunotherapy ,Cancer ,Hematology ,Women's Health ,Heart Disease ,Stem Cell Research ,Stem Cell Research - Nonembryonic - Human ,Estrogen ,Aging ,2.1 Biological and endogenous factors ,Good Health and Well Being ,Anthracyclines ,Antineoplastic Agents ,Cardiotoxicity ,Cardiovascular Diseases ,Female ,Heart Disease Risk Factors ,Hematopoietic Stem Cell Transplantation ,Hormone Replacement Therapy ,Humans ,Male ,Neoplasms ,Sex Characteristics ,anthracyclines ,cardiotoxicity ,heart failure ,thromboembolism ,Cardiorespiratory Medicine and Haematology ,Clinical Sciences ,Cardiovascular System & Hematology ,Cardiovascular medicine and haematology ,Clinical sciences - Abstract
In both cardiovascular disease and cancer, there are established sex-based differences in prevalence and outcomes. Males and females may also differ in terms of risk of cardiotoxicity following cancer therapy, including heart failure, cardiomyopathy, atherosclerosis, thromboembolism, arrhythmias, and myocarditis. Here, we describe sex-based differences in the epidemiology and pathophysiology of cardiotoxicity associated with anthracyclines, hematopoietic stem cell transplant (HCT), hormone therapy and immune therapy. Relative to males, the risk of anthracycline-induced cardiotoxicity is higher in prepubertal females, lower in premenopausal females, and similar in postmenopausal females. For autologous hematopoietic cell transplant, several studies suggest an increased risk of late heart failure in female lymphoma patients, but sex-based differences have not been shown for allogeneic hematopoietic cell transplant. Hormone therapies including GnRH (gonadotropin-releasing hormone) modulators, androgen receptor antagonists, selective estrogen receptor modulators, and aromatase inhibitors are associated with cardiotoxicity, including arrhythmia and venous thromboembolism. However, sex-based differences have not yet been elucidated. Evaluation of sex differences in cardiotoxicity related to immune therapy is limited, in part, due to low participation of females in relevant clinical trials. However, some studies suggest that females are at increased risk of immune checkpoint inhibitor myocarditis, although this has not been consistently demonstrated. For each of the aforementioned cancer therapies, we consider sex-based differences according to cardiotoxicity management. We identify knowledge gaps to guide future mechanistic and prospective clinical studies. Furthering our understanding of sex-based differences in cancer therapy cardiotoxicity can advance the development of targeted preventive and therapeutic cardioprotective strategies.
- Published
- 2022
3. Macrophages
- Author
-
Nishiga, Masataka and Wu, Joseph C
- Subjects
Biomedical and Clinical Sciences ,Cardiovascular Medicine and Haematology ,Clinical Sciences ,COVID-19 ,Humans ,Macrophages ,Myocardium ,SARS-CoV-2 ,Editorials ,macrophage ,myocyte ,cardiac ,pluripotent stem cell ,Cardiorespiratory Medicine and Haematology ,Cardiovascular System & Hematology ,Cardiovascular medicine and haematology ,Clinical sciences - Published
- 2021
4. Generation of Vascular Smooth Muscle Cells From Induced Pluripotent Stem Cells
- Author
-
Shen, Mengcheng, Quertermous, Thomas, Fischbein, Michael P, and Wu, Joseph C
- Subjects
Medical Biotechnology ,Biomedical and Clinical Sciences ,Cardiovascular ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Regenerative Medicine ,Stem Cell Research ,Stem Cell Research - Induced Pluripotent Stem Cell ,Stem Cell Research - Induced Pluripotent Stem Cell - Non-Human ,Aetiology ,2.1 Biological and endogenous factors ,Good Health and Well Being ,Animals ,Cell Differentiation ,Cellular Reprogramming Techniques ,Humans ,Induced Pluripotent Stem Cells ,Muscle ,Smooth ,Vascular ,Myocytes ,Smooth Muscle ,Signal Transduction ,developmental biology ,drug discovery ,pluripotent stem cell ,smooth muscle cell ,tissue engineering ,vascular diseases ,Cardiorespiratory Medicine and Haematology ,Clinical Sciences ,Cardiovascular System & Hematology ,Cardiovascular medicine and haematology ,Clinical sciences - 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.
- Published
- 2021
5. Towards Precision Medicine With Human iPSCs for Cardiac Channelopathies
- Author
-
Wu, Joseph C, Garg, Priyanka, Yoshida, Yoshinori, Yamanaka, Shinya, Gepstein, Lior, Hulot, Jean-Sébastien, Knollmann, Björn C, and Schwartz, Peter J
- Subjects
Biomedical and Clinical Sciences ,Cardiovascular Medicine and Haematology ,Clinical Sciences ,Rare Diseases ,Heart Disease ,Stem Cell Research ,Genetics ,Human Genome ,Cardiovascular ,Orphan Drug ,Stem Cell Research - Embryonic - Human ,Regenerative Medicine ,Clinical Research ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Stem Cell Research - Induced Pluripotent Stem Cell ,Genetic Testing ,Aetiology ,2.1 Biological and endogenous factors ,Good Health and Well Being ,Channelopathies ,Clustered Regularly Interspaced Short Palindromic Repeats ,Humans ,Induced Pluripotent Stem Cells ,Long QT Syndrome ,Precision Medicine ,genetic testing ,induced pluripotent stem cells ,long-QT syndrome ,mutations ,phenotype ,Cardiorespiratory Medicine and Haematology ,Cardiovascular System & Hematology ,Cardiovascular medicine and haematology ,Clinical sciences - Abstract
Long-QT syndrome, a frequently fatal inherited arrhythmia syndrome caused by genetic variants (congenital) or drugs (acquired), affects 1 in 2000 people worldwide. Its sentinel event is often sudden cardiac death, which makes preclinical diagnosis by genetic testing potentially life-saving. Unfortunately, clinical experience with genetic testing has shown that it is difficult to correctly identify genetic variants as disease causing. These current deficiencies in accurately assigning pathogenicity led to the discovery of increasing numbers of rare variants classified as variant of uncertain significance. To overcome these challenges, new technologies such as clustered regularly interspaced short palindromic repeats (CRISPR) genome editing can be combined with human induced pluripotent stem cell-derived cardiomyocytes to provide a new approach to decipher pathogenicity of variants of uncertain significance and to better predict arrhythmia risk. To that end, the overarching goal of our network is to establish the utility of induced pluripotent stem cell-based platforms to solve major clinical problems associated with long-QT syndrome by determining how to (1) differentiate pathogenic mutations from background genetic noise, (2) assess existing and novel variants associated with congenital and acquired long-QT syndrome, and (3) provide genotype- and phenotype- guided risk stratification and pharmacological management of long-QT syndrome. To achieve these goals and to further advance the use of induced pluripotent stem cells in disease modeling and drug discovery, our team of investigators for this Leducq Foundation Transatlantic Networks of Excellence proposal will work together to (1) improve differentiation efficiency, cellular maturation, and lineage specificity, (2) develop new assays for high throughput cellular phenotyping, and (3) train young investigators to clinically implement patient-specific genetic modeling.
