1
|
Zaragoza MV, Bui TA, Widyastuti HP, Mehrabi M, Cang Z, Sha Y, Grosberg A, Nie Q. LMNA-Related Dilated Cardiomyopathy: Single-Cell Transcriptomics during Patient-Derived iPSC Differentiation Support Cell Type and Lineage-Specific Dysregulation of Gene Expression and Development for Cardiomyocytes and Epicardium-Derived Cells with Lamin A/C Haploinsufficiency. Cells 2024; 13:1479. [PMID: 39273049 PMCID: PMC11394257 DOI: 10.3390/cells13171479] [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: 06/12/2024] [Revised: 08/14/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
LMNA-related dilated cardiomyopathy (DCM) is an autosomal-dominant genetic condition with cardiomyocyte and conduction system dysfunction often resulting in heart failure or sudden death. The condition is caused by mutation in the Lamin A/C (LMNA) gene encoding Type-A nuclear lamin proteins involved in nuclear integrity, epigenetic regulation of gene expression, and differentiation. The molecular mechanisms of the disease are not completely understood, and there are no definitive treatments to reverse progression or prevent mortality. We investigated possible mechanisms of LMNA-related DCM using induced pluripotent stem cells derived from a family with a heterozygous LMNA c.357-2A>G splice-site mutation. We differentiated one LMNA-mutant iPSC line derived from an affected female (Patient) and two non-mutant iPSC lines derived from her unaffected sister (Control) and conducted single-cell RNA sequencing for 12 samples (four from Patients and eight from Controls) across seven time points: Day 0, 2, 4, 9, 16, 19, and 30. Our bioinformatics workflow identified 125,554 cells in raw data and 110,521 (88%) high-quality cells in sequentially processed data. Unsupervised clustering, cell annotation, and trajectory inference found complex heterogeneity: ten main cell types; many possible subtypes; and lineage bifurcation for cardiac progenitors to cardiomyocytes (CMs) and epicardium-derived cells (EPDCs). Data integration and comparative analyses of Patient and Control cells found cell type and lineage-specific differentially expressed genes (DEGs) with enrichment, supporting pathway dysregulation. Top DEGs and enriched pathways included 10 ZNF genes and RNA polymerase II transcription in pluripotent cells (PP); BMP4 and TGF Beta/BMP signaling, sarcomere gene subsets and cardiogenesis, CDH2 and EMT in CMs; LMNA and epigenetic regulation, as well as DDIT4 and mTORC1 signaling in EPDCs. Top DEGs also included XIST and other X-linked genes, six imprinted genes (SNRPN, PWAR6, NDN, PEG10, MEG3, MEG8), and enriched gene sets related to metabolism, proliferation, and homeostasis. We confirmed Lamin A/C haploinsufficiency by allelic expression and Western blot. Our complex Patient-derived iPSC model for Lamin A/C haploinsufficiency in PP, CM, and EPDC provided support for dysregulation of genes and pathways, many previously associated with Lamin A/C defects, such as epigenetic gene expression, signaling, and differentiation. Our findings support disruption of epigenomic developmental programs, as proposed in other LMNA disease models. We recognized other factors influencing epigenetics and differentiation; thus, our approach needs improvement to further investigate this mechanism in an iPSC-derived model.
Collapse
Affiliation(s)
- Michael V. Zaragoza
- UCI Cardiogenomics Program, Pediatrics and Biological Chemistry, UC Irvine School of Medicine, Irvine, CA 92697, USA
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Thuy-Anh Bui
- UCI Cardiogenomics Program, Pediatrics and Biological Chemistry, UC Irvine School of Medicine, Irvine, CA 92697, USA
| | - Halida P. Widyastuti
- UCI Cardiogenomics Program, Pediatrics and Biological Chemistry, UC Irvine School of Medicine, Irvine, CA 92697, USA
| | - Mehrsa Mehrabi
- Biomedical Engineering and Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Zixuan Cang
- Mathematics and NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA
| | - Yutong Sha
- Mathematics and NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA
| | - Anna Grosberg
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA
- Biomedical Engineering and Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Qing Nie
- Mathematics and NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|
2
|
Velayutham N, Garbern JC, Elwell HLT, Zhuo Z, Rüland L, Elcure Alvarez F, Frontini S, Rodriguez Carreras Y, Eichholtz M, Ricci‐Blair E, Shaw JY, Bouffard AH, Sokol M, Mancheño Juncosa E, Rhoades S, van den Berg D, Kreymerman A, Aoyama J, Höfflin J, Ryan H, Ho Sui S, Lee RT. P53 Activation Promotes Maturational Characteristics of Pluripotent Stem Cell-Derived Cardiomyocytes in 3-Dimensional Suspension Culture Via FOXO-FOXM1 Regulation. J Am Heart Assoc 2024; 13:e033155. [PMID: 38934864 PMCID: PMC11255683 DOI: 10.1161/jaha.123.033155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 05/02/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Current protocols generate highly pure human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) in vitro that recapitulate characteristics of mature in vivo cardiomyocytes. Yet, a risk of arrhythmias exists when hiPSC-CMs are injected into large animal models. Thus, understanding hiPSC-CM maturational mechanisms is crucial for clinical translation. Forkhead box (FOX) transcription factors regulate postnatal cardiomyocyte maturation through a balance between FOXO and FOXM1. We also previously demonstrated that p53 activation enhances hiPSC-CM maturation. Here, we investigate whether p53 activation modulates the FOXO/FOXM1 balance to promote hiPSC-CM maturation in 3-dimensional suspension culture. METHODS AND RESULTS Three-dimensional cultures of hiPSC-CMs were treated with Nutlin-3a (p53 activator, 10 μM), LOM612 (FOXO relocator, 5 μM), AS1842856 (FOXO inhibitor, 1 μM), or RCM-1 (FOXM1 inhibitor, 1 μM), starting 2 days after onset of beating, with dimethyl sulfoxide (0.2% vehicle) as control. P53 activation promoted hiPSC-CM metabolic and electrophysiological maturation alongside FOXO upregulation and FOXM1 downregulation, in n=3 to 6 per group for all assays. FOXO inhibition significantly decreased expression of cardiac-specific markers such as TNNT2. In contrast, FOXO activation or FOXM1 inhibition promoted maturational characteristics such as increased contractility, oxygen consumption, and voltage peak maximum upstroke velocity, in n=3 to 6 per group for all assays. Further, by single-cell RNA sequencing of n=2 LOM612-treated cells compared with dimethyl sulfoxide, LOM612-mediated FOXO activation promoted expression of cardiac maturational pathways. CONCLUSIONS We show that p53 activation promotes FOXO and suppresses FOXM1 during 3-dimensional hiPSC-CM maturation. These results expand our understanding of hiPSC-CM maturational mechanisms in a clinically-relevant 3-dimensional culture system.
