1
|
Yu Z, Zhang S, Bogomolovas J, Chen J, Evans SM. Intronic RNAscope probes enable precise identification of cardiomyocyte nuclei and cell cycle activity. Commun Biol 2025; 8:577. [PMID: 40195462 PMCID: PMC11977257 DOI: 10.1038/s42003-025-08012-z] [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: 12/18/2024] [Accepted: 03/27/2025] [Indexed: 04/09/2025] Open
Abstract
Cardiac regeneration studies have been plagued by technical challenges in unequivocally identifying cardiomyocyte (CM) nuclei in cardiac sections, crucial for accurate identification of cycling CMs. The use of antibodies to sarcomeric proteins is error-prone, the CM specificity of common nuclear markers is controversial, and utilizing genetically modified mouse models poses risk of inducing unintended cardiac phenotypes. The application of RNAscope intronic probes overcomes the above shortcomings. Intronic probes label intronic RNAs within nuclei and can therefore be utilized as a method for nuclear localization. A Tnnt2 intronic RNAscope probe highly colocalized with Obscurin-H2B-GFP in adult mouse hearts, demonstrating CM specificity. Studies in embryos demonstrated that the Tnnt2 intronic RNAscope probe labeled CM nuclei that had undergone DNA replication, and remained closely associated with CM chromatin at all stages of mitosis, even with nuclear envelope breakdown. The efficiency, accuracy, and perdurance of the Tnnt2 intronic RNAscope probe even with nuclear envelope breakdown facilitated reliable investigation of dynamics of DNA synthesis and potential mitoses in CMs in both border and infarct zones after myocardial infarction (MI). Furthermore, we designed Myl2 and Myl4 intronic RNAscope probes, which labeled ventricular and atrial CM nuclei, respectively, and may help identify CM subtypes generated in vitro.
Collapse
Affiliation(s)
- Zhe Yu
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Sen Zhang
- Department of Pharmacology & Regenerative Medicine, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Julius Bogomolovas
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ju Chen
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Sylvia M Evans
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, 92093, USA.
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
- Department of Pharmacology, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
2
|
Jacyniak K, Barrera Jaimes K, Doan MH, Chartrand JM, Vickaryous MK. Squamate ventricular cardiomyocytes: Ploidy, proliferation, and heart muscle cell size in the leopard gecko (Eublepharis macularius). Dev Dyn 2025. [PMID: 40088131 DOI: 10.1002/dvdy.70015] [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: 05/30/2024] [Revised: 11/16/2024] [Accepted: 02/26/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND While heart function is broadly conserved across vertebrates, the cellular phenotype of muscle cells (cardiomyocytes) varies across taxa and throughout ontogeny. Emerging evidence suggests that some attributes may correlate with the capacity for spontaneous cardiomyocyte replacement following injury. For example, among non-regenerating taxa like adult mammals and birds, cardiomyocytes are polyploid, rarely proliferate, and are large in size. In contrast, in regeneration-competent zebrafish and amphibians, cardiomyocytes are diploid, spontaneously proliferate, and are comparatively small. For other species, less is known. RESULTS Here, we investigate these attributes in the squamate Eublepharis macularius, the leopard gecko. Using the nuclear counterstain DAPI to measure fluorescence intensity as a proxy for DNA content, we found that >90% of adult cardiomyocytes are diploid. Using serial histology and immunostaining for markers of DNA synthesis and mitosis, we determined that adult gecko cardiomyocytes spontaneously proliferate, albeit at significantly lower levels than previously reported in subadults. Furthermore, using wheat germ agglutinin, we found that the cross-sectional area is maintained across ontogeny and that gecko cardiomyocytes are 10× smaller than those of mice. CONCLUSIONS Taken together, our data show that gecko cardiomyocytes share several key cellular attributes with regeneration-competent species and that postnatal ventricular growth occurs via cardiomyocyte hyperplasia.
Collapse
Affiliation(s)
- Kathy Jacyniak
- Department of Biomedical Science, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Karemna Barrera Jaimes
- Department of Biomedical Science, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Minh Hanh Doan
- Department of Biomedical Science, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Jordyn M Chartrand
- Department of Biomedical Science, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Matthew K Vickaryous
- Department of Biomedical Science, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
3
|
Salahi‐Niri A, Zarand P, Mansouri N, Rastgou P, Yazdani O, Esbati R, Shojaeian F, Jahanbin B, Mohsenifar Z, Aghdaei HA, Ardalan FA, Safavi‐Naini SAA. Potential of Proliferative Markers in Pancreatic Cancer Management: A Systematic Review. Health Sci Rep 2025; 8:e70412. [PMID: 40051490 PMCID: PMC11882395 DOI: 10.1002/hsr2.70412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 11/12/2024] [Accepted: 01/13/2025] [Indexed: 03/09/2025] Open
Abstract
Background and Aims Pancreatic cancer is an aggressive malignancy with poor prognosis and limited treatment options. Chemotherapy remains a primary therapeutic approach, but patient responses vary significantly, emphasizing the need for reliable biomarkers. This review explores the potential role of proliferative markers, including Ki-67, PCNA, Cyclin D1, and PHH3, as predictive and prognostic indicators in pancreatic cancer management, aiming to enhance personalized treatment strategies. Methods We conducted a narrative review by searching Scopus, PubMed, and Google Scholar for studies focusing on Ki-67, PCNA, Cyclin D1, and PHH3 in relation to pancreatic cancer and chemotherapy. The literature was reviewed to evaluate the role of these markers in predicting chemotherapy response, tumor progression, and overall patient survival. Results The review highlights the clinical significance of these markers. Ki-67 and PCNA are associated with cell proliferation, while Cyclin D1 regulates cell cycle progression and PHH3 is linked to mitotic activity. High expression levels of these markers often correlate with increased tumor aggressiveness and poorer patient outcomes. Moreover, they show promise in predicting chemotherapy response, which can inform tailored therapeutic strategies. However, challenges remain, including standardization of detection methods and determination of optimal cutoff values. Conclusion Proliferative markers such as Ki-67, PCNA, Cyclin D1, and PHH3 hold potential as predictive and prognostic tools in pancreatic cancer management. Their integration into clinical practice could improve the accuracy of treatment decisions and enhance patient outcomes. Further research and validation are necessary to overcome existing challenges and optimize their application in personalized oncology.
Collapse
Affiliation(s)
- Aryan Salahi‐Niri
- Research, Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Paniz Zarand
- Research, Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Negar Mansouri
- Research, Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Parvaneh Rastgou
- School of MedicineTabriz University of Medical SciencesTabrizIran
| | - Omid Yazdani
- Research, Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Romina Esbati
- Research, Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Fatemeh Shojaeian
- Sidney Kimmel Comprehensive Cancer Research CenterJohns Hopkins School of MedicineBaltimoreMarylandUSA
| | - Behnaz Jahanbin
- Cancer Institute, Pathology Department, Imam Khomeini Hospital ComplexTehran University of Medical SciencesTehranIran
| | - Zhaleh Mohsenifar
- Department of Pathology, Ayatollah Taleghani Educational Hospital, Faculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Hamid Asadzadeh Aghdaei
- Research, Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Farid Azmoudeh Ardalan
- Pathology and Laboratory Medicine Department, Imam Khomeini Hospital ComplexTehran University of Medical SciencesTehranIran
| | | |
Collapse
|
4
|
Derks W, Rode J, Collin S, Rost F, Heinke P, Hariharan A, Pickel L, Simonova I, Lázár E, Graham E, Jashari R, Andrä M, Jeppsson A, Salehpour M, Alkass K, Druid H, Kyriakopoulos CP, Taleb I, Shankar TS, Selzman CH, Sadek H, Jovinge S, Brusch L, Frisén J, Drakos S, Bergmann O. A Latent Cardiomyocyte Regeneration Potential in Human Heart Disease. Circulation 2025; 151:245-256. [PMID: 39569515 PMCID: PMC11748904 DOI: 10.1161/circulationaha.123.067156] [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: 10/06/2023] [Accepted: 09/05/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Cardiomyocytes in the adult human heart show a regenerative capacity, with an annual renewal rate of ≈0.5%. Whether this regenerative capacity of human cardiomyocytes is employed in heart failure has been controversial. METHODS We determined cardiomyocyte renewal in 52 patients with advanced heart failure, 28 of whom received left ventricular assist device support. We measured the concentration of nuclear bomb test-derived 14C in cardiomyocyte genomic DNA and performed mathematical modeling to establish cardiomyocyte renewal in heart failure with and without LVAD unloading. RESULTS We show that cardiomyocyte generation is minimal in end-stage heart failure patients at rates 18 to 50× lower compared with the healthy heart. However, patients receiving left ventricle support device therapy, who showed significant functional and structural cardiac improvement, had a >6-fold increase in cardiomyocyte renewal relative to the healthy heart. CONCLUSIONS Our findings reveal a substantial cardiomyocyte regeneration potential in human heart disease, which could be exploited therapeutically.
