1
|
Hayashi Y, Kim S, Fujii T, Pedersen DD, Ozeki T, Jiang H, D’Amore A, Wagner WR. Placement of an elastic, biohybrid patch in a model of right heart failure with pulmonary artery banding. Front Bioeng Biotechnol 2025; 12:1485740. [PMID: 39902173 PMCID: PMC11788599 DOI: 10.3389/fbioe.2024.1485740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 12/09/2024] [Indexed: 02/05/2025] Open
Abstract
Introduction In a model of right heart failure secondary to pulmonary artery banding (PAB), a mechanical approach using an elastic, biodegradable epicardial patch with integrated extracellular matrix digest was evaluated for its potential to inhibit disease progression. Methods Adult male syngeneic Lewis rats aged 6-7 weeks old were used. Biohybrid cardiac patches were generated by co-processing biodegradable poly(ester carbonate urethane) urea (PECUU) and a digest of the porcine cardiac extracellular matrix. Three weeks after PAB, the cardiac patch was attached to the epicardium of the right ventricle (RV). Cardiac function was evaluated using echocardiography and catheterization for 9 weeks after PAB, comparing the patch (n = 7) and sham (n = 10) groups. Results Nine weeks after PAB, the RV wall was thickened, the RV cavity was enlarged with a reduced left ventricular cavity, and RV wall interstitial fibrosis was increased. However, these effects were diminished in the patch group. Left ventricular ejection fraction in the patch group was higher than in the sham group (p < 0.001), right end-systolic pressure was lower (p = 0.045), and tricuspid annular plane systolic excursion improved in the patch group (p = 0.007). In addition, von Willebrand factor expression was significantly greater in the patch group (p = 0.007). Conclusions The placement of a degradable, biohybrid patch onto the RV in a right ventricular failure model with fixed afterload improved myocardial output, moderated pressure stress, and was associated with reduced right ventricular fibrosis.
Collapse
Affiliation(s)
- Yasunari Hayashi
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Seungil Kim
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Agricultural and Biological Engineering, Mississippi State University, Mississippi State, MS, United States
| | - Taro Fujii
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Drake Dalton Pedersen
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Takahiro Ozeki
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Hongbin Jiang
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Antonio D’Amore
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
- Fondazione RiMED, Palermo, Italy
- Medicina di Precisione in Area Medica, Chirurgica e Critica, University of Palermo, Palermo, Italy
| | - William R. Wagner
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
2
|
Cao M, Liu Y, Sun Y, Han R, Jiang H. Current advances in human-induced pluripotent stem cell-based models and therapeutic approaches for congenital heart disease. Mol Cell Biochem 2025; 480:159-172. [PMID: 38635080 DOI: 10.1007/s11010-024-04997-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: 12/29/2023] [Accepted: 03/20/2024] [Indexed: 04/19/2024]
Abstract
Congenital heart disease (CHD) represents a significant risk factor with profound implications for neonatal survival rates and the overall well-being of adult patients. The emergence of induced pluripotent stem cells (iPSCs) and their derived cells, combined with CRISPR technology, high-throughput experimental techniques, and organoid technology, which are better suited to contemporary research demands, offer new possibilities for treating CHD. Prior investigations have indicated that the paracrine effect of exosomes may hold potential solutions for therapeutic intervention. This review provides a summary of the advancements in iPSC-based models and clinical trials associated with CHD while elucidating potential therapeutic mechanisms and delineating clinical constraints pertinent to iPSC-based therapy, thereby offering valuable insights for further deliberation.
Collapse
Affiliation(s)
- Meiling Cao
- Department of Neonatology, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China
| | - Yanshan Liu
- Department of Pediatrics, The First Hospital of China Medical University, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Ying Sun
- Department of Pediatrics, The First Hospital of China Medical University, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Ruiyi Han
- Department of Pediatrics, The First Hospital of China Medical University, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Hongkun Jiang
- Department of Pediatrics, The First Hospital of China Medical University, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
3
|
Zhang Z, Guo J. Deciphering Oxidative Stress in Cardiovascular Disease Progression: A Blueprint for Mechanistic Understanding and Therapeutic Innovation. Antioxidants (Basel) 2024; 14:38. [PMID: 39857372 PMCID: PMC11759168 DOI: 10.3390/antiox14010038] [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: 11/19/2024] [Revised: 12/20/2024] [Accepted: 12/27/2024] [Indexed: 01/27/2025] Open
Abstract
Oxidative stress plays a pivotal role in the pathogenesis and progression of cardiovascular diseases (CVDs). This review focuses on the signaling pathways of oxidative stress during the development of CVDs, delving into the molecular regulatory networks underlying oxidative stress in various disease stages, particularly apoptosis, inflammation, fibrosis, and metabolic imbalance. By examining the dual roles of oxidative stress and the influences of sex differences on oxidative stress levels and cardiovascular disease susceptibility, this study offers a comprehensive understanding of the pathogenesis of cardiovascular diseases. The study integrates key findings from current research in three comprehensive ways. First, it outlines the major CVDs associated with oxidative stress and their respective signaling pathways, emphasizing oxidative stress's central role in cardiovascular pathology. Second, it summarizes the cardiovascular protective effects, mechanisms of action, and animal models of various antioxidants, offering insights into future drug development. Third, it discusses the applications, advantages, limitations, and potential molecular targets of gene therapy in CVDs, providing a foundation for novel therapeutic strategies. These tables underscore the systematic and integrative nature of this study while offering a theoretical basis for precision treatment for CVDs. A major contribution of this study is the systematic review of the differential effects of oxidative stress across different stages of CVDs, in addition to the proposal of innovative, multi-level intervention strategies, which open new avenues for precision treatment of the cardiovascular system.
Collapse
Affiliation(s)
- Zhaoshan Zhang
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Jiawei Guo
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| |
Collapse
|
4
|
Robeson M, Goudy SL, Davis ME. Differential Effects of Four Canonical Notch-Activating Ligands on c-Kit+ Cardiac Progenitor Cells. Int J Mol Sci 2024; 25:11182. [PMID: 39456964 PMCID: PMC11508355 DOI: 10.3390/ijms252011182] [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: 08/16/2024] [Revised: 10/08/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Notch signaling, an important signaling pathway in cardiac development, has been shown to mediate the reparative functions of c-kit+ progenitor cells (CPCs). However, it is unclear how each of the four canonical Notch-activating ligands affects intracellular processes in c-kit+ cells when used as an external stimulus. Neonatal c-kit+ CPCs were stimulated using four different chimeric Notch-activating ligands tethered to Dynabeads, and the resulting changes were assessed using TaqMan gene expression arrays, with subsequent analysis by principal component analysis (PCA). Additionally, functional outcomes were measured using an endothelial cell tube formation assay and MSC migration assay to assess the paracrine capacity to stimulate new vessel formation and recruit other reparative cell types to the site of injury. Gene expression data showed that stimulation with Jagged-1 is associated with the greatest pro-angiogenic gene response, including the expression of VEGF and basement membrane proteins, while the other canonical ligands, Jagged-2, Dll-1, and Dll-4, are more associated with regulatory and epigenetic changes. The functional assay showed differential responses to the four ligands in terms of angiogenesis, while none of the ligands produced a robust change in migration. These data demonstrate how the four Notch-activating ligands differentially regulate CPC gene expression and function.
Collapse
Affiliation(s)
- Matthew Robeson
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA;
| | - Steven L. Goudy
- Department of Pediatric Otolaryngology, Emory University, Atlanta, GA 30322, USA;
| | - Michael E. Davis
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA;
- Children’s Heart Research and Outcomes (HeRO) Center, Emory University and Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| |
Collapse
|
5
|
Lin G, Pan W, He Y, Yi X, Zhou P, Lu J. Opportunities and Perspectives for Three Dimensional Culture of Mesenchymal Stem Cell-Derived Exosomes. TISSUE ENGINEERING. PART B, REVIEWS 2024; 30:462-476. [PMID: 38265004 DOI: 10.1089/ten.teb.2023.0253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
Exosomes are nanosized extracellular vesicles (EVs) that participate in intercellular communication through surface proteins and the delivery of internal cargo. The exosomes have gained attention for their potential as disease biomarkers and therapeutic agents. The therapeutic ability of exosomes has been verified by copious previous studies. Effective methods for extensive clinical applications are being researched for exosome-based regenerative therapies, including the application of 3D cultures to enhance exosome production and secretion, which can resolve limited exosome secretion from the parent cells. Cell culture has emerged as a crucial approach for biomedical research because of its many benefits. Both well-established continuous cell lines and primary cell cultures continue to be invaluable for basic research and clinical application. Previous studies have shown that three-dimensional cultured exosomes (3D-Exo) improve therapeutic properties and yields compared with traditional culture systems. Since the majority of studies have focused on exosomes derived from mesenchymal stem cells (MSC-Exo), this review will also concentrate on MSC-Exo. In this review, we will summarize the advantages of 3D-Exo and introduce the 3D culture system and methods of exosome isolation, providing scientific strategies for the diagnosis, treatment, and prognosis of a wide variety of diseases.