Collapse
Affiliation(s)
- Nivedhitha Velayutham
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell InstituteHarvard UniversityCambridgeMAUSA
| | - Jessica C. Garbern
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell InstituteHarvard UniversityCambridgeMAUSA
- Department of CardiologyBoston Children’s HospitalBostonMAUSA
| | - Hannah L. T. Elwell
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell InstituteHarvard UniversityCambridgeMAUSA
| | - Zhu Zhuo
- Bioinformatics Core, Department of BiostatisticsHarvard T.H. Chan School of Public HealthBostonMAUSA
| | - Laura Rüland
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell InstituteHarvard UniversityCambridgeMAUSA
| | - Farid Elcure Alvarez
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell InstituteHarvard UniversityCambridgeMAUSA
| | - Sara Frontini
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell InstituteHarvard UniversityCambridgeMAUSA
| | - Yago Rodriguez Carreras
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell InstituteHarvard UniversityCambridgeMAUSA
| | - Marie Eichholtz
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell InstituteHarvard UniversityCambridgeMAUSA
| | - Elisabeth Ricci‐Blair
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell InstituteHarvard UniversityCambridgeMAUSA
| | - Jeanna Y. Shaw
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell InstituteHarvard UniversityCambridgeMAUSA
| | - Aldric H. Bouffard
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell InstituteHarvard UniversityCambridgeMAUSA
| | - Morgan Sokol
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell InstituteHarvard UniversityCambridgeMAUSA
| | - Estela Mancheño Juncosa
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell InstituteHarvard UniversityCambridgeMAUSA
| | | | - Daphne van den Berg
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell InstituteHarvard UniversityCambridgeMAUSA
| | - Alexander Kreymerman
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell InstituteHarvard UniversityCambridgeMAUSA
| | - Junya Aoyama
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell InstituteHarvard UniversityCambridgeMAUSA
| | | | | | - Shannan Ho Sui
- Bioinformatics Core, Department of BiostatisticsHarvard T.H. Chan School of Public HealthBostonMAUSA
| | - Richard T. Lee
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell InstituteHarvard UniversityCambridgeMAUSA
- Division of Cardiovascular Medicine, Department of MedicineBrigham and Women’s Hospital and Harvard Medical SchoolBostonMAUSA
| |
Collapse
|
3
|
Saotome H, Yatsuka Y, Minowa O, Shinotsuka K, Tsuchida K, Hirose H, Dai K, Tokuno H, Hayakawa T, Hiranuma H, Hasegawa A, Nakatomi I, Okazaki A, Okazaki Y. Microstripe pattern substrate consisting of alternating planar and nanoprotrusive regions improved hiPSC-derived cardiomyocytes' unidirectional alignment and functional properties. Biomed Mater 2024; 19:045031. [PMID: 38815609 DOI: 10.1088/1748-605x/ad525d] [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: 03/12/2024] [Accepted: 05/30/2024] [Indexed: 06/01/2024]
Abstract
The alignment of each cell in human myocardium is considered critical for the efficient movement of cardiac tissue. We investigated 96-well microstripe-patterned plates to align human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CMs), which resemble fetal myocardium. The aligned CMs (ACMs) cultured on the microstripe-patterned plates exhibited pathology, motor function, gene expression, and drug response that more closely resembled those of adult cells than did unaligned CMs cultured on a flat plate (FCMs). We used these ACMs to evaluate drug side effects and efficacy, and to determine whether these were similar to adult-like responses. When CMs from patients with hypertrophic cardiomyopathy (HCMs) were seeded and cultured on the microstripe-patterned plates or layered on top of the ACMs, both sets of HCMs showed increased heart rate and synchronized contractions, indicating improved cardiac function. It is suggested that the ACMs could be used for drug screening as cells representative of adult-like CMs and be transplanted in the form of a cell sheet for regenerative treatment of heart failure.
