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 2024:S1742-7061(24)00719-0. [PMID: 39643223 DOI: 10.1016/j.actbio.2024.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
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
|
3
|
Furuno K, Suzuki K, Sakai S. Transduction and Genome Editing of the Heart with Adeno-Associated Viral Vectors Loaded onto Electrospun Polydioxanone Nonwoven Fabrics. Biomolecules 2024; 14:506. [PMID: 38672522 PMCID: PMC11047894 DOI: 10.3390/biom14040506] [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: 03/11/2024] [Revised: 04/05/2024] [Accepted: 04/19/2024] [Indexed: 04/28/2024] Open
Abstract
In this study, we introduce electrospun polydioxanone (PDO) nonwoven fabrics as a platform for the delivery of adeno-associated virus (AAV) vectors for transduction and genome editing by adhering them to organ surfaces, including the heart. AAV vectors were loaded onto the PDO fabrics by soaking the fabrics in a solution containing AAV vectors. In vitro, the amount of AAV vectors loaded onto the fabrics could be adjusted by changing their concentration in the solution, and the number of cells expressing the green fluorescent protein (GFP) encoded by the AAV vectors increased in correlation with the increasing amount of loaded AAV vectors. In vivo, both transduction and genome editing resulted in the observation of GFP expression around AAV vector-loaded PDO fabrics attached to the surfaces of mouse hearts, indicating effective transduction and expression at the target site. These results demonstrate the great potential of electrospun PDO nonwoven fabrics carrying therapeutic AAV vectors for gene therapy.
Collapse
Affiliation(s)
- Kotoko Furuno
- Department of Materials Engineering Science, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyama-cho, Toyonaka 560-8531, Japan;
| | - Keiichiro Suzuki
- Department of Materials Engineering Science, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyama-cho, Toyonaka 560-8531, Japan;
- Institute for Advanced Co-Creation Studies, Osaka University, 1-3 Machikaneyama-cho, Toyonaka 560-8531, Japan
- Graduate School of Frontier Bioscience, Osaka University, 1-3 Yamadaoka, Suita 565-0871, Japan
| | - Shinji Sakai
- Department of Materials Engineering Science, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyama-cho, Toyonaka 560-8531, Japan;
| |
Collapse
|
4
|
Moradikhah F, Shabani I, Tafazzoli Shadpour M. Fabrication of a tailor-made conductive polyaniline/ascorbic acid-coated nanofibrous mat as a conductive and antioxidant cell-free cardiac patch. Biofabrication 2024; 16:035004. [PMID: 38507809 DOI: 10.1088/1758-5090/ad35e9] [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: 09/30/2023] [Accepted: 03/20/2024] [Indexed: 03/22/2024]
Abstract
Polyaniline (PANI) wasin-situpolymerized on nanofibrous polycaprolactone mats as cell-free antioxidant cardiac patches (CPs), providing electrical conductivity and antioxidant properties. The fabricated CPs took advantage of intrinsic and additive antioxidant properties in the presence of PANI backbone and ascorbic acid as a biocompatible dopant of PANI. The antioxidant nature of CPs may reduce the serious repercussions of oxidative stress, produced during the ischemia-reperfusion (I/R) process following myocardial infarction. The polymerization parameters were considered as aniline (60 mM, 90 mM, and 120 mM), ascorbic acid concentrations ([aniline]:[ascorbic acid] = 3:0, 3:0.5, 3:1, 3:3), and polymerization time (1 h and 3 h). Mainly, the more aniline concentrations and polymerization time, the less sheet resistance was obtained. 1,1 diphenyl-2-picrylhydrazyl (DPPH) assay confirmed the dual antioxidant properties of prepared samples. The advantage of the employedin-situpolymerization was confirmed by the de-doping/re-doping process. Non-desirable groups were excluded based on their electrical conductivity, antioxidant properties, and biocompatibility. The remained groups protected H9c2 cells against oxidative stress and hypoxia conditions. Selected CPs reduced the intracellular reactive oxygen species content and mRNA level of caspase-3 while the Bcl-2 mRNA level was improved. Also, the selected cardiac patch could attenuate the hypertrophic impact of hydrogen peroxide on H9c2 cells. Thein vivoresults of the skin flap model confirmed the CP potency to attenuate the harmful impact of I/R.
Collapse
Affiliation(s)
- Farzad Moradikhah
- Department of Biomedical Engineering, Amirkabir University of Technology, 1591634311 Tehran, Iran
| | - Iman Shabani
- Department of Biomedical Engineering, Amirkabir University of Technology, 1591634311 Tehran, Iran
| | | |
Collapse
|
5
|
Rao K, Rochon E, Singh A, Jagannathan R, Peng Z, Mansoor H, Wang B, Moulik M, Zhang M, Saraf A, Corti P, Shiva S. Myoglobin modulates the Hippo pathway to promote cardiomyocyte differentiation. iScience 2024; 27:109146. [PMID: 38414852 PMCID: PMC10897895 DOI: 10.1016/j.isci.2024.109146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 09/30/2023] [Accepted: 02/01/2024] [Indexed: 02/29/2024] Open
Abstract
The endogenous mechanisms that propagate cardiomyocyte differentiation and prevent de-differentiation remain unclear. While the expression of the heme protein myoglobin increases by over 50% during cardiomyocyte differentiation, a role for myoglobin in regulating cardiomyocyte differentiation has not been tested. Here, we show that deletion of myoglobin in cardiomyocyte models decreases the gene expression of differentiation markers and stimulates cellular proliferation, consistent with cardiomyocyte de-differentiation. Mechanistically, the heme prosthetic group of myoglobin catalyzes the oxidation of the Hippo pathway kinase LATS1, resulting in phosphorylation and inactivation of yes-associated protein (YAP). In vivo, myoglobin-deficient zebrafish hearts show YAP dephosphorylation and accelerated cardiac regeneration after apical injury. Similarly, myoglobin knockdown in neonatal murine hearts shows increased YAP dephosphorylation and cardiomyocyte cycling. These data demonstrate a novel role for myoglobin as an endogenous driver of cardiomyocyte differentiation and highlight myoglobin as a potential target to enhance cardiac development and improve cardiac repair and regeneration.
Collapse
Affiliation(s)
- Krithika Rao
- Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Elizabeth Rochon
- Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Anuradha Singh
- Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Rajaganapathi Jagannathan
- Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Division of Cardiology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Zishan Peng
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Haris Mansoor
- Heart and Vascular Institute Division of Cardiology, Department of Medicine and Pediatrics, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Bing Wang
- Molecular Therapy Lab, Stem Cell Research Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Mousumi Moulik
- Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Division of Cardiology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Manling Zhang
- Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Division of Cardiology, Veteran Affair Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Anita Saraf
- Heart and Vascular Institute Division of Cardiology, Department of Medicine and Pediatrics, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Paola Corti
- Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Sruti Shiva
- Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
6
|
Broadwin M, Imarhia F, Oh A, Stone CR, Sellke FW, Bhowmick S, Abid MR. Exploring Electrospun Scaffold Innovations in Cardiovascular Therapy: A Review of Electrospinning in Cardiovascular Disease. Bioengineering (Basel) 2024; 11:218. [PMID: 38534492 DOI: 10.3390/bioengineering11030218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/15/2024] [Accepted: 02/22/2024] [Indexed: 03/28/2024] Open
Abstract
Cardiovascular disease (CVD) remains the leading cause of mortality worldwide. In particular, patients who suffer from ischemic heart disease (IHD) that is not amenable to surgical or percutaneous revascularization techniques have limited treatment options. Furthermore, after revascularization is successfully implemented, there are a number of pathophysiological changes to the myocardium, including but not limited to ischemia-reperfusion injury, necrosis, altered inflammation, tissue remodeling, and dyskinetic wall motion. Electrospinning, a nanofiber scaffold fabrication technique, has recently emerged as an attractive option as a potential therapeutic platform for the treatment of cardiovascular disease. Electrospun scaffolds made of biocompatible materials have the ability to mimic the native extracellular matrix and are compatible with drug delivery. These inherent properties, combined with ease of customization and a low cost of production, have made electrospun scaffolds an active area of research for the treatment of cardiovascular disease. In this review, we aim to discuss the current state of electrospinning from the fundamentals of scaffold creation to the current role of electrospun materials as both bioengineered extracellular matrices and drug delivery vehicles in the treatment of CVD, with a special emphasis on the potential clinical applications in myocardial ischemia.
Collapse
Affiliation(s)
- Mark Broadwin
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Frances Imarhia
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Amy Oh
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Christopher R Stone
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Frank W Sellke
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Sankha Bhowmick
- Department of Mechanical Engineering, University of Massachusetts Dartmouth, North Dartmouth, MA 02747, USA
| | - M Ruhul Abid
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA
| |
Collapse
|
7
|
Furuno K, Elvitigala KCML, Suzuki K, Sakai S. Local delivery of adeno-associated viral vectors with electrospun gelatin nanofiber mats. J Biomed Mater Res B Appl Biomater 2024; 112:e35345. [PMID: 37902433 DOI: 10.1002/jbm.b.35345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 08/28/2023] [Accepted: 10/14/2023] [Indexed: 10/31/2023]
Abstract
Adeno-associated viral (AAV) vectors play a significant role in gene therapy, yet the typical delivery methods, like systemic and local AAV injections, often lead to unintended off-target distribution and tissue damage due to injection. In this study, we propose a localized delivery approach for AAV vectors utilizing electrospun gelatin nanofiber mats, which are cross-linked with glutaraldehyde. The AAV vectors, which encoded a green fluorescent protein (GFP), were loaded onto the mats by immersing them in a solution containing the vectors. The amount of AAV vector loaded onto the mats increased as the vector concentration in the solution increased. The loaded AAV vector was steadily released into the cell culture medium over 3 days. The mats incubated for 3 days also showed the ability to transduce into the cells cultured on them. We evaluated the effectiveness of this delivery system by attaching the mats to mouse livers. GFP expression was visible on the surface of the liver beneath the attached mats, but not in areas in direct contact with the mats. These findings suggest that the attachment of AAV vector-loaded electrospun gelatin nanofiber mats to a target site present a promising solution for localized gene delivery while reducing off-target distribution.