Collapse
Affiliation(s)
- Guanyi Lin
- Southern Medical University, Guangzhou, China
| | - Wennuo Pan
- Southern Medical University, Guangzhou, China
| | - Yinde He
- Southern Medical University, Guangzhou, China
| | - Xiao Yi
- Department of Obstetrics & Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, China
| | - Pengyu Zhou
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jun Lu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
6
|
Hunter JD, Mesfin JM, Ahmed T, Chen A, Reimold K, Hancko A, Braden RL, Davis ME, Christman KL. Myocardial Matrix Hydrogels Mitigate Negative Remodeling and Improve Function in Right Heart Failure Model. JACC Basic Transl Sci 2024; 9:322-338. [PMID: 38559631 PMCID: PMC10978413 DOI: 10.1016/j.jacbts.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 04/04/2024]
Abstract
This study evaluates the effectiveness of myocardial matrix (MM) hydrogels in mitigating negative right ventricular (RV) remodeling in a rat model of RV heart failure. The goal was to assess whether a hydrogel derived from either the right or left ventricle could promote cardiac repair. Injured rat right ventricles were injected with either RV-or left ventricular-derived MM hydrogels. Both hydrogels improved RV function and morphology and reduced negative remodeling. This study supports the potential of injectable biomaterial therapies for treating RV heart failure.
Collapse
Affiliation(s)
- Jervaughn D. Hunter
- Shu Chien-Gene Lay Department of Bioengineering and Sanford Consortium for Regenerative Medicine, University of California-San Diego, San Diego, California, USA
| | - Joshua M. Mesfin
- Shu Chien-Gene Lay Department of Bioengineering and Sanford Consortium for Regenerative Medicine, University of California-San Diego, San Diego, California, USA
| | - Tanzeel Ahmed
- Shu Chien-Gene Lay Department of Bioengineering and Sanford Consortium for Regenerative Medicine, University of California-San Diego, San Diego, California, USA
| | - Alexander Chen
- Shu Chien-Gene Lay Department of Bioengineering and Sanford Consortium for Regenerative Medicine, University of California-San Diego, San Diego, California, USA
| | - Kate Reimold
- Shu Chien-Gene Lay Department of Bioengineering and Sanford Consortium for Regenerative Medicine, University of California-San Diego, San Diego, California, USA
| | - Arielle Hancko
- Shu Chien-Gene Lay Department of Bioengineering and Sanford Consortium for Regenerative Medicine, University of California-San Diego, San Diego, California, USA
| | - Rebecca L. Braden
- Shu Chien-Gene Lay Department of Bioengineering and Sanford Consortium for Regenerative Medicine, University of California-San Diego, San Diego, California, USA
| | - Michael E. Davis
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Karen L. Christman
- Shu Chien-Gene Lay Department of Bioengineering and Sanford Consortium for Regenerative Medicine, University of California-San Diego, San Diego, California, USA
| |
Collapse
|
7
|
Bheri S, Park HJ, Hoffman JR, Takaesu F, Davis ME. The Effect of Parent Cell Type on Small Extracellular Vesicle-Derived Vehicle Functionality. Adv Biol (Weinh) 2024; 8:e2300462. [PMID: 38143286 PMCID: PMC11043963 DOI: 10.1002/adbi.202300462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/20/2023] [Indexed: 12/26/2023]
Abstract
Cell therapies involving c-kit+ progenitor cells (CPCs) and mesenchymal stem cells (MSCs) have been actively studied for cardiac repair. The benefits of such therapies have more recently been attributed to the release of small extracellular vesicles (sEVs) from the parent cells. These sEVs are 30-180 nm vesicles containing protein/nucleic acid cargo encapsulated within an amphiphilic bilayer membrane. Despite their pro-reparative effects, sEV composition and cargo loading is highly variable, making it challenging to develop robust therapies with sEVs. Synthetic alternatives have been developed to allow cargo modulation, including prior work from the laboratory, to design sEV-like vehicles (ELVs). ELVs are synthesized from the sEV membrane but allow controlled cargo loading. It is previously shown that loading pro-angiogenic miR-126 into CPC-derived ELVs significantly increases endothelial cell angiogenesis compared to CPC-sEVs alone. Here, they expand on this work to design MSC-derived ELVs and study the role of the parent cell type on ELV composition and function. It is found that ELV origin does affect the ELV potency and that ELV membrane composition can affect outcomes. This study showcases the versatility of ELVs to be synthesized from different parent cells and highlights the importance of selecting ELV source cells based on the desired functional outcomes.
Collapse
Affiliation(s)
- Sruti Bheri
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, 30332, USA
| | - Hyun-Ji Park
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
| | - Jessica R Hoffman
- Molecular & Systems Pharmacology Graduate Training Program, Graduate Division of Biological & Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA, 30322, USA
| | - Felipe Takaesu
- Biochemistry, Cell and Developmental Biology Graduate Training Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA, 30332, USA
| | - Michael E Davis
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, 30332, USA
- Molecular & Systems Pharmacology Graduate Training Program, Graduate Division of Biological & Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA, 30322, USA
- Biochemistry, Cell and Developmental Biology Graduate Training Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA, 30332, USA
- Children's Heart Research & Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, GA, 30322, USA
| |
Collapse
|
8
|
Hoffman JR, Park HJ, Bheri S, Platt MO, Hare JM, Kaushal S, Bettencourt JL, Lai D, Slesnick TC, Mahle WT, Davis ME. Statistical modeling of extracellular vesicle cargo to predict clinical trial outcomes for hypoplastic left heart syndrome. iScience 2023; 26:107980. [PMID: 37868626 PMCID: PMC10589850 DOI: 10.1016/j.isci.2023.107980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 08/24/2023] [Accepted: 09/15/2023] [Indexed: 10/24/2023] Open
Abstract
Cardiac-derived c-kit+ progenitor cells (CPCs) are under investigation in the CHILD phase I clinical trial (NCT03406884) for the treatment of hypoplastic left heart syndrome (HLHS). The therapeutic efficacy of CPCs can be attributed to the release of extracellular vesicles (EVs). To understand sources of cell therapy variability we took a machine learning approach: combining bulk CPC-derived EV (CPC-EV) RNA sequencing and cardiac-relevant in vitro experiments to build a predictive model. We isolated CPCs from cardiac biopsies of patients with congenital heart disease (n = 29) and the lead-in patients with HLHS in the CHILD trial (n = 5). We sequenced CPC-EVs, and measured EV inflammatory, fibrotic, angiogeneic, and migratory responses. Overall, CPC-EV RNAs involved in pro-reparative outcomes had a significant fit to cardiac development and signaling pathways. Using a model trained on previously collected CPC-EVs, we predicted in vitro outcomes for the CHILD clinical samples. Finally, CPC-EV angiogenic performance correlated to clinical improvements in right ventricle performance.
Collapse
Affiliation(s)
- Jessica R. Hoffman
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine & Georgia Institute of Technology, Atlanta, GA 30322, USA
- Molecular & Systems Pharmacology Graduate Training Program, Laney Graduate School, Emory University, Atlanta, GA 30322, USA
| | - Hyun-Ji Park
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine & Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Sruti Bheri
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine & Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Manu O. Platt
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine & Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Joshua M. Hare
- Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Sunjay Kaushal
- Departments of Surgery and Pediatrics, Ann and Robert H. Lurie Children’s Hospital of Chicago, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Judith L. Bettencourt
- Coordinating Center for Clinical Trials, Department of Biostatistics and Data Science, University of Texas Health Science Center School of Public Health, Houston, TX 77030, USA
| | - Dejian Lai
- Coordinating Center for Clinical Trials, Department of Biostatistics and Data Science, University of Texas Health Science Center School of Public Health, Houston, TX 77030, USA
| | - Timothy C. Slesnick
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Children’s Heart Research & Outcomes (HeRO) Center, Children’s Healthcare of Atlanta & Emory University, Atlanta, GA 30322, USA
| | - William T. Mahle
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Children’s Heart Research & Outcomes (HeRO) Center, Children’s Healthcare of Atlanta & Emory University, Atlanta, GA 30322, USA
| | - Michael E. Davis
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine & Georgia Institute of Technology, Atlanta, GA 30322, USA
- Molecular & Systems Pharmacology Graduate Training Program, Laney Graduate School, Emory University, Atlanta, GA 30322, USA
- Children’s Heart Research & Outcomes (HeRO) Center, Children’s Healthcare of Atlanta & Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
9
|
Bragança J, Pinto R, Silva B, Marques N, Leitão HS, Fernandes MT. Charting the Path: Navigating Embryonic Development to Potentially Safeguard against Congenital Heart Defects. J Pers Med 2023; 13:1263. [PMID: 37623513 PMCID: PMC10455635 DOI: 10.3390/jpm13081263] [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: 07/06/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023] Open
Abstract
Congenital heart diseases (CHDs) are structural or functional defects present at birth due to improper heart development. Current therapeutic approaches to treating severe CHDs are primarily palliative surgical interventions during the peri- or prenatal stages, when the heart has fully developed from faulty embryogenesis. However, earlier interventions during embryonic development have the potential for better outcomes, as demonstrated by fetal cardiac interventions performed in utero, which have shown improved neonatal and prenatal survival rates, as well as reduced lifelong morbidity. Extensive research on heart development has identified key steps, cellular players, and the intricate network of signaling pathways and transcription factors governing cardiogenesis. Additionally, some reports have indicated that certain adverse genetic and environmental conditions leading to heart malformations and embryonic death may be amendable through the activation of alternative mechanisms. This review first highlights key molecular and cellular processes involved in heart development. Subsequently, it explores the potential for future therapeutic strategies, targeting early embryonic stages, to prevent CHDs, through the delivery of biomolecules or exosomes to compensate for faulty cardiogenic mechanisms. Implementing such non-surgical interventions during early gestation may offer a prophylactic approach toward reducing the occurrence and severity of CHDs.
