Journal articles on the topic 'Cardiosphere-derived cell'

To see the other types of publications on this topic, follow the link: Cardiosphere-derived cell.

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the top 50 journal articles for your research on the topic 'Cardiosphere-derived cell.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Browse journal articles on a wide variety of disciplines and organise your bibliography correctly.

1

Emani, Sitaram M., and Pedro J. del Nido. "Cell-Based Therapy With Cardiosphere-Derived Cardiocytes." Circulation Research 122, no. 7 (March 30, 2018): 916–17. http://dx.doi.org/10.1161/circresaha.118.312809.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Dergilev, K. V., Z. I. Tsokolaeva, Yu D. Vasilets, I. B. Beloglazova, and E. V. Parfenova. "Cardiac progenitor cell sheets secrete proangiogenic growth factors and locally activate capillarogenesis after infarction." Complex Issues of Cardiovascular Diseases 10, no. 3 (September 25, 2021): 34–43. http://dx.doi.org/10.17802/2306-1278-2021-10-3-34-43.

Full text
Abstract:
Background. The application of tissue-engineered constructs that simulate the natural microenvironment of cells, maintain their viability and functional properties, is a new promising route for the treatment of ischemic diseases. However, the mechanisms that ensure the effectiveness of this type of treatment and the principles of choosing the optimal population of progenitor cells remain poorly understood. Aim. To study the profile of secretion of proangiogenic growth factors of cardiosphere-derived cell sheet (CS), and to study the effect of their transplantation on postinfarction myocardial vascularization. Methods. Assembly of cardiosphere-derived cell sheets were performed on thermosensitive culture plates. Characterization of cell sheets was performed using immunofluorescence staining and a commercial kit for the determination of proangiogenic factors “Mouse Angiogenesis Antibody Array”. The evaluation of the angiogenic properties of the cell graft in vivo was carried out using a rat myocardial infarction model. Results. It was found that the cardiosphere-derived cell sheet secrete factors involved in the regulation of vasculo-/angiogenesis. At the same time, the cultivation of cell sheets under hypoxic conditions (3% O2) led to an increase in the secretion of proangigenic factors VEGF and pIgF, fGf-1, FGF-2, endothelin-1, as well as MMP-9, which is involved in extracellular matrix remodeling. Cell sheet transplantation on the epicardial surface of the heart after myocardial infarction ensures cell viability and local increase in capillarization of the damaged area. Conclusion. Thus, the application of cardiosphere-derived cell sheets, which have proangiogenic properties and ability to maintain post transplantation cell survival, can be considered as a promising approach for the development of new methods of therapy for heart diseases
APA, Harvard, Vancouver, ISO, and other styles
3

Pakzad, Khadijeh Kathy, Jun Jie Tan, Stephanie Anderson, Mary Board, Kieran Clarke, and Carolyn A. Carr. "Metabolic maturation of differentiating cardiosphere-derived cells." Stem Cell Research 54 (July 2021): 102422. http://dx.doi.org/10.1016/j.scr.2021.102422.

Full text
APA, Harvard, Vancouver, ISO, and other styles
4

Chen, Lijuan, Muhammad Ashraf, Yingjie Wang, Mi Zhou, John Zhang, Gangjian Qin, Jack Rubinstein, Neal L. Weintraub, and Yaoliang Tang. "The Role ofNotch 1Activation in Cardiosphere Derived Cell Differentiation." Stem Cells and Development 21, no. 12 (August 10, 2012): 2122–29. http://dx.doi.org/10.1089/scd.2011.0463.

Full text
APA, Harvard, Vancouver, ISO, and other styles
5

Xie, Yucai, Ahmed Ibrahim, Ke Cheng, Zhijun Wu, Wenbin Liang, Konstantinos Malliaras, Baiming Sun, et al. "Importance of Cell-Cell Contact in the Therapeutic Benefits of Cardiosphere-Derived Cells." STEM CELLS 32, no. 9 (August 18, 2014): 2397–406. http://dx.doi.org/10.1002/stem.1736.

Full text
APA, Harvard, Vancouver, ISO, and other styles
6

Martens, Andreas, Ina Gruh, Dimitrios Dimitroulis, Sebastian V. Rojas, Ingrid Schmidt-Richter, Christian Rathert, Nawid Khaladj, et al. "Rhesus monkey cardiosphere-derived cells for myocardial restoration." Cytotherapy 13, no. 7 (August 2011): 864–72. http://dx.doi.org/10.3109/14653249.2011.571247.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

Marbán, Eduardo. "Breakthroughs in Cell Therapy for Heart Disease: Focus on Cardiosphere-Derived Cells." Mayo Clinic Proceedings 89, no. 6 (June 2014): 850–58. http://dx.doi.org/10.1016/j.mayocp.2014.02.014.

Full text
APA, Harvard, Vancouver, ISO, and other styles
8

Fujita, Akira, Koji Ueno, Toshiro Saito, Masashi Yanagihara, Hiroshi Kurazumi, Ryo Suzuki, Akihito Mikamo, and Kimikazu Hamano. "Hypoxic-conditioned cardiosphere-derived cell sheet transplantation for chronic myocardial infarction." European Journal of Cardio-Thoracic Surgery 56, no. 6 (April 24, 2019): 1062–74. http://dx.doi.org/10.1093/ejcts/ezz122.

Full text
Abstract:
Abstract OBJECTIVES Cell therapy provides a suitable environment for regeneration through paracrine effects such as secretion of growth factors. Cardiosphere-derived cells (CDCs) have a high capacity for growth factor secretion and are an attractive target for clinical applications. In particular, a cell sheet technique was reported to have clinical advantages by covering a specific region. Here, we examined the effect of the hypoxic-conditioned (HC) autologous CDC sheet therapy on a rabbit chronic myocardial infarction model. METHODS CDC sheet function was assessed by the enzyme-linked immunosorbent assay and quantified by polymerase chain reaction in vitro (days 1–3 of conditioning). The rabbit chronic myocardial infarction model was established by left coronary ligation. Autologous CDCs were isolated from the left atrial specimen; CDC sheets with or without 2-day HC were transplanted onto the infarcted hearts at 4 weeks. The cardiac function was assessed by an echocardiography at 0, 4 and 8 weeks. A histological analysis of the host hearts was performed by tomato lectin staining at 8 weeks. RESULTS The optimal HC duration was 48 h. HC significantly increased the mRNA expression levels of VEGF and ANG2 on day 2 compared to the normoxic-conditioned (NC) group. The HC group showed significant improvement in the left ventricular ejection fraction (64.4% vs 58.8% and 53.4% in the NC and control) and a greater lectin-positive area in the ischaemic region (HC:NC:control = 13:8:2). CONCLUSIONS HC enhances the paracrine effect of a CDC sheet on angiogenesis to improve cardiac function in the chronic myocardial infarction model, which is essential for cardiomyocyte proliferation during cardiac regeneration.
APA, Harvard, Vancouver, ISO, and other styles
9

Bruyneel, Arne, Rabia Nazir, Qi Chen, Colleen Lopez, Jan Czernuszka, and Carolyn Carr. "164 Cardiosphere-Derived Cell-Seeded Porous Collagen Scaffolds for Cardiac Repair." Heart 102, Suppl 6 (June 2016): A116.1—A116. http://dx.doi.org/10.1136/heartjnl-2016-309890.164.