- Published
- 2019
6. Generation of Quiescent Cardiac Fibroblasts From Human Induced Pluripotent Stem Cells for In Vitro Modeling of Cardiac Fibrosis
- Author
-
Zhang, Hao, Tian, Lei, Shen, Mengcheng, Tu, Chengyi, Wu, Haodi, Gu, Mingxia, Paik, David T, and Wu, Joseph C
- Subjects
Regenerative Medicine ,Cardiovascular ,Stem Cell Research - Induced Pluripotent Stem Cell ,Stem Cell Research - Embryonic - Human ,Stem Cell Research ,Heart Disease ,Genetics ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,1.1 Normal biological development and functioning ,Aetiology ,Underpinning research ,2.1 Biological and endogenous factors ,Animals ,Antifibrinolytic Agents ,Cells ,Cultured ,Fibroblasts ,Fibrosis ,Humans ,Induced Pluripotent Stem Cells ,Mice ,Myocytes ,Cardiac ,fibroblasts ,fibrosis ,induced pluripotent stem cells ,transcriptome ,Cardiorespiratory Medicine and Haematology ,Clinical Sciences ,Cardiovascular System & Hematology - Abstract
RationaleActivated fibroblasts are the major cell type that secretes 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 fill such gaps of knowledge by enabling delineation of the developmental trajectories and identification of regulatory pathways of tissue-specific fibroblasts among different organs.ObjectiveThis study aims to define the transcriptome profiles of tissue-specific fibroblasts using recently reported mouse single-cell RNA-sequencing atlas and to develop a robust chemically defined protocol to derive cardiac fibroblasts (CFs) from human induced pluripotent stem cells for in vitro modeling of cardiac fibrosis and drug screening.Methods and resultsBy 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 induced pluripotent stem cells. 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 human induced pluripotent stem cell-derived cardiomyocytes and CFs via the atrial/brain natriuretic peptide-natriuretic peptide receptor-1 pathway is implicated in suppressing fibrogenesis.ConclusionsThis study uncovers unique gene signatures that define tissue-specific identities of fibroblasts. The bona fide quiescent CFs derived from human induced pluripotent stem cells can serve as a faithful in vitro platform to better understand the underlying mechanisms of cardiac fibrosis and to screen anti-fibrotic drugs.
- Published
- 2019
7. Convergences of Life Sciences and Engineering in Understanding and Treating Heart Failure.
- Author
-
Berry, Joel L, Zhu, Wuqiang, Tang, Yao Liang, Krishnamurthy, Prasanna, Ge, Ying, Cooke, John P, Chen, Yabing, Garry, Daniel J, Yang, Huang-Tian, Rajasekaran, Namakkal Soorapan, Koch, Walter J, Li, Song, Domae, Keitaro, Qin, Gangjian, Cheng, Ke, Kamp, Timothy J, Ye, Lei, Hu, Shijun, Ogle, Brenda M, Rogers, Jack M, Abel, E Dale, Davis, Michael E, Prabhu, Sumanth D, Liao, Ronglih, Pu, William T, Wang, Yibin, Ping, Peipei, Bursac, Nenad, Vunjak-Novakovic, Gordana, Wu, Joseph C, Bolli, Roberto, Menasché, Philippe, and Zhang, Jianyi
- Subjects
Myocardium ,Heart ,Animals ,Humans ,Interdisciplinary Communication ,Cooperative Behavior ,Biomedical Engineering ,Recovery of Function ,Regeneration ,Biomedical Research ,Diffusion of Innovation ,Heart Failure ,Biological Science Disciplines ,bioengineering ,heart failure ,myocardium ,stem cells ,tissue engineering ,Cardiovascular ,Heart Disease ,Regenerative Medicine ,Stem Cell Research ,Stem Cell Research - Nonembryonic - Human ,Bioengineering ,Cardiorespiratory Medicine and Haematology ,Clinical Sciences ,Cardiovascular System & Hematology - Abstract
On March 1 and 2, 2018, the National Institutes of Health 2018 Progenitor Cell Translational Consortium, Cardiovascular Bioengineering Symposium, was held at the University of Alabama at Birmingham. Convergence of life sciences and engineering to advance the understanding and treatment of heart failure was the theme of the meeting. Over 150 attendees were present, and >40 scientists presented their latest work on engineering human functional myocardium for disease modeling, drug development, and heart failure research. The scientists, engineers, and physicians in the field of cardiovascular sciences met and discussed the most recent advances in their work and proposed future strategies for overcoming the major roadblocks of cardiovascular bioengineering and therapy. Particular emphasis was given for manipulation and using of stem/progenitor cells, biomaterials, and methods to provide molecular, chemical, and mechanical cues to cells to influence their identity and fate in vitro and in vivo. Collectively, these works are profoundly impacting and progressing toward deciphering the mechanisms and developing novel treatments for left ventricular dysfunction of failing hearts. Here, we present some important perspectives that emerged from this meeting.
- Published
- 2019
8. Human Engineered Heart Muscles Engraft and Survive Long Term in a Rodent Myocardial Infarction Model.
- Author
-
Riegler, Johannes, Tiburcy, Malte, Ebert, Antje, Tzatzalos, Evangeline, Raaz, Uwe, Abilez, Oscar J, Shen, Qi, Kooreman, Nigel G, Neofytou, Evgenios, Chen, Vincent C, Wang, Mouer, Meyer, Tim, Tsao, Philip S, Connolly, Andrew J, Couture, Larry A, Gold, Joseph D, Zimmermann, Wolfram H, and Wu, Joseph C
- Subjects
Papillary Muscles ,Cell Line ,Myocytes ,Cardiac ,Animals ,Humans ,Rats ,Nude ,Rats ,Sprague-Dawley ,Myocardial Infarction ,Disease Models ,Animal ,Connexin 43 ,Immunosuppressive Agents ,Stroke Volume ,Heart Transplantation ,Tissue Engineering ,Transfection ,Cell Differentiation ,Cell Survival ,Graft Survival ,Myocardial Contraction ,Time Factors ,Male ,Embryonic Stem Cells ,Heterografts ,Biomarkers ,cardiac MRI ,cardiac function tests ,cell survival ,myocardial infarction ,myocardial ischemia ,tissue engineering ,transplantation ,Cardiovascular ,Heart Disease - Coronary Heart Disease ,Regenerative Medicine ,Stem Cell Research ,Heart Disease ,Transplantation ,Cardiorespiratory Medicine and Haematology ,Clinical Sciences ,Cardiovascular System & Hematology - Abstract
RationaleTissue engineering approaches may improve survival and functional benefits from human embryonic stem cell-derived cardiomyocyte transplantation, thereby potentially preventing dilative remodeling and progression to heart failure.ObjectiveAssessment of transport stability, long-term survival, structural organization, functional benefits, and teratoma risk of engineered heart muscle (EHM) in a chronic myocardial infarction model.Methods and resultsWe constructed EHMs from human embryonic stem cell-derived cardiomyocytes and released them for transatlantic shipping following predefined quality control criteria. Two days of shipment did not lead to adverse effects on cell viability or contractile performance of EHMs (n=3, P=0.83, P=0.87). One month after ischemia/reperfusion injury, EHMs were implanted onto immunocompromised rat hearts to simulate chronic ischemia. Bioluminescence imaging showed stable engraftment with no significant cell loss between week 2 and 12 (n=6, P=0.67), preserving ≤25% of the transplanted cells. Despite high engraftment rates and attenuated disease progression (change in ejection fraction for EHMs, -6.7±1.4% versus control, -10.9±1.5%; n>12; P=0.05), we observed no difference between EHMs containing viable and nonviable human cardiomyocytes in this chronic xenotransplantation model (n>12; P=0.41). Grafted cardiomyocytes showed enhanced sarcomere alignment and increased connexin 43 expression at 220 days after transplantation. No teratomas or tumors were found in any of the animals (n=14) used for long-term monitoring.ConclusionsEHM transplantation led to high engraftment rates, long-term survival, and progressive maturation of human cardiomyocytes. However, cell engraftment was not correlated with functional improvements in this chronic myocardial infarction model. Most importantly, the safety of this approach was demonstrated by the lack of tumor or teratoma formation.