Collapse
Affiliation(s)
- José Bragança
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Champalimaud Research Program, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | - Rute Pinto
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| | - Bárbara Silva
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- PhD Program in Biomedical Sciences, Faculty of Medicine and Biomedical Sciences, Universidade do Algarve, 8005-139 Faro, Portugal
| | - Nuno Marques
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| | - Helena S. Leitão
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| | - Mónica T. Fernandes
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- School of Health, University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| |
Collapse
|
10
|
Williams K, Khan A, Lee YS, Hare JM. Cell-based therapy to boost right ventricular function and cardiovascular performance in hypoplastic left heart syndrome: Current approaches and future directions. Semin Perinatol 2023; 47:151725. [PMID: 37031035 PMCID: PMC10193409 DOI: 10.1016/j.semperi.2023.151725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/10/2023]
Abstract
Congenital heart disease remains one of the most frequently diagnosed congenital diseases of the newborn, with hypoplastic left heart syndrome (HLHS) being considered one of the most severe. This univentricular defect was uniformly fatal until the introduction, 40 years ago, of a complex surgical palliation consisting of multiple staged procedures spanning the first 4 years of the child's life. While survival has improved substantially, particularly in experienced centers, ventricular failure requiring heart transplant and a number of associated morbidities remain ongoing clinical challenges for these patients. Cell-based therapies aimed at boosting ventricular performance are under clinical evaluation as a novel intervention to decrease morbidity associated with surgical palliation. In this review, we will examine the current burden of HLHS and current modalities for treatment, discuss various cells therapies as an intervention while delineating challenges and future directions for this therapy for HLHS and other congenital heart diseases.
Collapse
Affiliation(s)
- Kevin Williams
- Department of Pediatrics, University of Miami Miller School of Medicine. Miami FL, USA; Batchelor Children's Research Institute University of Miami Miller School of Medicine. Miami FL, USA
| | - Aisha Khan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami FL, USA
| | - Yee-Shuan Lee
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami FL, USA
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami FL, USA; Division of Cardiology, Department of Medicine, University of Miami Miller School of Medicine. Miami FL, USA.
| |
Collapse
|
11
|
Wang B, Zhai C, Li Y, Ma B, Li Z, Wang J. Sertoli cells-derived exosomal miR-30a-5p regulates ubiquitin E3 ligase Zeb2 to affect the spermatogonial stem cells proliferation and differentiation. Reprod Toxicol 2023; 117:108340. [PMID: 36731640 DOI: 10.1016/j.reprotox.2023.108340] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/19/2023] [Accepted: 01/22/2023] [Indexed: 02/01/2023]
Abstract
The role of spermatogonial stem cells (SSCs) is crucial in spermatogenesis, and extracellular vesicles (EVs) have been the focus of research as an important intercellular communication mechanism. Various endogenous regulatory factors secreted by Sertoli cells (SCs) can affect the self-maintenance and regeneration of SSCs, but little is known about the roles of SCs-derived exosomal microRNAs (miRNAs) on SSCs. In this study, we aimed to explore the regulation of the SCs-derived exosomal miR-30a-5p on SSCs proliferation and differentiation. EVs from the SCs were detected by electron microscopy and nanoparticle tracking analysis (NTA). Subsequently, the SSCs were treated with the SCs-derived extracellular vesicles (SCs-EVs). CCK-8 assay and EdU staining was applied to detect the cell proliferation, and the results indicated that SCs-EVs promoted the SSCs proliferation. Western blot detection of the SSCs markers (Gfrα1, Plzf, Stra8, and C-kit) indicated that SCs-EVs promoted the SSCs differentiation. Additionally, we found that SCs-EVs secreted miR-30a-5p to show the promoting effects. Besides, we discovered that miR-30a-5p targeted zinc finger E-box binding homeobox 2 (Zeb2) to regulate the ubiquitination of fibroblast growth factor 9 (Fgf9) in SSCs. miR-30a-3p/Zeb2/Fgf9 promoted the SSCs proliferation and differentiation by activating the mitogen‑activated protein kinase (MAPK) signaling pathway. Taken together, our study showed that SCs-EVs can transport miR-30a-5p to SSCs and affect SSCs proliferation and differentiation by regulating the MAPK signaling pathway via Zeb2/Fgf9. This paper disclosed a novel molecular mechanism that regulates SSCs proliferation and differentiation, which could be valuable for the treatment of male infertility.
Collapse
Affiliation(s)
- Bin Wang
- Department of Urology, People's Hospital of Yuxi City, The Sixth Affiliated Hospital of Kunming Medical University, Yuxi, Yunnan, 653100, China
| | - Chengxi Zhai
- Department of Urology, People's Hospital of Yuxi City, The Sixth Affiliated Hospital of Kunming Medical University, Yuxi, Yunnan, 653100, China
| | - Yingzhong Li
- Department of Urology, People's Hospital of Yuxi City, The Sixth Affiliated Hospital of Kunming Medical University, Yuxi, Yunnan, 653100, China
| | - Bo Ma
- Department of Urology, People's Hospital of Yuxi City, The Sixth Affiliated Hospital of Kunming Medical University, Yuxi, Yunnan, 653100, China
| | - Zhu Li
- Department of Urology, People's Hospital of Yuxi City, The Sixth Affiliated Hospital of Kunming Medical University, Yuxi, Yunnan, 653100, China
| | - Jian Wang
- Department of Urology, People's Hospital of Yuxi City, The Sixth Affiliated Hospital of Kunming Medical University, Yuxi, Yunnan, 653100, China.
| |
Collapse
|
12
|
Single-Cell RNA Sequencing Reveals Distinct Cardiac-Derived Stromal Cell Subpopulations. J Cardiovasc Dev Dis 2022; 9:jcdd9110374. [DOI: 10.3390/jcdd9110374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Human cardiac-derived c-kit+ stromal cells (CSCs) have demonstrated efficacy in preclinical trials for the treatment of heart failure and myocardial dysfunction. Unfortunately, large variability in patient outcomes and cell populations remains a problem. Previous research has demonstrated that the reparative capacity of CSCs may be linked to the age of the cells: CSCs derived from neonate patients increase cardiac function and reduce fibrosis. However, age-dependent differences between CSC populations have primarily been explored with bulk sequencing methods. In this work, we hypothesized that differences in CSC populations and subsequent cell therapy outcomes may arise from differing cell subtypes within donor CSC samples. We performed single-cell RNA sequencing on four neonatal CSC (nCSC) and five child CSC (cCSC) samples. Subcluster analysis revealed cCSC-enriched clusters upregulated in several fibrosis- and immune response-related genes. Module-based analysis identified upregulation of chemotaxis and ribosomal activity-related genes in nCSCs and upregulation of immune response and fiber synthesis genes in cCSCs. Further, we identified versican and integrin alpha 2 as potential markers for a fibrotic cell subtype. By investigating differences in patient-derived CSC populations at the single-cell level, this research aims to identify and characterize CSC subtypes to better optimize CSC-based therapy and improve patient outcomes.
Collapse
|
13
|
Hoffman JR, Park HJ, Bheri S, Jayaraman AR, Davis ME. Comparative computational RNA analysis of cardiac-derived progenitor cells and their extracellular vesicles. Genomics 2022; 114:110349. [PMID: 35346780 PMCID: PMC9510608 DOI: 10.1016/j.ygeno.2022.110349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 02/18/2022] [Accepted: 03/23/2022] [Indexed: 01/14/2023]
Abstract
Stem/progenitor cells, including cardiac-derived c-kit+ progenitor cells (CPCs), are under clinical evaluation for treatment of cardiac disease. Therapeutic efficacy of cardiac cell therapy can be attributed to paracrine signaling and the release of extracellular vesicles (EVs) carrying diverse cargo molecules. Despite some successes and demonstrated safety, large variation in cell populations and preclinical/clinical outcomes remains a problem. Here, we investigated this variability by sequencing coding and non-coding RNAs of CPCs and CPC-EVs from 30 congenital heart disease patients and used machine learning methods to determine potential mechanistic insights. CPCs retained RNAs related to extracellular matrix organization and exported RNAs related to various signaling pathways to CPC-EVs. CPC-EVs are enriched in miRNA clusters related to cell proliferation and angiogenesis. With network analyses, we identified differences in non-coding RNAs which give insight into age-dependent functionality of CPCs. By taking a quantitative computational approach, we aimed to uncover sources of CPC cell therapy variability.