Collapse
Affiliation(s)
- Hideo Saotome
- Diagnostics and Therapeutic of Intractable Diseases, Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Yukiko Yatsuka
- Diagnostics and Therapeutic of Intractable Diseases, Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Osamu Minowa
- Diagnostics and Therapeutic of Intractable Diseases, Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Kei Shinotsuka
- Strategic Planning Department, Innovation Promotion Division, Oji Holdings Corporation, Tokyo, Japan
| | - Katsuharu Tsuchida
- Diagnostics and Therapeutic of Intractable Diseases, Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Hitomi Hirose
- Diagnostics and Therapeutic of Intractable Diseases, Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Kotaro Dai
- Strategic Planning Department, Innovation Promotion Division, Oji Holdings Corporation, Tokyo, Japan
| | - Hisako Tokuno
- Strategic Planning Department, Innovation Promotion Division, Oji Holdings Corporation, Tokyo, Japan
| | - Tomohiro Hayakawa
- Diagnostics and Therapeutic of Intractable Diseases, Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Next Generation Medical Business Development Division, Sysmex Corporation, Kobe, Japan
| | - Hidenori Hiranuma
- Strategic Planning Department, Innovation Promotion Division, Oji Holdings Corporation, Tokyo, Japan
| | - Akari Hasegawa
- Strategic Planning Department, Innovation Promotion Division, Oji Holdings Corporation, Tokyo, Japan
| | - Ichiro Nakatomi
- Strategic Planning Department, Innovation Promotion Division, Oji Holdings Corporation, Tokyo, Japan
| | - Atsuko Okazaki
- Diagnostics and Therapeutic of Intractable Diseases, Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Yasushi Okazaki
- Diagnostics and Therapeutic of Intractable Diseases, Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| |
Collapse
|
4
|
Zaragoza MV, Bui TA, Widyastuti HP, Mehrabi M, Cang Z, Sha Y, Grosberg A, Nie Q. LMNA -Related Dilated Cardiomyopathy: Single-Cell Transcriptomics during Patient-derived iPSC Differentiation Support Cell type and Lineage-specific Dysregulation of Gene Expression and Development for Cardiomyocytes and Epicardium-Derived Cells with Lamin A/C Haploinsufficiency. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.598335. [PMID: 38915555 PMCID: PMC11195187 DOI: 10.1101/2024.06.12.598335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
LMNA -Related Dilated Cardiomyopathy (DCM) is an autosomal-dominant genetic condition with cardiomyocyte and conduction system dysfunction often resulting in heart failure or sudden death. The condition is caused by mutation in the Lamin A/C ( LMNA ) gene encoding Type-A nuclear lamin proteins involved in nuclear integrity, epigenetic regulation of gene expression, and differentiation. Molecular mechanisms of disease are not completely understood, and there are no definitive treatments to reverse progression or prevent mortality. We investigated possible mechanisms of LMNA -Related DCM using induced pluripotent stem cells derived from a family with a heterozygous LMNA c.357-2A>G splice-site mutation. We differentiated one LMNA mutant iPSC line derived from an affected female (Patient) and two non-mutant iPSC lines derived from her unaffected sister (Control) and conducted single-cell RNA sequencing for 12 samples (4 Patient and 8 Control) across seven time points: Day 0, 2, 4, 9, 16, 19, and 30. Our bioinformatics workflow identified 125,554 cells in raw data and 110,521 (88%) high-quality cells in sequentially processed data. Unsupervised clustering, cell annotation, and trajectory inference found complex heterogeneity: ten main cell types; many possible subtypes; and lineage bifurcation for Cardiac Progenitors to Cardiomyocytes (CM) and Epicardium-Derived Cells (EPDC). Data integration and comparative analyses of Patient and Control cells found cell type and lineage differentially expressed genes (DEG) with enrichment to support pathway dysregulation. Top DEG and enriched pathways included: 10 ZNF genes and RNA polymerase II transcription in Pluripotent cells (PP); BMP4 and TGF Beta/BMP signaling, sarcomere gene subsets and cardiogenesis, CDH2 and EMT in CM; LMNA and epigenetic regulation and DDIT4 and mTORC1 signaling in EPDC. Top DEG also included: XIST and other X-linked genes, six imprinted genes: SNRPN , PWAR6 , NDN , PEG10 , MEG3 , MEG8 , and enriched gene sets in metabolism, proliferation, and homeostasis. We confirmed Lamin A/C haploinsufficiency by allelic expression and Western blot. Our complex Patient-derived iPSC model for Lamin A/C haploinsufficiency in PP, CM, and EPDC provided support for dysregulation of genes and pathways, many previously associated with Lamin A/C defects, such as epigenetic gene expression, signaling, and differentiation. Our findings support disruption of epigenomic developmental programs as proposed in other LMNA disease models. We recognized other factors influencing epigenetics and differentiation; thus, our approach needs improvement to further investigate this mechanism in an iPSC-derived model.
Collapse
|
5
|
Desa DE, Amitrano MJ, Murphy WL, Skala MC. Optical redox imaging to screen synthetic hydrogels for stem cell-derived cardiomyocyte differentiation and maturation. BIOPHOTONICS DISCOVERY 2024; 1:015002. [PMID: 39036366 PMCID: PMC11258857 DOI: 10.1117/1.bios.1.1.015002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Significance Heart disease is the leading cause of death in the United States, yet research is limited by the inability to culture primary cardiac cells. Cardiomyocytes (CMs) derived from human induced pluripotent stem cells (iPSCs) are a promising solution for drug screening and disease modeling. Aim Induced pluripotent stem cell-derived CM (iPSC-CM) differentiation and maturation studies typically use heterogeneous substrates for growth and destructive verification methods. Reproducible, tunable substrates and touch-free monitoring are needed to identify ideal conditions to produce homogenous, functional CMs. Approach We generated synthetic polyethylene glycol-based hydrogels for iPSC-CM differentiation and maturation. Peptide concentrations, combinations, and gel stiffness were tuned independently. Label-free optical redox imaging (ORI) was performed on a widefield microscope in a 96-well screen of gel formulations. We performed live-cell imaging throughout differentiation and early to late maturation to identify key metabolic shifts. Results Label-free ORI confirmed the expected metabolic shifts toward oxidative phosphorylation throughout the differentiation and maturation processes of iPSC-CMs on synthetic hydrogels. Furthermore, ORI distinguished high and low differentiation efficiency cell batches in the cardiac progenitor stage. Conclusions We established a workflow for medium throughput screening of synthetic hydrogel conditions with the ability to perform repeated live-cell measurements and confirm expected metabolic shifts. These methods have implications for reproducible iPSC-CM generation in biomanufacturing.
