1
|
Lim AA, Pouyabahar D, Ashraf M, Huang K, Lohbihler M, Murareanu BM, Chang ML, Kwan M, Alibhai FJ, Tran T, Mazine A, Laflamme MA, Bader GD, Laksman Z, Protze S. Single-cell transcriptome analysis reveals CD34 as a marker of human sinoatrial node pacemaker cardiomyocytes. Nat Commun 2024; 15:10206. [PMID: 39604360 PMCID: PMC11603134 DOI: 10.1038/s41467-024-54337-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 11/08/2024] [Indexed: 11/29/2024] Open
Abstract
The sinoatrial node regulates the heart rate throughout life. Failure of this primary pacemaker results in life-threatening, slow heart rhythm. Despite its critical function, the cellular and molecular composition of the human sinoatrial node is not resolved. Particularly, no cell surface marker to identify and isolate sinoatrial node pacemaker cells has been reported. Here we use single-nuclei/cell RNA sequencing of fetal and human pluripotent stem cell-derived sinoatrial node cells to reveal that they consist of three subtypes of pacemaker cells: Core Pacemaker, Sinus Venosus, and Transitional Cells. Our study identifies a host of sinoatrial node pacemaker markers including MYH11, BMP4, and the cell surface antigen CD34. We demonstrate that sorting for CD34+ cells from stem cell differentiation cultures enriches for sinoatrial node cells exhibiting a functional pacemaker phenotype. This sinoatrial node pacemaker cell surface marker is highly valuable for stem cell-based disease modeling, drug discovery, cell replacement therapies, and the targeted delivery of therapeutics to sinoatrial node cells in vivo using antibody-drug conjugates.
Collapse
Affiliation(s)
- Amos A Lim
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Delaram Pouyabahar
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Mishal Ashraf
- Centre for Heart and Lung Innovation, University of British Columbia and St. Paul's Hospital, Vancouver, BC, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Kate Huang
- Centre for Heart and Lung Innovation, University of British Columbia and St. Paul's Hospital, Vancouver, BC, Canada
- Experimental Medicine Program, University of British Columbia, Vancouver, BC, Canada
| | - Michelle Lohbihler
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Brandon M Murareanu
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Matthew L Chang
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Maggie Kwan
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Faisal J Alibhai
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Thinh Tran
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amine Mazine
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Division of Cardiac Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Michael A Laflamme
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Gary D Bader
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Zachary Laksman
- Centre for Heart and Lung Innovation, University of British Columbia and St. Paul's Hospital, Vancouver, BC, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
- Experimental Medicine Program, University of British Columbia, Vancouver, BC, Canada
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Stephanie Protze
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
2
|
Gonzalez DM, Dariolli R, Moyett J, Song S, Shewale B, Bliley J, Clarke D, Ma'ayan A, Rentschler S, Feinberg A, Sobie E, Dubois NC. Transient Notch Activation Converts Pluripotent Stem Cell-Derived Cardiomyocytes Towards a Purkinje Fiber Fate. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.22.614353. [PMID: 39386729 PMCID: PMC11463678 DOI: 10.1101/2024.09.22.614353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Cardiac Purkinje fibers form the most distal part of the ventricular conduction system. They coordinate contraction and play a key role in ventricular arrhythmias. While many cardiac cell types can be generated from human pluripotent stem cells, methods to generate Purkinje fiber cells remain limited, hampering our understanding of Purkinje fiber biology and conduction system defects. To identify signaling pathways involved in Purkinje fiber formation, we analyzed single cell data from murine embryonic hearts and compared Purkinje fiber cells to trabecular cardiomyocytes. This identified several genes, processes, and signaling pathways putatively involved in cardiac conduction, including Notch signaling. We next tested whether Notch activation could convert human pluripotent stem cell-derived cardiomyocytes to Purkinje fiber cells. Following Notch activation, cardiomyocytes adopted an elongated morphology and displayed altered electrophysiological properties including increases in conduction velocity, spike slope, and action potential duration, all characteristic features of Purkinje fiber cells. RNA-sequencing demonstrated that Notch-activated cardiomyocytes undergo a sequential transcriptome shift, which included upregulation of key Purkinje fiber marker genes involved in fast conduction such as SCN5A, HCN4 and ID2, and downregulation of genes involved in contractile maturation. Correspondingly, we demonstrate that Notch-induced cardiomyocytes have decreased contractile force in bioengineered tissues compared to control cardiomyocytes. We next modified existing in silico models of human pluripotent stem cell-derived cardiomyocytes using our transcriptomic data and modeled the effect of several anti-arrhythmogenic drugs on action potential and calcium transient waveforms. Our models predicted that Purkinje fiber cells respond more strongly to dofetilide and amiodarone, while cardiomyocytes are more sensitive to treatment with nifedipine. We validated these findings in vitro, demonstrating that our new cell-specific in vitro model can be utilized to better understand human Purkinje fiber physiology and its relevance to disease.
Collapse
Affiliation(s)
- David M Gonzalez
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai, New York, NY 10029, USA
- Mindich Child Health and Development Institute, Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Mount Sinai, New York, NY 10029, USA
- Cardiovascular Research Institute, Mount Sinai, New York, NY 10029, USA
- Graduate School at the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA. Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rafael Dariolli
- Department of Pharmacology and Systems Biology, Mount Sinai Center for Bioinformatics, Department of Artificial Intelligence and Human Health, Mount Sinai, New York, NY 10029, USA
| | - Julia Moyett
- Duke University School of Medicine, Durham, NC 27710
| | - Stephanie Song
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai, New York, NY 10029, USA
- Mindich Child Health and Development Institute, Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Mount Sinai, New York, NY 10029, USA
- Cardiovascular Research Institute, Mount Sinai, New York, NY 10029, USA
| | - Bhavana Shewale
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai, New York, NY 10029, USA
- Mindich Child Health and Development Institute, Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Mount Sinai, New York, NY 10029, USA
- Cardiovascular Research Institute, Mount Sinai, New York, NY 10029, USA
- Graduate School at the Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA. Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Daniel Clarke
- Department of Pharmacology and Systems Biology, Mount Sinai Center for Bioinformatics, Department of Artificial Intelligence and Human Health, Mount Sinai, New York, NY 10029, USA
| | - Avi Ma'ayan
- Department of Pharmacology and Systems Biology, Mount Sinai Center for Bioinformatics, Department of Artificial Intelligence and Human Health, Mount Sinai, New York, NY 10029, USA
| | - Stacey Rentschler
- Washington University School of Medicine in St. Louis, Missouri MO 63110
| | | | - Eric Sobie
- Department of Pharmacology and Systems Biology, Mount Sinai Center for Bioinformatics, Department of Artificial Intelligence and Human Health, Mount Sinai, New York, NY 10029, USA
| | - Nicole C Dubois
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai, New York, NY 10029, USA
- Mindich Child Health and Development Institute, Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Mount Sinai, New York, NY 10029, USA
- Cardiovascular Research Institute, Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
3
|
Spanos M, Gokulnath P, Li G, Hutchins E, Meechoovet B, Sheng Q, Chatterjee E, Sharma R, Carnel-Amar N, Lin C, Azzam C, Ghaeli I, Amancherla KV, Victorino JF, Garcia-Mansfield K, Pfeffer R, Sahu P, Lindman BR, Elmariah S, Gamazon ER, Betti MJ, Bledsoe X, Lance ML, Absi T, Su YR, Do N, Contreras MG, Varrias D, Kladas M, Radulovic M, Tsiachris D, Spanos A, Tsioufis K, Ellinor PT, Tucker NR, Januzzi JL, Pirrotte P, Jovanovic-Talisman T, Van Keuren-Jensen K, Shah R, Das S. Cardiomyocyte-derived circulating extracellular vesicles allow a non-invasive liquid biopsy of myocardium in health and disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.19.24314009. [PMID: 39371135 PMCID: PMC11451713 DOI: 10.1101/2024.09.19.24314009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
The ability to track disease without tissue biopsy in patients is a major goal in biology and medicine. Here, we identify and characterize cardiomyocyte-derived extracellular vesicles in circulation (EVs; "cardiovesicles") through comprehensive studies of induced pluripotent stem cell-derived cardiomyocytes, genetic mouse models, and state-of-the-art mass spectrometry and low-input transcriptomics. These studies identified two markers (POPDC2, CHRNE) enriched on cardiovesicles for biotinylated antibody-based immunocapture. Captured cardiovesicles were enriched in canonical cardiomyocyte transcripts/pathways with distinct profiles based on human disease type (heart failure, myocardial infarction). In paired myocardial tissue-plasma from patients, highly expressed genes in cardiovesicles were largely cardiac-enriched (vs. "bulk" EVs, which were more organ non-specific) with high expression in myocardial tissue by single nuclear RNA-seq, largely in cardiomyocytes. These results demonstrate the first "liquid" biopsy discovery platform to interrogate cardiomyocyte states noninvasively in model systems and in human disease, allowing non-invasive characterization of cardiomyocyte biology for discovery and therapeutic applications.
Collapse
Affiliation(s)
- Michail Spanos
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Albert Einstein College of Medicine/ North Central Bronx/Jacobi Medical Center, New York City Health and Hospitals, The Bronx, NY, USA
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, Athens, Greece
| | - Priyanka Gokulnath
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Guoping Li
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Elizabeth Hutchins
- Division of Neurogenomics, The Translational Genomics Research Institute, Phoenix, AZ, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Bessie Meechoovet
- Division of Neurogenomics, The Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Quanhu Sheng
- Department of Biostatistics (Q.S.), Vanderbilt University Medical Center, Nashville, TN
| | - Emeli Chatterjee
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ritin Sharma
- Collaborative Center for Translational Mass Spectrometry, Translational Genomics Research Institute, Phoenix, AZ, USA
- Cancer & Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ
| | - Natacha Carnel-Amar
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Claire Lin
- Division of Cardiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Christopher Azzam
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ima Ghaeli
- Department of Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Kaushik V Amancherla
- Vanderbilt Translational and Clinical Research Center (R.S.), Vanderbilt University Medical Center, Nashville, TN, USA
| | - José Fabian Victorino
- Cancer & Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ
| | - Krystine Garcia-Mansfield
- Collaborative Center for Translational Mass Spectrometry, Translational Genomics Research Institute, Phoenix, AZ, USA
- Cancer & Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ
| | - Ryan Pfeffer
- Masonic Medical Research Institute, Utica, NY, USA 13501
| | - Parul Sahu
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Brian R. Lindman
- Vanderbilt Translational and Clinical Research Center (R.S.), Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sammy Elmariah
- Department of Medicine, Cardiovascular Division, University of California-San Francisco, San Francisco, CA, USA
| | - Eric R. Gamazon
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| | - Michael J. Betti
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| | - Xavier Bledsoe
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| | | | - Tarek Absi
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Yan Ru Su
- Vanderbilt Translational and Clinical Research Center (R.S.), Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ngoc Do
- Spectradyne LLC, Signal Hill, CA, USA
| | - Marta Garcia Contreras
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Dimitrios Varrias
- Albert Einstein College of Medicine/ Jacobi Medical Center, The Bronx, NY, USA
| | - Michail Kladas
- Albert Einstein College of Medicine/ North Central Bronx/Jacobi Medical Center, New York City Health and Hospitals, The Bronx, NY, USA
| | - Miroslav Radulovic
- Albert Einstein College of Medicine/ North Central Bronx/Jacobi Medical Center, New York City Health and Hospitals, The Bronx, NY, USA
| | - Dimitris Tsiachris
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, Athens, Greece
| | | | - Konstantinos Tsioufis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, Athens, Greece
| | - Patrick T. Ellinor
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - James L. Januzzi
- Division of Cardiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Baim Institute for Clinical Research, Boston, Massachusetts, USA
| | - Patrick Pirrotte
- Collaborative Center for Translational Mass Spectrometry, Translational Genomics Research Institute, Phoenix, AZ, USA
- Cancer & Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ
| | - Tijana Jovanovic-Talisman
- Department of Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | | | - Ravi Shah
- Vanderbilt Translational and Clinical Research Center (R.S.), Vanderbilt University Medical Center, Nashville, TN, USA
| | - Saumya Das
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
4
|
Hyams NA, Kerr CM, Arhontoulis DC, Ruddy JM, Mei Y. Improving human cardiac organoid design using transcriptomics. Sci Rep 2024; 14:20147. [PMID: 39209865 PMCID: PMC11362591 DOI: 10.1038/s41598-024-61554-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/07/2024] [Indexed: 09/04/2024] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide. To this end, human cardiac organoids (hCOs) have been developed for improved organotypic CVD modeling over conventional in vivo animal models. Utilizing human cells, hCOs hold great promise to bridge key gaps in CVD research pertaining to human-specific conditions. hCOs are multicellular 3D models which resemble heart structure and function. Varying hCOs fabrication techniques leads to functional and phenotypic differences. To investigate heterogeneity across hCO platforms, we performed a transcriptomic analysis utilizing bulk RNA-sequencing from four previously published unique hCO studies. We further compared selected hCOs to 2D and 3D hiPSC-derived cardiomyocytes (hiPSC-CMs), as well as fetal and adult human myocardium bulk RNA-sequencing samples. Upon investigation utilizing Principal Component Analysis, K-means clustering analysis of key genes, and further downstream analyses such as Gene Set Enrichment (GSEA), Gene Set Variation (GSVA), and GO term enrichment, we found that hCO fabrication method influences maturity and cellular heterogeneity across models. Thus, we propose that adjustment of fabrication method will result in an hCO with a defined maturity and transcriptomic profile to facilitate its specified applications, in turn maximizing its modeling potential.
Collapse
Affiliation(s)
- Nathaniel A Hyams
- Bioengineering Department, Clemson University, Clemson, SC, 29631, USA
| | - Charles M Kerr
- Molecular and Cellular Biology and Pathobiology Program, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Dimitrios C Arhontoulis
- Molecular and Cellular Biology and Pathobiology Program, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Jean Marie Ruddy
- Division of Vascular Surgery, Department of Surgery, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Ying Mei
- Bioengineering Department, Clemson University, Clemson, SC, 29631, USA.
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
5
|
Farboud SP, Fathi E, Valipour B, Farahzadi R. Toward the latest advancements in cardiac regeneration using induced pluripotent stem cells (iPSCs) technology: approaches and challenges. J Transl Med 2024; 22:783. [PMID: 39175068 PMCID: PMC11342568 DOI: 10.1186/s12967-024-05499-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 07/10/2024] [Indexed: 08/24/2024] Open
Abstract
A novel approach to treating heart failures was developed with the introduction of iPSC technology. Knowledge in regenerative medicine, developmental biology, and the identification of illnesses at the cellular level has exploded since the discovery of iPSCs. One of the most frequent causes of mortality associated with cardiovascular disease is the loss of cardiomyocytes (CMs), followed by heart failure. A possible treatment for heart failure involves restoring cardiac function and replacing damaged tissue with healthy, regenerated CMs. Significant strides in stem cell biology during the last ten years have transformed the in vitro study of human illness and enhanced our knowledge of the molecular pathways underlying human disease, regenerative medicine, and drug development. We seek to examine iPSC advancements in disease modeling, drug discovery, iPSC-Based cell treatments, and purification methods in this article.