Full text
APA, Harvard, Vancouver, ISO, and other styles
10

Grigorian-Shamagian, Lilian, Weixin Liu, Soraya Fereydooni, Ryan C. Middleton, Jackelyn Valle, Jae Hyung Cho, and Eduardo Marbán. "Cardiac and systemic rejuvenation after cardiosphere-derived cell therapy in senescent rats." European Heart Journal 38, no. 39 (August 14, 2017): 2957–67. http://dx.doi.org/10.1093/eurheartj/ehx454.

Full text
APA, Harvard, Vancouver, ISO, and other styles
11

Puluca, N., S. Doppler, H. Lahm, Z. Zhang, M. Dreßen, M. A. Deutsch, R. Lange, and M. Krane. "Cardiosphere-Derived Cells: A Possible Source for Regenerative Cell Therapy in Congenital Heart Diseases." Thoracic and Cardiovascular Surgeon 65, S 01 (February 3, 2017): S1—S110. http://dx.doi.org/10.1055/s-0037-1598857.

Full text
APA, Harvard, Vancouver, ISO, and other styles
12

Hensley, Michael Taylor, James Andrade, Bruce Keene, Kathryn Meurs, Junnan Tang, Zegen Wang, Thomas G. Caranasos, Jorge Piedrahita, Tao‐Sheng Li, and Ke Cheng. "Cardiac regenerative potential of cardiosphere‐derived cells from adult dog hearts." Journal of Cellular and Molecular Medicine 19, no. 8 (April 9, 2015): 1805–13. http://dx.doi.org/10.1111/jcmm.12585.

Full text
APA, Harvard, Vancouver, ISO, and other styles
13

Tomita, Yuichi, Keisuke Matsumura, Yoshio Wakamatsu, Yumi Matsuzaki, Isao Shibuya, Haruko Kawaguchi, Masaki Ieda, et al. "Cardiac neural crest cells contribute to the dormant multipotent stem cell in the mammalian heart." Journal of Cell Biology 170, no. 7 (September 26, 2005): 1135–46. http://dx.doi.org/10.1083/jcb.200504061.

Full text
Abstract:
Arodent cardiac side population cell fraction formed clonal spheroids in serum-free medium, which expressed nestin, Musashi-1, and multi-drug resistance transporter gene 1, markers of undifferentiated neural precursor cells. These markers were lost following differentiation, and were replaced by the expression of neuron-, glial-, smooth muscle cell–, or cardiomyocyte-specific proteins. Cardiosphere-derived cells transplanted into chick embryos migrated to the truncus arteriosus and cardiac outflow tract and contributed to dorsal root ganglia, spinal nerves, and aortic smooth muscle cells. Lineage studies using double transgenic mice encoding protein 0–Cre/Floxed-EGFP revealed undifferentiated and differentiated neural crest-derived cells in the fetal myocardium. Undifferentiated cells expressed GATA-binding protein 4 and nestin, but not actinin, whereas the differentiated cells were identified as cardiomyocytes. These results suggest that cardiac neural crest-derived cells migrate into the heart, remain there as dormant multipotent stem cells—and under the right conditions—differentiate into cardiomyocytes and typical neural crest-derived cells, including neurons, glia, and smooth muscle.
APA, Harvard, Vancouver, ISO, and other styles
14

Mentkowski, Kyle I., Asma Mursleen, Jonathan D. Snitzer, Lindsey M. Euscher, and Jennifer K. Lang. "CDC-derived extracellular vesicles reprogram inflammatory macrophages to an arginase 1-dependent proangiogenic phenotype." American Journal of Physiology-Heart and Circulatory Physiology 318, no. 6 (June 1, 2020): H1447—H1460. http://dx.doi.org/10.1152/ajpheart.00155.2020.

Full text
Abstract:
We hypothesized that in the window of therapeutic extracellular vesicle (EV) administration, inflammatory M1 macrophages are likely the primary target of cardiosphere-derived cell (CDC)-derived EVs. The effect of CDC-EVs on this population, however, is currently unknown. In this study, we demonstrate that CDC-derived EVs polarize M1 macrophages to a proangiogenic phenotype dependent on arginase 1 upregulation. These results provide insight into an immunomodulatory mechanism of CDC-EVs in a more physiologically relevant model of post-myocardial infarction (post-MI) macrophage polarization.
APA, Harvard, Vancouver, ISO, and other styles
15

Redgrave, R. E., B. Davison, M. Amirrasouli, B. Keavney, A. Blamire, and H. M. Arthur. "CARDIOSPHERE-DERIVED CELL TRANSPLANTATION RESCUES CARDIAC FUNCTION POST-MI INDEPENDENTLY OF ENDOGLIN EXPRESSION." Heart 98, Suppl 5 (November 2012): A1.2—A1. http://dx.doi.org/10.1136/heartjnl-2012-303148a.2.

Full text
APA, Harvard, Vancouver, ISO, and other styles
16

Middleton, Ryan C., Mario Fournier, Xuan Xu, Eduardo Marbán, and Michael I. Lewis. "Therapeutic benefits of intravenous cardiosphere-derived cell therapy in rats with pulmonary hypertension." PLOS ONE 12, no. 8 (August 24, 2017): e0183557. http://dx.doi.org/10.1371/journal.pone.0183557.

Full text
APA, Harvard, Vancouver, ISO, and other styles
17

Pagano, Francesca, Francesco Angelini, Clotilde Castaldo, Vittorio Picchio, Elisa Messina, Sebastiano Sciarretta, Ciro Maiello, et al. "Normal versus Pathological Cardiac Fibroblast-Derived Extracellular Matrix Differentially Modulates Cardiosphere-Derived Cell Paracrine Properties and Commitment." Stem Cells International 2017 (2017): 1–9. http://dx.doi.org/10.1155/2017/7396462.

