1
|
He F, Andrabi SM, Shi H, Son Y, Qiu H, Xie J, Zhu W. Sequential delivery of cardioactive drugs via microcapped microneedle patches for improved heart function in post myocardial infarction rats. Acta Biomater 2025; 192:235-247. [PMID: 39643223 PMCID: PMC11735313 DOI: 10.1016/j.actbio.2024.12.009] [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: 08/26/2024] [Revised: 11/19/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
After myocardial infarction, the heart undergoes adverse remodeling characterized by a series of pathological changes, including inflammation, apoptosis, fibrosis, and hypertrophy. In addition to cardiac catheter-based re-establishment of blood flow, patients typically receive multiple medications that aim to address these different mechanisms underlying left ventricular remodeling. The current study aims to establish a versatile multi-drug delivery platform for the controlled and sequential delivery of multiple therapeutic agents in a single treatment. Toward this goal, we generated a microcapped microneedle patch carrying methylprednisolone, interleukin-10, and vascular endothelial growth factor. In vitro characterization demonstrated a time-sequenced release pattern of these drug: methylprednisolone for the first 3 days, interleukin-10 from day 1 to 15, and vascular endothelial growth factor from day 3 to 25. The therapeutic effects of the microneedle patch were evaluated in a rat model of acute myocardial infarction induced by permanent ligation of left anterior descending coronary artery. Heart function was measured using trans-thoracic echocardiography. Heart inflammation, apoptosis, hypertrophy and angiogenesis were evaluated using histology. Our data indicated that, at 28 days after patch transplantation, animals receiving the microneedle patch with sequential release of these three agents showed reduced inflammation, apoptosis and cardiac hypertrophy compared to the animals receiving control patch without sequential release of these agents, which is associated with the improved angiogenesis and heart function. In conclusion, the microneedle patch can be utilized to deliver multiple therapeutic agents in a controlled and sequential manner that aligns with the pathological phases following myocardial infarction. STATEMENT OF SIGNIFICANCE: The post-myocardial infarction heart remodeling is characterized by a series of pathological events including acute inflammation, apoptosis, fibrosis, cardiac hypertrophy, and depressed heart function. In current clinical practice, multiple procedures and drugs given at different time points are necessary to combat these series of pathological events. In this study, we developed a novel microcapped microneedle patch for the controlled sequential delivery of triple cardioprotective drugs aiming to combat acute inflammation and cardiac hypertrophy, and promote angiogenesis. This study presents a comprehensive therapeutic approach, with the microneedle patch addressing multifaceted pathological processes during post-myocardial infarction left ventricular remodeling. This cardiac drug delivery system has the potential to improve patient treatment by delivering drugs in alignment with the series of time-dependent pathological phases following myocardial infarction, ultimately improving clinical outcomes.
Collapse
Affiliation(s)
- Fengpu He
- Department of Cardiovascular Medicine, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale, AZ, 85259, USA
| | - Syed Muntazir Andrabi
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Haiwang Shi
- Department of Cardiovascular Medicine, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale, AZ, 85259, USA
| | - Yura Son
- Department of Cardiovascular Medicine, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale, AZ, 85259, USA
| | - Huiliang Qiu
- Department of Cardiovascular Medicine, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale, AZ, 85259, USA
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Wuqiang Zhu
- Department of Cardiovascular Medicine, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale, AZ, 85259, USA.
| |
Collapse
|
2
|
Tang L, Qiu H, Xu B, Su Y, Nyarige V, Li P, Chen H, Killham B, Liao J, Adam H, Yang A, Yu A, Jang M, Rubart M, Xie J, Zhu W. Microparticle Mediated Delivery of Apelin Improves Heart Function in Post Myocardial Infarction Mice. Circ Res 2024; 135:777-798. [PMID: 39145385 PMCID: PMC11392624 DOI: 10.1161/circresaha.124.324608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Apelin is an endogenous prepropeptide that regulates cardiac homeostasis and various physiological processes. Intravenous injection has been shown to improve cardiac contractility in patients with heart failure. However, its short half-life prevents studying its impact on left ventricular remodeling in the long term. Here, we aim to study whether microparticle-mediated slow release of apelin improves heart function and left ventricular remodeling in mice with myocardial infarction (MI). METHODS A cardiac patch was fabricated by embedding apelin-containing microparticles in a fibrin gel scaffold. MI was induced via permanent ligation of the left anterior descending coronary artery in adult C57BL/6J mice followed by epicardial patch placement immediately after (acute MI) or 28 days (chronic MI) post-MI. Four groups were included in this study, namely sham, MI, MI plus empty microparticle-embedded patch treatment, and MI plus apelin-containing microparticle-embedded patch treatment. Cardiac function was assessed by transthoracic echocardiography. Cardiomyocyte morphology, apoptosis, and cardiac fibrosis were evaluated by histology. Cardioprotective pathways were determined by RNA sequencing, quantitative polymerase chain reaction, and Western blot. RESULTS The level of endogenous apelin was largely reduced in the first 7 days after MI induction and it was normalized by day 28. Apelin-13 encapsulated in poly(lactic-co-glycolic acid) microparticles displayed a sustained release pattern for up to 28 days. Treatment with apelin-containing microparticle-embedded patch inhibited cardiac hypertrophy and reduced scar size in both acute and chronic MI models, which is associated with improved cardiac function. Data from cellular and molecular analyses showed that apelin inhibits the activation and proliferation of cardiac fibroblasts by preventing transforming growth factor-β-mediated activation of Smad2/3 (supporessor of mothers against decapentaplegic 2/3) and downstream profibrotic gene expression. CONCLUSIONS Poly(lactic-co-glycolic acid) microparticles prolonged the apelin release time in the mouse hearts. Epicardial delivery of the apelin-containing microparticle-embedded patch protects mice from both acute and chronic MI-induced cardiac dysfunction, inhibits cardiac fibrosis, and improves left ventricular remodeling.
Collapse
Affiliation(s)
- Ling Tang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Huiliang Qiu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Bing Xu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Yajuan Su
- Department of Surgery-Transplant and Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha (Y.S., J.X.)
| | - Verah Nyarige
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Pengsheng Li
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Houjia Chen
- Department of Bioengineering, University of Texas at Arlington (H.C., B.K., J.L.)
| | - Brady Killham
- Department of Bioengineering, University of Texas at Arlington (H.C., B.K., J.L.)
| | - Jun Liao
- Department of Bioengineering, University of Texas at Arlington (H.C., B.K., J.L.)
| | - Henderson Adam
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Aaron Yang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Alexander Yu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Michelle Jang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Michael Rubart
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (M.R.)
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha (Y.S., J.X.)
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| |
Collapse
|
3
|
Fan C, Qin K, Iroegbu CD, Xiang K, Gong Y, Guan Q, Wang W, Peng J, Guo J, Wu X, Yang J. Magnesium lithospermate B enhances the potential of human-induced pluripotent stem cell-derived cardiomyocytes for myocardial repair. Chin Med J (Engl) 2024; 137:1857-1869. [PMID: 38221772 DOI: 10.1097/cm9.0000000000002867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Indexed: 01/16/2024] Open
Abstract
BACKGROUND We previously reported that activation of the cell cycle in human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) enhances their remuscularization capacity after human cardiac muscle patch transplantation in infarcted mouse hearts. Herein, we sought to identify the effect of magnesium lithospermate B (MLB) on hiPSC-CMs during myocardial repair using a myocardial infarction (MI) mouse model. METHODS In C57BL/6 mice, MI was surgically induced by ligating the left anterior descending coronary artery. The mice were randomly divided into five groups ( n = 10 per group); a MI group (treated with phosphate-buffered saline only), a hiPSC-CMs group, a MLB group, a hiPSC-CMs + MLB group, and a Sham operation group. Cardiac function and MLB therapeutic efficacy were evaluated by echocardiography and histochemical staining 4 weeks after surgery. To identify the associated mechanism, nuclear factor (NF)-κB p65 and intercellular cell adhesion molecule-1 (ICAM1) signals, cell adhesion ability, generation of reactive oxygen species, and rates of apoptosis were detected in human umbilical vein endothelial cells (HUVECs) and hiPSC-CMs. RESULTS After 4 weeks of transplantation, the number of cells that engrafted in the hiPSC-CMs + MLB group was about five times higher than those in the hiPSC-CMs group. Additionally, MLB treatment significantly reduced tohoku hospital pediatrics-1 (THP-1) cell adhesion, ICAM1 expression, NF-κB nuclear translocation, reactive oxygen species production, NF-κB p65 phosphorylation, and cell apoptosis in HUVECs cultured under hypoxia. Similarly, treatment with MLB significantly inhibited the apoptosis of hiPSC-CMs via enhancing signal transducer and activator of transcription 3 (STAT3) phosphorylation and B-cell lymphoma-2 (BCL2) expression, promoting STAT3 nuclear translocation, and downregulating BCL2-Associated X, dual specificity phosphatase 2 (DUSP2), and cleaved-caspase-3 expression under hypoxia. Furthermore, MLB significantly suppressed the production of malondialdehyde and lactate dehydrogenase and the reduction in glutathione content induced by hypoxia in both HUVECs and hiPSC-CMs in vitro . CONCLUSIONS MLB significantly enhanced the potential of hiPSC-CMs in repairing injured myocardium by improving endothelial cell function via the NF-κB/ICAM1 pathway and inhibiting hiPSC-CMs apoptosis via the DUSP2/STAT3 pathway.
Collapse
Affiliation(s)
- Chengming Fan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410078, China
- Hunan Fangsheng Pharmaceutical Co., Ltd., Changsha, Hunan 410000, China
| | - Kele Qin
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Chukwuemeka Daniel Iroegbu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Kun Xiang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Yibo Gong
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Qing Guan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Wenxiang Wang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 41000, China
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410078, China
| | - Jianjun Guo
- Hunan Fangsheng Pharmaceutical Co., Ltd., Changsha, Hunan 410000, China
| | - Xun Wu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 41000, China
| | - Jinfu Yang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
4
|
Song L, Jia K, Yang F, Wang J. Advanced Nanomedicine Approaches for Myocardial Infarction Treatment. Int J Nanomedicine 2024; 19:6399-6425. [PMID: 38952676 PMCID: PMC11215519 DOI: 10.2147/ijn.s467219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 06/04/2024] [Indexed: 07/03/2024] Open
Abstract
Myocardial infarction, usually caused by the rupture of atherosclerotic plaque, leads to irreversible ischemic cardiomyocyte death within hours followed by impaired cardiac performance or even heart failure. Current interventional reperfusion strategies for myocardial infarction still face high mortality with the development of heart failure. Nanomaterial-based therapy has made great progress in reducing infarct size and promoting cardiac repair after MI, although most studies are preclinical trials. This review focuses primarily on recent progress (2016-now) in the development of various nanomedicines in the treatment of myocardial infarction. We summarize these applications with the strategy of mechanism including anti-cardiomyocyte death strategy, activation of neovascularization, antioxidants strategy, immunomodulation, anti-cardiac remodeling, and cardiac repair.