Collapse
Affiliation(s)
- Seyedeh Parya Farboud
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Ezzatollah Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran.
| | - Behnaz Valipour
- Department of Anatomical Sciences, Sarab Faculty of Medical Sciences, Sarab, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
6
|
Bisson JA, Gordillo M, Kumar R, de Silva N, Yang E, Banks KM, Shi ZD, Lee K, Yang D, Chung WK, Huangfu D, Evans T. GATA6 regulates WNT and BMP programs to pattern precardiac mesoderm during the earliest stages of human cardiogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.09.602666. [PMID: 39026742 PMCID: PMC11257636 DOI: 10.1101/2024.07.09.602666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Haploinsufficiency for GATA6 is associated with congenital heart disease (CHD) with variable comorbidity of pancreatic or diaphragm defects, although the etiology of disease is not well understood. Here, we used cardiac directed differentiation from human embryonic stem cells (hESCs) as a platform to study GATA6 function during early cardiogenesis. GATA6 loss-of-function hESCs had a profound impairment in cardiac progenitor cell (CPC) specification and cardiomyocyte (CM) generation due to early defects during the mesendoderm and lateral mesoderm patterning stages. Profiling by RNA-seq and CUT&RUN identified genes of the WNT and BMP programs regulated by GATA6 during early mesoderm patterning. Furthermore, interactome analysis detected GATA6 binding with developmental transcription factors and chromatin remodelers suggesting cooperative regulation of cardiac lineage gene accessibility. We show that modulating WNT and BMP inputs during the first 48 hours of cardiac differentiation is sufficient to partially rescue CPC and CM defects in GATA6 heterozygous and homozygous mutant hESCs. This study provides evidence of the regulatory functions for GATA6 directing human precardiac mesoderm patterning during the earliest stages of cardiogenesis to further our understanding of haploinsufficiency causing CHD and the co-occurrence of cardiac and other organ defects caused by human GATA6 mutations.
Collapse
Affiliation(s)
- Joseph A. Bisson
- Department of Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Miriam Gordillo
- Department of Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Ritu Kumar
- Department of Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
- current address: Gladstone Institutes, San Francisco, CA, 94158, USA
| | - Neranjan de Silva
- Department of Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Ellen Yang
- Department of Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Kelly M. Banks
- Department of Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Zhong-Dong Shi
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, 10065, USA
| | - Kihyun Lee
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, 10065, USA
- current address: College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Dapeng Yang
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, 10065, USA
| | - Wendy K. Chung
- Childrens Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Danwei Huangfu
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, 10065, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
- Hartman Institute for Therapeutic Organ Regeneration, Weill Cornell Medicine, New York, NY 10065, USA
- Center for Genomic Health, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
7
|
Cai S, Dai Q. Progress in preclinical research on induced pluripotent stem cell therapy for acute myocardial infarction. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:244-253. [PMID: 38594961 PMCID: PMC11057988 DOI: 10.3724/zdxbyxb-2023-0402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 03/09/2024] [Indexed: 04/11/2024]
Abstract
Induced pluripotent stem cells (iPSCs) are obtained by introducing exogenous genes or adding chemicals to the culture medium to induce somatic cell differentiation. Similarly to embryonic stem cells, iPSCs have the ability to differentiate into all three embryonic cell lines. iPSCs can differentiate into cardiac muscle cells through two-dimensional differentiation methods such as monolayer cell culture and co-culture, or through embryoid body and scaffold-based three-dimensional differentiation methods. In addition, the process of iPSCs differentiation into cardiac muscle cells also requires activation or inhibition of specific signaling pathways,such as Wnt, BMP, Notch signaling pathways to mimic the development of the heart in vivo. In recent years, suspension culturing in bioreactors has been shown to produce large number of iPSCs derived cardiac muscle cells (iPSC-CMs). Before transplantation, it is necessary to purify iPSC-CMs through metabolic regulation or cell sorting to eliminate undifferentiated iPSCs, which may lead to teratoma formation. The transplantation methods for iPSC-CMs are mainly injection of cell suspension and transplantation of cell patches into the infarcted myocardium. Animal studies have shown that transplantation of iPSC-CMs into the infarcted myocardium can improve cardiac function. This article reviews the progress in preclinical studies on iPSC-CMs therapy for acute myocardial infarction and discusses the limitations and challenges of its clinical application to provide references for further clinical research and application.
Collapse
Affiliation(s)
- Songyan Cai
- Department of Cardiology, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China.
| | - Qingyuan Dai
- Department of Cardiology, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China.
| |
Collapse
|
8
|
Manda V, Pavelka J, Lau E. Proteomics applications in next generation induced pluripotent stem cell models. Expert Rev Proteomics 2024; 21:217-228. [PMID: 38511670 PMCID: PMC11065590 DOI: 10.1080/14789450.2024.2334033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 03/08/2024] [Indexed: 03/22/2024]
Abstract
INTRODUCTION Induced pluripotent stem (iPS) cell technology has transformed biomedical research. New opportunities now exist to create new organoids, microtissues, and body-on-a-chip systems for basic biology investigations and clinical translations. AREAS COVERED We discuss the utility of proteomics for attaining an unbiased view into protein expression changes during iPS cell differentiation, cell maturation, and tissue generation. The ability to discover cell-type specific protein markers during the differentiation and maturation of iPS-derived cells has led to new strategies to improve cell production yield and fidelity. In parallel, proteomic characterization of iPS-derived organoids is helping to realize the goal of bridging in vitro and in vivo systems. EXPERT OPINIONS We discuss some current challenges of proteomics in iPS cell research and future directions, including the integration of proteomic and transcriptomic data for systems-level analysis.
Collapse
Affiliation(s)
- Vyshnavi Manda
- Department of Medicine, Division of Cardiology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Consortium for Fibrosis Research and Translation, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Jay Pavelka
- Department of Medicine, Division of Cardiology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Consortium for Fibrosis Research and Translation, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Edward Lau
- Department of Medicine, Division of Cardiology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Consortium for Fibrosis Research and Translation, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
9
|
Nakanishi-Koakutsu M, Miki K, Naka Y, Sasaki M, Wakimizu T, Napier SC, Okubo C, Narita M, Nishikawa M, Hata R, Chonabayashi K, Hotta A, Imahashi K, Nishimoto T, Yoshida Y. CD151 expression marks atrial- and ventricular- differentiation from human induced pluripotent stem cells. Commun Biol 2024; 7:231. [PMID: 38418926 PMCID: PMC10901864 DOI: 10.1038/s42003-024-05809-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 01/11/2024] [Indexed: 03/02/2024] Open
Abstract
Current differentiation protocols for human induced pluripotent stem cells (hiPSCs) produce heterogeneous cardiomyocytes (CMs). Although chamber-specific CM selection using cell surface antigens enhances biomedical applications, a cell surface marker that accurately distinguishes between hiPSC-derived atrial CMs (ACMs) and ventricular CMs (VCMs) has not yet been identified. We have developed an approach for obtaining functional hiPSC-ACMs and -VCMs based on CD151 expression. For ACM differentiation, we found that ACMs are enriched in the CD151low population and that CD151 expression is correlated with the expression of Notch4 and its ligands. Furthermore, Notch signaling inhibition followed by selecting the CD151low population during atrial differentiation leads to the highly efficient generation of ACMs as evidenced by gene expression and electrophysiology. In contrast, for VCM differentiation, VCMs exhibiting a ventricular-related gene signature and uniform action potentials are enriched in the CD151high population. Our findings enable the production of high-quality ACMs and VCMs appropriate for hiPSC-derived chamber-specific disease models and other applications.
Collapse
Affiliation(s)
- Misato Nakanishi-Koakutsu
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
- Takeda-CiRA Joint program (T-CiRA), Fujisawa, 251-8555, Japan
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Surgery, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Kenji Miki
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan.
- Center for Organ Engineering, Department of Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA.
- Department of Surgery, Harvard Medical School, Boston, MA, 02114, USA.
- Premium Research Institute for Human Metaverse Medicine, Osaka University, Suita, 565-0871, Japan.
| | - Yuki Naka
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
- Takeda-CiRA Joint program (T-CiRA), Fujisawa, 251-8555, Japan
| | - Masako Sasaki
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
- Takeda-CiRA Joint program (T-CiRA), Fujisawa, 251-8555, Japan
| | - Takayuki Wakimizu
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
- Takeda-CiRA Joint program (T-CiRA), Fujisawa, 251-8555, Japan
| | - Stephanie C Napier
- Takeda-CiRA Joint program (T-CiRA), Fujisawa, 251-8555, Japan
- Global Advanced Platform, Takeda Pharmaceutical Company Limited, Fujisawa, 251-8555, Japan
| | - Chikako Okubo
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Megumi Narita
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Misato Nishikawa
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Reo Hata
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Kazuhisa Chonabayashi
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Akitsu Hotta
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
- Takeda-CiRA Joint program (T-CiRA), Fujisawa, 251-8555, Japan
| | - Kenichi Imahashi
- Takeda-CiRA Joint program (T-CiRA), Fujisawa, 251-8555, Japan
- Global Advanced Platform, Takeda Pharmaceutical Company Limited, Fujisawa, 251-8555, Japan
| | - Tomoyuki Nishimoto
- Takeda-CiRA Joint program (T-CiRA), Fujisawa, 251-8555, Japan
- Orizuru Therapeutics Incorporated, Fujisawa, 251-8555, Japan
| | - Yoshinori Yoshida
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan.
- Takeda-CiRA Joint program (T-CiRA), Fujisawa, 251-8555, Japan.
| |
Collapse
|
10
|
Hasegawa M, Miki K, Kawamura T, Takei Sasozaki I, Higashiyama Y, Tsuchida M, Kashino K, Taira M, Ito E, Takeda M, Ishida H, Higo S, Sakata Y, Miyagawa S. Gene correction and overexpression of TNNI3 improve impaired relaxation in engineered heart tissue model of pediatric restrictive cardiomyopathy. Dev Growth Differ 2024; 66:119-132. [PMID: 38193576 PMCID: PMC11457505 DOI: 10.1111/dgd.12909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 12/20/2023] [Accepted: 12/24/2023] [Indexed: 01/10/2024]
Abstract
Research on cardiomyopathy models using engineered heart tissue (EHT) created from disease-specific induced pluripotent stem cells (iPSCs) is advancing rapidly. However, the study of restrictive cardiomyopathy (RCM), a rare and intractable cardiomyopathy, remains at the experimental stage because there is currently no established method to replicate the hallmark phenotype of RCM, particularly diastolic dysfunction, in vitro. In this study, we generated iPSCs from a patient with early childhood-onset RCM harboring the TNNI3 R170W mutation (R170W-iPSCs). The properties of R170W-iPSC-derived cardiomyocytes (CMs) and EHTs were evaluated and compared with an isogenic iPSC line in which the mutation was corrected. Our results indicated altered calcium kinetics in R170W-iPSC-CMs, including prolonged tau, and an increased ratio of relaxation force to contractile force in R170W-EHTs. These properties were reversed in the isogenic line, suggesting that our model recapitulates impaired relaxation of RCM, i.e., diastolic dysfunction in clinical practice. Furthermore, overexpression of wild-type TNNI3 in R170W-iPSC-CMs and -EHTs effectively rescued impaired relaxation. These results highlight the potential efficacy of EHT, a modality that can accurately recapitulate diastolic dysfunction in vitro, to elucidate the pathophysiology of RCM, as well as the possible benefits of gene therapies for patients with RCM.
Collapse
Affiliation(s)
- Moyu Hasegawa
- Department of Cardiovascular SurgeryOsaka University Graduate School of MedicineOsakaJapan
| | - Kenji Miki
- Premium Research Institute for Human Metaverse MedicineOsaka UniversityOsakaJapan
| | - Takuji Kawamura
- Department of Cardiovascular SurgeryOsaka University Graduate School of MedicineOsakaJapan
| | - Ikue Takei Sasozaki
- Department of Cardiovascular SurgeryOsaka University Graduate School of MedicineOsakaJapan
| | - Yuki Higashiyama
- Department of Cardiovascular SurgeryOsaka University Graduate School of MedicineOsakaJapan
| | - Masaru Tsuchida
- NTT Communication Science LaboratoriesMedia Information Research DepartmentKanagawaJapan
| | - Kunio Kashino
- Premium Research Institute for Human Metaverse MedicineOsaka UniversityOsakaJapan
- NTT Communication Science LaboratoriesMedia Information Research DepartmentKanagawaJapan
| | - Masaki Taira
- Department of Cardiovascular SurgeryOsaka University Graduate School of MedicineOsakaJapan
| | - Emiko Ito
- Department of Cardiovascular SurgeryOsaka University Graduate School of MedicineOsakaJapan
| | - Maki Takeda
- Department of Cardiovascular SurgeryOsaka University Graduate School of MedicineOsakaJapan
| | - Hidekazu Ishida
- Department of PediatricsOsaka University Graduate School of MedicineOsakaJapan
| | - Shuichiro Higo
- Department of Medical Therapeutics for Heart FailureOsaka University Graduate School of MedicineOsakaJapan
| | - Yasushi Sakata
- Department of Cardiovascular MedicineOsaka University Graduate School of MedicineOsakaJapan
| | - Shigeru Miyagawa
- Department of Cardiovascular SurgeryOsaka University Graduate School of MedicineOsakaJapan
- Premium Research Institute for Human Metaverse MedicineOsaka UniversityOsakaJapan
| |
Collapse
|
11
|
Wolnik J, Adamska P, Oleksy A, Dulak J, Biniecka M. Enriching Cardiomyocytes Derived from hiPSCs by Magnetic-Activated Cell Sorting (MACS). Methods Mol Biol 2024; 2835:83-98. [PMID: 39105908 DOI: 10.1007/978-1-0716-3995-5_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
Cardiomyocytes (CMs) derived from human-induced pluripotent stem cells (hiPSCs) are considered a promising platform for multiple applications, including disease modeling, regenerative medicine, screening of drug toxicity and investigation of cardiomyogenesis. Despite remarkable improvement in methodology enabling differentiation of hiPSCs into CMs, applied protocols generate heterogeneous cell populations composed of CMs along with differentiated non-cardiac cell-types and undifferentiated hiPSCs. Here we describea procedure of automated Magnetic-Activated Cell Sorting (autoMACS) for the purification of hiPSCs-derived CMs under sterile culture conditions. We illustrate that this approach led to a robust depletion of non-cardiac cells and enrichment of CMs, a result particularly crucial for hiPSC lines with poor cardiac differentiation efficiencies.
Collapse
Affiliation(s)
- Jan Wolnik
- Departament of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
- Jagiellonian University, Doctoral School of Exact and Natural Sciences, Krakow, Poland
| | - Patrycja Adamska
- Departament of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
- Jagiellonian University, Doctoral School of Exact and Natural Sciences, Krakow, Poland
| | - Aleksandra Oleksy
- Departament of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Józef Dulak
- Departament of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Monika Biniecka
- Departament of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.