Full text
Abstract:
Human resident cardiac progenitor cells (CPCs) isolated as cardiosphere-derived cells (CDCs) are under clinical evaluation as a therapeutic product for cardiac regenerative medicine. Unfortunately, limited engraftment and differentiation potential of transplanted cells significantly hamper therapeutic success. Moreover, maladaptive remodelling of the extracellular matrix (ECM) during heart failure progression provides impaired biological and mechanical signals to cardiac cells, including CPCs. In this study, we aimed at investigating the differential effect on the phenotype of human CDCs of cardiac fibroblast-derived ECM substrates from healthy or diseased hearts, named, respectively, normal or pathological cardiogel (CG-N/P). After 7 days of culture, results show increased levels of cardiogenic gene expression (NKX2.5, CX43) on both decellularized cardiogels compared to control, while the proportion and staining patterns of GATA4, OCT4, NKX2.5, ACTA1, VIM, and CD90-positive CPCs were not affected, as assessed by immunofluorescence microscopy and flow cytometry analyses. Nonetheless, CDCs cultured on CG-N secreted significantly higher levels of osteopontin, FGF6, FGF7, NT-3, IGFBP4, and TIMP-2 compared to those cultured on CG-P, suggesting overall a reduced trophic and antiremodelling paracrine profile of CDCs when in contact with ECM from pathological cardiac fibroblasts. These results provide novel insights into the bidirectional interplay between cardiac ECM and CPCs, potentially affecting CPC biology and regenerative potential.
APA, Harvard, Vancouver, ISO, and other styles
18

Malliaras, Konstantinos, Tao-Sheng Li, Daniel Luthringer, John Terrovitis, Ke Cheng, Tarun Chakravarty, Giselle Galang, et al. "Safety and Efficacy of Allogeneic Cell Therapy in Infarcted Rats Transplanted With Mismatched Cardiosphere-Derived Cells." Circulation 125, no. 1 (January 2012): 100–112. http://dx.doi.org/10.1161/circulationaha.111.042598.

Full text
APA, Harvard, Vancouver, ISO, and other styles
19

Gago-Lopez, Nuria, Obinna Awaji, Yiqiang Zhang, Christopher Ko, Ali Nsair, David Liem, April Stempien-Otero, and W. Robb MacLellan. "THY-1 Receptor Expression Differentiates Cardiosphere-Derived Cells with Divergent Cardiogenic Differentiation Potential." Stem Cell Reports 2, no. 5 (May 2014): 576–91. http://dx.doi.org/10.1016/j.stemcr.2014.03.003.

Full text
APA, Harvard, Vancouver, ISO, and other styles
20

Nazari, Hojjatollah, Mousa Kehtari, Iman Rad, Behnaz Ashtari, and Mohammad Taghi Joghataei. "Electrical stimulation induces differentiation of human cardiosphere-derived cells (hCDCs) to committed cardiomyocyte." Molecular and Cellular Biochemistry 470, no. 1-2 (May 9, 2020): 29–39. http://dx.doi.org/10.1007/s11010-020-03742-6.

Full text
APA, Harvard, Vancouver, ISO, and other styles
21

Wang, Siyuan, Weidan Chen, Li Ma, Minghui Zou, Wenyan Dong, Haili Yang, Lei Sun, Xinxin Chen, and Jinzhu Duan. "Infant cardiosphere-derived cells exhibit non-durable heart protection in dilated cardiomyopathy rats." Cytotechnology 71, no. 6 (October 3, 2019): 1043–52. http://dx.doi.org/10.1007/s10616-019-00328-z.

Full text
APA, Harvard, Vancouver, ISO, and other styles
22

Gómez-Cid, Lidia, Marina Moro-López, Ana de la Nava, Ismael Hernández-Romero, Ana Fernández, Susana Suárez-Sancho, Felipe Atienza, Lilian Grigorian-Shamagian, and Francisco Fernández-Avilés. "Electrophysiological Effects of Extracellular Vesicles Secreted by Cardiosphere-Derived Cells: Unraveling the Antiarrhythmic Properties of Cell Therapies." Processes 8, no. 8 (August 2, 2020): 924. http://dx.doi.org/10.3390/pr8080924.

Full text
Abstract:
Although cell-based therapies show potential antiarrhythmic effects that could be mediated by their paracrine action, the mechanisms and the extent of these effects were not deeply explored. We investigated the antiarrhythmic mechanisms of extracellular vesicles secreted by cardiosphere-derived cell extracellular vesicles (CDC-EVs) on the electrophysiological properties and gene expression profile of HL1 cardiomyocytes. HL-1 cultures were primed with CDC-EVs or serum-free medium alone for 48 h, followed by optical mapping and gene expression analysis. In optical mapping recordings, CDC-EVs reduced the activation complexity of the cardiomyocytes by 40%, increased rotor meandering, and reduced rotor curvature, as well as induced an 80% increase in conduction velocity. HL-1 cells primed with CDC-EVs presented higher expression of SCN5A, CACNA1C, and GJA1, coding for proteins involved in INa, ICaL, and Cx43, respectively. Our results suggest that CDC-EVs reduce activation complexity by increasing conduction velocity and modifying rotor dynamics, which could be driven by an increase in expression of SCN5A and CACNA1C genes, respectively. Our results provide new insights into the antiarrhythmic mechanisms of cell therapies, which should be further validated using other models.
APA, Harvard, Vancouver, ISO, and other styles
23

Bonios, Michael, Connie Y. Chang, Aurelio Pinheiro, Veronica Lea Dimaano, Takahiro Higuchi, Christina Melexopoulou, Frank Bengel, John Terrovitis, Theodore P. Abraham, and M. Roselle Abraham. "Cardiac Resynchronization by Cardiosphere-Derived Stem Cell Transplantation in an Experimental Model of Myocardial Infarction." Journal of the American Society of Echocardiography 24, no. 7 (July 2011): 808–14. http://dx.doi.org/10.1016/j.echo.2011.03.003.

Full text
APA, Harvard, Vancouver, ISO, and other styles
24

Hsiao, Lien-Cheng, Filippo Perbellini, Renata S. M. Gomes, Jun Jie Tan, Silvia Vieira, Giuseppe Faggian, Kieran Clarke, and Carolyn A. Carr. "Murine Cardiosphere-Derived Cells Are Impaired by Age but Not by Cardiac Dystrophic Dysfunction." Stem Cells and Development 23, no. 9 (May 2014): 1027–36. http://dx.doi.org/10.1089/scd.2013.0388.

Full text
APA, Harvard, Vancouver, ISO, and other styles
25

Kawaguchi, Nanako, Mitsuyo Machida, Kota Hatta, Toshio Nakanishi, and Yohtaroh Takagaki. "Cell Shape and Cardiosphere Differentiation: A Revelation by Proteomic Profiling." Biochemistry Research International 2013 (2013): 1–9. http://dx.doi.org/10.1155/2013/730874.

Full text
Abstract:
Stem cells (embryonic stem cells, somatic stem cells such as neural stem cells, and cardiac stem cells) and cancer cells are known to aggregate and form spheroid structures. This behavior is common in undifferentiated cells and may be necessary for adapting to certain conditions such as low-oxygen levels or to maintain undifferentiated status in microenvironments including stem cell niches. In order to decipher the meaning of this spheroid structure, we established a cardiosphere clone (CSC-21E) derived from the rat heart which can switch its morphology between spheroid and nonspheroid. Two forms, floating cardiospheres and dish-attached flat cells, could be switched reversibly by changing the cell culture condition. We performed differential proteome analysis studies and obtained protein profiles distinct between spherical forms and flat cells. From protein profiling analysis, we found upregulation of glycolytic enzymes in spheroids with some stress proteins switched in expression levels between these two forms. Evidence has been accumulating that certain chaperone/stress proteins are upregulated in concert with cellular changes including proliferation and differentiation. We would like to discuss the possible mechanism of how these aggregates affect cell differentiation and/or other cellular functions.
APA, Harvard, Vancouver, ISO, and other styles
26

Farrugia, Georgiana, and Rena Balzan. "Stem Cell Repair for Cardiac Muscle Regeneration: A Review of the Literature." International Journal of Medical Students 4, no. 1 (April 30, 2016): 19–25. http://dx.doi.org/10.5195/ijms.2016.145.