Collapse
Affiliation(s)
- Jessica R. Hoffman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, USA.,Molecular & Systems Pharmacology Graduate Training Program, Graduate Division of Biological & Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA 30322, USA
| | - Hyun-Ji Park
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, USA
| | - Sruti Bheri
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, USA
| | - Arun R. Jayaraman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, USA
| | - Michael E. Davis
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, USA.,Molecular & Systems Pharmacology Graduate Training Program, Graduate Division of Biological & Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA 30322, USA.,Children’s Heart Research & Outcomes (HeRO) Center, Children’s Healthcare of Atlanta & Emory University, Atlanta, Georgia, USA.,Corresponding author at: Professor of Biomedical Engineering, 2015 Uppergate Drive, 310, Atlanta, GA 30322, USA,
| |
Collapse
|
14
|
Prasad V, Makkaoui N, Rajan R, Patel A, Mainali B, Bagchi P, Kumar R, Rogers J, Diamond J, Maxwell JT. Loss of cardiac myosin light chain kinase contributes to contractile dysfunction in right ventricular pressure overload. Physiol Rep 2022; 10:e15238. [PMID: 35384363 PMCID: PMC8981447 DOI: 10.14814/phy2.15238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 03/03/2022] [Accepted: 03/06/2022] [Indexed: 06/14/2023] Open
Abstract
Nearly 1 in every 100 children born have a congenital heart defect. Many of these defects primarily affect the right heart causing pressure overload of the right ventricle (RV). The RV maintains function by adapting to the increased pressure; however, many of these adaptations eventually lead to RV hypertrophy and failure. In this study, we aim to identify the cellular and molecular mechanisms of these adaptions. We utilized a surgical animal model of pulmonary artery banding (PAB) in juvenile rats that has been shown to accurately recapitulate the physiology of right ventricular pressure overload in young hearts. Using this model, we examined changes in cardiac myocyte protein expression as a result of pressure overload with mass spectrometry 4 weeks post-banding. We found pressure overload of the RV induced significant downregulation of cardiac myosin light chain kinase (cMLCK). Single myocyte calcium and contractility recordings showed impaired contraction and relaxation in PAB RV myocytes, consistent with the loss of cMLCK. In the PAB myocytes, calcium transients were of smaller amplitude and decayed at a slower rate compared to controls. We also identified miR-200c, which has been shown to regulate cMLCK expression, as upregulated in the RV in response to pressure overload. These results indicate the loss of cMLCK is a critical maladaptation of the RV to pressure overload and represents a novel target for therapeutic approaches to treat RV hypertrophy and failure associated with congenital heart defects.
Collapse
Affiliation(s)
- Vidhya Prasad
- Division of Pediatric CardiologyDepartment of PediatricsEmory University School of MedicineAtlantaGeorgiaUSA
- Children’s Heart Research & Outcomes (HeRO) CenterChildren’s Healthcare of Atlanta & Emory UniversityAtlantaGeorgiaUSA
| | - Nour Makkaoui
- Division of Pediatric CardiologyDepartment of PediatricsEmory University School of MedicineAtlantaGeorgiaUSA
- Children’s Heart Research & Outcomes (HeRO) CenterChildren’s Healthcare of Atlanta & Emory UniversityAtlantaGeorgiaUSA
| | - Rohan Rajan
- Emory University College of Arts and SciencesAtlantaGeorgiaUSA
| | - Alisha Patel
- Emory University College of Arts and SciencesAtlantaGeorgiaUSA
| | - Bipul Mainali
- Emory University College of Arts and SciencesAtlantaGeorgiaUSA
| | - Pritha Bagchi
- Department of BiochemistryEmory University School of MedicineAtlantaGeorgiaUSA
| | - Rhea Kumar
- Emory University College of Arts and SciencesAtlantaGeorgiaUSA
| | - Julia Rogers
- Emory University College of Arts and SciencesAtlantaGeorgiaUSA
| | - Jake Diamond
- Emory University College of Arts and SciencesAtlantaGeorgiaUSA
| | - Joshua T. Maxwell
- Division of Pediatric CardiologyDepartment of PediatricsEmory University School of MedicineAtlantaGeorgiaUSA
- Children’s Heart Research & Outcomes (HeRO) CenterChildren’s Healthcare of Atlanta & Emory UniversityAtlantaGeorgiaUSA
| |
Collapse
|
15
|
Gao L, Li X, Tan R, Cui J, Schmull S. Human-derived decellularized extracellular matrix scaffold incorporating autologous bone marrow stem cells from patients with congenital heart disease for cardiac tissue engineering. Biomed Mater Eng 2022; 33:407-421. [PMID: 35180106 DOI: 10.3233/bme-211368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Stem cells are used as an alternative treatment option for patients with congenital heart disease (CHD) due to their regenerative potential, but they are subject to low retention rate in the injured myocardium. Also, the diseased microenvironment in the injured myocardium may not provide healthy cues for optimal stem cell function. OBJECTIVE In this study, we prepared a novel human-derived cardiac scaffold to improve the functional behaviors of stem cells. METHODS Decellularized extracellular matrix (ECM) scaffolds were fabricated by removing cells of human-derived cardiac appendage tissues. Then, bone marrow c-kit+ progenitor cells from patients with congenital heart disease were seeded on the cardiac ECM scaffolds. Cell adhesion, survival, proliferation and cardiac differentiation on human cardiac decellularized ECM scaffold were evaluated in vitro. Label-free mass spectrometry was applied to analyze cardiac ECM proteins regulating cell behaviors. RESULTS It was shown that cardiac ECM scaffolds promoted stem cell adhesion and proliferation. Importantly, bone marrow c-kit+ progenitor cells cultured on cardiac ECM scaffold for 14 days differentiated into cardiomyocyte-like cells without supplement with any inducible factors, as confirmed by the increased protein level of Gata4 and upregulated gene levels of Gata4, Nkx2.5, and cTnT. Proteomic analysis showed the proteins in cardiac ECM functioned in multiple biological activities, including regulation of cell proliferation, regulation of cell differentiation, and cardiovascular system development. CONCLUSION The human-derived cardiac scaffold constructed in this study may help repair the damaged myocardium and hold great potential for tissue engineering application in pediatric patients with CHD.
Collapse
Affiliation(s)
- Liping Gao
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu, China.,National Demonstration Center for Experiment Basic Medical Science Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xuexia Li
- Department of Endocrinology, Xuzhou Cancer Hospital, Xuzhou, Jiangsu, China
| | - Rubin Tan
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu, China.,National Demonstration Center for Experiment Basic Medical Science Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jie Cui
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu, China.,National Demonstration Center for Experiment Basic Medical Science Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Sebastian Schmull
- Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
16
|
Vermeulen S, Birgani ZT, Habibovic P. Biomaterial-induced pathway modulation for bone regeneration. Biomaterials 2022; 283:121431. [DOI: 10.1016/j.biomaterials.2022.121431] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/28/2022] [Accepted: 02/17/2022] [Indexed: 12/18/2022]
|
17
|
Streeter BW, Brown ME, Shakya P, Park HJ, Qiu J, Xia Y, Davis ME. Using computational methods to design patient-specific electrospun cardiac patches for pediatric heart failure. Biomaterials 2022; 283:121421. [DOI: 10.1016/j.biomaterials.2022.121421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 01/12/2022] [Accepted: 02/17/2022] [Indexed: 12/15/2022]
|
18
|
Bejleri D, Robeson MJ, Brown ME, Hunter J, Maxwell JT, Streeter BW, Brazhkina O, Park HJ, Christman KL, Davis ME. In vivo evaluation of bioprinted cardiac patches composed of cardiac-specific extracellular matrix and progenitor cells in a model of pediatric heart failure. Biomater Sci 2022; 10:444-456. [PMID: 34878443 PMCID: PMC8772587 DOI: 10.1039/d1bm01539g] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Pediatric patients with congenital heart defects (CHD) often present with heart failure from increased load on the right ventricle (RV) due to both surgical methods to treat CHD and the disease itself. Patients with RV failure often require transplantation, which is limited due to lack of donor availability and rejection. Previous studies investigating the development and in vitro assessment of a bioprinted cardiac patch composed of cardiac extracellular matrix (cECM) and human c-kit + progenitor cells (hCPCs) showed that the construct has promise in treating cardiac dysfunction. The current study investigates in vivo cardiac outcomes of patch implantation in a rat model of RV failure. Patch parameters including cECM-inclusion and hCPC-inclusion are investigated. Assessments include hCPC retention, RV function, and tissue remodeling (vascularization, hypertrophy, and fibrosis). Animal model evaluation shows that both cell-free and neonatal hCPC-laden cECM-gelatin methacrylate (GelMA) patches improve RV function and tissue remodeling compared to other patch groups and controls. Inclusion of cECM is the most influential parameter driving therapeutic improvements, with or without cell inclusion. This study paves the way for clinical translation in treating pediatric heart failure using bioprinted GelMA-cECM and hCPC-GelMA-cECM patches.
Collapse
Affiliation(s)
- Donald Bejleri
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 1760 Haygood Dr, Atlanta, GA, 30322, USA.
| | - Matthew J Robeson
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 1760 Haygood Dr, Atlanta, GA, 30322, USA.
| | - Milton E Brown
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 1760 Haygood Dr, Atlanta, GA, 30322, USA.
| | - Jervaughn Hunter
- Department of Bioengineering and Sanford Consortium for Regenerative Medicine, University of California, San Diego, 2880 Torrey Pines Scenic Dr, La Jolla, CA, 92037, USA
| | - Joshua T Maxwell
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Dr, Atlanta, GA, 30322, USA
| | - Benjamin W Streeter
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 1760 Haygood Dr, Atlanta, GA, 30322, USA.
| | - Olga Brazhkina
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 1760 Haygood Dr, Atlanta, GA, 30322, USA.
| | - Hyun-Ji Park
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 1760 Haygood Dr, Atlanta, GA, 30322, USA.
| | - Karen L Christman
- Department of Bioengineering and Sanford Consortium for Regenerative Medicine, University of California, San Diego, 2880 Torrey Pines Scenic Dr, La Jolla, CA, 92037, USA
| | - Michael E Davis
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 1760 Haygood Dr, Atlanta, GA, 30322, USA.