Collapse
Affiliation(s)
- Kotoko Furuno
- Department of Materials Engineering Science, Graduate School of Engineering Science, Osaka University, Osaka, Japan
| | | | - Keiichiro Suzuki
- Department of Materials Engineering Science, Graduate School of Engineering Science, Osaka University, Osaka, Japan
- Institute for Advanced Co-Creation Studies, Osaka University, Osaka, Japan
- Graduate School of Frontier Bioscience, Osaka University, Osaka, Japan
| | - Shinji Sakai
- Department of Materials Engineering Science, Graduate School of Engineering Science, Osaka University, Osaka, Japan
| |
Collapse
|
8
|
Wu C, Zhang Y, Xu Y, Long L, Hu X, Zhang J, Wang Y. Injectable polyaniline nanorods/alginate hydrogel with AAV9-mediated VEGF overexpression for myocardial infarction treatment. Biomaterials 2023; 296:122088. [PMID: 36898222 DOI: 10.1016/j.biomaterials.2023.122088] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 02/28/2023] [Accepted: 03/06/2023] [Indexed: 03/08/2023]
Abstract
Intramyocardial injection of hydrogels possesses great potential in the minimally invasive treatment of myocardial infarction (MI), but the current injectable hydrogels lack conductivity, long-term angiogenesis inductive ability, and reactive oxygen species (ROS)-scavenging ability, which are essential for myocardium repair. In this study, lignosulfonate-doped polyaniline (PANI/LS) nanorods and adeno-associated virus encoding vascular endothelial growth factor (AAV9-VEGF) are incorporated in the calcium-crosslinked alginate hydrogel to develop an injectable conductive hydrogel with excellent antioxidative and angiogenic ability (Alg-P-AAV hydrogel). Due to the special nanorod morphology, a conductive network is constructed in the hydrogel with the conductivity matching the native myocardium for excitation conduction. The PANI/LS nanorod network may also have large specific surfaces and effectively scavenges ROS to protect cardiomyocytes from oxidative stress damage. AAV9-VEGF transfects the surrounding cardiomyocytes for continuously expressing VEGF, which significantly promotes the proliferation, migration and tube formation of endothelial cells. After injecting the Alg-P-AAV hydrogel around the MI area in rats, the generation of gap junctions and angiogenesis are greatly improved with reduced infarct area and recovered cardiac function. The remarkable therapeutic effect indicates the promising potential of this multi-functional hydrogel for MI treatment.
Collapse
Affiliation(s)
- Can Wu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Yuxin Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Yuanyuan Xu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Linyu Long
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Xuefeng Hu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Jieyu Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China.
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China.
| |
Collapse
|
9
|
Kong B, Liu R, Guo J, Lu L, Zhou Q, Zhao Y. Tailoring micro/nano-fibers for biomedical applications. Bioact Mater 2023; 19:328-347. [PMID: 35892003 PMCID: PMC9301605 DOI: 10.1016/j.bioactmat.2022.04.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/31/2022] [Accepted: 04/13/2022] [Indexed: 12/02/2022] Open
Abstract
Nano/micro fibers have evoked much attention of scientists and have been researched as cutting edge and hotspot in the area of fiber science in recent years due to the rapid development of various advanced manufacturing technologies, and the appearance of fascinating and special functions and properties, such as the enhanced mechanical strength, high surface area to volume ratio and special functionalities shown in the surface, triggered by the nano or micro-scale dimensions. In addition, these outstanding and special characteristics of the nano/micro fibers impart fiber-based materials with wide applications, such as environmental engineering, electronic and biomedical fields. This review mainly focuses on the recent development in the various nano/micro fibers fabrication strategies and corresponding applications in the biomedical fields, including tissue engineering scaffolds, drug delivery, wound healing, and biosensors. Moreover, the challenges for the fabrications and applications and future perspectives are presented.
Collapse
Affiliation(s)
- Bin Kong
- Department of Cardio-Thoracic Surgery, Institute of Translational Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 210008, Nanjing, China
| | - Rui Liu
- Department of Cardio-Thoracic Surgery, Institute of Translational Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 210008, Nanjing, China
| | - Jiahui Guo
- Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 210008, Nanjing, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, China
| | - Ling Lu
- Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 210008, Nanjing, China
| | - Qing Zhou
- Department of Cardio-Thoracic Surgery, Institute of Translational Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 210008, Nanjing, China
| | - Yuanjin Zhao
- Department of Cardio-Thoracic Surgery, Institute of Translational Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 210008, Nanjing, China
- Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 210008, Nanjing, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Science, 100101, Beijing, China
| |
Collapse
|
10
|
Bioabsorbable, elastomer-coated magnesium alloy coils for treating saccular cerebrovascular aneurysms. Biomaterials 2022; 290:121857. [DOI: 10.1016/j.biomaterials.2022.121857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/06/2022] [Accepted: 10/09/2022] [Indexed: 11/19/2022]
|
11
|
Xu C, Hong Y. Rational design of biodegradable thermoplastic polyurethanes for tissue repair. Bioact Mater 2022; 15:250-271. [PMID: 35386346 PMCID: PMC8940769 DOI: 10.1016/j.bioactmat.2021.11.029] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 11/09/2021] [Accepted: 11/24/2021] [Indexed: 12/25/2022] Open
Abstract
As a type of elastomeric polymers, non-degradable polyurethanes (PUs) have a long history of being used in clinics, whereas biodegradable PUs have been developed in recent decades, primarily for tissue repair and regeneration. Biodegradable thermoplastic (linear) PUs are soft and elastic polymeric biomaterials with high mechanical strength, which mimics the mechanical properties of soft and elastic tissues. Therefore, biodegradable thermoplastic polyurethanes are promising scaffolding materials for soft and elastic tissue repair and regeneration. Generally, PUs are synthesized by linking three types of changeable blocks: diisocyanates, diols, and chain extenders. Alternating the combination of these three blocks can finely tailor the physio-chemical properties and generate new functional PUs. These PUs have excellent processing flexibilities and can be fabricated into three-dimensional (3D) constructs using conventional and/or advanced technologies, which is a great advantage compared with cross-linked thermoset elastomers. Additionally, they can be combined with biomolecules to incorporate desired bioactivities to broaden their biomedical applications. In this review, we comprehensively summarized the synthesis, structures, and properties of biodegradable thermoplastic PUs, and introduced their multiple applications in tissue repair and regeneration. A whole picture of their design and applications along with discussions and perspectives of future directions would provide theoretical and technical supports to inspire new PU development and novel applications.
Collapse
Affiliation(s)
- Cancan Xu
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, 76019, USA
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, 76019, USA
| |
Collapse
|
12
|
Molecularly Imprinted Nanoparticles towards MMP9 for Controlling Cardiac ECM after Myocardial Infarction: A Predictive Experimental-Computational Chemistry Investigation. Biomedicines 2022; 10:biomedicines10092070. [PMID: 36140171 PMCID: PMC9495980 DOI: 10.3390/biomedicines10092070] [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: 06/24/2022] [Revised: 08/14/2022] [Accepted: 08/20/2022] [Indexed: 11/17/2022] Open
Abstract
The recent advances in nanotechnology are revolutionizing preventive and therapeutic approaches to treating cardiovascular diseases. Controlling the extracellular matrix metalloproteinase (MMP) activation and expression in the failing human left ventricular myocardium represents a significant therapeutic target for heart disease. In this study, we used molecularly imprinting polymers (MIPs) to restore the correct balance between MMPs and their tissue inhibitors (TIMPs), and explored the potential of this technique exhaustively through chemical synthesis, physicochemical and biological characterizations, and computational chemistry methods. By molecular dynamics simulations based on classical force fields, we simulated the early stages of the imprinting process in solution disclosing the pivotal interaction established between the monomers and the MMP9 protein template. The average interaction energies of methacrylic acid (MAA) and poly (ethylene glycol) ethyl ether methacrylate (PEG) units were in the ranges 17–22 and 30–37 kcal/mol, respectively. At low coverage, the PEG monomers seemed firmly anchored to the protein surface and were not displaced by water, while only about 20% of MAA was replaced by water. The synthesis of MIPs was successfully with a monomer conversion higher than 99% and the production of spherical particles with average diameter of 344 ± 33 nm. HPLC analysis showed a specific recognition factor of MMP9 on MIPs of about 1.3. FT-IR Chemical Imaging confirmed the mechanisms necessary to generate a “selective memory” of the MIPs towards the enzyme. HPLC results indicated that the rebound amount of both TIMP1 and MMP2 to MIPs is lower than that of the template, showing a selectivity factor of 2.1 and 2.3, respectively. Preliminary tests on the effect of MIPs on H9C2 cells revealed that this treatment has no cytotoxic effects.