Collapse
Affiliation(s)
- Lin Song
- School of Basic Medicine, Qingdao University, Qingdao, People’s Republic of China
| | - Kangwei Jia
- School of Basic Medicine, Qingdao University, Qingdao, People’s Republic of China
| | - Fuqing Yang
- School of Basic Medicine, Qingdao University, Qingdao, People’s Republic of China
| | - Jianxun Wang
- School of Basic Medicine, Qingdao University, Qingdao, People’s Republic of China
| |
Collapse
|
5
|
Liu T, Hao Y, Zhang Z, Zhou H, Peng S, Zhang D, Li K, Chen Y, Chen M. Advanced Cardiac Patches for the Treatment of Myocardial Infarction. Circulation 2024; 149:2002-2020. [PMID: 38885303 PMCID: PMC11191561 DOI: 10.1161/circulationaha.123.067097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Myocardial infarction is a cardiovascular disease characterized by a high incidence rate and mortality. It leads to various cardiac pathophysiological changes, including ischemia/reperfusion injury, inflammation, fibrosis, and ventricular remodeling, which ultimately result in heart failure and pose a significant threat to global health. Although clinical reperfusion therapies and conventional pharmacological interventions improve emergency survival rates and short-term prognoses, they are still limited in providing long-lasting improvements in cardiac function or reversing pathological progression. Recently, cardiac patches have gained considerable attention as a promising therapy for myocardial infarction. These patches consist of scaffolds or loaded therapeutic agents that provide mechanical reinforcement, synchronous electrical conduction, and localized delivery within the infarct zone to promote cardiac restoration. This review elucidates the pathophysiological progression from myocardial infarction to heart failure, highlighting therapeutic targets and various cardiac patches. The review considers the primary scaffold materials, including synthetic, natural, and conductive materials, and the prevalent fabrication techniques and optimal properties of the patch, as well as advanced delivery strategies. Last, the current limitations and prospects of cardiac patch research are considered, with the goal of shedding light on innovative products poised for clinical application.
Collapse
Affiliation(s)
- Tailuo Liu
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases (T.L., Y.H., H.Z., S.P., D.Z., Y.C., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
- Department of Cardiology (T.L., S.P., D.Z., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, PR China (T.L., K.L., Y.C.)
| | - Ying Hao
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases (T.L., Y.H., H.Z., S.P., D.Z., Y.C., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
| | - Zixuan Zhang
- West China School of Public Health/West China Fourth Hospital, Sichuan University, Chengdu, PR China (Z.Z.)
| | - Hao Zhou
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases (T.L., Y.H., H.Z., S.P., D.Z., Y.C., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
| | - Shiqin Peng
- Department of Cardiology (T.L., S.P., D.Z., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
| | - Dingyi Zhang
- Department of Cardiology (T.L., S.P., D.Z., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
| | - Ka Li
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, PR China (T.L., K.L., Y.C.)
| | - Yuwen Chen
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, PR China (T.L., K.L., Y.C.)
| | - Mao Chen
- Department of Cardiology (T.L., S.P., D.Z., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
| |
Collapse
|
6
|
Wei Q, Xiao Y, Du L, Li Y. Advances in Nanoparticles in the Prevention and Treatment of Myocardial Infarction. Molecules 2024; 29:2415. [PMID: 38893291 PMCID: PMC11173599 DOI: 10.3390/molecules29112415] [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: 03/26/2024] [Revised: 05/17/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Myocardial infarction (MI) is one of the most prevalent types of cardiovascular disease. During MI, myocardial cells become ischemic and necrotic due to inadequate blood perfusion, leading to irreversible damage to the heart. Despite the development of therapeutic strategies for the prevention and treatment of MI, their effects are still unsatisfactory. Nanoparticles represent a new strategy for the pre-treatment and treatment of MI, and novel multifunctional nanoparticles with preventive and therapeutic capabilities hold promise for the prevention and treatment of this disease. This review summarizes the common types and properties of nanoparticles, and focuses on the research progress of nanoparticles for the prevention and treatment of MI.
Collapse
Affiliation(s)
| | | | | | - Ya Li
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Q.W.); (Y.X.); (L.D.)
| |
Collapse
|
7
|
Esmaeili H, Patino-Guerrero A, Nelson RA, Karamanova N, M Fisher T, Zhu W, Perreault F, Migrino RQ, Nikkhah M. Engineered Gold and Silica Nanoparticle-Incorporated Hydrogel Scaffolds for Human Stem Cell-Derived Cardiac Tissue Engineering. ACS Biomater Sci Eng 2024; 10:2351-2366. [PMID: 38323834 PMCID: PMC11075803 DOI: 10.1021/acsbiomaterials.3c01256] [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] [Indexed: 02/08/2024]
Abstract
Electrically conductive biomaterials and nanomaterials have demonstrated great potential in the development of functional and mature cardiac tissues. In particular, gold nanomaterials have emerged as promising candidates due to their biocompatibility and ease of fabrication for cardiac tissue engineering utilizing rat- or stem cell-derived cardiomyocytes (CMs). However, despite significant advancements, it is still not clear whether the enhancement in cardiac tissue function is primarily due to the electroconductivity features of gold nanoparticles or the structural changes of the scaffold resulting from the addition of these nanoparticles. To address this question, we developed nanoengineered hydrogel scaffolds comprising gelatin methacrylate (GelMA) embedded with either electrically conductive gold nanorods (GNRs) or nonconductive silica nanoparticles (SNPs). This enabled us to simultaneously assess the roles of electrically conductive and nonconductive nanomaterials in the functionality and fate of human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Our studies revealed that both GNR- and SNP-incorporated hydrogel scaffolds exhibited excellent biocompatibility and similar cardiac cell attachment. Although the expression of sarcomere alpha-actinin did not significantly differ among the conditions, a more organized sarcomere structure was observed within the GNR-embedded hydrogels compared to the nonconductive nanoengineered scaffolds. Furthermore, electrical coupling was notably improved in GNR-embedded scaffolds, as evidenced by the synchronous calcium flux and enhanced calcium transient intensity. While we did not observe a significant difference in the gene expression profile of human cardiac tissues formed on the conductive GNR- and nonconductive SNP-incorporated hydrogels, we noticed marginal improvements in the expression of some calcium and structural genes in the nanomaterial-embedded hydrogel groups as compared to the control condition. Given that the cardiac tissues formed atop the nonconductive SNP-based scaffolds (used as the control for conductivity) also displayed similar levels of gene expression as compared to the conductive hydrogels, it suggests that the electrical conductivity of nanomaterials (i.e., GNRs) may not be the sole factor influencing the function and fate of hiPSC-derived cardiac tissues when cells are cultured atop the scaffolds. Overall, our findings provide additional insights into the role of electrically conductive gold nanoparticles in regulating the functionalities of hiPSC-CMs.
Collapse
Affiliation(s)
- Hamid Esmaeili
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States
| | - Alejandra Patino-Guerrero
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic, Scottsdale, Arizona 85259, United States
| | - Ronald A Nelson
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States
| | - Nina Karamanova
- Phoenix Veterans Affairs Health Care System, Phoenix, Arizona 85022, United States
| | - Taylor M Fisher
- School of Sustainable Engineering and the Built Environment, Arizona State University, Tempe, Arizona 85287, United States
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic, Scottsdale, Arizona 85259, United States
| | - François Perreault
- School of Sustainable Engineering and the Built Environment, Arizona State University, Tempe, Arizona 85287, United States
| | - Raymond Q Migrino
- Phoenix Veterans Affairs Health Care System, Phoenix, Arizona 85022, United States
- University of Arizona College of Medicine, Phoenix, Arizona 85004, United States
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States
- Biodesign Virginia G. Piper Center for Personalized Diagnosis, Arizona State University, Tempe, Arizona 85287, United States
| |
Collapse
|
8
|
Cheng X, Liu Y, Qi B, Wang Y, Zheng Y, Liang X, Chang Y, Ning M, Gao W, Li T. Glycyrrhizic acid alleviated MI/R-induced injuries by inhibiting Hippo/YAP signaling pathways. Cell Signal 2024; 115:111036. [PMID: 38185229 DOI: 10.1016/j.cellsig.2024.111036] [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: 10/16/2023] [Revised: 12/22/2023] [Accepted: 01/02/2024] [Indexed: 01/09/2024]
Abstract
BACKGROUND Previous research has demonstrated that glycyrrhizic acid (GA) exhibits antioxidant, anti-inflammatory, and antiapoptotic characteristics. Using myocardial ischemia/reperfusion injury as a case study, this study aims to clarify the functional significance of GA and to elucidate the mechanisms involved. MATERIALS AND METHODS In this study, an MI/R injury model was established both in vivo and in vitro to investigate the impact of GA on MI/R injury. The viability of H9c2 cells was evaluated using the Cell Counting Kit-8. Myocardial damage was assessed through the measurement of creatine kinase myocardial band (CK-MB) levels and lactate dehydrogenase (LDH), HE staining, and MASSON staining. Inflammatory cytokine levels (IL-6, IL-1β, IL-10, and TNF-α) were measured to determine the presence of inflammation. Cellular oxidative stress was evaluated by measuring ROS and MMP levels, while cardiac function was assessed using cardiac color Doppler ultrasound. Immunofluorescence staining to determine the nuclear translocation of YAP, TUNEL to determine apoptosis, and western blotting to determine gene expression. RESULTS GA treatment effectively alleviated myocardial injury induced by MI/R, as evidenced by reduced levels of inflammatory cytokines (IL-1β, IL-6, IL-10, and TNF-α) and cardiac biomarkers (CK-MB, LDH) in MI/R rats. Moreover, There was a significant increase in cell viability in vitro after GA treatment and inhibited reactive oxygen species (ROS) during oxidative stress, while also increasing mitochondrial membrane potential (MMP) in vitro. The Western blot findings indicate that GA treatment effectively suppressed apoptosis in both in vivo and in vitro settings. Additionally, GA demonstrated inhibitory effects on the activation of the Hippo/YAP signaling pathway triggered by MI/R and facilitated YAP nuclear translocation both in vitro and in vivo. It has been found, however, in vitro, that silencing the YAP gene negates GA's protective effect against hypoxia/reoxygenation-induced myocardial injury. CONCLUSION This study suggests that GA regulates YAP nuclear translocation by inhibiting the Hippo/YAP signaling pathway, which protects ists against MI/R injury. This finding may present a novel therapeutic approach for the treatment of MI/R.
Collapse
Affiliation(s)
- Xian Cheng
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China.
| | - Yanwu Liu
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Bingcai Qi
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Yuchao Wang
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; School of Medicine, Nankai University, Tianjin 300071, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Yue Zheng
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; School of Medicine, Nankai University, Tianjin 300071, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Xiaoyu Liang
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Yun Chang
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Meng Ning
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Wenqing Gao
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; School of Medicine, Nankai University, Tianjin 300071, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China.
| | - Tong Li
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; School of Medicine, Nankai University, Tianjin 300071, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China.
| |
Collapse
|
9
|
Vo QD, Saito Y, Nakamura K, Iida T, Yuasa S. Induced Pluripotent Stem Cell-Derived Cardiomyocytes Therapy for Ischemic Heart Disease in Animal Model: A Meta-Analysis. Int J Mol Sci 2024; 25:987. [PMID: 38256060 PMCID: PMC10815661 DOI: 10.3390/ijms25020987] [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: 12/08/2023] [Revised: 01/07/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Ischemic heart disease (IHD) poses a significant challenge in cardiovascular health, with current treatments showing limited success. Induced pluripotent derived-cardiomyocyte (iPSC-CM) therapy within regenerative medicine offers potential for IHD patients, although its clinical impacts remain uncertain. This study utilizes meta-analysis to assess iPSC-CM outcomes in terms of efficacy and safety in IHD animal model studies. A meta-analysis encompassing PUBMED, ScienceDirect, Web of Science, and the Cochrane Library databases, from inception until October 2023, investigated iPSC therapy effects on cardiac function and safety outcomes. Among 51 eligible studies involving 1012 animals, despite substantial heterogeneity, the iPSC-CM transplantation improved left ventricular ejection fraction (LVEF) by 8.23% (95% CI, 7.15 to 9.32%; p < 0.001) compared to control groups. Additionally, cell-based treatment reduced the left ventricle fibrosis area and showed a tendency to reduce left ventricular end-systolic volume (LVESV) and end-diastolic volume (LVEDV). No significant differences emerged in mortality and arrhythmia risk between iPSC-CM treatment and control groups. In conclusion, this meta-analysis indicates iPSC-CM therapy's promise as a safe and beneficial intervention for enhancing heart function in IHD. However, due to observed heterogeneity, the efficacy of this treatment must be further explored through large randomized controlled trials based on rigorous research design.