Collapse
Affiliation(s)
- Danielle E. Desa
- Morgridge Institute for Research, Madison, Wisconsin, United States
| | - Margot J. Amitrano
- University of Wisconsin-Madison, Department of Biomedical Engineering, Madison, Wisconsin, United States
| | - William L. Murphy
- University of Wisconsin-Madison, Department of Biomedical Engineering, Madison, Wisconsin, United States
- University of Wisconsin-Madison, Department of Orthopedics and Rehabilitation, Madison, Wisconsin, United States
| | - Melissa C. Skala
- Morgridge Institute for Research, Madison, Wisconsin, United States
- University of Wisconsin-Madison, Department of Biomedical Engineering, Madison, Wisconsin, United States
| |
Collapse
|
6
|
Johnson BB, Cosson MV, Tsansizi LI, Holmes TL, Gilmore T, Hampton K, Song OR, Vo NTN, Nasir A, Chabronova A, Denning C, Peffers MJ, Merry CLR, Whitelock J, Troeberg L, Rushworth SA, Bernardo AS, Smith JGW. Perlecan (HSPG2) promotes structural, contractile, and metabolic development of human cardiomyocytes. Cell Rep 2024; 43:113668. [PMID: 38198277 DOI: 10.1016/j.celrep.2023.113668] [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: 05/22/2023] [Revised: 11/01/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Perlecan (HSPG2), a heparan sulfate proteoglycan similar to agrin, is key for extracellular matrix (ECM) maturation and stabilization. Although crucial for cardiac development, its role remains elusive. We show that perlecan expression increases as cardiomyocytes mature in vivo and during human pluripotent stem cell differentiation to cardiomyocytes (hPSC-CMs). Perlecan-haploinsuffient hPSCs (HSPG2+/-) differentiate efficiently, but late-stage CMs have structural, contractile, metabolic, and ECM gene dysregulation. In keeping with this, late-stage HSPG2+/- hPSC-CMs have immature features, including reduced ⍺-actinin expression and increased glycolytic metabolism and proliferation. Moreover, perlecan-haploinsuffient engineered heart tissues have reduced tissue thickness and force generation. Conversely, hPSC-CMs grown on a perlecan-peptide substrate are enlarged and display increased nucleation, typical of hypertrophic growth. Together, perlecan appears to play the opposite role of agrin, promoting cellular maturation rather than hyperplasia and proliferation. Perlecan signaling is likely mediated via its binding to the dystroglycan complex. Targeting perlecan-dependent signaling may help reverse the phenotypic switch common to heart failure.
Collapse
Affiliation(s)
- Benjamin B Johnson
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK
| | - Marie-Victoire Cosson
- The Francis Crick Institute, London NW1 1AT, UK; NHLI, Imperial College London, London, UK
| | - Lorenza I Tsansizi
- The Francis Crick Institute, London NW1 1AT, UK; NHLI, Imperial College London, London, UK
| | - Terri L Holmes
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK
| | | | - Katherine Hampton
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK
| | - Ok-Ryul Song
- The Francis Crick Institute, London NW1 1AT, UK; High-Throughput Screening Science Technology Platform, The Francis Crick Institute, London NW1 1AT, UK
| | - Nguyen T N Vo
- School of Medicine, Regenerating and Modelling Tissues, Biodiscovery Institute, University Park, University of Nottingham, Nottingham NG7 2RD, UK
| | - Aishah Nasir
- School of Medicine, Regenerating and Modelling Tissues, Biodiscovery Institute, University Park, University of Nottingham, Nottingham NG7 2RD, UK
| | - Alzbeta Chabronova
- Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
| | - Chris Denning
- School of Medicine, Regenerating and Modelling Tissues, Biodiscovery Institute, University Park, University of Nottingham, Nottingham NG7 2RD, UK
| | - Mandy J Peffers
- Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
| | - Catherine L R Merry
- School of Medicine, Regenerating and Modelling Tissues, Biodiscovery Institute, University Park, University of Nottingham, Nottingham NG7 2RD, UK; Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - John Whitelock
- School of Medicine, Regenerating and Modelling Tissues, Biodiscovery Institute, University Park, University of Nottingham, Nottingham NG7 2RD, UK; Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Linda Troeberg
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK
| | - Stuart A Rushworth
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK
| | - Andreia S Bernardo
- The Francis Crick Institute, London NW1 1AT, UK; NHLI, Imperial College London, London, UK.
| | - James G W Smith
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK.
| |
Collapse
|
7
|
Mishra A, Tavasoli M, Sokolenko S, McMaster CR, Pasumarthi KB. Atrial natriuretic peptide signaling co-regulates lipid metabolism and ventricular conduction system gene expression in the embryonic heart. iScience 2024; 27:108748. [PMID: 38235330 PMCID: PMC10792247 DOI: 10.1016/j.isci.2023.108748] [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: 05/15/2023] [Revised: 09/15/2023] [Accepted: 12/12/2023] [Indexed: 01/19/2024] Open
Abstract
It has been shown that atrial natriuretic peptide (ANP) and its high affinity receptor (NPRA) are involved in the formation of ventricular conduction system (VCS). Inherited genetic variants in fatty acid oxidation (FAO) genes are known to cause conduction abnormalities in newborn children. Although the effect of ANP on energy metabolism in noncardiac cell types is well documented, the role of lipid metabolism in VCS cell differentiation via ANP/NPRA signaling is not known. In this study, histological sections and primary cultures obtained from E11.5 mouse ventricles were analyzed to determine the role of metabolic adaptations in VCS cell fate determination and maturation. Exogenous treatment of E11.5 ventricular cells with ANP revealed a significant increase in lipid droplet accumulation, FAO and higher expression of VCS marker Cx40. Using specific inhibitors, we further identified PPARγ and FAO as critical downstream regulators of ANP-mediated regulation of metabolism and VCS formation.