Collapse
|
13
|
Nguyen-Truong M, Kim S, Doherty C, Frederes M, LeBar K, Ghosh S, Hematti P, Chinnadurai R, Wagner WR, Wang Z. Pro-angiogenic Potential of Mesenchymal Stromal Cells Regulated by Matrix Stiffness and Anisotropy Mimicking Right Ventricles. Biomacromolecules 2022; 23:2353-2361. [PMID: 35502841 DOI: 10.1021/acs.biomac.2c00132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Capillary rarefaction is a hallmark of right ventricle (RV) failure. Mesenchymal stromal cell (MSC)-based therapy offers a potential treatment due to its pro-angiogenic function. However, the impact of RV tissue mechanics on MSC behavior is unclear, especially when referring to RV end-diastolic stiffness and mechanical anisotropy. In this study, we assessed MSC behavior on electrospun scaffolds with varied stiffness (normal vs failing RV) and anisotropy (isotropic vs anisotropic). In individual MSCs, we observed the highest vascular endothelial growth factor (VEGF) production and total tube length in the failing, isotropic group (2.00 ± 0.37, 1.53 ± 0.24), which was greater than the normal, isotropic group (0.70 ± 0.15, 0.55 ± 0.07; p < 0.05). The presence of anisotropy led to trends of increased VEGF production on normal groups (0.75 ± 0.09 vs 1.20 ± 0.17), but this effect was absent on failing groups. Our findings reveal synergistic effects of RV-like stiffness and anisotropy on MSC pro-angiogenic function and may guide MSC-based therapies for heart failure.
Collapse
Affiliation(s)
- Michael Nguyen-Truong
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Seungil Kim
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States.,Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Courtney Doherty
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado 80523, United States.,Department of Mechanical Engineering, Colorado State University, Fort Collins, Colorado 80523-1376, United States
| | - Megan Frederes
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado 80523, United States.,Department of Mechanical Engineering, Colorado State University, Fort Collins, Colorado 80523-1376, United States
| | - Kristen LeBar
- Department of Mechanical Engineering, Colorado State University, Fort Collins, Colorado 80523-1376, United States
| | - Soham Ghosh
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado 80523, United States.,Department of Mechanical Engineering, Colorado State University, Fort Collins, Colorado 80523-1376, United States
| | - Peiman Hematti
- Department of Medicine, University of Wisconsin, Madison-School of Medicine and Public Health, Madison, Wisconsin 53726, United States
| | - Raghavan Chinnadurai
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia 31207, United States
| | - William R Wagner
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States.,Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Zhijie Wang
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado 80523, United States.,Department of Mechanical Engineering, Colorado State University, Fort Collins, Colorado 80523-1376, United States
| |
Collapse
|
14
|
Pedersen DD, Kim S, Wagner WR. Biodegradable polyurethane scaffolds in regenerative medicine: Clinical translation review. J Biomed Mater Res A 2022; 110:1460-1487. [PMID: 35481723 DOI: 10.1002/jbm.a.37394] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/07/2022] [Accepted: 04/09/2022] [Indexed: 12/14/2022]
Abstract
Early explorations of tissue engineering and regenerative medicine concepts commonly utilized simple polyesters such as polyglycolide, polylactide, and their copolymers as scaffolds. These biomaterials were deemed clinically acceptable, readily accessible, and provided processability and a generally known biological response. With experience and refinement of approaches, greater control of material properties and integrated bioactivity has received emphasis and a broadened palette of synthetic biomaterials has been employed. Biodegradable polyurethanes (PUs) have emerged as an attractive option for synthetic scaffolds in a variety of tissue applications because of their flexibility in molecular design and ability to fulfill mechanical property objectives, particularly in soft tissue applications. Biodegradable PUs are highly customizable based on their composition and processability to impart tailored mechanical and degradation behavior. Additionally, bioactive agents can be readily incorporated into these scaffolds to drive a desired biological response. Enthusiasm for biodegradable PU scaffolds has soared in recent years, leading to rapid growth in the literature documenting novel PU chemistries, scaffold designs, mechanical properties, and aspects of biocompatibility. Despite the enthusiasm in the field, there are still few examples of biodegradable PU scaffolds that have achieved regulatory approval and routine clinical use. However, there is a growing literature where biodegradable PU scaffolds are being specifically developed for a wide range of pathologies and where relevant pre-clinical models are being employed. The purpose of this review is first to highlight examples of clinically used biodegradable PU scaffolds, and then to summarize the growing body of reports on pre-clinical applications of biodegradable PU scaffolds.
Collapse
Affiliation(s)
- Drake D Pedersen
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Seungil Kim
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - William R Wagner
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Chemical Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
15
|
Kashiyama N, Kormos RL, Matsumura Y, D'Amore A, Miyagawa S, Sawa Y, Wagner WR. Adipose-derived stem cell sheet under an elastic patch improves cardiac function in rats after myocardial infarction. J Thorac Cardiovasc Surg 2022; 163:e261-e272. [PMID: 32636026 DOI: 10.1016/j.jtcvs.2020.04.150] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 04/12/2020] [Accepted: 04/14/2020] [Indexed: 01/28/2023]
Abstract
OBJECTIVES Although adipose-derived stem cells (ADSCs) have shown promise in cardiac regeneration, stable engraftment is still challenging. Acellular bioengineered cardiac patches have shown promise in positively altering ventricular remodeling in ischemic cardiomyopathy. We hypothesized that combining an ADSC sheet approach with a bioengineered patch would enhance ADSC engraftment and positively promote cardiac function compared with either therapy alone in a rat ischemic cardiomyopathy model. METHODS Cardiac patches were generated from poly(ester carbonate urethane) urea and porcine decellularized cardiac extracellular matrix. ADSCs constitutively expressing green fluorescent protein were established from F344 rats and transplanted as a cell sheet over the left ventricle 3 days after left anterior descending artery ligation with or without an overlying cardiac patch. Cardiac function was serially evaluated using echocardiography for 8 weeks, comparing groups with combined cells and patch (group C, n = 9), ADSCs alone (group A, n = 7), patch alone (group P, n = 6) or sham groups (n = 7). RESULTS Much greater numbers of ADSCs survived in the C versus A groups (P < .01). At 8 weeks posttransplant, the percentage fibrotic area was lower (P < .01) in groups C and P compared with the other groups and vasculature in the peri-infarct zone was greater in group C versus other groups (P < .01), and hepatocyte growth factor expression was higher in group C than in other groups (P < .05). Left ventricular ejection fraction was higher in group C versus other groups. CONCLUSIONS A biodegradable cardiac patch enhanced ADSC engraftment, which was associated with greater cardiac function and neovascularization in the peri-infarct zone following subacute myocardial infarction.
Collapse
Affiliation(s)
- Noriyuki Kashiyama
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pa; Heart and Vascular Institute, University of Pittsburgh Medical Center, Pittsburgh, Pa; Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pa; Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita-city, Osaka, Japan
| | - Robert L Kormos
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pa; Heart and Vascular Institute, University of Pittsburgh Medical Center, Pittsburgh, Pa; Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - Yasumoto Matsumura
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pa
| | - Antonio D'Amore
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pa; Fondazione RiMED, Palermo, Italy
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita-city, Osaka, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita-city, Osaka, Japan
| | - William R Wagner
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pa; Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pa; Department of Surgery, University of Pittsburgh, Pittsburgh, Pa.
| |
Collapse
|
16
|
Puhl DL, Mohanraj D, Nelson DW, Gilbert RJ. Designing electrospun fiber platforms for efficient delivery of genetic material and genome editing tools. Adv Drug Deliv Rev 2022; 183:114161. [PMID: 35183657 PMCID: PMC9724629 DOI: 10.1016/j.addr.2022.114161] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/29/2022] [Accepted: 02/11/2022] [Indexed: 02/06/2023]
Abstract
Electrospun fibers are versatile biomaterial platforms with great potential to support regeneration. Electrospun fiber characteristics such as fiber diameter, degree of alignment, rate of degradation, and surface chemistry enable the creation of unique, tunable scaffolds for various drug or gene delivery applications. The delivery of genetic material and genome editing tools via viral and non-viral vectors are approaches to control cellular protein production. However, immunogenicity, off-target effects, and low delivery efficiencies slow the progression of gene delivery strategies to clinical settings. The delivery of genetic material from electrospun fibers overcomes such limitations by allowing for localized, tunable delivery of genetic material. However, the process of electrospinning is harsh, and care must be taken to retain genetic material bioactivity. This review presents an up-to-date summary of strategies to incorporate genetic material onto or within electrospun fiber platforms to improve delivery efficiency and enhance the regenerative potential of electrospun fibers for various tissue engineering applications.