Collapse
Affiliation(s)
- Quan Duy Vo
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (Q.D.V.); (T.I.); (S.Y.)
| | - Yukihiro Saito
- Department of Cardiovascular Medicine, Okayama University Hospital, Okayama 700-8558, Japan
| | - Kazufumi Nakamura
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (Q.D.V.); (T.I.); (S.Y.)
| | - Toshihiro Iida
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (Q.D.V.); (T.I.); (S.Y.)
| | - Shinsuke Yuasa
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (Q.D.V.); (T.I.); (S.Y.)
| |
Collapse
|
10
|
Katili PA, Karima AP, Azwani W, Antarianto RD, Djer MM. Application of Human Induced Pluripotent Stem Cells for Tissue Engineered Cardiomyocyte Modelling. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2023; 9:431-446. [DOI: 10.1007/s40883-023-00294-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 01/02/2023] [Accepted: 01/06/2023] [Indexed: 01/06/2025]
Abstract
Abstract
Purpose
Cardiac
tissue engineering opens up opportunities for regenerative therapy in heart diseases. Current technologies improve engineered cardiac tissue characteristics by combining human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) with non-cardiomyocytes, selective biomaterials, and additional growth factors. Animal models are still required to determine cardiac patches’ overall in vivo effect before initiating human trials. Here, we review the current in vivo studies of cardiac patches using hiPSC-CMs.
Methods
We performed a literature search for studies on cardiac patch in vivo application and compared outcomes based on cell engraftment, functional changes, and safety profiles.
Results
Present studies confirm the beneficial results of combining hiPSC-CMs with other cardiac cell lineages and biomaterials. They improved the functional capacity of the heart, showed a reduction in infarct size, and initiated an adaptive inflammatory process through neovascularisation.
Conclusion
The cardiac patch is currently the most effective delivery system, proving safety and improvements in animal models, which are suggested to be the role of the paracrine mechanism. Further studies should focus on honing in vitro patch characteristics to achieve ideal results.
Lay Summary
Cardiac tissue engineering answers the demand for regenerative therapy in heart diseases. Combining human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) with biomaterials and growth factors in cardiac patches improves the heart’s structural and functional characteristics. This delivery system is safe and efficient for delivering many cells and minimising cellular loss in vivo. Rat and porcine models of ischemic and non-ischemic heart diseases demonstrated the benefits of this therapy, which include cell engraftment, reduced infarct size, and increased left ventricular (LV) systolic function, with no reported critical adverse events. These reports sufficiently provide evidence of feasible improvements to proceed towards further trials.
Collapse
|
11
|
Kistamás K, Müller A, Muenthaisong S, Lamberto F, Zana M, Dulac M, Leal F, Maziz A, Costa P, Bernotiene E, Bergaud C, Dinnyés A. Multifactorial approaches to enhance maturation of human iPSC-derived cardiomyocytes. J Mol Liq 2023; 387:122668. [DOI: 10.1016/j.molliq.2023.122668] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
12
|
Zhang F, Xu X, Hou J, Xiao H, Guo F, Li X, Yang H. Cardioprotective efficacy of Xin-shu-bao tablet in heart failure with reduced ejection fraction by modulating THBD/ARRB1/FGF1/STIM1 signaling. Biomed Pharmacother 2023; 165:115119. [PMID: 37423168 DOI: 10.1016/j.biopha.2023.115119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/20/2023] [Accepted: 07/01/2023] [Indexed: 07/11/2023] Open
Abstract
Traditional Chinese medicine offer unique advantages in mitigating and preventing early or intermediate stage for treating heart failure (HF). The purpose of this study was to assess the in vivo therapeutic efficacy of Xin-shu-bao (XSB) at different stages of HF following induction of a myocardial infarction (MI) in mice and use mass spectrometry-based proteomics to identify potential therapeutic targets for different stages of HF based on the molecular changes following XSB treatment. XSB had high cardioprotective efficacy in the pre-HF with reduced ejection fraction (HFrEF) stages, but had a weak or no effect in the post-HFrEF stages. This was supported by echocardiographic measurements showing that XSB decreased ejection fraction and fractional shortening in HF. XSB administration improved cardiac function in the pre- and post-HFrEF mouse model, ameliorated deleterious changes to the morphology and subcellular structure of cardiomyocytes, and reduced cardiac fibrosis. Proteomics analysis showed that XSB intervention exclusively targeted thrombomodulin (THBD) and stromal interaction molecule 1 (STIM1) proteins when administered to the mice for both 8 and 6 weeks. Furthermore, XSB intervention for 8, 6, and 4 weeks after MI induction increased the expression of fibroblast growth factor 1 (FGF1) and decreased arrestin β1 (ARRB1), which are classic biomarkers of cardiac fibroblast transformation and collagen synthesis, respectively. Overall, the study suggests that early intervention with XSB could be an effective strategy for preventing HFrEF and highlights potential therapeutic targets for further investigation into HFrEF remediation strategies.
Collapse
Affiliation(s)
- Fengrong Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xingyue Xu
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jinli Hou
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Honghe Xiao
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Feifei Guo
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xianyu Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Hongjun Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China; China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
13
|
Patino-Guerrero A, Esmaeili H, Migrino RQ, Nikkhah M. Nanoengineering of gold nanoribbon-embedded isogenic stem cell-derived cardiac organoids. RSC Adv 2023; 13:16985-17000. [PMID: 37288383 PMCID: PMC10243308 DOI: 10.1039/d3ra01811c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 05/29/2023] [Indexed: 06/09/2023] Open
Abstract
Cardiac tissue engineering is an emerging field providing tools to treat and study cardiovascular diseases (CVDs). In the past years, the integration of stem cell technologies with micro- and nanoengineering techniques has enabled the creation of novel engineered cardiac tissues (ECTs) with potential applications in disease modeling, drug screening, and regenerative medicine. However, a major unaddressed limitation of stem cell-derived ECTs is their immature state, resembling a neonatal phenotype and genotype. The modulation of the cellular microenvironment within the ECTs has been proposed as an efficient mechanism to promote cellular maturation and improve features such as cellular coupling and synchronization. The integration of biological and nanoscale cues in the ECTs could serve as a tool for the modification and control of the engineered tissue microenvironment. Here we present a proof-of-concept study for the integration of biofunctionalized gold nanoribbons (AuNRs) with hiPSC-derived isogenic cardiac organoids to enhance tissue function and maturation. We first present extensive characterization of the synthesized AuNRs, their PEGylation and cytotoxicity evaluation. We then evaluated the functional contractility and transcriptomic profile of cardiac organoids fabricated with hiPSC-derived cardiomyocytes (mono-culture) as well as with hiPSC-derived cardiomyocytes and cardiac fibroblasts (co-culture). We demonstrated that PEGylated AuNRs are biocompatible and do not induce cell death in hiPSC-derived cardiac cells and organoids. We also found an improved transcriptomic profile of the co-cultured organoids indicating maturation of the hiPSC-derived cardiomyocytes in the presence of cardiac fibroblasts. Overall, we present for the first time the integration of AuNRs into cardiac organoids, showing promising results for improved tissue function.
Collapse
Affiliation(s)
| | - Hamid Esmaeili
- School of Biological and Health Systems Engineering, Arizona State University Tempe AZ 8528 USA
| | - Raymond Q Migrino
- Phoenix Veterans Affairs Health Care System Phoenix AZ 85012 USA
- University of Arizona College of Medicine Phoenix AZ 85004 USA
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering, Arizona State University Tempe AZ 8528 USA
- Center for Personalized Diagnostics Biodesign Institute, Arizona State University Tempe AZ 85281 USA
| |
Collapse
|
14
|
Lou X, Tang Y, Ye L, Pretorius D, Fast VG, Kahn-Krell AM, Zhang J, Zhang J, Qiao A, Qin G, Kamp T, Thomson JA, Zhang J. Cardiac muscle patches containing four types of cardiac cells derived from human pluripotent stem cells improve recovery from cardiac injury in mice. Cardiovasc Res 2023; 119:1062-1076. [PMID: 36647784 PMCID: PMC10153642 DOI: 10.1093/cvr/cvad004] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 10/24/2022] [Accepted: 11/04/2022] [Indexed: 01/18/2023] Open
Abstract
AIMS We have shown that human cardiac muscle patches (hCMPs) containing three different types of cardiac cells-cardiomyocytes (CMs), smooth muscle cells (SMCs), and endothelial cells (ECs), all of which were differentiated from human pluripotent stem cells (hPSCs)-significantly improved cardiac function, infarct size, and hypertrophy in a pig model of myocardial infarction (MI). However, hPSC-derived CMs (hPSC-CMs) are phenotypically immature, which may lead to arrhythmogenic concerns; thus, since hPSC-derived cardiac fibroblasts (hPSC-CFs) appear to enhance the maturity of hPSC-CMs, we compared hCMPs containing hPSC-CMs, -SMCs, -ECs, and -CFs (4TCC-hCMPs) with a second hCMP construct that lacked hPSC-CFs but was otherwise identical [hCMP containing hPSC-CMs, -AECs, and -SMCs (3TCC-hCMPs)]. METHODS AND RESULTS hCMPs were generated in a fibrin scaffold. MI was induced in severe combined immunodeficiency (SCID) mice through permanent coronary artery (left anterior descending) ligation, followed by treatment with cardiac muscle patches. Animal groups included: MI heart treated with 3TCC-hCMP; with 4TCC-hCMP; MI heart treated with no patch (MI group) and sham group. Cardiac function was evaluated using echocardiography, and cell engraftment rate and infarct size were evaluated histologically at 4 weeks after patch transplantation. The results from experiments in cultured hCMPs demonstrate that the inclusion of cardiac fibroblast in 4TCC-hCMPs had (i) better organized sarcomeres; (ii) abundant structural, metabolic, and ion-channel markers of CM maturation; and (iii) greater conduction velocities (31 ± 3.23 cm/s, P < 0.005) and action-potential durations (APD50 = 365 ms ± 2.649, P < 0.0001; APD = 408 ms ± 2.757, P < 0.0001) than those (velocity and APD time) in 3TCC-hCMPs. Furthermore, 4TCC-hCMPs transplantation resulted in better cardiac function [ejection fraction (EF) = 49.18% ± 0.86, P < 0.05], reduced infarct size (22.72% ± 0.98, P < 0.05), and better engraftment (15.99% ± 1.56, P < 0.05) when compared with 3TCC-hCMPs (EF = 41.55 ± 0.92%, infarct size = 39.23 ± 4.28%, and engraftment = 8.56 ± 1.79%, respectively). CONCLUSION Collectively, these observations suggest that the inclusion of hPSC-CFs during hCMP manufacture promotes hPSC-CM maturation and increases the potency of implanted hCMPs for improving cardiac recovery in mice model of MI.