Collapse
Affiliation(s)
- Abhishek Mishra
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Mahtab Tavasoli
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Stanislav Sokolenko
- Department of Process Engineering and Applied Science, Dalhousie University, Halifax, NS, Canada
| | | | | |
Collapse
|
8
|
Dingare C, Steventon B. Gastruloids - a minimalistic model to study complex developmental metabolism. Emerg Top Life Sci 2023; 7:455-464. [PMID: 38108463 PMCID: PMC10754324 DOI: 10.1042/etls20230082] [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: 09/16/2023] [Revised: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 12/19/2023]
Abstract
Metabolic networks are well placed to orchestrate the coordination of multiple cellular processes associated with embryonic development such as cell growth, proliferation, differentiation and cell movement. Here, we discuss the advantages that gastruloids, aggregates of mammalian embryonic stem cells that self-assemble a rudimentary body plan, have for uncovering the instructive role of metabolic pathways play in directing developmental processes. We emphasise the importance of using such reductionist systems to link specific pathways to defined events of early mammalian development and their utility for obtaining enough material for metabolomic studies. Finally, we review the ways in which the basic gastruloid protocol can be adapted to obtain specific models of embryonic cell types, tissues and regions. Together, we propose that gastruloids are an ideal system to rapidly uncover new mechanistic links between developmental signalling pathways and metabolic networks, which can then inform precise in vivo studies to confirm their function in the embryo.
Collapse
Affiliation(s)
- Chaitanya Dingare
- Department of Genetics, University of Cambridge, Downing Site, Cambridge CB2 3EH, U.K
| | - Ben Steventon
- Department of Genetics, University of Cambridge, Downing Site, Cambridge CB2 3EH, U.K
| |
Collapse
|
9
|
Bobori SN, Zhu Y, Saarinen A, Liuzzo AJ, Folmes CDL. Metabolic Remodeling during Early Cardiac Lineage Specification of Pluripotent Stem Cells. Metabolites 2023; 13:1086. [PMID: 37887411 PMCID: PMC10608731 DOI: 10.3390/metabo13101086] [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: 09/15/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 10/28/2023] Open
Abstract
Growing evidence indicates that metabolites and energy metabolism play an active rather than consequential role in regulating cellular fate. Cardiac development requires dramatic metabolic remodeling from relying primarily on glycolysis in pluripotent stem cells (PSCs) to oxidizing a wide array of energy substrates to match the high bioenergetic demands of continuous contraction in the developed heart. However, a detailed analysis of how remodeling of energy metabolism contributes to human cardiac development is lacking. Using dynamic multiple reaction monitoring metabolomics of central carbon metabolism, we evaluated temporal changes in energy metabolism during human PSC 3D cardiac lineage specification. Significant metabolic remodeling occurs during the complete differentiation, yet temporal analysis revealed that most changes occur during transitions from pluripotency to mesoderm (day 1) and mesoderm to early cardiac (day 5), with limited maturation of cardiac metabolism beyond day 5. Real-time metabolic analysis demonstrated that while hPSC cardiomyocytes (hPSC-CM) showed elevated rates of oxidative metabolism compared to PSCs, they still retained high glycolytic rates, confirming an immature metabolic phenotype. These observations support the opportunity to metabolically optimize the differentiation process to support lineage specification and maturation of hPSC-CMs.
Collapse
Affiliation(s)
| | | | | | | | - Clifford D. L. Folmes
- Departments of Biochemistry and Molecular Biology and Cardiovascular Medicine, Center for Regenerative Biotherapeutics, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA; (S.N.B.)
| |
Collapse
|
10
|
Lamberto F, Shashikadze B, Elkhateib R, Lombardo SD, Horánszky A, Balogh A, Kistamás K, Zana M, Menche J, Fröhlich T, Dinnyés A. Low-dose Bisphenol A exposure alters the functionality and cellular environment in a human cardiomyocyte model. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 335:122359. [PMID: 37567409 DOI: 10.1016/j.envpol.2023.122359] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/26/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023]
Abstract
Early embryonic development represents a sensitive time-window during which the foetus might be vulnerable to the exposure of environmental contaminants, potentially leading to heart diseases also later in life. Bisphenol A (BPA), a synthetic chemical widely used in plastics manufacturing, has been associated with heart developmental defects, even in low concentrations. This study aims to investigate the effects of environmentally relevant doses of BPA on developing cardiomyocytes using a human induced pluripotent stem cell (hiPSC)-derived model. Firstly, a 2D in vitro differentiation system to obtain cardiomyocytes from hiPSCs (hiPSC-CMs) have been established and characterised to provide a suitable model for the early stages of cardiac development. Then, the effects of a repeated BPA exposure, starting from the undifferentiated stage throughout the differentiation process, were evaluated. The chemical significantly decreased the beat rate of hiPSC-CMs, extending the contraction and relaxation time in a dose-dependent manner. Quantitative proteomics analysis revealed a high abundance of basement membrane (BM) components (e.g., COL4A1, COL4A2, LAMC1, NID2) and a significant increase in TNNC1 and SERBP1 proteins in hiPSC-CMs treated with BPA. Network analysis of proteomics data supported altered extracellular matrix remodelling and provided a disease-gene association with well-known pathological conditions of the heart. Furthermore, upon hypoxia-reoxygenation challenge, hiPSC-CMs treated with BPA showed higher rate of apoptotic events. Taken together, our results revealed that a long-term treatment, even with low doses of BPA, interferes with hiPSC-CMs functionality and alters the surrounding cellular environment, providing new insights about diseases that might arise upon the toxin exposure. Our study contributes to the current understanding of BPA effects on developing human foetal cardiomyocytes, in correlation with human clinical observations and animal studies, and it provides a suitable model for New Approach Methodologies (NAMs) for environmental chemical hazard and risk assessment.