Collapse
Affiliation(s)
- Devan L Puhl
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 1623 15th Street, Troy, NY 12180, USA.
| | - Divya Mohanraj
- Department of Biological Sciences, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 1623 15th Street, Troy, NY 12180, USA.
| | - Derek W Nelson
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 1623 15th Street, Troy, NY 12180, USA.
| | - Ryan J Gilbert
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 1623 15th Street, Troy, NY 12180, USA.
| |
Collapse
|
17
|
Li M, Wu H, Yuan Y, Hu B, Gu N. Recent fabrications and applications of cardiac patch in myocardial infarction treatment. VIEW 2022. [DOI: 10.1002/viw.20200153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Mei Li
- School of Biomedical Engineering and Informatics Nanjing Medical University Nanjing China
- The Laboratory Center for Basic Medical Sciences Nanjing Medical University Nanjing China
| | - Hao Wu
- School of Biomedical Engineering and Informatics Nanjing Medical University Nanjing China
| | - Yuehui Yuan
- School of Biomedical Engineering and Informatics Nanjing Medical University Nanjing China
| | - Benhui Hu
- School of Biomedical Engineering and Informatics Nanjing Medical University Nanjing China
| | - Ning Gu
- School of Biomedical Engineering and Informatics Nanjing Medical University Nanjing China
- State Key Laboratory of Bioelectronics Jiangsu Key Laboratory for Biomaterials and Devices School of Biological Sciences and Medical Engineering Southeast University Nanjing China
| |
Collapse
|
18
|
Perveen S, Rossin D, Vitale E, Rosso R, Vanni R, Cristallini C, Rastaldo R, Giachino C. Therapeutic Acellular Scaffolds for Limiting Left Ventricular Remodelling-Current Status and Future Directions. Int J Mol Sci 2021; 22:ijms222313054. [PMID: 34884856 PMCID: PMC8658014 DOI: 10.3390/ijms222313054] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 12/14/2022] Open
Abstract
Myocardial infarction (MI) is one of the leading causes of heart-related deaths worldwide. Following MI, the hypoxic microenvironment triggers apoptosis, disrupts the extracellular matrix and forms a non-functional scar that leads towards adverse left ventricular (LV) remodelling. If left untreated this eventually leads to heart failure. Besides extensive advancement in medical therapy, complete functional recovery is never accomplished, as the heart possesses limited regenerative ability. In recent decades, the focus has shifted towards tissue engineering and regenerative strategies that provide an attractive option to improve cardiac regeneration, limit adverse LV remodelling and restore function in an infarcted heart. Acellular scaffolds possess attractive features that have made them a promising therapeutic candidate. Their application in infarcted areas has been shown to improve LV remodelling and enhance functional recovery in post-MI hearts. This review will summarise the updates on acellular scaffolds developed and tested in pre-clinical and clinical scenarios in the past five years with a focus on their ability to overcome damage caused by MI. It will also describe how acellular scaffolds alone or in combination with biomolecules have been employed for MI treatment. A better understanding of acellular scaffolds potentialities may guide the development of customised and optimised therapeutic strategies for MI treatment.
Collapse
Affiliation(s)
- Sadia Perveen
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (S.P.); (D.R.); (E.V.); (R.R.); (R.V.); (C.G.)
| | - Daniela Rossin
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (S.P.); (D.R.); (E.V.); (R.R.); (R.V.); (C.G.)
| | - Emanuela Vitale
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (S.P.); (D.R.); (E.V.); (R.R.); (R.V.); (C.G.)
| | - Rachele Rosso
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (S.P.); (D.R.); (E.V.); (R.R.); (R.V.); (C.G.)
| | - Roberto Vanni
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (S.P.); (D.R.); (E.V.); (R.R.); (R.V.); (C.G.)
| | | | - Raffaella Rastaldo
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (S.P.); (D.R.); (E.V.); (R.R.); (R.V.); (C.G.)
- Correspondence:
| | - Claudia Giachino
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (S.P.); (D.R.); (E.V.); (R.R.); (R.V.); (C.G.)
| |
Collapse
|
19
|
Recent Advances in Cardiac Tissue Engineering for the Management of Myocardium Infarction. Cells 2021; 10:cells10102538. [PMID: 34685518 PMCID: PMC8533887 DOI: 10.3390/cells10102538] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/16/2021] [Accepted: 09/21/2021] [Indexed: 12/26/2022] Open
Abstract
Myocardium Infarction (MI) is one of the foremost cardiovascular diseases (CVDs) causing death worldwide, and its case numbers are expected to continuously increase in the coming years. Pharmacological interventions have not been at the forefront in ameliorating MI-related morbidity and mortality. Stem cell-based tissue engineering approaches have been extensively explored for their regenerative potential in the infarcted myocardium. Recent studies on microfluidic devices employing stem cells under laboratory set-up have revealed meticulous events pertaining to the pathophysiology of MI occurring at the infarcted site. This discovery also underpins the appropriate conditions in the niche for differentiating stem cells into mature cardiomyocyte-like cells and leads to engineering of the scaffold via mimicking of native cardiac physiological conditions. However, the mode of stem cell-loaded engineered scaffolds delivered to the site of infarction is still a challenging mission, and yet to be translated to the clinical setting. In this review, we have elucidated the various strategies developed using a hydrogel-based system both as encapsulated stem cells and as biocompatible patches loaded with cells and applied at the site of infarction.
Collapse
|
20
|
Gupta N, Kamath S M, Rao SK, D J, Patil S, Gupta N, Arunachalam KD. Kaempferol loaded albumin nanoparticles and dexamethasone encapsulation into electrospun polycaprolactone fibrous mat – Concurrent release for cartilage regeneration. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102666] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
21
|
Tavakol DN, Fleischer S, Vunjak-Novakovic G. Harnessing organs-on-a-chip to model tissue regeneration. Cell Stem Cell 2021; 28:993-1015. [PMID: 34087161 DOI: 10.1016/j.stem.2021.05.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tissue engineering has markedly matured since its early beginnings in the 1980s. In addition to the original goal to regenerate damaged organs, the field has started to explore modeling of human physiology "in a dish." Induced pluripotent stem cell (iPSC) technologies now enable studies of organ regeneration and disease modeling in a patient-specific context. We discuss the potential of "organ-on-a-chip" systems to study regenerative therapies with focus on three distinct organ systems: cardiac, respiratory, and hematopoietic. We propose that the combinatorial studies of human tissues at these two scales would help realize the translational potential of tissue engineering.
Collapse
Affiliation(s)
| | - Sharon Fleischer
- Department of Biomedical Engineering, Columbia University, New York, NY
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY; Department of Medicine, Columbia University, New York, NY.
| |
Collapse
|
22
|
Borrelli MA, Turnquist HR, Little SR. Biologics and their delivery systems: Trends in myocardial infarction. Adv Drug Deliv Rev 2021; 173:181-215. [PMID: 33775706 PMCID: PMC8178247 DOI: 10.1016/j.addr.2021.03.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/14/2021] [Accepted: 03/20/2021] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease is the leading cause of death around the world, in which myocardial infarction (MI) is a precipitating event. However, current therapies do not adequately address the multiple dysregulated systems following MI. Consequently, recent studies have developed novel biologic delivery systems to more effectively address these maladies. This review utilizes a scientometric summary of the recent literature to identify trends among biologic delivery systems designed to treat MI. Emphasis is placed on sustained or targeted release of biologics (e.g. growth factors, nucleic acids, stem cells, chemokines) from common delivery systems (e.g. microparticles, nanocarriers, injectable hydrogels, implantable patches). We also evaluate biologic delivery system trends in the entire regenerative medicine field to identify emerging approaches that may translate to the treatment of MI. Future developments include immune system targeting through soluble factor or chemokine delivery, and the development of advanced delivery systems that facilitate the synergistic delivery of biologics.
Collapse
Affiliation(s)
- Matthew A Borrelli
- Department of Chemical Engineering, University of Pittsburgh, 940 Benedum Hall, 3700 O'Hara Street, Pittsburgh, PA 15213, USA.
| | - Heth R Turnquist
- Starzl Transplantation Institute, 200 Darragh St, Pittsburgh, PA 15213, USA; Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Immunology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA.
| | - Steven R Little
- Department of Chemical Engineering, University of Pittsburgh, 940 Benedum Hall, 3700 O'Hara Street, Pittsburgh, PA 15213, USA; Department of Bioengineering, University of Pittsburgh, 302 Benedum Hall, 3700 O'Hara Street, Pittsburgh, PA 15213, USA; Department of Clinical and Translational Science, University of Pittsburgh, Forbes Tower, Suite 7057, Pittsburgh, PA 15213, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA 15219, USA; Department of Immunology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA; Department of Pharmaceutical Science, University of Pittsburgh, 3501 Terrace Street, Pittsburgh, PA 15213, USA; Department of Ophthalmology, University of Pittsburgh, 203 Lothrop Street, Pittsburgh, PA 15213, USA.
| |
Collapse
|
23
|
Feng J, Shi H, Yang X, Xiao S. Self-Adhesion Conductive Sub-micron Fiber Cardiac Patch from Shape Memory Polymers to Promote Electrical Signal Transduction Function. ACS APPLIED MATERIALS & INTERFACES 2021; 13:19593-19602. [PMID: 33900060 DOI: 10.1021/acsami.0c22844] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Myocardial infarction (MI) constitutes the first cause of morbidity and mortality in our life, so using highly conductive and elastic materials to produce an engineered cardiac patch is an effective way to improve the myocardium infarction area function. Here, shape memory polymers of the polyurethane/polyaniline/silicon oxide (PU/PANI/SiO2) electrospinning sub-micron fiber patch were precisely produced in the case of the hydrogen bonding effect and interaction between the carboxyl groups to provide compatibility, phase mixing/miscibility, and stability. The sub-micron fiber patch prepared by our group has some remarkable characteristics, such as sub-micron fibers, 3D porous structure, special thickness to simulate the extracellular matrix (ECM), elastic deformation, good properties in conducting weak electrical signals, stability to maintain the whole structure, and self-adhesion. This sub-micron fiber material has been proven to be effective, easy, and reliable. Through precise design of the material system, structure regulation, and performance optimization, the aim is to produce a sub-micron fiber cardiac patch to simulate the myocardium ECM and improve conductive signal transduction for potential MI therapy.