Collapse
Affiliation(s)
- Xi Lou
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, 1670 University Boulevard, Volker Hall G094J, Birmingham, AL 35294, USA
| | - Yawen Tang
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, 1670 University Boulevard, Volker Hall G094J, Birmingham, AL 35294, USA
| | - Lei Ye
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, 1670 University Boulevard, Volker Hall G094J, Birmingham, AL 35294, USA
| | - Danielle Pretorius
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, 1670 University Boulevard, Volker Hall G094J, Birmingham, AL 35294, USA
| | - Vladimir G Fast
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, 1670 University Boulevard, Volker Hall G094J, Birmingham, AL 35294, USA
| | - Asher M Kahn-Krell
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, 1670 University Boulevard, Volker Hall G094J, Birmingham, AL 35294, USA
| | - Jue Zhang
- Morgridge Institute for Research, Madison, WI 53715, USA
| | - Jianhua Zhang
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - Aijun Qiao
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, 1670 University Boulevard, Volker Hall G094J, Birmingham, AL 35294, USA
| | - Gangjian Qin
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, 1670 University Boulevard, Volker Hall G094J, Birmingham, AL 35294, USA
| | - Timothy Kamp
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - James A Thomson
- Morgridge Institute for Research, Madison, WI 53715, USA
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - Jianyi Zhang
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, 1670 University Boulevard, Volker Hall G094J, Birmingham, AL 35294, USA
- Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
15
|
Gil-Cabrerizo P, Scaccheti I, Garbayo E, Blanco-Prieto MJ. Cardiac tissue engineering for myocardial infarction treatment. Eur J Pharm Sci 2023; 185:106439. [PMID: 37003408 DOI: 10.1016/j.ejps.2023.106439] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/26/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023]
Abstract
Myocardial infarction is one of the major causes of morbidity and mortality worldwide. Current treatments can relieve the symptoms of myocardial ischemia but cannot repair the necrotic myocardial tissue. Novel therapeutic strategies based on cellular therapy, extracellular vesicles, non-coding RNAs and growth factors have been designed to restore cardiac function while inducing cardiomyocyte cycle re-entry, ensuring angiogenesis and cardioprotection, and preventing ventricular remodeling. However, they face low stability, cell engraftment issues or enzymatic degradation in vivo, and it is thus essential to combine them with biomaterial-based delivery systems. Microcarriers, nanocarriers, cardiac patches and injectable hydrogels have yielded promising results in preclinical studies, some of which are currently being tested in clinical trials. In this review, we cover the recent advances made in cellular and acellular therapies used for cardiac repair after MI. We present current trends in cardiac tissue engineering related to the use of microcarriers, nanocarriers, cardiac patches and injectable hydrogels as biomaterial-based delivery systems for biologics. Finally, we discuss some of the most crucial aspects that should be addressed in order to advance towards the clinical translation of cardiac tissue engineering approaches.
Collapse
Affiliation(s)
- Paula Gil-Cabrerizo
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, C/Irunlarrea 1, E-31080, Spain.; Navarra Institute for Health Research, IdiSNA, Pamplona, C/Irunlarrea 3, E-31008 Pamplona, Spain
| | - Ilaria Scaccheti
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, C/Irunlarrea 1, E-31080, Spain
| | - Elisa Garbayo
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, C/Irunlarrea 1, E-31080, Spain.; Navarra Institute for Health Research, IdiSNA, Pamplona, C/Irunlarrea 3, E-31008 Pamplona, Spain..
| | - María J Blanco-Prieto
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, C/Irunlarrea 1, E-31080, Spain.; Navarra Institute for Health Research, IdiSNA, Pamplona, C/Irunlarrea 3, E-31008 Pamplona, Spain..
| |
Collapse
|
16
|
Patino-Guerrero A, Ponce Wong RD, Kodibagkar VD, Zhu W, Migrino RQ, Graudejus O, Nikkhah M. Development and Characterization of Isogenic Cardiac Organoids from Human-Induced Pluripotent Stem Cells Under Supplement Starvation Regimen. ACS Biomater Sci Eng 2023; 9:944-958. [PMID: 36583992 DOI: 10.1021/acsbiomaterials.2c01290] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The prevalence of cardiovascular risk factors is expected to increase the occurrence of cardiovascular diseases (CVDs) worldwide. Cardiac organoids are promising candidates for bridging the gap between in vitro experimentation and translational applications in drug development and cardiac repair due to their attractive features. Here we present the fabrication and characterization of isogenic scaffold-free cardiac organoids derived from human induced pluripotent stem cells (hiPSCs) formed under a supplement-deprivation regimen that allows for metabolic synchronization and maturation of hiPSC-derived cardiac cells. We propose the formation of coculture cardiac organoids that include hiPSC-derived cardiomyocytes and hiPSC-derived cardiac fibroblasts (hiPSC-CMs and hiPSC-CFs, respectively). The cardiac organoids were characterized through extensive morphological assessment, evaluation of cellular ultrastructures, and analysis of transcriptomic and electrophysiological profiles. The morphology and transcriptomic profile of the organoids were improved by coculture of hiPSC-CMs with hiPSC-CFs. Specifically, upregulation of Ca2+ handling-related genes, such as RYR2 and SERCA, and structure-related genes, such as TNNT2 and MYH6, was observed. Additionally, the electrophysiological characterization of the organoids under supplement deprivation shows a trend for reduced conduction velocity for coculture organoids. These studies help us gain a better understanding of the role of other isogenic cells such as hiPSC-CFs in the formation of mature cardiac organoids, along with the introduction of exogenous chemical cues, such as supplement starvation.
Collapse
Affiliation(s)
- Alejandra Patino-Guerrero
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona8528, United States
| | | | - Vikram D Kodibagkar
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona8528, United States
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Mayo Clinic Arizona, Scottsdale, Arizona85259, United States
| | - Raymond Q Migrino
- Phoenix Veterans Affairs Health Care System, Phoenix, Arizona85012, United States.,University of Arizona College of Medicine, Phoenix, Arizona85004, United States
| | - Oliver Graudejus
- BMSEED, Mesa, Arizona85201, United States.,School of Molecular Sciences, Arizona State University, Tempe, Arizona85287, United States
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona8528, United States.,Center for Personalized Diagnostics Biodesign Institute, Arizona State University, Tempe, Arizona85281, United States
| |
Collapse
|
17
|
He X, Liang J, Paul C, Huang W, Dutta S, Wang Y. Advances in Cellular Reprogramming-Based Approaches for Heart Regenerative Repair. Cells 2022; 11:3914. [PMID: 36497171 PMCID: PMC9740402 DOI: 10.3390/cells11233914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/18/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Continuous loss of cardiomyocytes (CMs) is one of the fundamental characteristics of many heart diseases, which eventually can lead to heart failure. Due to the limited proliferation ability of human adult CMs, treatment efficacy has been limited in terms of fully repairing damaged hearts. It has been shown that cell lineage conversion can be achieved by using cell reprogramming approaches, including human induced pluripotent stem cells (hiPSCs), providing a promising therapeutic for regenerative heart medicine. Recent studies using advanced cellular reprogramming-based techniques have also contributed some new strategies for regenerative heart repair. In this review, hiPSC-derived cell therapeutic methods are introduced, and the clinical setting challenges (maturation, engraftment, immune response, scalability, and tumorigenicity), with potential solutions, are discussed. Inspired by the iPSC reprogramming, the approaches of direct cell lineage conversion are merging, such as induced cardiomyocyte-like cells (iCMs) and induced cardiac progenitor cells (iCPCs) derived from fibroblasts, without induction of pluripotency. The studies of cellular and molecular pathways also reveal that epigenetic resetting is the essential mechanism of reprogramming and lineage conversion. Therefore, CRISPR techniques that can be repurposed for genomic or epigenetic editing become attractive approaches for cellular reprogramming. In addition, viral and non-viral delivery strategies that are utilized to achieve CM reprogramming will be introduced, and the therapeutic effects of iCMs or iCPCs on myocardial infarction will be compared. After the improvement of reprogramming efficiency by developing new techniques, reprogrammed iCPCs or iCMs will provide an alternative to hiPSC-based approaches for regenerative heart therapies, heart disease modeling, and new drug screening.
Collapse
Affiliation(s)
- Xingyu He
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Jialiang Liang
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Christian Paul
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Wei Huang
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Suchandrima Dutta
- Department of Internal Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Yigang Wang
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| |
Collapse
|
18
|
Resveratrol and FGF1 Synergistically Ameliorates Doxorubicin-Induced Cardiotoxicity via Activation of SIRT1-NRF2 Pathway. Nutrients 2022; 14:nu14194017. [PMID: 36235670 PMCID: PMC9572068 DOI: 10.3390/nu14194017] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/16/2022] [Accepted: 09/20/2022] [Indexed: 11/30/2022] Open
Abstract
Doxorubicin (DOX) has received attention due to dose-dependent cardiotoxicity through abnormal redox cycling. Native fibroblast growth factor 1 (FGF1) is known for its anti-oxidative benefits in cardiovascular diseases, but possesses a potential tumorigenic risk. Coincidentally, the anti-proliferative properties of resveratrol (RES) have attracted attention as alternatives or auxiliary therapy when combined with other chemotherapeutic drugs. Therefore, the purpose of this study is to explore the therapeutic potential and underlying mechanisms of co-treatment of RES and FGF1 in a DOX-treated model. Here, various cancer cells were applied to determine whether RES could antagonize the oncogenesis effect of FGF1. In addition, C57BL/6J mice and H9c2 cells were used to testify the therapeutic potential of a co-treatment of RES and FGF1 against DOX-induced cardiotoxicity. We found RES could reduce the growth-promoting activity of FGF1. Additionally, the co-treatment of RES and FGF1 exhibits a more powerful cardio-antioxidative capacity in a DOX-treated model. The inhibition of SIRT1/NRF2 abolished RES in combination with FGF1 on cardioprotective action. Further mechanism analysis demonstrated that SIRT1 and NRF2 might form a positive feedback loop to perform the protective effect on DOX-induced cardiotoxicity. These favorable anti-oxidative activities and reduced proliferative properties of the co-treatment of RES and FGF1 provided a promising therapy for anthracycline cardiotoxicity during chemotherapy.
Collapse
|
19
|
Matsiukevich D, House SL, Weinheimer C, Kovacs A, Ornitz DM. Fibroblast growth factor receptor signaling in cardiomyocytes is protective in the acute phase following ischemia-reperfusion injury. Front Cardiovasc Med 2022; 9:1011167. [PMID: 36211556 PMCID: PMC9539275 DOI: 10.3389/fcvm.2022.1011167] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 08/29/2022] [Indexed: 11/29/2022] Open
Abstract
Fibroblast growth factor receptors (FGFRs) are expressed in multiple cell types in the adult heart. Previous studies have shown a cardioprotective effect of some FGF ligands in cardiac ischemia-reperfusion (I/R) injury and a protective role for endothelial FGFRs in post-ischemic vascular remodeling. To determine the direct role FGFR signaling in cardiomyocytes in acute cardiac I/R injury, we inactivated Fgfr1 and Fgfr2 (CM-DCKO) or activated FGFR1 (CM-caFGFR1) in cardiomyocytes in adult mice prior to I/R injury. In the absence of injury, inactivation of Fgfr1 and Fgfr2 in adult cardiomyocytes had no effect on cardiac morphometry or function. When subjected to I/R injury, compared to controls, CM-DCKO mice had significantly increased myocyte death 1 day after reperfusion, and increased infarct size, cardiac dysfunction, and myocyte hypertrophy 7 days after reperfusion. No genotype-dependent effect was observed on post-ischemic cardiomyocyte cross-sectional area and vessel density in areas remote to the infarct. By contrast, transient activation of FGFR1 signaling in cardiomyocytes just prior to the onset of ischemia did not affect outcomes after cardiac I/R injury at 1 day and 7 days after reperfusion. These data demonstrate that endogenous cell-autonomous cardiomyocyte FGFR signaling supports the survival of cardiomyocytes in the acute phase following cardiac I/R injury and that this cardioprotection results in continued improved outcomes during cardiac remodeling. Combined with the established protective role of some FGF ligands and endothelial FGFR signaling in I/R injury, this study supports the development of therapeutic strategies that promote cardiomyocyte FGF signaling after I/R injury.