- Published
- 2015
9. Impaired Human Cardiac Cell Development due to NOTCH1 Deficiency
- Author
-
Shiqiao Ye, Cankun Wang, Zhaohui Xu, Hui Lin, Xiaoping Wan, Yang Yu, Subhodip Adhicary, Joe Z. Zhang, Yang Zhou, Chun Liu, Matthew Alonzo, Jianli Bi, Angelina Ramirez-Navarro, Isabelle Deschenes, Qin Ma, Vidu Garg, Joseph C. Wu, and Ming-Tao Zhao
- Subjects
Physiology ,Cardiology and Cardiovascular Medicine - Abstract
Background: NOTCH1 pathogenic variants are implicated in multiple types of congenital heart defects including hypoplastic left heart syndrome, where the left ventricle is underdeveloped. It is unknown how NOTCH1 regulates human cardiac cell lineage determination and cardiomyocyte proliferation. In addition, mechanisms by which NOTCH1 pathogenic variants lead to ventricular hypoplasia in hypoplastic left heart syndrome remain elusive. Methods: CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats)/Cas9 genome editing was utilized to delete NOTCH1 in human induced pluripotent stem cells. Cardiac differentiation was carried out by sequential modulation of WNT signaling, and NOTCH1 knockout and wild-type differentiating cells were collected at day 0, 2, 5, 10, 14, and 30 for single-cell RNA-seq. Results: Human NOTCH1 knockout induced pluripotent stem cells are able to generate functional cardiomyocytes and endothelial cells, suggesting that NOTCH1 is not required for mesoderm differentiation and cardiovascular development in vitro. However, disruption of NOTCH1 blocks human ventricular-like cardiomyocyte differentiation but promotes atrial-like cardiomyocyte generation through shortening the action potential duration. NOTCH1 deficiency leads to defective proliferation of early human cardiomyocytes, and transcriptomic analysis indicates that pathways involved in cell cycle progression and mitosis are downregulated in NOTCH1 knockout cardiomyocytes. Single-cell transcriptomic analysis reveals abnormal cell lineage determination of cardiac mesoderm, which is manifested by the biased differentiation toward epicardial and second heart field progenitors at the expense of first heart field progenitors in NOTCH1 knockout cell populations. Conclusions: NOTCH1 is essential for human ventricular-like cardiomyocyte differentiation and proliferation through balancing cell fate determination of cardiac mesoderm and modulating cell cycle progression. Because first heart field progenitors primarily contribute to the left ventricle, we speculate that pathogenic NOTCH1 variants lead to biased differentiation of first heart field progenitors, blocked ventricular-like cardiomyocyte differentiation, and defective cardiomyocyte proliferation, which collaboratively contribute to left ventricular hypoplasia in hypoplastic left heart syndrome.
- Published
- 2023
10. Microfluidic Single-Cell Analysis Shows That Porcine Induced Pluripotent Stem Cell–Derived Endothelial Cells Improve Myocardial Function by Paracrine Activation
- Author
-
Gu, Mingxia, Nguyen, Patricia K, Lee, Andrew S, Xu, Dan, Hu, Shijun, Plews, Jordan R, Han, Leng, Huber, Bruno C, Lee, Won Hee, Gong, Yongquan, de Almeida, Patricia E, Lyons, Jennifer, Ikeno, Fumi, Pacharinsak, Cholawat, Connolly, Andrew J, Gambhir, Sanjiv S, Robbins, Robert C, Longaker, Michael T, and Wu, Joseph C
- Subjects
Medical Biotechnology ,Biomedical and Clinical Sciences ,Stem Cell Research ,Cardiovascular ,Heart Disease - Coronary Heart Disease ,Heart Disease ,Stem Cell Research - Induced Pluripotent Stem Cell ,Biotechnology ,Regenerative Medicine ,Stem Cell Research - Induced Pluripotent Stem Cell - Non-Human ,Transplantation ,Development of treatments and therapeutic interventions ,5.2 Cellular and gene therapies ,Animals ,Cell Differentiation ,Cell Survival ,Cell Transplantation ,Cells ,Cultured ,Echocardiography ,Endothelium ,Vascular ,Female ,Heart ,Magnetic Resonance Imaging ,Mice ,Mice ,SCID ,Microfluidic Analytical Techniques ,Models ,Animal ,Myocardial Infarction ,Myocytes ,Cardiac ,Neovascularization ,Physiologic ,Paracrine Communication ,Pluripotent Stem Cells ,Swine ,Swine ,Miniature ,Cardiorespiratory Medicine and Haematology ,Clinical Sciences ,Cardiovascular System & Hematology ,Cardiovascular medicine and haematology ,Clinical sciences - Abstract
RationaleInduced pluripotent stem cells (iPSCs) hold great promise for the development of patient-specific therapies for cardiovascular disease. However, clinical translation will require preclinical optimization and validation of large-animal iPSC models.ObjectiveTo successfully derive endothelial cells from porcine iPSCs and demonstrate their potential utility for the treatment of myocardial ischemia.Methods and resultsPorcine adipose stromal cells were reprogrammed to generate porcine iPSCs (piPSCs). Immunohistochemistry, quantitative PCR, microarray hybridization, and angiogenic assays confirmed that piPSC-derived endothelial cells (piPSC-ECs) shared similar morphological and functional properties as endothelial cells isolated from the autologous pig aorta. To demonstrate their therapeutic potential, piPSC-ECs were transplanted into mice with myocardial infarction. Compared with control, animals transplanted with piPSC-ECs showed significant functional improvement measured by echocardiography (fractional shortening at week 4: 27.2±1.3% versus 22.3±1.1%; P
- Published
- 2012
11. Abstract P3060: Modeling Immune Checkpoint Inhibitor Associated Myocarditis In Vitro
- Author
-
Jensen, Garrett, primary, Wang, Xinjie, additional, Zhao, Wei, additional, chen, Zhishi, additional, Yu, Wei, additional, Palaskas, Nicolas L, additional, Stephan, Clifford, additional, Powell, Reid, additional, Wu, Joseph C, additional, Luo, Weijia, additional, and Chang, Jiang, additional
- Published
- 2023
- Full Text
- View/download PDF
12. Abstract P1033: Derivation Of Large Biorepository Of Human Ipsc Lines For Open Access Sharing Among Academic Investigators
- Author
-
Yan, Christopher D, primary, Lai, Celine J, additional, Shenoy, Sushma P, additional, Lee, Chelsea, additional, Guevara, Julio V, additional, Zhuge, Yan, additional, Pang, Paul D, additional, and Wu, Joseph C, additional
- Published
- 2023
- Full Text
- View/download PDF
13. Tumor Repressor Circular RNA as a New Target for Preventative Gene Therapy Against Doxorubicin-Induced Cardiotoxicity
- Author
-
Jahng, James W.S., Liu, Lichao, and Wu, Joseph C.