Collapse
Affiliation(s)
- Jianyong Feng
- College of Textile Science and Engineering, Zhejiang Sci-Tech University, No. 928, 2nd Street, Xiasha Higher Education Zone, Hangzhou 310018, China
| | - Hui Shi
- College of Media Engineering, Communication University of Zhejiang, 998 Xue Yuan Street, Higher Education Zone, Hangzhou 310018, China
| | - Xiaoyuan Yang
- College of Textile Science and Engineering, Zhejiang Sci-Tech University, No. 928, 2nd Street, Xiasha Higher Education Zone, Hangzhou 310018, China
| | - Shuang Xiao
- College of Textile Science and Engineering, Zhejiang Sci-Tech University, No. 928, 2nd Street, Xiasha Higher Education Zone, Hangzhou 310018, China
| |
Collapse
|
24
|
A perfusable, multifunctional epicardial device improves cardiac function and tissue repair. Nat Med 2021; 27:480-490. [PMID: 33723455 DOI: 10.1038/s41591-021-01279-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/04/2021] [Indexed: 02/07/2023]
Abstract
Despite advances in technologies for cardiac repair after myocardial infarction (MI), new integrated therapeutic approaches still need to be developed. In this study, we designed a perfusable, multifunctional epicardial device (PerMed) consisting of a biodegradable elastic patch (BEP), permeable hierarchical microchannel networks (PHMs) and a system to enable delivery of therapeutic agents from a subcutaneously implanted pump. After its implantation into the epicardium, the BEP is designed to provide mechanical cues for ventricular remodeling, and the PHMs are designed to facilitate angiogenesis and allow for infiltration of reparative cells. In a rat model of MI, implantation of the PerMed improved ventricular function. When connected to a pump, the PerMed enabled targeted, sustained and stable release of platelet-derived growth factor-BB, amplifying the efficacy of cardiac repair as compared to the device without a pump. We also demonstrated the feasibility of minimally invasive surgical PerMed implantation in pigs, demonstrating its promise for clinical translation to treat heart disease.
Collapse
|
25
|
Valera IC, Wacker AL, Hwang HS, Holmes C, Laitano O, Landstrom AP, Parvatiyar MS. Essential roles of the dystrophin-glycoprotein complex in different cardiac pathologies. Adv Med Sci 2021; 66:52-71. [PMID: 33387942 DOI: 10.1016/j.advms.2020.12.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 12/12/2020] [Accepted: 12/17/2020] [Indexed: 12/20/2022]
Abstract
The dystrophin-glycoprotein complex (DGC), situated at the sarcolemma dynamically remodels during cardiac disease. This review examines DGC remodeling as a common denominator in diseases affecting heart function and health. Dystrophin and the DGC serve as broad cytoskeletal integrators that are critical for maintaining stability of muscle membranes. The presence of pathogenic variants in genes encoding proteins of the DGC can cause absence of the protein and/or alterations in other complex members leading to muscular dystrophies. Targeted studies have allowed the individual functions of affected proteins to be defined. The DGC has demonstrated its dynamic function, remodeling under a number of conditions that stress the heart. Beyond genetic causes, pathogenic processes also impinge on the DGC, causing alterations in the abundance of dystrophin and associated proteins during cardiac insult such as ischemia-reperfusion injury, mechanical unloading, and myocarditis. When considering new therapeutic strategies, it is important to assess DGC remodeling as a common factor in various heart diseases. The DGC connects the internal F-actin-based cytoskeleton to laminin-211 of the extracellular space, playing an important role in the transmission of mechanical force to the extracellular matrix. The essential functions of dystrophin and the DGC have been long recognized. DGC based therapeutic approaches have been primarily focused on muscular dystrophies, however it may be a beneficial target in a number of disorders that affect the heart. This review provides an account of what we now know, and discusses how this knowledge can benefit persistent health conditions in the clinic.
Collapse
Affiliation(s)
- Isela C Valera
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA
| | - Amanda L Wacker
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA
| | - Hyun Seok Hwang
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA
| | - Christina Holmes
- Department of Chemical and Biomedical Engineering, Florida A&M University-Florida State University College of Engineering, Tallahassee, FL, USA
| | - Orlando Laitano
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA
| | - Andrew P Landstrom
- Department of Pediatrics, Division of Cardiology, Duke University School of Medicine, Durham, NC, USA; Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| | - Michelle S Parvatiyar
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA.
| |
Collapse
|
26
|
Wang Y, Bruggeman KF, Franks S, Gautam V, Hodgetts SI, Harvey AR, Williams RJ, Nisbet DR. Is Viral Vector Gene Delivery More Effective Using Biomaterials? Adv Healthc Mater 2021; 10:e2001238. [PMID: 33191667 DOI: 10.1002/adhm.202001238] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/03/2020] [Indexed: 12/16/2022]
Abstract
Gene delivery has been extensively investigated for introducing foreign genetic material into cells to promote expression of therapeutic proteins or to silence relevant genes. This approach can regulate genetic or epigenetic disorders, offering an attractive alternative to pharmacological therapy or invasive protein delivery options. However, the exciting potential of viral gene therapy has yet to be fully realized, with a number of clinical trials failing to deliver optimal therapeutic outcomes. Reasons for this include difficulty in achieving localized delivery, and subsequently lower efficacy at the target site, as well as poor or inconsistent transduction efficiency. Thus, ongoing efforts are focused on improving local viral delivery and enhancing its efficiency. Recently, biomaterials have been exploited as an option for more controlled, targeted and programmable gene delivery. There is a growing body of literature demonstrating the efficacy of biomaterials and their potential advantages over other delivery strategies. This review explores current limitations of gene delivery and the progress of biomaterial-mediated gene delivery. The combination of biomaterials and gene vectors holds the potential to surmount major challenges, including the uncontrolled release of viral vectors with random delivery duration, poorly localized viral delivery with associated off-target effects, limited viral tropism, and immune safety concerns.
Collapse
Affiliation(s)
- Yi Wang
- Laboratory of Advanced Biomaterials Research School of Engineering The Australian National University Canberra ACT 2601 Australia
| | - Kiara F. Bruggeman
- Laboratory of Advanced Biomaterials Research School of Engineering The Australian National University Canberra ACT 2601 Australia
| | - Stephanie Franks
- Laboratory of Advanced Biomaterials Research School of Engineering The Australian National University Canberra ACT 2601 Australia
| | - Vini Gautam
- Department of Biomedical Engineering The University of Melbourne Melbourne Victoria 3010 Australia
| | - Stuart I. Hodgetts
- School of Human Sciences The University of Western Australia Perth WA 6009 Australia
- Perron Institute for Neurological and Translational Science Perth WA 6009 Australia
| | - Alan R. Harvey
- School of Human Sciences The University of Western Australia Perth WA 6009 Australia
- Perron Institute for Neurological and Translational Science Perth WA 6009 Australia
| | - Richard J. Williams
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT) School of Medicine Deakin University Waurn Ponds VIC 3216 Australia
- Biofab3D St. Vincent's Hospital Fitzroy 3065 Australia
| | - David R. Nisbet
- Laboratory of Advanced Biomaterials Research School of Engineering The Australian National University Canberra ACT 2601 Australia
- Biofab3D St. Vincent's Hospital Fitzroy 3065 Australia
| |
Collapse
|
27
|
Lugin ML, Lee RT, Kwon YJ. Synthetically Engineered Adeno-Associated Virus for Efficient, Safe, and Versatile Gene Therapy Applications. ACS NANO 2020; 14:14262-14283. [PMID: 33073995 DOI: 10.1021/acsnano.0c03850] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Gene therapy directly targets mutations causing disease, allowing for a specific treatment at a molecular level. Adeno-associated virus (AAV) has been of increasing interest as a gene delivery vehicle, as AAV vectors are safe, effective, and capable of eliciting a relatively contained immune response. With the recent FDA approval of two AAV drugs for treating rare genetic diseases, AAV vectors are now on the market and are being further explored for other therapies. While showing promise in immune privileged tissue, the use of AAV for systemic delivery is still limited due to the high prevalence of neutralizing antibodies (nAbs). To avoid nAb-mediated inactivation, engineered AAV vectors with modified protein capsids, materials tethered to the capsid surface, or fully encapsulated in a second, larger carrier have been explored. Many of these engineered AAVs have added benefits, including avoided immune response, overcoming the genome size limit, targeted and stimuli-responsive delivery, and multimodal therapy of two or more therapeutic modalities in one platform. Native and engineered AAV vectors have been tested to treat a broad range of diseases, including spinal muscular atrophy, retinal diseases, cancers, and tissue damage. This review will cover the benefits of AAV as a promising gene vector by itself, the progress and advantages of engineered AAV vectors, particularly synthetically engineered ones, and the current state of their clinical translation in therapy.
Collapse
|
28
|
Abstract
Therapeutic viral gene delivery is an emerging technology which aims to correct genetic mutations by introducing new genetic information to cells either to correct a faulty gene or to initiate cell death in oncolytic treatments. In recent years, significant scientific progress has led to several clinical trials resulting in the approval of gene therapies for human treatment. However, successful therapies remain limited due to a number of challenges such as inefficient cell uptake, low transduction efficiency (TE), limited tropism, liver toxicity and immune response. To adress these issues and increase the number of available therapies, additives from a broad range of materials like polymers, peptides, lipids, nanoparticles, and small molecules have been applied so far. The scope of this review is to highlight these selected delivery systems from a materials perspective.