Collapse
Affiliation(s)
- Federica Lamberto
- BioTalentum Ltd., Aulich Lajos Str. 26, Gödöllő, H-2100, Hungary; Department of Physiology and Animal Health, Institute of Physiology and Animal Nutrition, Hungarian University of Agriculture and Life Sciences, Páter Károly Str. 1, H-2100, Gödöllő, Hungary
| | - Bachuki Shashikadze
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, 81377, Munich, Germany
| | - Radwa Elkhateib
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, 81377, Munich, Germany
| | - Salvo Danilo Lombardo
- Max Perutz Labs, Vienna Biocenter Campus (VBC), 1030, Vienna, Austria; Department of Structural and Computational Biology, Center for Molecular Biology, University of Vienna, 1030, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria
| | - Alex Horánszky
- BioTalentum Ltd., Aulich Lajos Str. 26, Gödöllő, H-2100, Hungary; Department of Physiology and Animal Health, Institute of Physiology and Animal Nutrition, Hungarian University of Agriculture and Life Sciences, Páter Károly Str. 1, H-2100, Gödöllő, Hungary
| | - Andrea Balogh
- BioTalentum Ltd., Aulich Lajos Str. 26, Gödöllő, H-2100, Hungary
| | - Kornél Kistamás
- BioTalentum Ltd., Aulich Lajos Str. 26, Gödöllő, H-2100, Hungary
| | - Melinda Zana
- BioTalentum Ltd., Aulich Lajos Str. 26, Gödöllő, H-2100, Hungary
| | - Jörg Menche
- Max Perutz Labs, Vienna Biocenter Campus (VBC), 1030, Vienna, Austria; Department of Structural and Computational Biology, Center for Molecular Biology, University of Vienna, 1030, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria; Faculty of Mathematics, University of Vienna, 1090, Vienna, Austria
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, 81377, Munich, Germany
| | - András Dinnyés
- BioTalentum Ltd., Aulich Lajos Str. 26, Gödöllő, H-2100, Hungary; Department of Physiology and Animal Health, Institute of Physiology and Animal Nutrition, Hungarian University of Agriculture and Life Sciences, Páter Károly Str. 1, H-2100, Gödöllő, Hungary; Department of Cell Biology and Molecular Medicine, University of Szeged, H-6720, Szeged, Hungary.
| |
Collapse
|
11
|
Fenimore JM, Springer DA, Romero ME, Edmondson EF, McVicar DW, Yanpallewar S, Sanford M, Spindel T, Engle E, Meyer TJ, Valencia JC, Young HA. IFN-γ and androgens disrupt mitochondrial function in murine myocytes. J Pathol 2023; 260:276-288. [PMID: 37185821 PMCID: PMC10330777 DOI: 10.1002/path.6081] [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: 08/15/2022] [Revised: 02/11/2023] [Accepted: 03/15/2023] [Indexed: 05/17/2023]
Abstract
The effect of cytokines on non-traditional immunological targets under conditions of chronic inflammation is an ongoing subject of study. Fatigue is a symptom often associated with autoimmune diseases. Chronic inflammatory response and activated cell-mediated immunity are associated with cardiovascular myopathies which can be driven by muscle weakness and fatigue. Thus, we hypothesize that immune dysfunction-driven changes in myocyte mitochondria may play a critical role in fatigue-related pathogenesis. We show that persistent low-level expression of IFN-γ in designated IFN-γ AU-Rich Element deletion mice (ARE mice) under androgen exposure resulted in mitochondrial and metabolic deficiencies in myocytes from male or castrated ARE mice. Most notably, echocardiography unveiled that low ejection fraction in the left ventricle post-stress correlated with mitochondrial deficiencies, explaining how heart function decreases under stress. We report that inefficiencies and structural changes in mitochondria, with changes to expression of mitochondrial genes, are linked to male-biased fatigue and acute cardiomyopathy under stress. Our work highlights how male androgen hormone backgrounds and active autoimmunity reduce mitochondrial function and the ability to cope with stress and how pharmacological blockade of stress signal protects heart function. These studies provide new insight into the diverse actions of IFN-γ in fatigue, energy metabolism, and autoimmunity. © 2023 The Pathological Society of Great Britain and Ireland. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA.
Collapse
Affiliation(s)
- John M Fenimore
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Danielle A Springer
- Murine Phenotyping Core, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | | | - Elijah F Edmondson
- Pathology and Histology Lab, National Cancer Institute, Frederick, MD, USA
| | - Dan W McVicar
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Sudhirkumar Yanpallewar
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Michael Sanford
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Thea Spindel
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Elizabeth Engle
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Thomas J Meyer
- CCR Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Julio C Valencia
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Howard A Young
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| |
Collapse
|
12
|
Cho SW, Kim HK, Sung JH, Kim Y, Kim JH, Han J. Mitochondrial energy metabolic transcriptome profiles during cardiac differentiation from mouse and human pluripotent stem cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY 2022; 26:357-365. [PMID: 36039736 PMCID: PMC9437366 DOI: 10.4196/kjpp.2022.26.5.357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/13/2022] [Accepted: 07/21/2022] [Indexed: 11/15/2022]
Abstract
Simultaneous myofibril and mitochondrial development is crucial for the cardiac differentiation of pluripotent stem cells (PSCs). Specifically, mitochondrial energy metabolism (MEM) development in cardiomyocytes is essential for the beating function. Although previous studies have reported that MEM is correlated with cardiac differentiation, the process and timing of MEM regulation for cardiac differentiation remain poorly understood. Here, we performed transcriptome analysis of cells at specific stages of cardiac differentiation from mouse embryonic stem cells (mESCs) and human induced PSCs (hiPSCs). We selected MEM genes strongly upregulated at cardiac lineage commitment and in a time-dependent manner during cardiac maturation and identified the protein-protein interaction networks. Notably, MEM proteins were found to interact closely with cardiac maturation-related proteins rather than with cardiac lineage commitment-related proteins. Furthermore, MEM proteins were found to primarily interact with cardiac muscle contractile proteins rather than with cardiac transcription factors. We identified several candidate MEM regulatory genes involved in cardiac lineage commitment (Cck, Bdnf, Fabp4, Cebpα, and Cdkn2a in mESC-derived cells, and CCK and NOS3 in hiPSC-derived cells) and cardiac maturation (Ppargc1α, Pgam2, Cox6a2, and Fabp3 in mESC-derived cells, and PGAM2 and SLC25A4 in hiPSC-derived cells). Therefore, our findings show the importance of MEM in cardiac maturation.