- Published
- 2020
- Full Text
- View/download PDF
14. RNA Sequencing Analysis of Induced Pluripotent Stem Cell-Derived Cardiomyocytes From Congenital Heart Disease Patients
- Author
-
Kitani, Tomoya, Tian, Lei, Zhang, Tiejun, Itzhaki, Ilanit, Zhang, Joe Z., Ma, Ning, Liu, Chun, Rhee, June-Wha, Romfh, Anitra W., Lui, George K., and Wu, Joseph C.
- Published
- 2020
- Full Text
- View/download PDF
15. KMT2D-NOTCH Mediates Coronary Abnormalities in Hypoplastic Left Heart Syndrome
- Author
-
Zhiyun Yu, Xin Zhou, Ziyi Liu, Victor Pastrana-Gomez, Yu Liu, Minzhe Guo, Lei Tian, Timothy J. Nelson, Nian Wang, Seema Mital, David Chitayat, Joseph C. Wu, Marlene Rabinovitch, Sean M. Wu, Michael P. Snyder, Yifei Miao, and Mingxia Gu
- Subjects
Heart Defects, Congenital ,JAG1 ,medicine.medical_specialty ,education.field_of_study ,Heart disease ,Physiology ,business.industry ,Population ,Notch signaling pathway ,Hemodynamics ,medicine.disease ,Coronary Vessels ,Hypoplastic left heart syndrome ,medicine.anatomical_structure ,Ventricle ,Internal medicine ,Hypoplastic Left Heart Syndrome ,medicine ,Cardiology ,Humans ,Cardiology and Cardiovascular Medicine ,education ,business ,Artery - Abstract
Hypoplastic left heart syndrome (HLHS) is a severe form of single ventricle congenital heart disease characterized by an underdevelopment of the left ventricle. Early serial postmortem examinations revealed high rate of coronary artery abnormalities in HLHS fetal hearts, which may impact ventricular development and intra-cardiac hemodynamics, leading to a poor prognosis after surgical palliations. Previous study reported that endothelial cells (ECs) lining the coronary vessels showed DNA damage in the left ventricle of human fetal heart with HLHS, indicating that EC dysfunction may contribute to the coronary abnormalities in HLHS. To investigate the underlying mechanism of HLHS coronary artery abnormalities, we profiled both human fetal heart with an underdeveloped left ventricle (ULV) and ECs differentiated from induced pluripotent stem cells (iPSCs) derived from HLHS patients at single cell resolution. CD144+/NPR3- vascular ECs were selected and further classified as venous EC (NR2F2high), arterial EC (EFNB2high) and late arterial EC (GJA5high) subclusters based on previously reported marker genes. To study the arterial phenotype, we specifically generated iPSC-arterial ECs (AECs, CD34+CDH5+CXCR4+NT5E-/low) derived from three HLHS patients and three age-matched healthy controls to further dissect the phenotype of HLHS-AECs. As compared to normal human heart and control iPSC-ECs respectively, ULV late arterial EC subcluster and HLHS iPSC-EC arterial clusters showed significantly reduced expression of arterial genes GJA5, DLL4, and HEY1. Pathway enrichment analysis based on differentially expressed genes revealed several defects in late AEC cluster from ULV compared to normal human heart, such as impaired endothelial proliferation, development and Notch signaling. HLHS iPSCs exhibited impaired AEC differentiation as evidenced by the significantly reduced CXCR4+NT5E-/low AEC progenitor population. Consistent with human heart transcriptomic data, matured HLHS iPSC-AECs also showed a lower expression of the arterial genes such as GJA5, DLL4, HEY1 which are also downstream of NOTCH signaling. Additionally, matured HLHS iPSC-AECs showed significantly decreased expression of cell proliferation marker Ki67 and G1/S transition genes (CCND1, CCND2) compared with controls. Interestingly, NOTCH ligand Jag1 treatment significantly rescued this cell proliferation defect in HLHS AECs, accompanied by upregulation of various G1/S transition genes. In summary, we found that coronary AECs from HLHS showed impaired arterial development and proliferation downstream of NOTCH signaling. These functional defects in HLHS coronary AECs could contribute to the vascular structure malformation and impaired ventricular development.
- Published
- 2023
16. Cardiac Myocyte-Specific Excision of the β1 Integrin Gene Results in Myocardial Fibrosis and Cardiac Failure
- Author
-
Shai, Shaw-Yung, Harpf, Alice E, Babbitt, Christopher J, Jordan, Maria C, Fishbein, Michael C, Chen, Ju, Omura, Michelle, Leil, Tarek A, Becker, K David, Jiang, Meisheng, Smith, Desmond J, Cherry, Simon R, Loftus, Joseph C, and Ross, Robert S
- Subjects
Medical Physiology ,Biomedical and Clinical Sciences ,Heart Disease ,Cardiovascular ,Aetiology ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Underpinning research ,Animals ,Cardiomyopathy ,Dilated ,Cell Membrane ,Cell Membrane Permeability ,Disease Progression ,Fibrosis ,Gene Targeting ,Glucose ,Heart Failure ,Heart Ventricles ,Homozygote ,Integrases ,Integrin beta1 ,Mice ,Mice ,Knockout ,Myocardium ,Organ Specificity ,Talin ,Viral Proteins ,extracellular matrix ,homologous recombination ,Cre recombinase ,heart ,positron emission tomography ,Cardiorespiratory Medicine and Haematology ,Clinical Sciences ,Cardiovascular System & Hematology ,Cardiovascular medicine and haematology ,Clinical sciences - Abstract
Integrins link the extracellular matrix to the cellular cytoskeleton and serve important roles in cell growth, differentiation, migration, and survival. Ablation of beta1 integrin in all murine tissues results in peri-implantation embryonic lethality. To investigate the role of beta1 integrin in the myocardium, we used Cre-LoxP technology to inactivate the beta1 integrin gene exclusively in ventricular cardiac myocytes. Animals with homozygous ventricular myocyte beta1 integrin gene excision were born in appropriate numbers and grew into adulthood. These animals had 18% of control levels of beta1D integrin protein in the heart and displayed myocardial fibrosis. High-fidelity micromanometer-tipped catheterization of the intact 5-week-old beta1 integrin knockout mice showed depressed left ventricular basal and dobutamine-stimulated contractility and relaxation (LV dP/dt(max) and LV dP/dt(min)) as compared with control groups (n=8 to 10 of each, P
- Published
- 2002
17. Cellular and Engineered Organoids for Cardiovascular Models
- Author
-
Dilip Thomas, Suji Choi, Christina Alamana, Kevin Kit Parker, and Joseph C. Wu
- Subjects
Organoids ,Pluripotent Stem Cells ,Physiology ,Induced Pluripotent Stem Cells ,Models, Cardiovascular ,Humans ,Cell Differentiation ,Heart ,Cardiology and Cardiovascular Medicine - Abstract
An ensemble of in vitro cardiac tissue models has been developed over the past several decades to aid our understanding of complex cardiovascular disorders using a reductionist approach. These approaches often rely on recapitulating single or multiple clinically relevant end points in a dish indicative of the cardiac pathophysiology. The possibility to generate disease-relevant and patient-specific human induced pluripotent stem cells has further leveraged the utility of the cardiac models as screening tools at a large scale. To elucidate biological mechanisms in the cardiac models, it is critical to integrate physiological cues in form of biochemical, biophysical, and electromechanical stimuli to achieve desired tissue-like maturity for a robust phenotyping. Here, we review the latest advances in the directed stem cell differentiation approaches to derive a wide gamut of cardiovascular cell types, to allow customization in cardiac model systems, and to study diseased states in multiple cell types. We also highlight the recent progress in the development of several cardiovascular models, such as cardiac organoids, microtissues, engineered heart tissues, and microphysiological systems. We further expand our discussion on defining the context of use for the selection of currently available cardiac tissue models. Last, we discuss the limitations and challenges with the current state-of-the-art cardiac models and highlight future directions.