Full text
Abstract:
The notion that the human adult heart is a quiescent organ incapable of self-regeneration has been successfully challenged. It is now evident that the heart possesses a significant ability for repair and regeneration. Stem cells of endogenous cardiac origin are currently considered to possess the greatest ability to differentiate into cardiomyocytes. The major types of cardiac stem cells that show a promising potential to replace damaged cardiomyocytes include C-KIT positive (C-KIT+) cardiac progenitor cells, cardiosphere-derived progenitor cells, islet-1 (Isl1+) cardiac progenitor cells, side-population cardiac progenitor cells, epicardium-derived progenitor cells and stem cell antigen-1 (SCA1+) cardiac progenitor cells. Moreover, stem cells of extra-cardiac origin are also thought to restore contractility and vascularization of the myocardium. These include skeletal myoblasts, bone marrow mononuclear cells, mesenchymal stem cells, endothelial progenitor cells as well as embryonic stem cells. The need for further investigation on cardiac stem cell therapeutic strategies still remains.
APA, Harvard, Vancouver, ISO, and other styles
27

Marunouchi, Tetsuro, Emi Yano, and Kouichi Tanonaka. "Effects of cardiosphere-derived cell transplantation on cardiac mitochondrial oxygen consumption after myocardial infarction in rats." Biomedicine & Pharmacotherapy 108 (December 2018): 883–92. http://dx.doi.org/10.1016/j.biopha.2018.09.117.

Full text
APA, Harvard, Vancouver, ISO, and other styles
28

Namazi, Helia, Elham Mohit, Iman Namazi, Sarah Rajabi, Azam Samadian, Ensiyeh Hajizadeh-Saffar, Nasser Aghdami, and Hossein Baharvand. "Exosomes secreted by hypoxic cardiosphere-derived cells enhance tube formation and increase pro-angiogenic miRNA." Journal of Cellular Biochemistry 119, no. 5 (January 22, 2018): 4150–60. http://dx.doi.org/10.1002/jcb.26621.

Full text
APA, Harvard, Vancouver, ISO, and other styles
29

Sun, Yong, Di Chi, Miaoxin Tan, Kai Kang, Maomao Zhang, Xiangyuan Jin, Xiaoping Leng, et al. "Cadaveric cardiosphere-derived cells can maintain regenerative capacity and improve the heart function of cardiomyopathy." Cell Cycle 15, no. 9 (April 8, 2016): 1248–56. http://dx.doi.org/10.1080/15384101.2016.1160973.

Full text
APA, Harvard, Vancouver, ISO, and other styles
30

Nana-Leventaki, E., M. Nana, N. Poulianitis, D. Sampaziotis, D. Perrea, D. Sanoudou, D. Rontogianni, and K. Malliaras. "Cardiosphere-Derived Cells Attenuate Inflammation, Preserve Systolic Function, and Prevent Adverse Remodeling in Rat Hearts With Experimental Autoimmune Myocarditis." Journal of Cardiovascular Pharmacology and Therapeutics 24, no. 1 (July 30, 2018): 70–77. http://dx.doi.org/10.1177/1074248418784287.

Full text
Abstract:
Background: Cardiosphere-derived cells (CDCs) have yielded promising efficacy signals in early-phase clinical trials of ischemic and nonischemic cardiomyopathy. The potential efficacy of CDCs in acute myocarditis, an inflammatory cardiomyopathy without effective therapy, remains unexplored. Given that CDCs produce regenerative, cardioprotective, anti-inflammatory, and anti-fibrotic effects (all of which could be beneficial in acute myocarditis), we investigated the efficacy of intracoronary delivery of CDCs in a rat model of experimental autoimmune myocarditis. Methods: Lewis rats underwent induction of experimental autoimmune myocarditis by subcutaneous footpad injection of purified porcine cardiac myosin supplemented with Mycobacterium tuberculosis on days 1 and 7. On day 10, rats were randomly assigned to receive global intracoronary delivery of 500 000 CDCs or vehicle. Global intracoronary delivery was performed by injection of cells or vehicle into the left ventricular (LV) cavity during transient occlusion of the aortic root. Rats were euthanized 18 days after infusion. Cardiac volumes and systolic function were assessed by serial echocardiography, performed on days 1, 10, and 28. Myocardial inflammation, T-cell infiltration, and cardiac fibrosis were evaluated by histology. Results: Experimental autoimmune myocarditis was successfully induced in 14/14 rats that completed follow-up. Left ventricular ejection fraction (LVEF) and volumes were comparable on days 1 and 10 between groups. CDC infusion resulted in increased LVEF (81.5% ± 3% vs 65.4% ± 8%, P < .001) and decreased LV end-systolic volume (43 ± 15 vs 100 ± 24 μL, P < .001) compared to placebo administration at 18 days post-infusion. Cardiosphere-derived cell infusion decreased myocardial inflammation (7.4% ± 7% vs 20.7% ± 4% of myocardium, P = .007), cardiac fibrosis (16.6% ± 13% vs 38.1% ± 3% of myocardium, P = .008), and myocardial T-cell infiltration (30.4 ± 29 vs 125.8 ± 49 cells per field, P = .005) at 18 days post-infusion compared to placebo administration. Conclusion: Intracoronary delivery of CDCs attenuates myocardial inflammation, T-cell infiltration, and fibrosis while preventing myocarditis-induced systolic dysfunction and adverse remodeling in rats with experimental autoimmune myocarditis.
APA, Harvard, Vancouver, ISO, and other styles
31

Lapchak, Paul A., Paul D. Boitano, Geoffrey de Couto, and Eduardo Marbán. "Intravenous xenogeneic human cardiosphere-derived cell extracellular vesicles (exosomes) improves behavioral function in small-clot embolized rabbits." Experimental Neurology 307 (September 2018): 109–17. http://dx.doi.org/10.1016/j.expneurol.2018.06.007.

Full text
APA, Harvard, Vancouver, ISO, and other styles
32

Suzuki, Gen, Rebeccah F. Young, Merced M. Leiker, and Takayuki Suzuki. "Heart-Derived Stem Cells in Miniature Swine with Coronary Microembolization: Novel Ischemic Cardiomyopathy Model to Assess the Efficacy of Cell-Based Therapy." Stem Cells International 2016 (2016): 1–14. http://dx.doi.org/10.1155/2016/6940195.