Collapse
Affiliation(s)
- Wouter Derks
- Centers for Regenerative Therapies Dresden (W.D., F.R., P.H., A.H., L.P., I.S., O.B.), Technische Universität Dresden, Germany
| | - Julian Rode
- Information Services and High-Performance Computing (J.R., F.R., L.B.), Technische Universität Dresden, Germany
| | - Sofia Collin
- Departments of Cell and Molecular Biology (S.C., E.L., E.G., J.F., O.B.), Stockholm, Sweden
| | - Fabian Rost
- Centers for Regenerative Therapies Dresden (W.D., F.R., P.H., A.H., L.P., I.S., O.B.), Technische Universität Dresden, Germany
- Information Services and High-Performance Computing (J.R., F.R., L.B.), Technische Universität Dresden, Germany
- DRESDEN-Concept Genome Center, Technology Platform at the Center for Molecular and Cellular Bioengineering (F.R.), Technische Universität Dresden, Germany
| | - Paula Heinke
- Centers for Regenerative Therapies Dresden (W.D., F.R., P.H., A.H., L.P., I.S., O.B.), Technische Universität Dresden, Germany
| | - Anjana Hariharan
- Centers for Regenerative Therapies Dresden (W.D., F.R., P.H., A.H., L.P., I.S., O.B.), Technische Universität Dresden, Germany
| | - Lauren Pickel
- Centers for Regenerative Therapies Dresden (W.D., F.R., P.H., A.H., L.P., I.S., O.B.), Technische Universität Dresden, Germany
| | - Irina Simonova
- Centers for Regenerative Therapies Dresden (W.D., F.R., P.H., A.H., L.P., I.S., O.B.), Technische Universität Dresden, Germany
| | - Enikő Lázár
- Departments of Cell and Molecular Biology (S.C., E.L., E.G., J.F., O.B.), Stockholm, Sweden
| | - Evan Graham
- Departments of Cell and Molecular Biology (S.C., E.L., E.G., J.F., O.B.), Stockholm, Sweden
| | | | - Michaela Andrä
- Department of Cardiothoracic and Vascular Surgery, Klinikum Klagenfurt and Section for Surgical Research Medical University Graz, Austria (M.A.)
| | - Anders Jeppsson
- Department of Cardiothoracic Surgery, Sahlgrenska University Hospital (A.J.), Gothenburg, Sweden
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg (A.J.), Gothenburg, Sweden
| | - Mehran Salehpour
- Department of Physics and Astronomy, Applied Nuclear Physics, Uppsala University, Uppsala, Sweden (M.S.)
| | - Kanar Alkass
- Oncology-Pathology (K.A., H.D.), Karolinska Institute, Stockholm, Sweden
- National Board of Forensic Medicine (K.A., H.D.), Stockholm, Sweden
| | - Henrik Druid
- Oncology-Pathology (K.A., H.D.), Karolinska Institute, Stockholm, Sweden
- National Board of Forensic Medicine (K.A., H.D.), Stockholm, Sweden
| | - Christos P. Kyriakopoulos
- Divisions of Cardiovascular Medicine and Cardiothoracic Surgery, University of Utah Health and School of Medicine (C.P.K., I.T., C.H.S., S.D.), Salt Lake City
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah (C.P.K., I.T., T.S.S., C.H.S., S.D.), Salt Lake City
| | - Iosif Taleb
- Divisions of Cardiovascular Medicine and Cardiothoracic Surgery, University of Utah Health and School of Medicine (C.P.K., I.T., C.H.S., S.D.), Salt Lake City
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah (C.P.K., I.T., T.S.S., C.H.S., S.D.), Salt Lake City
| | - Thirupura S. Shankar
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah (C.P.K., I.T., T.S.S., C.H.S., S.D.), Salt Lake City
| | - Craig H. Selzman
- Divisions of Cardiovascular Medicine and Cardiothoracic Surgery, University of Utah Health and School of Medicine (C.P.K., I.T., C.H.S., S.D.), Salt Lake City
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah (C.P.K., I.T., T.S.S., C.H.S., S.D.), Salt Lake City
| | - Hesham Sadek
- The Sarver Heart Center and The Department of Internal Medicine/Cardiology, The University of Arizona College of Medicine Tucson, Arizona (H.S.)
| | - Stefan Jovinge
- Spectrum Health Frederik Meijer Heart and Vascular Institute and Van Andel Institute, Grand Rapids, MI (S.J.)
| | - Lutz Brusch
- Information Services and High-Performance Computing (J.R., F.R., L.B.), Technische Universität Dresden, Germany
| | - Jonas Frisén
- Departments of Cell and Molecular Biology (S.C., E.L., E.G., J.F., O.B.), Stockholm, Sweden
| | - Stavros Drakos
- Divisions of Cardiovascular Medicine and Cardiothoracic Surgery, University of Utah Health and School of Medicine (C.P.K., I.T., C.H.S., S.D.), Salt Lake City
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah (C.P.K., I.T., T.S.S., C.H.S., S.D.), Salt Lake City
| | - Olaf Bergmann
- Centers for Regenerative Therapies Dresden (W.D., F.R., P.H., A.H., L.P., I.S., O.B.), Technische Universität Dresden, Germany
- Departments of Cell and Molecular Biology (S.C., E.L., E.G., J.F., O.B.), Stockholm, Sweden
- Department of Pharmacology and Toxicology, University Medical Center Goettingen (O.B.), Germany
- DZHK (German Centre for Cardiovascular Research), Lower Saxony Partner Site (O.B.), Germany
| |
Collapse
|
5
|
Wang X, Kulik K, Wan TC, Lough JW, Auchampach JA. Histone H2A.Z Deacetylation and Dedifferentiation in Infarcted/Tip60-depleted Cardiomyocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.01.11.575312. [PMID: 38260622 PMCID: PMC10802610 DOI: 10.1101/2024.01.11.575312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Myocardial infarction (MI) results in the loss of billions of cardiomyocytes (CMs), resulting in cardiac dysfunction. To re-muscularize injured myocardium, new CMs must be generated via renewed proliferation of surviving CMs. Approaches to induce proliferation of CMs after injury have been insufficient. Toward this end we are targeting the acetyltransferase Tip60, encoded by the Kat5 gene, based on the rationale that its pleiotropic functions combine to block CM proliferation at multiple checkpoints. We previously demonstrated that genetic depletion of Tip60 in a mouse model after MI reduces scarring, retains cardiac function, and activates the CM cell-cycle, although it remains unclear whether this culminates in the generation of daughter CMs. In order for pre-existing CMs in the adult heart to undergo proliferation, it has become accepted that they must first dedifferentiate, a process highlighted by loss of maturity, epithelial to mesenchymal transitioning (EMT), and reversion from fatty acid oxidation to glycolytic metabolism, accompanied by softening of the myocardial extracellular matrix (ECM). Based on recently published findings that Tip60 induces and maintains the differentiated state of hematopoietic stem cells and neurons via site-specific acetylation of the histone variant H2A.Z, we assessed levels of acetylated H2A.Z and dedifferentiation markers after depleting Tip60 in CMs post-MI. We report that genetic depletion of Tip60 from CMs after MI results in the near obliteration of acetylated H2A.Z in CM nuclei, accompanied by the altered expression of genes indicative of EMT induction, ECM softening, decreased fatty acid oxidation, and depressed expression of genes that regulate the TCA cycle. In accord with the possibility that site-specific acetylation of H2A.Z maintains adult CMs in a mature state of differentiation, CUT&Tag revealed enrichment of H2A.ZacK4/K7 in genetic motifs and in GO terms respectively associated with CM transcription factor binding and muscle development/differentiation. Along with our previous findings, these results support the notion that Tip60 has multiple targets in CMs that combine to maintain the differentiated state and prevent proliferation.
Collapse
Affiliation(s)
- Xinrui Wang
- Department of Pharmacology and Toxicology
- Cardiovascular Center Medical College of Wisconsin Milwaukee, WI 53226
| | - Katherine Kulik
- Department of Cell Biology Neurobiology and Anatomy
- Cardiovascular Center Medical College of Wisconsin Milwaukee, WI 53226
| | - Tina C. Wan
- Department of Pharmacology and Toxicology
- Cardiovascular Center Medical College of Wisconsin Milwaukee, WI 53226
| | - John W. Lough
- Department of Cell Biology Neurobiology and Anatomy
- Cardiovascular Center Medical College of Wisconsin Milwaukee, WI 53226
| | - John A. Auchampach
- Department of Pharmacology and Toxicology
- Cardiovascular Center Medical College of Wisconsin Milwaukee, WI 53226
| |
Collapse
|
6
|
Li J, Gong G, Zhang Y, Zheng Y, He Y, Chen M, He X, Zheng X, Gong X, Liu L, Zhou K, Zhao Z, Iv CWS, Hua Y, Li Y, Guo J. Polyphenol-Nanoengineered Monocyte Biohybrids for Targeted Cardiac Repair and Immunomodulation. Adv Healthc Mater 2025; 14:e2403595. [PMID: 39526529 DOI: 10.1002/adhm.202403595] [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: 09/20/2024] [Revised: 11/02/2024] [Indexed: 11/16/2024]
Abstract
Myocardial infarction is one of the leading cause of cardiovascular death worldwide. Invasive interventional procedures and medications are applied to attenuate the attacks associated with ischemic heart disease by reestablishing blood flow and restoring oxygen supply. However, the overactivation of inflammatory responses and unsatisfactory drug delivery efficiency in the infarcted regions prohibit functional improvement. Here, a nanoengineered monocyte (MO)-based biohybrid system, referred to as CTAs @MOs, for the heart-targeted delivery of combinational therapeutic agents (CTAs) containing anti-inflammatory IL-10 and cardiomyogenic miR-19a to overcome the limitation of malperfusion within the infarcted myocardium through a polyphenol-mediated interfacial assembly, is reported. Systemic administration of CTAs@MOs bypasses extensive thoracotomy and intramyocardial administration risks, leading to infarcted heart-specific accumulation and sustained release of therapeutic agents, enabling immunomodulation of the proinflammatory microenvironment and promoting cardiomyocyte proliferation in sequence. Moreover, CTAs@MOs, which serve as a cellular biohybrid-based therapy, significantly improve cardiac function as evidenced by enhanced ejection fractions, increased fractional shortening, and diminished infarct sizes. This polyphenol nanoengineered biohybrid system represents a general and potent platform for the efficient treatment of cardiovascular disorders.