- Published
- 2022
18. Use of Human Induced Pluripotent Stem Cell–Derived Cardiomyocytes in Preclinical Cancer Drug Cardiotoxicity Testing: A Scientific Statement From the American Heart Association
- Author
-
Gintant, Gary, Burridge, Paul, Gepstein, Lior, Harding, Sian, Herron, Todd, Hong, Charles, Jalife, José, and Wu, Joseph C.
- Published
- 2019
- Full Text
- View/download PDF
19. Workshop Report: FDA Workshop on Improving Cardiotoxicity Assessment With Human-Relevant Platforms
- Author
-
Pang, Li, Sager, Philip, Yang, Xi, Shi, Hong, Sannajust, Frederick, Brock, Mathew, Wu, Joseph C., Abi-Gerges, Najah, Lyn-Cook, Beverly, Berridge, Brian R., and Stockbridge, Norman
- Published
- 2019
- Full Text
- View/download PDF
20. Transcriptomic Profiling of the Developing Cardiac Conduction System at Single-Cell Resolution
- Author
-
Goodyer, William R., Beyersdorf, Benjamin M., Paik, David T., Tian, Lei, Li, Guang, Buikema, Jan W., Chirikian, Orlando, Choi, Shannon, Venkatraman, Sneha, Adams, Eliza L., Tessier-Lavigne, Marc, Wu, Joseph C., and Wu, Sean M.
- Published
- 2019
- Full Text
- View/download PDF
21. Identifying the Transcriptome Signatures of Calcium Channel Blockers in Human Induced Pluripotent Stem Cell–Derived Cardiomyocytes
- Author
-
Lam, Chi Keung, Tian, Lei, Belbachir, Nadjet, Wnorowski, Alexa, Shrestha, Rajani, Ma, Ning, Kitani, Tomoya, Rhee, June-Wha, and Wu, Joseph C.
- Published
- 2019
- Full Text
- View/download PDF
22. Proteasome-Dependent Regulation of Distinct Metabolic States During Long-Term Culture of Human iPSC-Derived Cardiomyocytes
- Author
-
Ebert, Antje, Joshi, Amit U., Andorf, Sandra, Dai, Yuanyuan, Sampathkumar, Shrivatsan, Chen, Haodong, Li, Yingxin, Garg, Priyanka, Toischer, Karl, Hasenfuss, Gerd, Mochly-Rosen, Daria, and Wu, Joseph C.
- Published
- 2019
- Full Text
- View/download PDF
23. Stanford Cardiovascular Institute: At the Forefront of Cardiovascular Research
- Author
-
Wu, Joseph C., Woo, Y. Joseph, Mayerle, Megan, Harrington, Robert A., and Quertermous, Thomas
- Published
- 2019
- Full Text
- View/download PDF
24. Impaired Human Cardiac Cell Development due to NOTCH1 Deficiency
- Author
-
Ye, Shiqiao, primary, Wang, Cankun, additional, Xu, Zhaohui, additional, Lin, Hui, additional, Wan, Xiaoping, additional, Yu, Yang, additional, Adhicary, Subhodip, additional, Zhang, Joe Z., additional, Zhou, Yang, additional, Liu, Chun, additional, Alonzo, Matthew, additional, Bi, Jianli, additional, Ramirez-Navarro, Angelina, additional, Deschenes, Isabelle, additional, Ma, Qin, additional, Garg, Vidu, additional, Wu, Joseph C., additional, and Zhao, Ming-Tao, additional
- Published
- 2023
- Full Text
- View/download PDF
25. Ferroptosis of Pacemaker Cells in COVID-19
- Author
-
Masataka Nishiga, James W.S. Jahng, and Joseph C. Wu
- Subjects
Physiology ,Cell Line, Tumor ,COVID-19 ,Ferroptosis ,Humans ,Myocytes, Cardiac ,Cardiology and Cardiovascular Medicine - Published
- 2022
26. From the BCVS Chair: Joseph C. Wu
- Author
-
Wu, Joseph C.
- Published
- 2018
- Full Text
- View/download PDF
27. Strategies for Improving the Maturity of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes
- Author
-
Tu, Chengyi, Chao, Benjamin S., and Wu, Joseph C.
- Published
- 2018
- Full Text
- View/download PDF
28. Large-Scale Single-Cell RNA-Seq Reveals Molecular Signatures of Heterogeneous Populations of Human Induced Pluripotent Stem Cell-Derived Endothelial Cells
- Author
-
Paik, David T., Tian, Lei, Lee, Jaecheol, Sayed, Nazish, Chen, Ian Y., Rhee, Siyeon, Rhee, June-Wha, Kim, Youngkyun, Wirka, Robert C., Buikema, Jan W., Wu, Sean M., Red-Horse, Kristy, Quertermous, Thomas, and Wu, Joseph C.
- Published
- 2018
- Full Text
- View/download PDF
29. Human Induced Pluripotent Stem Cell–Derived Cardiomyocytes as Models for Cardiac Channelopathies: A Primer for Non-Electrophysiologists
- Author
-
Garg, Priyanka, Garg, Vivek, Shrestha, Rajani, Sanguinetti, Michael C., Kamp, Timothy J., and Wu, Joseph C.
- Published
- 2018
- Full Text
- View/download PDF
30. Global Overview of the Transnational Alliance for Regenerative Therapies in Cardiovascular Syndromes (TACTICS) Recommendations: A Comprehensive Series of Challenges and Priorities of Cardiovascular Regenerative Medicine
- Author
-
Fernández-Avilés, Francisco, Sanz-Ruiz, Ricardo, Climent, Andreu M., Badimon, Lina, Bolli, Roberto, Charron, Dominique, Fuster, Valentin, Janssens, Stefan, Kastrup, Jens, Kim, Hyo-Soo, Lüscher, Thomas F., Martin, John F., Menasché, Philippe, Pinto, Fausto J., Simari, Robert D., Stone, Gregg W., Terzic, Andre, Willerson, James T., and Wu, Joseph C.
- Published
- 2018
- Full Text
- View/download PDF
31. Cardiac Cell Cycle Activation as a Strategy to Improve iPSC-Derived Cardiomyocyte Therapy
- Author
-
Rhee, June-Wha and Wu, Joseph C.