Full text
Abstract:
A major problem in translating stem cell therapeutics is the difficulty of producing stable, long-term severe left ventricular (LV) dysfunction in a large animal model. For that purpose, extensive infarction was created in sinclair miniswine by injecting microspheres (1.5 × 106microspheres, 45 μm diameter) in LAD. At 2 months after embolization, animals (n=11) were randomized to receive allogeneic cardiosphere-derived cells derived from atrium (CDCs: 20 × 106,n=5) or saline (untreated,n=6). Four weeks after therapy myocardial function, myocyte proliferation (Ki67), mitosis (phosphor-Histone H3; pHH3), apoptosis, infarct size (TTC), myocyte nuclear density, and cell size were evaluated. CDCs injected into infarcted and remodeled remote myocardium (global infusion) increased regional function and global function contrasting no change in untreated animals. CDCs reduced infarct volume and stimulated Ki67 and pHH3 positive myocytes in infarct and remote regions. As a result, myocyte number (nuclear density) increased and myocyte cell diameter decreased in both infarct and remote regions. Coronary microembolization produces stable long-term ischemic cardiomyopathy. Global infusion of CDCs stimulates myocyte regeneration and improves left ventricular ejection fraction. Thus, global infusion of CDCs could become a new therapy to reverse LV dysfunction in patients with asymptomatic heart failure.
APA, Harvard, Vancouver, ISO, and other styles
33

Lin, Yen-Nien, Thassio Mesquita, Lizbeth Sanchez, Yin-Huei Chen, Weixin Liu, Chang Li, Russell Rogers, et al. "Extracellular vesicles from immortalized cardiosphere-derived cells attenuate arrhythmogenic cardiomyopathy in desmoglein-2 mutant mice." European Heart Journal 42, no. 35 (July 29, 2021): 3558–71. http://dx.doi.org/10.1093/eurheartj/ehab419.

Full text
Abstract:
Abstract Aims Arrhythmogenic cardiomyopathy (ACM) is characterized by progressive loss of cardiomyocytes, and fibrofatty tissue replacement. Extracellular vesicles (EVs) secreted by cardiosphere-derived cells, immortalized, and engineered to express high levels of β-catenin, exert anti-inflammatory, and anti-fibrotic effects. The aim of the current study was to assess efficacy of EVs in an ACM murine model. Methods and results Four-week-old homozygous knock-in mutant desmoglein-2 (Dsg2mt/mt) were randomized to receive weekly EVs or vehicle for 4 weeks. After 4 weeks, DSG2mt/mt mice receiving EVs showed improved biventricular function (left, P &lt; 0.0001; right, P = 0.0037) and less left ventricular dilation (P &lt; 0.0179). Electrocardiography revealed abbreviated QRS duration (P = 0.0003) and QTc interval (P = 0.0006) in EV-treated DSG2mt/mt mice. Further electrophysiology testing in the EV group showed decreased burden (P = 0.0042) and inducibility of ventricular arrhythmias (P = 0.0037). Optical mapping demonstrated accelerated repolarization (P = 0.0290) and faster conduction (P = 0.0274) in Dsg2mt/mt mice receiving EVs. DSG2mt/mt hearts exhibited reduced fibrosis, less cell death, and preserved connexin 43 expression after EV treatment. Hearts of Dsg2mt/mt mice expressed markedly increased levels of inflammatory cytokines that were, in part, attenuated by EV therapy. The pan-inflammatory transcription factor nuclear factor-κB (NF-κB), the inflammasome sensor NLRP3, and the macrophage marker CD68 were all reduced in EV-treated animals. Blocking EV hsa-miR-4488 in vitro and in vivo reactivates NF-κB and blunts the beneficial effects of EVs. Conclusions Extracellular vesicle treatment improved cardiac function, reduced cardiac inflammation, and suppressed arrhythmogenesis in ACM. Further studies are needed prior to translating the present findings to human forms of this heterogenous disease.
APA, Harvard, Vancouver, ISO, and other styles
34

Thej, Charan, and Raj Kishore. "Unfathomed Nanomessages to the Heart: Translational Implications of Stem Cell-Derived, Progenitor Cell Exosomes in Cardiac Repair and Regeneration." Cells 10, no. 7 (July 17, 2021): 1811. http://dx.doi.org/10.3390/cells10071811.

Full text
Abstract:
Exosomes formed from the endosomal membranes at the lipid microdomains of multivesicular bodies (MVBs) have become crucial structures responsible for cell communication. This paracrine communication system between a myriad of cell types is essential for maintaining homeostasis and influencing various biological functions in immune, vasculogenic, and regenerative cell types in multiple organs in the body, including, but not limited to, cardiac cells and tissues. Characteristically, exosomes are identifiable by common proteins that participate in their biogenesis; however, many different proteins, mRNA, miRNAs, and lipids, have been identified that mediate intercellular communication and elicit multiple functions in other target cells. Although our understanding of exosomes is still limited, the last decade has seen a steep surge in translational studies involving the treatment of cardiovascular diseases with cell-free exosome fractions from cardiomyocytes (CMs), cardiosphere-derived cells (CDCs), endothelial cells (ECs), mesenchymal stromal cells (MSCs), or their combinations. However, most primary cells are difficult to culture in vitro and to generate sufficient exosomes to treat cardiac ischemia or promote cardiac regeneration effectively. Pluripotent stem cells (PSCs) offer the possibility of an unlimited supply of either committed or terminally differentiated cells and their exosomes for treating cardiovascular diseases (CVDs). This review discusses the promising prospects of treating CVDs using exosomes from cardiac progenitor cells (CPCs), endothelial progenitor cells (EPCs), MSCs, and cardiac fibroblasts derived from PSCs.
APA, Harvard, Vancouver, ISO, and other styles
35

Saha, Progyaparamita, Sudhish Sharma, Laxminarayana Korutla, Srinivasa Raju Datla, Farnaz Shoja-Taheri, Rachana Mishra, Grace E. Bigham, et al. "Circulating exosomes derived from transplanted progenitor cells aid the functional recovery of ischemic myocardium." Science Translational Medicine 11, no. 493 (May 22, 2019): eaau1168. http://dx.doi.org/10.1126/scitranslmed.aau1168.