Collapse
Affiliation(s)
- Dzmitry Matsiukevich
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
- Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Stacey L. House
- Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
- Department of Emergency Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Carla Weinheimer
- Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Attila Kovacs
- Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - David M. Ornitz
- Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| |
Collapse
|
20
|
Xiao Y, Chen Y, Shao C, Wang Y, Hu S, Lei W. Strategies to improve the therapeutic effect of pluripotent stem cell-derived cardiomyocytes on myocardial infarction. Front Bioeng Biotechnol 2022; 10:973496. [PMID: 35992358 PMCID: PMC9388750 DOI: 10.3389/fbioe.2022.973496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/11/2022] [Indexed: 11/20/2022] Open
Abstract
Myocardial infarction (MI) is a common cardiovascular disease caused by permanent loss of cardiomyocytes and the formation of scar tissue due to myocardial ischemia. Mammalian cardiomyocytes lose their ability to proliferate almost completely in adulthood and are unable to repair the damage caused by MI. Therefore, transplantation of exogenous cells into the injured area for treatment becomes a promising strategy. Pluripotent stem cells (PSCs) have the ability to proliferate and differentiate into various cellular populations indefinitely, and pluripotent stem cell-derived cardiomyocytes (PSC-CMs) transplanted into areas of injury can compensate for part of the injuries and are considered to be one of the most promising sources for cell replacement therapy. However, the low transplantation rate and survival rate of currently transplanted PSC-CMs limit their ability to treat MI. This article focuses on the strategies of current research for improving the therapeutic efficacy of PSC-CMs, aiming to provide some inspiration and ideas for subsequent researchers to further enhance the transplantation rate and survival rate of PSC-CMs and ultimately improve cardiac function.
Collapse
Affiliation(s)
- Yang Xiao
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| | - Yihuan Chen
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| | - Chunlai Shao
- Department of Cardiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yaning Wang
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| | - Shijun Hu
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
- *Correspondence: Wei Lei, ; Shijun Hu,
| | - Wei Lei
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
- *Correspondence: Wei Lei, ; Shijun Hu,
| |
Collapse
|
21
|
Xu B, Li F, Zhang W, Su Y, Tang L, Li P, Joshi J, Yang A, Li D, Wang Z, Wang S, Xie J, Gu H, Zhu W. Identification of metabolic pathways underlying FGF1 and CHIR99021-mediated cardioprotection. iScience 2022; 25:104447. [PMID: 35707727 PMCID: PMC9189130 DOI: 10.1016/j.isci.2022.104447] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/16/2022] [Accepted: 05/18/2022] [Indexed: 12/05/2022] Open
Abstract
Acute myocardial infarction is a leading cause of death worldwide. We have previously identified two cardioprotective molecules — FGF1 and CHIR99021— that confer cardioprotection in mouse and pig models of acute myocardial infarction. Here, we aimed to determine if improved myocardial metabolism contributes to this cardioprotection. Nanofibers loaded with FGF1 and CHIR99021 were intramyocardially injected to ischemic myocardium of adult mice immediately following surgically induced myocardial infarction. Animals were euthanized 3 and 7 days later. Our data suggested that FGF1/CHIR99021 nanofibers enhanced the heart’s capacity to utilize glycolysis as an energy source and reduced the accumulation of branched-chain amino acids in ischemic myocardium. The impact of FGF1/CHIR99021 on metabolism was more obvious in the first three days post myocardial infarction. Taken together, these findings suggest that FGF1/CHIR99021 protects the heart against ischemic injury via improving myocardial metabolism which may be exploited for treatment of acute myocardial infarction in humans. FGF1/CHIR confer cardioprotection in myocardial infarction animals FGF1/CHIR enhance the capability of ischemic hearts to produce energy via glycolysis FGF1/CHIR reduce the abundance of branched chain amino acids in ischemic hearts This study reveals a novel approach to correct metabolic disorders in ischemic hearts
Collapse
Affiliation(s)
- Bing Xu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, USA 85259.,Department of Cardiology, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou 225001, China
| | - Fan Li
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, USA 85259.,Department of Kinesiology, South China Normal University, Guangzhou 510631, China
| | - Wenjing Zhang
- Center for Translational Science, Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Port St. Lucie, FL 34987, USA.,College of Health Solutions, Arizona State University, Phoenix, AZ 85287, USA
| | - Yajuan Su
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ling Tang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, USA 85259
| | - Pengsheng Li
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, USA 85259
| | - Jyotsna Joshi
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, USA 85259
| | - Aaron Yang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, USA 85259
| | - Dong Li
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, USA 85259
| | - Zhao Wang
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Shu Wang
- College of Health Solutions, Arizona State University, Phoenix, AZ 85287, USA
| | - Jingwei Xie
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Haiwei Gu
- Center for Translational Science, Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Port St. Lucie, FL 34987, USA.,College of Health Solutions, Arizona State University, Phoenix, AZ 85287, USA
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, USA 85259
| |
Collapse
|
22
|
Zhu W, Sun J, Bishop SP, Sadek H, Zhang J. Turning back the clock: A concise viewpoint of cardiomyocyte cell cycle activation for myocardial regeneration and repair. J Mol Cell Cardiol 2022; 170:15-21. [PMID: 35660800 PMCID: PMC9391298 DOI: 10.1016/j.yjmcc.2022.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 04/18/2022] [Accepted: 05/14/2022] [Indexed: 11/25/2022]
Abstract
Patients with acute myocardial infarction (MI) could progress to end-stage congestive heart failure, which is one of the most significant problems in public health. From the molecular and cellular perspective, heart failure often results from the loss of cardiomyocytes-the fundamental contractile unit of the heart-and the damage caused by myocardial injury in adult mammals cannot be repaired, in part because mammalian cardiomyocytes undergo cell-cycle arrest during the early perinatal period. However, recent studies in the hearts of neonatal small and large mammals suggest that the onset of cardiomyocyte cell-cycle arrest can be reversed, which may lead to the development of entirely new strategies for the treatment of heart failure. In this Viewpoint, we summarize these and other provocative findings about the cellular and molecular mechanisms that regulate cardiomyocyte proliferation and how they may be targeted to turn back the clock of cardiomyocyte cell-cycle arrest and improve recovery from cardiac injury and disease.
Collapse
Affiliation(s)
- Wuqiang Zhu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic, Scottsdale, AZ 85259, United States of America
| | - Jiacheng Sun
- Department of Biomedical Engineering, School of Medicine and School of Engineering, the University of Alabama at Birmingham, Birmingham, AL 35294, United States of America
| | - Sanford P Bishop
- Department of Biomedical Engineering, School of Medicine and School of Engineering, the University of Alabama at Birmingham, Birmingham, AL 35294, United States of America
| | - Hesham Sadek
- Division of Cardiovascular Diseases, UT Southwestern Medical Center, United States of America
| | - Jianyi Zhang
- Department of Biomedical Engineering, School of Medicine and School of Engineering, the University of Alabama at Birmingham, Birmingham, AL 35294, United States of America; Department of Medicine, Division of Cardiovascular Diseases, School of Medicine, the University of Alabama at Birmingham, Birmingham, AL 35294, United States of America.
| |
Collapse
|
23
|
Xu M, Guo YY, Li D, Cen XF, Qiu HL, Ma YL, Huang SH, Tang QZ. Screening of Lipid Metabolism-Related Gene Diagnostic Signature for Patients With Dilated Cardiomyopathy. Front Cardiovasc Med 2022; 9:853468. [PMID: 35433888 PMCID: PMC9010535 DOI: 10.3389/fcvm.2022.853468] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/16/2022] [Indexed: 11/24/2022] Open
Abstract
Background Dilated cardiomyopathy (DCM) is characterized by enlarged ventricular dimensions and systolic dysfunction and poor prognosis. Myocardial lipid metabolism appears abnormal in DCM. However, the mechanism of lipid metabolism disorders in DCM remains unclear. Methods A gene set variation analysis (GSVA) were performed to estimate pathway activity related to DCM progression. Three datasets and clinical data downloaded from the Gene Expression Omnibus (GEO), including dilated cardiomyopathy and donor hearts, were integrated to obtain gene expression profiles and identify differentially expressed genes related to lipid metabolism. GO enrichment analyses of differentially expressed lipid metabolism-related genes (DELs) were performed. The clinical information used in this study were obtained from GSE21610 dataset. Data from the EGAS00001003263 were used for external validation and our hospital samples were also tested the expression levels of these genes through RT-PCR. Subsequently, logistic regression model with the LASSO method for DCM prediction was established basing on the 7 DELs. Results GSVA analysis showed that the fatty acid metabolism was closely related to DCM progression. The integrated dataset identified 19 DELs, including 8 up-regulated and 11 down-regulated genes. A total of 7 DELs were identified by further external validation of the data from the EGAS00001003263 and verified by RT-PCR. By using the LASSO model, 6 genes, including CYP2J2, FGF1, ETNPPL, PLIN2, LPCAT3, and DGKG, were identified to construct a logistic regression model. The area under curve (AUC) values over 0.8 suggested the good performance of the model. Conclusion Integrated bioinformatic analysis of gene expression in DCM and the effective logistic regression model construct in our study may contribute to the early diagnosis and prevention of DCM in people with high risk of the disease.
Collapse
Affiliation(s)
- Man Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Ying-ying Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Dan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Xian-feng Cen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Hong-liang Qiu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Yu-lan Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Si-hui Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Qi-zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
- *Correspondence: Qi-zhu Tang,
| |
Collapse
|
24
|
Chen K, Rao Z, Dong S, Chen Y, Wang X, Luo Y, Gong F, Li X. Roles of the fibroblast growth factor signal transduction system in tissue injury repair. BURNS & TRAUMA 2022; 10:tkac005. [PMID: 35350443 PMCID: PMC8946634 DOI: 10.1093/burnst/tkac005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 12/13/2021] [Indexed: 12/13/2022]
Abstract
Following injury, tissue autonomously initiates a complex repair process, resulting in either partial recovery or regeneration of tissue architecture and function in most organisms. Both the repair and regeneration processes are highly coordinated by a hierarchy of interplay among signal transduction pathways initiated by different growth factors, cytokines and other signaling molecules under normal conditions. However, under chronic traumatic or pathological conditions, the reparative or regenerative process of most tissues in different organs can lose control to different extents, leading to random, incomplete or even flawed cell and tissue reconstitution and thus often partial restoration of the original structure and function, accompanied by the development of fibrosis, scarring or even pathogenesis that could cause organ failure and death of the organism. Ample evidence suggests that the various combinatorial fibroblast growth factor (FGF) and receptor signal transduction systems play prominent roles in injury repair and the remodeling of adult tissues in addition to embryonic development and regulation of metabolic homeostasis. In this review, we attempt to provide a brief update on our current understanding of the roles, the underlying mechanisms and clinical application of FGFs in tissue injury repair.