- Silesian Park of Medical Technology, Kardio-Med Silesia, Zabrze, Poland.
| |
Collapse
|
12
|
Liu S, Fang C, Zhong C, Li J, Xiao Q. Recent advances in pluripotent stem cell-derived cardiac organoids and heart-on-chip applications for studying anti-cancer drug-induced cardiotoxicity. Cell Biol Toxicol 2023; 39:2527-2549. [PMID: 37889357 DOI: 10.1007/s10565-023-09835-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/11/2023] [Indexed: 10/28/2023]
Abstract
Cardiovascular disease (CVD) caused by anti-cancer drug-induced cardiotoxicity is now the second leading cause of mortality among cancer survivors. It is necessary to establish efficient in vitro models for early predicting the potential cardiotoxicity of anti-cancer drugs, as well as for screening drugs that would alleviate cardiotoxicity during and post treatment. Human induced pluripotent stem cells (hiPSCs) have opened up new avenues in cardio-oncology. With the breakthrough of tissue engineering technology, a variety of hiPSC-derived cardiac microtissues or organoids have been recently reported, which have shown enormous potential in studying cardiotoxicity. Moreover, using hiPSC-derived heart-on-chip for studying cardiotoxicity has provided novel insights into the underlying mechanisms. Herein, we summarize different types of anti-cancer drug-induced cardiotoxicities and present an extensive overview on the applications of hiPSC-derived cardiac microtissues, cardiac organoids, and heart-on-chips in cardiotoxicity. Finally, we highlight clinical and translational challenges around hiPSC-derived cardiac microtissues/organoids/heart-on chips and their applications in anti-cancer drug-induced cardiotoxicity. • Anti-cancer drug-induced cardiotoxicities represent pressing challenges for cancer treatments, and cardiovascular disease is the second leading cause of mortality among cancer survivors. • Newly reported in vitro models such as hiPSC-derived cardiac microtissues/organoids/chips show enormous potential for studying cardio-oncology. • Emerging evidence supports that hiPSC-derived cardiac organoids and heart-on-chip are promising in vitro platforms for predicting and minimizing anti-cancer drug-induced cardiotoxicity.
Collapse
Affiliation(s)
- Silin Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Heart Centre, Charterhouse Square, London, EC1M 6BQ, UK
- Guangdong Provincial Clinical Research Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Chongkai Fang
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Heart Centre, Charterhouse Square, London, EC1M 6BQ, UK
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Chong Zhong
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Guangdong Provincial Clinical Research Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jing Li
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- Guangdong Provincial Clinical Research Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- Faculty of Biological Sciences, University of Leeds, Leeds, UK.
| | - Qingzhong Xiao
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Heart Centre, Charterhouse Square, London, EC1M 6BQ, UK.
- Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences, Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
13
|
Feng Y, Huang C, Wang Y, Chen J. SIRPα: A key player in innate immunity. Eur J Immunol 2023; 53:e2350375. [PMID: 37672390 DOI: 10.1002/eji.202350375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 07/15/2023] [Accepted: 09/05/2023] [Indexed: 09/08/2023]
Abstract
Signal regulatory protein alpha (SIRPα) is a crucial inhibitory regulator expressed on the surface of myeloid cells, including macrophages, dendritic cells, monocytes, neutrophils, and microglia. SIRPα plays an indispensable role in innate immune and adoptive immune responses in cancer immunology, tissue homeostasis, and other physiological or phycological conditions. This review provides an overview of the research history, ligands, signal transduction pathways, and functional mechanisms associated with SIRPα. Additionally, we summarize the therapeutic implications of targeting SIRPα as a promising novel strategy in immuno-oncology.
Collapse
Affiliation(s)
- Yongyi Feng
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chunliu Huang
- Molecular Imaging Center, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Yingzhao Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jun Chen
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, China
- Jinfeng Laboratory, Chongqing, China
| |
Collapse
|
14
|
Oh MS, Lee SG, Lee GH, Kim CY, Song JH, Yu BY, Chung HM. Verification of Therapeutic Effect through Accelerator Mass Spectrometry-Based Single Cell Level Quantification of hESC-Endothelial Cells Distributed into an Ischemic Model. Adv Healthc Mater 2023; 12:e2300476. [PMID: 37068221 DOI: 10.1002/adhm.202300476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/01/2023] [Indexed: 04/19/2023]
Abstract
As the potential of pluripotent stem cell-derived differentiated cells has been demonstrated in regenerative medicine, differentiated vascular endothelial cells (ECs) are emerging as a therapeutic agent for the cardiovascular system. To verify the therapeutic efficacy of differentiated ECs in an ischemic model, human embryonic stem cells (hESCs) are induced as EC lineage and produce high-purity ECs through fluorescence-activated cell sorting (FACS). When hESC-ECs are transplanted into a hindlimb ischemic model, it is confirmed that blood flow and muscle regeneration are further improved by creating new blood vessels together with autologous ECs than the primary cell as cord blood endothelial progenitor cells (CB-EPCs). In addition, previously reported studies show the detection of transplanted cells engrafted in blood vessels through various tracking methods, but fail to provide accurate quantitative values over time. In this study, it is demonstrated that hESC-ECs are engrafted approximately sevenfold more than CB-EPCs by using an accelerator mass spectrometry (AMS)-based cell tracking technology that can perform quantification at the single cell level. An accurate quantification index is suggested. It has never been reported in in vivo kinetics of hESC-ECs that can act as therapeutic agents.
Collapse
Affiliation(s)
- Min-Seok Oh
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, Seoul, 05029, Republic of Korea
- Advanced Analysis and Data Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Seul-Gi Lee
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, Seoul, 05029, Republic of Korea
| | - Gwan-Ho Lee
- Advanced Analysis and Data Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - C-Yoon Kim
- College of Veterinary Medicine, Konkuk University, Gwangjin-Gu, Seoul, 05029, Republic of Korea
| | - Jong Han Song
- Advanced Analysis and Data Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Byung-Yong Yu
- Advanced Analysis and Data Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Hyung Min Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-Ro, Gwangjin-Gu, Seoul, 05029, Republic of Korea
- Mirae Cell Bio Co. Ltd, Seoul, 04795, Republic of Korea
| |
Collapse
|
15
|
Li CY, Liang Z, Liu L, Kuang Y. Intracellular Molecules Induced Extracellular Peptide Self-Assembly for Efficient and Effective In Situ Cell Purification. Angew Chem Int Ed Engl 2023; 62:e202306533. [PMID: 37483172 DOI: 10.1002/anie.202306533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/18/2023] [Accepted: 07/21/2023] [Indexed: 07/25/2023]
Abstract
Synthetic messenger RNA (mRNA) switches are powerful tools for in situ cell purification, especially for cells derived from stem cells. However, the retention effectiveness of the target cells is limited by the leaky expression of toxic protein. The elimination efficiency of non-target cells is also constrained due to the lack of signal amplification. In this study, we designed a novel approach that uses synthetic mRNA switch to convey intracellular marker molecule information into spatially controlled extracellular toxic assembly formation. The approach bypasses the use of toxic protein to ensure high target cell recovery effectiveness. Meanwhile, the marker molecule information is amplified at multiple levels to ensure high non-target cell elimination effectiveness. Our approach can be tailored to meet various in situ cell purification needs, promising high-quality in situ cell purification for a wide range of biomedical applications.
Collapse
Affiliation(s)
- Cheuk Yin Li
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, Hong Kong
| | - Zhenghua Liang
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, Hong Kong
| | - Lejian Liu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, Hong Kong
| | - Yi Kuang
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, Hong Kong
| |
Collapse
|
16
|
Rebs S, Streckfuss-Bömeke K. How can we use stem cell-derived cardiomyocytes to understand the involvement of energetic metabolism in alterations of cardiac function? FRONTIERS IN MOLECULAR MEDICINE 2023; 3:1222986. [PMID: 39086669 PMCID: PMC11285589 DOI: 10.3389/fmmed.2023.1222986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/15/2023] [Indexed: 08/02/2024]
Abstract
Mutations in the mitochondrial-DNA or mitochondria related nuclear-encoded-DNA lead to various multisystemic disorders collectively termed mitochondrial diseases. One in three cases of mitochondrial disease affects the heart muscle, which is called mitochondrial cardiomyopathy (MCM) and is associated with hypertrophic, dilated, and noncompact cardiomyopathy. The heart is an organ with high energy demand, and mitochondria occupy 30%-40% of its cardiomyocyte-cell volume. Mitochondrial dysfunction leads to energy depletion and has detrimental effects on cardiac performance. However, disease development and progression in the context of mitochondrial and nuclear DNA mutations, remains incompletely understood. The system of induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CM) is an excellent platform to study MCM since the unique genetic identity to their donors enables a robust recapitulation of the predicted phenotypes in a dish on a patient-specific level. Here, we focus on recent insights into MCM studied by patient-specific iPSC-CM and further discuss research gaps and advances in metabolic maturation of iPSC-CM, which is crucial for the study of mitochondrial dysfunction and to develop novel therapeutic strategies.
Collapse
Affiliation(s)
- Sabine Rebs
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
- Clinic for Cardiology and Pneumology, University Medicine Göttingen and DZHK (German Centre for Cardiovascular Research), Göttingen, Germany
| | - Katrin Streckfuss-Bömeke
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
- Clinic for Cardiology and Pneumology, University Medicine Göttingen and DZHK (German Centre for Cardiovascular Research), Göttingen, Germany
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany
| |
Collapse
|
17
|
Kizub IV. Induced pluripotent stem cells for cardiovascular therapeutics: Progress and perspectives. REGULATORY MECHANISMS IN BIOSYSTEMS 2023; 14:451-468. [DOI: 10.15421/10.15421/022366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
The discovery of methods for reprogramming adult somatic cells into induced pluripotent stem cells (iPSCs) opens up prospects of developing personalized cell-based therapy options for a variety of human diseases as well as disease modeling and new drug discovery. Like embryonic stem cells, iPSCs can give rise to various cell types of the human body and are amenable to genetic correction. This allows usage of iPSCs in the development of modern therapies for many virtually incurable human diseases. The review summarizes progress in iPSC research in the context of application in the cardiovascular field including modeling cardiovascular disease, drug study, tissue engineering, and perspectives for personalized cardiovascular medicine.
Collapse
|
18
|
Li X, La Salvia S, Liang Y, Adamiak M, Kohlbrenner E, Jeong D, Chepurko E, Ceholski D, Lopez-Gordo E, Yoon S, Mathiyalagan P, Agarwal N, Jha D, Lodha S, Daaboul G, Phan A, Raisinghani N, Zhang S, Zangi L, Gonzalez-Kozlova E, Dubois N, Dogra N, Hajjar RJ, Sahoo S. Extracellular Vesicle-Encapsulated Adeno-Associated Viruses for Therapeutic Gene Delivery to the Heart. Circulation 2023; 148:405-425. [PMID: 37409482 DOI: 10.1161/circulationaha.122.063759] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 05/16/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND Adeno-associated virus (AAV) has emerged as one of the best tools for cardiac gene delivery due to its cardiotropism, long-term expression, and safety. However, a significant challenge to its successful clinical use is preexisting neutralizing antibodies (NAbs), which bind to free AAVs, prevent efficient gene transduction, and reduce or negate therapeutic effects. Here we describe extracellular vesicle-encapsulated AAVs (EV-AAVs), secreted naturally by AAV-producing cells, as a superior cardiac gene delivery vector that delivers more genes and offers higher NAb resistance. METHODS We developed a 2-step density-gradient ultracentrifugation method to isolate highly purified EV-AAVs. We compared the gene delivery and therapeutic efficacy of EV-AAVs with an equal titer of free AAVs in the presence of NAbs, both in vitro and in vivo. In addition, we investigated the mechanism of EV-AAV uptake in human left ventricular and human induced pluripotent stem cell-derived cardiomyocytes in vitro and mouse models in vivo using a combination of biochemical techniques, flow cytometry, and immunofluorescence imaging. RESULTS Using cardiotropic AAV serotypes 6 and 9 and several reporter constructs, we demonstrated that EV-AAVs deliver significantly higher quantities of genes than AAVs in the presence of NAbs, both to human left ventricular and human induced pluripotent stem cell-derived cardiomyocytes in vitro and to mouse hearts in vivo. Intramyocardial delivery of EV-AAV9-sarcoplasmic reticulum calcium ATPase 2a to infarcted hearts in preimmunized mice significantly improved ejection fraction and fractional shortening compared with AAV9-sarcoplasmic reticulum calcium ATPase 2a delivery. These data validated NAb evasion by and therapeutic efficacy of EV-AAV9 vectors. Trafficking studies using human induced pluripotent stem cell-derived cells in vitro and mouse hearts in vivo showed significantly higher expression of EV-AAV6/9-delivered genes in cardiomyocytes compared with noncardiomyocytes, even with comparable cellular uptake. Using cellular subfraction analyses and pH-sensitive dyes, we discovered that EV-AAVs were internalized into acidic endosomal compartments of cardiomyocytes for releasing and acidifying AAVs for their nuclear uptake. CONCLUSIONS Together, using 5 different in vitro and in vivo model systems, we demonstrate significantly higher potency and therapeutic efficacy of EV-AAV vectors compared with free AAVs in the presence of NAbs. These results establish the potential of EV-AAV vectors as a gene delivery tool to treat heart failure.
Collapse
Affiliation(s)
- Xisheng Li
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Sabrina La Salvia
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Yaxuan Liang
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, China (Y.L.)
| | - Marta Adamiak
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Erik Kohlbrenner
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
- Spark Therapeutics, Philadelphia, PA (E.K.)
| | - Dongtak Jeong
- Department of Molecular and Life Science, College of Science and Convergence Technology, Hanyang University-ERICA, Ansan, South Korea (D.J.)
| | - Elena Chepurko
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Delaine Ceholski
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Estrella Lopez-Gordo
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Seonghun Yoon
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Neha Agarwal
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Divya Jha
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Shweta Lodha
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Anh Phan
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Nikhil Raisinghani
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Shihong Zhang
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Lior Zangi
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Edgar Gonzalez-Kozlova
- Department of Oncological Sciences (E.G.-K.), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Nicole Dubois
- Department of Cell, Developmental and Regenerative Biology (N. Dubois), Icahn School of Medicine at Mount Sinai, New York, NY
- Mindich Child Health and Development Institute (N. Dubois), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Navneet Dogra
- Department of Pathology and Laboratory Medicine (N. Dogra), Icahn School of Medicine at Mount Sinai, New York, NY
- Icahn Genomics Institute (N.Dogra), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Roger J Hajjar
- Gene and Cell Therapy Institute, Massachusetts General Brigham, Boston (R.J.H.)
| | - Susmita Sahoo
- Cardiovascular Research Institute (X.L., S.L.S., M.A., E.C., D.C., E.L.-G., S.Y., N.A., D.J., S.L., A.P., N.R., S.Z., L.Z., S.S.), Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
19
|
Lam YY, Chan CH, Geng L, Wong N, Keung W, Cheung YF. APLNR marks a cardiac progenitor derived with human induced pluripotent stem cells. Heliyon 2023; 9:e18243. [PMID: 37539315 PMCID: PMC10395470 DOI: 10.1016/j.heliyon.2023.e18243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 07/06/2023] [Accepted: 07/12/2023] [Indexed: 08/05/2023] Open
Abstract
Cardiomyocytes can be readily derived from human induced pluripotent stem cell (hiPSC) lines, yet its efficacy varies across different batches of the same and different hiPSC lines. To unravel the inconsistencies of in vitro cardiac differentiation, we utilized single cell transcriptomics on hiPSCs undergoing cardiac differentiation and identified cardiac and extra-cardiac lineages throughout differentiation. We further identified APLNR as a surface marker for in vitro cardiac progenitors and immunomagnetically isolated them. Differentiation of isolated in vitro APLNR+ cardiac progenitors derived from multiple hiPSC lines resulted in predominantly cardiomyocytes accompanied with cardiac mesenchyme. Transcriptomic analysis of differentiating in vitro APLNR+ cardiac progenitors revealed transient expression of cardiac progenitor markers before further commitment into cardiomyocyte and cardiac mesenchyme. Analysis of in vivo human and mouse embryo single cell transcriptomic datasets have identified APLNR expression in early cardiac progenitors of multiple lineages. This platform enables generation of in vitro cardiac progenitors from multiple hiPSC lines without genetic manipulation, which has potential applications in studying cardiac development, disease modelling and cardiac regeneration.