Collapse
Affiliation(s)
- Jiawen Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatric Dentistry, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Guidong Gong
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Yue Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatric Dentistry, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Yanjiang Zheng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatric Dentistry, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Yunxiang He
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Mei Chen
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Xianglian He
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Xiaolan Zheng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatric Dentistry, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Xue Gong
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatric Dentistry, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Lei Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatric Dentistry, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Kaiyu Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatric Dentistry, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Zongmin Zhao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - C Wyatt Shields Iv
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Colorado, 80303, USA
| | - Yimin Hua
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatric Dentistry, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatric Dentistry, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Junling Guo
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
- Department of Chemical and Biological Engineering, The University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| |
Collapse
|
7
|
Huang S, Wei G, Jia X, Tang Z, Chen Q, Li C, Yan W, Jin M, Li X, Chen Y, Zheng H, Chen G, Liao W, Liao Y, Wang Y, Li J, Bin J. CircRNA-RBAC induces cardiac repair by promoting ribosome biogenesis and cell cycle progression in cardiomyocytes. Int J Biol Macromol 2025; 287:138406. [PMID: 39643169 DOI: 10.1016/j.ijbiomac.2024.138406] [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: 06/23/2024] [Revised: 11/24/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Ribosome biogenesis (RiBi) is an essential process that controls the protein synthesis rate, but its function in regulating endogenous cardiac regeneration remains unknown. Herein, we investigated the function and underlying mechanism of RiBi-associated circRNAs in cardiomyocyte (CM) proliferation and cardiac regeneration. We used high-throughput sequencing, quantitative PCR and in situ hybridization techniques to identify an adult downregulated circRNA, RiBi-associated circRNA (RBAC), in CMs. A functional study further revealed that RBAC overexpression increased ribosome biogenesis activity and cell cycle progression in CMs, while silencing RBAC decreased ribosome biogenesis activity and cell cycle progression. Moreover, RBAC overexpression induced adult CM proliferation and improved cardiac function after myocardial infarction in adult mice. Mechanistically, RBAC controlled ribosome biogenesis and cell proliferation by regulating the proteasome-dependent degradation of Ddx21, thereby altering the localization of Rps14 and reducing Rb expression. Our findings indicate that RBAC upregulation might be a plausible therapeutic strategy to induce endogenous cardiac regeneration.
Collapse
Affiliation(s)
- Senlin Huang
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515 Guangzhou, China
| | - Guoquan Wei
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515 Guangzhou, China
| | - Xiaoqian Jia
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515 Guangzhou, China
| | - Zhenquan Tang
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515 Guangzhou, China
| | - Qiqi Chen
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515 Guangzhou, China
| | - Chuling Li
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515 Guangzhou, China
| | - Wen Yan
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515 Guangzhou, China
| | - Ming Jin
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515 Guangzhou, China
| | - Xinzhong Li
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515 Guangzhou, China
| | - Yanmei Chen
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515 Guangzhou, China
| | - Hao Zheng
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515 Guangzhou, China
| | - Guojun Chen
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515 Guangzhou, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China
| | - Yulin Liao
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515 Guangzhou, China
| | - Yuegang Wang
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515 Guangzhou, China
| | - Jianyong Li
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515 Guangzhou, China.
| | - Jianping Bin
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515 Guangzhou, China.
| |
Collapse
|
8
|
Liu Y, Liu L, Zhuang P, Zou J, Chen X, Wu H, Lu B, Wang WE. A meta-analysis and systematic review of myocardial infarction-induced cardiomyocyte proliferation in adult mouse heart. BMC Med 2024; 22:603. [PMID: 39736615 DOI: 10.1186/s12916-024-03822-0] [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: 07/18/2024] [Accepted: 12/13/2024] [Indexed: 01/01/2025] Open
Abstract
BACKGROUND The proliferation capacity of adult cardiomyocytes is very limited in the normal adult mammalian heart. Previous studies implied that cardiomyocyte proliferation increases after injury stimulation, but the result is controversial partly due to different methodologies. We aim to evaluate whether myocardial infarction (MI) stimulates cardiomyocyte proliferation in adult mice. METHODS A comprehensive literature search was conducted through PubMed/Medline, Embase, and Web of Science databases from 1 January 2000 to 21 December 2023. The SYRCLE's Risk of Bias tool for animal experiments was used to evaluate the quality of the literature by two independent reviewers. Twenty-six studies with cell cycle indicators (Ki67+, PH3+, BrdU/EdU+, and AurkB+) to evaluate cycling cardiomyocytes were collected for a meta-analysis. Another 10 studies with genetic reporter/tracing systems to evaluate cardiomyocyte proliferation were collected for a systematic review. RESULTS Evaluating cardiomyocyte proliferation by immunostaining of the cell cycle indicators on heart tissue, the meta-analysis showed that differences of Ki67+, PH3+, and BrdU/EdU+ cycling cardiomyocytes between MI and Sham groups were not statistically significant. In the post-MI heart, the percentages of PH3+, BrdU/EdU+, and AurkB+ cardiomyocytes were not significantly different between the infarct border zone and remote zone. The percentage of Ki67+ cardiomyocytes in the infarct border zone was statistically higher than that in the remote zone. Most of the studies (6 out of 10) using genetic reporter/tracing mouse systems showed that the difference in cardiomyocyte proliferation between MI and Sham groups was not statistically significant. Among the other 4 studies, at least 3 studies could not demonstrate that MI stimulates bona fide cardiomyocyte proliferation because of methodological shortages. CONCLUSIONS MI injury increases Ki67+ cycling adult mouse cardiomyocytes in infarct border zone. Very little overwhelming evidence shows that MI stimulates bona fide proliferation in the adult heart.
Collapse
Affiliation(s)
- Ya Liu
- Department of Geriatrics, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Street, Chongqing, 400038, China
| | - Lingyan Liu
- Department of Geriatrics, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Street, Chongqing, 400038, China
| | - Pengcheng Zhuang
- Department of Geriatrics, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Street, Chongqing, 400038, China
| | - Jiamin Zou
- Department of Geriatrics, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Street, Chongqing, 400038, China
| | - Xiaokang Chen
- Department of Geriatrics, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Street, Chongqing, 400038, China
| | - Hao Wu
- Department of Geriatrics, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Street, Chongqing, 400038, China
| | - Bingjun Lu
- Department of Geriatrics, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Street, Chongqing, 400038, China
| | - Wei Eric Wang
- Department of Geriatrics, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Street, Chongqing, 400038, China.
| |
Collapse
|
9
|
Lin XL, Lin JH, Cao Y, Zhang H, He SY, Wu HY, Ye ZB, Zheng L, Qi XF. Cardiomyocyte proliferation and heart regeneration in adult Xenopus tropicalis evidenced by a transgenic reporter line. NPJ Regen Med 2024; 9:40. [PMID: 39702515 DOI: 10.1038/s41536-024-00384-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024] Open
Abstract
Cardiomyocyte proliferation in adult Xenopus tropicalis during heart regeneration has remained largely contentious due to the absence of genetic evidence. Here, we generated a transgenic reporter line Tg(mlc2:H2C) expressing mCherry specifically in cardiomyocyte nuclei driven by the promoter of myosin light chain 2 (mlc2). Using the reporter line, we found that traditional whole-cell staining is not a rigorous way to identify cardiomyocytes in adult Xenopus tropicalis when using a cryosection with common thickness (5 μm) which leading to a high error, but this deviation could be reduced by increasing section thickness. In addition, the reporter line confirmed that apex resection injury greatly increased the proliferation of mlc2+ cardiomyocytes at 3-30 days post-resection (dpr), thereby regenerating the lost cardiac muscle by 30 dpr in adult Xenopus tropicalis. Our findings from the reporter line have rigorously defined cardiomyocyte proliferation in adult heart upon injury, thereby contributing heart regeneration in adult Xenopus tropicalis.
Collapse
Affiliation(s)
- Xiao-Lin Lin
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Biology, College of Life Science and Technology, Department of Cardiology, The Affiliated Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, 510632, China
| | - Jin-Hua Lin
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Biology, College of Life Science and Technology, Department of Cardiology, The Affiliated Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, 510632, China
| | - Yan Cao
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Biology, College of Life Science and Technology, Department of Cardiology, The Affiliated Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, 510632, China
| | - Han Zhang
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Biology, College of Life Science and Technology, Department of Cardiology, The Affiliated Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, 510632, China
| | - Si-Yi He
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Biology, College of Life Science and Technology, Department of Cardiology, The Affiliated Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, 510632, China
| | - Hai-Yan Wu
- Department of Hematology, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Ze-Bing Ye
- Department of Cardiology, The Affiliated Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, China.
| | - Li Zheng
- School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou, China.
| | - Xu-Feng Qi
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Biology, College of Life Science and Technology, Department of Cardiology, The Affiliated Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
10
|
Huang H, Park S, Ross I, Moreno J, Khyeam S, Simmons J, Huang GN, Payumo AY. Quantitative label-free digital holographic imaging of cardiomyocyte optical volume, nucleation, and cell division. J Mol Cell Cardiol 2024; 196:94-104. [PMID: 39251060 PMCID: PMC11801258 DOI: 10.1016/j.yjmcc.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/18/2024] [Accepted: 09/05/2024] [Indexed: 09/11/2024]
Abstract
Cardiac regeneration in newborn rodents depends on the ability of pre-existing cardiomyocytes to proliferate and divide. This capacity is lost within the first week of postnatal development when these cells rapidly switch from hyperplasia to hypertrophy, withdraw from the cell cycle, become binucleated, and increase in size. How these dynamic changes in cell size and nucleation impact cardiomyocyte proliferative potential is not well understood. In this study, we innovate the application of a commercially available digital holographic imaging microscope, the Holomonitor M4, to evaluate the proliferative responses of mononucleated and binucleated cardiomyocytes after CHIR99021 treatment, a model proliferative stimulus. This system enables long-term label-free quantitative tracking of primary cardiomyocyte dynamics in real-time with single-cell resolution. Our results confirm that chemical inhibition of glycogen synthase kinase 3 with CHIR99021 promotes complete cell division of both mononucleated and binucleated cardiomyocytes with high frequency. Quantitative tracking of cardiomyocyte volume dynamics during these proliferative events revealed that both mononucleated and binucleated cardiomyocytes reach a similar size-increase threshold prior to attempted cell division. Binucleated cardiomyocytes attempt to divide with lower frequency than mononucleated cardiomyocytes, which may be associated with inadequate increases in cell size. By defining the interrelationship between cardiomyocyte size, nucleation, and cell cycle control, we may better understand the cellular mechanisms that drive the loss of mammalian cardiac regenerative capacity after birth.