- Published
- 2018
- Full Text
- View/download PDF
32. Abstract P3005: Cannabinoid Receptor 1 Antagonist Genistein Attenuates Marijuana-Induced Vascular Inflammation
- Author
-
Mark Chandy, Tzu-tang T Wei, Masataka Nishiga, Angela Zhang, Kaavya K Kumar, Dilip Thomas, Amit Manhas, Siyeon Rhee, Johanne M Justesen, Ian Y Chen, Hung-Ta Wo, Johnson Y Yang, Saereh Khanamiri, Frederick Seidl, Noah Burns, Chun Liu, Nazish Sayed, Jiun-Jie Shie, Chih Fan Yeh, Kai-chien YANG, Edward Lau, Kara Lynch, Manuel Rivas, Brian Kobilka, and Joseph C Wu
- Subjects
Physiology ,Cardiology and Cardiovascular Medicine - Abstract
Epidemiological studies reveal that marijuana increases the risk of cardiovascular disease (CVD); however, little is known about the mechanism. Δ 9 -tetrahydrocannabinol (Δ 9 -THC), the psychoactive component of marijuana, binds cannabinoid receptor 1 (CB1/CNR1) in the vasculature and is implicated in CVD. A UK Biobank analysis found that cannabis is an independent risk factor for CVD. We found that marijuana smoking activated inflammatory cytokines implicated in CVD. In silico virtual screening identified genistein, a soybean isoflavone, as a putative CB1 antagonist. Human-induced pluripotent stem cell-derived endothelial cells (hiPSC-ECs) were used to model Δ 9 -THC induced inflammation and oxidative stress via NF-κB signaling. Knockdown of the CB1 receptor with siRNA, CRISPR interference (CRISPRi), and genistein attenuate the effects of Δ 9 -THC. In mice, genistein blocked Δ 9 -THC-induced endothelial dysfunction in wire myograph, reduced atherosclerotic plaque, and had minimal penetration of the central nervous system (CNS). Genistein is a peripherally restricted CB1 antagonist that attenuates Δ 9 -THC-induced atherosclerosis.
- Published
- 2022
33. Abstract P2109: Elaborating Safety Margins To Predict Drug Proarrhythmia Using Deep Learning And Patient-derived IPSCs
- Author
-
Ricardo Serrano, Dries Feyen, Arne A Bruyneel, Anna Hnatiuk Hnatiuk, Michelle Vu, Prashila Amatya, Isaac Perea Gil, Maricela Prado, Timon Seeger, Joseph C Wu, Ioannis Karakikes, and Mark Mercola
- Subjects
Physiology ,Cardiology and Cardiovascular Medicine - Abstract
Introduction: Drug-induced arrhythmias are a common cause for drug attrition during development and for restricted use or withdrawal from the market. Cell based assays to assess arrhythmia risk typically rely on the quantification of waveform features - e.g. action potential prolongation and after depolarizations - in the cells’ action potential. However, the predictive power of these approaches is limited. Hypothesis: We hypothesize that deep learning can extract features relevant to discriminating input classes in a systematic and unbiased manner, effectively removing the need for human-defined metrics. This can lead to a new model to estimate torsadogenic risk of drugs and evaluate the influence of myopathic gene variants on drug-induced arrhythmia. Methods: We optically recorded action potentials optically recorded for 40 drugs - characterized high, intermediate, and low or no torsadogenic risk in patients- at 8 concentrations in hiPSC-CMs from 3 healthy donors and 5 hiPSC-CMs isogenic lines carrying 5 gene variants that cause dilated and hypertrophic cardiomyopathies. We designed a convolutional neural network (CNN) to classify non-arrhythmic, arrhythmic and asystolic traces in hiPSC-CMs. Using the class probabilities measured by the CNN, we created torsadogenic and asystolic safety margins for each drug and cell line. Results: The arrhythmic class probability computed by the CNN, provided a continuous, dose-dependent metric of the proarrhythmic risk of drugs in healthy and cardiomyopathic hiPSC-CMs. We used this metric to estimate safety margins for drug-induced arrhythmia and achieved a 0.942 AUC in classifying drugs of high-intermediate risk from safe ones. We used this approach to discern the contribution of putative genetic risk factors to arrhythmia susceptibility by comparing the risk profiles of the same drugs in healthy and isogenic hiPSC-CMs carrying causal HCM and DCM gene variants that are associated with arrhythmia in patients. Conclusions: We conclude that deep learning algorithms can effectively evaluate proarrhythmic risk of small molecules. Moreover, they can also be used to discern heightened arrhythmic risk caused by genetic mutations that increase the propensity for drug-induced arrhythmia in patients.
- Published
- 2022
34. Abstract P3120: Cardio And Neurotoxicity Of Repurposed Anti-COVID-19 Drugs
- Author
-
Martin Nicholson, Ching-Ying Huang, Jyun-Yuan Wang, Ting Chien-Yu, Yu-Che Cheng, Darien Chan, Yi-Chan Lee, Ching-Chuan Hsu, Cindy M Chang, Marvin L. Hsieh, Yuan-Yuan Cheng, Yi-Ling Lin, Chien-Hsiun Chen, Ying-Ta Wu, Timothy A Hacker, Joseph C Wu, Timothy Kamp, and Patrick C Hsieh
- Subjects
Physiology ,Cardiology and Cardiovascular Medicine - Abstract
In December 2019, the novel coronavirus disease 2019 (COVID-19), caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) spread around the globe resulting in ~435 million confirmed cases and ~6 million related deaths as of March 2022, according to the World Health Organization. To combat COVID-19 quickly, there have been many attempts to repurpose current FDA-approved drugs or to revive old drugs with anti-viral properties. However, aside from the biological stress imposed by the virus, many of the current treatment options have been known to cause adverse drug reactions. We established a population-based human induced pluripotent stem cell drug screening platform to assess the toxicity of the first line of anti-COVID-19 drugs and to understand viral infection of cardiomyocytes and neurons. We found that iPSC-derived cardiomyocytes express the ACE2 receptor which correlated with a higher infection of the SARS-CoV-2 virus (r=0.86). However, ACE2 expression was undetectable in neurons which correlated with low infection of neurons. We then assessed the toxicity of anti-COVID-19 drugs and identified two cardiotoxic compounds (remdesivir and arbidol) and 4 neurotoxic compounds (arbidol, remdesivir, hydroxychloroquine, and chloroquine) which were validated by dose-response curves. These data show that this platform can quickly and easily be employed to further our understanding of cell-specific infection and identify drug toxicity of potential treatment options helping clinicians better decide on treatment options.