Collapse
Affiliation(s)
- Sung Woo Cho
- Division of Cardiology, Department of Internal Medicine, Inje University College of Medicine, Ilsan Paik Hospital, Cardiac & Vascular Center, Goyang 10380, Korea
- Cardiovascular and Metabolic Disease Center, Smart Marine Therapeutics Center, Inje University College of Medicine, Busan 47392, Korea
| | - Hyoung Kyu Kim
- Cardiovascular and Metabolic Disease Center, Smart Marine Therapeutics Center, Inje University College of Medicine, Busan 47392, Korea
- Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, Inje University College of Medicine, Busan 47392, Korea
| | - Ji Hee Sung
- Cardiovascular and Metabolic Disease Center, Smart Marine Therapeutics Center, Inje University College of Medicine, Busan 47392, Korea
- Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, Inje University College of Medicine, Busan 47392, Korea
| | - Yeseul Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Jae Ho Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Korea
| | - Jin Han
- Cardiovascular and Metabolic Disease Center, Smart Marine Therapeutics Center, Inje University College of Medicine, Busan 47392, Korea
- Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, Inje University College of Medicine, Busan 47392, Korea
| |
Collapse
|
13
|
Xu Q, Li G, Osorio D, Zhong Y, Yang Y, Lin YT, Zhang X, Cai JJ. scInTime: A Computational Method Leveraging Single-Cell Trajectory and Gene Regulatory Networks to Identify Master Regulators of Cellular Differentiation. Genes (Basel) 2022; 13:371. [PMID: 35205415 PMCID: PMC8872487 DOI: 10.3390/genes13020371] [Citation(s) in RCA: 4] [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: 12/29/2021] [Revised: 02/05/2022] [Accepted: 02/11/2022] [Indexed: 02/01/2023] Open
Abstract
Trajectory inference (TI) or pseudotime analysis has dramatically extended the analytical framework of single-cell RNA-seq data, allowing regulatory genes contributing to cell differentiation and those involved in various dynamic cellular processes to be identified. However, most TI analysis procedures deal with individual genes independently while overlooking the regulatory relations between genes. Integrating information from gene regulatory networks (GRNs) at different pseudotime points may lead to more interpretable TI results. To this end, we introduce scInTime-an unsupervised machine learning framework coupling inferred trajectory with single-cell GRNs (scGRNs) to identify master regulatory genes. We validated the performance of our method by analyzing multiple scRNA-seq data sets. In each of the cases, top-ranking genes predicted by scInTime supported their functional relevance with corresponding signaling pathways, in line with the results of available functional studies. Overall results demonstrated that scInTime is a powerful tool to exploit pseudotime-series scGRNs, allowing for a clear interpretation of TI results toward more significant biological insights.
Collapse
Affiliation(s)
- Qian Xu
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA;
| | - Guanxun Li
- Department of Statistics, Texas A&M University, College Station, TX 77843, USA;
| | - Daniel Osorio
- Department of Oncology, Institutes of Livestrong Cancer, Dell Medical School, University of Texas at Austin, Austin, TX 78701, USA;
| | - Yan Zhong
- Key Laboratory of Advanced Theory and Application in Statistics and Data Science-MOE, School of Statistics, East China Normal University, Shanghai 200062, China;
| | - Yongjian Yang
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX 77843, USA;
| | - Yu-Te Lin
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 10617, Taiwan;
| | - Xiuren Zhang
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX 77843, USA;
| | - James J. Cai
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA;
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX 77843, USA;
| |
Collapse
|
14
|
Sato T, Ichise N, Kobayashi T, Fusagawa H, Yamazaki H, Kudo T, Tohse N. Enhanced glucose metabolism through activation of HIF-1α covers the energy demand in a rat embryonic heart primordium after heartbeat initiation. Sci Rep 2022; 12:74. [PMID: 34996938 PMCID: PMC8741773 DOI: 10.1038/s41598-021-03832-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 12/08/2021] [Indexed: 12/15/2022] Open
Abstract
The initiation of heartbeat is an essential step in cardiogenesis in the heart primordium, but it remains unclear how intracellular metabolism responds to increased energy demands after heartbeat initiation. In this study, embryos in Wistar rats at embryonic day 10, at which heartbeat begins in rats, were divided into two groups by the heart primordium before and after heartbeat initiation and their metabolic characteristics were assessed. Metabolome analysis revealed that increased levels of ATP, a main product of glucose catabolism, and reduced glutathione, a by-product of the pentose phosphate pathway, were the major determinants in the heart primordium after heartbeat initiation. Glycolytic capacity and ATP synthesis-linked mitochondrial respiration were significantly increased, but subunits in complexes of mitochondrial oxidative phosphorylation were not upregulated in the heart primordium after heartbeat initiation. Hypoxia-inducible factor (HIF)-1α was activated and a glucose transporter and rate-limiting enzymes of the glycolytic and pentose phosphate pathways, which are HIF-1α-downstream targets, were upregulated in the heart primordium after heartbeat initiation. These results suggest that the HIF-1α-mediated enhancement of glycolysis with activation of the pentose phosphate pathway, potentially leading to antioxidant defense and nucleotide biosynthesis, covers the increased energy demand in the beating and developing heart primordium.