Collapse
Affiliation(s)
| | | | - Siyang Dong
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Department of breast surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yajing Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xulan Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yongde Luo
- Correspondence. Xiaokun Li, ; Fanghua Gong, ; Yongde Luo,
| | - Fanghua Gong
- Correspondence. Xiaokun Li, ; Fanghua Gong, ; Yongde Luo,
| | - Xiaokun Li
- Correspondence. Xiaokun Li, ; Fanghua Gong, ; Yongde Luo,
| |
Collapse
|
25
|
Stüdemann T, Weinberger F. The Guinea Pig Model in Cardiac Regeneration Research; Current Tissue Engineering Approaches and Future Directions. ADVANCED TECHNOLOGIES IN CARDIOVASCULAR BIOENGINEERING 2022:103-122. [DOI: 10.1007/978-3-030-86140-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
26
|
Fazil MHUT, Prasannan P, Wong BHS, Kottaiswamy A, Salim NSBM, Sze SK, Verma NK. GSK3β Interacts With CRMP2 and Notch1 and Controls T-Cell Motility. Front Immunol 2021; 12:680071. [PMID: 34975828 PMCID: PMC8718691 DOI: 10.3389/fimmu.2021.680071] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 11/30/2021] [Indexed: 11/22/2022] Open
Abstract
The trafficking of T-cells through peripheral tissues and into afferent lymphatic vessels is essential for immune surveillance and an adaptive immune response. Glycogen synthase kinase 3β (GSK3β) is a serine/threonine kinase and regulates numerous cell/tissue-specific functions, including cell survival, metabolism, and differentiation. Here, we report a crucial involvement of GSK3β in T-cell motility. Inhibition of GSK3β by CHIR-99021 or siRNA-mediated knockdown augmented the migratory behavior of human T-lymphocytes stimulated via an engagement of the T-cell integrin LFA-1 with its ligand ICAM-1. Proteomics and protein network analysis revealed ongoing interactions among GSK3β, the surface receptor Notch1 and the cytoskeletal regulator CRMP2. LFA-1 stimulation in T-cells reduced Notch1-dependent GSK3β activity by inducing phosphorylation at Ser9 and its nuclear translocation accompanied by the cleaved Notch1 intracellular domain and decreased GSK3β-CRMP2 association. LFA-1-induced or pharmacologic inhibition of GSK3β in T-cells diminished CRMP2 phosphorylation at Thr514. Although substantial amounts of CRMP2 were localized to the microtubule-organizing center in resting T-cells, this colocalization of CRMP2 was lost following LFA-1 stimulation. Moreover, the migratory advantage conferred by GSK3β inhibition in T-cells by CHIR-99021 was lost when CRMP2 expression was knocked-down by siRNA-induced gene silencing. We therefore conclude that GSK3β controls T-cell motility through interactions with CRMP2 and Notch1, which has important implications in adaptive immunity, T-cell mediated diseases and LFA-1-targeted therapies.
Collapse
Affiliation(s)
| | - Praseetha Prasannan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| | - Brandon Han Siang Wong
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
- Interdisciplinary Graduate Programme, NTU Institute for Health Technologies (HealthTech NTU), Nanyang Technological University Singapore, Singapore, Singapore
| | - Amuthavalli Kottaiswamy
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| | | | - Siu Kwan Sze
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore, Singapore
| | - Navin Kumar Verma
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
- *Correspondence: Navin Kumar Verma,
| |
Collapse
|
27
|
Chang T, Liu C, Lu K, Wu Y, Xu M, Yu Q, Shen Z, Jiang T, Zhang Y. Biomaterials based cardiac patches for the treatment of myocardial infarction. JOURNAL OF MATERIALS SCIENCE & TECHNOLOGY 2021; 94:77-89. [DOI: 10.1016/j.jmst.2021.03.062] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
28
|
Wu X, Wang D, Qin K, Iroegbu CD, Xiang K, Zhou Y, Guan Q, Tang W, Peng J, Guo J, Yang J, Fan C. Cardiac Repair With Echocardiography-Guided Multiple Percutaneous Left Ventricular Intramyocardial Injection of hiPSC-CMs After Myocardial Infarction. Front Cardiovasc Med 2021; 8:768873. [PMID: 34805322 PMCID: PMC8600116 DOI: 10.3389/fcvm.2021.768873] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/15/2021] [Indexed: 12/15/2022] Open
Abstract
Objective: We investigated the potency of cardiac repair based on echocardiography-guided multiple percutaneous left ventricular intramyocardial injection of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) after myocardial infarction (MI). Methods: Mice with surgically induced MI were randomly divided into three groups (n = 8 in each group) and subjected to echocardiography-guided percutaneous left ventricular infarcted border injection of hiPSC-CMs (single dose; 10 μl 3 × 105 cells) or repeated injections of hiPSC-CMs at post-MI weeks 1 and 2 (multiple doses). The sham group of animals underwent all surgical procedures necessary for MI induction except for ligation. Then 4 weeks after MI, heart function was measured with transthoracic echocardiography. Engraftment was evaluated through the detection of human-specific cardiac troponin T. Infarct size and collagen volume were calculated with Sirius Red/Fast Green staining. Angiogenesis was evaluated with isolectin B4 staining. Cardiac remodeling was evaluated from the cardiomyocyte minimal fiber diameter in the infarcted border zone. Apoptosis was detected via TdT-mediated dUTP Nick-End Labeling (TUNEL) staining in cardiomyocytes from the infarcted border zone. Results: No mice died after echocardiography-guided percutaneous left ventricular intramyocardial injection. hiPSC-CMs were about nine-fold higher in the multiple-dose group at week 4 compared to the single-dose group. Multiple-dose transplantation was associated with significant improvement in left ventricular function, infarct size, angiogenesis, cardiac remodeling, and cardiomyocyte apoptosis. Conclusion: Echocardiography-guided multiple percutaneous left ventricular intramyocardial injection is a feasible, satisfactory, repeatable, relatively less invasive, and effective method of delivering cell therapy. The delivery of hiPSC-CMs indicates a novel therapy for MI.
Collapse
Affiliation(s)
- Xun Wu
- Department of Cardiovascular Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Di Wang
- Hunan Provincial Key Laboratory of Cardiovascular Research, Changsha, China
| | - Kele Qin
- Department of Cardiovascular Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Chukwuemeka Daniel Iroegbu
- Department of Cardiovascular Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Kun Xiang
- Department of Cardiovascular Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yuanjing Zhou
- Hunan Provincial Key Laboratory of Cardiovascular Research, Changsha, China
| | - Qing Guan
- Department of Cardiovascular Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Weijie Tang
- Department of Cardiovascular Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Jun Peng
- Hunan Provincial Key Laboratory of Cardiovascular Research, Changsha, China
| | - Jianjun Guo
- Hunan Fangsheng Pharmaceutical Co., Ltd., Changsha, China
| | - Jinfu Yang
- Department of Cardiovascular Surgery, Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Provincial Key Laboratory of Cardiovascular Research, Changsha, China
| | - Chengming Fan
- Department of Cardiovascular Surgery, Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Provincial Key Laboratory of Cardiovascular Research, Changsha, China.,Hunan Fangsheng Pharmaceutical Co., Ltd., Changsha, China
| |
Collapse
|
29
|
Yu D, Wang X, Ye L. Cardiac Tissue Engineering for the Treatment of Myocardial Infarction. J Cardiovasc Dev Dis 2021; 8:jcdd8110153. [PMID: 34821706 PMCID: PMC8617685 DOI: 10.3390/jcdd8110153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/04/2021] [Indexed: 11/26/2022] Open
Abstract
Poor cell engraftment rate is one of the primary factors limiting the effectiveness of cell transfer therapy for cardiac repair. Recent studies have shown that the combination of cell-based therapy and tissue engineering technology can improve stem cell engraftment and promote the therapeutic effects of the treatment for myocardial infarction. This mini-review summarizes the recent progress in cardiac tissue engineering of cardiovascular cells from differentiated human pluripotent stem cells (PSCs), highlights their therapeutic applications for the treatment of myocardial infarction, and discusses the present challenges of cardiac tissue engineering and possible future directions from a clinical perspective.
Collapse
Affiliation(s)
- Dongmin Yu
- Department of Cardiovascular Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China;
| | - Xiaowei Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China;
- Correspondence: (X.W.); (L.Y.); Tel.: +86-02568303105 (X.W.); +65-67042193 2 (L.Y.)
| | - Lei Ye
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore 169609, Singapore
- Correspondence: (X.W.); (L.Y.); Tel.: +86-02568303105 (X.W.); +65-67042193 2 (L.Y.)
| |
Collapse
|
30
|
Eschenhagen T, Ridders K, Weinberger F. How to repair a broken heart with pluripotent stem cell-derived cardiomyocytes. J Mol Cell Cardiol 2021; 163:106-117. [PMID: 34687723 DOI: 10.1016/j.yjmcc.2021.10.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/08/2021] [Accepted: 10/12/2021] [Indexed: 01/14/2023]
Abstract
Heart regeneration addresses a central problem in cardiology, the irreversibility of the loss of myocardium that eventually leads to heart failure. True restoration of heart function can only be achieved by remuscularization, i.e. replacement of lost myocardium by new, force-developing heart muscle. With the availability of principally unlimited human cardiomyocytes from pluripotent stem cells, one option to remuscularize the injured heart is to produce large numbers of cardiomyocytes plus/minus other cardiovascular cell types or progenitors ex vivo and apply them to the heart, either by injection or application as a patch. Exciting progress over the past decade has led to the first clinical applications, but important questions remain. Academic and increasingly corporate activity is ongoing to answer them and optimize the approach to finally develop a true regenerative therapy of heart failure.
Collapse
Affiliation(s)
- Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| | | | - Florian Weinberger
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| |
Collapse
|
31
|
Hesselbarth R, Esser TU, Roshanbinfar K, Schrüfer S, Schubert DW, Engel FB. CHIR99021 Promotes hiPSC-Derived Cardiomyocyte Proliferation in Engineered 3D Microtissues. Adv Healthc Mater 2021; 10:e2100926. [PMID: 34499814 PMCID: PMC11468594 DOI: 10.1002/adhm.202100926] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/30/2021] [Indexed: 02/05/2023]
Abstract
Cardiac tissue engineering is a promising strategy to generate human cardiac tissues for modeling cardiac diseases, screening for therapeutic drugs, and repairing the injured heart. Yet, several issues remain to be resolved including the generation of tissues with high cardiomyocyte density. Here, it is shown that the integration of the glycogen synthase kinase-3β inhibitor CHIR99021 in collagen I hydrogels promotes proliferation of human-induced pluripotent stem cell-derived (hiPSC) cardiomyocytes post-fabrication improving contractility of and calcium flow in engineered 3D cardiac microtissues. CHIR99021 has no effect on the gelation kinetics or the mechanical properties of collagen I hydrogels. Analysis of cell density and proliferation based on Ki-67 staining indicates that integration of CHIR99021 together with external CHIR99021 stimulation increases hiPSC-cardiomyocyte number by ≈2-fold within 7 d post-fabrication. Analysis of the contractility of engineered cardiac tissues after another 3 d in the absence of external CHIR99021 shows that CHIR99021-induced hiPSC-cardiomyocyte proliferation results in synchronized calcium flow, rhythmic beating, increased speed of contraction and contraction amplitude, and reduced peak-to-peak time. The CHIR99021-stimulated engineered cardiac microtissues exhibit spontaneous rhythmic contractions for at least 35 d. Collectively, the data demonstrate the potential of induced cardiomyocyte proliferation to enhance engineered cardiac microtissues by increasing cardiomyocyte density.