Collapse
Affiliation(s)
- Kübra Kaygisiz
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | | |
Collapse
|
29
|
Nguyen-Truong M, Li YV, Wang Z. Mechanical Considerations of Electrospun Scaffolds for Myocardial Tissue and Regenerative Engineering. Bioengineering (Basel) 2020; 7:E122. [PMID: 33022929 PMCID: PMC7711753 DOI: 10.3390/bioengineering7040122] [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: 09/01/2020] [Revised: 09/25/2020] [Accepted: 10/01/2020] [Indexed: 12/13/2022] Open
Abstract
Biomaterials to facilitate the restoration of cardiac tissue is of emerging importance. While there are many aspects to consider in the design of biomaterials, mechanical properties can be of particular importance in this dynamically remodeling tissue. This review focuses on one specific processing method, electrospinning, that is employed to generate materials with a fibrous microstructure that can be combined with material properties to achieve the desired mechanical behavior. Current methods used to fabricate mechanically relevant micro-/nanofibrous scaffolds, in vivo studies using these scaffolds as therapeutics, and common techniques to characterize the mechanical properties of the scaffolds are covered. We also discuss the discrepancies in the reported elastic modulus for physiological and pathological myocardium in the literature, as well as the emerging area of in vitro mechanobiology studies to investigate the mechanical regulation in cardiac tissue engineering. Lastly, future perspectives and recommendations are offered in order to enhance the understanding of cardiac mechanobiology and foster therapeutic development in myocardial regenerative medicine.
Collapse
Affiliation(s)
- Michael Nguyen-Truong
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA; (M.N.-T.); (Y.V.L.)
| | - Yan Vivian Li
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA; (M.N.-T.); (Y.V.L.)
- Department of Design and Merchandising, Colorado State University, Fort Collins, CO 80523, USA
- School of Advanced Materials Discovery, Colorado State University, Fort Collins, CO 80523, USA
| | - Zhijie Wang
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA; (M.N.-T.); (Y.V.L.)
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
30
|
Ippel B, Van Haaften EE, Bouten CVC, Dankers PYW. Impact of Additives on Mechanical Properties of Supramolecular Electrospun Scaffolds. ACS APPLIED POLYMER MATERIALS 2020; 2:3742-3748. [PMID: 32954355 PMCID: PMC7497720 DOI: 10.1021/acsapm.0c00658] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/18/2020] [Indexed: 06/11/2023]
Abstract
The mechanical properties of scaffolds used for mechanically challenging applications such as cardiovascular implants are unequivocally important. Here, the effect of supramolecular additive functionalization on mechanical behavior of electrospun scaffolds was investigated for one bisurea-based model additive and two previously developed antifouling additives. The model additive has no effect on the mechanical properties of the bulk material, whereas the stiffness of electrospun scaffolds was slightly decreased compared to pristine PCL-BU following the addition of the three different additives. These results show the robustness of supramolecular additives used in biomedical applications, in which mechanical properties are important, such as vascular grafts and heart valve constructs.
Collapse
Affiliation(s)
- Bastiaan
D. Ippel
- Institute
for Complex Molecular Systems, Eindhoven
University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
- Laboratory
for Cell and Tissue Engineering, Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
| | - Eline E. Van Haaften
- Institute
for Complex Molecular Systems, Eindhoven
University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
- Laboratory
for Cell and Tissue Engineering, Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
| | - Carlijn V. C. Bouten
- Institute
for Complex Molecular Systems, Eindhoven
University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
- Laboratory
for Cell and Tissue Engineering, Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
| | - Patricia Y. W. Dankers
- Institute
for Complex Molecular Systems, Eindhoven
University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
- Laboratory
for Cell and Tissue Engineering, Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
- Laboratory
of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|
31
|
Wang Z, Cui W. Two Sides of Electrospun Fiber in Promoting and Inhibiting Biomedical Processes. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000096] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Zhen Wang
- Shanghai Institute of Traumatology and Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases Ruijin Hospital Shanghai Jiao Tong University School of Medicine 197 Ruijin 2nd Road Shanghai 200025 P. R. China
| | - Wenguo Cui
- Shanghai Institute of Traumatology and Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases Ruijin Hospital Shanghai Jiao Tong University School of Medicine 197 Ruijin 2nd Road Shanghai 200025 P. R. China
| |
Collapse
|
32
|
Jain R, Shetty S, Yadav KS. Unfolding the electrospinning potential of biopolymers for preparation of nanofibers. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101604] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
33
|
Shi H, Xue T, Yang Y, Jiang C, Huang S, Yang Q, Lei D, You Z, Jin T, Wu F, Zhao Q, Ye X. Microneedle-mediated gene delivery for the treatment of ischemic myocardial disease. SCIENCE ADVANCES 2020; 6:eaaz3621. [PMID: 32596444 PMCID: PMC7299628 DOI: 10.1126/sciadv.aaz3621] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 05/04/2020] [Indexed: 06/01/2023]
Abstract
Cardiovascular disorders are still the primary cause of mortality worldwide. Although intramyocardial injection can effectively deliver agents to the myocardium, this approach is limited because of its restriction to needle-mediated injection and the minor retention of agents in the myocardium. Here, we engineered phase-transition microneedles (MNs) coated with adeno-associated virus (AAV) and achieved homogeneous distribution of AAV delivery. Bioluminescence imaging revealed the successful delivery and transfection of AAV-luciferase. AAV-green fluorescent protein-transfected cardiomyocytes were homogeneously distributed on postoperative day 28. AAV-vascular endothelial growth factor (VEGF)-loaded MNs improved heart function by enhancing VEGF expression, promoting functional angiogenesis, and activating the Akt signaling pathway. The results indicated the superiority of MNs over direct muscle injection. Consequently, MNs might emerge as a promising tool with great versatility for delivering various agents to treat ischemic myocardial disease.
Collapse
Affiliation(s)
- Hongpeng Shi
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Tong Xue
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Yang Yang
- Department of Cardiothoracic Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P. R. China
| | - Chenyu Jiang
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Shixing Huang
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Qi Yang
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Dong Lei
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, International Joint Laboratory for Advanced Fiber and Low-Dimension Materials, College of Materials Science and Engineering Donghua University, Shanghai 201620, P. R. China
| | - Zhengwei You
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, International Joint Laboratory for Advanced Fiber and Low-Dimension Materials, College of Materials Science and Engineering Donghua University, Shanghai 201620, P. R. China
| | - Tuo Jin
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Fei Wu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Qiang Zhao
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Xiaofeng Ye
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| |
Collapse
|
34
|
McMahan S, Taylor A, Copeland KM, Pan Z, Liao J, Hong Y. Current advances in biodegradable synthetic polymer based cardiac patches. J Biomed Mater Res A 2020; 108:972-983. [PMID: 31895482 DOI: 10.1002/jbm.a.36874] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/19/2019] [Accepted: 12/26/2019] [Indexed: 12/21/2022]
Abstract
The number of people affected by heart disease such as coronary artery disease and myocardial infarction increases at an alarming rate each year. Currently, the methods to treat these diseases are restricted to lifestyle change, pharmaceuticals, and eventually heart transplant if the condition is severe enough. While these treatment options are the standard for caring for patients who suffer from heart disease, limited regenerative ability of the heart restricts the effectiveness of treatment and may lead to other heart-related health problems in the future. Because of the increasing need for more effective therapeutic technologies for treating diseased heart tissue, cardiac patches are now a large focus for researchers. The cardiac patches are designed to be integrated into the patients' natural tissue to introduce mechanical support and healing to the damaged areas. As a promising alternative, synthetic biodegradable polymer based biomaterials can be easily manipulated to customize material properties, as well as possess certain desired characteristics for cardiac patch use. This comprehensive review summarizes recent works on synthetic biodegradable cardiac patches implanted into infarcted animal models. In addition, this review describes the basic requirements that should be met for cardiac patch development, and discusses the inspirations to designing new biomaterials and technologies for cardiac patches.
Collapse
Affiliation(s)
- Sara McMahan
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas
| | - Alan Taylor
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas
| | - Katherine M Copeland
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas
| | - Zui Pan
- College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, Texas
| | - Jun Liao
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas
| |
Collapse
|
35
|
Yao Y, Ding J, Wang Z, Zhang H, Xie J, Wang Y, Hong L, Mao Z, Gao J, Gao C. ROS-responsive polyurethane fibrous patches loaded with methylprednisolone (MP) for restoring structures and functions of infarcted myocardium in vivo. Biomaterials 2019; 232:119726. [PMID: 31901502 DOI: 10.1016/j.biomaterials.2019.119726] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 12/03/2019] [Accepted: 12/22/2019] [Indexed: 12/13/2022]
Abstract
Reactive oxygen species (ROS) play an important role in the pathogenesis of numerous diseases including atherosclerosis, diabetes, inflammation and myocardial infarction (MI). In this study, a ROS-responsive biodegradable elastomeric polyurethane containing thioketal (PUTK) linkages was synthesized from polycaprolactone diol (PCL-diol ), 1,6-hexamethylene diisocyanate (HDI), and ROS-cleavable chain extender. The PUTK was electrospun into fibrous patches with the option to load glucocorticoid methylprednisolone (MP), which were then used to treat MI of rats in vivo. The fibrous patches exhibited suitable mechanical properties and high elasticity. The molecular weight of PUTK was decreased significantly after incubation in 1 mM H2O2 solution for 2 weeks due to the degradation of thioketal bonds on the polymer backbone. Both the PUTK and PUTK/MP fibrous patches showed good antioxidant property in an oxidative environment in vitro. Implantation of the ROS-responsive polyurethane patches in MI of rats in vivo could better protect cardiomyocytes from death in the earlier stage (24 h) than the non ROS-responsive ones. Implantation of the PUTK/MP fibrous patches for 28 days could effectively improve the reconstruction of cardiac functions including increased ejection fraction, decreased infarction size, and enhanced revascularization of the infarct myocardium.