Full text
Abstract:
The stem cell field is hindered by its inability to noninvasively monitor transplanted cells within the target organ in a repeatable, time-sensitive, and condition-specific manner. We hypothesized that quantifying and characterizing transplanted cell–derived exosomes in the recipient plasma would enable reliable, noninvasive surveillance of the conditional activity of the transplanted cells. To test this hypothesis, we used a human-into-rat xenogeneic myocardial infarction model comparing two well-studied progenitor cell types: cardiosphere-derived cells (CDCs) and c-kit+ cardiac progenitor cells (CPCs), both derived from the right atrial appendage of adults undergoing cardiopulmonary bypass. CPCs outperformed the CDCs in cell-based and in vivo regenerative assays. To noninvasively monitor the activity of transplanted CDCs or CPCs in vivo, we purified progenitor cell–specific exosomes from recipient total plasma exosomes. Seven days after transplantation, the concentration of plasma CPC-specific exosomes increased about twofold compared to CDC-specific exosomes. Computational pathway analysis failed to link CPC or CDC cellular messenger RNA (mRNA) with observed myocardial recovery, although recovery was linked to the microRNA (miRNA) cargo of CPC exosomes purified from recipient plasma. We further identified mechanistic pathways governing specific outcomes related to myocardial recovery associated with transplanted CPCs. Collectively, these findings demonstrate the potential of circulating progenitor cell–specific exosomes as a liquid biopsy that provides a noninvasive window into the conditional state of the transplanted cells. These data implicate the surveillance potential of cell-specific exosomes for allogeneic cell therapies.
APA, Harvard, Vancouver, ISO, and other styles
36

de Couto, Geoffrey, Ervin Jaghatspanyan, Matthew DeBerge, Weixin Liu, Kristin Luther, Yizhou Wang, Jie Tang, Edward B. Thorp, and Eduardo Marbán. "Mechanism of Enhanced MerTK-Dependent Macrophage Efferocytosis by Extracellular Vesicles." Arteriosclerosis, Thrombosis, and Vascular Biology 39, no. 10 (October 2019): 2082–96. http://dx.doi.org/10.1161/atvbaha.119.313115.

Full text
Abstract:
Objective: Extracellular vesicles secreted by cardiosphere-derived cells (CDC ev ) polarize macrophages toward a distinctive phenotype with enhanced phagocytic capacity (M CDCev ). These changes underlie cardioprotection by CDC ev and by the parent CDCs, notably attenuating the no-reflow phenomenon following myocardial infarction, but the mechanisms are unclear. Here, we tested the hypothesis that M CDCev are especially effective at scavenging debris from dying cells (ie, efferocytosis) to attenuate irreversible damage post-myocardial infarction. Approach and Results: In vitro efferocytosis assays with bone marrow-derived macrophages, and in vivo transgenic rodent models of myocardial infarction, demonstrate enhanced apoptotic cell clearance with M CDCev . CDC ev exposure induces sustained MerTK expression in M CDCev through extracellular vesicle transfer of microRNA-26a (via suppression of Adam17 ); the cardioprotective response is lost in animals deficient in MerTK. Single-cell RNA-sequencing revealed phagocytic pathway activation in M CDCev , with increased expression of complement factor C1qa , a phagocytosis facilitator. Conclusions: Together, these data demonstrate that extracellular vesicle modulation of MerTK and C1qa expression leads to enhanced macrophage efferocytosis and cardioprotection.
APA, Harvard, Vancouver, ISO, and other styles
37

Ostovaneh, Mohammad R., Raj R. Makkar, Bharath Ambale-Venkatesh, Deborah Ascheim, Tarun Chakravarty, Timothy D. Henry, Glen Kowalchuk, et al. "Effect of cardiosphere-derived cells on segmental myocardial function after myocardial infarction: ALLSTAR randomised clinical trial." Open Heart 8, no. 2 (July 2021): e001614. http://dx.doi.org/10.1136/openhrt-2021-001614.

Full text
Abstract:
BackgroundMost cell therapy trials failed to show an improvement in global left ventricular (LV) function measures after myocardial infarction (MI). Myocardial segments are heterogeneously impacted by MI. Global LV function indices are not able to detect the small treatment effects on segmental myocardial function which may have prognostic implications for cardiac events. We aimed to test the efficacy of allogeneic cardiosphere-derived cells (CDCs) for improving regional myocardial function and contractility.MethodsIn this exploratory analysis of a randomised clinical trial, 142 patients with post-MI with LVEF <45% and 15% or greater LV scar size were randomised in 2:1 ratio to receive intracoronary infusion of allogenic CDCs or placebo, respectively. Change in segmental myocardial circumferential strain (Ecc) by MRI from baseline to 6 months was compared between CDCs and placebo groups.ResultsIn total, 124 patients completed the 6-month follow-up (mean (SD) age 54.3 (10.8) and 108 (87.1%) men). Segmental Ecc improvement was significantly greater in patients receiving CDC (−0.5% (4.0)) compared with placebo (0.2% (3.7), p=0.05). The greatest benefit for improvement in segmental Ecc was observed in segments containing scar tissue (change in segmental Ecc of −0.7% (3.5) in patients receiving CDC vs 0.04% (3.7) in the placebo group, p=0.04).ConclusionsIn patients with post-MI LV dysfunction, CDC administration resulted in improved segmental myocardial function. Our findings highlight the importance of segmental myocardial function indices as an endpoint in future clinical trials of patients with post-MI.Trial registration numberNCT01458405.
APA, Harvard, Vancouver, ISO, and other styles
38

Li, Tao-Sheng, Ke Cheng, Konstantinos Malliaras, Rachel Ruckdeschel Smith, Yiqiang Zhang, Baiming Sun, Noriko Matsushita, et al. "Direct Comparison of Different Stem Cell Types and Subpopulations Reveals Superior Paracrine Potency and Myocardial Repair Efficacy With Cardiosphere-Derived Cells." Journal of the American College of Cardiology 59, no. 10 (March 2012): 942–53. http://dx.doi.org/10.1016/j.jacc.2011.11.029.

Full text
APA, Harvard, Vancouver, ISO, and other styles
39

Aghila Rani, Koippallil GopalakrishnanNair, and Chandrasekharan Cheranellore Kartha. "Effects of epidermal growth factor on proliferation and migration of cardiosphere-derived cells expanded from adult human heart." Growth Factors 28, no. 3 (February 19, 2010): 157–65. http://dx.doi.org/10.3109/08977190903512628.

Full text
APA, Harvard, Vancouver, ISO, and other styles
40

Ekhteraei-Tousi, Samaneh, Bahram Mohammad-Soltani, Majid Sadeghizadeh, Seyed Javad Mowla, Sepideh Parsi, and Masoud Soleimani. "Inhibitory Effect of Hsa-miR-590-5p on Cardiosphere-derived Stem Cells Differentiation Through Downregulation of TGFB Signaling." Journal of Cellular Biochemistry 116, no. 1 (November 11, 2014): 179–91. http://dx.doi.org/10.1002/jcb.24957.

Full text
APA, Harvard, Vancouver, ISO, and other styles
41

Fu, Wenbin, and Chunyu Zeng. "GW28-e0089 Metformin Promotes the Survival of Transplanted Cardiosphere-Derived Cell thereby Enhancing Its Therapeutic Effect against Myocardial Infarction." Journal of the American College of Cardiology 70, no. 16 (October 2017): C2. http://dx.doi.org/10.1016/j.jacc.2017.07.007.