Collapse
Affiliation(s)
- Yin-Yu Lam
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, China
| | - Chun-Ho Chan
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, China
| | - Lin Geng
- – Dr. Li Dak-Sum Research Centre, HKU-KI Collaboration in Regenerative Medicine, The University of Hong Kong, China
| | - Nicodemus Wong
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, China
| | - Wendy Keung
- – Dr. Li Dak-Sum Research Centre, HKU-KI Collaboration in Regenerative Medicine, The University of Hong Kong, China
| | - Yiu-Fai Cheung
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, China
- – Dr. Li Dak-Sum Research Centre, HKU-KI Collaboration in Regenerative Medicine, The University of Hong Kong, China
| |
Collapse
|
20
|
Chepeleva EV, Pavlova SV, Bgatova NP, Volkov AM, Kazanskaya GM, Sergeevichev DS. Functional Activity of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes on a Mouse Renal Subcapsular Xenograft Model. Int J Mol Sci 2023; 24:9792. [PMID: 37372940 DOI: 10.3390/ijms24129792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/02/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
In the treatment of coronary heart disease, the most promising approach for replacing lost contractile elements involves obtaining cardiomyocytes through cardiac differentiation of pluripotent cells. The objective of this study is to develop a technology for creating a functional layer of cardiomyocytes derived from iPSCs, capable of generating rhythmic activity and synchronous contractions. To expedite the maturation of cardiomyocytes, a renal subcapsular transplantation model was employed in SCID mice. Following explantation, the formation of the cardiomyocyte contractile apparatus was assessed using fluorescence and electron microscopy, while the cytoplasmic oscillation of calcium ions was evaluated through visualization using the fluorescent calcium binding dye Fluo-8. The results demonstrate that transplanted human iPSC-derived cardiomyocyte cell layers, placed under the fibrous capsules of SCID mouse kidneys (for up to 6 weeks), initiate the development of an organized contractile apparatus and retain functional activity along with the ability to generate calcium ion oscillations even after removal from the body.
Collapse
Affiliation(s)
- Elena V Chepeleva
- Federal State Budgetary Institution National Medical Research Center Named after Academician E.N. Meshalkin of the Ministry of Health of the Russian Federation, 15, Rechkunovskaya Str., 630055 Novosibirsk, Russia
- Research Institute of Clinical and Experimental Lymphology-Branch of the Institute of Cytology and Genetics Siberian Branch of Russian Academy of Sciences, 2, Timakova Str., 630060 Novosibirsk, Russia
| | - Sophia V Pavlova
- Federal State Budgetary Institution National Medical Research Center Named after Academician E.N. Meshalkin of the Ministry of Health of the Russian Federation, 15, Rechkunovskaya Str., 630055 Novosibirsk, Russia
- Federal Research Center Institute of Cytology and Genetics Siberian Branch of Russian Academy of Sciences, 10, Ac. Lavrentiev Ave., 630090 Novosibirsk, Russia
| | - Nataliya P Bgatova
- Research Institute of Clinical and Experimental Lymphology-Branch of the Institute of Cytology and Genetics Siberian Branch of Russian Academy of Sciences, 2, Timakova Str., 630060 Novosibirsk, Russia
| | - Alexander M Volkov
- Federal State Budgetary Institution National Medical Research Center Named after Academician E.N. Meshalkin of the Ministry of Health of the Russian Federation, 15, Rechkunovskaya Str., 630055 Novosibirsk, Russia
| | - Galina M Kazanskaya
- Institute of Molecular Biology and Biophysics-Subdivision of FRC FTM, 2/12, Timakova Str., 630060 Novosibirsk, Russia
| | - David S Sergeevichev
- Federal State Budgetary Institution National Medical Research Center Named after Academician E.N. Meshalkin of the Ministry of Health of the Russian Federation, 15, Rechkunovskaya Str., 630055 Novosibirsk, Russia
| |
Collapse
|
21
|
Kishino Y, Tohyama S, Morita Y, Soma Y, Tani H, Okada M, Kanazawa H, Fukuda K. Cardiac Regenerative Therapy Using Human Pluripotent Stem Cells for Heart Failure: A State-of-the-Art Review. J Card Fail 2023; 29:503-513. [PMID: 37059512 DOI: 10.1016/j.cardfail.2022.10.433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/07/2022] [Accepted: 10/09/2022] [Indexed: 04/16/2023]
Abstract
Heart transplantation (HT) is the only definitive treatment available for patients with end-stage heart failure who are refractory to medical and device therapies. However, HT as a therapeutic option, is limited by a significant shortage of donors. To overcome this shortage, regenerative medicine using human pluripotent stem cells (hPSCs), such as human embryonic stem cells and human-induced pluripotent stem cells (hiPSCs), has been considered an alternative to HT. Several issues, including the methods of large-scale culture and production of hPSCs and cardiomyocytes, the prevention of tumorigenesis secondary to contamination of undifferentiated stem cells and non-cardiomyocytes, and the establishment of an effective transplantation strategy in large-animal models, need to be addressed to fulfill this unmet need. Although post-transplantation arrhythmia and immune rejection remain problems, the ongoing rapid technological advances in hPSC research have been directed toward the clinical application of this technology. Cell therapy using hPSC-derived cardiomyocytes is expected to serve as an integral component of realistic medicine in the near future and is being potentially viewed as a treatment that would revolutionize the management of patients with severe heart failure.
Collapse
Affiliation(s)
- Yoshikazu Kishino
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Shugo Tohyama
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan.
| | - Yuika Morita
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Yusuke Soma
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Hidenori Tani
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Marina Okada
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Hideaki Kanazawa
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
22
|
Wang BZ, Nash TR, Zhang X, Rao J, Abriola L, Kim Y, Zakharov S, Kim M, Luo LJ, Morsink M, Liu B, Lock RI, Fleischer S, Tamargo MA, Bohnen M, Welch CL, Chung WK, Marx SO, Surovtseva YV, Vunjak-Novakovic G, Fine BM. Engineered cardiac tissue model of restrictive cardiomyopathy for drug discovery. Cell Rep Med 2023; 4:100976. [PMID: 36921598 PMCID: PMC10040415 DOI: 10.1016/j.xcrm.2023.100976] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 12/19/2022] [Accepted: 02/21/2023] [Indexed: 03/16/2023]
Abstract
Restrictive cardiomyopathy (RCM) is defined as increased myocardial stiffness and impaired diastolic relaxation leading to elevated ventricular filling pressures. Human variants in filamin C (FLNC) are linked to a variety of cardiomyopathies, and in this study, we investigate an in-frame deletion (c.7416_7418delGAA, p.Glu2472_Asn2473delinAsp) in a patient with RCM. Induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) with this variant display impaired relaxation and reduced calcium kinetics in 2D culture when compared with a CRISPR-Cas9-corrected isogenic control line. Similarly, mutant engineered cardiac tissues (ECTs) demonstrate increased passive tension and impaired relaxation velocity compared with isogenic controls. High-throughput small-molecule screening identifies phosphodiesterase 3 (PDE3) inhibition by trequinsin as a potential therapy to improve cardiomyocyte relaxation in this genotype. Together, these data demonstrate an engineered cardiac tissue model of RCM and establish the translational potential of this precision medicine approach to identify therapeutics targeting myocardial relaxation.
Collapse
Affiliation(s)
- Bryan Z Wang
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Trevor R Nash
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Xiaokan Zhang
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Jenny Rao
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Laura Abriola
- Yale Center for Molecular Discovery, Yale University, New Haven, CT 06520, USA
| | - Youngbin Kim
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Sergey Zakharov
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Michael Kim
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Lori J Luo
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Margaretha Morsink
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Bohao Liu
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Roberta I Lock
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Sharon Fleischer
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Manuel A Tamargo
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Michael Bohnen
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Carrie L Welch
- Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | - Wendy K Chung
- Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | - Steven O Marx
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Yulia V Surovtseva
- Yale Center for Molecular Discovery, Yale University, New Haven, CT 06520, USA
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA; Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY 10032, USA; College of Dental Medicine, Columbia University, New York, NY 10032, USA
| | - Barry M Fine
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
23
|
Kawaguchi N, Nakanishi T. Animal Disease Models and Patient-iPS-Cell-Derived In Vitro Disease Models for Cardiovascular Biology-How Close to Disease? BIOLOGY 2023; 12:468. [PMID: 36979160 PMCID: PMC10045735 DOI: 10.3390/biology12030468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/22/2023]
Abstract
Currently, zebrafish, rodents, canines, and pigs are the primary disease models used in cardiovascular research. In general, larger animals have more physiological similarities to humans, making better disease models. However, they can have restricted or limited use because they are difficult to handle and maintain. Moreover, animal welfare laws regulate the use of experimental animals. Different species have different mechanisms of disease onset. Organs in each animal species have different characteristics depending on their evolutionary history and living environment. For example, mice have higher heart rates than humans. Nonetheless, preclinical studies have used animals to evaluate the safety and efficacy of human drugs because no other complementary method exists. Hence, we need to evaluate the similarities and differences in disease mechanisms between humans and experimental animals. The translation of animal data to humans contributes to eliminating the gap between these two. In vitro disease models have been used as another alternative for human disease models since the discovery of induced pluripotent stem cells (iPSCs). Human cardiomyocytes have been generated from patient-derived iPSCs, which are genetically identical to the derived patients. Researchers have attempted to develop in vivo mimicking 3D culture systems. In this review, we explore the possible uses of animal disease models, iPSC-derived in vitro disease models, humanized animals, and the recent challenges of machine learning. The combination of these methods will make disease models more similar to human disease.
Collapse
Affiliation(s)
- Nanako Kawaguchi
- Department of Pediatric Cardiology and Adult Congenital Cardiology, Tokyo Women’s Medical University, Tokyo 162-8666, Japan;
| | | |
Collapse
|
24
|
Kishino Y, Fukuda K. Unlocking the Pragmatic Potential of Regenerative Therapies in Heart Failure with Next-Generation Treatments. Biomedicines 2023; 11:biomedicines11030915. [PMID: 36979894 PMCID: PMC10046277 DOI: 10.3390/biomedicines11030915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/28/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023] Open
Abstract
Patients with chronic heart failure (HF) have a poor prognosis due to irreversible impairment of left ventricular function, with 5-year survival rates <60%. Despite advances in conventional medicines for HF, prognosis remains poor, and there is a need to improve treatment further. Cell-based therapies to restore the myocardium offer a pragmatic approach that provides hope for the treatment of HF. Although first-generation cell-based therapies using multipotent cells (bone marrow-derived mononuclear cells, mesenchymal stem cells, adipose-derived regenerative cells, and c-kit-positive cardiac cells) demonstrated safety in preclinical models of HF, poor engraftment rates, and a limited ability to form mature cardiomyocytes (CMs) and to couple electrically with existing CMs, meant that improvements in cardiac function in double-blind clinical trials were limited and largely attributable to paracrine effects. The next generation of stem cell therapies uses CMs derived from human embryonic stem cells or, increasingly, from human-induced pluripotent stem cells (hiPSCs). These cell therapies have shown the ability to engraft more successfully and improve electromechanical function of the heart in preclinical studies, including in non-human primates. Advances in cell culture and delivery techniques promise to further improve the engraftment and integration of hiPSC-derived CMs (hiPSC-CMs), while the use of metabolic selection to eliminate undifferentiated cells will help minimize the risk of teratomas. Clinical trials of allogeneic hiPSC-CMs in HF are now ongoing, providing hope for vast numbers of patients with few other options available.
Collapse
Affiliation(s)
| | - Keiichi Fukuda
- Correspondence: ; Tel.: +81-3-5363-3874; Fax: +81-3-5363-3875
| |
Collapse
|
25
|
Shah PP, Keough KC, Gjoni K, Santini GT, Abdill RJ, Wickramasinghe NM, Dundes CE, Karnay A, Chen A, Salomon REA, Walsh PJ, Nguyen SC, Whalen S, Joyce EF, Loh KM, Dubois N, Pollard KS, Jain R. An atlas of lamina-associated chromatin across twelve human cell types reveals an intermediate chromatin subtype. Genome Biol 2023; 24:16. [PMID: 36691074 PMCID: PMC9869549 DOI: 10.1186/s13059-023-02849-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 01/05/2023] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Association of chromatin with lamin proteins at the nuclear periphery has emerged as a potential mechanism to coordinate cell type-specific gene expression and maintain cellular identity via gene silencing. Unlike many histone modifications and chromatin-associated proteins, lamina-associated domains (LADs) are mapped genome-wide in relatively few genetically normal human cell types, which limits our understanding of the role peripheral chromatin plays in development and disease. RESULTS To address this gap, we map LAMIN B1 occupancy across twelve human cell types encompassing pluripotent stem cells, intermediate progenitors, and differentiated cells from all three germ layers. Integrative analyses of this atlas with gene expression and repressive histone modification maps reveal that lamina-associated chromatin in all twelve cell types is organized into at least two subtypes defined by differences in LAMIN B1 occupancy, gene expression, chromatin accessibility, transposable elements, replication timing, and radial positioning. Imaging of fluorescently labeled DNA in single cells validates these subtypes and shows radial positioning of LADs with higher LAMIN B1 occupancy and heterochromatic histone modifications primarily embedded within the lamina. In contrast, the second subtype of lamina-associated chromatin is relatively gene dense, accessible, dynamic across development, and positioned adjacent to the lamina. Most genes gain or lose LAMIN B1 occupancy consistent with cell types along developmental trajectories; however, we also identify examples where the enhancer, but not the gene body and promoter, changes LAD state. CONCLUSIONS Altogether, this atlas represents the largest resource to date for peripheral chromatin organization studies and reveals an intermediate chromatin subtype.
Collapse
Affiliation(s)
- Parisha P Shah
- Departments of Medicine and Cell and Developmental Biology, Penn CVI, Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Smilow TRC, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Kathleen C Keough
- University of California, San Francisco, CA, 94117, USA
- Gladstone Institute of Data Science and Biotechnology, 1650 Owens Street, San Francisco, CA, 94158, USA
| | - Ketrin Gjoni
- University of California, San Francisco, CA, 94117, USA
- Gladstone Institute of Data Science and Biotechnology, 1650 Owens Street, San Francisco, CA, 94158, USA
| | - Garrett T Santini
- Departments of Medicine and Cell and Developmental Biology, Penn CVI, Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Smilow TRC, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Richard J Abdill
- Departments of Medicine and Cell and Developmental Biology, Penn CVI, Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Smilow TRC, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Nadeera M Wickramasinghe
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Carolyn E Dundes
- Department of Developmental Biology and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Ashley Karnay
- Departments of Medicine and Cell and Developmental Biology, Penn CVI, Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Smilow TRC, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Angela Chen
- Department of Developmental Biology and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Rachel E A Salomon
- Department of Developmental Biology and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Patrick J Walsh
- Department of Genetics, Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Son C Nguyen
- Department of Genetics, Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sean Whalen
- Gladstone Institute of Data Science and Biotechnology, 1650 Owens Street, San Francisco, CA, 94158, USA
| | - Eric F Joyce
- Department of Genetics, Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kyle M Loh
- Department of Developmental Biology and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Nicole Dubois
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Katherine S Pollard
- University of California, San Francisco, CA, 94117, USA.