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Dr, Atlanta, GA, 30322, USA
| |
Collapse
|
19
|
Shakya P, Brown ME, Davis ME. Encapsulation of Pediatric Cardiac-Derived C-Kit + Cells in Cardiac Extracellular Matrix Hydrogel for Echocardiography-Directed Intramyocardial Injection in Rodents. Methods Mol Biol 2022; 2485:269-278. [PMID: 35618912 DOI: 10.1007/978-1-0716-2261-2_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Pediatric cardiac-derived c-kit+ cell therapies represent an innovative approach for cardiac tissue repair that have demonstrated promising improvements in recent studies and offer multiple benefits, such as easy isolation and autologous transplant. However, concerns about failure of engraftment and transient paracrine effects have thus far limited their use. To overcome these issues, an appropriate cell delivery vehicle such as a cardiac extracellular matrix (cECM) hydrogel can be utilized. This naturally derived biomaterial can support embedded cells, allowing for local diffusion of paracrine factors, and provide a healthy microenvironment for optimal cellular function. This protocol focuses on combining cardiac-derived c-kit+ cells and a cECM hydrogel to prepare a minimally invasive, dual therapeutic for in vivo delivery. We also outline a detailed method for ultrasound-guided intramyocardial injection of cell-laden hydrogels in a rodent model. Additional steps for labeling cells with a fluorescent dye for in vivo cell tracking are provided.
Collapse
Affiliation(s)
- Preety Shakya
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
| | - Milton E Brown
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Michael E Davis
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA.
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.
- Children's Heart Research and Outcomes (HeRO) Center, Children's Healthcare of Atlanta and Emory University, Atlanta, GA, USA.
| |
Collapse
|
20
|
Yu W, Li S, Guan X, Zhang N, Xie X, Zhang K, Bai Y. Higher yield and enhanced therapeutic effects of exosomes derived from MSCs in hydrogel-assisted 3D culture system for bone regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2022; 133:112646. [DOI: 10.1016/j.msec.2022.112646] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 12/09/2021] [Accepted: 01/03/2022] [Indexed: 02/07/2023]
|
21
|
Zhong X, Wang T, Xie Y, Wang M, Zhang W, Dai L, Lai J, Nie X, He X, Madhusudhan T, Zeng H, Wang H. Activated Protein C Ameliorates Diabetic Cardiomyopathy via Modulating OTUB1/YB-1/MEF2B Axis. Front Cardiovasc Med 2021; 8:758158. [PMID: 34778410 PMCID: PMC8585767 DOI: 10.3389/fcvm.2021.758158] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/04/2021] [Indexed: 12/12/2022] Open
Abstract
Aims: The pathogenesis of diabetic cardiomyopathy (DCM) is complex and the detailed mechanism remains unclear. Coagulation protease activated Protein C (aPC) has been reported to have a protective effect in diabetic microvascular disease. Here, we investigated whether aPC could play a protective role in the occurrence and development of major diabetic complication DCM, and its underlying molecular mechanism. Methods and Results: In a mouse model of streptozotocin (STZ) induced DCM, endogenous aPC levels were reduced. Restoring aPC levels by exogenous administration of zymogen protein C (PC) improved cardiac function of diabetic mice measured by echocardiography and invasive hemodynamics. The cytoprotective effect of aPC in DCM is mediated by transcription factor Y-box binding protein-1 (YB-1). Mechanistically, MEF2B lies downstream of YB-1 and YB-1/MEF2B interaction restrains deleterious MEF2B promoter activity in DCM. The regulation of YB-1 on MEF2B transcription was analyzed by dual-luciferase and chromatin immunoprecipitation assays. In diabetic mice, aPC ameliorated YB-1 degradation via reducing its K48 ubiquitination through deubiquitinating enzyme otubain-1 (OTUB1) and improving the interaction between YB-1 and OTUB1. Using specific agonists and blocking antibodies, PAR1 and EPCR were identified as crucial receptors for aPC's dependent cytoprotective signaling. Conclusion: These data identify that the cytoprotective aPC signaling via PAR1/EPCR maintains YB-1 levels by preventing the ubiquitination and subsequent proteasomal degradation of YB-1 via OTUB1. By suppressing MEF2B transcription, YB-1 can protect against DCM. Collectively, the current study uncovered the important role of OTUB1/YB-1/MEF2B axis in DCM and targeting this pathway might offer a new therapeutic strategy for DCM. Translational Perspective: DCM is emerging at epidemic rate recently and the underlying mechanism remains unclear. This study explored the protective cell signaling mechanisms of aPC in mouse models of DCM. As a former FDA approved anti-sepsis drug, aPC along with its derivatives can be applied from bench to bed and can be explored as a new strategy for personalized treatment for DCM. Mechanistically, OTUB1/YB-1/MEF2B axis plays a critical role in the occurrence and development of DCM and offers a potential avenue for therapeutic targeting of DCM.
Collapse
Affiliation(s)
- Xiaodan Zhong
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Tao Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Yang Xie
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Mengwen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Wenjun Zhang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Lei Dai
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Jinsheng Lai
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Xiang Nie
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Xingwei He
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Thati Madhusudhan
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Hesong Zeng
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Hongjie Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| |
Collapse
|
22
|
Gomez AH, Joshi S, Yang Y, Tune JD, Zhao MT, Yang H. Bioengineering Systems for Modulating Notch Signaling in Cardiovascular Development, Disease, and Regeneration. J Cardiovasc Dev Dis 2021; 8:125. [PMID: 34677194 PMCID: PMC8541010 DOI: 10.3390/jcdd8100125] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/27/2021] [Accepted: 09/27/2021] [Indexed: 12/13/2022] Open
Abstract
The Notch intercellular signaling pathways play significant roles in cardiovascular development, disease, and regeneration through modulating cardiovascular cell specification, proliferation, differentiation, and morphogenesis. The dysregulation of Notch signaling leads to malfunction and maldevelopment of the cardiovascular system. Currently, most findings on Notch signaling rely on animal models and a few clinical studies, which significantly bottleneck the understanding of Notch signaling-associated human cardiovascular development and disease. Recent advances in the bioengineering systems and human pluripotent stem cell-derived cardiovascular cells pave the way to decipher the role of Notch signaling in cardiovascular-related cells (endothelial cells, cardiomyocytes, smooth muscle cells, fibroblasts, and immune cells), and intercellular crosstalk in the physiological, pathological, and regenerative context of the complex human cardiovascular system. In this review, we first summarize the significant roles of Notch signaling in individual cardiac cell types. We then cover the bioengineering systems of microfluidics, hydrogel, spheroid, and 3D bioprinting, which are currently being used for modeling and studying Notch signaling in the cardiovascular system. At last, we provide insights into ancillary supports of bioengineering systems, varied types of cardiovascular cells, and advanced characterization approaches in further refining Notch signaling in cardiovascular development, disease, and regeneration.
Collapse
Affiliation(s)
- Angello Huerta Gomez
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207, USA; (A.H.G.); (S.J.); (Y.Y.)
| | - Sanika Joshi
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207, USA; (A.H.G.); (S.J.); (Y.Y.)
- Texas Academy of Mathematics and Science, University of North Texas, Denton, TX 76201, USA
| | - Yong Yang
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207, USA; (A.H.G.); (S.J.); (Y.Y.)
| | - Johnathan D. Tune
- Department of Physiology & Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA;
| | - Ming-Tao Zhao
- Center for Cardiovascular Research, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43215, USA;
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH 43215, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Huaxiao Yang
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207, USA; (A.H.G.); (S.J.); (Y.Y.)
| |
Collapse
|
23
|
Park HJ, De Jesus Morales KJ, Bheri S, Kassouf BP, Davis ME. Bidirectional relationship between cardiac extracellular matrix and cardiac cells in ischemic heart disease. Stem Cells 2021; 39:1650-1659. [PMID: 34480804 DOI: 10.1002/stem.3445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 08/10/2021] [Indexed: 11/07/2022]
Abstract
Ischemic heart diseases (IHDs), including myocardial infarction and cardiomyopathies, are a leading cause of mortality and morbidity worldwide. Cardiac-derived stem and progenitor cells have shown promise as a therapeutic for IHD but are limited by poor cell survival, limited retention, and rapid washout. One mechanism to address this is to encapsulate the cells in a matrix or three-dimensional construct, so as to provide structural support and better mimic the cells' physiological microenvironment during administration. More specifically, the extracellular matrix (ECM), the native cellular support network, has been a strong candidate for this purpose. Moreover, there is a strong consensus that the ECM and its residing cells, including cardiac stem cells, have a constant interplay in response to tissue development, aging, disease progression, and repair. When externally stimulated, the cells and ECM work together to mutually maintain the local homeostasis by initially altering the ECM composition and stiffness, which in turn alters the cellular response and behavior. Given this constant interplay, understanding the mechanism of bidirectional cell-ECM interaction is essential to develop better cell implantation matrices to enhance cell engraftment and cardiac tissue repair. This review summarizes current understanding in the field, elucidating the signaling mechanisms between cardiac ECM and residing cells in response to IHD onset. Furthermore, this review highlights recent advances in native ECM-mimicking cardiac matrices as a platform for modulating cardiac cell behavior and inducing cardiac repair.