- Published
- 2022
35. Abstract P2115: Differential Cardiac Remodeling Profile Of Immunosuppression Drugs
- Author
-
Karim Sallam, Dilip Thomas, Sadhana Gaddam, Nicole Lopez, Aimee Beck, Ryan Dexheimer, Leila Y Beach, Albert J Rogers, Hao Zhang, Ian Y Chen, Mo Ameen, William Hiesinger, Jeffrey Teuteberg, June W Rhee, Kevin Wang, Nazish Sayed, and Joseph C Wu
- Subjects
Physiology ,Cardiology and Cardiovascular Medicine - Abstract
Introduction: Heart transplantation provides lifesaving therapy for patients with end-stage heart failure. The longevity of the therapy is limited by Cardiac Graft Dysfunction (CGD), which is an acquired cardiomyopathy affecting transplanted hearts associated with diastolic and/or systolic dysfunction. Some clinical risk factors for CGD have been identified, but none of them are easily modifiable. An unexplored potential contributor to CGD is the choice of immunosuppression agent used despite multiple clinical reports suggesting reduced adverse cardiac remodeling with mammalian target of rapamycin (mTOR) inhibitors compared to calcineurin inhibitors (CNI). This study examines mechanisms of differential cardiac remodeling effects of CNI versus mTOR inhibitors in a human cellular cardiac model. Methods/Results: We utilized 3D cardiac spheres composed of induced pluripotent stem cell-derived cardiomyocytes, cardiac fibroblasts, and endothelial cells (cardiac organoids). Cardiac organoids were treated with 5 days of vehicle, tacrolimus (CNI), or sirolimus (mTOR inhibitor). We did not observe a significant difference in surrogates of systolic or diastolic function in treated cardiac organoids. We pursued single cell-RNA sequencing of drug-treated cardiac organoids and identified gene expression changes consistent with increased extracellular matrix deposition and fibroblast activity in response to CNI treatment. In addition, CNI-treated cardiac organoids cellular composition was notable for increased proportion of fibroblasts and less cardiomyocytes compared to mTOR inhibitor-treated cardiac organoids. To validate gene expression changes observed, we treated cardiac fibroblasts with drugs and observed an increase in collagen production in response to CNI treatment and a reduction in fibroblast number and collagen production in response to mTOR inhibitor treatment. Furthermore, we observed increased ATP production in CNI-treated cardiac fibroblasts, but a reduction in mTOR-treated counterparts. Conclusion: We identify reduced extracellular matrix deposition and cardiac fibroblast proliferation in response to mTOR inhibitor as a potential mechanism for the more favorable remodeling profile observed clinically.
- Published
- 2022
36. Cardiopulmonary Consequences of Vaping in Adolescents: A Scientific Statement From the American Heart Association
- Author
-
Loren E, Wold, Robert, Tarran, Laura E, Crotty Alexander, Naomi M, Hamburg, Farrah, Kheradmand, Gideon, St Helen, and Joseph C, Wu
- Subjects
Smokers ,Vaping ,Humans ,Smoking Cessation ,American Heart Association ,Electronic Nicotine Delivery Systems - Abstract
Although the US Food and Drug Administration has not approved e-cigarettes as a cessation aid, industry has at times positioned their products in that way for adults trying to quit traditional cigarettes; however, their novelty and customizability have driven them into the hands of unintended users, particularly adolescents. Most new users of e-cigarette products have never smoked traditional cigarettes; therefore, understanding the respiratory and cardiovascular consequences of e-cigarette use has become of increasing interest to the research community. Most studies have been performed on adult e-cigarette users, but the majority of these study participants are either former traditional smokers or smokers who have used e-cigarettes to switch from traditional smoking. Therefore, the respiratory and cardiovascular consequences in this population are not attributable to e-cigarette use alone. Preclinical studies have been used to study the effects of naive e-cigarette use on various organ systems; however, almost all of these studies have used adult animals, which makes translation of health effects to adolescents problematic. Given that inhalation of any foreign substance can have effects on the respiratory and cardiovascular systems, a more holistic understanding of the pathways involved in toxicity could help to guide researchers to novel therapeutic treatment strategies. The goals of this scientific statement are to provide salient background information on the cardiopulmonary consequences of e-cigarette use (vaping) in adolescents, to guide therapeutic and preventive strategies and future research directions, and to inform public policymakers on the risks, both short and long term, of vaping.
- Published
- 2022
37. Abstract P3005: Cannabinoid Receptor 1 Antagonist Genistein Attenuates Marijuana-Induced Vascular Inflammation
- Author
-
Chandy, Mark, primary, Wei, Tzu-tang T, additional, Nishiga, Masataka, additional, Zhang, Angela, additional, Kumar, Kaavya K, additional, Thomas, Dilip, additional, Manhas, Amit, additional, Rhee, Siyeon, additional, Justesen, Johanne M, additional, Chen, Ian Y, additional, Wo, Hung-Ta, additional, Yang, Johnson Y, additional, Khanamiri, Saereh, additional, Seidl, Frederick, additional, Burns, Noah, additional, Liu, Chun, additional, Sayed, Nazish, additional, Shie, Jiun-Jie, additional, Yeh, Chih Fan, additional, YANG, Kai-chien, additional, Lau, Edward, additional, Lynch, Kara, additional, Rivas, Manuel, additional, Kobilka, Brian, additional, and Wu, Joseph C, additional
- Published
- 2022
- Full Text
- View/download PDF
38. Abstract P3120: Cardio And Neurotoxicity Of Repurposed Anti-COVID-19 Drugs
- Author
-
Nicholson, Martin, primary, Huang, Ching-Ying, additional, Wang, Jyun-Yuan, additional, Chien-Yu, Ting, additional, Cheng, Yu-Che, additional, Chan, Darien, additional, Lee, Yi-Chan, additional, Hsu, Ching-Chuan, additional, Chang, Cindy M, additional, Hsieh, Marvin L., additional, Cheng, Yuan-Yuan, additional, Lin, Yi-Ling, additional, Chen, Chien-Hsiun, additional, Wu, Ying-Ta, additional, Hacker, Timothy A, additional, Wu, Joseph C, additional, Kamp, Timothy, additional, and Hsieh, Patrick C, additional
- Published
- 2022
- Full Text
- View/download PDF
39. Abstract P2115: Differential Cardiac Remodeling Profile Of Immunosuppression Drugs
- Author
-
Sallam, Karim, primary, Thomas, Dilip, additional, Gaddam, Sadhana, additional, Lopez, Nicole, additional, Beck, Aimee, additional, Dexheimer, Ryan, additional, Beach, Leila Y, additional, Rogers, Albert J, additional, Zhang, Hao, additional, Chen, Ian Y, additional, Ameen, Mo, additional, Hiesinger, William, additional, Teuteberg, Jeffrey, additional, Rhee, June W, additional, Wang, Kevin, additional, Sayed, Nazish, additional, and Wu, Joseph C, additional
- Published
- 2022
- Full Text
- View/download PDF
40. Abstract P2109: Elaborating Safety Margins To Predict Drug Proarrhythmia Using Deep Learning And Patient-derived IPSCs.
- Author
-
Serrano, Ricardo, primary, Feyen, Dries, additional, Bruyneel, Arne A, additional, Hnatiuk Hnatiuk, Anna, additional, Vu, Michelle, additional, Amatya, Prashila, additional, Perea Gil, Isaac, additional, Prado, Maricela, additional, Seeger, Timon, additional, Wu, Joseph C, additional, Karakikes, Ioannis, additional, and Mercola, Mark, additional
- Published
- 2022
- Full Text
- View/download PDF
41. KMT2D-NOTCH Mediates Coronary Abnormalities in Hypoplastic Left Heart Syndrome
- Author
-
Yu, Zhiyun, primary, Zhou, Xin, additional, Liu, Ziyi, additional, Pastrana-Gomez, Victor, additional, Liu, Yu, additional, Guo, Minzhe, additional, Tian, Lei, additional, Nelson, Timothy J., additional, Wang, Nian, additional, Mital, Seema, additional, Chitayat, David, additional, Wu, Joseph C., additional, Rabinovitch, Marlene, additional, Wu, Sean M., additional, Snyder, Michael P., additional, Miao, Yifei, additional, and Gu, Mingxia, additional
- Published
- 2022
- Full Text
- View/download PDF
42. Cellular and Engineered Organoids for Cardiovascular Models
- Author
-
Thomas, Dilip, primary, Choi, Suji, additional, Alamana, Christina, additional, Parker, Kevin Kit, additional, and Wu, Joseph C., additional
- Published
- 2022
- Full Text
- View/download PDF
43. Cell Type-Specific Chromatin Signatures Underline Regulatory DNA Elements in Human Induced Pluripotent Stem Cells and Somatic Cells
- Author
-
Zhao, Ming-Tao, Shao, Ning-Yi, Hu, Shijun, Ma, Ning, Srinivasan, Rajini, Jahanbani, Fereshteh, Lee, Jaecheol, Zhang, Sophia L., Snyder, Michael P., and Wu, Joseph C.