- Gladstone Institute of Data Science and Biotechnology, 1650 Owens Street, San Francisco, CA, 94158, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, 94158, USA.
| | - Rajan Jain
- Departments of Medicine and Cell and Developmental Biology, Penn CVI, Penn Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Smilow TRC, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA.
- Smilow TRC, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA.
| |
Collapse
|
26
|
Shim JV, Xiong Y, Dhanan P, Dariolli R, Azeloglu EU, Hu B, Jayaraman G, Schaniel C, Birtwistle MR, Iyengar R, Dubois NC, Sobie EA. Predicting individual-specific cardiotoxicity responses induced by tyrosine kinase inhibitors. Front Pharmacol 2023; 14:1158222. [PMID: 37101545 PMCID: PMC10123273 DOI: 10.3389/fphar.2023.1158222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/27/2023] [Indexed: 04/28/2023] Open
Abstract
Introduction: Tyrosine kinase inhibitor drugs (TKIs) are highly effective cancer drugs, yet many TKIs are associated with various forms of cardiotoxicity. The mechanisms underlying these drug-induced adverse events remain poorly understood. We studied mechanisms of TKI-induced cardiotoxicity by integrating several complementary approaches, including comprehensive transcriptomics, mechanistic mathematical modeling, and physiological assays in cultured human cardiac myocytes. Methods: Induced pluripotent stem cells (iPSCs) from two healthy donors were differentiated into cardiac myocytes (iPSC-CMs), and cells were treated with a panel of 26 FDA-approved TKIs. Drug-induced changes in gene expression were quantified using mRNA-seq, changes in gene expression were integrated into a mechanistic mathematical model of electrophysiology and contraction, and simulation results were used to predict physiological outcomes. Results: Experimental recordings of action potentials, intracellular calcium, and contraction in iPSC-CMs demonstrated that modeling predictions were accurate, with 81% of modeling predictions across the two cell lines confirmed experimentally. Surprisingly, simulations of how TKI-treated iPSC-CMs would respond to an additional arrhythmogenic insult, namely, hypokalemia, predicted dramatic differences between cell lines in how drugs affected arrhythmia susceptibility, and these predictions were confirmed experimentally. Computational analysis revealed that differences between cell lines in the upregulation or downregulation of particular ion channels could explain how TKI-treated cells responded differently to hypokalemia. Discussion: Overall, the study identifies transcriptional mechanisms underlying cardiotoxicity caused by TKIs, and illustrates a novel approach for integrating transcriptomics with mechanistic mathematical models to generate experimentally testable, individual-specific predictions of adverse event risk.
Collapse
Affiliation(s)
- Jaehee V. Shim
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Yuguang Xiong
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Priyanka Dhanan
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Rafael Dariolli
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Evren U. Azeloglu
- Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Bin Hu
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Gomathi Jayaraman
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Christoph Schaniel
- Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | - Ravi Iyengar
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- *Correspondence: Ravi Iyengar, ; Eric A. Sobie,
| | - Nicole C. Dubois
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Eric A. Sobie
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- *Correspondence: Ravi Iyengar, ; Eric A. Sobie,
| |
Collapse
|
27
|
Hong Y, Zhao Y, Li H, Yang Y, Chen M, Wang X, Luo M, Wang K. Engineering the maturation of stem cell-derived cardiomyocytes. Front Bioeng Biotechnol 2023; 11:1155052. [PMID: 37034258 PMCID: PMC10073467 DOI: 10.3389/fbioe.2023.1155052] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/06/2023] [Indexed: 04/11/2023] Open
Abstract
The maturation of human stem cell-derived cardiomyocytes (hSC-CMs) has been a major challenge to further expand the scope of their application. Over the past years, several strategies have been proven to facilitate the structural and functional maturation of hSC-CMs, which include but are not limited to engineering the geometry or stiffness of substrates, providing favorable extracellular matrices, applying mechanical stretch, fluidic or electrical stimulation, co-culturing with niche cells, regulating biochemical cues such as hormones and transcription factors, engineering and redirecting metabolic patterns, developing 3D cardiac constructs such as cardiac organoid or engineered heart tissue, or culturing under in vivo implantation. In this review, we summarize these maturation strategies, especially the recent advancements, and discussed their advantages as well as the pressing problems that need to be addressed in future studies.
Collapse
Affiliation(s)
- Yi Hong
- Key Laboratory of Molecular Cardiovascular Science, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Ministry of Education, Beijing, China
| | - Yun Zhao
- Key Laboratory of Molecular Cardiovascular Science, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Ministry of Education, Beijing, China
| | - Hao Li
- Key Laboratory of Molecular Cardiovascular Science, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Ministry of Education, Beijing, China
| | - Yunshu Yang
- Key Laboratory of Molecular Cardiovascular Science, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Ministry of Education, Beijing, China
| | - Meining Chen
- Key Laboratory of Molecular Cardiovascular Science, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Ministry of Education, Beijing, China
| | - Xi Wang
- Key Laboratory of Molecular Cardiovascular Science, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Ministry of Education, Beijing, China
- *Correspondence: Kai Wang, ; Mingyao Luo, ; Xi Wang,
| | - Mingyao Luo
- Center of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Vascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, Yunnan, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Beijing, China
- *Correspondence: Kai Wang, ; Mingyao Luo, ; Xi Wang,
| | - Kai Wang
- Key Laboratory of Molecular Cardiovascular Science, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Ministry of Education, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Beijing, China
- *Correspondence: Kai Wang, ; Mingyao Luo, ; Xi Wang,
| |
Collapse
|
28
|
Berg Luecke L, Waas M, Littrell J, Wojtkiewicz M, Castro C, Burkovetskaya M, Schuette EN, Buchberger AR, Churko JM, Chalise U, Waknitz M, Konfrst S, Teuben R, Morrissette-McAlmon J, Mahr C, Anderson DR, Boheler KR, Gundry RL. Surfaceome mapping of primary human heart cells with CellSurfer uncovers cardiomyocyte surface protein LSMEM2 and proteome dynamics in failing hearts. NATURE CARDIOVASCULAR RESEARCH 2023; 2:76-95. [PMID: 36950336 PMCID: PMC10030153 DOI: 10.1038/s44161-022-00200-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 11/29/2022] [Indexed: 01/19/2023]
Abstract
Cardiac cell surface proteins are drug targets and useful biomarkers for discriminating among cellular phenotypes and disease states. Here we developed an analytical platform, CellSurfer, that enables quantitative cell surface proteome (surfaceome) profiling of cells present in limited quantities, and we apply it to isolated primary human heart cells. We report experimental evidence of surface localization and extracellular domains for 1,144 N-glycoproteins, including cell-type-restricted and region-restricted glycoproteins. We identified a surface protein specific for healthy cardiomyocytes, LSMEM2, and validated an anti-LSMEM2 monoclonal antibody for flow cytometry and imaging. Surfaceome comparisons among pluripotent stem cell derivatives and their primary counterparts highlighted important differences with direct implications for drug screening and disease modeling. Finally, 20% of cell surface proteins, including LSMEM2, were differentially abundant between failing and non-failing cardiomyocytes. These results represent a rich resource to advance development of cell type and organ-specific targets for drug delivery, disease modeling, immunophenotyping and in vivo imaging.
Collapse
Affiliation(s)
- Linda Berg Luecke
- CardiOmics Program, Center for Heart and Vascular Research and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE USA
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI USA
| | - Matthew Waas
- CardiOmics Program, Center for Heart and Vascular Research and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE USA
- Present Address: Princess Margaret Cancer Centre, University Health Network, Toronto, ON Canada
| | - Jack Littrell
- CardiOmics Program, Center for Heart and Vascular Research and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE USA
| | - Melinda Wojtkiewicz
- CardiOmics Program, Center for Heart and Vascular Research and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE USA
| | - Chase Castro
- CardiOmics Program, Center for Heart and Vascular Research and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE USA
| | - Maria Burkovetskaya
- CardiOmics Program, Center for Heart and Vascular Research and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE USA
| | - Erin N. Schuette
- CardiOmics Program, Center for Heart and Vascular Research and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE USA
| | - Amanda Rae Buchberger
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI USA
- Present Address: Department of Chemistry, University of Wisconsin-Madison, Madison, WI USA
| | - Jared M. Churko
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ USA
| | - Upendra Chalise
- CardiOmics Program, Center for Heart and Vascular Research and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE USA
| | - Michelle Waknitz
- CardiOmics Program, Center for Heart and Vascular Research and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE USA
| | - Shelby Konfrst
- CardiOmics Program, Center for Heart and Vascular Research and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE USA
| | - Roald Teuben
- Department of Biomedical Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD USA
| | - Justin Morrissette-McAlmon
- Department of Biomedical Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD USA
| | - Claudius Mahr
- Department of Mechanical Engineering, Division of Cardiology, University of Washington, Seattle, WA USA
| | - Daniel R. Anderson
- Division of Cardiovascular Medicine, University of Nebraska Medical Center, Omaha, NE USA
| | - Kenneth R. Boheler
- Department of Biomedical Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD USA
- Department of Medicine, Division of Cardiology, The Johns Hopkins University, Baltimore, MD USA
| | - Rebekah L. Gundry
- CardiOmics Program, Center for Heart and Vascular Research and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE USA
- Division of Cardiovascular Medicine, University of Nebraska Medical Center, Omaha, NE USA
| |
Collapse
|
29
|
Ho BX, Pang JKS, Chen Y, Loh YH, An O, Yang HH, Seshachalam VP, Koh JLY, Chan WK, Ng SY, Soh BS. Robust generation of human-chambered cardiac organoids from pluripotent stem cells for improved modelling of cardiovascular diseases. Stem Cell Res Ther 2022; 13:529. [PMID: 36544188 PMCID: PMC9773542 DOI: 10.1186/s13287-022-03215-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Tissue organoids generated from human pluripotent stem cells are valuable tools for disease modelling and to understand developmental processes. While recent progress in human cardiac organoids revealed the ability of these stem cell-derived organoids to self-organize and intrinsically formed chamber-like structure containing a central cavity, it remained unclear the processes involved that enabled such chamber formation. METHODS Chambered cardiac organoids (CCOs) differentiated from human embryonic stem cells (H7) were generated by modulation of Wnt/ß-catenin signalling under fully defined conditions, and several growth factors essential for cardiac progenitor expansion. Transcriptomic profiling of day 8, day 14 and day 21 CCOs was performed by quantitative PCR and single-cell RNA sequencing. Endothelin-1 (EDN1) known to induce oxidative stress in cardiomyocytes was used to induce cardiac hypertrophy in CCOs in vitro. Functional characterization of cardiomyocyte contractile machinery was performed by immunofluorescence staining and analysis of brightfield and fluorescent video recordings. Quantitative PCR values between groups were compared using two-tailed Student's t tests. Cardiac organoid parameters comparison between groups was performed using two-tailed Mann-Whitney U test when sample size is small; otherwise, Welch's t test was used. Comparison of calcium kinetics parameters derived from the fluorescent data was performed using two-tailed Student's t tests. RESULTS Importantly, we demonstrated that a threshold number of cardiac progenitor was essential to line the circumference of the inner cavity to ensure proper formation of a chamber within the organoid. Single-cell RNA sequencing revealed improved maturation over a time course, as evidenced from increased mRNA expression of cardiomyocyte maturation genes, ion channel genes and a metabolic shift from glycolysis to fatty acid ß-oxidation. Functionally, CCOs recapitulated clinical cardiac hypertrophy by exhibiting thickened chamber walls, reduced fractional shortening, and increased myofibrillar disarray upon treatment with EDN1. Furthermore, electrophysiological assessment of calcium transients displayed tachyarrhythmic phenotype observed as a consequence of rapid depolarization occurring prior to a complete repolarization. CONCLUSIONS Our findings shed novel insights into the role of progenitors in CCO formation and pave the way for the robust generation of cardiac organoids, as a platform for future applications in disease modelling and drug screening in vitro.
Collapse
Affiliation(s)
- Beatrice Xuan Ho
- grid.418812.60000 0004 0620 9243Disease Modelling and Therapeutics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673 Singapore ,grid.4280.e0000 0001 2180 6431Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore, 117543 Singapore
| | - Jeremy Kah Sheng Pang
- grid.418812.60000 0004 0620 9243Disease Modelling and Therapeutics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673 Singapore ,grid.4280.e0000 0001 2180 6431Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore, 117543 Singapore
| | - Ying Chen
- grid.4280.e0000 0001 2180 6431Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore, 117543 Singapore ,grid.4280.e0000 0001 2180 6431Integrative Sciences and Engineering Programme, National University of Singapore, 21 Lower Kent Ridge Road, Singapore, 119077 Singapore ,grid.418812.60000 0004 0620 9243Epigenetics and Cell Fates Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673 Singapore
| | - Yuin-Han Loh
- grid.4280.e0000 0001 2180 6431Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore, 117543 Singapore ,grid.418812.60000 0004 0620 9243Epigenetics and Cell Fates Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673 Singapore
| | - Omer An
- grid.4280.e0000 0001 2180 6431Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore, 117599 Singapore
| | - Henry He Yang
- grid.4280.e0000 0001 2180 6431Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore, 117599 Singapore
| | - Veerabrahma Pratap Seshachalam
- grid.510300.7Computational Phenomics Group, Experimental Drug Development Centre (EDDC), Agency for Science, Technology and Research (A*STAR), Singapore, 138670 Singapore
| | - Judice L. Y. Koh
- grid.510300.7Computational Phenomics Group, Experimental Drug Development Centre (EDDC), Agency for Science, Technology and Research (A*STAR), Singapore, 138670 Singapore
| | - Woon-Khiong Chan
- grid.4280.e0000 0001 2180 6431Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore, 117543 Singapore
| | - Shi Yan Ng
- grid.418812.60000 0004 0620 9243Neurotherapeutics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673 Singapore ,grid.4280.e0000 0001 2180 6431Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Drive, Singapore, 117456 Singapore ,grid.276809.20000 0004 0636 696XNational Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
| | - Boon Seng Soh
- grid.418812.60000 0004 0620 9243Disease Modelling and Therapeutics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673 Singapore ,grid.4280.e0000 0001 2180 6431Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore, 117543 Singapore
| |
Collapse
|
30
|
Sylvén C, Wärdell E, Månsson-Broberg A, Cingolani E, Ampatzis K, Larsson L, Björklund Å, Giacomello S. High cardiomyocyte diversity in human early prenatal heart development. iScience 2022; 26:105857. [PMID: 36624836 PMCID: PMC9823232 DOI: 10.1016/j.isci.2022.105857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 07/19/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022] Open
Abstract
Cardiomyocytes play key roles during cardiogenesis, but have poorly understood features, especially in prenatal stages. Here, we characterized human prenatal cardiomyocytes, 6.5-7 weeks post-conception, by integrating single-cell RNA sequencing, spatial transcriptomics, and ligand-receptor interaction information. Using a computational workflow developed to dissect cell type heterogeneity, localize cell types, and explore their molecular interactions, we identified eight types of developing cardiomyocyte, more than double compared to the ones identified in the Human Developmental Cell Atlas. These have high variability in cell cycle activity, mitochondrial content, and connexin gene expression, and are differentially distributed in the ventricles, including outflow tract, and atria, including sinoatrial node. Moreover, cardiomyocyte ligand-receptor crosstalk is mainly with non-cardiomyocyte cell types, encompassing cardiogenesis-related pathways. Thus, early prenatal human cardiomyocytes are highly heterogeneous and develop unique location-dependent properties, with complex ligand-receptor crosstalk. Further elucidation of their developmental dynamics may give rise to new therapies.