Collapse
Affiliation(s)
- Hyun-Ji Park
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine & Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Kenneth J De Jesus Morales
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine & Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Sruti Bheri
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine & Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Brandon P Kassouf
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine & Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Michael E Davis
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine & Georgia Institute of Technology, Atlanta, Georgia, USA.,Children's Heart Research and Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, Georgia, USA
| |
Collapse
|
24
|
Ali MK, Ichimura K, Spiekerkoetter E. Promising therapeutic approaches in pulmonary arterial hypertension. Curr Opin Pharmacol 2021; 59:127-139. [PMID: 34217109 DOI: 10.1016/j.coph.2021.05.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/12/2021] [Accepted: 05/11/2021] [Indexed: 12/19/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a debilitating multifactorial disease characterized by progressive pulmonary vascular remodeling, elevated pulmonary arterial pressure, and pulmonary vascular resistance, resulting in right ventricular failure and subsequent death. Current available therapies do not reverse the disease, resulting in a persistent high morbidity and mortality. Thus, there is an urgent unmet medical need for novel effective therapies to better treat patients with PAH. Over the past few years, enthusiastic attempts have been made to identify novel effective therapies that address the essential roots of PAH with targeting key signaling pathways in both preclinical models and patients with PAH. This review aims to discuss the most emerging and promising therapeutic interventions in PAH pathogenesis.
Collapse
Affiliation(s)
- Md Khadem Ali
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford Medical School, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, USA
| | - Kenzo Ichimura
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford Medical School, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, USA
| | - Edda Spiekerkoetter
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford Medical School, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, USA.
| |
Collapse
|
25
|
Klinke A, Schubert T, Müller M, Legchenko E, Zelt JGE, Shimauchi T, Napp LC, Rothman AMK, Bonnet S, Stewart DJ, Hansmann G, Rudolph V. Emerging therapies for right ventricular dysfunction and failure. Cardiovasc Diagn Ther 2020; 10:1735-1767. [PMID: 33224787 PMCID: PMC7666928 DOI: 10.21037/cdt-20-592] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/27/2020] [Indexed: 12/17/2022]
Abstract
Therapeutic options for right ventricular (RV) dysfunction and failure are strongly limited. Right heart failure (RHF) has been mostly addressed in the context of pulmonary arterial hypertension (PAH), where it is not possible to discern pulmonary vascular- and RV-directed effects of therapeutic approaches. In part, opposing pathomechanisms in RV and pulmonary vasculature, i.e., regarding apoptosis, angiogenesis and proliferation, complicate addressing RHF in PAH. Therapy effective for left heart failure is not applicable to RHF, e.g., inhibition of adrenoceptor signaling and of the renin-angiotensin system had no or only limited success. A number of experimental studies employing animal models for PAH or RV dysfunction or failure have identified beneficial effects of novel pharmacological agents, with most promising results obtained with modulators of metabolism and reactive oxygen species or inflammation, respectively. In addition, established PAH agents, in particular phosphodiesterase-5 inhibitors and soluble guanylate cyclase stimulators, may directly address RV integrity. Promising results are furthermore derived with microRNA (miRNA) and long non-coding RNA (lncRNA) blocking or mimetic strategies, which can target microvascular rarefaction, inflammation, metabolism or fibrotic and hypertrophic remodeling in the dysfunctional RV. Likewise, pre-clinical data demonstrate that cell-based therapies using stem or progenitor cells have beneficial effects on the RV, mainly by improving the microvascular system, however clinical success will largely depend on delivery routes. A particular option for PAH is targeted denervation of the pulmonary vasculature, given the sympathetic overdrive in PAH patients. Finally, acute and durable mechanical circulatory support are available for the right heart, which however has been tested mostly in RHF with concomitant left heart disease. Here, we aim to review current pharmacological, RNA- and cell-based therapeutic options and their potential to directly target the RV and to review available data for pulmonary artery denervation and mechanical circulatory support.
Collapse
Affiliation(s)
- Anna Klinke
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Torben Schubert
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Marion Müller
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Ekaterina Legchenko
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover, Germany
| | - Jason G. E. Zelt
- Division of Cardiology, University of Ottawa Heart Institute and the Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Canada
| | - Tsukasa Shimauchi
- Pulmonary Hypertension Research Group, Centre de recherche de IUCPQ/Laval University, Quebec, Canada
| | - L. Christian Napp
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | | | - Sébastien Bonnet
- Pulmonary Hypertension Research Group, Centre de recherche de IUCPQ/Laval University, Quebec, Canada
| | - Duncan J. Stewart
- Division of Cardiology, University of Ottawa Heart Institute and the Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Canada
| | - Georg Hansmann
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover, Germany
| | - Volker Rudolph
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| |
Collapse
|
26
|
Monsanto MM, Wang BJ, Ehrenberg ZR, Echeagaray O, White KS, Alvarez R, Fisher K, Sengphanith S, Muliono A, Gude NA, Sussman MA. Enhancing myocardial repair with CardioClusters. Nat Commun 2020; 11:3955. [PMID: 32769998 PMCID: PMC7414230 DOI: 10.1038/s41467-020-17742-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 07/14/2020] [Indexed: 12/15/2022] Open
Abstract
Cellular therapy to treat heart failure is an ongoing focus of intense research, but progress toward structural and functional recovery remains modest. Engineered augmentation of established cellular effectors overcomes impediments to enhance reparative activity. Such 'next generation' implementation includes delivery of combinatorial cell populations exerting synergistic effects. Concurrent isolation and expansion of three distinct cardiac-derived interstitial cell types from human heart tissue, previously reported by our group, prompted design of a 3D structure that maximizes cellular interaction, allows for defined cell ratios, controls size, enables injectability, and minimizes cell loss. Herein, mesenchymal stem cells (MSCs), endothelial progenitor cells (EPCs) and c-Kit+ cardiac interstitial cells (cCICs) when cultured together spontaneously form scaffold-free 3D microenvironments termed CardioClusters. scRNA-Seq profiling reveals CardioCluster expression of stem cell-relevant factors, adhesion/extracellular-matrix molecules, and cytokines, while maintaining a more native transcriptome similar to endogenous cardiac cells. CardioCluster intramyocardial delivery improves cell retention and capillary density with preservation of cardiomyocyte size and long-term cardiac function in a murine infarction model followed 20 weeks. CardioCluster utilization in this preclinical setting establish fundamental insights, laying the framework for optimization in cell-based therapeutics intended to mitigate cardiomyopathic damage.
Collapse
Affiliation(s)
- Megan M Monsanto
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Bingyan J Wang
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Zach R Ehrenberg
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Oscar Echeagaray
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Kevin S White
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Roberto Alvarez
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Kristina Fisher
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Sharon Sengphanith
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Alvin Muliono
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Natalie A Gude
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Mark A Sussman
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA.
| |
Collapse
|
27
|
Martinez J, Zoretic S, Moreira A, Moreira A. Safety and efficacy of cell therapies in pediatric heart disease: a systematic review and meta-analysis. Stem Cell Res Ther 2020; 11:272. [PMID: 32641168 PMCID: PMC7341627 DOI: 10.1186/s13287-020-01764-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/04/2020] [Accepted: 06/08/2020] [Indexed: 12/09/2022] Open
Abstract
BACKGROUND Adult clinical trials have reported safety and the therapeutic potential of stem cells for cardiac disease. These observations have now translated to the pediatric arena. We conducted a meta-analysis to assess safety and efficacy of cell-based therapies in animal and human studies of pediatric heart disease. METHODS AND RESULTS A literature search was conducted to examine the effects of cell-based therapies on: (i) safety and (ii) cardiac function. In total, 18 pre-clinical and 13 human studies were included. Pre-clinical: right ventricular dysfunction was the most common animal model (80%). Cardiac-derived (28%) and umbilical cord blood (24%) cells were delivered intravenously (36%) or intramyocardially (35%). Mortality was similar between cell-based and control groups (OR 0.94; 95% CI 0.05, 17.41). Cell-based treatments preserved ejection fraction by 6.9% (p < 0.01), while intramyocardial at a dose of 1-10 M cells/kg optimized ejection fraction. Clinical: single ventricle physiology was the most common cardiac disease (n = 9). Cardiac tissue was a frequent cell source, dosed from 3.0 × 105 to 2.4 × 107 cells/kg. A decrease in adverse events occurred in the cell-based cohort (OR 0.17, p < 0.01). Administration of cell-based therapies improved ejection fraction (MD 4.84; 95% CI 1.62, 8.07; p < 0.01). CONCLUSIONS In this meta-analysis, cell-based therapies were safe and improved specific measures of cardiac function. Implications from this review may provide methodologic recommendations (source, dose, route, timing) for future clinical trials. Of note, many of the results described in this study pattern those seen in adult stem cell reviews and meta-analyses.
Collapse
Affiliation(s)
- John Martinez
- Department of Pediatrics, University of Texas Health San Antonio, San Antonio, TX, 77229, USA
| | - Sarah Zoretic
- Department of Pediatrics, University of Texas Health San Antonio, San Antonio, TX, 77229, USA
| | - Axel Moreira
- Department of Pediatrics, University of Texas Health San Antonio, San Antonio, TX, 77229, USA
- Department of Pediatrics, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Alvaro Moreira
- Department of Pediatrics, University of Texas Health San Antonio, San Antonio, TX, 77229, USA.