Collapse
Affiliation(s)
- Yuejun Yao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jie Ding
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Zhaoyi Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Haolan Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jieqi Xie
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yingchao Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Liangjie Hong
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China.
| | - Jianqing Gao
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
36
|
Ippel BD, Arts B, Keizer HM, Dankers PYW. Combinatorial functionalization with bisurea-peptides and antifouling bisurea additives of a supramolecular elastomeric biomaterial. JOURNAL OF POLYMER SCIENCE. PART B, POLYMER PHYSICS 2019; 57:1725-1735. [PMID: 32025088 PMCID: PMC6988465 DOI: 10.1002/polb.24907] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/08/2019] [Accepted: 11/08/2019] [Indexed: 12/13/2022]
Abstract
The bioactive additive toolbox to functionalize supramolecular elastomeric materials expands rapidly. Here we have set an explorative step toward screening of complex combinatorial functionalization with antifouling and three peptide-containing additives in a bisurea-based supramolecular system. Thorough investigation of surface properties of thin films with contact angle measurements, X-ray photoelectron spectroscopy and atomic force microscopy, was correlated to cell-adhesion of endothelial and smooth muscle cells to apprehend their respective predictive values for functional biomaterial development. Peptides were presented at the surface alone, and in combinatorial functionalization with the oligo(ethylene glycol)-based non-cell adhesive additive. The bisurea-RGD additive was cell-adhesive in all conditions, whereas the endothelial cell-specific bisurea-REDV showed limited bioactive properties in all chemical nano-environments. Also, aspecific functionality was observed for a bisurea-SDF1α peptide. These results emphasize that special care should be taken in changing the chemical nano-environment with peptide functionalization. © 2019 The Authors. Journal of Polymer Science Part B: Polymer Physics published by Wiley Periodicals, Inc. J. Polym. Sci., Part B: Polym. Phys. 2019, 57, 1725-1735.
Collapse
Affiliation(s)
- Bastiaan D. Ippel
- Institute for Complex Molecular SystemsEindhoven University of TechnologyPO Box 513 5600EindhovenManitobaThe Netherlands
- Department of Biomedical Engineering, Laboratory for Cell and Tissue EngineeringEindhoven University of TechnologyPO Box 513 5600EindhovenManitobaThe Netherlands
| | - Boris Arts
- Institute for Complex Molecular SystemsEindhoven University of TechnologyPO Box 513 5600EindhovenManitobaThe Netherlands
- Department of Biomedical Engineering, Laboratory of Chemical BiologyEindhoven University of TechnologyPO Box 513, 5600EindhovenManitobaThe Netherlands
| | - Henk M. Keizer
- SyMO‐Chem B.VDen Dolech 2, 5612EindhovenArizonaThe Netherlands
| | - Patricia Y. W. Dankers
- Institute for Complex Molecular SystemsEindhoven University of TechnologyPO Box 513 5600EindhovenManitobaThe Netherlands
- Department of Biomedical Engineering, Laboratory for Cell and Tissue EngineeringEindhoven University of TechnologyPO Box 513 5600EindhovenManitobaThe Netherlands
- Department of Biomedical Engineering, Laboratory of Chemical BiologyEindhoven University of TechnologyPO Box 513, 5600EindhovenManitobaThe Netherlands
| |
Collapse
|
37
|
A viscoelastic adhesive epicardial patch for treating myocardial infarction. Nat Biomed Eng 2019; 3:632-643. [PMID: 30988471 DOI: 10.1038/s41551-019-0380-9] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 03/05/2019] [Indexed: 01/10/2023]
Abstract
Acellular epicardial patches that treat myocardial infarction by increasing the mechanical integrity of damaged left ventricular tissues exhibit widely scattered therapeutic efficacy. Here, we introduce a viscoelastic adhesive patch, made of an ionically crosslinked transparent hydrogel, that accommodates the cyclic deformation of the myocardium and outperforms most existing acellular epicardial patches in reversing left ventricular remodelling and restoring heart function after both acute and subacute myocardial infarction in rats. The superior performance of the patch results from its relatively low dynamic modulus, designed at the so-called 'gel point' via finite-element simulations of left ventricular remodelling so as to balance the fluid and solid properties of the material.
Collapse
|
38
|
Abstract
Polymeric matrices inherently protect viral vectors from pre-existing immune conditions, limit dissemination to off-target sites, and can sustain vector release. Advancing methodologies in development of particulate based vehicles have led to improved encapsulation of viral vectors. Polymeric delivery systems have contributed to increasing cellular transduction, responsive release mechanisms, cellular infiltration, and cellular signaling. Synthetic polymers are easily customizable, and are capable of balancing matrix retention with cellular infiltration. Natural polymers contain inherent biorecognizable motifs adding therapeutic efficacy to the incorporated viral vector. Recombinant polymers use highly conserved motifs to carefully engineer matrices, allowing for precise design including elements of vector retention and responsive release mechanisms. Composite polymer systems provide opportunities to create matrices with unique properties. Carefully designed matrices can control spatiotemporal release patterns that synergize with approaches in regenerative medicine and antitumor therapies.
Collapse
Affiliation(s)
- Douglas Steinhauff
- Utah Center for Nanomedicine , Nano Institute of Utah , 36 South Wasatch Drive , Salt Lake City , Utah 84112 , United States
| | - Hamidreza Ghandehari
- Utah Center for Nanomedicine , Nano Institute of Utah , 36 South Wasatch Drive , Salt Lake City , Utah 84112 , United States
| |
Collapse
|
39
|
Liang S, Zhang Y, Wang H, Xu Z, Chen J, Bao R, Tan B, Cui Y, Fan G, Wang W, Wang W, Liu W. Paintable and Rapidly Bondable Conductive Hydrogels as Therapeutic Cardiac Patches. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1704235. [PMID: 29687502 DOI: 10.1002/adma.201704235] [Citation(s) in RCA: 240] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 02/02/2018] [Indexed: 06/08/2023]
Abstract
In recent years, cardiac patches have been developed for the treatment of myocardial infarction. However, the fixation approaches onto the tissue through suture or phototriggered reaction inevitably cause new tissue damage. Herein, a paintable hydrogel is constructed based on Fe3+ -triggered simultaneous polymerization of covalently linked pyrrole and dopamine in the hyperbranched chains where the in situ formed conductive polypyrrole also uniquely serves to crosslink network. This conductive and adhesive hydrogel can be conveniently painted as a patch onto the heart surface without adverse liquid leakage. The functional patch whose conductivity is equivalent to that of normal myocardium is strongly bonded to the beating heart within 4 weeks, accordingly efficiently boosting the transmission of electrophysiological signals. Eventually, the reconstruction of cardiac function and revascularization of the infarct myocardium are remarkably improved. The translatable suture-free strategy reported in this work is promising to address the human clinical challenges in cardiac tissue engineering.
Collapse
Affiliation(s)
- Shuang Liang
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300072, China
| | - Yinyu Zhang
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300072, China
| | - Hongbo Wang
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300072, China
| | - Ziyang Xu
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300072, China
| | - Jingrui Chen
- Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, P. R. China
| | - Rui Bao
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300072, China
| | - Baoyu Tan
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300072, China
| | - Yuanlu Cui
- Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, P. R. China
| | - Guanwei Fan
- Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, P. R. China
| | - Wenxin Wang
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300072, China
| | - Wei Wang
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300072, China
| | - Wenguang Liu
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300072, China
| |
Collapse
|
40
|
Yoo SY, Jeong SN, Kang JI, Lee SW. Chimeric Adeno-Associated Virus-Mediated Cardiovascular Reprogramming for Ischemic Heart Disease. ACS OMEGA 2018; 3:5918-5925. [PMID: 30023931 PMCID: PMC6044635 DOI: 10.1021/acsomega.8b00904] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 05/22/2018] [Indexed: 05/28/2023]
Abstract
Here, we demonstrated chimeric adeno-associated virus (chimeric AAV), AAV-DJ-mediated cardiovascular reprogramming strategy to generate new cardiomyocytes and limit collagen deposition in cardiac fibroblasts by inducing synergism of chimeric AAV-expressing Gata4, Mef2c, Tbx5 (AAV-GMT)-mediated heart reprogramming and chimeric AAV-expressing thymosin β4 (AAV-Tβ4)-mediated heart regeneration. AAV-GMT promoted a gradual increase in expression of cardiac-specific genes, including Actc1, Gja1, Myh6, Ryr2, and cTnT, with a gradual decrease in expression of a fibrosis-specific gene, procollagen type I and here AAV-Tβ4 help to induce GMT expression, providing a chimeric AAV-mediated therapeutic cell reprogramming strategy for ischemic heart diseases.