Collapse
Affiliation(s)
- Christer Sylvén
- Department of Medicine, Karolinska Institute, Huddinge, Sweden,Corresponding author
| | - Eva Wärdell
- Department of Medicine, Karolinska Institute, Huddinge, Sweden
| | | | | | | | - Ludvig Larsson
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Åsa Björklund
- Department of Cell and Molecular Biology, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Stefania Giacomello
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden,Corresponding author
| |
Collapse
|
31
|
Dias TP, Baltazar T, Pinto SN, Fernandes TG, Fernandes F, Diogo MM, Prieto M, Cabral JMS. Xeno-Free Integrated Platform for Robust Production of Cardiomyocyte Sheets from hiPSCs. Stem Cells Int 2022; 2022:4542719. [PMID: 36467280 PMCID: PMC9712013 DOI: 10.1155/2022/4542719] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 10/17/2022] [Accepted: 11/02/2022] [Indexed: 08/05/2024] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) can be efficiently differentiated into cardiomyocytes (CMs), which can be used for cardiac disease modeling, for drug screening, and to regenerate damaged myocardium. Implementation of xeno-free culture systems is essential to fully explore the potential of these cells. However, differentiation using xeno-free adhesion matrices often results in low CM yields and lack of functional CM sheets, capable of enduring additional maturation stages. Here, we established a xeno-free CM differentiation platform using TeSR/Synthemax, including a replating step and integrated with two versatile purification/enrichment metabolic approaches. Results showed that the replating step was essential to reestablish a fully integrated, closely-knit CM sheet. In addition, replating contributed to increase the cTnT expression from 65% to 75% and the output from 2.2 to 3.1 CM per hiPSC, comparable with the efficiency observed when using TeSR/Matrigel. In addition, supplementation with PluriSin1 and Glu-Lac+ medium allowed increasing the CM content over 80% without compromising CM sheet integrity or functionality. Thus, this xeno-free differentiation platform is a reliable and robust method to produce hiPSC-derived CMs, increasing the possibility of using these cells safely for a wide range of applications.
Collapse
Affiliation(s)
- Tiago P. Dias
- iBB—Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Tânia Baltazar
- iBB—Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Sandra N. Pinto
- iBB—Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Tiago G. Fernandes
- iBB—Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Fábio Fernandes
- iBB—Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Maria Margarida Diogo
- iBB—Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Manuel Prieto
- iBB—Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Joaquim M. S. Cabral
- iBB—Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| |
Collapse
|
32
|
Sart S, Liu C, Zeng EZ, Xu C, Li Y. Downstream bioprocessing of human pluripotent stem cell-derived therapeutics. Eng Life Sci 2022; 22:667-680. [PMID: 36348655 PMCID: PMC9635003 DOI: 10.1002/elsc.202100042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/08/2021] [Accepted: 08/16/2021] [Indexed: 11/30/2022] Open
Abstract
With the advancement in lineage-specific differentiation from human pluripotent stem cells (hPSCs), downstream cell separation has now become a critical step to produce hPSC-derived products. Since differentiation procedures usually result in a heterogeneous cell population, cell separation needs to be performed either to enrich the desired cell population or remove the undesired cell population. This article summarizes recent advances in separation processes for hPSC-derived cells, including the standard separation technologies, such as magnetic-activated cell sorting, as well as the novel separation strategies, such as those based on adhesion strength and metabolic flux. Specifically, the downstream bioprocessing flow and the identification of surface markers for various cell lineages are discussed. While challenges remain for large-scale downstream bioprocessing of hPSC-derived cells, the rational quality-by-design approach should be implemented to enhance the understanding of the relationship between process and the product and to ensure the safety of the produced cells.
Collapse
Affiliation(s)
- Sebastien Sart
- Laboratory of Physical Microfluidics and BioengineeringDepartment of Genome and GeneticsInstitut PasteurParisFrance
| | - Chang Liu
- Department of Chemical and Biomedical EngineeringFAMU‐FSU College of EngineeringFlorida State UniversityTallahasseeFLUSA
| | - Eric Z. Zeng
- Department of Chemical and Biomedical EngineeringFAMU‐FSU College of EngineeringFlorida State UniversityTallahasseeFLUSA
| | - Chunhui Xu
- Department of PediatricsEmory University School of Medicine and Children's Healthcare of AtlantaAtlantaGAUSA
| | - Yan Li
- Department of Chemical and Biomedical EngineeringFAMU‐FSU College of EngineeringFlorida State UniversityTallahasseeFLUSA
| |
Collapse
|
33
|
Hu C, Li T, Xu Y, Zhang X, Li F, Bai J, Chen J, Jiang W, Yang K, Ou Q, Li X, Wang P, Zhang Y. CellMarker 2.0: an updated database of manually curated cell markers in human/mouse and web tools based on scRNA-seq data. Nucleic Acids Res 2022; 51:D870-D876. [PMID: 36300619 PMCID: PMC9825416 DOI: 10.1093/nar/gkac947] [Citation(s) in RCA: 298] [Impact Index Per Article: 99.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 09/29/2022] [Accepted: 10/11/2022] [Indexed: 01/30/2023] Open
Abstract
CellMarker 2.0 (http://bio-bigdata.hrbmu.edu.cn/CellMarker or http://117.50.127.228/CellMarker/) is an updated database that provides a manually curated collection of experimentally supported markers of various cell types in different tissues of human and mouse. In addition, web tools for analyzing single cell sequencing data are described. We have updated CellMarker 2.0 with more data and several new features, including (i) Appending 36 300 tissue-cell type-maker entries, 474 tissues, 1901 cell types and 4566 markers over the previous version. The current release recruits 26 915 cell markers, 2578 cell types and 656 tissues, resulting in a total of 83 361 tissue-cell type-maker entries. (ii) There is new marker information from 48 sequencing technology sources, including 10X Chromium, Smart-Seq2 and Drop-seq, etc. (iii) Adding 29 types of cell markers, including protein-coding gene lncRNA and processed pseudogene, etc. Additionally, six flexible web tools, including cell annotation, cell clustering, cell malignancy, cell differentiation, cell feature and cell communication, were developed to analysis and visualization of single cell sequencing data. CellMarker 2.0 is a valuable resource for exploring markers of various cell types in different tissues of human and mouse.
Collapse
Affiliation(s)
| | | | | | - Xinxin Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Feng Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Jing Bai
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Jing Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Wenqi Jiang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Kaiyue Yang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Qi Ou
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Xia Li
- Correspondence may also be addressed to Xia Li.
| | - Peng Wang
- Correspondence may also be addressed to Peng Wang.
| | - Yunpeng Zhang
- To whom correspondence should be addressed. Tel: +86 451 86615922;
| |
Collapse
|
34
|
The negative regulation of gene expression by microRNAs as key driver of inducers and repressors of cardiomyocyte differentiation. Clin Sci (Lond) 2022; 136:1179-1203. [PMID: 35979890 PMCID: PMC9411751 DOI: 10.1042/cs20220391] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 11/28/2022]
Abstract
Cardiac muscle damage-induced loss of cardiomyocytes (CMs) and dysfunction of the remaining ones leads to heart failure, which nowadays is the number one killer worldwide. Therapies fostering effective cardiac regeneration are the holy grail of cardiovascular research to stop the heart failure epidemic. The main goal of most myocardial regeneration protocols is the generation of new functional CMs through the differentiation of endogenous or exogenous cardiomyogenic cells. Understanding the cellular and molecular basis of cardiomyocyte commitment, specification, differentiation and maturation is needed to devise innovative approaches to replace the CMs lost after injury in the adult heart. The transcriptional regulation of CM differentiation is a highly conserved process that require sequential activation and/or repression of different genetic programs. Therefore, CM differentiation and specification have been depicted as a step-wise specific chemical and mechanical stimuli inducing complete myogenic commitment and cell-cycle exit. Yet, the demonstration that some microRNAs are sufficient to direct ESC differentiation into CMs and that four specific miRNAs reprogram fibroblasts into CMs show that CM differentiation must also involve negative regulatory instructions. Here, we review the mechanisms of CM differentiation during development and from regenerative stem cells with a focus on the involvement of microRNAs in the process, putting in perspective their negative gene regulation as a main modifier of effective CM regeneration in the adult heart.
Collapse
|
35
|
Laoharawee K, Johnson MJ, Lahr WS, Sipe CJ, Kleinboehl E, Peterson JJ, Lonetree CL, Bell JB, Slipek NJ, Crane AT, Webber BR, Moriarity BS. A Pan-RNase Inhibitor Enabling CRISPR-mRNA Platforms for Engineering of Primary Human Monocytes. Int J Mol Sci 2022; 23:9749. [PMID: 36077152 PMCID: PMC9456164 DOI: 10.3390/ijms23179749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/16/2022] [Accepted: 08/26/2022] [Indexed: 11/23/2022] Open
Abstract
Monocytes and their downstream effectors are critical components of the innate immune system. Monocytes are equipped with chemokine receptors, allowing them to migrate to various tissues, where they can differentiate into macrophage and dendritic cell subsets and participate in tissue homeostasis, infection, autoimmune disease, and cancer. Enabling genome engineering in monocytes and their effector cells will facilitate a myriad of applications for basic and translational research. Here, we demonstrate that CRISPR-Cas9 RNPs can be used for efficient gene knockout in primary human monocytes. In addition, we demonstrate that intracellular RNases are likely responsible for poor and heterogenous mRNA expression as incorporation of pan-RNase inhibitor allows efficient genome engineering following mRNA-based delivery of Cas9 and base editor enzymes. Moreover, we demonstrate that CRISPR-Cas9 combined with an rAAV vector DNA donor template mediates site-specific insertion and expression of a transgene in primary human monocytes. Finally, we demonstrate that SIRPa knock-out monocyte-derived macrophages have enhanced activity against cancer cells, highlighting the potential for application in cellular immunotherapies.
Collapse
Affiliation(s)
- Kanut Laoharawee
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Matthew J. Johnson
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Walker S. Lahr
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Christopher J. Sipe
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Evan Kleinboehl
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Joseph J. Peterson
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Cara-lin Lonetree
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jason B. Bell
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Nicholas J. Slipek
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Andrew T. Crane
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Beau R. Webber
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Branden S. Moriarity
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
36
|
Fang J, Li JJ, Zhong X, Zhou Y, Lee RJ, Cheng K, Li S. Engineering stem cell therapeutics for cardiac repair. J Mol Cell Cardiol 2022; 171:56-68. [PMID: 35863282 DOI: 10.1016/j.yjmcc.2022.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 05/18/2022] [Accepted: 06/25/2022] [Indexed: 10/17/2022]
Abstract
Cardiovascular disease is the leading cause of death in the world. Stem cell-based therapies have been widely investigated for cardiac regeneration in patients with heart failure or myocardial infarction (MI) and surged ahead on multiple fronts over the past two decades. To enhance cellular therapy for cardiac regeneration, numerous engineering techniques have been explored to engineer cells, develop novel scaffolds, make constructs, and deliver cells or their derivatives. This review summarizes the state-of-art stem cell-based therapeutics for cardiac regeneration and discusses the emerged bioengineering approaches toward the enhancement of therapeutic efficacy of stem cell therapies in cardiac repair. We cover the topics in stem cell source and engineering, followed by stem cell-based therapies such as cell aggregates and cell sheets, and biomaterial-mediated stem cell therapies such as stem cell delivery with injectable hydrogel, three-dimensional scaffolds, and microneedle patches. Finally, we discuss future directions and challenges of engineering stem cell therapies for clinical translation.
Collapse
Affiliation(s)
- Jun Fang
- Department of Bioengineering, Department of Medicine, University of California, Los Angeles, Los Angeles, California 90095, USA; School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Jennifer J Li
- Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; Department of Medicine, Cardiovascular Research Institute and Institute for Regeneration Medicine, University of California, San Francisco, CA 94143, USA
| | - Xintong Zhong
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yue Zhou
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Randall J Lee
- Department of Medicine, Cardiovascular Research Institute and Institute for Regeneration Medicine, University of California, San Francisco, CA 94143, USA
| | - Ke Cheng
- Department of Biomedical Engineering, North Carolina State University, NC, USA
| | - Song Li
- Department of Bioengineering, Department of Medicine, University of California, Los Angeles, Los Angeles, California 90095, USA; Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, California 90095, USA.
| |
Collapse
|
37
|
Zhang J, Gregorich ZR, Tao R, Kim GC, Lalit PA, Carvalho JL, Markandeya Y, Mosher DF, Palecek SP, Kamp TJ. Cardiac differentiation of human pluripotent stem cells using defined extracellular matrix proteins reveals essential role of fibronectin. eLife 2022; 11:e69028. [PMID: 35758861 PMCID: PMC9236614 DOI: 10.7554/elife.69028] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/05/2022] [Indexed: 11/13/2022] Open
Abstract
Research and therapeutic applications using human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) require robust differentiation strategies. Efforts to improve hPSC-CM differentiation have largely overlooked the role of extracellular matrix (ECM). The present study investigates the ability of defined ECM proteins to promote hPSC cardiac differentiation. Fibronectin (FN), laminin-111, and laminin-521 enabled hPSCs to attach and expand. However, only addition of FN promoted cardiac differentiation in response to growth factors Activin A, BMP4, and bFGF in contrast to the inhibition produced by laminin-111 or laminin-521. hPSCs in culture produced endogenous FN which accumulated in the ECM to a critical level necessary for effective cardiac differentiation. Inducible shRNA knockdown of FN prevented Brachyury+ mesoderm formation and subsequent hPSC-CM generation. Antibodies blocking FN binding integrins α4β1 or αVβ1, but not α5β1, inhibited cardiac differentiation. Furthermore, inhibition of integrin-linked kinase led to a decrease in phosphorylated AKT, which was associated with increased apoptosis and inhibition of cardiac differentiation. These results provide new insights into defined matrices for culture of hPSCs that enable production of FN-enriched ECM which is essential for mesoderm formation and efficient cardiac differentiation.