- Published
- 2017
- Full Text
- View/download PDF
44. Genome Editing in Cardiovascular Biology
- Author
-
Seeger, Timon, Porteus, Matthew, and Wu, Joseph C.
- Published
- 2017
- Full Text
- View/download PDF
45. Macrophages: Potential Therapeutic Target of Myocardial Injury in COVID-19
- Author
-
Masataka Nishiga and Joseph C. Wu
- Subjects
2019-20 coronavirus outbreak ,Coronavirus disease 2019 (COVID-19) ,Physiology ,business.industry ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Macrophage ,Medicine ,Cardiology and Cardiovascular Medicine ,Induced pluripotent stem cell ,business ,Virology - Published
- 2021
46. Abstract P445: A Combinatorial Approach Comprehensively Improves The Maturation Of Human Induced Pluripotent Stem Cell Derived Atrial Cardiomyocytes
- Author
-
Seock Won Youn, Liang Hong, Joseph C. Wu, Khaled Abdelhady, Malek G. Massad, Brandon Chalazan, Arvind Sridhar, Dawood Darbar, Mark Maienschein-Cline, Mahmud Arif Pavel, Yong Duk Han, Grace E. Brown, Hanna Chen, Xinge Wang, Olivia T Ly, Jalees Rehman, and Salman R. Khetani
- Subjects
Physiology ,Biology ,Cardiology and Cardiovascular Medicine ,Induced pluripotent stem cell ,Cell biology - Abstract
Introduction: The limited success of pharmacological approaches to atrial fibrillation ( AF ) is due to limitations of in vitro and in vivo models and inaccessibility of human atrial tissue. Patient-specific induced pluripotent stem cell-derived atrial cardiomyocytes (iPSC-aCMs) are a robust platform to model the heterogeneous myocardial substrate of AF, but their immaturity limits their fidelity. Objective: We hypothesized that a combinatorial approach of biochemical (triiodothyronine [ T3 ], insulin-like growth factor-1 [ IGF-1 ], and dexamethasone; collectively TID ), bioenergetic (fatty acids [ FA ]), and electrical stimulation ( ES ) will enhance electrophysiological ( EP ), structural, and metabolic maturity of iPSC- a CMs. Methods: We assessed maturation with whole cell patch clamping, calcium transients, immunofluorescence (IF), Seahorse Analyzer, contractility assay, RT-PCR, Western Blotting, and RNA sequencing (RNAseq). Using a time series with RNAseq we identified signaling pathways and transcriptional regulation that drive EP, structural, and metabolic atrial development and compared iPSC-aCM maturity with human aCMs (haCMs) obtained from the same patient. Results: TID+FA+ES significantly improved structural organization and cell morphology ( Fig. 1a ), enhanced membrane potential stability and improved depolarization ( Fig. 1b ), improved Ca 2+ kinetics with faster and increased Ca 2+ release from sarcoplasmic reticulum ( Fig. 1c ), and increased expression of Na + , Ca 2+ , and K + channels, markers of structural maturity, FA metabolism, and oxidative phosphorylation ( Fig. 1d ). There was no difference in each parameter between TID+FA+ES iPSC-aCMs and haCMs from the same patient. Conclusion: Our optimized, combinatorial TID+FA+ES approach markedly enhanced EP, structural, and metabolic maturity of human iPSC-aCMs, which will be useful for elucidating the genetic basis of AF developing precision drug therapies.
- Published
- 2021
47. Matrigel Mattress: A Method for the Generation of Single Contracting Human-Induced Pluripotent Stem Cell–Derived Cardiomyocytes
- Author
-
Feaster, Tromondae K., Cadar, Adrian G., Wang, Lili, Williams, Charles H., Chun, Young Wook, Hempel, Jonathan E., Bloodworth, Nathaniel, Merryman, W. David, Lim, Chee Chew, Wu, Joseph C., Knollmann, Björn C., and Hong, Charles C.
- Published
- 2015
- Full Text
- View/download PDF
48. Abstract P445: A Combinatorial Approach Comprehensively Improves The Maturation Of Human Induced Pluripotent Stem Cell Derived Atrial Cardiomyocytes
- Author
-
Ly, Olivia T, primary, Brown, Grace, additional, Chen, Hanna, additional, Hong, Liang, additional, Wang, Xinge, additional, Han, Yong Duk, additional, Pavel, Mahmud Arif, additional, Sridhar, Arvind, additional, Maienschein-Cline, Mark, additional, Youn, Seock Won, additional, Chalazan, Brandon, additional, Massad, Malek G, additional, Abdelhady, khaled, additional, Wu, Joseph C, additional, Rehman, Jalees, additional, KHETANI, SALMAN, additional, and Darbar, Dawood, additional
- Published
- 2021
- Full Text
- View/download PDF
49. Tumor Repressor Circular RNA as a New Target for Preventative Gene Therapy Against Doxorubicin-Induced Cardiotoxicity
- Author
-
Lichao Liu, James W.S. Jahng, and Joseph C. Wu
- Subjects
Cardiotoxicity ,Physiology ,business.industry ,Genetic enhancement ,Repressor ,Circular RNA ,microRNA ,Cancer research ,medicine ,Doxorubicin ,Cardiology and Cardiovascular Medicine ,Induced pluripotent stem cell ,business ,medicine.drug - Published
- 2020
50. Workshop Report
- Author
-
Xi Yang, Brian R. Berridge, Joseph C. Wu, Frederick Sannajust, Hong Shi, Philip T. Sager, Mathew Brock, Norman Stockbridge, Beverly Lyn-Cook, Najah Abi-Gerges, and Li Pang
- Subjects
Cardiotoxicity ,Government ,Cardiovascular safety ,Physiology ,business.industry ,Pre-clinical development ,White paper ,Drug development ,Risk analysis (engineering) ,Cardiotoxicities ,Medicine ,Cardiology and Cardiovascular Medicine ,Risk assessment ,business - Abstract
Given that cardiovascular safety concerns remain the leading cause of drug attrition at the preclinical drug development stage, the National Center for Toxicological Research of the US Food and Drug Administration hosted a workshop to discuss current gaps and challenges in translating preclinical cardiovascular safety data to humans. This white paper summarizes the topics presented by speakers from academia, industry, and government intended to address the theme of improving cardiotoxicity assessment in drug development. The main conclusion is that to reduce cardiovascular safety liabilities of new therapeutic agents, there is an urgent need to integrate human-relevant platforms/approaches into drug development. Potential regulatory applications of human-derived cardiomyocytes and future directions in employing human-relevant platforms to fill the gaps and overcome barriers and challenges in preclinical cardiovascular safety assessment were discussed. This paper is intended to serve as an initial step in a public-private collaborative development program for human-relevant cardiotoxicity tools, particularly for cardiotoxicities characterized by contractile dysfunction or structural injury.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.