Collapse
Affiliation(s)
- Ramona Hesselbarth
- Experimental Renal and Cardiovascular ResearchDepartment of NephropathologyInstitute of PathologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)Muscle Research Center Erlangen (MURCE)Erlangen91054Germany
| | - Tilman U. Esser
- Experimental Renal and Cardiovascular ResearchDepartment of NephropathologyInstitute of PathologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)Muscle Research Center Erlangen (MURCE)Erlangen91054Germany
| | - Kaveh Roshanbinfar
- Experimental Renal and Cardiovascular ResearchDepartment of NephropathologyInstitute of PathologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)Muscle Research Center Erlangen (MURCE)Erlangen91054Germany
| | - Stefan Schrüfer
- Institute of Polymer MaterialsFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)Martensstraße 7Erlangen91058Germany
| | - Dirk W. Schubert
- Institute of Polymer MaterialsFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)Martensstraße 7Erlangen91058Germany
| | - Felix B. Engel
- Experimental Renal and Cardiovascular ResearchDepartment of NephropathologyInstitute of PathologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)Muscle Research Center Erlangen (MURCE)Erlangen91054Germany
| |
Collapse
|
32
|
Marchini A, Gelain F. Synthetic scaffolds for 3D cell cultures and organoids: applications in regenerative medicine. Crit Rev Biotechnol 2021; 42:468-486. [PMID: 34187261 DOI: 10.1080/07388551.2021.1932716] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Three-dimensional (3D) cell cultures offer an unparalleled platform to recreate spatial arrangements of cells in vitro. 3D cell culture systems have gained increasing interest due to their evident advantages in providing more physiologically relevant information and more predictive data compared to their two-dimensional (2D) counterpart. Design and well-established fabrication of organoids (a particular type of 3D cell culture system) are nowadays considered a pivotal achievement for their ability to replicate in vitro cytoarchitecture and the functionalities of an organ. In this condition, pluripotent stem cells self-organize into 3D structures mimicking the in vivo microenvironments, architectures and multi-lineage differentiation. Scaffolds are key supporting elements in these 3D constructs, and Matrigel is the most commonly used matrix despite its relevant translation limitations including animal-derived sources, non-defined composition, batch-to-batch variability and poorly tailorable properties. Alternatively, 3D synthetic scaffolds, including self-assembling peptides, show promising biocompatibility and biomimetic properties, and can be tailored on specific target tissue/cells. In this review, we discuss the recent advances on 3D cell culture systems and organoids, promising tools for tissue engineering and regenerative medicine applications. For this purpose, we will describe the current state-of-the-art on 3D cell culture systems and organoids based on currently available synthetic-based biomaterials (including tailored self-assembling peptides) either tested in in vivo animal models or developed in vitro with potential application in the field of tissue engineering, with the aim to inspire researchers to take on such promising platforms for clinical applications in the near future.
Collapse
Affiliation(s)
- Amanda Marchini
- Tissue Engineering Unit, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies-ISBReMIT, Fondazione IRCSS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Fabrizio Gelain
- Tissue Engineering Unit, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies-ISBReMIT, Fondazione IRCSS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.,Center for Nanomedicine and Tissue Engineering (CNTE), ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| |
Collapse
|
33
|
Tang S, Fan C, Iroegbu CD, Zhou W, Zhang Z, Wu M, Chen W, Wu X, Peng J, Li Z, Yang J. TMSB4 Overexpression Enhances the Potency of Marrow Mesenchymal Stromal Cells for Myocardial Repair. Front Cell Dev Biol 2021; 9:670913. [PMID: 34178995 PMCID: PMC8221609 DOI: 10.3389/fcell.2021.670913] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/06/2021] [Indexed: 01/08/2023] Open
Abstract
Objective The actin-sequestering proteins, thymosin beta-4 (Tβ4) and hypoxia-inducible factor (HIF)-1α, are known to be associated with angiogenesis after myocardial infarction (MI). Herein, we aimed to identify the mechanism of HIF-1α induction by Tβ4 and investigate the effects of bone marrow mesenchymal stromal cells (BMMSCs) transfected with the Tβ4 gene (TMSB4) in a rat model of MI. Methods Rat BMMSCs were isolated, cultured, and transfected with the TMSB4 gene by using the lentivirus-mediated method. Rats with surgically induced MI were randomly divided into three groups (n = 9/group); after 1 week, the rats were injected at the heart infarcted border zone with TMSB4-overexpressed BMMSCs (BMMSC-TMSB4OE), wild-type BMMSCs that expressed normal levels of TMSB4 (BMMSC-TMSB4WT), or medium (MI). The fourth group of animals (n = 9) underwent all surgical procedures necessary for MI induction except for the ligation step (Sham). Four weeks after the injection, heart function was measured using transthoracic echocardiography. Infarct size was calculated by TTC staining, and collagen volume was measured by Masson staining. Angiogenesis in the infarcted heart area was evaluated by CD31 immunofluorescence histochemistry. In vitro experiments were carried out to observe the effect of exogenous Tβ4 on HIF-1α and explore the various possible mechanism(s). Results In vivo experiments showed that vascular density 4 weeks after treatment was about twofold higher in BMMSC-TMSB4OE-treated animals than in BMMSC-TMSB4WT-treated animals (p < 0.05). The cardiac function and infarct size significantly improved in both cell-treatment groups compared to controls. Notably, the cardiac function and infarct size were most prominent in BMMSC-TMSB4OE-treated animals (both p < 0.05). HIF-1α and phosphorylated HIF-1α (p-HIF-1α) in vitro were significantly enhanced by exogenous Tβ4, which was nonetheless blocked by the factor-inhibiting HIF (FIH) promoter (YC-1). The expression of prolyl hydroxylase domain proteins (PHD) was decreased upon treatment with Tβ4 and further decreased with the combined treatment of Tβ4 and FG-4497 (a specific PHD inhibitor). Conclusion TMSB4-transfected BMMSCs might significantly improve recovery from myocardial ischemia and promote the generation of HIF-1α and p-HIF-1α via the AKT pathway, and inhibit the degradation of HIF-1α via the PHD and FIH pathways.
Collapse
Affiliation(s)
- Shiyuan Tang
- Department of the Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chengming Fan
- Department of the Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chukwuemeka Daniel Iroegbu
- Department of the Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wenwu Zhou
- Department of the Cardiovascular Surgery of the Hunan Provincial People's Hospital, Changsha, China
| | - Zhigong Zhang
- Department of the Cardiovascular Surgery of the Hunan Provincial People's Hospital, Changsha, China
| | - Ming Wu
- Department of the Cardiovascular Surgery of the Hunan Provincial People's Hospital, Changsha, China
| | - Wangping Chen
- Department of the Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoming Wu
- Department of the Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jun Peng
- Hunan Provincial Key Laboratory of Cardiovascular Research, Changsha, China
| | - Zhihong Li
- Institute of Senile and Aging Diseases, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jinfu Yang
- Department of the Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Provincial Key Laboratory of Cardiovascular Research, Changsha, China
| |
Collapse
|
34
|
Bian W, Chen W, Nguyen T, Zhou Y, Zhang J. miR-199a Overexpression Enhances the Potency of Human Induced-Pluripotent Stem-Cell-Derived Cardiomyocytes for Myocardial Repair. Front Pharmacol 2021; 12:673621. [PMID: 34149424 PMCID: PMC8209326 DOI: 10.3389/fphar.2021.673621] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 05/18/2021] [Indexed: 01/07/2023] Open
Abstract
Mammalian cardiomyocytes exit the cell cycle during the perinatal period, and although cardiomyocytes differentiated from human induced-pluripotent stem cells (hiPSC-CMs) are phenotypically immature, their intrinsic cell-cycle activity remains limited. Thus, neither endogenous cardiomyocytes nor the small number of transplanted hiPSC-CMs that are engrafted by infarcted hearts can remuscularize the myocardial scar. microRNAs are key regulators of cardiomyocyte proliferation, and when adeno-associated viruses coding for microRNA-199a (miR-199a) expression were injected directly into infarcted pig hearts, measures of cardiac function and fibrosis significantly improved, but the treatment was also associated with lethal arrhythmia. For the studies reported here, the same vector (AAV6-miR-199a) was transduced into hiPSC-CMs, and the cells were subsequently evaluated in a mouse model of myocardial infarction. AAV6-mediated miR-199a overexpression increased proliferation in both cultured and transplanted hiPSC-CMs, and measures of left ventricular ejection fraction, fractional shortening, and scar size were significantly better in mice treated with miR-199a-overexpressing hiPSC-CMs than with hiPSC-CMs that had been transduced with a control vector. Furthermore, although this investigation was not designed to characterize the safety of transplanted AAV6-miR-199a-transduced hiPSC-CMs, there was no evidence of sudden death. Collectively, these results support future investigations of miR-199a-overexpressing hiPSC-CMs in large animals.
Collapse
Affiliation(s)
- Weihua Bian
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Wangping Chen
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Thanh Nguyen
- Informatics Institute, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yang Zhou
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jianyi Zhang
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Medicine/Cardiovascular Diseases, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
35
|
Khosravi F, Ahmadvand N, Bellusci S, Sauer H. The Multifunctional Contribution of FGF Signaling to Cardiac Development, Homeostasis, Disease and Repair. Front Cell Dev Biol 2021; 9:672935. [PMID: 34095143 PMCID: PMC8169986 DOI: 10.3389/fcell.2021.672935] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/20/2021] [Indexed: 12/13/2022] Open
Abstract
The current focus on cardiovascular research reflects society’s concerns regarding the alarming incidence of cardiac-related diseases and mortality in the industrialized world and, notably, an urgent need to combat them by more efficient therapies. To pursue these therapeutic approaches, a comprehensive understanding of the mechanism of action for multifunctional fibroblast growth factor (FGF) signaling in the biology of the heart is a matter of high importance. The roles of FGFs in heart development range from outflow tract formation to the proliferation of cardiomyocytes and the formation of heart chambers. In the context of cardiac regeneration, FGFs 1, 2, 9, 16, 19, and 21 mediate adaptive responses including restoration of cardiac contracting rate after myocardial infarction and reduction of myocardial infarct size. However, cardiac complications in human diseases are correlated with pathogenic effects of FGF ligands and/or FGF signaling impairment. FGFs 2 and 23 are involved in maladaptive responses such as cardiac hypertrophic, fibrotic responses and heart failure. Among FGFs with known causative (FGFs 2, 21, and 23) or protective (FGFs 2, 15/19, 16, and 21) roles in cardiac diseases, FGFs 15/19, 21, and 23 display diagnostic potential. The effective role of FGFs on the induction of progenitor stem cells to cardiac cells during development has been employed to boost the limited capacity of postnatal cardiac repair. To renew or replenish damaged cardiomyocytes, FGFs 1, 2, 10, and 16 were tested in (induced-) pluripotent stem cell-based approaches and for stimulation of cell cycle re-entry in adult cardiomyocytes. This review will shed light on the wide range of beneficiary and detrimental actions mediated by FGF ligands and their receptors in the heart, which may open new therapeutic avenues for ameliorating cardiac complications.
Collapse
Affiliation(s)
- Farhad Khosravi
- Department of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Negah Ahmadvand
- Cardio-Pulmonary Institute, Justus Liebig University Giessen, Giessen, Germany
| | - Saverio Bellusci
- Cardio-Pulmonary Institute, Justus Liebig University Giessen, Giessen, Germany
| | - Heinrich Sauer
- Department of Physiology, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
36
|
Zhou NQ, Fang ZX, Huang N, Zuo Y, Qiu Y, Guo LJ, Song P, Xu J, Wan GR, Tian XQ, Yin YL, Li P. aFGF Targeted Mediated by Novel Nanoparticles-Microbubble Complex Combined With Ultrasound-Targeted Microbubble Destruction attenuates Doxorubicin-Induced Heart Failure via Anti-Apoptosis and Promoting Cardiac Angiogenesis. Front Pharmacol 2021; 12:607785. [PMID: 33986662 PMCID: PMC8111001 DOI: 10.3389/fphar.2021.607785] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 02/15/2021] [Indexed: 11/30/2022] Open
Abstract
The purpose of this study was to evaluate the protective effect of acidic fibroblast growth factor targeted mediated by novel nanoparticles–cationic lipid microbubbles complex (aFGF–NP + CPMBs) combined with ultrasound targeted microbubble destruction (UTMD)on doxorubicin–induced heart failure (HF)and its mechanism. Heart failure rats induced by intraperitoneal injection with doxorubicin (DOX) to achieve cummulative dose of 15mg/kg for continuous 6 weeks showed left ventricular dysfunction, seriously oxidative stress, cardiomyocyte apoptosis, and decrease of myocardial vascular density. In contrast, aFGF–NP + CPMBs combined with UTMD therapy (3ug/kg, caudal vein injection, twice a week, 6weeks)prominently ameliorated left ventricular dysfunction by increased ejection fraction (EF) and fractional shortening (FS), decreased brain natriuretic peptide (BNP); strengthened the ability of antioxidant stress confirmed by increasing the activity of SOD and reducing the production of MDA; exerted the effect of anti–cardiomyocyte apoptosis and promotion angiogenesis by inhibited Bax expression and increased Bcl–2 expression and platelet endothelial cell adhesion molecule (CD31) expression. Taken together, the research suggested that aFGF targeted mediated by novel nanoparticles–cationic lipid microbubbles complex combined with UTMD should be a promising targeted treatment for heart failure.