Collapse
Affiliation(s)
- Jianhua Zhang
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
- Stem Cell and Regenerative Medicine Center, University of Wisconsin - MadisonMadisonUnited States
| | - Zachery R Gregorich
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
| | - Ran Tao
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
| | - Gina C Kim
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
| | - Pratik A Lalit
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
| | - Juliana L Carvalho
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
- Department of Genomic Sciences and Biotechnology, University of BrasíliaBrasíliaBrazil
| | - Yogananda Markandeya
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
| | - Deane F Mosher
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
- Morgridge Institute for ResearchMadisonUnited States
- Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin-MadisonMadisonUnited States
| | - Sean P Palecek
- Stem Cell and Regenerative Medicine Center, University of Wisconsin - MadisonMadisonUnited States
- Department of Chemical and Biological Engineering, College of Engineering, University of WisconsinMadisonUnited States
| | - Timothy J Kamp
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
- Stem Cell and Regenerative Medicine Center, University of Wisconsin - MadisonMadisonUnited States
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
| |
Collapse
|
38
|
Tsujisaka Y, Hatani T, Okubo C, Ito R, Kimura A, Narita M, Chonabayashi K, Funakoshi S, Lucena-Cacace A, Toyoda T, Osafune K, Kimura T, Saito H, Yoshida Y. Purification of human iPSC-derived cells at large scale using microRNA switch and magnetic-activated cell sorting. Stem Cell Reports 2022; 17:1772-1785. [PMID: 35688152 PMCID: PMC9287667 DOI: 10.1016/j.stemcr.2022.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 11/26/2022] Open
Abstract
For regenerative cell therapies using pluripotent stem cell (PSC)-derived cells, large quantities of purified cells are required. Magnetic-activated cell sorting (MACS) is a powerful approach to collect target antigen-positive cells; however, it remains a challenge to purify various cell types efficiently at large scale without using antibodies specific to the desired cell type. Here we develop a technology that combines microRNA (miRNA)-responsive mRNA switch (miR-switch) with MACS (miR-switch-MACS) to purify large amounts of PSC-derived cells rapidly and effectively. We designed miR-switches that detect specific miRNAs expressed in target cells and controlled the translation of a CD4-coding transgene as a selection marker for MACS. For the large-scale purification of induced PSC-derived cardiomyocytes (iPSC-CMs), we transferred miR-208a-CD4 switch-MACS and obtained purified iPSC-CMs efficiently. Moreover, miR-375-CD4 switch-MACS highly purified pancreatic insulin-producing cells and their progenitors expressing Chromogranin A. Overall, the miR-switch-MACS method can efficiently purify target PSC-derived cells for cell replacement therapy. MiR-208a-CD4 switch-MACS can purify a large amount of iPSC-CMs in a short time MiR-208a switch can purify iPSC-CMs in each subtype-specific protocol MiR-375-CD4 switch-MACS can be applied to pancreatic endocrine precursor cells MiR-switch-MACS method can be efficient for large-scale target cell purification
Collapse
Affiliation(s)
- Yuta Tsujisaka
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan; Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Takeshi Hatani
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan; Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Chikako Okubo
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Ryo Ito
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Azuma Kimura
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Megumi Narita
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Kazuhisa Chonabayashi
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan; Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Shunsuke Funakoshi
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan; Takeda-CiRA Joint Program (T-CiRA), Fujisawa 251-0012, Japan
| | - Antonio Lucena-Cacace
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Taro Toyoda
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Kenji Osafune
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Takeshi Kimura
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Hirohide Saito
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan.
| | - Yoshinori Yoshida
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan; Takeda-CiRA Joint Program (T-CiRA), Fujisawa 251-0012, Japan.
| |
Collapse
|
39
|
Shin JH, Seo BG, Lee IW, Kim HJ, Seo EC, Lee KM, Jeon SB, Baek SK, Kim TS, Lee JH, Choi JW, Hwangbo C, Lee JH. Functional Characterization of Endothelial Cells Differentiated from Porcine Epiblast Stem Cells. Cells 2022; 11:1524. [PMID: 35563830 PMCID: PMC9104949 DOI: 10.3390/cells11091524] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/24/2022] [Accepted: 04/29/2022] [Indexed: 12/12/2022] Open
Abstract
Endothelial cells (ECs), lining blood vessels' lumen, play an essential role in regulating vascular functions. As multifunctional components of vascular structures, pluripotent stem cells (PSCs) are the promising source for potential therapeutic applications in various vascular diseases. Our laboratory has previously established an approach for differentiating porcine epiblast stem cells (pEpiSCs) into ECs, representing an alternative and potentially superior cell source. However, the condition of pEpiSCs-derived ECs growth has yet to be determined, and whether pEpiSCs differentiate into functional ECs remained unclear. Changes in morphology, proliferation and functional endothelial marker were assessed in pEpiSCs-derived ECs in vitro. pEpiSCs-derived ECs were subjected to magnetic-activated cell sorting (MACS) to collect CD-31+ of ECs. We found that sorted ECs showed the highest proliferation rate in differentiation media in primary culture and M199 media in the subculture. Next, sorted ECs were examined for their ability to act as typical vascular ECs through capillary-like structure formation assay, Dil-acetylated low-density lipoprotein (Dil-Ac-LDL) uptake, and three-dimensional spheroid sprouting. Consequently, pEpiSCs-derived ECs function as typical vascular ECs, indicating that pEpiSC-derived ECs might be used to develop cell therapeutics for vascular disease.
Collapse
Affiliation(s)
- Joon-Hong Shin
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52828, Korea; (J.-H.S.); (I.-W.L.); (S.-B.J.); (S.-K.B.); (T.-S.K.)
- Division of Applied Life Science (BK21), PMBBRC and Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, Korea; (B.-G.S.); (H.-J.K.); (E.-C.S.); (K.-M.L.)
| | - Bo-Gyeong Seo
- Division of Applied Life Science (BK21), PMBBRC and Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, Korea; (B.-G.S.); (H.-J.K.); (E.-C.S.); (K.-M.L.)
- Division of Life Science, College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea
| | - In-Won Lee
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52828, Korea; (J.-H.S.); (I.-W.L.); (S.-B.J.); (S.-K.B.); (T.-S.K.)
- Division of Applied Life Science (BK21), PMBBRC and Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, Korea; (B.-G.S.); (H.-J.K.); (E.-C.S.); (K.-M.L.)
| | - Hyo-Jin Kim
- Division of Applied Life Science (BK21), PMBBRC and Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, Korea; (B.-G.S.); (H.-J.K.); (E.-C.S.); (K.-M.L.)
- Division of Life Science, College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea
| | - Eun-Chan Seo
- Division of Applied Life Science (BK21), PMBBRC and Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, Korea; (B.-G.S.); (H.-J.K.); (E.-C.S.); (K.-M.L.)
- Division of Life Science, College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea
| | - Kwang-Min Lee
- Division of Applied Life Science (BK21), PMBBRC and Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, Korea; (B.-G.S.); (H.-J.K.); (E.-C.S.); (K.-M.L.)
- Division of Life Science, College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea
| | - Soo-Been Jeon
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52828, Korea; (J.-H.S.); (I.-W.L.); (S.-B.J.); (S.-K.B.); (T.-S.K.)
| | - Sang-Ki Baek
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52828, Korea; (J.-H.S.); (I.-W.L.); (S.-B.J.); (S.-K.B.); (T.-S.K.)
| | - Tae-Suk Kim
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52828, Korea; (J.-H.S.); (I.-W.L.); (S.-B.J.); (S.-K.B.); (T.-S.K.)
| | - Jeong-Hyung Lee
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon 24414, Korea;
| | - Jung-Woo Choi
- College of Animal Life Science, Kangwon National University, Chuncheon 24414, Korea;
| | - Cheol Hwangbo
- Division of Applied Life Science (BK21), PMBBRC and Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, Korea; (B.-G.S.); (H.-J.K.); (E.-C.S.); (K.-M.L.)
- Division of Life Science, College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea
| | - Joon-Hee Lee
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52828, Korea; (J.-H.S.); (I.-W.L.); (S.-B.J.); (S.-K.B.); (T.-S.K.)
- Institute of Agriculture & Life Science, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52828, Korea
| |
Collapse
|
40
|
Synthetic RNA-based post-transcriptional expression control methods and genetic circuits. Adv Drug Deliv Rev 2022; 184:114196. [PMID: 35288218 DOI: 10.1016/j.addr.2022.114196] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/27/2022] [Accepted: 03/08/2022] [Indexed: 12/19/2022]
Abstract
RNA-based synthetic genetic circuits provide an alternative for traditional transcription-based circuits in applications where genomic integration is to be avoided. Incorporating various post-transcriptional control methods into such circuits allows for controlling the behaviour of the circuit through the detection of certain biomolecular inputs or reconstituting defined circuit behaviours, thus manipulating cellular functions. In this review, recent developments of various types of post-transcriptional control methods in mammalian cells are discussed as well as auxiliary components that allow for the creation and development of mRNA-based switches. How such post-transcriptional switches are combined into synthetic circuits as well as their applications in biomedical and preclinical settings are also described. Finally, we examine the challenges that need to be surmounted before RNA-based synthetic circuits can be reliably deployed into clinical settings.
Collapse
|
41
|
Opportunities and challenges in cardiac tissue engineering from an analysis of two decades of advances. Nat Biomed Eng 2022; 6:327-338. [PMID: 35478227 DOI: 10.1038/s41551-022-00885-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 03/08/2022] [Indexed: 12/20/2022]
Abstract
Engineered human cardiac tissues facilitate progress in regenerative medicine, disease modelling and drug development. In this Perspective, we reflect on the most notable advances in cardiac tissue engineering from the past two decades by analysing pivotal studies and critically examining the most consequential developments. This retrospective analysis led us to identify key milestones and to outline a set of opportunities, along with their associated challenges, for the further advancement of engineered human cardiac tissues.
Collapse
|
42
|
Xu X, Jin K, Bais AS, Zhu W, Yagi H, Feinstein TN, Nguyen PK, Criscione JD, Liu X, Beutner G, Karunakaran KB, Rao KS, He H, Adams P, Kuo CK, Kostka D, Pryhuber GS, Shiva S, Ganapathiraju MK, Porter GA, Lin JHI, Aronow B, Lo CW. Uncompensated mitochondrial oxidative stress underlies heart failure in an iPSC-derived model of congenital heart disease. Cell Stem Cell 2022; 29:840-855.e7. [PMID: 35395180 PMCID: PMC9302582 DOI: 10.1016/j.stem.2022.03.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 11/19/2021] [Accepted: 03/08/2022] [Indexed: 12/14/2022]
Abstract
Hypoplastic left heart syndrome (HLHS) is a severe congenital heart disease with 30% mortality from heart failure (HF) in the first year of life, but the cause of early HF remains unknown. Induced pluripotent stem-cell-derived cardiomyocytes (iPSC-CM) from patients with HLHS showed that early HF is associated with increased apoptosis, mitochondrial respiration defects, and redox stress from abnormal mitochondrial permeability transition pore (mPTP) opening and failed antioxidant response. In contrast, iPSC-CM from patients without early HF showed normal respiration with elevated antioxidant response. Single-cell transcriptomics confirmed that early HF is associated with mitochondrial dysfunction accompanied with endoplasmic reticulum (ER) stress. These findings indicate that uncompensated oxidative stress underlies early HF in HLHS. Importantly, mitochondrial respiration defects, oxidative stress, and apoptosis were rescued by treatment with sildenafil to inhibit mPTP opening or TUDCA to suppress ER stress. Together these findings point to the potential use of patient iPSC-CM for modeling clinical heart failure and the development of therapeutics.
Collapse
Affiliation(s)
- Xinxiu Xu
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kang Jin
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Biomedical Informatics, University of Cincinnati, Cincinnati, OH, USA
| | - Abha S Bais
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Wenjuan Zhu
- Centre for Cardiovascular Genomics and Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Hisato Yagi
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Timothy N Feinstein
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Phong K Nguyen
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA; Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Joseph D Criscione
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Xiaoqin Liu
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gisela Beutner
- Departments of Pediatrics and Environmental Medicine University of Rochester Medical Center Rochester, NY USA
| | - Kalyani B Karunakaran
- Supercomputer Education and Research Centre, Indian Institute of Science, Bangalore, India
| | - Krithika S Rao
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Haoting He
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Phillip Adams
- Anesthesiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Catherine K Kuo
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA; Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA; Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dennis Kostka
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Computational & Systems Biology and Pittsburgh Center for Evolutionary Biology and Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gloria S Pryhuber
- Departments of Pediatrics and Environmental Medicine University of Rochester Medical Center Rochester, NY USA
| | - Sruti Shiva
- Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA; Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - George A Porter
- Pediatrics, Pharmacology, and Physiology, Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Jiuann-Huey Ivy Lin
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bruce Aronow
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Electrical Engineering and Computer Science, University of Cincinnati, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH 45256, USA
| | - Cecilia W Lo
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
43
|
Wickramasinghe NM, Sachs D, Shewale B, Gonzalez DM, Dhanan-Krishnan P, Torre D, LaMarca E, Raimo S, Dariolli R, Serasinghe MN, Mayourian J, Sebra R, Beaumont K, Iyengar S, French DL, Hansen A, Eschenhagen T, Chipuk JE, Sobie EA, Jacobs A, Akbarian S, Ischiropoulos H, Ma'ayan A, Houten SM, Costa K, Dubois NC. PPARdelta activation induces metabolic and contractile maturation of human pluripotent stem cell-derived cardiomyocytes. Cell Stem Cell 2022; 29:559-576.e7. [PMID: 35325615 PMCID: PMC11072853 DOI: 10.1016/j.stem.2022.02.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 06/30/2021] [Accepted: 02/24/2022] [Indexed: 02/09/2023]
Abstract
Pluripotent stem-cell-derived cardiomyocytes (PSC-CMs) provide an unprecedented opportunity to study human heart development and disease, but they are functionally and structurally immature. Here, we induce efficient human PSC-CM (hPSC-CM) maturation through metabolic-pathway modulations. Specifically, we find that peroxisome-proliferator-associated receptor (PPAR) signaling regulates glycolysis and fatty acid oxidation (FAO) in an isoform-specific manner. While PPARalpha (PPARa) is the most active isoform in hPSC-CMs, PPARdelta (PPARd) activation efficiently upregulates the gene regulatory networks underlying FAO, increases mitochondrial and peroxisome content, enhances mitochondrial cristae formation, and augments FAO flux. PPARd activation further increases binucleation, enhances myofibril organization, and improves contractility. Transient lactate exposure, which is frequently used for hPSC-CM purification, induces an independent cardiac maturation program but, when combined with PPARd activation, still enhances oxidative metabolism. In summary, we investigate multiple metabolic modifications in hPSC-CMs and identify a role for PPARd signaling in inducing the metabolic switch from glycolysis to FAO in hPSC-CMs.