Collapse
Affiliation(s)
- So Young Yoo
- BIO-IT
Foundry Technology Institute, Pusan National
University, Busan 46241, Republic of Korea
- Research
Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| | - Su-Nam Jeong
- BIO-IT
Foundry Technology Institute, Pusan National
University, Busan 46241, Republic of Korea
| | - Jeong-In Kang
- Research
Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
- Control
and Instrumentation Engineering, Korea Maritime
and Ocean University, Busan 49112, Republic of Korea
| | - Seung-Wuk Lee
- Bioengineering,
University of California, Berkeley, Lawrence
Berkeley National Laboratory, Berkeley, California 94720, United States
| |
Collapse
|
41
|
Becker M, Maring JA, Schneider M, Herrera Martin AX, Seifert M, Klein O, Braun T, Falk V, Stamm C. Towards a Novel Patch Material for Cardiac Applications: Tissue-Specific Extracellular Matrix Introduces Essential Key Features to Decellularized Amniotic Membrane. Int J Mol Sci 2018; 19:E1032. [PMID: 29596384 PMCID: PMC5979550 DOI: 10.3390/ijms19041032] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 03/26/2018] [Accepted: 03/27/2018] [Indexed: 12/18/2022] Open
Abstract
There is a growing need for scaffold material with tissue-specific bioactivity for use in regenerative medicine, tissue engineering, and for surgical repair of structural defects. We developed a novel composite biomaterial by processing human cardiac extracellular matrix (ECM) into a hydrogel and combining it with cell-free amniotic membrane via a dry-coating procedure. Cardiac biocompatibility and immunogenicity were tested in vitro using human cardiac fibroblasts, epicardial progenitor cells, murine HL-1 cells, and human immune cells derived from buffy coat. Processing of the ECM preserved important matrix proteins as demonstrated by mass spectrometry. ECM coating did not alter the mechanical characteristics of decellularized amniotic membrane but did cause a clear increase in adhesion capacity, cell proliferation and viability. Activated monocytes secreted less pro-inflammatory cytokines, and both macrophage polarization towards the pro-inflammatory M1 type and T cell proliferation were prevented. We conclude that the incorporation of human cardiac ECM hydrogel shifts and enhances the bioactivity of decellularized amniotic membrane, facilitating its use in future cardiac applications.
Collapse
Affiliation(s)
- Matthias Becker
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany.
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353 Berlin, Germany.
| | - Janita A Maring
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany.
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353 Berlin, Germany.
| | - Maria Schneider
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany.
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353 Berlin, Germany.
| | - Aarón X Herrera Martin
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany.
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, 13353 Berlin, Germany.
| | - Martina Seifert
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany.
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353 Berlin, Germany.
| | - Oliver Klein
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany.
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353 Berlin, Germany.
| | - Thorsten Braun
- Department of Obstetrics and Gynecology, Charite Medical University, 13353 Berlin, Germany.
| | - Volkmar Falk
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353 Berlin, Germany.
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, 13316 Berlin, Germany.
- Deutsches Herzzentrum Berlin (DHZB), Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Christof Stamm
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany.
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353 Berlin, Germany.
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, 13316 Berlin, Germany.
- Deutsches Herzzentrum Berlin (DHZB), Augustenburger Platz 1, 13353 Berlin, Germany.
| |
Collapse
|
42
|
An injectable conductive hydrogel encapsulating plasmid DNA-eNOs and ADSCs for treating myocardial infarction. Biomaterials 2018; 160:69-81. [PMID: 29396380 DOI: 10.1016/j.biomaterials.2018.01.021] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 01/14/2018] [Indexed: 12/15/2022]
Abstract
Myocardial infarction (MI) leads to the mass death of cardiomyocytes accompanying with the unfavorable alternation of microenvironment, a fibrosis scar deprived of electrical communications, and the lack of blood supply in the infarcted myocardium. The three factors are inextricably intertwined and thus result in a conservative MI therapy efficacy in clinic. A holistic approach pertinently targeted to these three key points would be favorable to rebuild the heart functions. Here, an injectable conductive hydrogel was constructed via in situ Michael addition reaction between multi-armed conductive crosslinker tetraaniline-polyethylene glycol diacrylate (TA-PEG) and thiolated hyaluronic acid (HA-SH). The resultant soft conductive hydrogel with equivalent myocardial conductivity and anti-fatigue performance was loaded with plasmid DNA encoding eNOs (endothelial nitric oxide synthase) nanocomplexes and adipose derived stem cells (ADSCs) for treating MI. The TA-PEG/HA-SH/ADSCs/Gene hydrogel-based holistic system was injected into the infarcted myocardium of SD rats. We demonstrated an increased expression of eNOs in myocardial tissue the heightening of nitrite concentration, accompanied with upregulation of proangiogenic growth factors and myocardium related mRNA. The results of electrocardiography, cardiogram, and histological analysis convincingly revealed a distinct increase of ejection fraction (EF), shortened QRS interval, smaller infarction size, less fibrosis area, and higher vessel density, indicating a significant improvement of heart functions. This conception of combination approach by a conductive injectable hydrogel loaded with stem cells and gene-encoding eNOs nanoparticles will become a robust therapeutic strategy for the treatment of MI.
Collapse
|
43
|
Li W, Wu D, Tan J, Liu Z, Lu L, Zhou C. A gene-activating skin substitute comprising PLLA/POSS nanofibers and plasmid DNA encoding ANG and bFGF promotes in vivo revascularization and epidermalization. J Mater Chem B 2018; 6:6977-6992. [DOI: 10.1039/c8tb02006j] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
A gene-activated porous nanofibrous scaffold for effectively promoting vascularization, epidermalization and dermal wound healing by sustained release of dual plasmid DNAs.
Collapse
Affiliation(s)
- Wenqiang Li
- Department of Materials Science and Engineering
- Jinan University
- Guangzhou 510632
- China
- College of Life Science and Technology
| | - Dongwei Wu
- Department of Materials Science and Engineering
- Jinan University
- Guangzhou 510632
- China
| | - Jianwang Tan
- Department of Materials Science and Engineering
- Jinan University
- Guangzhou 510632
- China
| | - Zhibin Liu
- Department of Materials Science and Engineering
- Jinan University
- Guangzhou 510632
- China
| | - Lu Lu
- Department of Materials Science and Engineering
- Jinan University
- Guangzhou 510632
- China
- Engineering Research Center of Artificial Organs and Materials
| | - Changren Zhou
- Department of Materials Science and Engineering
- Jinan University
- Guangzhou 510632
- China
- Engineering Research Center of Artificial Organs and Materials
| |
Collapse
|
44
|
Yavvari PS, Pal S, Kumar S, Kar A, Awasthi AK, Naaz A, Srivastava A, Bajaj A. Injectable, Self-Healing Chimeric Catechol-Fe(III) Hydrogel for Localized Combination Cancer Therapy. ACS Biomater Sci Eng 2017; 3:3404-3413. [DOI: 10.1021/acsbiomaterials.7b00741] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Prabhu S. Yavvari
- Department
of Chemistry, Indian Institute of Science Education and Research, Bhopal By-pass Road, Bhauri, Bhopal 462066, Madhya Pradesh, India
| | - Sanjay Pal
- Laboratory
of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Cluster, 3rd Milestone Faridabad-Gurgaon
Expressway, Faridabad 121001, Haryana India
- Kalinga Institute of Industrial Technology, KIIT Road, Patia, Bhubaneswar 751024, Odisha, India
| | - Sandeep Kumar
- Laboratory
of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Cluster, 3rd Milestone Faridabad-Gurgaon
Expressway, Faridabad 121001, Haryana India
- Manipal University, Madhav Nagar,
Near Tiger Circle, Manipal 576104, Karnataka, India
| | - Animesh Kar
- Laboratory
of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Cluster, 3rd Milestone Faridabad-Gurgaon
Expressway, Faridabad 121001, Haryana India
| | - Anand Kumar Awasthi
- Department
of Chemistry, Indian Institute of Science Education and Research, Bhopal By-pass Road, Bhauri, Bhopal 462066, Madhya Pradesh, India
| | - Aaliya Naaz
- Laboratory
of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Cluster, 3rd Milestone Faridabad-Gurgaon
Expressway, Faridabad 121001, Haryana India
| | - Aasheesh Srivastava
- Department
of Chemistry, Indian Institute of Science Education and Research, Bhopal By-pass Road, Bhauri, Bhopal 462066, Madhya Pradesh, India
| | - Avinash Bajaj
- Laboratory
of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Cluster, 3rd Milestone Faridabad-Gurgaon
Expressway, Faridabad 121001, Haryana India
| |
Collapse
|
45
|
Xue J, Lin H, Bean A, Tang Y, Tan J, Tuan RS, Wang B. One-Step Fabrication of Bone Morphogenetic Protein-2 Gene-Activated Porous Poly-L-Lactide Scaffold for Bone Induction. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 7:50-59. [PMID: 29018836 PMCID: PMC5626914 DOI: 10.1016/j.omtm.2017.08.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 08/31/2017] [Indexed: 01/18/2023]
Abstract
Bone morphogenetic protein 2 (BMP2) is an efficacious inducer for the osteogenesis of mesenchymal stem cells (MSCs). Conventional applications of BMP2 have involved either the direct incorporation of BMP2 protein or ex vivo BMP2 gene transfer into stem cells prior to their transplantation. These approaches are able to promote bone formation to some extent; however, they are hampered by either the lack of stability and sustainability of BMP2 protein or the time-consuming and cost-prohibitive in vitro cell culture procedure. To overcome these limitations, we have developed a gene-activated poly-L-lactide acid (PLLA) scaffold with the encapsulation of recombinant adeno-associated viral (AAV) vector encoding a full-length cDNA of human BMP2 using an ice-based microparticle porogenization method that was recently developed. Results showed continuous release of AAV particles from the micropores of scaffolds for up to 1 week, subsequently transducing embedded human MSCs and producing functional BMP2. MSCs within scaffolds underwent efficacious osteogenesis, on the basis of osteoinductive gene expression and osteogenic differentiation, which resulted in robust new bone formation in vivo at 4 weeks. These findings show the potential of the technology toward developing clinical applications of a rapid, cost-effective, and potentially point-of-care approach for the repair of bone defects.
Collapse
Affiliation(s)
- Jingwen Xue
- Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.,Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.,School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Hang Lin
- Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.,Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Allison Bean
- Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.,Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Ying Tang
- Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.,Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Jian Tan
- Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.,Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.,Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.,McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Bing Wang
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.,McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| |
Collapse
|