- Department of Pediatrics, UT Health San Antonio, 7703 Floyd Curl Drive, MC 7812, San Antonio, TX, 78229, USA.
| |
Collapse
|
28
|
Abstract
Right-sided heart failure (RHF) occurs from impaired contractility of the right ventricle caused by pressure, volume overload, or intrinsic myocardial contractile dysfunction. The development of subclinical right ventricle (RV) dysfunction or overt RHF is a negative prognostic indicator. Recent attention has focused on RV-specific inflammatory growth factors and mediators of myocardial fibrosis to elucidate the mechanisms leading to RHF and potentially guide the development of novel therapeutics. This article focuses on the distinct changes in RV structure, mechanics, and function, as well as molecular and inflammatory mediators involved in the pathophysiology of acute and chronic RHF.
Collapse
Affiliation(s)
| | - Kalyan R Chitturi
- Houston Methodist DeBakey Heart & Vascular Center, 6550 Fannin Street, Houston, TX 77030, USA
| | - Ashrith Guha
- Houston Methodist DeBakey Heart & Vascular Center, 6550 Fannin Street, Houston, TX 77030, USA.
| |
Collapse
|
29
|
Xuan W, Khan M, Ashraf M. Extracellular Vesicles From Notch Activated Cardiac Mesenchymal Stem Cells Promote Myocyte Proliferation and Neovasculogenesis. Front Cell Dev Biol 2020; 8:11. [PMID: 32154243 PMCID: PMC7047205 DOI: 10.3389/fcell.2020.00011] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 01/10/2020] [Indexed: 01/08/2023] Open
Abstract
Cardiac mesenchymal stem cells (C-MSCs) are a novel mesenchymal stem cell (MSC) subpopulation derived from cardiac tissue, which are reported to be responsible for cardiac regeneration. Notch signaling is believed to aid in cardiac repair following myocardial injury. In this study, we have investigated the role of extracellular vesicles (EVs) from Notch1 engineered C-MSCs on angiogenesis and cardiomyocyte (CM) proliferation in ischemic myocardium. C-MSCs were isolated from Notch1flox mice (C-MSCNotch1 FF). Notch1 gene deletion was accomplished by adenoviral vector-mediated Cre recombination, and Notch1 overexpression was achieved by overexpression of Notch1 intracellular domain (N1ICD). EVs were isolated by using the size exclusion column method. Proteomic composition of EV was carried out by mass spectrometry. A mouse myocardial infarction (MI) model was generated by permanent left anterior descending (LAD) coronary artery ligation. Intramyocardial transplantation of Notch1 knockout C-MSCs (C-MSCsNotch1 KO) did not have any effect on cardiac function and scar size. On the other hand, transplantation of N1ICD-overexpressing C-MSCs (C-MSCsN1ICD) showed significant improvement in cardiac function and attenuation of fibrosis as compared to the control (PBS) group and non-modified C-MSC groups. C-MSCsN1ICD differentiated into smooth muscle cells and formed new vessels. Proteomics profiling identified several proteins, such as lysyl oxidase homolog-2 and biglycan, as highly enriched proteins in EV-C-MSCsN1ICD. Go term analysis indicated that EV-C-MSCsN1ICD were enriched with bioactive factors, potent pro-repair proteins responsible for cell migration and proliferation. EV-C-MSCsNotch1FF and EV-C-MSCsN1ICD were strongly proangiogenic under both in vitro and in vivo conditions. EV-C-MSCsN1ICD caused dense tube formation in vitro and increased neovasculogenesis in the peri-infarct area in vivo. Furthermore, EV-C-MSCsN1ICD attenuated endothelial cell (EC) and CM apoptosis under oxidative stress and ischemic injury. Similarly, EV-C-MSCNotch1 FF and EV-C-MSCN1ICD treatment improved cardiac function and decreased fibrosis in mice post-MI. EV-C-MSCsN1ICD were very effective in improving cardiac function and decreasing fibrosis. Notch1 signaling is a strong stimulus for cardiac regeneration by C-MSCs. EVs secreted by Notch1-overexpressing C-MSCs were highly effective in preventing cell death, promoting angiogenesis and CM proliferation, and restoring cardiac function post-MI. Overall, these results suggest that Notch1 overexpression may further enhance the effectiveness of EVs secreted by C-MSCs in cell-free therapy.
Collapse
Affiliation(s)
- Wanling Xuan
- Vascular Biology Center, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Mahmood Khan
- Department of Emergency Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Muhammad Ashraf
- Vascular Biology Center, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
30
|
Bejoy J, Bijonowski B, Marzano M, Jeske R, Ma T, Li Y. Wnt-Notch Signaling Interactions During Neural and Astroglial Patterning of Human Stem Cells. Tissue Eng Part A 2019; 26:419-431. [PMID: 31686622 DOI: 10.1089/ten.tea.2019.0202] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The human brain formation involves complicated processing, which is regulated by a gene regulatory network influenced by different signaling pathways. The cross-regulatory interactions between elements of different pathways affect the process of cell fate assignment during neural and astroglial tissue patterning. In this study, the interactions between Wnt and Notch pathways, the two major pathways that influence neural and astroglial differentiation of human induced pluripotent stem cells (hiPSCs) individually, were investigated. In particular, the synergistic effects of Wnt-Notch pathway on the neural patterning processes along the anterior-posterior or dorsal-ventral axis of hiPSC-derived cortical spheroids were explored. The human cortical spheroids derived from hiPSCs were treated with Wnt activator CHIR99021 (CHIR), Wnt inhibitor IWP4, and Notch inhibitor (N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester [DAPT]) individually, or in combinations (CHIR + DAPT, IWP4 + DAPT). The results suggest that CHIR + DAPT can promote Notch signaling, similar or higher than CHIR alone, whereas IWP4 + DAPT reduces Notch activity compared to IWP4 alone. Also, CHIR + DAPT promoted hindbrain marker HOXB4 expression more consistently than CHIR alone, while IWP4 + DAPT promoted Olig2 expression, indicating the synergistic effects distinctly different from that of the individual small molecule. In addition, IWP4 simultaneously promoted dorsal and ventral identity. The patterned neural spheroids can be switched for astroglial differentiation using bone morphogenetic protein 4. This study should advance the derivations of neurons, astroglial cells, and brain region-specific organoids from hiPSCs for disease modeling, drug screening, as well as for hiPSC-based therapies. Impact Statement Wnt signaling plays a central role in neural patterning of human pluripotent stem cells. It can interact with Notch signaling in defining dorsal-ventral and rostral-caudal (or anterior-posterior) axis of brain organoids. This study investigates novel Wnt and Notch interactions (i.e., Wntch) in neural patterning of dorsal forebrain spheroids or organoids derived from human induced pluripotent stem cells. The synergistic effects of Wnt activator or inhibitor with Notch inhibitor were observed. This study should advance the derivations of neurons, astroglial cells, and brain region-specific organoids from human stem cells for disease modeling and drug screening, as well as for stem cell-based therapies. The results can be used to establish better in vitro culture methods for efficiently mimicking in vivo structure of central nervous system.
Collapse
Affiliation(s)
- Julie Bejoy
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida
| | - Brent Bijonowski
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida
| | - Mark Marzano
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida
| | - Richard Jeske
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida
| | - Teng Ma
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida
| |
Collapse
|
31
|
Murphy JF, Mayourian J, Stillitano F, Munawar S, Broughton KM, Agullo-Pascual E, Sussman MA, Hajjar RJ, Costa KD, Turnbull IC. Adult human cardiac stem cell supplementation effectively increases contractile function and maturation in human engineered cardiac tissues. Stem Cell Res Ther 2019; 10:373. [PMID: 31801634 PMCID: PMC6894319 DOI: 10.1186/s13287-019-1486-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 10/25/2019] [Accepted: 11/05/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Delivery of stem cells to the failing heart is a promising therapeutic strategy. However, the improvement in cardiac function in animal studies has not fully translated to humans. To help bridge the gap between species, we investigated the effects of adult human cardiac stem cells (hCSCs) on contractile function of human engineered cardiac tissues (hECTs) as a species-specific model of the human myocardium. METHODS Human induced pluripotent stem cell-derived cardiomyoctes (hCMs) were mixed with Collagen/Matrigel to fabricate control hECTs, with an experimental group of hCSC-supplemented hECT fabricated using a 9:1 ratio of hCM to hCSC. Functional testing was performed starting on culture day 6, under spontaneous conditions and also during electrical pacing from 0.25 to 1.0 Hz, measurements repeated at days 8 and 10. hECTs were then frozen and processed for gene analysis using a Nanostring assay with a cardiac targeted custom panel. RESULTS The hCSC-supplemented hECTs displayed a twofold higher developed force vs. hCM-only controls by day 6, with approximately threefold higher developed stress and maximum rates of contraction and relaxation during pacing at 0.75 Hz. The spontaneous beat rate characteristics were similar between groups, and hCSC supplementation did not adversely impact beat rate variability. The increased contractility persisted through days 8 and 10, albeit with some decrease in the magnitude of the difference of the force by day 10, but with developed stress still significantly higher in hCSC-supplemented hECT; these findings were confirmed with multiple hCSC and hCM cell lines. The force-frequency relationship, while negative for both, control (- 0.687 Hz- 1; p = 0.013 vs. zero) and hCSC-supplemented (- 0.233 Hz- 1;p = 0.067 vs. zero) hECTs, showed a significant rectification in the regression slope in hCSC-supplemented hECT (p = 0.011 vs. control). Targeted gene exploration (59 genes) identified a total of 14 differentially expressed genes, with increases in the ratios of MYH7/MHY6, MYL2/MYL7, and TNNI3/TNNI1 in hCSC-supplemented hECT versus controls. CONCLUSIONS For the first time, hCSC supplementation was shown to significantly improve human cardiac tissue contractility in vitro, without evidence of proarrhythmic effects, and was associated with increased expression of markers of cardiac maturation. These findings provide new insights about adult cardiac stem cells as contributors to functional improvement of human myocardium.