Collapse
Affiliation(s)
- Nadeera M Wickramasinghe
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - David Sachs
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bhavana Shewale
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - David M Gonzalez
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Priyanka Dhanan-Krishnan
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Denis Torre
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Elizabeth LaMarca
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Serena Raimo
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Rafael Dariolli
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Madhavika N Serasinghe
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joshua Mayourian
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Robert Sebra
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kristin Beaumont
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Srinivas Iyengar
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Deborah L French
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Arne Hansen
- University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | | | - Jerry E Chipuk
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Eric A Sobie
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Adam Jacobs
- Department of Obstetrics and Gynecology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Schahram Akbarian
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Harry Ischiropoulos
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Avi Ma'ayan
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sander M Houten
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kevin Costa
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nicole C Dubois
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
44
|
Jiang CL, Goyal Y, Jain N, Wang Q, Truitt RE, Coté AJ, Emert B, Mellis IA, Kiani K, Yang W, Jain R, Raj A. Cell type determination for cardiac differentiation occurs soon after seeding of human-induced pluripotent stem cells. Genome Biol 2022; 23:90. [PMID: 35382863 PMCID: PMC8985385 DOI: 10.1186/s13059-022-02654-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 03/16/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Cardiac differentiation of human-induced pluripotent stem (hiPS) cells consistently produces a mixed population of cardiomyocytes and non-cardiac cell types, even when using well-characterized protocols. We sought to determine whether different cell types might result from intrinsic differences in hiPS cells prior to the onset of differentiation. RESULTS By associating individual differentiated cells that share a common hiPS cell precursor, we tested whether expression variability is predetermined from the hiPS cell state. In a single experiment, cells that shared a progenitor were more transcriptionally similar to each other than to other cells in the differentiated population. However, when the same hiPS cells were differentiated in parallel, we did not observe high transcriptional similarity across differentiations. Additionally, we found that substantial cell death occurs during differentiation in a manner that suggested all cells were equally likely to survive or die, suggesting that there is no intrinsic selection bias for cells descended from particular hiPS cell progenitors. We thus wondered how cells grow spatially during differentiation, so we labeled cells by expression of marker genes and found that cells expressing the same marker tended to occur in patches. Our results suggest that cell type determination across multiple cell types, once initiated, is maintained in a cell-autonomous manner for multiple divisions. CONCLUSIONS Altogether, our results show that while substantial heterogeneity exists in the initial hiPS cell population, it is not responsible for the variability observed in differentiated outcomes; instead, factors specifying the various cell types likely act during a window that begins shortly after the seeding of hiPS cells for differentiation.
Collapse
Affiliation(s)
- Connie L Jiang
- Genetics and Epigenetics, Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yogesh Goyal
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center for Synthetic Biology, Northwestern University, Evanston, IL, USA
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Naveen Jain
- Genetics and Epigenetics, Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Qiaohong Wang
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Rachel E Truitt
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Allison J Coté
- Cell Biology, Physiology, and Metabolism, Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Benjamin Emert
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ian A Mellis
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Karun Kiani
- Genetics and Epigenetics, Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wenli Yang
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rajan Jain
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Arjun Raj
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
45
|
Cardiac Cell Therapy with Pluripotent Stem Cell-Derived Cardiomyocytes: What Has Been Done and What Remains to Do? Curr Cardiol Rep 2022; 24:445-461. [PMID: 35275365 PMCID: PMC9068652 DOI: 10.1007/s11886-022-01666-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/05/2022] [Indexed: 01/14/2023]
Abstract
PURPOSE OF REVIEW Exciting pre-clinical data presents pluripotent stem cell-derived cardiomyocytes (PSC-CM) as a novel therapeutic prospect following myocardial infarction, and worldwide clinical trials are imminent. However, despite notable advances, several challenges remain. Here, we review PSC-CM pre-clinical studies, identifying key translational hurdles. We further discuss cell production and characterization strategies, identifying markers that may help generate cells which overcome these barriers. RECENT FINDINGS PSC-CMs can robustly repopulate infarcted myocardium with functional, force generating cardiomyocytes. However, current differentiation protocols produce immature and heterogenous cardiomyocytes, creating related issues such as arrhythmogenicity, immunogenicity and poor engraftment. Recent efforts have enhanced our understanding of cardiovascular developmental biology. This knowledge may help implement novel differentiation or gene editing strategies that could overcome these limitations. PSC-CMs are an exciting therapeutic prospect. Despite substantial recent advances, limitations of the technology remain. However, with our continued and increasing biological understanding, these issues are addressable, with several worldwide clinical trials anticipated in the coming years.
Collapse
|
46
|
Floy ME, Shabnam F, Simmons AD, Bhute VJ, Jin G, Friedrich WA, Steinberg AB, Palecek SP. Advances in Manufacturing Cardiomyocytes from Human Pluripotent Stem Cells. Annu Rev Chem Biomol Eng 2022; 13:255-278. [PMID: 35320695 DOI: 10.1146/annurev-chembioeng-092120-033922] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The emergence of human pluripotent stem cell (hPSC) technology over the past two decades has provided a source of normal and diseased human cells for a wide variety of in vitro and in vivo applications. Notably, hPSC-derived cardiomyocytes (hPSC-CMs) are widely used to model human heart development and disease and are in clinical trials for treating heart disease. The success of hPSC-CMs in these applications requires robust, scalable approaches to manufacture large numbers of safe and potent cells. Although significant advances have been made over the past decade in improving the purity and yield of hPSC-CMs and scaling the differentiation process from 2D to 3D, efforts to induce maturation phenotypes during manufacturing have been slow. Process monitoring and closed-loop manufacturing strategies are just being developed. We discuss recent advances in hPSC-CM manufacturing, including differentiation process development and scaling and downstream processes as well as separation and stabilization. Expected final online publication date for the Annual Review of Chemical and Biomolecular Engineering, Volume 13 is October 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Martha E Floy
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA; , , , , ,
| | - Fathima Shabnam
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA; , , , , ,
| | - Aaron D Simmons
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA; , , , , ,
| | - Vijesh J Bhute
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, Wisconsin, USA; , .,Department of Chemical Engineering, Imperial College London, London, United Kingdom
| | - Gyuhyung Jin
- Department of Chemical Engineering, Purdue University, West Lafayette, Indiana, USA;
| | - Will A Friedrich
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA; , , , , ,
| | - Alexandra B Steinberg
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA; , , , , ,
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA; , , , , ,
| |
Collapse
|
47
|
Park S, Kim H, Lee S, Kim J, Jung T, Choi SW, Park B, Kang S, Elliott DA, Stanley EG, Elefanty AG, Ban K, Park H, Moon S. Effect and application of cryopreserved three‐dimensional microcardiac spheroids in myocardial infarction therapy. Clin Transl Med 2022; 12:e721. [PMID: 35092703 PMCID: PMC8800481 DOI: 10.1002/ctm2.721] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/09/2022] [Accepted: 01/14/2022] [Indexed: 01/13/2023] Open
Affiliation(s)
- Soon‐Jung Park
- Department of Medicine Konkuk University School of Medicine Seoul Korea
- Research Institute T&R Biofab Co. Ltd Siheung Korea
| | - Hyeok Kim
- Department of Medical Life Science, College of Medicine The Catholic University of Korea Seoul Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital The Catholic University of Korea Seoul Korea
| | - Sunghun Lee
- Department of Biomedical Sciences City University of Hong Kong Kowloon Hong Kong SAR
| | - Jongsoo Kim
- Department of Medicine Konkuk University School of Medicine Seoul Korea
- Department of Surgery, Wexner Medical Center Ohio State University Columbus Ohio USA
| | - Taek‐Hee Jung
- Department of Medicine Konkuk University School of Medicine Seoul Korea
- Research Institute T&R Biofab Co. Ltd Siheung Korea
| | - Seong Woo Choi
- Department of Physiology, Department of Biomedical Sciences, College of Medicine Seoul National University Seoul Korea
| | - Bong‐Woo Park
- Department of Medical Life Science, College of Medicine The Catholic University of Korea Seoul Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital The Catholic University of Korea Seoul Korea
| | - Sun‐Woong Kang
- Research Group for Biomimetic Advanced Technology Korea Institute of Toxicology Daejeon Korea
| | - David A. Elliott
- Monash Immunology and Stem Cell Laboratories Monash University Clayton Victoria Australia
| | - Edouard G. Stanley
- Monash Immunology and Stem Cell Laboratories Monash University Clayton Victoria Australia
| | - Andrew G. Elefanty
- Monash Immunology and Stem Cell Laboratories Monash University Clayton Victoria Australia
| | - Kiwon Ban
- Department of Biomedical Sciences City University of Hong Kong Kowloon Hong Kong SAR
| | - Hun‐Jun Park
- Department of Medical Life Science, College of Medicine The Catholic University of Korea Seoul Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital The Catholic University of Korea Seoul Korea
- Cell Death Disease Research Center College of Medicine The Catholic University of Korea Seoul Korea
| | - Sung‐Hwan Moon
- Department of Medicine Konkuk University School of Medicine Seoul Korea
- Research Institute T&R Biofab Co. Ltd Siheung Korea
| |
Collapse
|
48
|
Stem Cell Studies in Cardiovascular Biology and Medicine: A Possible Key Role of Macrophages. BIOLOGY 2022; 11:biology11010122. [PMID: 35053119 PMCID: PMC8773242 DOI: 10.3390/biology11010122] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/26/2021] [Accepted: 01/06/2022] [Indexed: 02/04/2023]
Abstract
Simple Summary Stem cells are used in cardiovascular biology and biomedicine and this field of research is expanding. Two types of stem cells have been used in research: induced pluripotent and somatic stem cells. Induced pluripotent stem cells (iPSCs) are similar to embryonic stem cells (ESCs) in that they can differentiate into somatic cells. Bone marrow stem/stromal cells (BMSCs), adipose-derived stem cells (ASCs), and cardiac stem cells (CSCs) are somatic stem cells that have been used for cardiac regeneration. Recent studies have indicated that exosomes and vesicles from BMSCs and ASCs can be used in regenerative medicine and diagnostics. Chemokines and exosomes can contribute to the communication between inflammatory cells and stem cells to differentiate stem cells into the cell types required for tissue regeneration or repair. In this review, we address these issues based on our research and previous publications. Abstract Stem cells are used in cardiovascular biology and biomedicine, and research in this field is expanding. Two types of stem cells have been used in research: induced pluripotent and somatic stem cells. Stem cell research in cardiovascular medicine has developed rapidly following the discovery of different types of stem cells. Induced pluripotent stem cells (iPSCs) possess potent differentiation ability, unlike somatic stem cells, and have been postulated for a long time. However, differentiating into adult-type mature and functional cardiac myocytes (CMs) remains difficult. Bone marrow stem/stromal cells (BMSCs), adipose-derived stem cells (ASCs), and cardiac stem cells (CSCs) are somatic stem cells used for cardiac regeneration. Among somatic stem cells, bone marrow stem/stromal cells (BMSCs) were the first to be discovered and are relatively well-characterized. BMSCs were once thought to have differentiation ability in infarcted areas of the heart, but it has been identified that paracrine cytokines and micro-RNAs derived from BMSCs contributed to that effect. Moreover, vesicles and exosomes from these cells have similar effects and are effective in cardiac repair. The molecular signature of exosomes can also be used for diagnostics because exosomes have the characteristics of their origin cells. Cardiac stem cells (CSCs) differentiate into cardiomyocytes, smooth muscle cells, and endothelial cells, and supply cardiomyocytes during myocardial infarction by differentiating into newly formed cardiomyocytes. Stem cell niches and inflammatory cells play important roles in stem cell regulation and the recovery of damaged tissues. In particular, chemokines can contribute to the communication between inflammatory cells and stem cells. In this review, we present the current status of this exciting and promising research field.
Collapse
|
49
|
Lee CS, Kim J, Cho HJ, Kim HS. Cardiovascular Regeneration via Stem Cells and Direct Reprogramming: A Review. Korean Circ J 2022; 52:341-353. [PMID: 35502566 PMCID: PMC9064703 DOI: 10.4070/kcj.2022.0005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/16/2022] [Accepted: 02/22/2022] [Indexed: 11/24/2022] Open
Abstract
Despite recent advancements in treatment strategies, cardiovascular disease such as heart failure remains a significant source of global mortality. Stem cell technology and cellular reprogramming are rapidly growing fields that will continue to prove useful in cardiac regenerative therapeutics. This review provides information on the role of human pluripotent stem cells (hPSCs) in cardiac regeneration and discusses the practical applications of hPSC-derived cardiomyocytes (CMCs). Moreover, we discuss the practical applications of hPSC-derived CMCs while outlining the relevance of directly-reprogrammed CMCs in regenerative medicine. This review critically summarizes the most recent advances in the field will help to guide future research in this developing area. Cardiovascular disease (CVD) is the leading causes of morbidity and death globally. In particular, a heart failure remains a major problem that contributes to global mortality. Considerable advancements have been made in conventional pharmacological therapies and coronary intervention surgery for cardiac disorder treatment. However, more than 15% of patients continuously progress to end-stage heart failure and eventually require heart transplantation. Over the past year, numerous numbers of protocols to generate cardiomyocytes (CMCs) from human pluripotent stem cells (hPSCs) have been developed and applied in clinical settings. Number of studies have described the therapeutic effects of hPSCs in animal models and revealed the underlying repair mechanisms of cardiac regeneration. In addition, biomedical engineering technologies have improved the therapeutic potential of hPSC-derived CMCs in vivo. Recently substantial progress has been made in driving the direct differentiation of somatic cells into mature CMCs, wherein an intermediate cellular reprogramming stage can be bypassed. This review provides information on the role of hPSCs in cardiac regeneration and discusses the practical applications of hPSC-derived CMCs; furthermore, it outlines the relevance of directly reprogrammed CMCs in regenerative medicine.
Collapse
Affiliation(s)
- Choon-Soo Lee
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea
| | - Joonoh Kim
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea
| | - Hyun-Jai Cho
- Division of Cardiology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Hyo-Soo Kim
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Medicine or College of Pharmacy, Seoul National University, Seoul, Korea
| |
Collapse
|
50
|
Soma Y, Morita Y, Kishino Y, Kanazawa H, Fukuda K, Tohyama S. The Present State and Future Perspectives of Cardiac Regenerative Therapy Using Human Pluripotent Stem Cells. Front Cardiovasc Med 2021; 8:774389. [PMID: 34957258 PMCID: PMC8692665 DOI: 10.3389/fcvm.2021.774389] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 10/25/2021] [Indexed: 12/13/2022] Open
Abstract
The number of patients with heart failure (HF) is increasing with aging in our society worldwide. Patients with HF who are resistant to medication and device therapy are candidates for heart transplantation (HT). However, the shortage of donor hearts is a serious issue. As an alternative to HT, cardiac regenerative therapy using human pluripotent stem cells (hPSCs), such as human embryonic stem cells and induced pluripotent stem cells, is expected to be realized. Differentiation of hPSCs into cardiomyocytes (CMs) is facilitated by mimicking normal heart development. To prevent tumorigenesis after transplantation, it is important to eliminate non-CMs, including residual hPSCs, and select only CMs. Among many CM selection systems, metabolic selection based on the differences in metabolism between CMs and non-CMs is favorable in terms of cost and efficacy. Large-scale culture systems have been developed because a large number of hPSC-derived CMs (hPSC-CMs) are required for transplantation in clinical settings. In large animal models, hPSC-CMs transplanted into the myocardium improved cardiac function in a myocardial infarction model. Although post-transplantation arrhythmia and immune rejection remain problems, their mechanisms and solutions are under investigation. In this manner, the problems of cardiac regenerative therapy are being solved individually. Thus, cardiac regenerative therapy with hPSC-CMs is expected to become a safe and effective treatment for HF in the near future. In this review, we describe previous studies related to hPSC-CMs and discuss the future perspectives of cardiac regenerative therapy using hPSC-CMs.
Collapse
Affiliation(s)
- Yusuke Soma
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Yuika Morita
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Yoshikazu Kishino
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Hideaki Kanazawa
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Shugo Tohyama
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|