Collapse
Affiliation(s)
- Tatsuya Sato
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo, 060-8556, Japan.
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan.
| | - Nobutoshi Ichise
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Takeshi Kobayashi
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Hiroyori Fusagawa
- Department of Orthopedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hiroya Yamazaki
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Taiki Kudo
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Noritsugu Tohse
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo, 060-8556, Japan
| |
Collapse
|
15
|
Modelling Metabolic Shifts during Cardiomyocyte Differentiation, Iron Deficiency and Transferrin Rescue Using Human Pluripotent Stem Cells. Metabolites 2021; 12:metabo12010009. [PMID: 35050131 PMCID: PMC8778576 DOI: 10.3390/metabo12010009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/03/2021] [Accepted: 12/20/2021] [Indexed: 01/13/2023] Open
Abstract
Cardiomyocytes rely on specialised metabolism to meet the high energy demand of the heart. During heart development, metabolism matures and shifts from the predominant utilisation of glycolysis and glutamine oxidation towards lactate and fatty acid oxidation. Iron deficiency (ID) leads to cellular metabolism perturbations. However, the exact alterations in substrate metabolism during ID are poorly defined. Using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM), the present study investigated changes in major metabolic substrate utilisation in the context of ID or upon transferrin rescue. Typically, during hiPSC-CM differentiation, the greatest increase in total metabolic output and rate was seen in fatty acid metabolism. When ID was induced, hiPSC-CMs displayed increased reliance on glycolytic metabolism, and six TCA cycle, five amino acid, and four fatty acid substrates were significantly impaired. Transferrin rescue was able to improve TCA cycle substrate metabolism, but the amino acid and fatty acid metabolism remained perturbed. Replenishing iron stores partially reverses the adverse metabolic changes that occur during ID. Understanding the changes in metabolic substrate utilisation and their modification may provide potential for discovery of new biomarkers and therapeutic targets in cardiovascular diseases.
Collapse
|
16
|
Proteomic Analysis of Exosomes during Cardiogenic Differentiation of Human Pluripotent Stem Cells. Cells 2021; 10:cells10102622. [PMID: 34685602 PMCID: PMC8533815 DOI: 10.3390/cells10102622] [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: 09/10/2021] [Revised: 09/23/2021] [Accepted: 09/28/2021] [Indexed: 11/26/2022] Open
Abstract
Efforts to direct the specification of human pluripotent stem cells (hPSCs) to therapeutically important somatic cell types have focused on identifying proper combinations of soluble cues. Yet, whether exosomes, which mediate intercellular communication, play a role in the differentiation remains unexplored. We took a first step toward addressing this question by subjecting hPSCs to stage-wise specification toward cardiomyocytes (CMs) in scalable stirred-suspension cultures and collecting exosomes. Samples underwent liquid chromatography (LC)/mass spectrometry (MS) and subsequent proteomic analysis revealed over 300 unique proteins from four differentiation stages including proteins such as PPP2CA, AFM, MYH9, MYH10, TRA2B, CTNNA1, EHD1, ACTC1, LDHB, and GPC4, which are linked to cardiogenic commitment. There was a significant correlation of the protein composition of exosomes with the hPSC line and stage of commitment. Differentiating hPSCs treated with exosomes from hPSC-derived CMs displayed improved efficiency of CM formation compared to cells without exogenously added vesicles. Collectively, these results demonstrate that exosomes from hPSCs induced along the CM lineage contain proteins linked to the specification process with modulating effects and open avenues for enhancing the biomanufacturing of stem cell products for cardiac diseases.
Collapse
|
17
|
Ding Q, Qi Y, Tsang SY. Mitochondrial Biogenesis, Mitochondrial Dynamics, and Mitophagy in the Maturation of Cardiomyocytes. Cells 2021; 10:cells10092463. [PMID: 34572112 PMCID: PMC8466139 DOI: 10.3390/cells10092463] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/09/2021] [Accepted: 09/15/2021] [Indexed: 01/01/2023] Open
Abstract
Pluripotent stem cells (PSCs) can undergo unlimited self-renewal and can differentiate into all the cell types present in our body, including cardiomyocytes. Therefore, PSCs can be an excellent source of cardiomyocytes for future regenerative medicine and medical research studies. However, cardiomyocytes obtained from PSC differentiation culture are regarded as immature structurally, electrophysiologically, metabolically, and functionally. Mitochondria are organelles responsible for various cellular functions such as energy metabolism, different catabolic and anabolic processes, calcium fluxes, and various signaling pathways. Cells can respond to cellular needs to increase the mitochondrial mass by mitochondrial biogenesis. On the other hand, cells can also degrade mitochondria through mitophagy. Mitochondria are also dynamic organelles that undergo continuous fusion and fission events. In this review, we aim to summarize previous findings on the changes of mitochondrial biogenesis, mitophagy, and mitochondrial dynamics during the maturation of cardiomyocytes. In addition, we intend to summarize whether changes in these processes would affect the maturation of cardiomyocytes. Lastly, we aim to discuss unanswered questions in the field and to provide insights for the possible strategies of enhancing the maturation of PSC-derived cardiomyocytes.
Collapse
Affiliation(s)
- Qianqian Ding
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China;
| | - Yanxiang Qi
- State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong, China;
| | - Suk-Ying Tsang
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China;
- State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong, China;
- Key Laboratory for Regenerative Medicine, Ministry of Education, The Chinese University of Hong Kong, Hong Kong, China
- The Institute for Tissue Engineering and Regenerative Medicine (iTERM), The Chinese University of Hong Kong, Hong Kong, China
- Correspondence: ; Tel.: +852-39431020
| |
Collapse
|