Collapse
Affiliation(s)
- Jack F Murphy
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Pl, Box 1030, New York, NY, 10029, USA
| | - Joshua Mayourian
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Pl, Box 1030, New York, NY, 10029, USA
| | - Francesca Stillitano
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Pl, Box 1030, New York, NY, 10029, USA
| | - Sadek Munawar
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Pl, Box 1030, New York, NY, 10029, USA
| | | | | | - Mark A Sussman
- San Diego Heart Research Institute, San Diego State University, San Diego, USA
| | | | - Kevin D Costa
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Pl, Box 1030, New York, NY, 10029, USA
| | - Irene C Turnbull
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Pl, Box 1030, New York, NY, 10029, USA.
| |
Collapse
|
32
|
Maxwell JT, Trac D, Shen M, Brown ME, Davis ME, Chao MS, Supapannachart KJ, Zaladonis CA, Baker E, Li ML, Zhao J, Jacobs DI. Electrical Stimulation of pediatric cardiac-derived c-kit + progenitor cells improves retention and cardiac function in right ventricular heart failure. Stem Cells 2019; 37:1528-1541. [PMID: 31574184 PMCID: PMC6916193 DOI: 10.1002/stem.3088] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 07/18/2019] [Accepted: 08/29/2019] [Indexed: 12/20/2022]
Abstract
Nearly 1 in every 120 children born has a congenital heart defect. Although surgical therapy has improved survival, many of these children go on to develop right ventricular heart failure (RVHF). The emergence of cardiovascular regenerative medicine as a potential therapeutic strategy for pediatric HF has provided new avenues for treatment with a focus on repairing or regenerating the diseased myocardium to restore cardiac function. Although primarily tried using adult cells and adult disease models, stem cell therapy is relatively untested in the pediatric population. Here, we investigate the ability of electrical stimulation (ES) to enhance the retention and therapeutic function of pediatric cardiac-derived c-kit+ progenitor cells (CPCs) in an animal model of RVHF. Human CPCs isolated from pediatric patients were exposed to chronic ES and implanted into the RV myocardium of rats. Cardiac function and cellular retention analysis showed electrically stimulated CPCs (ES-CPCs) were retained in the heart at a significantly higher level and longer time than control CPCs and also significantly improved right ventricular functional parameters. ES also induced upregulation of extracellular matrix and adhesion genes and increased in vitro survival and adhesion of cells. Specifically, upregulation of β1 and β5 integrins contributed to the increased retention of ES-CPCs. Lastly, we show that ES induces CPCs to release higher levels of pro-reparative factors in vitro. These findings suggest that ES can be used to increase the retention, survival, and therapeutic effect of human c-kit+ progenitor cells and can have implications on a variety of cell-based therapies. Stem Cells 2019;37:1528-1541.
Collapse
Affiliation(s)
- Joshua T. Maxwell
- Division of Pediatric Cardiology, Department of PediatricsEmory University School of MedicineAtlantaGeorgiaUSA
- Children's Heart Research & Outcomes (HeRO) CenterChildren's Healthcare of Atlanta & Emory UniversityAtlantaGeorgiaUSA
| | - David Trac
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology & Emory University School of MedicineAtlantaGeorgiaUSA
| | - Ming Shen
- Division of Pediatric Cardiology, Department of PediatricsEmory University School of MedicineAtlantaGeorgiaUSA
- Children's Heart Research & Outcomes (HeRO) CenterChildren's Healthcare of Atlanta & Emory UniversityAtlantaGeorgiaUSA
| | - Milton E. Brown
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology & Emory University School of MedicineAtlantaGeorgiaUSA
| | - Michael E. Davis
- Division of Pediatric Cardiology, Department of PediatricsEmory University School of MedicineAtlantaGeorgiaUSA
- Children's Heart Research & Outcomes (HeRO) CenterChildren's Healthcare of Atlanta & Emory UniversityAtlantaGeorgiaUSA
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology & Emory University School of MedicineAtlantaGeorgiaUSA
| | - Myra S. Chao
- Emory University College of Arts and SciencesAtlantaGeorgiaUSA
| | | | | | - Emily Baker
- Emory University College of Arts and SciencesAtlantaGeorgiaUSA
| | - Martin L. Li
- Emory University College of Arts and SciencesAtlantaGeorgiaUSA
| | - Jennifer Zhao
- Cornell University College of Arts and SciencesIthacaNew YorkUSA
| | | |
Collapse
|
33
|
Trac D, Hoffman JR, Bheri S, Maxwell JT, Platt MO, Davis ME. Predicting Functional Responses of Progenitor Cell Exosome Potential with Computational Modeling. Stem Cells Transl Med 2019; 8:1212-1221. [PMID: 31385648 PMCID: PMC6811701 DOI: 10.1002/sctm.19-0059] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/17/2019] [Indexed: 01/08/2023] Open
Abstract
Congenital heart disease can lead to severe right ventricular heart failure (RVHF). We have shown that aggregated c‐kit+ progenitor cells (CPCs) can improve RVHF repair, likely due to exosome‐mediated effects. Here, we demonstrate that miRNA content from monolayer (2D) and aggregated (3D) CPC exosomes can be related to in vitro angiogenesis and antifibrosis responses using partial least squares regression (PLSR). PLSR reduced the dimensionality of the data set to the top 40 miRNAs with the highest weighted coefficients for the in vitro biological responses. Target pathway analysis of these top 40 miRNAs demonstrated significant fit to cardiac angiogenesis and fibrosis pathways. Although the model was trained on in vitro data, we demonstrate that the model can predict angiogenesis and fibrosis responses to exosome treatment in vivo with a strong correlation with published in vivo responses. These studies demonstrate that PLSR modeling of exosome miRNA content has the potential to inform preclinical trials and predict new promising CPC therapies. stem cells translational medicine2019;8:1212–1221
Collapse
Affiliation(s)
- David Trac
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jessica R Hoffman
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Sruti Bheri
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, USA
| | - Joshua T Maxwell
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Manu O Platt
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, USA
| | - Michael E Davis
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, USA.,Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA.,Children's Heart Research & Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, Georgia, USA
| |
Collapse
|
34
|
Tan Y, Fei D, He X, Dai J, Xu R, Xu X, Wu J, Li B. L-type voltage-gated calcium channels in stem cells and tissue engineering. Cell Prolif 2019; 52:e12623. [PMID: 31115100 PMCID: PMC6669004 DOI: 10.1111/cpr.12623] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/26/2019] [Accepted: 03/29/2019] [Indexed: 12/22/2022] Open
Abstract
L-type voltage-gated calcium ion channels (L-VGCCs) have been demonstrated to be the mediator of several significant intracellular activities in excitable cells, such as neurons, chromaffin cells and myocytes. Recently, an increasing number of studies have investigated the function of L-VGCCs in non-excitable cells, particularly stem cells. However, there appear to be no systematic reviews of the relationship between L-VGCCs and stem cells, and filling this gap is prescient considering the contribution of L-VGCCs to the proliferation and differentiation of several types of stem cells. This review will discuss the possible involvement of L-VGCCs in stem cells, mainly focusing on osteogenesis mediated by mesenchymal stem cells (MSCs) from different tissues and neurogenesis mediated by neural stem/progenitor cells (NSCs). Additionally, advanced applications that use these channels as the target for tissue engineering, which may offer the hope of tissue regeneration in the future, will also be explored.
Collapse
Affiliation(s)
- Yi‐zhou Tan
- Department of Periodontology, State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, School of StomatologyThe Fourth Military Medical UniversityXi’anChina
| | - Dong‐dong Fei
- Department of Periodontology, State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, School of StomatologyThe Fourth Military Medical UniversityXi’anChina
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue EngineeringFourth Military Medical UniversityXi’anChina
| | - Xiao‐ning He
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue EngineeringFourth Military Medical UniversityXi’anChina
| | - Ji‐min Dai
- Doctoral students of eight-year programThe Fourth Military Medical UniversityXi’anChina
| | - Rong‐chen Xu
- Doctoral students of eight-year programThe Fourth Military Medical UniversityXi’anChina
| | - Xin‐yue Xu
- Department of Periodontology, State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, School of StomatologyThe Fourth Military Medical UniversityXi’anChina
| | - Jun‐jie Wu
- Department of Orthodontics, State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, School of StomatologyThe Fourth Military Medical UniversityXi’anChina
| | - Bei Li
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue EngineeringFourth Military Medical UniversityXi’anChina
| |
Collapse
|