Full text
APA, Harvard, Vancouver, ISO, and other styles
42

Lo, Chi Y., Brian R. Weil, Beth A. Palka, Arezoo Momeni, John M. Canty, and Sriram Neelamegham. "Cell surface glycoengineering improves selectin-mediated adhesion of mesenchymal stem cells (MSCs) and cardiosphere-derived cells (CDCs): Pilot validation in porcine ischemia-reperfusion model." Biomaterials 74 (January 2016): 19–30. http://dx.doi.org/10.1016/j.biomaterials.2015.09.026.

Full text
APA, Harvard, Vancouver, ISO, and other styles
43

Ishigami, Shuta, Toshikazu Sano, Sunaya Krishnapura, Tatsuo Ito, and Shunji Sano. "An overview of stem cell therapy for paediatric heart failure." European Journal of Cardio-Thoracic Surgery 58, no. 5 (June 26, 2020): 881–87. http://dx.doi.org/10.1093/ejcts/ezaa155.

Full text
Abstract:
Abstract Significant achievements in paediatric cardiology, surgical treatment and intensive care of congenital heart disease have drastically changed clinical outcomes for paediatric patients. Nevertheless, late-onset heart failure in children after staged surgeries still remains a serious concern in the medical community. Heart transplantation is an option for treatment; however, the shortage of available organs is a persistent problem in many developed countries. In order to resolve these issues, advanced technologies, such as innovative mechanical circulatory support devices and regenerative therapies, are strongly desired. Accumulated evidence regarding cell-based cardiac regenerative therapies has suggested their safety and efficacy in treating adult heart failure. Given that young children seem to have a higher regenerative capacity than adults, stem cell-based therapies appear a promising treatment option for paediatric heart failure as well. Based on the findings from past trials and studies, we present the potential of various different types of stem cells, ranging from bone marrow mononuclear cells to cardiosphere-derived stem cells for use in paediatric cell-based therapies. Here, we assess both the current challenges associated with cell-based therapies and novel strategies that may be implemented in the future to advance stem cell therapy in the paediatric population.
APA, Harvard, Vancouver, ISO, and other styles
44

Yap, Jonathan, Hector A. Cabrera-Fuentes, Jason Irei, Derek J. Hausenloy, and William A. Boisvert. "Role of Macrophages in Cardioprotection." International Journal of Molecular Sciences 20, no. 10 (May 19, 2019): 2474. http://dx.doi.org/10.3390/ijms20102474.

Full text
Abstract:
Cardiovascular diseases are the leading cause of mortality worldwide. It is widely known that non-resolving inflammation results in atherosclerotic conditions, which are responsible for a host of downstream pathologies including thrombosis, myocardial infarction (MI), and neurovascular events. Macrophages, as part of the innate immune response, are among the most important cell types in every stage of atherosclerosis. In this review we discuss the principles governing macrophage function in the healthy and infarcted heart. More specifically, how cardiac macrophages participate in myocardial infarction as well as cardiac repair and remodeling. The intricate balance between phenotypically heterogeneous populations of macrophages in the heart have profound and highly orchestrated effects during different phases of myocardial infarction. In the early “inflammatory” stage of MI, resident cardiac macrophages are replaced by classically activated macrophages derived from the bone marrow and spleen. And while the macrophage population shifts towards an alternatively activated phenotype, the inflammatory response subsides giving way to the “reparative/proliferative” phase. Lastly, we describe the therapeutic potential of cardiac macrophages in the context of cell-mediated cardio-protection. Promising results demonstrate innovative concepts; one employing a subset of yolk sac-derived, cardiac macrophages that have complete restorative capacity in the injured myocardium of neonatal mice, and in another example, post-conditioning of cardiac macrophages with cardiosphere-derived cells significantly improved patient’s post-MI diagnoses.
APA, Harvard, Vancouver, ISO, and other styles
45

Suzuki, Gen, Brian R. Weil, Rebeccah F. Young, James A. Fallavollita, and John M. Canty. "Nonocclusive multivessel intracoronary infusion of allogeneic cardiosphere-derived cells early after reperfusion prevents remote zone myocyte loss and improves global left ventricular function in swine with myocardial infarction." American Journal of Physiology-Heart and Circulatory Physiology 317, no. 2 (August 1, 2019): H345—H356. http://dx.doi.org/10.1152/ajpheart.00124.2019.

Full text
Abstract:
Intracoronary cardiosphere-derived cells (icCDCs) infused into the infarct-related artery reduce scar volume but do not improve left ventricular (LV) ejection fraction (LVEF). We tested the hypothesis that this reflects the inability of regional delivery to prevent myocyte death or promote myocyte proliferation in viable myocardium remote from the infarct. Swine ( n = 23) pretreated with oral cyclosporine (200 mg/day) underwent a 1-h left anterior descending coronary artery (LAD) occlusion, which reduced LVEF from 61.6 ± 1.0 to 45.3 ± 1.5% 30 min after reperfusion. At that time, animals received global infusion of allogeneic icCDCs ( n = 8), regional infusion of icCDCs restricted to the LAD using the stop-flow technique ( n = 8), or vehicle ( n = 7). After 1 mo, global icCDCs increased LVEF from 44.8 ± 1.9 to 60.8 ± 3.8% ( P < 0.05) with no significant change after LAD stop-flow icCDCs (44.8 ± 3.6 to 50.9 ± 3.1%) or vehicle (46.5 ± 2.5 to 47.7 ± 2.6%). In contrast, global icCDCs did not alter infarct volume (%LV mass) assessed at 2 days (11.2 ± 2.3 vs. 12.6 ± 2.3%), whereas it was reduced after LAD stop-flow icCDCs (7.1 ± 1.1%, P < 0.05). Histopathological analysis of remote myocardium after global icCDCs demonstrated a significant increase in myocyte proliferation (147 ± 32 vs. 14 ± 10 nuclei/106 myocytes, P < 0.05) and a reduction in myocyte apoptosis (15 ± 9 vs. 46 ± 10 nuclei/106 myocytes, P < 0.05) that increased myocyte nuclear density (1,264 ± 39 vs. 1,157 ± 33 nuclei/mm2, P < 0.05) and decreased myocyte diameter (13.2 ± 0.2 vs. 14.5 ± 0.3 μm, P < 0.05) compared with vehicle-treated controls. In contrast, remote zone changes after regional LAD icCDCs were no different from vehicle. These data indicate that changes in global LVEF after icCDCs are dependent upon preventing myocyte loss and hypertrophy in myocardium remote from the infarct. These arise from stimulating myocyte proliferation and reducing myocyte apoptosis indicating the importance of directing cell therapy to viable remote regions. NEW & NOTEWORTHY Administration of allogeneic cardiosphere-derived cells to the entire heart via global intracoronary infusion shortly after myocardial infarction favorably influenced left ventricular ejection fraction by preventing myocyte death and promoting myocyte proliferation in remote, noninfarcted myocardium in swine. In contrast, regional intracoronary cell infusion did not significantly affect remote zone myocyte remodeling. Global cell administration targeting viable myocardium remote from the infarct may be an effective approach to prevent adverse ventricular remodeling after myocardial infarction.
APA, Harvard, Vancouver, ISO, and other styles
46

Takehara, Naofumi, Yoshiaki Tsutsumi, Kento Tateishi, Takehiro Ogata, Hideo Tanaka, Tomomi Ueyama, Tomosaburo Takahashi, et al. "Controlled Delivery of Basic Fibroblast Growth Factor Promotes Human Cardiosphere-Derived Cell Engraftment to Enhance Cardiac Repair for Chronic Myocardial Infarction." Journal of the American College of Cardiology 52, no. 23 (December 2008): 1858–65. http://dx.doi.org/10.1016/j.jacc.2008.06.052.