Collapse
Affiliation(s)
- Herman Huang
- Department of Biological Sciences, San Jose State University, San Jose, CA 95192, USA
| | - Sangsoon Park
- Cardiovascular Research Institute & Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94158, USA; BAKAR Aging Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ines Ross
- Department of Biological Sciences, San Jose State University, San Jose, CA 95192, USA
| | - Joseph Moreno
- Cardiovascular Research Institute & Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94158, USA; BAKAR Aging Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Sheamin Khyeam
- Cardiovascular Research Institute & Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94158, USA; BAKAR Aging Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jacquelyn Simmons
- Department of Biological Sciences, San Jose State University, San Jose, CA 95192, USA
| | - Guo N Huang
- Cardiovascular Research Institute & Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94158, USA; BAKAR Aging Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Alexander Y Payumo
- Department of Biological Sciences, San Jose State University, San Jose, CA 95192, USA.
| |
Collapse
|
11
|
Schuetz T, Dolejsi T, Beck E, Fugger F, Bild A, Duin MT, Gavranovic-Novakovic J, Hilbold E, Hoffmann T, Zuber J, Bauer A, Ruschitzka F, Bär C, Penninger JM, Haubner BJ. Murine neonatal cardiac regeneration depends on Insulin-like growth factor 1 receptor signaling. Sci Rep 2024; 14:22661. [PMID: 39349545 PMCID: PMC11443045 DOI: 10.1038/s41598-024-72783-4] [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: 01/08/2024] [Accepted: 09/10/2024] [Indexed: 10/02/2024] Open
Abstract
Unlike adult mammals, the hearts of neonatal mice possess the ability to completely regenerate from myocardial infarction (MI). This observation has sparked vast interest in deciphering the potentially lifesaving and morbidity-reducing mechanisms involved in neonatal cardiac regeneration. In mice, the regenerative potential is lost within the first week of life and coincides with a reduction of Insulin-like growth factor 1 receptor (Igf1r) expression in the heart. Igf1r is a well-known regulator of cardiomyocyte maturation and proliferation in neonatal mice. To test the role of Igf1r as a pivotal factor in cardiac regeneration, we knocked down (KD) Igf1r specifically in cardiomyocytes using recombinant adeno-associated virus (rAAV) delivery and troponin T promotor driven shRNAmirs. Cardiomyocyte specific Igf1r KD versus control mice were subjected to experimental MI by permanent ligation of the left anterior descending artery (LAD). Cardiac functional and morphological data were analyzed over a 21-day period. Neonatal Igf1r KD mice showed reduced systolic cardiac function and increased fibrotic cardiac remodeling 21 days post injury. This cardiac phenotype was associated with reduced cardiomyocyte nuclei mitosis and decreased AKT and ERK phosphorylation in Igf1r KD, compared to control neonatal mouse hearts. Our in vivo murine data show that Igf1r KD shifts neonatal cardiac regeneration to a more adult-like scarring phenotype, identifying cardiomyocyte-specific Igf1r signaling as a crucial component of neonatal cardiac regeneration.
Collapse
Affiliation(s)
- Thomas Schuetz
- Department of Internal Medicine III (Cardiology and Angiology), Innsbruck Medical University, Innsbruck, Austria
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna, Austria
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Theresa Dolejsi
- Department of Internal Medicine III (Cardiology and Angiology), Innsbruck Medical University, Innsbruck, Austria
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Eva Beck
- Department of Internal Medicine III (Cardiology and Angiology), Innsbruck Medical University, Innsbruck, Austria
| | - Fabio Fugger
- Department of Internal Medicine III (Cardiology and Angiology), Innsbruck Medical University, Innsbruck, Austria
| | - Alexander Bild
- Department of Internal Medicine III (Cardiology and Angiology), Innsbruck Medical University, Innsbruck, Austria
| | - Marie-Theres Duin
- Department of Internal Medicine III (Cardiology and Angiology), Innsbruck Medical University, Innsbruck, Austria
| | - Jasmina Gavranovic-Novakovic
- Department of Internal Medicine III (Cardiology and Angiology), Innsbruck Medical University, Innsbruck, Austria
| | - Erika Hilbold
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | | | | | - Axel Bauer
- Department of Internal Medicine III (Cardiology and Angiology), Innsbruck Medical University, Innsbruck, Austria
| | - Frank Ruschitzka
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Christian Bär
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Josef Martin Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna, Austria.
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.
- Helmholtz Centre for Infection Research, Braunschweig, Germany.
| | - Bernhard Johannes Haubner
- Department of Internal Medicine III (Cardiology and Angiology), Innsbruck Medical University, Innsbruck, Austria.
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna, Austria.
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
- Department of Cardiology, University Heart Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland.
| |
Collapse
|
12
|
Swift SK, Purdy AL, Buddell T, Lovett JJ, Chanjeevaram SV, Arkatkar A, O'Meara CC, Patterson M. A broadly applicable method for quantifying cardiomyocyte cell division identifies proliferative events following myocardial infarction. CELL REPORTS METHODS 2024; 4:100860. [PMID: 39255794 PMCID: PMC11440799 DOI: 10.1016/j.crmeth.2024.100860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/12/2024] [Accepted: 08/15/2024] [Indexed: 09/12/2024]
Abstract
Cardiomyocyte proliferation is a challenging metric to assess. Current methodologies have limitations in detecting the generation of new cardiomyocytes and technical challenges that reduce widespread applicability. Here, we describe an improved cell suspension and imaging-based methodology that can be broadly employed to assess cardiomyocyte cell division in standard laboratories across a multitude of model organisms and experimental conditions. We highlight additional metrics that can be gathered from the same cell preparations to enable additional relevant analyses to be performed. We incorporate additional antibody stains to address potential technical concerns of miscounting. Finally, we employ this methodology with a dual-thymidine analog-labeling approach to a post-infarction murine model, which allowed us to robustly identify unique cycling events, such as cardiomyocytes undergoing multiple rounds of cell division.
Collapse
Affiliation(s)
- Samantha K Swift
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Alexandra L Purdy
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Tyler Buddell
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jerrell J Lovett
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Smrithi V Chanjeevaram
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Anooj Arkatkar
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Caitlin C O'Meara
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Michaela Patterson
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
13
|
Akins KA, Flinn MA, Swift SK, Chanjeevaram SV, Purdy AL, Buddell T, Kolell ME, Andresen KG, Paddock S, Buday SL, Veldman MB, O'Meara CC, Patterson M. Runx1 is sufficient but not required for cardiomyocyte cell-cycle activation. Am J Physiol Heart Circ Physiol 2024; 327:H377-H389. [PMID: 38847758 PMCID: PMC11442100 DOI: 10.1152/ajpheart.00782.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 05/08/2024] [Accepted: 05/30/2024] [Indexed: 07/23/2024]
Abstract
Factors responsible for cardiomyocyte proliferation could serve as potential therapeutics to stimulate endogenous myocardial regeneration following insult, such as ischemic injury. A previously published forward genetics approach on cardiomyocyte cell cycle and ploidy led us to the transcription factor, Runx1. Here, we examine the effect of Runx1 on cardiomyocyte cell cycle during postnatal development and cardiac regeneration using cardiomyocyte-specific gain- and loss-of-function mouse models. RUNX1 is expressed in cardiomyocytes during early postnatal life, decreases to negligible levels by 3 wk of age, and increases upon myocardial injury, all consistent with observed rates of cardiomyocyte cell-cycle activity. Loss of Runx1 transiently stymied cardiomyocyte cell-cycle activity during normal postnatal development, a result that corrected itself and did not extend to the context of neonatal heart regeneration. On the other hand, cardiomyocyte-specific Runx1 overexpression resulted in an expansion of diploid cardiomyocytes in uninjured hearts and expansion of 4 N cardiomyocytes in the context of neonatal cardiac injury, suggesting Runx1 overexpression is sufficient to induce cardiomyocyte cell-cycle responses. Persistent overexpression of Runx1 for >1 mo continued to promote cardiomyocyte cell-cycle activity resulting in substantial hyperpolyploidization (≥8 N DNA content). This persistent cell-cycle activation was accompanied by ventricular dilation and adverse remodeling, raising the concern that continued cardiomyocyte cell cycling can have detrimental effects.NEW & NOTEWORTHY Runx1 is sufficient but not required for cardiomyocyte cell cycle.
Collapse
Grants
- R01 HL170547 NHLBI NIH HHS
- T32 HL007852 NHLBI NIH HHS
- R01HL155085 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL155085 NHLBI NIH HHS
- F32HL150958 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- S10OD025038 HHS | NIH | NIH Office of the Director (OD)
- F31HL162468 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- T32 HL134643 NHLBI NIH HHS
- R01HL170547 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R21 HL156022 NHLBI NIH HHS
- R01HL155085-S1 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL141159 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- T32HL007852 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- T32HL034643 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- F32 HL150958 NHLBI NIH HHS
- F31 HL162468 NHLBI NIH HHS
- R01 HL141159 NHLBI NIH HHS
- S10 OD025038 NIH HHS
Collapse
Affiliation(s)
- Kaelin A Akins
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Michael A Flinn
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Samantha K Swift
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Smrithi V Chanjeevaram
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Alexandra L Purdy
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Tyler Buddell
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Mary E Kolell
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Kaitlyn G Andresen
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Samantha Paddock
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Sydney L Buday
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Matthew B Veldman
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Caitlin C O'Meara
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Michaela Patterson
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| |
Collapse
|
14
|
Cottle B, Tiwari S, Kaza A, Sachse FB, Hitchcock R. Intraoperative characterization of cardiac tissue: the potential of light scattering spectroscopy. JOURNAL OF BIOMEDICAL OPTICS 2024; 29:066005. [PMID: 38841076 PMCID: PMC11152447 DOI: 10.1117/1.jbo.29.6.066005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/22/2024] [Accepted: 05/13/2024] [Indexed: 06/07/2024]
Abstract
Significance Damage to the cardiac conduction system remains one of the most significant risks associated with surgical interventions to correct congenital heart disease. This work demonstrates how light-scattering spectroscopy (LSS) can be used to non-destructively characterize cardiac tissue regions. Aim To present an approach for associating tissue composition information with location-specific LSS data and further evaluate an LSS and machine learning system as a method for non-destructive tissue characterization. Approach A custom LSS probe was used to gather spectral data from locations across 14 excised human pediatric nodal tissue samples (8 sinus nodes, 6 atrioventricular nodes). The LSS spectra were used to train linear and neural-network-based regressor models to predict tissue composition characteristics derived from the 3D models. Results Nodal tissue region nuclear densities were reported. A linear model trained to regress nuclear density from spectra achieved a prediction r-squared of 0.64 and a concordance correlation coefficient of 0.78. Conclusions These methods build on previous studies suggesting that LSS measurements combined with machine learning signal processing can provide clinically relevant cardiac tissue composition.
Collapse
Affiliation(s)
- Brian Cottle
- University of Utah, Department of Biomedical Engineering, Salt Lake City, Utah, United States
| | - Sarthak Tiwari
- University of Utah, Department of Biomedical Engineering, Salt Lake City, Utah, United States
| | - Aditya Kaza
- Boston Children’s Hospital, Boston, Massachusetts, United States
| | - Frank B. Sachse
- University of Utah, Department of Biomedical Engineering, Salt Lake City, Utah, United States
| | - Robert Hitchcock
- University of Utah, Department of Biomedical Engineering, Salt Lake City, Utah, United States
| |
Collapse
|
15
|
Zhu C, Yuan T, Krishnan J. Targeting cardiomyocyte cell cycle regulation in heart failure. Basic Res Cardiol 2024; 119:349-369. [PMID: 38683371 PMCID: PMC11142990 DOI: 10.1007/s00395-024-01049-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 03/11/2024] [Accepted: 03/29/2024] [Indexed: 05/01/2024]
Abstract
Heart failure continues to be a significant global health concern, causing substantial morbidity and mortality. The limited ability of the adult heart to regenerate has posed challenges in finding effective treatments for cardiac pathologies. While various medications and surgical interventions have been used to improve cardiac function, they are not able to address the extensive loss of functioning cardiomyocytes that occurs during cardiac injury. As a result, there is growing interest in understanding how the cell cycle is regulated and exploring the potential for stimulating cardiomyocyte proliferation as a means of promoting heart regeneration. This review aims to provide an overview of current knowledge on cell cycle regulation and mechanisms underlying cardiomyocyte proliferation in cases of heart failure, while also highlighting established and novel therapeutic strategies targeting this area for treatment purposes.
Collapse
Affiliation(s)
- Chaonan Zhu
- Department of Medicine III, Cardiology/Angiology/Nephrology, Goethe University Hospital, 60590, Frankfurt am Main, Germany
- Institute for Cardiovascular Regeneration, Goethe University, 60590, Frankfurt am Main, Germany
| | - Ting Yuan
- Department of Medicine III, Cardiology/Angiology/Nephrology, Goethe University Hospital, 60590, Frankfurt am Main, Germany.
- Institute for Cardiovascular Regeneration, Goethe University, 60590, Frankfurt am Main, Germany.
- German Center for Cardiovascular Research, Partner Site Rhein-Main, 60590, Frankfurt am Main, Germany.
- Cardio-Pulmonary Institute, Goethe University Hospital, 60590, Frankfurt am Main, Germany.
| | - Jaya Krishnan
- Department of Medicine III, Cardiology/Angiology/Nephrology, Goethe University Hospital, 60590, Frankfurt am Main, Germany.
- Institute for Cardiovascular Regeneration, Goethe University, 60590, Frankfurt am Main, Germany.
- German Center for Cardiovascular Research, Partner Site Rhein-Main, 60590, Frankfurt am Main, Germany.
- Cardio-Pulmonary Institute, Goethe University Hospital, 60590, Frankfurt am Main, Germany.
| |
Collapse
|
16
|
Yang Y, Ma D, Liu B, Sun X, Fu W, Lv F, Qiu C. E3 Ubiquitin Ligase ASB14 Inhibits Cardiomyocyte Proliferation by Regulating MAPRE2 Ubiquitination. Cell Biochem Biophys 2024; 82:715-727. [PMID: 38319584 DOI: 10.1007/s12013-024-01223-x] [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: 11/30/2023] [Accepted: 01/19/2024] [Indexed: 02/07/2024]
Abstract
The ubiquitin proteasome system is a highly specific and selective protein regulatory system that plays an essential role in the regulation of the cell cycle. Despite its significance, the role of ubiquitination in cardiomyocyte proliferation remains largely unclear. This study aimed to investigate the potential impact of E3 ubiquitin ligase ASB14 (Ankyrin Repeat And SOCS Box Containing 14) on cardiac regeneration. We conducted a microarray analysis of apical resection ventricle tissues, and our findings revealed that ASB14 was down-regulated during the cardiac regenerative response. Subsequently, we examined the effect of ASB14 silencing on cardiomyocyte nuclear proliferation both in vitro and in vivo. Our results indicated that ASB14 silencing promoted cardiomyocyte nuclear proliferation, suggesting that ASB14 may play a role in regulating cardiac regeneration. To further investigate the potential therapeutic implications of ASB14 deficiency, we examined the cardiac function of mice with ASB14 deficiency in response to ischemic injury. Our findings showed that mice with ASB14 deficiency exhibited preserved cardiac function and a therapeutic effect in response to ischemic injury, which was attributed to the enhancement of cardiomyocyte nuclear proliferation. To elucidate the underlying mechanisms, we investigated the effect of ASB14 on microtubule-associated protein RP/EB family member 2 (MAPRE2) protein degradation. Our results indicated that the loss of ASB14 decreased the degradation of MAPRE2 protein, subsequently promoting cardiomyocyte nuclear proliferation and enhancing cardiac repair after myocardial infarction (MI). In conclusion, our study provides evidence that inhibition of ASB14-mediated MAPRE2 ubiquitination promotes cardiomyocyte nuclear proliferation, which may serve as a potential target for treating heart failure induced by MI injury.
Collapse
Affiliation(s)
- Yanpeng Yang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dongpu Ma
- Cardiac Care Unit, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Bo Liu
- Cardiac Care Unit, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Xu Sun
- Department of Integrated Chinese and Western Medicine, Henan Cancer Hospital and Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Fu
- Tuberculosis Department No. 1 Ward, Henan Provincial Chest Hospital, Zhengzhou University, Zhengzhou, China
| | - Feifei Lv
- Cardiac Care Unit, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Chunguang Qiu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
17
|
Yang X, Li L, Zeng C, Wang WE. The characteristics of proliferative cardiomyocytes in mammals. J Mol Cell Cardiol 2023; 185:50-64. [PMID: 37918322 DOI: 10.1016/j.yjmcc.2023.10.010] [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: 12/31/2022] [Revised: 10/03/2023] [Accepted: 10/16/2023] [Indexed: 11/04/2023]
Abstract
Better understanding of the mechanisms regulating the proliferation of pre-existing cardiomyocyte (CM) should lead to better options for regenerating injured myocardium. The absence of a perfect research model to definitively identify newly formed mammalian CMs is lacking. However, methodologies are being developed to identify and enrich proliferative CMs. These methods take advantages of the different proliferative states of CMs during postnatal development, before and after injury in the neonatal heart. New approaches use CMs labeled in lineage tracing animals or single cell technique-based CM clusters. This review aims to provide a timely update on the characteristics of the proliferative CMs, including their structural, functional, genetic, epigenetic and metabolic characteristics versus non-proliferative CMs. A better understanding of the characteristics of proliferative CMs should lead to the mechanisms for inducing endogenous CMs to self-renew, which is a promising therapeutic strategy to treat cardiac diseases that cause CM death in humans.
Collapse
Affiliation(s)
- Xinyue Yang
- Department of Geriatrics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Liangpeng Li
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| | - Wei Eric Wang
- Department of Geriatrics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
18
|
Zhao X, Li D, Song Y, Xu J, Xiang FL. Drug Discovery for Adult Cardiomyocyte Regeneration: Opportunities and Challenges. Antioxid Redox Signal 2023; 39:1070-1087. [PMID: 37166381 DOI: 10.1089/ars.2023.0319] [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] [Indexed: 05/12/2023]
Abstract
Significance: Cardiovascular disease is a major contributor to human mortality and morbidity. The cardiac tissue undergoes fibrotic healing after injury because of the limited regenerative capacity of adult mammalian cardiomyocyte (CM). Extensive research has been performed to identify therapeutic targets for CM regeneration, as the success of promoting adult human CM regeneration to repair the injured heart is considered the Holy Grail in the field. Recent Advances: To date, more than 30 target genes have been shown to regulate adult mammalian CM proliferation. More than 20 targets have been validated in adult mouse myocardial infarction (MI) model in a therapeutic setting. In this review, the translational efficacy readouts from 17 selected pharmaceutical targets are summarized, among which the Hippo-yes-associated protein (Yap) pathway is the most extensively investigated and fits the criteria for a promising target for pro-CM-regeneration therapy development. Critical Issues and Future Directions: As the pro-CM-regeneration potential of current drug treatment for cardiovascular patients is limited, to help identify and fill the gap between basic research and drug discovery in this specific field, details regarding target identification, validation in mouse MI models, high-throughput screening assay development, and preclinical in vivo efficacy model optimization are discussed. Finally, suggestions and recommendations are also provided to help establish a common guideline for in vivo translational studies for drug discovery focusing on CM regeneration. Antioxid. Redox Signal. 39, 1070-1087.
Collapse
Affiliation(s)
- Xu Zhao
- Department of Anesthesiology and the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Donghua Li
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yiyan Song
- Department of Anesthesiology and the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jie Xu
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Fu-Li Xiang
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
19
|
Park S, Huang H, Ross I, Moreno J, Khyeam S, Simmons J, Huang GN, Payumo AY. Quantitative Three-dimensional Label-free Digital Holographic Imaging of Cardiomyocyte Size, Ploidy, and Cell Division. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.02.565407. [PMID: 37961676 PMCID: PMC10635088 DOI: 10.1101/2023.11.02.565407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Cardiac regeneration in newborn rodents depends on the ability of pre-existing cardiomyocytes to proliferate and divide. This capacity is lost within the first week of postnatal development when these cells rapidly switch from hyperplasia to hypertrophy, withdraw from the cell cycle, become binucleated, and increase in size. How these dynamic changes in size and ploidy impact cardiomyocyte proliferative potential is not well understood. In this study, we innovate the application of a commercially available digital holographic imaging microscope, the Holomonitor M4, to evaluate the proliferative responses of mononucleated diploid and binucleated tetraploid cardiomyocytes. This instrument coupled with the powerful Holomonitor App Suite software enables long-term label-free quantitative three-dimensional tracking of primary cardiomyocyte dynamics in real-time with single-cell resolution. Our digital holographic imaging results provide direct evidence that mononucleated cardiomyocytes retain significant proliferative potential as most can successfully divide with high frequency. In contrast, binucleated cardiomyocytes exhibit a blunted response to a proliferative stimulus with the majority not attempting to divide at all. Nevertheless, some binucleated cardiomyocytes were capable of complete division, suggesting that these cells still do retain limited proliferative capacity. By quantitatively tracking cardiomyocyte volume dynamics during these proliferative responses, we reveal that both mononucleated and binucleated cells reach a unique size threshold prior to attempted cell division. The absolute threshold is increased by binucleation, which may limit the ability of binucleated cardiomyocytes to divide. By defining the interrelationship between cardiomyocyte size, ploidy, and cell cycle control, we will better understand the cellular mechanisms that drive the loss of mammalian cardiac regenerative capacity after birth.
Collapse
|
20
|
Purdy AL, Swift SK, Sucov HM, Patterson M. Tnni3k influences cardiomyocyte S-phase activity and proliferation. J Mol Cell Cardiol 2023; 183:22-26. [PMID: 37597489 PMCID: PMC11645536 DOI: 10.1016/j.yjmcc.2023.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/25/2023] [Accepted: 08/14/2023] [Indexed: 08/21/2023]
Abstract
Cardiomyocyte proliferation is a difficult phenomenon to capture and prove. Here we employ a retrospective analysis of single cell ventricular suspensions to definitively identify cardiomyocytes that have completed cell division. Through this analysis we determined that the capacity of cardiomyocytes to re-enter the cell cycle and complete cell division after injury are separate and variable traits. Further, we provide evidence that Tnni3k definitively influences both early and final stages of the cell cycle.
Collapse
Affiliation(s)
- Alexandra L Purdy
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Samantha K Swift
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Henry M Sucov
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States of America
| | - Michaela Patterson
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States of America; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States of America.
| |
Collapse
|
21
|
Derks W, Rode J, Collin S, Rost F, Heinke P, Hariharan A, Pickel L, Simonova I, Lázár E, Graham E, Jashari R, Andrä M, Jeppsson A, Salehpour M, Alkass K, Druid H, Kyriakopoulos CP, Taleb I, Shankar TS, Selzman CH, Sadek H, Jovinge S, Brusch L, Frisén J, Drakos S, Bergmann O. A latent cardiomyocyte regeneration potential in human heart disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.14.557681. [PMID: 37745322 PMCID: PMC10515906 DOI: 10.1101/2023.09.14.557681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Cardiomyocytes in the adult human heart show a regenerative capacity, with an annual renewal rate around 0.5%. Whether this regenerative capacity of human cardiomyocytes is employed in heart failure has been controversial. Using retrospective 14C birth dating we analyzed cardiomyocyte renewal in patients with end-stage heart failure. We show that cardiomyocyte generation is minimal in end-stage heart failure patients at rates 18-50 times lower compared to the healthy heart. However, patients receiving left ventricle support device therapy, who showed significant functional and structural cardiac improvement, had a >6-fold increase in cardiomyocyte renewal relative to the healthy heart. Our findings reveal a substantial cardiomyocyte regeneration potential in human heart disease, which could be exploited therapeutically.
Collapse
Affiliation(s)
- Wouter Derks
- Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
| | - Julian Rode
- Center of Information Services and High-Performance Computing, TU Dresden, Dresden, Germany
| | - Sofia Collin
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Stockholm, Sweden
| | - Fabian Rost
- Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
- Center of Information Services and High-Performance Computing, TU Dresden, Dresden, Germany
- DRESDEN-concept Genome Center, Technology Platform at the Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
| | - Paula Heinke
- Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
| | - Anjana Hariharan
- Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
| | - Lauren Pickel
- Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
| | - Irina Simonova
- Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
| | - Enikő Lázár
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Stockholm, Sweden
| | - Evan Graham
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Stockholm, Sweden
| | | | - Michaela Andrä
- Department of Cardiothoracic and Vascular Surgery, Klinikum Klagenfurt and Section for Surgical Research Medical University Graz, 9020 Graz, Austria
| | - Anders Jeppsson
- Department of Cardiothoracic Surgery, Sahlgrenska University Hospital, SE-413 45 Gothenburg, Sweden
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Mehran Salehpour
- Department of Physics and Astronomy, Applied Nuclear Physics, Uppsala University, SE-751 20 Uppsala, Sweden
| | - Kanar Alkass
- Department of Oncology-Pathology, Karolinska Institute, SE-171 77 Stockholm and National Board of Forensic Medicine, SE-171 65 Stockholm, Sweden
| | - Henrik Druid
- Department of Oncology-Pathology, Karolinska Institute, SE-171 77 Stockholm and National Board of Forensic Medicine, SE-171 65 Stockholm, Sweden
| | - Christos P. Kyriakopoulos
- Divisions of Cardiovascular Medicine and Cardiothoracic Surgery, University of Utah Health & School of Medicine, Salt Lake City, Utah, United States
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States
| | - Iosif Taleb
- Divisions of Cardiovascular Medicine and Cardiothoracic Surgery, University of Utah Health & School of Medicine, Salt Lake City, Utah, United States
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States
| | - Thirupura S. Shankar
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States
| | - Craig H. Selzman
- Divisions of Cardiovascular Medicine and Cardiothoracic Surgery, University of Utah Health & School of Medicine, Salt Lake City, Utah, United States
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States
| | - Hesham Sadek
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Stefan Jovinge
- Spectrum Health Frederik Meijer Heart & Vascular Institute and Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Lutz Brusch
- Center of Information Services and High-Performance Computing, TU Dresden, Dresden, Germany
| | - Jonas Frisén
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Stockholm, Sweden
| | - Stavros Drakos
- Divisions of Cardiovascular Medicine and Cardiothoracic Surgery, University of Utah Health & School of Medicine, Salt Lake City, Utah, United States
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States
| | - Olaf Bergmann
- Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Stockholm, Sweden
- Pharmacology and Toxicology, Department of Pharmacology and Toxicology University Medical Center Goettingen, Goettingen, Germany
| |
Collapse
|
22
|
Luo J, Farris SD, Helterline D, Stempien-Otero A. Cardiomyocyte nuclear remodeling after mechanical unloading. Am J Physiol Heart Circ Physiol 2023; 325:H244-H251. [PMID: 37204870 PMCID: PMC10393327 DOI: 10.1152/ajpheart.00545.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 04/07/2023] [Accepted: 04/10/2023] [Indexed: 05/21/2023]
Abstract
Cardiomyocytes increase DNA content in response to stress in humans. DNA content is reported to decrease in association with increased markers of proliferation in cardiomyocytes following left ventricular assist device (LVAD) unloading. However, cardiac recovery resulting in LVAD explant is rare. Thus, we sought to test the hypothesis that changes in DNA content with mechanical unloading occurs independent of cardiomyocyte proliferation by quantifying cardiomyocyte nuclear number, cell size, DNA content, and the frequency of cell-cycling markers using a novel imaging flow cytometry methodology comparing human subjects undergoing LVAD implantation or primary transplantation. We found that cardiomyocyte size was 15% smaller in unloaded versus loaded samples without differences in the percentage of mono-, bi-, or multinuclear cells. DNA content per nucleus was significantly decreased in unloaded hearts versus loaded controls. Cell-cycle markers, Ki67 and phospho-histon3 (H3P), were not increased in unloaded samples. In conclusion, unloading of failing hearts is associated with decreased DNA content of nuclei independent of nucleation state within the cell. As these changes were associated with a trend to decreased cell size but not increased cell-cycle markers, they may represent a regression of hypertrophic nuclear remodeling and not proliferation.NEW & NOTEWORTHY Our data suggest that increases in DNA content that occur with cardiomyocyte hypertrophy in heart failure may reverse with mechanical unloading.
Collapse
Affiliation(s)
- Jun Luo
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States
| | - Stephen D Farris
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States
| | - Deri Helterline
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States
| | - April Stempien-Otero
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington, United States
| |
Collapse
|
23
|
Bazgir F, Nau J, Nakhaei-Rad S, Amin E, Wolf MJ, Saucerman JJ, Lorenz K, Ahmadian MR. The Microenvironment of the Pathogenesis of Cardiac Hypertrophy. Cells 2023; 12:1780. [PMID: 37443814 PMCID: PMC10341218 DOI: 10.3390/cells12131780] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/22/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
Pathological cardiac hypertrophy is a key risk factor for the development of heart failure and predisposes individuals to cardiac arrhythmia and sudden death. While physiological cardiac hypertrophy is adaptive, hypertrophy resulting from conditions comprising hypertension, aortic stenosis, or genetic mutations, such as hypertrophic cardiomyopathy, is maladaptive. Here, we highlight the essential role and reciprocal interactions involving both cardiomyocytes and non-myocardial cells in response to pathological conditions. Prolonged cardiovascular stress causes cardiomyocytes and non-myocardial cells to enter an activated state releasing numerous pro-hypertrophic, pro-fibrotic, and pro-inflammatory mediators such as vasoactive hormones, growth factors, and cytokines, i.e., commencing signaling events that collectively cause cardiac hypertrophy. Fibrotic remodeling is mediated by cardiac fibroblasts as the central players, but also endothelial cells and resident and infiltrating immune cells enhance these processes. Many of these hypertrophic mediators are now being integrated into computational models that provide system-level insights and will help to translate our knowledge into new pharmacological targets. This perspective article summarizes the last decades' advances in cardiac hypertrophy research and discusses the herein-involved complex myocardial microenvironment and signaling components.
Collapse
Affiliation(s)
- Farhad Bazgir
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (F.B.); (J.N.)
| | - Julia Nau
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (F.B.); (J.N.)
| | - Saeideh Nakhaei-Rad
- Stem Cell Biology, and Regenerative Medicine Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad 91779-48974, Iran;
| | - Ehsan Amin
- Institute of Neural and Sensory Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| | - Matthew J. Wolf
- Department of Medicine and Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA;
| | - Jeffry J. Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA;
| | - Kristina Lorenz
- Institute of Pharmacology and Toxicology, University of Würzburg, Leibniz Institute for Analytical Sciences, 97078 Würzburg, Germany;
| | - Mohammad Reza Ahmadian
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (F.B.); (J.N.)
| |
Collapse
|
24
|
Ostadal B, Kolar F, Ostadalova I, Sedmera D, Olejnickova V, Hlavackova M, Alanova P. Developmental Aspects of Cardiac Adaptation to Increased Workload. J Cardiovasc Dev Dis 2023; 10:jcdd10050205. [PMID: 37233172 DOI: 10.3390/jcdd10050205] [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: 04/05/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 05/27/2023] Open
Abstract
The heart is capable of extensive adaptive growth in response to the demands of the body. When the heart is confronted with an increased workload over a prolonged period, it tends to cope with the situation by increasing its muscle mass. The adaptive growth response of the cardiac muscle changes significantly during phylogenetic and ontogenetic development. Cold-blooded animals maintain the ability for cardiomyocyte proliferation even in adults. On the other hand, the extent of proliferation during ontogenetic development in warm-blooded species shows significant temporal limitations: whereas fetal and neonatal cardiac myocytes express proliferative potential (hyperplasia), after birth proliferation declines and the heart grows almost exclusively by hypertrophy. It is, therefore, understandable that the regulation of the cardiac growth response to the increased workload also differs significantly during development. The pressure overload (aortic constriction) induced in animals before the switch from hyperplastic to hypertrophic growth leads to a specific type of left ventricular hypertrophy which, in contrast with the same stimulus applied in adulthood, is characterized by hyperplasia of cardiomyocytes, capillary angiogenesis and biogenesis of collagenous structures, proportional to the growth of myocytes. These studies suggest that timing may be of crucial importance in neonatal cardiac interventions in humans: early definitive repairs of selected congenital heart disease may be more beneficial for the long-term results of surgical treatment.
Collapse
Affiliation(s)
- Bohuslav Ostadal
- Institute of Physiology of the Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Frantisek Kolar
- Institute of Physiology of the Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Ivana Ostadalova
- Institute of Physiology of the Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - David Sedmera
- Institute of Physiology of the Czech Academy of Sciences, 142 20 Prague, Czech Republic
- Institute of Anatomy, First Faculty of Medicine, Charles University, 128 00 Prague, Czech Republic
| | - Veronika Olejnickova
- Institute of Physiology of the Czech Academy of Sciences, 142 20 Prague, Czech Republic
- Institute of Anatomy, First Faculty of Medicine, Charles University, 128 00 Prague, Czech Republic
| | - Marketa Hlavackova
- Institute of Physiology of the Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Petra Alanova
- Institute of Physiology of the Czech Academy of Sciences, 142 20 Prague, Czech Republic
| |
Collapse
|
25
|
Anatskaya OV, Runov AL, Ponomartsev SV, Vonsky MS, Elmuratov AU, Vinogradov AE. Long-Term Transcriptomic Changes and Cardiomyocyte Hyperpolyploidy after Lactose Intolerance in Neonatal Rats. Int J Mol Sci 2023; 24:7063. [PMID: 37108224 PMCID: PMC10138443 DOI: 10.3390/ijms24087063] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/02/2023] [Accepted: 04/08/2023] [Indexed: 04/29/2023] Open
Abstract
Many cardiovascular diseases originate from growth retardation, inflammation, and malnutrition during early postnatal development. The nature of this phenomenon is not completely understood. Here we aimed to verify the hypothesis that systemic inflammation triggered by neonatal lactose intolerance (NLI) may exert long-term pathologic effects on cardiac developmental programs and cardiomyocyte transcriptome regulation. Using the rat model of NLI triggered by lactase overloading with lactose and the methods of cytophotometry, image analysis, and mRNA-seq, we evaluated cardiomyocyte ploidy, signs of DNA damage, and NLI-associated long-term transcriptomic changes of genes and gene modules that differed qualitatively (i.e., were switched on or switched off) in the experiment vs. the control. Our data indicated that NLI triggers the long-term animal growth retardation, cardiomyocyte hyperpolyploidy, and extensive transcriptomic rearrangements. Many of these rearrangements are known as manifestations of heart pathologies, including DNA and telomere instability, inflammation, fibrosis, and reactivation of fetal gene program. Moreover, bioinformatic analysis identified possible causes of these pathologic traits, including the impaired signaling via thyroid hormone, calcium, and glutathione. We also found transcriptomic manifestations of increased cardiomyocyte polyploidy, such as the induction of gene modules related to open chromatin, e.g., "negative regulation of chromosome organization", "transcription" and "ribosome biogenesis". These findings suggest that ploidy-related epigenetic alterations acquired in the neonatal period permanently rewire gene regulatory networks and alter cardiomyocyte transcriptome. Here we provided first evidence indicating that NLI can be an important trigger of developmental programming of adult cardiovascular disease. The obtained results can help to develop preventive strategies for reducing the NLI-associated adverse effects of inflammation on the developing cardiovascular system.
Collapse
Affiliation(s)
| | - Andrey L. Runov
- The D.I. Mendeleev All-Russian Institute for Metrology (VNIIM), Moskovsky ave 19, Saint Petersburg 190005, Russia
- Almazov Medical Research Centre, Akkuratova Street 2, Saint Petersburg 197341, Russia
| | | | - Maxim S. Vonsky
- The D.I. Mendeleev All-Russian Institute for Metrology (VNIIM), Moskovsky ave 19, Saint Petersburg 190005, Russia
- Almazov Medical Research Centre, Akkuratova Street 2, Saint Petersburg 197341, Russia
| | - Artem U. Elmuratov
- Medical Genetics Centre Genotek, Nastavnichesky Alley 17-1-15, Moscow 105120, Russia
| | | |
Collapse
|
26
|
Watanabe H, Tao G, Gan P, Westbury BC, Cox KD, Tjen K, Song R, Fishman GI, Makita T, Sucov HM. Purkinje Cardiomyocytes of the Adult Ventricular Conduction System Are Highly Diploid but Not Uniquely Regenerative. J Cardiovasc Dev Dis 2023; 10:161. [PMID: 37103040 PMCID: PMC10140853 DOI: 10.3390/jcdd10040161] [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: 03/02/2023] [Revised: 03/31/2023] [Accepted: 04/05/2023] [Indexed: 04/28/2023] Open
Abstract
Adult hearts are characterized by inefficient regeneration after injury, thus, the features that support or prevent cardiomyocyte (CM) proliferation are important to clarify. Diploid CMs are a candidate cell type that may have unique proliferative and regenerative competence, but no molecular markers are yet known that selectively identify all or subpopulations of diploid CMs. Here, using the conduction system expression marker Cntn2-GFP and the conduction system lineage marker Etv1CreERT2, we demonstrate that Purkinje CMs that comprise the adult ventricular conduction system are disproportionately diploid (33%, vs. 4% of bulk ventricular CMs). These, however, represent only a small proportion (3%) of the total diploid CM population. Using EdU incorporation during the first postnatal week, we demonstrate that bulk diploid CMs found in the later heart enter and complete the cell cycle during the neonatal period. In contrast, a significant fraction of conduction CMs persist as diploid cells from fetal life and avoid neonatal cell cycle activity. Despite their high degree of diploidy, the Purkinje lineage had no enhanced competence to support regeneration after adult heart infarction.
Collapse
Affiliation(s)
- Hirofumi Watanabe
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ge Tao
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Peiheng Gan
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Baylee C. Westbury
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Kristie D. Cox
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Kelsey Tjen
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ruolan Song
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Glenn I. Fishman
- Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Takako Makita
- Darby Children’s Research Institute, Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Henry M. Sucov
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
27
|
Swift SK, Purdy AL, Kolell ME, Andresen KG, Lahue C, Buddell T, Akins KA, Rau CD, O'Meara CC, Patterson M. Cardiomyocyte ploidy is dynamic during postnatal development and varies across genetic backgrounds. Development 2023; 150:dev201318. [PMID: 36912240 PMCID: PMC10113957 DOI: 10.1242/dev.201318] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 03/06/2023] [Indexed: 03/14/2023]
Abstract
Somatic polyploidization, an adaptation by which cells increase their DNA content to support growth, is observed in many cell types, including cardiomyocytes. Although polyploidization is believed to be beneficial, progression to a polyploid state is often accompanied by loss of proliferative capacity. Recent work suggests that genetics heavily influence cardiomyocyte ploidy. However, the developmental course by which cardiomyocytes reach their final ploidy state has only been investigated in select backgrounds. Here, we assessed cardiomyocyte number, cell cycle activity, and ploidy dynamics across two divergent mouse strains: C57BL/6J and A/J. Both strains are born and reach adulthood with comparable numbers of cardiomyocytes; however, the end composition of ploidy classes and developmental progression to reach the final state differ substantially. We expand on previous findings that identified Tnni3k as a mediator of cardiomyocyte ploidy and uncover a role for Runx1 in ploidy dynamics and cardiomyocyte cell division, in both developmental and injury contexts. These data provide novel insights into the developmental path to cardiomyocyte polyploidization and challenge the paradigm that hypertrophy is the sole mechanism for growth in the postnatal heart.
Collapse
Affiliation(s)
- Samantha K Swift
- Medical College of Wisconsin, Department of Cell Biology, Neurobiology, and Anatomy, Milwaukee, WI 53226, USA
| | - Alexandra L Purdy
- Medical College of Wisconsin, Department of Cell Biology, Neurobiology, and Anatomy, Milwaukee, WI 53226, USA
| | - Mary E Kolell
- Medical College of Wisconsin, Department of Cell Biology, Neurobiology, and Anatomy, Milwaukee, WI 53226, USA
| | - Kaitlyn G Andresen
- Medical College of Wisconsin, Department of Cell Biology, Neurobiology, and Anatomy, Milwaukee, WI 53226, USA
| | - Caitlin Lahue
- University of North Carolina School of Medicine, Department of Genetics, Chapel Hill, NC 27599, USA
| | - Tyler Buddell
- Medical College of Wisconsin, Department of Cell Biology, Neurobiology, and Anatomy, Milwaukee, WI 53226, USA
- Medical College of Wisconsin, Cardiovascular Center, Milwaukee, WI 53226, USA
| | - Kaelin A Akins
- Medical College of Wisconsin, Department of Cell Biology, Neurobiology, and Anatomy, Milwaukee, WI 53226, USA
| | - Christoph D Rau
- University of North Carolina School of Medicine, Department of Genetics, Chapel Hill, NC 27599, USA
| | - Caitlin C O'Meara
- Medical College of Wisconsin, Cardiovascular Center, Milwaukee, WI 53226, USA
- Medical College of Wisconsin, Department of Physiology, Milwaukee, WI 53226, USA
| | - Michaela Patterson
- Medical College of Wisconsin, Department of Cell Biology, Neurobiology, and Anatomy, Milwaukee, WI 53226, USA
- Medical College of Wisconsin, Cardiovascular Center, Milwaukee, WI 53226, USA
| |
Collapse
|
28
|
Gene Therapy for Cardiomyocyte Renewal: Cell Cycle, a Potential Therapeutic Target. Mol Diagn Ther 2023; 27:129-140. [PMID: 36512179 PMCID: PMC10123801 DOI: 10.1007/s40291-022-00625-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2022] [Indexed: 12/14/2022]
Abstract
Heart disease is the primary cause of death worldwide. Even though extensive research has been done, and many pharmacological and surgical treatments have been introduced to treat heart disease, the mortality rate still remains high. Gene therapy is widely used to understand molecular mechanisms of myocardial infarction and to treat cardiomyocyte loss. It was reported that adult cardiomyocytes proliferate at a very low rate; thus, targeting their proliferation has become a new regenerative therapeutic approach. Currently, re-activating cardiomyocyte proliferation appears to be one of the most promising methods to promote adult cardiomyocyte renewal. In this article, we highlight gene therapeutic targets of cell proliferation presently being pursued to re-activate the cell cycle of cardiomyocytes, including cell cycle regulators, transcription factors, microRNAs, signal transduction, and other contributing factors. We also summarize gene delivery vectors that have been used in cardiac research and major challenges to be overcome in the translation to the clinical approach and future directions.
Collapse
|
29
|
Lindsey ML. Getting it Right. Am J Physiol Heart Circ Physiol 2022; 323:H421-H423. [PMID: 35930444 DOI: 10.1152/ajpheart.00400.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Merry L Lindsey
- School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, United States
| |
Collapse
|
30
|
Wilson AJ, Sands GB, LeGrice IJ, Young AA, Ennis DB. Myocardial mesostructure and mesofunction. Am J Physiol Heart Circ Physiol 2022; 323:H257-H275. [PMID: 35657613 PMCID: PMC9273275 DOI: 10.1152/ajpheart.00059.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/23/2022] [Accepted: 05/23/2022] [Indexed: 11/22/2022]
Abstract
The complex and highly organized structural arrangement of some five billion cardiomyocytes directs the coordinated electrical activity and mechanical contraction of the human heart. The characteristic transmural change in cardiomyocyte orientation underlies base-to-apex shortening, circumferential shortening, and left ventricular torsion during contraction. Individual cardiomyocytes shorten ∼15% and increase in diameter ∼8%. Remarkably, however, the left ventricular wall thickens by up to 30-40%. To accommodate this, the myocardium must undergo significant structural rearrangement during contraction. At the mesoscale, collections of cardiomyocytes are organized into sheetlets, and sheetlet shear is the fundamental mechanism of rearrangement that produces wall thickening. Herein, we review the histological and physiological studies of myocardial mesostructure that have established the sheetlet shear model of wall thickening. Recent developments in tissue clearing techniques allow for imaging of whole hearts at the cellular scale, whereas magnetic resonance imaging (MRI) and computed tomography (CT) can image the myocardium at the mesoscale (100 µm to 1 mm) to resolve cardiomyocyte orientation and organization. Through histology, cardiac diffusion tensor imaging (DTI), and other modalities, mesostructural sheetlets have been confirmed in both animal and human hearts. Recent in vivo cardiac DTI methods have measured reorientation of sheetlets during the cardiac cycle. We also examine the role of pathological cardiac remodeling on sheetlet organization and reorientation, and the impact this has on ventricular function and dysfunction. We also review the unresolved mesostructural questions and challenges that may direct future work in the field.
Collapse
Affiliation(s)
- Alexander J Wilson
- Department of Radiology, Stanford University, Stanford, California
- Stanford Cardiovascular Institute, Stanford University, Stanford, California
| | - Gregory B Sands
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Ian J LeGrice
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
- Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Alistair A Young
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
- Department of Biomedical Engineering, King's College London, London, United Kingdom
| | - Daniel B Ennis
- Department of Radiology, Stanford University, Stanford, California
- Veterans Administration Palo Alto Health Care System, Palo Alto, California
| |
Collapse
|
31
|
Watanabe H, Sucov HM. Cardiomyocyte proliferation by calcineurin inhibition. NATURE CARDIOVASCULAR RESEARCH 2022; 1:599-600. [PMID: 39196241 DOI: 10.1038/s44161-022-00100-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Affiliation(s)
- Hirofumi Watanabe
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Henry M Sucov
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA.
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
32
|
Velayutham N, Yutzey KE. Porcine Models of Heart Regeneration. J Cardiovasc Dev Dis 2022; 9:jcdd9040093. [PMID: 35448069 PMCID: PMC9025077 DOI: 10.3390/jcdd9040093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 01/11/2023] Open
Abstract
Swine are popular large mammals for cardiac preclinical testing due to their similarities with humans in terms of organ size and physiology. Recent studies indicate an early neonatal regenerative capacity for swine hearts similar to small mammal laboratory models such as rodents, inspiring exciting possibilities for studying cardiac regeneration with the goal of improved clinical translation to humans. However, while swine hearts are anatomically similar to humans, fundamental differences exist in growth mechanisms, nucleation, and the maturation of pig cardiomyocytes, which could present difficulties for the translation of preclinical findings in swine to human therapeutics. In this review, we discuss the maturational dynamics of pig cardiomyocytes and their capacity for proliferative cardiac regeneration during early neonatal development to provide a perspective on swine as a preclinical model for developing cardiac gene- and cell-based regenerative therapeutics.
Collapse
Affiliation(s)
- Nivedhitha Velayutham
- The Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA;
- Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Katherine E. Yutzey
- The Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA;
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Correspondence:
| |
Collapse
|