Collapse
Affiliation(s)
- Nan-Qian Zhou
- Department of Ultrasonography, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,.Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Zhi-Xin Fang
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,.Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.,College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Ning Huang
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,.Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.,College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Yue Zuo
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yue Qiu
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,.Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.,College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Li-Juan Guo
- Department of Oncology, First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Ping Song
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,.Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.,College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Jian Xu
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,.Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.,College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Guang-Rui Wan
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,.Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.,College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Xin-Qiao Tian
- Department of Ultrasonography, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Ya-Ling Yin
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Peng Li
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,.Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.,College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
37
|
Regulatory role of endogenous and exogenous fibroblast growth factor 1 in the cardiovascular system and related diseases. Pharmacol Res 2021; 169:105596. [PMID: 33831565 DOI: 10.1016/j.phrs.2021.105596] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 03/07/2021] [Accepted: 03/31/2021] [Indexed: 12/15/2022]
Abstract
Fibroblast growth factor 1 (FGF1) has a critical regulatory role in the development of the cardiovascular system (CVS) and is strongly associated with the progression or treatment of cardiovascular diseases (CVDs). However, the regulatory mechanisms of FGF1 in CVS and CVDs have not yet been fully elucidated. Therefore, this review article summarized the existing literature reports on the role of FGF1 in CVS under physiological and pathological conditions. First, the expression and physiological functions of endogenous FGF1 is fully demonstrated. Then, we analyzed the role of exogenous FGF1 in normal CVS and related pathological processes. Specifically, the potential signaling pathways might be mediated by FGF1 in CVDs treatment is discussed in detail. In addition, the barriers and feasible solutions for the application of FGF1 are further analyzed. Finally, we highlight therapeutic considerations of FGF1 for CVDs in the future. Thus, this article may be as a reference to provide some ideas for the follow-up research.
Collapse
|
38
|
Pretorius D, Serpooshan V, Zhang J. Nano-Medicine in the Cardiovascular System. Front Pharmacol 2021; 12:640182. [PMID: 33746761 PMCID: PMC7969876 DOI: 10.3389/fphar.2021.640182] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 01/19/2021] [Indexed: 01/19/2023] Open
Abstract
Nano-medicines that include nanoparticles, nanocomposites, small molecules, and exosomes represent new viable sources for future therapies for the dysfunction of cardiovascular system, as well as the other important organ systems. Nanomaterials possess special properties ranging from their intrinsic physicochemical properties, surface energy and surface topographies which can illicit advantageous cellular responses within the cardiovascular system, making them exceptionally valuable in future clinical translation applications. The success of nano-medicines as future cardiovascular theranostic agents requires a comprehensive understanding of the intersection between nanomaterial and the biomedical fields. In this review, we highlight some of the major types of nano-medicine systems that are currently being explored in the cardiac field. This review focusses on the major differences between the systems, and how these differences affect the specific therapeutic or diagnostic applications. The important concerns relevant to cardiac nano-medicines, including cellular responses, toxicity of the different nanomaterials, as well as cardio-protective and regenerative capabilities are discussed. In this review an overview of the current development of nano-medicines specific to the cardiac field is provided, discussing the diverse nature and applications of nanomaterials as therapeutic and diagnostic agents.
Collapse
Affiliation(s)
- Danielle Pretorius
- Department of Biomedical Engineering, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States.,Department of Biomedical Engineering, School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Vahid Serpooshan
- Emory Children's Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Jianyi Zhang
- Department of Biomedical Engineering, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States.,Department of Biomedical Engineering, School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
39
|
Wang L, Serpooshan V, Zhang J. Engineering Human Cardiac Muscle Patch Constructs for Prevention of Post-infarction LV Remodeling. Front Cardiovasc Med 2021; 8:621781. [PMID: 33718449 PMCID: PMC7952323 DOI: 10.3389/fcvm.2021.621781] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 02/04/2021] [Indexed: 12/20/2022] Open
Abstract
Tissue engineering combines principles of engineering and biology to generate living tissue equivalents for drug testing, disease modeling, and regenerative medicine. As techniques for reprogramming human somatic cells into induced pluripotent stem cells (iPSCs) and subsequently differentiating them into cardiomyocytes and other cardiac cells have become increasingly efficient, progress toward the development of engineered human cardiac muscle patch (hCMP) and heart tissue analogs has accelerated. A few pilot clinical studies in patients with post-infarction LV remodeling have been already approved. Conventional methods for hCMP fabrication include suspending cells within scaffolds, consisting of biocompatible materials, or growing two-dimensional sheets that can be stacked to form multilayered constructs. More recently, advanced technologies, such as micropatterning and three-dimensional bioprinting, have enabled fabrication of hCMP architectures at unprecedented spatiotemporal resolution. However, the studies working on various hCMP-based strategies for in vivo tissue repair face several major obstacles, including the inadequate scalability for clinical applications, poor integration and engraftment rate, and the lack of functional vasculature. Here, we review many of the recent advancements and key concerns in cardiac tissue engineering, focusing primarily on the production of hCMPs at clinical/industrial scales that are suitable for administration to patients with myocardial disease. The wide variety of cardiac cell types and sources that are applicable to hCMP biomanufacturing are elaborated. Finally, some of the key challenges remaining in the field and potential future directions to address these obstacles are discussed.
Collapse
Affiliation(s)
- Lu Wang
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Vahid Serpooshan
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA, United States
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Children's Healthcare of Atlanta, Atlanta, GA, United States
| | - Jianyi Zhang
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
40
|
Smagul S, Kim Y, Smagulova A, Raziyeva K, Nurkesh A, Saparov A. Biomaterials Loaded with Growth Factors/Cytokines and Stem Cells for Cardiac Tissue Regeneration. Int J Mol Sci 2020; 21:E5952. [PMID: 32824966 PMCID: PMC7504169 DOI: 10.3390/ijms21175952] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 12/17/2022] Open
Abstract
Myocardial infarction causes cardiac tissue damage and the release of damage-associated molecular patterns leads to activation of the immune system, production of inflammatory mediators, and migration of various cells to the site of infarction. This complex response further aggravates tissue damage by generating oxidative stress, but it eventually heals the infarction site with the formation of fibrotic tissue and left ventricle remodeling. However, the limited self-renewal capability of cardiomyocytes cannot support sufficient cardiac tissue regeneration after extensive myocardial injury, thus, leading to an irreversible decline in heart function. Approaches to improve cardiac tissue regeneration include transplantation of stem cells and delivery of inflammation modulatory and wound healing factors. Nevertheless, the harsh environment at the site of infarction, which consists of, but is not limited to, oxidative stress, hypoxia, and deficiency of nutrients, is detrimental to stem cell survival and the bioactivity of the delivered factors. The use of biomaterials represents a unique and innovative approach for protecting the loaded factors from degradation, decreasing side effects by reducing the used dosage, and increasing the retention and survival rate of the loaded cells. Biomaterials with loaded stem cells and immunomodulating and tissue-regenerating factors can be used to ameliorate inflammation, improve angiogenesis, reduce fibrosis, and generate functional cardiac tissue. In this review, we discuss recent findings in the utilization of biomaterials to enhance cytokine/growth factor and stem cell therapy for cardiac tissue regeneration in small animals with myocardial infarction.
Collapse
Affiliation(s)
| | | | | | | | | | - Arman Saparov
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (S.S.); (Y.K.); (A.S.); (K.R.); (A.N.)
| |
Collapse
|
41
|
Fan C, Joshi J, Li F, Xu B, Khan M, Yang J, Zhu W. Nanoparticle-Mediated Drug Delivery for Treatment of Ischemic Heart Disease. Front Bioeng Biotechnol 2020; 8:687. [PMID: 32671049 PMCID: PMC7326780 DOI: 10.3389/fbioe.2020.00687] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 06/02/2020] [Indexed: 12/26/2022] Open
Abstract
The regenerative capacity of an adult cardiac tissue is insufficient to repair the massive loss of heart tissue, particularly cardiomyocytes (CMs), following ischemia or other catastrophic myocardial injuries. The delivery methods of therapeutics agents, such as small molecules, growth factors, exosomes, cells, and engineered tissues have significantly advanced in medical science. Furthermore, with the controlled release characteristics, nanoparticle (NP) systems carrying drugs are promising in enhancing the cardioprotective potential of drugs in patients with cardiac ischemic events. NPs can provide sustained exposure precisely to the infarcted heart via direct intramyocardial injection or intravenous injection with active targets. In this review, we present the recent advances and challenges of different types of NPs loaded with agents for the repair of myocardial infarcted heart tissue.
Collapse
Affiliation(s)
- Chengming Fan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jyotsna Joshi
- Department of Cardiovascular Diseases, Mayo Clinic, Scottsdale, AZ, United States
| | - Fan Li
- Department of Cardiovascular Diseases, Mayo Clinic, Scottsdale, AZ, United States
| | - Bing Xu
- Department of Cardiovascular Diseases, Mayo Clinic, Scottsdale, AZ, United States
| | - Mahmood Khan
- Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Jinfu Yang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Mayo Clinic, Scottsdale, AZ, United States.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
42
|
Shi C, Xie H, Ma Y, Yang Z, Zhang J. Nanoscale Technologies in Highly Sensitive Diagnosis of Cardiovascular Diseases. Front Bioeng Biotechnol 2020; 8:531. [PMID: 32582663 PMCID: PMC7289988 DOI: 10.3389/fbioe.2020.00531] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 05/04/2020] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular diseases (CVD) are the leading cause of death and morbidity in the world and are a major contributor to healthcare costs. Although enormous progress has been made in diagnosing CVD, there is an urgent need for more efficient early detection and the development of novel diagnostic tools. Currently, CVD diagnosis relies primarily on clinical symptoms based on molecular imaging (MOI) or biomarkers associated with CVDs. However, sensitivity, specificity, and accuracy of the assay are still challenging for early-stage CVDs. Nanomaterial platform has been identified as a promising candidate for improving the practical usage of diagnostic tools because of their unique physicochemical properties. In this review article, we introduced cardiac biomarkers and imaging techniques that are currently used for CVD diagnosis. We presented the applications of various nanotechnologies on diagnosis within cardiac immunoassays (CIAs) and molecular imaging. We also summarized and compared different cardiac immunoassays based on their sensitivities and working ranges of biomarkers.
Collapse
Affiliation(s)
- Chaohong Shi
- Department of Rehabilitation Medicine, The First People’s Hospital of Wenling, Wenzhou Medical University, Wenling, China
| | - Haotian Xie
- Department of Mathematics, The Ohio State University, Columbus, OH, United States
| | - Yifan Ma
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, United States
| | - Zhaogang Yang
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Jingjing Zhang
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, United States
| |
Collapse
|