Full text
APA, Harvard, Vancouver, ISO, and other styles
47

Bonios, Michael, Connie Yachan Chang, John Terrovitis, Aurelio Pinheiro, Andreas Barth, Peihong Dong, Miguel Santaularia, et al. "Constitutive HIF-1α Expression Blunts the Beneficial Effects of Cardiosphere-Derived Cell Therapy in the Heart by Altering Paracrine Factor Balance." Journal of Cardiovascular Translational Research 4, no. 3 (May 3, 2011): 363–72. http://dx.doi.org/10.1007/s12265-011-9265-3.

Full text
APA, Harvard, Vancouver, ISO, and other styles
48

Straface, Elisabetta, Lucrezia Gambardella, Francesca Pagano, Francesco Angelini, Barbara Ascione, Rosa Vona, Elena De Falco, et al. "Sex Differences of Human Cardiac Progenitor Cells in the Biological Response to TNF-α Treatment." Stem Cells International 2017 (2017): 1–9. http://dx.doi.org/10.1155/2017/4790563.

Full text
Abstract:
Adult cardiac progenitor cells (CPCs), isolated as cardiosphere-derived cells (CDCs), represent promising candidates for cardiac regenerative therapy. CDCs can be expanded in vitro manyfolds without losing their differentiation potential, reaching numbers that are appropriate for clinical applications. Since mechanisms of successful CDC survival and engraftment in the damaged myocardium are still critical and unresolved issues, we aimed at deciphering possible key factors capable of bolstering CDC function. In particular, the response and the phenotype of CDCs exposed to low concentrations of the multifunctional cytokine tumor necrosis factor α (TNF-α), known to be capable of activating cell survival pathways, have been investigated. Furthermore, differential biological responses of CDCs from male and female donors, in terms of cell cycle progression and cell spreading, have also been assessed. The results obtained indicate that (i) the intracellular signaling activated in our experimental conditions is most likely due to the prosurvival and proliferative signaling of TNF-α receptor 2 and that (ii) cells from female patients appear more responsive to TNF-α treatment in terms of cell cycle progression and migration ability. In conclusion, the present report highlights the hypothesis that TNF-stimulated CDCs isolated from females may represent a promising candidate for cardiac regenerative therapy applications.
APA, Harvard, Vancouver, ISO, and other styles
49

Sousonis, Vasileios, Titika Sfakianaki, Argirios Ntalianis, Ioannis Nanas, Christos Kontogiannis, Dionysios Aravantinos, Chris Kapelios, et al. "Intracoronary Administration of Allogeneic Cardiosphere-Derived Cells Immediately Prior to Reperfusion in Pigs With Acute Myocardial Infarction Reduces Infarct Size and Attenuates Adverse Cardiac Remodeling." Journal of Cardiovascular Pharmacology and Therapeutics 26, no. 1 (July 17, 2020): 88–99. http://dx.doi.org/10.1177/1074248420941672.

Full text
Abstract:
Background: Allogeneic cardiosphere-derived cells (CDCs) exert cardioprotective effects when administered intracoronarily after reperfusion in animal models of acute myocardial infarction (AMI). The “no-reflow” phenomenon develops rapidly post-reperfusion and may undermine the efficacy of cell therapy, due to poor cell delivery in areas of microvascular obstruction (MVO). We hypothesized that CDC-induced cardioprotection would be enhanced by cell administration prior to reperfusion, when microvasculature is still relatively intact, to facilitate widespread cell delivery within the ischemic area. Methods and Results: We studied 81 farm pigs; 55 completed the specified protocols. A dose-optimization study in infarcted pigs demonstrated that the doses of 5 million and 10 million CDCs are the maximum safe doses that can be administered intracoronarily at 5 minutes prior to and at 5 minutes post-reperfusion, respectively, without aggravating MVO. Quantification of acute cell retention by polymerase chain reaction demonstrated that cell delivery prior to reperfusion resulted in higher cardiac cell retention compared to delivery post-reperfusion. We then performed a randomized, placebo-controlled study to assess the long-term efficacy of intracoronary infusion of 5 million allogeneic CDCs, delivered at 5 minutes prior to reperfusion, in a porcine model of AMI. The CDC therapy resulted in decreased scar size, improved regional systolic function, and attenuation of adverse cardiac remodeling (manifested as preserved global systolic function, preserved end-systolic volume, and decreased interstitial fibrosis) compared to placebo at 30 days post-MI. Conclusions: Dose-optimized intracoronary infusion of allogeneic CDCs prior to reperfusion in a porcine model of AMI is feasible, safe and confers long-term benefits.
APA, Harvard, Vancouver, ISO, and other styles
50

Zeng, Wendy R., and Pauline M. Doran. "Interactivity of biochemical and physical stimuli during epigenetic conditioning and cardiomyocytic differentiation of stem and progenitor cells derived from adult hearts." Integrative Biology 13, no. 3 (March 2021): 73–85. http://dx.doi.org/10.1093/intbio/zyab003.

Full text
Abstract:
Abstract Mixed populations of cardiosphere-derived stem and progenitor cells containing proliferative and cardiomyogenically committed cells were obtained from adult rat hearts. The cells were cultured in either static 2D monolayers or dynamic 3D scaffold systems with fluid flow. Cardiomyocyte lineage commitment in terms of GATA4 and Nkx2.5 expression was significantly enhanced in the dynamic 3D cultures compared with static 2D conditions. Treatment of the cells with 5-azacytidine (5-aza) produced different responses in the two culture systems, as activity of this chemical epigenetic conditioning agent depended on the cell attachment and hydrodynamic conditions provided during culture. Cell growth was unaffected by 5-aza in the static 2D cultures but was significantly reduced under dynamic 3D conditions relative to untreated controls. Myogenic differentiation measured as Mef2c expression was markedly upregulated by 5-aza in the dynamic 3D cultures but downregulated in the static 2D cultures. The ability of the physical environment to modulate the cellular cardiomyogenic response to 5-aza underscores the interactivity of biochemical and physical stimuli applied for cell differentiation. Accordingly, observations about the efficacy of 5-aza as a cardiomyocyte induction agent may not be applicable across different culture systems. Overall, use of dynamic 3D rather than static 2D culture was more beneficial for cardio-specific myogenesis than 5-aza treatment, which generated a more ambiguous differentiation response.
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography